Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cell Immunol ; 383: 104657, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36603504

RESUMO

Mature IL-33 (MIL33) acting through its receptor, ST2, is known to regulate fibrosis. The precursor, full-length IL-33 (FLIL33), may function differently from MIL33 and independently of ST2. Here we report that genetic deletion of either IL-33 or ST2 attenuates pulmonary fibrosis in the bleomycin model, as does Cre-induced IL-33 deficiency in response to either acute or chronic bleomycin challenge. However, adenovirus-mediated gene delivery of FLIL33, but not MIL33, to the lungs of either wild-type or ST2-deficient mice potentiates the profibrotic effect of bleomycin without inducing a Th2 phenotype. In cultured mouse lung cells, FLIL33 overexpression induces moderate and distinct transcriptomic changes compared with a robust response induced by MIL33, whereas ST2 deletion abrogates the effects of both IL-33 forms. Thus, FLIL33 may contribute to fibrosis in an ST2-independent, Th2-independent, non-transcriptomic fashion, suggesting that pharmacological targeting of both FLIL33 and MIL33 may prove efficacious in patients with pulmonary fibrosis.


Assuntos
Fibrose Pulmonar , Camundongos , Animais , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Interleucina-33/genética , Proteína 1 Semelhante a Receptor de Interleucina-1/genética , Fibrose , Bleomicina , Camundongos Endogâmicos C57BL
2.
Am J Respir Cell Mol Biol ; 66(2): 146-157, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34668840

RESUMO

Some previous studies in tissue fibrosis have suggested a profibrotic contribution from elevated expression of a protein termed either RGCC (regulator of cell cycle) or RGC-32 (response gene to complement 32 protein). Our analysis of public gene expression datasets, by contrast, revealed a consistent decrease in RGCC mRNA levels in association with pulmonary fibrosis. Consistent with this observation, we found that stimulating primary adult human lung fibroblasts with transforming growth factor (TGF)-ß in cell cultures elevated collagen expression and simultaneously attenuated RGCC mRNA and protein levels. Moreover, overexpression of RGCC in cultured lung fibroblasts attenuated the stimulating effect of TGF-ß on collagen levels. Similar to humans with pulmonary fibrosis, the levels of RGCC were also decreased in vivo in lung tissues of wild-type mice challenged with bleomycin in both acute and chronic models. Mice with constitutive RGCC gene deletion accumulated more collagen in their lungs in response to chronic bleomycin challenge than did wild-type mice. RNA-Seq analyses of lung fibroblasts revealed that RGCC overexpression alone had a modest transcriptomic effect, but in combination with TGF-ß stimulation, induced notable transcriptomic changes that negated the effects of TGF-ß, including on extracellular matrix-related genes. At the level of intracellular signaling, RGCC overexpression delayed early TGF-ß-induced Smad2/3 phosphorylation, elevated the expression of total and phosphorylated antifibrotic mediator STAT1, and attenuated the expression of a profibrotic mediator STAT3. We conclude that RGCC plays a protective role in pulmonary fibrosis and that its decline permits collagen accumulation. Restoration of RGCC expression may have therapeutic potential in pulmonary fibrosis.


Assuntos
Fibroblastos/metabolismo , Pulmão/metabolismo , Proteínas Nucleares/fisiologia , Fibrose Pulmonar/prevenção & controle , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta3/metabolismo , Animais , Ciclo Celular , Células Cultivadas , Feminino , Fibroblastos/patologia , Humanos , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Fibrose Pulmonar/etiologia , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Proteína Smad2/genética , Transcriptoma , Fator de Crescimento Transformador beta3/genética
3.
Cell Immunol ; 357: 104203, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32977155

RESUMO

IL-33 has emerged as a central mediator of immune, inflammatory, and fibrotic responses. Many studies have focused on mature IL-33, but elevated expression of the precursor, full-length IL-33 (FLIL33), has also been implicated in a spectrum of diseases, including tissue fibrosis. We previously reported and now confirmed that overexpression of FLIL33 induced phosphorylation of the key profibrotic signaling mediator of TGF-ß, Smad3, in primary human lung fibroblasts from healthy donors and idiopathic pulmonary fibrosis patients. Presently, we demonstrate that FLIL33-induced Smad3 phosphorylation was not abrogated by anti-TGF-ß antibody but was abrogated by ALK5/TGFBR1-specific and Smad3-specific inhibition, indicating that FLIL33 effect was independent of TGF-ß but dependent on its receptor, TGFBR. Western blotting analyses revealed that FLIL33 overexpression increased levels, but did not affect subcellular distribution, of the AP2A1 and AP2B1 subunits of the adaptor protein complex 2 (AP2), a known TGFBR binding partner. siRNA-mediated inhibition of these subunits blocked FLIL33-induced Smad3 phosphorylation, whereas AP2 subunit overexpression induced Smad3 phosphorylation even in the absence of FLIL33. RNA-Seq transcriptomic analyses revealed that fibroblast stimulation with TGF-ß induced major changes in expression levels of numerous genes, whereas overexpression of FLIL33 induced modest expression changes in a small number of genes. Furthermore, qRT-PCR tests demonstrated that despite inducing Smad3 phosphorylation, FLIL33 did not induce collagen gene transcription and even mildly attenuated TGF-ß-induced levels of collagen I and III mRNAs. We conclude that FLIL33 induces Smad3 phosphorylation through a TGF-ß-independent but TGF-ß receptor- and AP2- dependent mechanism and has limited downstream transcriptomic consequences.


Assuntos
Proteínas de Ligação a Ácido Graxo/metabolismo , Interleucina-33/metabolismo , Proteína Smad3/metabolismo , Adulto , Feminino , Fibroblastos/metabolismo , Fibrose/fisiopatologia , Humanos , Fibrose Pulmonar Idiopática/fisiopatologia , Masculino , Fosforilação , Ligação Proteica , Transporte Proteico , Receptor do Fator de Crescimento Transformador beta Tipo I/genética , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
4.
Cytokine ; 119: 1-6, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30856600

RESUMO

Proteolytic activation of the IL-33 precursor, full-length interleukin-33 (FLIL33), at multiple sites within the sensor domain (aa 95-109) yields several functionally mature (MIL33) forms. Unlike nuclear FLIL33, intracellular MIL33 occurs in the cytoplasm, is secreted from source cells, and exerts biological effects by activating the ST2 receptor on target cells. Previous studies and our findings in this report indicated that IL-33 forms that are substantially longer than those produced by cleavage within the sensor domain are biologically indistinguishable from classical MIL33. We utilized a series of human and mouse N-terminal FLIL33 mutants to narrow down the boundaries of the nuclear localization sequence to aa 46-67, a segment known to include a portion of the chromatin-binding motif as well as another site controlling intracellular stability of FLIL33 in an importin-5-dependent fashion. The N-terminal FLIL33 deletion mutants starting prior to this region were intranuclear, non-secreted in cell culture, and manifested modest functional activity in vivo, similar to FLIL33. By contrast, the mutants starting after this region were cytoplasmic, secreted from cells in culture, and overtly biologically active in vivo, similar to MIL33. The deletion mutants starting within this region manifested an intermediate phenotype between FLIL33 and MIL33. Thus, this segment of IL-33 molecule controls multiple aspects of its biology, including subcellular localization, extracellular secretion, and functional maturation into the longest possible form of mature IL-33 cytokine. Future anti-IL-33 therapies may be based on interfering with this segment, thus restraining extracellular release and maturation of IL-33 into the active cytokine.


Assuntos
Interleucina-33/metabolismo , Animais , Transporte Biológico/fisiologia , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica/fisiologia
5.
J Biol Chem ; 292(52): 21653-21661, 2017 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-29127199

RESUMO

Human mature IL-33 is a member of the IL-1 family and a potent regulator of immunity through its pro-T helper cell 2 activity. Its precursor form, full-length interleukin-33 (FLIL33), is an intranuclear protein in many cell types, including fibroblasts, and its intracellular levels can change in response to stimuli. However, the mechanisms controlling the nuclear localization of FLIL33 or its stability in cells are not understood. Here, we identified importin-5 (IPO5), a member of the importin family of nuclear transport proteins, as an intracellular binding partner of FLIL33. By overexpressing various FLIL33 protein segments and variants in primary human lung fibroblasts and HEK293T cells, we show that FLIL33, but not mature interleukin-33, physically interacts with IPO5 and that this interaction localizes to a cluster of charged amino acids (positions 46-56) but not to an adjacent segment (positions 61-67) in the FLIL33 N-terminal region. siRNA-mediated IPO5 knockdown in cell culture did not affect nuclear localization of FLIL33. However, the IPO5 knockdown significantly decreased the intracellular levels of overexpressed FLIL33, reversed by treatment with the 20S proteasome inhibitor bortezomib. Furthermore, FLIL33 variants deficient in IPO5 binding remained intranuclear and exhibited decreased levels, which were also restored by the bortezomib treatment. These results indicate that the interaction between FLIL33 and IPO5 is localized to a specific segment of the FLIL33 protein, is not required for nuclear localization of FLIL33, and protects FLIL33 from proteasome-dependent degradation.


Assuntos
Interleucina-33/metabolismo , beta Carioferinas/metabolismo , Sequência de Aminoácidos , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células HEK293 , Células HeLa , Humanos , Interleucina-33/genética , Sinais de Localização Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Proteólise , beta Carioferinas/genética
6.
Am J Physiol Lung Cell Mol Physiol ; 312(6): L945-L958, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28385812

RESUMO

Pulmonary fibrosis is a severe condition with no cure and limited therapeutic options. A better understanding of its pathophysiology is needed. Recent studies have suggested that pulmonary fibrosis may be driven by accelerated aging-related mechanisms. Sirtuins (SIRTs), particularly SIRT1, SIRT3, and SIRT6, are well-known mediators of aging; however, limited data exist on the contribution of sirtuins to lung fibrosis. We assessed the mRNA and protein levels of all seven known sirtuins in primary lung fibroblasts from patients with idiopathic pulmonary fibrosis (IPF) and systemic sclerosis-associated interstitial lung disease (SSc-ILD) in comparison with lung fibroblasts from healthy controls. These unbiased tests revealed a tendency for all sirtuins to be expressed at lower levels in fibroblasts from patients compared with controls, but the greatest decrease was observed with SIRT7. Similarly, SIRT7 was decreased in lung tissues of bleomycin-challenged mice. Inhibition of SIRT7 with siRNA in cultured lung fibroblasts resulted in an increase in collagen and α-smooth muscle actin (α-SMA). Reciprocally, overexpression of SIRT7 resulted in lower basal and TGF-ß-induced levels of COL1A1, COL1A2, COL3A1, and α-SMA mRNAs, as well as collagen and α-SMA proteins. Induced changes in SIRT7 had no effect on endogenous TGF-ß mRNA levels or latent TGF-ß activation, but overexpression of SIRT7 reduced the levels of Smad3 mRNA and protein. In conclusion, the decline in SIRT7 in lung fibroblasts has a profibrotic effect, which is mediated by changes in Smad3 levels.


Assuntos
Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Pulmão/patologia , Sirtuínas/metabolismo , Actinas/metabolismo , Adulto , Animais , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Colágeno/metabolismo , Derme/patologia , Feminino , Fibroblastos/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Recém-Nascido , Camundongos Endogâmicos C57BL , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sirtuínas/genética , Proteína Smad3/metabolismo , Frações Subcelulares/metabolismo , Fator de Crescimento Transformador beta/farmacologia
7.
J Immunol ; 194(7): 3127-35, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25716994

RESUMO

UV radiation (UVR) induces DNA damage, leading to the accumulation of mutations in epidermal keratinocytes and immunosuppression, which contribute to the development of nonmelanoma skin cancer. We reported previously that the TLR4-MyD88 signaling axis is necessary for UV-induced apoptosis. In the dinitrofluorobenzene contact hypersensitivity model, UV-irradiated MyD88-deficient (MyD88(-/-)) C57BL/6 mice had intact ear swelling, exaggerated inflammation, and higher levels of dinitrofluorobenzene-specific IgG2a compared with wild-type (WT) mice. Even with normal UV-induced, dendritic cell migration, DNA damage in the local lymph nodes was less pronounced in MyD88(-/-) mice compared with WT mice. Cultured, UV-irradiated WT APCs showed cleavage (inactivation) of the DNA damage-recognition molecule PARP, whereas PARP persisted in MyD88(-/-) and TLR4(-/-) APCs. Epidermal DNA from in vivo UV-irradiated MyD88(-/-) mice had an increased resolution rate of cyclobutane pyrimidine dimers. Both in vitro treatment of MyD88(-/-) APCs with and intradermal in vivo injections of PARP inhibitor, PJ-34, caused WT-level cyclobutane pyrimidine dimer repair. Lymphoblasts deficient in DNA repair (derived from a xeroderma pigmentosum group A patient) failed to augment DNA repair after MyD88 knockdown after UVR, in contrast to lymphoblasts from a healthy control. These data suggest that interference with the TLR4/MyD88 pathway may be a useful tool in promoting DNA repair and maintaining immune responses following UVR-induced damage.


Assuntos
Reparo do DNA , Terapia de Imunossupressão , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais/efeitos da radiação , Pele/imunologia , Pele/metabolismo , Receptor 4 Toll-Like/metabolismo , Raios Ultravioleta , Animais , Microambiente Celular/genética , Microambiente Celular/imunologia , Microambiente Celular/efeitos da radiação , Dano ao DNA/efeitos da radiação , Feminino , Humanos , Linfonodos/imunologia , Linfonodos/metabolismo , Linfonodos/efeitos da radiação , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Pele/efeitos da radiação , Receptor 4 Toll-Like/genética
8.
Exp Dermatol ; 25(12): 949-955, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27676214

RESUMO

UVR-induced apoptosis in cutaneous antigen presenting cells (APC) causes systemic immune suppression and is dependent on TLR4/MyD88 signalling, but the apoptotic signalling pathways have not been defined. Macrophages pretreated with lipopolysaccharide (LPS) were unresponsive to subsequent LPS treatment, however, but were susceptible to UVR-induced apoptosis. Macrophage survival and apoptotic events after UVR were also unaffected by treatment with TLR4 antagonists, a blocking IgG or a TLR4 analog antagonist, suggesting that UVR cell death is independent of a soluble ligand. After UVR, IRAK4KDKI (catalytically inactive IRAK4) and wild-type (WT) macrophages show equivalent levels of survival, as measured by MTT assay, and apoptosis, as measured by cleaved caspase-3. Furthermore, in macrophages from both mice, UVR activated caspase-8 and PARP, while inactivating Rip3. This finding is supported by a lack of IRAK1 degradation after UVR, compared to treatment with TLR2 or TLR4 agonists. UVR induced association of MyD88 with FADD, an extrinsic apoptotic pathway protein, but not IRAK4. UVR-induced migration of FADD to the cell membrane of WT macrophages, but not MyD88-/- macrophages, was observed (confocal microscopy). Co-immunoprecipitation using an epitope-tagged MyD88 revealed that FADD, but not TRADD, was recruited to MyD88 within 30 minutes of UVR exposure. UVR engages TLR4/MyD88 as a death signalling complex, rather than the classical inflammatory signalling pathway triggered by PAMP recognition of TLR4. These studies provide the rationale for the future development of topical TLR4 modulating therapies to interfere with this UVB-mediated apoptosis and the associated negative consequences of immune suppression.


Assuntos
Apoptose , Proteína de Domínio de Morte Associada a Fas/metabolismo , Macrófagos Peritoneais/efeitos da radiação , Fator 88 de Diferenciação Mieloide/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Células Cultivadas , Feminino , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Lipopolissacarídeos , Macrófagos Peritoneais/metabolismo , Camundongos Endogâmicos C57BL
9.
J Immunol ; 187(4): 1664-73, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21765012

RESUMO

Imiquimod is a TLR7/8 agonist that has anticancer therapeutic efficacy in the treatment of precancerous skin lesions and certain nonmelanoma skin cancers. To test our hypothesis that imiquimod enhances DNA repair as a mechanism for its anticancer activity, the nucleotide excision repair genes were studied in bone marrow-derived cells. Imiquimod enhanced the expression of xeroderma pigmentosum (XP) A and other DNA repair genes (quantitative real-time PCR analysis) and resulted in an increased nuclear localization of the DNA repair enzyme XPA. This was dependent on MyD88, as bone marrow-derived cells from MyD88(-/-) mice did not increase XPA gene expression and did not enhance the survival of MyD88(-/-)-derived bone marrow-derived cells after UV B exposure as was observed in bone marrow-derived cells from MyD88(+/+) mice. Imiquimod also enhanced DNA repair of UV light (UVL)-irradiated gene expression constructs and accelerated the resolution of cyclobutane pyrimidine dimers after UVL exposures in P388 and XS52. Lastly, topical treatment of mouse skin with 5% imiquimod cream prior to UVL irradiation resulted in a decrease in the number of cyclobutane pyridimine dimer-positive APC that were found in local lymph nodes 24 h after UVL irradiation in both wild-type and IL-12 gene-targeted mice. In total, these data support the idea that TLR7 agonists such as imiquimod enhance DNA repair in bone marrow-derived cells. This property is likely to be an important mechanism for its anticancer effects because it protects cutaneous APC from the deleterious effects of UVL.


Assuntos
Aminoquinolinas/farmacologia , Antineoplásicos/farmacologia , Células da Medula Óssea/imunologia , Dano ao DNA , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/imunologia , Transdução de Sinais , Neoplasias Cutâneas/imunologia , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/imunologia , Raios Ultravioleta/efeitos adversos , Animais , Células da Medula Óssea/metabolismo , Linhagem Celular , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/imunologia , Dano ao DNA/efeitos da radiação , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/genética , Reparo do DNA/imunologia , Reparo do DNA/efeitos da radiação , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/imunologia , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Imiquimode , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Dímeros de Pirimidina/genética , Dímeros de Pirimidina/imunologia , Dímeros de Pirimidina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Transdução de Sinais/efeitos da radiação , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/genética , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/metabolismo , Proteína de Xeroderma Pigmentoso Grupo A/biossíntese , Proteína de Xeroderma Pigmentoso Grupo A/genética , Proteína de Xeroderma Pigmentoso Grupo A/imunologia
10.
Exp Dermatol ; 21(12): 915-20, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23171451

RESUMO

Conventional CD8+ T-lymphocytes are thought to be major effector cells in allergic contact dermatitis (ACD). However, previous work has demonstrated a significant population of invariant natural killer T-cells (iNKT-cells) in the elicitation phase of ACD. In this study, we investigate whether iNKT-cells have the capacity to serve as effector lymphocytes in ACD. Using in situ staining of skin biopsy specimens from ACD lesions, we observed intra-epidermal iNKT-cells. Presence of these cells provides the possibility of interactions with keratinocytes (KC), Langerhans cells (LC) and CD1d-bearing antigen-presenting cells (APC). Investigation into gene expression profiles of cytotoxic effector molecules in seven different cases of ACD found that the expression of perforin and granzymes A, B and K were significantly elevated in ACD relative to paired clinically normal skin. Immunostaining of ACD skin biopsy specimens revealed that these cytotoxic granules indeed localized to iNKT-cells. Studies of antigen presentation of KC to iNKT-cells show that these epithelial cells do not activate the expression of cytotoxicity effector genes in resting iNKT-cells, but had the capacity to serve as targets for activated iNKT-cells, which was dependent on CD1d expression. Mature LC were not able to present glycolipids to iNKT-cells and did not up-regulate CD1d in vitro to a variety of maturational stimuli or in vivo during ACD. These data suggest that iNKT-cells can serve as effector cells during human ACD and provide the rationale for developing inhibitory glycolipids as therapeutic agents for ACD.


Assuntos
Antígenos CD1d/imunologia , Comunicação Celular/imunologia , Dermatite Alérgica de Contato/imunologia , Queratinócitos/imunologia , Células T Matadoras Naturais/imunologia , Adulto , Idoso , Antígenos CD1d/genética , Antígenos CD1d/metabolismo , Biópsia , Células Cultivadas , Grânulos Citoplasmáticos/imunologia , Grânulos Citoplasmáticos/metabolismo , Grânulos Citoplasmáticos/patologia , Dermatite Alérgica de Contato/metabolismo , Dermatite Alérgica de Contato/patologia , Epiderme/imunologia , Epiderme/metabolismo , Epiderme/patologia , Feminino , Glicolipídeos/imunologia , Glicolipídeos/metabolismo , Humanos , Queratinócitos/patologia , Células de Langerhans/imunologia , Células de Langerhans/patologia , Masculino , Pessoa de Meia-Idade , Células T Matadoras Naturais/patologia , Peptídeo Hidrolases/imunologia , Peptídeo Hidrolases/metabolismo , Perforina/imunologia , Perforina/metabolismo , RNA Interferente Pequeno/genética
11.
J Immunol ; 183(3): 1657-66, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19592659

RESUMO

CD1d is a nonclassical Ag-presenting molecule that presents glycolipid Ags to NKT cells that are involved in immune defense and tumor rejection. It also plays a role in immunoregulatory functions in the epidermis. The mechanisms controlling the expression of CD1d are not well understood. Therefore, we cloned the CD1d gene promoter and characterized its activities in primary human keratinocytes and other cell lines of epithelial origin. We found that a CCAAT box in the CD1d promoter is required for its expression in keratinocytes. We show here that transcription factor C/EBP-beta binds to the CCAAT box in the CD1d promoter in vitro and in vivo. Consistent with these observations, deletion of the gene encoding for C/EBP-beta caused a loss of CD1d expression. The in vivo regulation of CD1d has significant implications for the pathologic mechanisms of certain immunologic skin diseases in which NKT cells play a role, such as allergic contact dermatitis and psoriasis. Together, these data show a central role for C/EBP-beta in regulating CD1d transcription.


Assuntos
Antígenos CD1d/genética , Proteína beta Intensificadora de Ligação a CCAAT/fisiologia , Regulação da Expressão Gênica , Queratinócitos/metabolismo , Transcrição Gênica , Sítios de Ligação , Linhagem Celular , Clonagem Molecular , Células Epiteliais , Humanos , Células T Matadoras Naturais , Regiões Promotoras Genéticas , Dermatopatias/etiologia , Dermatopatias/imunologia
12.
J Dermatol Sci ; 53(2): 112-9, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19004620

RESUMO

BACKGROUND: Skin lesions commonly affect AIDS patients. The pathogenesis of certain dermatologic disorders primarily associated to HIV-1 is unclear, and better forms of therapy for these conditions need to be discovered. Transgenic animal models represent a novel approach for the study of these disorders and for the quest of more effective forms of treatment. OBJECTIVE: Characterize this HIV-1 transgenic rat as a model to study skin diseases related to HIV/AIDS. METHODS: A transgenic rat was developed, using an HIV-1 construct with deleted gag and pol genes. Morphological and genotypical evaluations were followed by cytokine profile characterization of the lesions. RESULTS: We report the characterization of a colony of HIV-1 transgenic rats that developed skin lesions in a frequency of 22.5%. Cutaneous expression of functional HIV-1 transgenes correlated precisely with the severity of the phenotype. In early stages, rats manifested localized areas of xerosis and dispersed papulosquamous lesions. These hyperplastic manifestations were observed in conjunction with an increased epidermal expression of tat protein and a Th1/Th2 profile of cytokines. As the lesions progressed, they formed inflammatory plaques that subsequently ulcerated. Histologically, these lesions displayed a profound lymphocytic infiltrate, epidermal necrosis, and a marked increase of both Th1 and Th2 derived cytokines. Moreover, the presence of circulating IgG antibodies against HIV-1 gp120 was detected. CONCLUSION: This animal model as other HIV-1 transgenic mice described in the past, is not able to fully explain the myriad of skin findings that can occur in HIV-infected humans; however, it represents a potential animal model system for the study of immune-mediated inflammatory skin diseases.


Assuntos
Dermatite/patologia , Epiderme/patologia , Infecções por HIV/complicações , HIV-1/genética , Animais , Citocinas/genética , Citocinas/metabolismo , Dermatite/imunologia , Dermatite/virologia , Modelos Animais de Doenças , Epiderme/imunologia , Epiderme/virologia , Eritema Multiforme/patologia , Eritema Multiforme/virologia , Genótipo , Anticorpos Anti-HIV/sangue , Proteína gp120 do Envelope de HIV/genética , Proteína gp120 do Envelope de HIV/imunologia , Infecções por HIV/imunologia , Infecções por HIV/patologia , Infecções por HIV/virologia , HIV-1/imunologia , Hiperplasia , Necrose , Fenótipo , RNA Mensageiro/metabolismo , Ratos , Ratos Transgênicos , Índice de Gravidade de Doença , Úlcera Cutânea/patologia , Úlcera Cutânea/virologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
13.
J Invest Dermatol ; 126(4): 821-31, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16439962

RESUMO

Imiquimod (1-(2-methylpropyl)-1H-imidazo[4,5-c]quinolin-4-amine) is a TLR7 agonist that induces cytokine production in TLR7 bearing antigen-presenting cells (APCs), including IL-12, a cytokine that has been demonstrated to be a critical effector molecule for contact hypersensitivity (CHS). To test our hypothesis that topical applications of imiquimod may protect the skin immune system against the deleterious effects of UV light exposures, we treated animals with this agent, or its vehicle or nothing before UV exposures. Although topical imiquimod exposures before UV light did not prevent the depletion of epidermal Langerhans cells, it did prevent the loss of CHS. IL-12 was important in the protective role of imiquimod in preventing UV-induced loss of CHS, as systemic treatment of mice with an anti-IL-12 p70 monoclonal antibody blocked the protective effects of imiquimod. Additionally, only imiquimod-treated mice were resistant to hapten-specific tolerance induction after UV irradiation at the site of the initial sensitization with the hapten 2,4 dinitro-1-fluorobenzene. To model for the effects of TLR7 activation on the UV effect on antigen-APCs, XS52 cell line was used to study this interaction in an in vitro model system. This cell line expressed mRNA for TLR7, downregulated IkappaB, phosphorylated c-Jun N-terminal kinase, and secreted cytokines after exposure to imiquimod or lipopolysaccharide. Activation of the TLR7 signaling pathway on XS52 before UV-light exposures enhanced IL-12p70 secretion by this cell line. Similarly, activation of TLR7 on XS52 before UV-light exposure also prevented the UV-induced loss of IFN-gamma triggering in T cells during an allogeneic mixed lymphocyte reaction. Imiquimod-treated, UV-irradiated XS52 triggered a more vigorous IFN-gamma production than did either imiquimod-treated XS52 or UV-irradiated XS52, again suggesting a synergy between the two treatments. Lastly, enriched lymph node CD11c+ APCs from mice treated with UV irradiation, imiquimod alone or the combination of UV irradiation and imiquimod indicated the same in vivo synergy between imiquimod irradiation and UV irradiation in enhancing IL-12p70 production. These data suggest that topical imiquimod applications may play a role in preventing UV-induced impairment of the skin immune system, which is thought to be one of the critical events that allow the development of UV-induced skin cancers.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Aminoquinolinas/administração & dosagem , Dermatite de Contato/prevenção & controle , Tolerância Imunológica/efeitos dos fármacos , Células de Langerhans/efeitos dos fármacos , Pele/efeitos dos fármacos , Raios Ultravioleta/efeitos adversos , Animais , Anticorpos Monoclonais/farmacologia , Feminino , Proteínas I-kappa B/metabolismo , Imiquimode , Interferon gama/metabolismo , Interleucina-12/antagonistas & inibidores , Interleucina-12/metabolismo , Células de Langerhans/efeitos da radiação , Glicoproteínas de Membrana/agonistas , Camundongos , Camundongos Endogâmicos C57BL , Pele/efeitos da radiação , Receptor 7 Toll-Like/agonistas
14.
J Dermatol Sci ; 74(3): 236-41, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24636351

RESUMO

BACKGROUND: UV-exposures result in accumulation of genetic lesions that facilitate the development of skin cancer. Numerous pharmacologic agents are currently under development to both inhibit formation of DNA lesions and enhance repair. Drugs must be evaluated in vitro, currently performed in cell culture systems, before being tested on humans. Current systems do not account for the architecture and diverse cellularity of intact human skin. OBJECTIVE: To establish a novel, functionally viable, and reproducible in vitro skin organ culture system for studying the effects of various pharmacologic agents on DNA repair. METHODS: Human skin was obtained from neonatal foreskins. Intact skin punches derived from foreskins were cultured in vitro prior to exposure to UV-irradiation, and evaluated for DNA-damage using a DNA dot blot. Serial skin biopsies were obtained from patients with actinic keratoses treated with topical imiquimod. Expression of immune-stimulating and DNA repair genes was evaluated in ex vivo and in vitro samples. RESULTS: DNA dot blots revealed active repair of UV induced lesions in our in vitro skin organ culture. The photo-protective effect of sunscreen was detected, while imiquimod treatment did not enhance DNA repair in vitro. The DNA repair molecules XPA and XPF were up-regulated in the skin of imiquimod treated patients with actinic keratoses and imiquimod treated bone marrow-derived cell lines, but not keratinocytes. CONCLUSION: Our in vitro human skin organ culture model detected repair of UV-induced DNA lesions, and may be easily adapted to investigate various photo-protective drugs intended to prevent or treat skin cancer.


Assuntos
Reparo do DNA , Técnicas de Cultura de Órgãos , Pele , Aminoquinolinas , Humanos , Imiquimode , Dímeros de Pirimidina/metabolismo , Raios Ultravioleta , Regulação para Cima
15.
Innate Immun ; 20(5): 529-39, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24048771

RESUMO

UV irradiation-induced cellular damage is classically associated with apoptosis and is known to result in systemic immunosuppression. How the decision to undergo apoptosis is made following UV is not fully understood. We hypothesize that a central mediator of TLR signaling, MyD88, determines cell fate after UV exposure. Survival after UV of immortalized bone marrow-derived macrophages (BMDM) and ex vivo peritoneal macrophages (PM) from MyD88 germline-deficient mice (MyD88(-/-)) was significantly higher than wild type (WT) PM. UV-induced apoptosis (DNA laddering) in PM and epidermis of MyD88(-/-) animals versus WT was decreased. In MyD88(-/-) PM, decreased cleavage of caspase 3, as well as pro-necroptotic protein, RIP1, and a significant increase in transcription and release of pro-inflammatory TNF-α, suggest that necroptosis, rather than apoptosis, has been initiated. In vivo studies confirm this hypothesis after UV, showing low apoptosis by TUNEL and inflammation in MyD88(-/-) skin sections. Considering that MyD88 participates in many TLR pathways, BMDM from TLR2(-/-), TLR4(-/-) and WT mice were compared for evidence of UV-induced apoptosis. Only TLR4(-/-) BMDM and PM had a similar phenotype to MyD88(-/-), suggesting that the TLR4-MyD88 axis importantly contributes to cell fate decision. Our study describes a new cellular consequence of MyD88 signaling after UV, and may provide rationale for therapies to mitigate UV-induced immunosuppression.


Assuntos
Apoptose/genética , Apoptose/efeitos da radiação , Morte Celular/genética , Morte Celular/efeitos da radiação , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/fisiologia , Necrose/genética , Animais , Células da Medula Óssea/efeitos da radiação , Caspase 3/biossíntese , Caspase 3/genética , Humanos , Terapia de Imunossupressão , Camundongos , Camundongos Endogâmicos C57BL , Receptor 4 Toll-Like/biossíntese , Receptor 4 Toll-Like/genética , Fator de Necrose Tumoral alfa/metabolismo , Raios Ultravioleta
16.
J Invest Dermatol ; 134(7): 1903-1911, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24487305

RESUMO

The cytokine IL-9, derived primarily from T-helper 9 (Th9) lymphocytes, promotes expansion of the Th2 subset and is implicated in the mechanisms of allergic asthma. We hypothesize that IL-9 also has a role in human allergic contact dermatitis (ACD). To investigate this hypothesis, skin biopsy specimens of positive patch-test sites from non-atopic patients were assayed using quantitative PCR and immunohistochemistry. The cytokines IFN-γ, IL-4, IL-17A, IL-9, and PU.1, a Th9 associated transcription factor, were elevated when compared with paired normal skin. Immunohistochemistry on ACD skin biopsies identified PU.1+ CD3+ and PU.1+ CD4+ cells, consistent with Th9 lymphocytes, in the inflammatory infiltrate. Peripheral blood mononuclear cells from nickel-allergic patients, but not nonallergic controls, show significant IL-9 production in response to nickel. Blocking studies with mAbs to HLA-DR (but not HLA-A, -B, -C) or chloroquine significantly reduced this nickel-specific IL-9 production. In addition, blockade of IL-9 or IL-4 enhanced allergen-specific IFN-γ production. A contact hypersensitivity model using IL-9(-/-) mice shows enhanced Th1 lymphocyte immune responses, when compared with wild-type mice, consistent with our human in vitro data. This study demonstrates that IL-9, through its direct effects on Th1 and ability to promote IL-4 secretion, has a regulatory role for Th1 lymphocytes in ACD.


Assuntos
Dermatite Alérgica de Contato/imunologia , Interleucina-9/imunologia , Níquel/imunologia , Células Th1/imunologia , Adulto , Idoso , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Dermatite Alérgica de Contato/genética , Dermatite Alérgica de Contato/metabolismo , Feminino , Humanos , Interleucina-4/imunologia , Interleucina-4/metabolismo , Interleucina-9/genética , Interleucina-9/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas/metabolismo , Células Th1/metabolismo , Transativadores/metabolismo
19.
J Invest Dermatol ; 128(6): 1460-9, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18079745

RESUMO

The purpose of this study is to identify invariant natural killer T cells (NKT cells) in cellular infiltrate of human allergic contact dermatitis (ACD) skin challenge sites. Skin biopsy specimens were taken from positive patch test reactions from 10 different patients (9 different allergens) and studied by immunochemistry, real-time PCR, nested PCR, and in situ hybridization to identify NKT cells and the cytokines associated with this cell type. Invariant NKT cells were identified in all the 10 skin biopsy specimens studied, ranging from 1.72 to 33% of the cellular infiltrate. These NKT cells were activated in all cases, as they expressed cytokine transcripts for IFN-gamma and IL-4. Invariant NKT cells are present in ACD, regardless of the allergen that triggers the reaction, and are in an activated state. We conclude that innate immunity plays a role in late phases of type IV hypersensitivity reactions and may be responding to self-lipids released during allergic inflammation. These data complement the previous work by other investigators that suggest that NKT cells are important in the early cellular response during primary immune responses to allergens. Herein, it is demonstrated that NKT cells are constantly present during the late elicitation phase of human type IV hypersensitivity reactions.


Assuntos
Dermatite Alérgica de Contato/patologia , Células Matadoras Naturais/citologia , Pele/patologia , Linfócitos T/citologia , Adulto , Idoso , Células Apresentadoras de Antígenos/imunologia , Biópsia , Feminino , Humanos , Imunidade Inata , Interferon gama/metabolismo , Interleucina-4/metabolismo , Masculino , Pessoa de Meia-Idade
20.
J Invest Dermatol ; 128(9): 2190-7, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18385757

RESUMO

PKCzeta (protein kinase C-zeta), a member of protein kinase C family, plays an important role in cell proliferation, differentiation, and apoptosis. It acts as a downstream molecule for TNF-alpha (tumor necrosis factor) signal transduction and also regulates the expression of CD1d, an HLA-class I-like molecule. The interaction of CD1d with natural killer T (NKT) cells has been shown to be important in their Th1 cytokine production in psoriasis. In this study, we examined PKCzeta in psoriasis in order to define its role in the pathogenesis of the disease. We found that T-cell receptor (TCR) V alpha24+ V beta11+ NKT cells and CD1d molecules within psoriatic skin were increased. Moreover, there was an associated increase in PKCzeta mRNA and protein expression with membrane translocation in psoriasis lesions compared to uninvolved skin. Furthermore, cultured keratinocytes exhibited increased PKCzeta activity and membrane translocation upon stimulation by TNF-alpha, a cytokine known to play an important role in the pathogenesis of psoriasis. These results implied that PKCzeta is an important transduction molecule downstream of TNF-alpha signaling and is associated with increased expression of CD1d that may enhance CD1d-NKT cell interactions in psoriasis lesions. This makes PKCzeta a tempting target for possible pharmacological intervention in modifying the downstream effects of TNF-alpha in psoriasis.


Assuntos
Queratinócitos/enzimologia , Proteína Quinase C/metabolismo , Psoríase/enzimologia , Antígenos CD1/metabolismo , Antígenos CD1d , Linhagem Celular , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/patologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Masculino , Psoríase/etiologia , Psoríase/patologia , RNA Mensageiro/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa