Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Drug Metab Dispos ; 45(10): 1093-1100, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28790147

RESUMO

The epidermal growth factor receptor (EGFR) regulates cellular expression levels of breast cancer resistance protein (humans: ABCG2, rodents: Abcg2) via its downstream signaling pathways. Drugs that inhibit EGFR signaling (e.g., tyrosine kinase inhibitors, antibodies) may lead to ABCG2-mediated drug-drug interactions (DDIs) by changing the disposition of concomitantly administered ABCG2 substrate drugs. In this study, we used positron emission tomography and magnetic resonance imaging to compare disposition of the model Abcg2 substrate [11C]erlotinib in a mouse model of hepatocyte-specific deletion of EGFR (EGFR∆hep mice, n = 5) with EGFRfl/fl control mice (n = 6), which have normal EGFR expression levels in all tissues. Integration plot analysis was used to estimate the rate constants for transfer of radioactivity from the liver into bile (kbile) and from the kidney into urine (kurine). EGFR∆hep mice showed significantly lower radioactivity concentrations in the intestine (1.6-fold) and higher radioactivity concentrations in the urinary bladder (3.2-fold) compared with EGFRfl/fl mice. Kbile was significantly decreased (3.0-fold) in EGFR∆hep mice, whereas kurine was by 2.2-fold increased. Western blot analysis of liver tissue confirmed deletion of EGFR and showed significant decreases in Abcg2 and increases in P-glycoprotein (Abcb1a/b) expression levels in EGFR∆hep versus EGFRfl/fl mice. Our data show that EGFR deletion in hepatocytes leads to a reduction in Abcg2-mediated hepatobiliary clearance of a probe substrate accompanied by a shift to renal excretion of the drug, which raises the possibility that EGFR-inhibiting drugs may cause ABCG2-mediated DDIs.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Receptores ErbB/metabolismo , Cloridrato de Erlotinib/metabolismo , Hepatócitos/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Isótopos de Carbono/metabolismo , Interações Medicamentosas/fisiologia , Mucosa Intestinal/metabolismo , Rim/metabolismo , Fígado/metabolismo , Camundongos , Tomografia por Emissão de Pósitrons/métodos , Inibidores de Proteínas Quinases/metabolismo , Eliminação Renal/fisiologia , Transdução de Sinais/fisiologia
2.
Exp Dermatol ; 24(7): 522-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25828150

RESUMO

Chemokines are small chemotactic proteins that have a crucial role in leukocyte recruitment into tissue. Targeting these mediators has been suggested as a potential therapeutic option in inflammatory skin diseases such as psoriasis. Using quantitative RT-PCR, we found CCL7, a chemokine ligand known to interact with multiple C-C chemokine receptors, to be markedly increased in lesional psoriasis as opposed to atopic dermatitis, lichen planus, non-lesional psoriatic and normal control skin. Surprisingly, this increase in CCL7 mRNA expression exceeded that of all other chemokines investigated, and keratinocytes and dermal blood endothelial cells were identified as its likely cellular sources. In an imiquimod-induced psoriasis-like mouse model, CCL7 had a profound impact on myeloid cell inflammation as well as on the upregulation of key pro-psoriatic cytokines such as CCL20, IL-12p40 and IL-17C, while its blockade led to an increase in the antipsoriatic cytokine IL-4. In humans receiving the TNF-α-blocker infliximab, CCL7 was downregulated in lesional psoriatic skin already within 16 hours after a single intravenous infusion. These data suggest that CCL7 acts as a driver of TNF-α-dependent Th1/Th17-mediated inflammation in lesional psoriatic skin.


Assuntos
Quimiocina CCL7/metabolismo , Psoríase/etiologia , Fator de Necrose Tumoral alfa/metabolismo , Adolescente , Adulto , Animais , Estudos de Casos e Controles , Quimiocina CCL7/genética , Dermatite Atópica/imunologia , Modelos Animais de Doenças , Regulação para Baixo , Células Endoteliais/imunologia , Humanos , Mediadores da Inflamação/metabolismo , Infliximab/farmacologia , Interleucina-1beta/metabolismo , Queratinócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Psoríase/imunologia , Psoríase/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Dermatopatias/imunologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Adulto Jovem
3.
J Invest Dermatol ; 142(9): 2446-2454.e3, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35300973

RESUMO

The cytokine TGFß1 induces epidermal Langerhans cell (LC) differentiation from human precursors, an effect mediated through BMPR1a/ALK3 signaling, as revealed from ectopic expression and receptor inhibition studies. Whether TGFß1‒BMPR1a signaling is required for LC differentiation in vivo remained incompletely understood. We found that TGFß1-deficient mice show defective perinatal expansion and differentiation of LCs. LCs can be identified within the normal healthy human epidermis by anti-BMPR1a immunohistology staining. Deletion of BMPR1a in all (vav+) hematopoietic cells revealed that BMPR1a is required for the efficient TGFß1-dependent generation of CD207+ LC-like cells from CD11c+ intermediates in vitro. Similarly, BMPR1a was required for the optimal induction of CD207 by preformed major histocompatibility complex II‒positive epidermal resident LC precursors in the steady state. BMPR1a expression is strongly upregulated in epidermal cells in psoriatic lesions, and BMPR1aΔCD11c mice showed a defect in the resolution phase of allergic and psoriatic skin inflammation. Moreover, whereas LCs from these mice expressed CD207, BMPR1a counteracted LC activation and migration from skin explant cultures. Therefore, TGFß1‒BMPR1a signaling seems to be required for the efficient induction of CD207 during LC differentiation in the steady state, and bone marrow‒derived lesional CD11c+ cells may limit established skin inflammation through enhanced BMPR1a signaling.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I , Dermatite , Células de Langerhans , Animais , Antígenos CD/metabolismo , Antígenos de Superfície , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Antígenos CD11 , Antígeno CD11c/metabolismo , Diferenciação Celular , Dermatite/metabolismo , Epiderme/metabolismo , Inflamação/metabolismo , Células de Langerhans/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Lectinas de Ligação a Manose/metabolismo , Camundongos
4.
Sci Adv ; 6(2): eaax5849, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31934626

RESUMO

Psoriasis is a common chronic skin disorder characterized by keratinocyte hyperproliferation with altered differentiation accompanied by inflammation and increased angiogenesis. It remains unclear whether the first events that initiate psoriasis development occur in keratinocytes or inflammatory cells. Here, using different psoriasis mouse models, we showed that conditional deletion of Flt1 or Nrp1 in epidermal cells inhibited psoriasis mediated by Vegfa overexpression or c-Jun/JunB deletion. Administration of anti-Nrp1 antibody reverted the psoriasis phenotype. Using transcriptional and chromatin profiling of epidermal cells following Vegfa overexpression together with Flt1 or Nrp1 deletion, we identified the gene regulatory network regulated by Vegfa/Nrp1/Flt1 during psoriasis development and uncovered a key role of Fosl1 in regulating the chromatin remodeling mediated by Vegfa overexpression in keratinocytes. In conclusion, our study identifies an epidermal autonomous function of Vegfa/Nrp1/Flt1 that mediates psoriatic-like disease and demonstrates the clinical relevance of blocking Vegfa/Nrp1/Flt1 axis in psoriasis.


Assuntos
Células Epidérmicas/metabolismo , Neuropilina-1/metabolismo , Psoríase/metabolismo , Psoríase/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Anticorpos Bloqueadores/farmacologia , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Células Epidérmicas/efeitos dos fármacos , Células Epidérmicas/patologia , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Camundongos Knockout , Fenótipo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Psoríase/genética , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos
5.
Commun Biol ; 3(1): 252, 2020 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-32444775

RESUMO

Tumors have evolved mechanisms to escape anti-tumor immunosurveillance. They limit humoral and cellular immune activities in the stroma and render tumors resistant to immunotherapy. Sensitizing tumor cells to immune attack is an important strategy to revert immunosuppression. However, the underlying mechanisms of immune escape are still poorly understood. Here we discover Indoleamine-2,3-dioxygenase-1 (IDO1)+ Paneth cells in the stem cell niche of intestinal crypts and tumors, which promoted immune escape of colorectal cancer (CRC). Ido1 expression in Paneth cells was strictly Stat1 dependent. Loss of IDO1+ Paneth cells in murine intestinal adenomas with tumor cell-specific Stat1 deletion had profound effects on the intratumoral immune cell composition. Patient samples and TCGA expression data suggested corresponding cells in human colorectal tumors. Thus, our data uncovered an immune escape mechanism of CRC and identify IDO1+ Paneth cells as a target for immunotherapy.


Assuntos
Neoplasias Colorretais/patologia , Tolerância Imunológica/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Neoplasias Intestinais/patologia , Celulas de Paneth/imunologia , Fator de Transcrição STAT1/fisiologia , Animais , Neoplasias Colorretais/etiologia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Neoplasias Intestinais/imunologia , Neoplasias Intestinais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
6.
J Anat ; 215(1): 60-8, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19467150

RESUMO

The insulin/insulin-like growth factor (IGF) system regulates fetal and placental growth and development. In maternal diabetes, components of this system including insulin, IGF1, IGF2 and various IGF-binding proteins are deregulated in the maternal or fetal circulation, or in the placenta. The placenta expresses considerable amounts of insulin and IGF1 receptors at distinct locations on both placental surfaces. This makes the insulin and the IGF1 receptor accessible to fetal and/or maternal insulin, IGF1 and IGF2. Unlike the receptor for IGF1, the insulin receptor undergoes a gestational change in expression site from the trophoblast at the beginning of pregnancy to the endothelium at term. Insulin and IGFs are implicated in the receptor-mediated regulation of placental growth and transport, trophoblast invasion and placental angiogenesis. The dysregulation of the growth factors and their receptors may be involved in placental and fetal changes observed in diabetes, i.e. enhanced placental and fetal growth, placental hypervascularization and higher levels of fetal plasma amino acids.


Assuntos
Insulina/fisiologia , Placentação , Gravidez em Diabéticas/fisiopatologia , Somatomedinas/fisiologia , Feminino , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/fisiologia , Placenta/metabolismo , Gravidez , Receptor IGF Tipo 1/metabolismo
7.
EMBO Mol Med ; 10(11)2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30361264

RESUMO

Osteosarcoma (OS) is a rare tumor of the bone occurring mainly in young adults accounting for 5% of all childhood cancers. Because of the limited therapeutic options, there has been no survival improvement for OS patients in the past 40 years. The epidermal growth factor receptor (EGFR) is highly expressed in OS; however, its clinical relevance is unclear. Here, we employed an autochthonous c-Fos-dependent OS mouse model (H2-c-fosLTR) and human OS tumor biopsies for preclinical studies aimed at identifying novel biomarkers and therapeutic benefits of anti-EGFR therapies. We show that EGFR deletion/inhibition results in reduced tumor formation in H2-c-fosLTR mice by directly inhibiting the proliferation of cancer-initiating osteoblastic cells by a mechanism involving RSK2/CREB-dependent c-Fos expression. Furthermore, OS patients with co-expression of EGFR and c-Fos exhibit reduced overall survival. Preclinical studies using human OS xenografts revealed that only tumors expressing both EGFR and c-Fos responded to anti-EGFR therapy demonstrating that c-Fos can be considered as a novel biomarker predicting response to anti-EGFR treatment in OS patients.


Assuntos
Neoplasias Ósseas/metabolismo , Carcinogênese/metabolismo , Carcinogênese/patologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Receptores ErbB/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Transdução de Sinais , Animais , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Receptores ErbB/genética , Deleção de Genes , Humanos , Ligantes , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Osteoblastos/metabolismo , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-fos/genética , Análise de Sobrevida
8.
Cell Death Differ ; 25(6): 1094-1106, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29445126

RESUMO

Mice deficient in epidermal growth factor receptor (Egfr-/- mice) are growth retarded and exhibit severe bone defects that are poorly understood. Here we show that EGFR-deficient mice are osteopenic and display impaired endochondral and intramembranous ossification resulting in irregular mineralization of their bones. This phenotype is recapitulated in mice lacking EGFR exclusively in osteoblasts, but not in mice lacking EGFR in osteoclasts indicating that osteoblasts are responsible for the bone phenotype. Experiments are presented demonstrating that signaling via EGFR stimulates osteoblast proliferation and inhibits their differentiation by suppression of the IGF-1R/mTOR-pathway via ERK1/2-dependent up-regulation of IGFBP-3. Osteoblasts from Egfr-/- mice show increased levels of IGF-1R and hyperactivation of mTOR-pathway proteins, including enhanced phosphorylation of 4E-BP1 and S6. The same changes are also seen in Egfr-/- bones. Importantly, pharmacological inhibition of mTOR with rapamycin decreases osteoblasts differentiation as well as rescues the low bone mass phenotype of Egfr-/- fetuses. Our results demonstrate that suppression of the IGF-1R/mTOR-pathway by EGFR/ERK/IGFBP-3 signaling is necessary for balanced osteoblast maturation providing a mechanism for the skeletal phenotype observed in EGFR-deficient mice.


Assuntos
Desenvolvimento Ósseo , Proliferação de Células , Receptores ErbB/metabolismo , Osteoblastos/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Animais , Receptores ErbB/genética , Camundongos , Camundongos Knockout , Osteoblastos/citologia , Serina-Treonina Quinases TOR/genética
9.
FEBS J ; 285(17): 3175-3196, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30028091

RESUMO

Mice lacking the epidermal growth factor receptor (EGFR) develop an early postnatal degeneration of the frontal cortex and olfactory bulbs and show increased cortical astrocyte apoptosis. The poor health and early lethality of EGFR-/- mice prevented the analysis of mechanisms responsible for the neurodegeneration and function of the EGFR in the adult brain. Here, we show that postnatal EGFR-deficient neural stem cells are impaired in their self-renewal potential and lack clonal expansion capacity in vitro. Mice lacking the EGFR in the brain (EGFRΔbrain ) show low penetrance of cortical degeneration compared to EGFR-/- mice despite genetic recombination of the conditional allele. Adult EGFRΔ mice establish a proper blood-brain barrier and perform reactive astrogliosis in response to mechanical and infectious brain injury, but are more sensitive to Kainic acid-induced epileptic seizures. EGFR-deficient cortical astrocytes, but not midbrain astrocytes, have reduced expression of glutamate transporters Glt1 and Glast, and show reduced glutamate uptake in vitro, illustrating an excitotoxic mechanism to explain the hypersensitivity to Kainic acid and region-specific neurodegeneration observed in EGFR-deficient brains.


Assuntos
Astrócitos/patologia , Encéfalo/patologia , Receptores ErbB/fisiologia , Ácido Glutâmico/metabolismo , Hipersensibilidade/complicações , Células-Tronco Neurais/patologia , Convulsões/etiologia , Animais , Astrócitos/metabolismo , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Encéfalo/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Convulsões/patologia
10.
Cancer Cell ; 33(6): 1061-1077.e6, 2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29894692

RESUMO

How fully differentiated cells that experience carcinogenic insults become proliferative cancer progenitors that acquire multiple initiating mutations is not clear. This question is of particular relevance to hepatocellular carcinoma (HCC), which arises from differentiated hepatocytes. Here we show that one solution to this problem is provided by CD44, a hyaluronic acid receptor whose expression is rapidly induced in carcinogen-exposed hepatocytes in a STAT3-dependent manner. Once expressed, CD44 potentiates AKT activation to induce the phosphorylation and nuclear translocation of Mdm2, which terminates the p53 genomic surveillance response. This allows DNA-damaged hepatocytes to escape p53-induced death and senescence and respond to proliferative signals that promote fixation of mutations and their transmission to daughter cells that go on to become HCC progenitors.


Assuntos
Carcinoma Hepatocelular/genética , Receptores de Hialuronatos/genética , Neoplasias Hepáticas/genética , Proteína Supressora de Tumor p53/genética , Idoso , Animais , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Células Cultivadas , Dano ao DNA , Feminino , Regulação Neoplásica da Expressão Gênica , Hepatócitos/metabolismo , Humanos , Receptores de Hialuronatos/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Transdução de Sinais/genética , Proteína Supressora de Tumor p53/metabolismo
11.
Methods Mol Biol ; 1267: 217-50, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25636471

RESUMO

The skin is the largest organ of the mammalian body, made up of multiple layers, which include the epidermis, dermis, and subcutis (Alam and Ratner, N Engl J Med 344(13):975-983, 2001). The human interfollicular epidermis can be subdivided into five different layers: (1) stratum basale, (2) stratum spinosum, (3) stratum granulosum, (4) stratum lucidum, and (5) stratum corneum, all originating from basal keratinocytes by differentiation (Hameetman et al., BMC cancer 13:58, 2013; Ramirez et al., Differentiation 58(1):53-64, 1994). The epidermis is also able to generate different appendages: hair follicles (HF) and their associated sebaceous glands (Sibilia et al., Cell 102(2):211-220, 2000) as well as sweat glands (Luetteke et al., Genes Dev 8(4):399-413, 1994). The skin has important functions in several biological processes like environmental barrier, tissue regeneration, hair cycling, and wound repair. During these processes, stem cells from the interfollicular epidermis and from the hair follicle bulge are activated to renew the epidermis or hair. The epidermis and hair undergo continuous homeostatic regeneration and mutations, upon mutations which disturb the balance of homeostatic regeneration of epidermis and hair and lead to enhanced proliferation of keratinocytes, development of skin cancer is developed. Tumors that arise in the skin are mainly of three types: malignant melanoma, arising from melanocytes, basal cell carcinoma (BCC), and squamous cell carcinoma (SCC), the latter two both arising from keratinocytes or hair follicle cells. In this chapter, we will describe some genetically engineered mouse models (GEMM) that aim at modeling human BCC and SCC and their respective precancerous lesions. We will describe the experimental approaches used in our laboratory to analyze tumor-bearing mice focusing on methods necessary for the induction of tumor growth as well as for the molecular and histological analysis of tumor tissue.


Assuntos
Neoplasias Cutâneas , 9,10-Dimetil-1,2-benzantraceno/farmacologia , Animais , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Técnicas de Cultura de Células , DNA/genética , DNA/isolamento & purificação , Epiderme/efeitos dos fármacos , Epiderme/patologia , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/patologia , Camundongos , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Coloração e Rotulagem , Tamoxifeno/farmacologia , Acetato de Tetradecanoilforbol/farmacologia
12.
EMBO Mol Med ; 6(10): 1312-27, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25216727

RESUMO

Several subtypes of APCs are found in psoriasis patients, but their involvement in disease pathogenesis is poorly understood. Here, we investigated the contribution of Langerhans cells (LCs) and plasmacytoid DCs (pDCs) in psoriasis. In human psoriatic lesions and in a psoriasis mouse model (DKO* mice), LCs are severely reduced, whereas pDCs are increased. Depletion of pDCs in DKO* mice prior to psoriasis induction resulted in a milder phenotype, whereas depletion during active disease had no effect. In contrast, while depletion of Langerin-expressing APCs before disease onset had no effect, depletion from diseased mice aggravated psoriasis symptoms. Disease aggravation was due to the absence of LCs, but not other Langerin-expressing APCs. LCs derived from DKO* mice produced increased IL-10 levels, suggesting an immunosuppressive function. Moreover, IL-23 production was high in psoriatic mice and further increased in the absence of LCs. Conversely, pDC depletion resulted in reduced IL-23 production, and therapeutic inhibition of IL-23R signaling ameliorated disease symptoms. Therefore, LCs have an anti-inflammatory role during active psoriatic disease, while pDCs exert an instigatory function during disease initiation.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Células Dendríticas/imunologia , Células de Langerhans/imunologia , Psoríase/imunologia , Adjuvantes Imunológicos/farmacologia , Aminoquinolinas/farmacologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Células Apresentadoras de Antígenos/metabolismo , Transplante de Medula Óssea , Células Dendríticas/metabolismo , Progressão da Doença , Citometria de Fluxo , Humanos , Imiquimode , Interleucina-10/imunologia , Interleucina-10/metabolismo , Interleucina-23/imunologia , Interleucina-23/metabolismo , Células de Langerhans/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Proteínas Proto-Oncogênicas c-jun/deficiência , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/imunologia , Psoríase/genética , Psoríase/prevenção & controle , Receptores de Interleucina/imunologia , Receptores de Interleucina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Pele/efeitos dos fármacos , Pele/imunologia , Pele/patologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia
14.
Diabetes ; 57(1): 150-7, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17928399

RESUMO

OBJECTIVE: In pregestational diabetes, the placenta at term of gestation is characterized by various structural and functional changes. Whether similar alterations occur in the first trimester has remained elusive. Placental development requires proper trophoblast invasion and tissue remodeling, processes involving matrix metalloproteinases (MMPs) of which the membrane-anchored members (MT-MMPs) such as MT1-MMPs are key players. Here, we hypothesize a dysregulation of placental MT1-MMP in the first trimester of type 1 diabetic pregnancies induced by the diabetic environment. RESEARCH DESIGN AND METHODS: MT1-MMP protein was measured in first-trimester placentas of healthy (n = 13) and type 1 diabetic (n = 13) women. To identify potential regulators, first-trimester trophoblasts were cultured under hyperglycemia and various insulin, IGF-I, IGF-II, and tumor necrosis factor-alpha (TNF-alpha) concentrations in presence or absence of signaling pathway inhibitors. RESULTS: MT1-MMP was strongly expressed in first-trimester trophoblasts. In type 1 diabetes, placental pro-MT1-MMP was upregulated, whereas active MT1-MMP expression was only increased in late first trimester. In isolated primary trophoblasts, insulin, IGF-I, IGF-II, and TNF-alpha upregulated MT1-MMP expression, whereas glucose had no effect. The insulin effect was dependent on phosphatidylinositol 3-kinase, the IGF-I effect on mitogen-activated protein kinase, and the IGF-II effect on both. CONCLUSIONS: This is the first study reporting alterations in the first-trimester placenta in type 1 diabetes. The upregulated MT1-MMP expression in type 1 diabetes may be the result of higher maternal insulin and TNF-alpha levels. We speculate that the elevated MT1-MMP will affect placental development and may thus contribute to long-term structural alterations in the placenta in pregestational diabetes.


Assuntos
Diabetes Mellitus Tipo 1/genética , Regulação Enzimológica da Expressão Gênica , Insulina/sangue , Metaloproteinase 14 da Matriz/genética , Placenta/enzimologia , Complicações na Gravidez/genética , Fator de Necrose Tumoral alfa/sangue , Aborto Espontâneo/enzimologia , Aborto Espontâneo/genética , Técnicas de Cultura de Células , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/enzimologia , Feminino , Humanos , Gravidez , Complicações na Gravidez/enzimologia , Primeiro Trimestre da Gravidez , Valores de Referência , Trofoblastos/citologia , Trofoblastos/enzimologia , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA