Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Biomedicines ; 12(4)2024 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-38672274

RESUMO

Oral squamous cell carcinoma (OSCC) presents significant treatment challenges due to its poor survival and intense pain at the primary cancer site. Cancer pain is debilitating, contributes to diminished quality of life, and causes opioid tolerance. The stimulator of interferon genes (STING) agonism has been investigated as an anti-cancer strategy. We have developed STINGel, an extended-release formulation that prolongs the availability of STING agonists, which has demonstrated an enhanced anti-tumor effect in OSCC compared to STING agonist injection. This study investigates the impact of intra-tumoral STINGel on OSCC-induced pain using two separate OSCC models and nociceptive behavioral assays. Intra-tumoral STINGel significantly reduced mechanical allodynia in the orofacial cancer model and alleviated thermal and mechanical hyperalgesia in the hind paw model. To determine the cellular signaling cascade contributing to the antinociceptive effect, we performed an in-depth analysis of immune cell populations via single-cell RNA-seq. We demonstrated an increase in M1-like macrophages and N1-like neutrophils after STINGel treatment. The identified regulatory pathways controlled immune response activation, myeloid cell differentiation, and cytoplasmic translation. Functional pathway analysis demonstrated the suppression of translation at neuron synapses and the negative regulation of neuron projection development in M2-like macrophages after STINGel treatment. Importantly, STINGel treatment upregulated TGF-ß pathway signaling between various cell populations and peripheral nervous system (PNS) macrophages and enhanced TGF-ß signaling within the PNS itself. Overall, this study sheds light on the mechanisms underlying STINGel-mediated antinociception and anti-tumorigenic impact.

2.
ACS Biomater Sci Eng ; 7(2): 415-421, 2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33470801

RESUMO

We evaluated a peptide-based immunotherapy termed SynerGel: an injectable, biomaterial-based platform for intratumoral drug delivery. A drug-mimicking peptide hydrogel named L-NIL-MDP was loaded with an antitumor cyclic dinucleotide (CDN) immunotherapy agonist. The biomaterial combines inducible nitric oxide synthase (iNOS) inhibition with controlled delivery of CDNs, demonstrating between 4- and 20-fold slower drug release than commercially available hydrogels. SynerGel allowed for immune-mediated elimination of established treatment-resistant oral tumors in a murine model, with a median survival of 67.5 days compared with 44 days in no-treatment control. This report details findings for a promising therapy showing improved efficacy over previous hydrogel systems.


Assuntos
Materiais Biocompatíveis , Neoplasias Bucais , Animais , Liberação Controlada de Fármacos , Hidrogéis , Imunoterapia , Camundongos , Neoplasias Bucais/terapia
3.
J Cell Biochem ; 108(4): 802-15, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19711367

RESUMO

Understanding the underlying mechanisms by which a normal cell avoids the oncogenic potential of MUC1 signaling requires further definition of the pathways by which the MUC1 cytoplasmic tail is processed in both normal and tumor-derived cells. In the present study we describe the processing pathway initiated by TACE/ADAM17 cleavage of MUC1. Utilizing the human uterine epithelial cell line, HES, derived from normal endometrium, we show that endogenous full length MUC1 undergoes regulated intramembranous proteolysis mediated by presenillin-dependent gamma-secretase. Cytokine-stimulated HES cells exposed to gamma-secretase inhibitors accumulated a membrane-associated 15 kDa fragment of the MUC1 C-terminal subunit (CTF15). Inhibitors of TACE/ADAM17-mediated shedding inhibited accumulation of MUC1-CTF15 and MUC1 ectodomain release to a similar extent consistent with MUC1-CTF15 being a product of TACE/ADAM17 action. Reduction of catalytically active gamma-secretase complex by nicastrin siRNA treatment also resulted in CTF15 accumulation. Furthermore, mature nicastrin, the substrate receptor for gamma-secretase, co-immunoprecipitated with CTF15 in the presence of gamma-secretase inhibitors indicating the formation of CTF15: nicastrin complexes. MUC1-CTF15 accumulation in response to gamma-secretase inhibition was demonstrated in both normal and tumor-derived cells from humans and mice indicating that this processing pathway exists in many cell contexts. We did not detect products of MUC1 cleavage by gamma-secretase in the presence of various proteasomal inhibitors indicating that subsequent degradation is either non-proteasomal or extremely efficient. We suggest that this efficient pathway attenuates potential signaling mediated by cytoplasmic tail fragments.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Implantação do Embrião , Endométrio/metabolismo , Mucina-1/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAM17 , Secretases da Proteína Precursora do Amiloide/química , Animais , Linhagem Celular , Citocinas/metabolismo , Dimerização , Feminino , Humanos , Glicoproteínas de Membrana/química , Camundongos , Complexo de Endopeptidases do Proteassoma/química , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais
4.
Biol Reprod ; 81(6): 1182-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19684334

RESUMO

Embryo implantation involves direct interaction of the blastocyst with the luminal epithelium of the receptive uterus. MUC1, a transmembrane mucin expressed at the apical surface of uterine epithelia, acts as a barrier to microbial infection and enzymatic attack. Loss of MUC1 is believed to be a prerequisite for a functionally receptive uterus across many species. Human and murine MUC1 regulation by steroid hormones displays important differences. Estrogen (E2) stimulates MUC1 expression in mice, and progesterone (P4) antagonizes E2 action in this regard. MUC1 expression is severely reduced during the receptive uterine state in mice. In contrast, human MUC1 expression is maximal at the receptive or midluteal phase, when P4 levels are high. No information is available regarding regulation of human MUC1 in vivo at the site of embryo attachment. Our aim was to better understand regulation of human MUC1 during early pregnancy in vivo. For this purpose, we used a transgenic mouse carrying full-length human MUC1 gene (Tg(MUC1)79.24Gend) as well as endogenous MUC1 as a model system. Human MUC1 was detected by real-time RT-PCR, Western blotting, and immunohistochemistry during early pregnancy. Our data indicate that human MUC1 persists at reduced (20% relative to Day 1 postcoitum) levels in receptive-phase uteri, including the site of embryo attachment. In contrast, mouse MUC1 was much more severely (>98% relative to Day 1 postcoitum) reduced in the same context. These observations are consistent with distinct regulation between the human and mouse genes. Because these genes are expressed in the same transcriptional context (i.e., mouse uterine epithelia), structural differences between human and murine genes must account for these differences in MUC1 regulation.


Assuntos
Implantação do Embrião/genética , Endométrio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Mucina-1/genética , Prenhez , Animais , Western Blotting , Células Epiteliais/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Mucina-1/metabolismo , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie
5.
Tissue Eng Part C Methods ; 25(3): 160-167, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30747042

RESUMO

IMPACT STATEMENT: Maxillofacial defects often present the clinical challenge of a compromised wound bed. Preclinical evaluation of tissue engineering techniques developed to facilitate healing and reconstruction typically involves animal models with ideal wound beds. The healthy wound bed scenario does not fully mimic the complex clinical environment in patients, which can lead to technology failure when translating from preclinical in vivo research to clinical use. The reported preclinical animal model of compromised wound healing enables investigation of tissue engineering technologies in a more clinically relevant scenario, potentially fostering translation of promising results in preclinical research to patients.


Assuntos
Modelos Animais de Doenças , Traumatismos Maxilofaciais/terapia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Cicatrização , Animais , Masculino , Coelhos , Raios Ultravioleta
6.
J Immunother Cancer ; 7(1): 216, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31409394

RESUMO

BACKGROUND: Immune checkpoint inhibitors (ICIs) for solid tumors, including those targeting programmed cell death 1 (PD-1) and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), have shown impressive clinical efficacy, however, most patients do not achieve durable responses. One major therapeutic obstacle is the immunosuppressive tumor immune microenvironment (TIME). Thus, we hypothesized that a strategy combining tumor-directed radiation with TIME immunomodulation could improve ICI response rates in established solid tumors. METHODS: Using a syngeneic mouse model of human papillomavirus (HPV)-associated head and neck cancer, mEER, we developed a maximally effective regimen combining PD-1 and CTLA-4 inhibition, tumor-directed radiation, and two existing immunomodulatory drugs: cyclophosphamide (CTX) and a small-molecule inducible nitric oxide synthase (iNOS) inhibitor, L-n6-(1-iminoethyl)-lysine (L-NIL). We compared the effects of the various combinations of this regimen on tumor growth, overall survival, establishment of immunologic memory, and immunologic changes with flow cytometry and quantitative multiplex immunofluorescence. RESULTS: We found PD-1 and CTLA-4 blockade, and radiotherapy alone or in combination, incapable of clearing established tumors or reversing the unfavorable balance of effector to suppressor cells in the TIME. However, modulation of the TIME with cyclophosphamide (CTX) and L-NIL in combination with dual checkpoint inhibition and radiation led to rejection of over 70% of established mEER tumors and doubled median survival in the B16 melanoma model. Anti-tumor activity was CD8+ T cell-dependent and led to development of immunologic memory against tumor-associated HPV antigens. Immune profiling revealed that CTX/L-NIL induced remodeling of myeloid cell populations in the TIME and tumor-draining lymph node and drove subsequent activation and intratumoral infiltration of CD8+ effector T cells. CONCLUSIONS: Overall, this study demonstrates that modulation of the immunosuppressive TIME is required to unlock the benefits of ICIs and radiotherapy to induce immunologic rejection of treatment-refractory established solid tumors.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Animais , Anticorpos Monoclonais/farmacologia , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Microambiente Tumoral
7.
Oncoimmunology ; 8(4): e1568809, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30906661

RESUMO

Immunotherapeutic treatments in head and neck cancer clinical trials include cancer vaccines targeting foreign viral antigens or mutational neoantigens derived from cancer-expressed proteins. Anti-tumor immune responses place cancer cells under selective pressure to lose or downregulate target antigens; therefore, vaccination against virus- or host- "driver" oncogenes are proposed as a strategy to overcome immune escape. Herein, we demonstrate the impact of immunogenic viral antigens on anti-tumor response and immune editing in MOC2-E6E7, a syngeneic murine oral cancer cell line expressing HPV-16 E6 and E7 oncoproteins. Using orthotopic syngeneic models, we observed in vivo tumor growth kinetics of MOC2-E6E7 is delayed in immunocompetent mice compared to parental MOC2 tumors. In contrast, tumor growth remained similar in Rag1-/- mice lacking adaptive immunity. MOC2-E6E7 tumors demonstrated an "inflamed" or immune-activated tumor microenvironment and greater infiltration of CD8+ T cells compared to MOC2. By real-time PCR, we detected downregulation of E6 and E7 genes in MOC2-E6E7 tumors only in immunocompetent mice, suggesting the loss of ectopic viral antigen expression due to immune editing. We then assessed the efficacy of a biomaterials-based mesoporous silica rod (MSR) cancer vaccine targeting HPV-16 E7 in our model. Vaccination induced robust infiltration of antigen-specific CD8+ T cells, which led to tumor growth delay and modestly prolonged survival in MOC2-E6E7 tumors. Increased efficacy was seen in a separate head and neck cancer tumor model, mEER, which obligately expresses E7 antigen. Collectively, our data highlight the need for both immunogenicity and 'driver' status of target antigens to be considered in cancer vaccine design.

8.
Mol Endocrinol ; 21(11): 2725-37, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17717071

RESUMO

Mucin 1 (MUC1) is a transmembrane glycoprotein that modulates the interaction between the embryo and the uterine epithelial cell surface. MUC1 also is a tumor marker and has been implicated in the protection of cancer cells from immune cell attack as well as in cell signaling in some tumors. We and others have shown that MUC1 expression is activated by progesterone (P), TNF-alpha, and interferon-gamma (IFN-gamma). Here we demonstrate that MUC1 expression is down-regulated by overexpression of members of the protein inhibitor of activated signal transducer and activator of transcription (PIAS) family, PIAS1, PIAS3, PIASxalpha, PIASxbeta, and PIASy, in human uterine epithelial cell lines HES and HEC-1A and in a breast cancer cell line, T47D. Treatments with P, TNF-alpha, and IFN-gamma were unable to overcome the repression by PIASy. PIASy repression of basal, P-, and TNF-alpha-stimulated MUC1 promoter activity was not dependent on the PIASy sumoylation domain. In contrast, PIASy suppression of IFN-gamma-activated MUC1 promoter activity was dependent on the PIASy sumoylation domain. PIASy and P receptor B were localized to the nucleus upon P treatment, and small interfering RNA knockdown of PIASy resulted in an increase in P-mediated stimulation of MUC1 protein expression. Overexpression of PIASy did not affect P receptor B binding to the MUC1 promoter but surprisingly led to a loss of nuclear receptor corepressor (NCoR), which was recruited to the promoter in response to P. Collectively, these data indicate that PIASy may be a useful target for down-regulation of MUC1 expression in various contexts.


Assuntos
Regulação Neoplásica da Expressão Gênica , Mucina-1/biossíntese , Proteínas Inibidoras de STAT Ativados/metabolismo , Biomarcadores Tumorais , Neoplasias da Mama/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Endométrio/metabolismo , Feminino , Humanos , Interferon gama/metabolismo , Ligantes , Mucina-1/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose , Progesterona/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Útero/metabolismo
9.
Biomaterials ; 163: 67-75, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29454236

RESUMO

Recent advancements in the field of immunotherapy have yielded encouraging results for the treatment of advanced cancers. Cyclic dinucleotides (CDNs) are a powerful new class of immunotherapy drugs known as STING (Stimulator of Interferon Genes) agonists, currently in clinical trials. However, previous studies of CDNs in murine cancer models have required multiple injections, and improve survival only in relatively nonaggressive tumor models. Therefore, we sought to improve the efficacy of CDN immunotherapy by developing a novel biomaterial we call "STINGel." STINGel is an injectable peptide hydrogel that localizes and provides controlled release of CDN delivery, showing an 8-fold slower release rate compared to a standard collagen hydrogel. The carrier hydrogel is a positively charged, MultiDomain Peptide (MDP) which self-assembles to form a nanofibrous matrix and is easily delivered by syringe. The highly localized delivery of CDN from this nanostructured biomaterial affects the local histological response in a subcutaneous model, and dramatically improves overall survival in a challenging murine model of head and neck cancer compared to CDN alone or CDN delivered from a collagen hydrogel. This study demonstrates the feasibility of biomaterial-based immunotherapy platforms like STINGel as strategies for increasing the efficacy of CDN immunotherapies.


Assuntos
Antineoplásicos/química , Neoplasias/terapia , Nucleotídeos Cíclicos/química , Peptídeos/química , Animais , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Sobrevivência Celular , Colágeno/química , Preparações de Ação Retardada , Dimerização , Portadores de Fármacos/química , Liberação Controlada de Fármacos , Feminino , Humanos , Hidrogéis , Imunoterapia/métodos , Injeções Intralesionais , Camundongos Endogâmicos C57BL , Nanofibras/química , Neoplasias/imunologia , Nucleotídeos Cíclicos/administração & dosagem , Tamanho da Partícula , Eletricidade Estática
10.
Endocrinology ; 145(9): 4192-203, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15142990

RESUMO

Regulation of MUC1 expression and removal is a salient feature of embryo implantation, bacterial clearance, and tumor progression. In some species, embryo implantation is accompanied by a transcriptional decline in uterine epithelial expression of MUC1. In other species, MUC1 is locally removed at blastocyst attachment sites, suggesting a proteolytic activity. Previously, we demonstrated that MUC1 is proteolytically released from the surface of a human uterine epithelial cell line, HES, and identified TNFalpha converting enzyme/a disintegrin and metalloprotease 17 as a constitutive and phorbol ester-stimulated MUC1 sheddase. The aims of the current study were to test the ability of soluble factors elevated during the periimplantation interval in vivo to stimulate ectodomain shedding of MUC1 from HES uterine epithelial cells and to characterize the nature of this proteolytic activity(ies). We identified TNFalpha as a prospective endogenous stimulus of MUC1 ectodomain release and of MUC1 and TNFalpha converting enzyme/a disintegrin and metalloprotease 17 expression. Moreover, we established that TNFalpha-stimulated MUC1 shedding occurs independently of increased de novo protein synthesis and demonstrated that the TNFalpha-induced increase in MUC1 gene expression is mediated through the kappaB site in the MUC1 promoter. Finally, we determined that the TNFalpha-sensitive MUC1 sheddase is inhibited by the metalloprotease inhibitor, TNFalpha protease inhibitor (TAPI), and the endogenous tissue inhibitor of metalloprotease-3. Collectively, these studies provide the initial in vitro characterization of a putative physiological stimulus of MUC1 ectodomain release and establish the nature of the metalloproteolytic activity(ies) involved.


Assuntos
Antineoplásicos/farmacologia , Células Epiteliais/fisiologia , Mucina-1/genética , Fator de Necrose Tumoral alfa/farmacologia , Útero/citologia , Antígenos de Superfície/metabolismo , Linhagem Celular , Citoplasma/metabolismo , Dipeptídeos/farmacologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Ácidos Hidroxâmicos/farmacologia , Mucina-1/química , Mucina-1/metabolismo , NF-kappa B/metabolismo , Regiões Promotoras Genéticas/fisiologia , Proteína Quinase C/metabolismo , Estrutura Terciária de Proteína , Inibidor Tecidual de Metaloproteinase-1/farmacologia , Inibidor Tecidual de Metaloproteinase-3/farmacologia , Fator de Transcrição RelA
11.
Ophthalmic Genet ; 24(3): 161-5, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12868034

RESUMO

In order to understand the underlying molecular genetic defect causing aniridia in India, eight probands from sporadic cases were screened for all 14 exons of the PAX6 gene using polymerase chain reaction-single strand conformation polymorphism (PCR-SSCP). Direct sequencing of the SSCP variants revealed a nonsense mutation (R317X) in the eleventh exon leading to a premature termination of the PAX6 protein in the proline-serine-threonine (PST)-rich domain in two probands. Another proband exhibited an intronic polymorphism (IVS 9-12 C-T). The mutation resulted in loss of function of the PAX6 protein along with variable phenotypic manifestations in the probands. This is the first report describing a PAX6 gene mutation in aniridia cases from India and highlights the variable expressivity in phenotypes due to haploinsufficiency.


Assuntos
Aniridia/genética , Éxons/genética , Proteínas de Homeodomínio/genética , Mutação , Criança , Análise Mutacional de DNA , Proteínas do Olho , Genótipo , Humanos , Índia , Pressão Intraocular , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados , Fenótipo , Reação em Cadeia da Polimerase , Polimorfismo Conformacional de Fita Simples , Proteínas Repressoras
12.
Expert Rev Endocrinol Metab ; 6(6): 835-848, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22201009

RESUMO

Membrane-tethered mucin glycoproteins are abundantly expressed at the apical surfaces of simple epithelia, where they play important roles in lubricating and protecting tissues from pathogens and enzymatic attack. Notable examples of these mucins are MUC1, MUC4 and MUC16 (also known as cancer antigen 125). In adenocarcinomas, apical mucin restriction is lost and overall expression is often highly increased. High-level mucin expression protects tumors from killing by the host immune system, as well as by chemotherapeutic agents, and affords protection from apoptosis. Mucin expression can increase as the result of gene duplication and/or in response to hormones, cytokines and growth factors prevalent in the tumor milieu. Rises in the normally low levels of mucin fragments in serum have been used as markers of disease, such as tumor burden, for many years. Currently, several approaches are being examined that target mucins for immunization or nanomedicine using mucin-specific antibodies.

13.
Mol Endocrinol ; 24(12): 2253-66, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20962044

RESUMO

Mucin 1 (MUC1), a transmembrane mucin expressed at the apical surface of uterine epithelia, is a barrier to microbial infection and enzymatic attack. MUC1 loss at implantation sites appears to be required to permit embryo attachment and implantation in most species. MUC1 expression is regulated by progesterone (P) and proinflammatory cytokines, including TNFα and interferon γ (IFNγ). TNFα and IFNγ are highly expressed in uterine tissues under conditions where MUC1 expression is also high and activate MUC1 expression via their downstream transcription factors, nuclear factor (NF) κB and signal transducers and activators of transcription. P receptor (PR) regulates MUC1 gene expression in a PR isoform-specific fashion. Here we demonstrate that interactions among PR isoforms and cytokine-activated transcription factors cooperatively regulate MUC1 expression in a human uterine epithelial cell line, HES. Low doses of IFNγ and TNFα synergistically stimulate MUC1 promoter activity, enhance PRB stimulation of MUC1 promoter activity and cooperate with PRA to stimulate MUC1 promoter activity. Cooperative stimulation of MUC1 promoter activity requires the DNA-binding domain of the PR isoforms. MUC1 mRNA and protein expression is increased by cytokine and P treatment in HES cells stably expressing PRB. Using chromatin immunoprecipitation assays, we demonstrate efficient recruitment of NFκB, p300, SRC3 (steroid receptor coactivator 3), and PR to the MUC1 promoter. Collectively, our studies indicate a dynamic interplay among cytokine-activated transcription factors, PR isoforms and transcriptional coregulators in modulating MUC1 expression. This interplay may have important consequences in both normal and pathological contexts, e.g. implantation failure and recurrent miscarriages.


Assuntos
Interferon gama/farmacologia , Mucina-1/genética , Receptores de Progesterona/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interferon gama/metabolismo , Mucina-1/biossíntese , Mucina-1/metabolismo , Progesterona/farmacologia , Regiões Promotoras Genéticas , Isoformas de Proteínas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Progesterona/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Útero/efeitos dos fármacos , Útero/metabolismo , Útero/fisiologia
14.
Mol Endocrinol ; 24(7): 1368-79, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20484415

RESUMO

Mucin 1 (MUC1) is a type I transmembrane glycoprotein abundantly expressed on nearly all epithelial tissues and overexpressed by many cancer cells. Previous studies from our lab showed that progesterone receptor (PR)B is a strong stimulator of MUC1 gene expression. It is reported that liganded peroxisome proliferator-activated receptor gamma (PPARgamma) stimulates Muc1 expression in murine trophoblast. Here, we demonstrate that although the PPARgamma ligand, rosiglitazone, stimulates the murine Muc1 promoter in HEC1A, a human uterine epithelial cell line, rosiglitazone alone, has no significant effect on basal human MUC1 promoter activity. In fact, rosiglitazone treatment antagonizes progesterone-stimulated human MUC1 promoter activity and protein expression in two human uterine epithelial cell lines and T47D human breast cancer cells. This response is antagonized by the PPARgamma antagonist, GW9662, as well as a dominant-negative form of PPARgamma, demonstrating the response is mediated by PPARgamma. Additional studies indicate that PPARgamma activation does not change PR binding to the MUC1 promoter but generally antagonizes progesterone activity by stimulating PRB degradation and inhibiting progesterone-induced PRB phosphorylation. Collectively, these studies indicate that PPARgamma activation inhibits PRB activity through both acute (phosphorylation) and long-term (PRB degradation) pathways.


Assuntos
Mucina-1/metabolismo , PPAR gama/metabolismo , Progesterona/farmacologia , Anilidas/farmacologia , Western Blotting , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Humanos , Mucina-1/genética , PPAR alfa/agonistas , PPAR alfa/metabolismo , PPAR gama/agonistas , PPAR gama/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Receptores de Progesterona/metabolismo , Receptores X de Retinoides/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rosiglitazona , Tiazolidinedionas/farmacologia
15.
Expert Rev Endocrinol Metab ; 3(4): 463-471, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30290438

RESUMO

MUC1 is a multifunctional cell surface glycoprotein that modulates cell adhesion, protects mucosa from infection and enzymatic attack, lubricates cell surfaces, participates in multiple signal-transduction pathways and is overexpressed by many tumors. MUC1 levels change dynamically in various cellular contexts. The primary mechanism for controlling MUC1 expression appears to be transcriptional through a complex combination of often overlapping regulatory motifs that control both tissue specificity and overall rate of transcription. This review will summarize the current knowledge of the factors known to control MUC1 transcriptional regulation, including cytokines, steroid hormones and the growth factors they stimulate, as well as suggest how this information may be exploited in the future to control MUC1 expression in specific biological contexts.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA