Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brain Res ; 1776: 147747, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34864044

RESUMO

Glucose uptake in the brain is critically important to brain health. Using two widely used cell line model systems, we have found that siramesine, a lysosomotropic agent and ligand for the sigma-2 receptor, inhibits glucose uptake and decreases pools of the GLUT1 glucose transporter at the plasma membrane. Siramesine induces autophagy but also disrupts degradation of autophagy substrates, providing a potential mechanism for its action on glucose uptake. In other cell systems, many of the effects of siramesine can be suppressed by α -tocopherol, a type of vitamin E and potent antioxidant, and α-tocopherol also suppressed the effect of siramesine on glucose uptake, suggesting a role for reactive oxygen species and membrane maintenance. We have also identified a novel mechanism for siramesine in which it inhibited plasma membrane levels of GAPDH, a key protein in glycolysis which localizes to the plasma membrane in some cell types. Indeed, GAPDH inhibitors decreased glucose uptake, like siramesine, likely through an overlapping pathway with siramesine. GAPDH inhibitors induced autophagy but inhibited degradation of autophagy targets. Thus, we have identified novel mechanisms required for glucose uptake which may have important implications in disease.


Assuntos
Autofagia/fisiologia , Membrana Celular/metabolismo , Glucose/metabolismo , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Autofagia/efeitos dos fármacos , Transporte Biológico/efeitos dos fármacos , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Humanos , Indóis/farmacologia , Lisossomos/metabolismo , Compostos de Espiro/farmacologia
2.
Front Neurosci ; 14: 668, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32733189

RESUMO

Insulin signaling is an integral component of healthy brain function, with evidence of positive insulin-mediated alterations in synaptic integrity, cerebral blood flow, inflammation, and memory. However, the specific pathways targeted by this peptide remain unclear. Previously, our lab used a molecular approach to characterize the impact of insulin signaling on voltage-gated calcium channels and has also shown that acute insulin administration reduces calcium-induced calcium release in hippocampal neurons. Here, we explore the relationship between insulin receptor signaling and glucose metabolism using similar methods. Mixed, primary hippocampal cultures were infected with either a control lentivirus or one containing a constitutively active human insulin receptor (IRß). 2-NBDG imaging was used to obtain indirect measures of glucose uptake and utilization. Other outcome measures include Western immunoblots of GLUT3 and GLUT4 on total membrane and cytosolic subcellular fractions. Glucose imaging data indicate that neurons expressing IRß show significant elevations in uptake and rates of utilization compared to controls. As expected, astrocytes did not respond to the IRß treatment. Quantification of Western immunoblots show that IRß is associated with significant elevations in GLUT3 expression, particularly in the total membrane subcellular fraction, but did not alter GLUT4 expression in either fraction. Our work suggests that insulin plays a significant role in mediating neuronal glucose metabolism, potentially through an upregulation in the expression of GLUT3. This provides further evidence for a potential therapeutic mechanism underlying the beneficial impact of intranasal insulin in the clinic.

3.
Mol Pharmacol ; 94(1): 665-673, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29674524

RESUMO

The insulin receptor (IR) is a ligand-activated receptor tyrosine kinase that has a key role in metabolism, cellular survival, and proliferation. Progesterone receptor membrane component 1 (PGRMC1) promotes cellular signaling via receptor trafficking and is essential for some elements of tumor growth and metastasis. In the present study, we demonstrate that PGRMC1 coprecipitates with IR. Furthermore, we show that PGRMC1 increases plasma membrane IR levels in multiple cell lines and decreases insulin binding at the cell surface. The findings have therapeutic applications because a small-molecule PGRMC1 ligand, AG205, also decreases plasma membrane IR levels. However, PGRMC1 knockdown via short hairpin RNA expression and AG205 treatment potentiated insulin-mediated phosphorylation of the IR signaling mediator AKT. Finally, PGRMC1 also increased plasma membrane levels of two key glucose transporters, GLUT-4 and GLUT-1. Our data support a role for PGRMC1 maintaining plasma membrane pools of the receptor, modulating IR signaling and function.


Assuntos
Antígenos CD/metabolismo , Membrana Celular/metabolismo , Proteínas de Membrana/metabolismo , Receptor de Insulina/metabolismo , Receptores de Progesterona/metabolismo , Células A549 , Linhagem Celular Tumoral , Transportador de Glucose Tipo 1/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Humanos , Progesterona/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/fisiologia
4.
Adv Lung Cancer (Irvine) ; 4(3): 37-51, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27867772

RESUMO

Cancer is one of the leading causes of death in America, and there is an urgent need for new therapeutic approaches. The progesterone receptor membrane component 1 (PGRMC1) is a cytoch-rome b5 related protein that binds heme and is associated with signaling, apoptotic suppression and autophagy. PGRMC1 is essential for tumor formation, invasion and metastasis, and is upregulated in breast, colon, lung and thyroid tumors. In the present study, we have analyzed PGRMC1 levels in over 600 tumor sections, including a larger cohort of lung tumors than in previous studies, and report the first clinical analysis of PGRMC1 levels in human oral cavity and ovarian tumors compared to corresponding nonmalignant tissues. PGRMC1 was highly expressed in lung and ovarian cancers and correlated with patient survival. PGRMC1 has been previously associated with drug resistance, a characteristic of cancer stem cells. The stem cell theory proposes that a subset of cancerous stem cells contribute to drug resistance and tumor maintenance, and PGRMC1 was detected in lung-tumor derived stem cells. Drug treatment with a PGRMC1 inhibitor, AG-205, triggered stem cell death whereas treatment with erlotinib and the ERK inhibitor, PD98059, did not, suggesting a specific role for PGRMC1 in cancer stem cell viability. Together, our data demonstrate PGRMC1 as a potential tumor biomarker across a variety of tumors, as well as a therapeutic target for cancer stem cells.

5.
Oncoscience ; 1(8): 504-12, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25594057

RESUMO

EGFR (epidermal growth factor receptor) is activated through changes in expression or mutations in a number of tumors and is a driving force in cancer progression. EGFR is targeted by numerous inhibitors, including chimeric antibodies targeting the extracellular domain and small molecule kinase domain inhibitors. The kinase domain inhibitors are particularly active against mutant forms of the receptor, and subsequent mutations drive resistance to the inhibitors. Here, we review recent developments on the trafficking of wild-type and mutant EGFR, focusing on the roles of MIG6, SPRY2, ITSN, SHP2, S2R(PGRMC1) and RAK. Some classes of EGFR regulators affect wild-type and mutant EGFR equally, while others are specific for either the wild-type or mutant form of the receptor. Below we summarize multiple signaling-associated pathways that are important in trafficking wild-type and mutant EGFR with the goal being stimulation of new approaches for targeting the distinct forms of the receptor.

6.
Autophagy ; 9(10): 1566-78, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24113030

RESUMO

Autophagy resembles a recycling process in which proteins, organelles, or regions of the cytoplasm are enveloped and degraded. We have found that two of the central autophagy proteins, MAP1LC3 (microtubule-associated protein 1 light chain 3, also described as LC3) and UVRAG (UV radiation resistance associated/UV radiation associated gene), complex with PGRMC1/S2R (progesterone receptor membrane component 1, also known as sigma-2 receptor). PGRMC1 is a cytochrome that is induced in cancer and is essential for tumor formation, invasion, and metastasis. Autophagy contributes to the turnover of long-lived and/or ubiquitinated proteins and the clearance of damaged organelles, and we have shown that PGRMC1 promotes both processes. Inhibition of PGRMC1 by RNAi or small molecule inhibitors causes autophagy substrates to increase and aberrant mitochondria to accumulate. We propose that this disruption of autophagy upon PGRMC1 inhibition increases AMPK activation, elevating the levels of TSC1 (tuberous sclerosis complex) and TSC2 and inactivating MTOR and RPS6KB/p70S6K, causing cleaved MAP1LC3B levels to increase. Thus, PGRMC1 binds to key components of the autophagy machinery and is required for the degradative activity of autophagy.


Assuntos
Autofagia/fisiologia , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Receptores de Progesterona/metabolismo , Receptores sigma/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Humanos , Proteólise , Receptores de Progesterona/efeitos dos fármacos , Receptores sigma/efeitos dos fármacos , Proteína Sequestossoma-1 , Esclerose Tuberosa/metabolismo
7.
Biochem Res Int ; 2012: 268504, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23213529
8.
J Biol Chem ; 287(18): 14494-501, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22418433

RESUMO

Tumor invasion is a critical step in the spread of cancer. S2R (sigma-2 receptor)/Pgrmc1 (progesterone receptor membrane component 1) is a cytochrome b(5)-related drug-binding orphan receptor essential for tumor formation and invasion. Secretory proteins drive these processes, so we screened for S2R(Pgrmc1)-dependent secreted proteins using antibody arrays. S2R(Pgrmc1) markedly regulated the expression of NGAL/LCN2 (neutrophil gelatinase-associated lipocalin/lipocalin 2), a secreted glycoprotein that binds to MMP-9 (matrix metalloproteinase 9) and protects it from degradation. S2R(Pgrmc1) knock-down blocked NGAL/LCN2 expression at the protein and RNA levels and decreased MMP9 activity. NGAL expression was required for MMP-9 activity and tumor formation. S2R(Pgrmc1) associates with EGFR and increases EGFR levels at the plasma membrane, and the EGFR inhibitors erlotinib and AG1478, as well as Akt and ERK inhibitors, suppressed the NGAL/LCN2 RNA and protein levels. NGAL is transcriptionally regulated by NFκB, and S2R(Pgrmc1) knock-down decreased the NFκB subunit p65/RelA acetylation, phosphorylation, and activation. In S2R(Pgrmc1) knock-down cells, p65 acetylation was reversed by inhibitors of histone deacetylase 1, and the inhibitors partially restored NGAL levels. Our results are consistent with a model in which S2R(Pgrmc1) increases NGAL/LCN2 levels by activating NFκB via EGFR.


Assuntos
Proteínas de Fase Aguda/biossíntese , Membrana Celular/metabolismo , Regulação Neoplásica da Expressão Gênica , Lipocalinas/biossíntese , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas/biossíntese , Receptores de Progesterona/metabolismo , Acetilação/efeitos dos fármacos , Proteínas de Fase Aguda/genética , Animais , Linhagem Celular Tumoral , Membrana Celular/genética , Membrana Celular/patologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Cloridrato de Erlotinib , Feminino , Histona Desacetilase 1/antagonistas & inibidores , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Humanos , Lipocalina-2 , Lipocalinas/genética , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Nus , Modelos Biológicos , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Neoplasias/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Ligação Proteica , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas/genética , Quinazolinas/farmacologia , Receptores de Progesterona/genética , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética , Transplante Heterólogo , Tirfostinas/farmacologia
9.
Expert Opin Drug Metab Toxicol ; 8(3): 361-70, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22292588

RESUMO

INTRODUCTION: S2R (sigma-2 receptor)/Pgrmc1 (progesterone receptor membrane component 1) is a cytochrome-related protein that binds directly to heme and various pharmacological compounds. S2R(Pgrmc1) also associates with cytochrome P450 proteins, the EGFR receptor tyrosine kinase and the RNA-binding protein PAIR-BP1. S2R(Pgrmc1) is induced in multiple types of cancer, where it regulates tumor growth and is implicated in progesterone signaling. S2R(Pgrmc1) also increases cholesterol synthesis in non-cancerous cells and may have a role in modulating drug metabolizing P450 proteins. AREAS COVERED: This review covers the independent identification of S2R and Pgrmc1 and their induction in cancers, as well as the role of S2R(Pgrmc1) in increasing cholesterol metabolism and P450 activity. This article was formed through a PubMed literature search using, but not limited to, the terms sigma-2 receptor, Pgrmc1, Dap1, cholesterol and aromatase. EXPERT OPINION: Multiple laboratories have shown that S2R(Pgrmc1) associates with various P450 proteins and increases cholesterol synthesis via Cyp51. However, the lipogenic role of S2R(Pgrmc1) is tissue-specific. Furthermore, the role of S2R(Pgrmc1) in regulating P450 proteins other than Cyp51 appears to be highly selective, with modest inhibitory activity for Cyp3A4 in vitro and a complex regulatory pattern for Cyp21. Cyp19/aromatase is a therapeutic target in breast cancer, and S2R(Pgrmc1) activated Cyp19 significantly in vitro but modestly in biochemical assays. In summary, S2R(Pgrmc1) is a promising therapeutic target for cancer and possibly cholesterol synthesis but research to date has not identified a major role in P450-mediated drug metabolism.


Assuntos
Colesterol/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Citocromos b5/metabolismo , Hormônios/metabolismo , Proteínas de Membrana/metabolismo , Receptores de Progesterona/metabolismo , Receptores sigma/metabolismo , Transdução de Sinais , Animais , Biotransformação , Clonagem Molecular , Dislipidemias/tratamento farmacológico , Dislipidemias/metabolismo , Humanos , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/genética , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Progesterona/metabolismo , Receptores de Progesterona/efeitos dos fármacos , Receptores de Progesterona/genética , Receptores sigma/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
10.
Int J Cancer ; 131(2): E1-9, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21918976

RESUMO

Cancer is one of the leading causes of death, and there is an urgent need for new biomarkers and therapeutic targets. The progesterone receptor membrane component 1 (Pgrmc1) protein is upregulated in multiple types of cancer, and Pgrmc1 is required for tumor cell proliferation, motility and tumor formation in vivo. Furthermore, a small molecule inhibitor of Pgrmc1 suppressed the growth of lung, breast and cervical cancer cell lines. Recently, Pgrmc1 was identified as the sigma-2 receptor, a putative type of opioid receptor, and sigma-2 receptors are induced in cancers. However, Pgrmc1 shares no homology with known opioid or hormone receptors but is related to cytochrome b(5), and Pgrmc1 binds to heme and has reducing activity. In this study, we have analyzed Pgrmc1 levels in clinical tumor samples from squamous cell lung cancers (SCLC) and lung adenocarcinomas compared to corresponding nonmalignant tissue. Pgrmc1 levels increased significantly (p ≤ 0.05) in 12/15 SCLC samples and was elevated in poorly differentiated tumors. Pgrmc1 was highly expressed in SCLC cell lines, and SCLC cell survival was inhibited by siRNA knockdown of Pgrmc1 or the Pgrmc1 inhibitor AG-205. In adenocarcinomas, 6/15 tumors significantly had elevated Pgrmc1 levels, which correlated with patient survival. Pgrmc1 localizes to secretory vesicles in cancer cells, and Pgrmc1 was secreted by lung cancer cells. Furthermore, Pgrmc1 was significantly elevated in the plasma of lung cancer patients compared to noncancer patients. Together, the results demonstrate that Pgrmc1 is a potential tumor and serum biomarker, as well as a therapeutic target, for lung cancer.


Assuntos
Adenocarcinoma/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Membrana/metabolismo , Neoplasias de Células Escamosas/metabolismo , Receptores de Progesterona/metabolismo , Adenocarcinoma/sangue , Adenocarcinoma de Pulmão , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Neoplasias Pulmonares/sangue , Masculino , Proteínas de Membrana/sangue , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Neoplasias de Células Escamosas/sangue , Interferência de RNA , RNA Interferente Pequeno , Receptores de Progesterona/sangue , Receptores de Progesterona/genética , Receptores sigma/sangue , Receptores sigma/genética , Receptores sigma/metabolismo
11.
J Biol Chem ; 285(32): 24775-82, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20538600

RESUMO

Tumorigenesis requires the concerted action of multiple pathways, including pathways that stimulate proliferation and metabolism. Epidermal growth factor receptor (EGFR) is a transmembrane receptor-tyrosine kinase that is associated with cancer progression, and the EGFR inhibitors erlotinib/tarceva and tyrphostin/AG-1478 are potent anti-cancer therapeutics. Pgrmc1 (progesterone receptor membrane component 1) is a cytochrome b(5)-related protein that is up-regulated in tumors and promotes cancer growth. Pgrmc1 and its homologues have been implicated in cell signaling, and we show here that Pgrmc1 increases susceptibility to AG-1478 and erlotinib, increases plasma membrane EGFR levels, and co-precipitates with EGFR. Pgrmc1 co-localizes with EGFR in cytoplasmic vesicles and co-fractionates with EGFR in high density microsomes. The findings have therapeutic potential because a Pgrmc1 small molecule ligand, which inhibits growth in a variety of cancer cell types, de-stabilized EGFR in multiple tumor cell lines. EGFR is one of the most potent receptor-tyrosine kinases driving tumorigenesis, and our data support a role for Pgrmc1 in promoting several cancer phenotypes at least in part by binding EGFR and stabilizing plasma membrane pools of the receptor.


Assuntos
Receptores ErbB/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Proteínas de Membrana/metabolismo , Quinazolinas/farmacologia , Receptores de Progesterona/metabolismo , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Citocromos b5/química , Citoplasma/metabolismo , Cloridrato de Erlotinib , Feminino , Humanos , Concentração Inibidora 50 , Neoplasias Pulmonares/metabolismo , Modelos Biológicos , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA
12.
J Pharmacol Exp Ther ; 333(2): 564-73, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20164297

RESUMO

Tumorigenesis requires the concerted action of multiple pathways, including pathways that stimulate proliferation and increase metabolism. Progesterone receptor membrane component 1 (Pgrmc1) is related to cytochrome b5, binds to heme, and is associated with DNA damage resistance and apoptotic suppression. Pgrmc1 is induced by carcinogens, including dioxin, and is up-regulated in multiple types of cancer. In the present study, we found that Pgrmc1 increased in vivo tumor growth, anchorage-independent growth, and migration. Pgrmc1 also promoted proliferation in the absence of serum in A549 non-small cell lung cancer cells but enhanced proliferation regardless of serum concentration in MDA-MB-468 breast cancer cells. Pgrmc1 promotes cholesterol synthesis and binds to Insig (insulin-induced gene), Scap (sterol regulatory element binding protein cleavage activating protein), and P450 proteins, but Pgrmc1 did not affect cholesterol synthesis in lung cancer cells. Pgrmc1 is also associated with progesterone signaling and plasminogen activator inhibitor (PAI1) RNA binding protein, but neither progesterone activity nor PAI1 transcript levels were altered in Pgrmc1-knockdown lung cancer cells. Pgrmc1 homologues bind to aryl ligands identified in an in silico screen, and we have found that a Pgrmc1 ligand induced cell death in a Pgrmc1-specific manner in multiple breast and lung tumor cell lines. Our data support a role for Pgrmc1 in promoting cancer-associated phenotypes and provide a therapeutic approach for targeting Pgrmc1 with a small molecule in lung and breast cancer.


Assuntos
Proteínas de Membrana/farmacologia , Neoplasias/induzido quimicamente , Animais , Neoplasias da Mama/fisiopatologia , Carcinoma Pulmonar de Células não Pequenas/fisiopatologia , Linhagem Celular Tumoral , Ensaios de Migração Celular , Proliferação de Células/efeitos dos fármacos , Colesterol/biossíntese , Feminino , Heme/análogos & derivados , Heme/fisiologia , Humanos , Neoplasias Pulmonares/fisiopatologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias/fisiopatologia , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/fisiopatologia , Estrutura Terciária de Proteína , Receptores de Progesterona/antagonistas & inibidores , Receptores de Progesterona/fisiologia
13.
Cancer Cell ; 15(4): 304-14, 2009 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-19345329

RESUMO

Expression of the PTEN tumor suppressor is frequently lost in breast cancer in the absence of mutation or promoter methylation through as yet undetermined mechanisms. In this study, we demonstrate that the Rak tyrosine kinase physically interacts with PTEN and phosphorylates PTEN on Tyr336. Knockdown of Rak enhanced the binding of PTEN to its E3 ligase NEDD4-1 and promoted PTEN polyubiquitination, leading to PTEN protein degradation. Notably, ectopic expression of Rak effectively suppressed breast cancer cell proliferation, invasion, and colony formation in vitro and tumor growth in vivo. Furthermore, Rak knockdown was sufficient to transform normal mammary epithelial cells. Therefore, Rak acts as a bona fide tumor suppressor gene through the mechanism of regulating PTEN protein stability and function.


Assuntos
Genes Supressores de Tumor/fisiologia , Proteínas de Neoplasias/fisiologia , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Tirosina Quinases/fisiologia , Quinases da Família src/fisiologia , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Mama/citologia , Mama/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células , Transformação Celular Neoplásica , Células Cultivadas , Complexos Endossomais de Distribuição Requeridos para Transporte , Feminino , Humanos , Immunoblotting , Imunoprecipitação , Camundongos , Camundongos Nus , Mutação , Ubiquitina-Proteína Ligases Nedd4 , Invasividade Neoplásica , Osteossarcoma/genética , Osteossarcoma/metabolismo , Osteossarcoma/patologia , PTEN Fosfo-Hidrolase/genética , Fosforilação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
14.
Pharmacol Ther ; 121(1): 14-9, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18992768

RESUMO

Hormone signaling is important in a number of disease states, and hormone receptors are effective therapeutic targets. PGRMC1 (progesterone receptor membrane component 1) is a member of a multi-protein complex that binds to progesterone and other steroids, as well as pharmaceutical compounds. In spite of its name, PGRMC1 shares homology with cytochrome b5-related proteins rather than hormone receptors, and heme binding is the sole biochemical activity of PGRMC1. PGRMC1 and its homologues regulate cholesterol synthesis by activating the P450 protein Cyp51/lanosterol demethylase, and the cholesterol synthetic pathway is an important target in cardiovascular disease and in treating infections. PGRMC1 binding partners include multiple P450 proteins, PAIR-BP1, Insig, and an uncharacterized hormone/drug-binding protein. PGRMC1 is induced in a spectrum of cancers, where it promotes cell survival and damage resistance, and PGRMC1 is also expressed in the nervous system and tissues involved in drug metabolism, cholesterol synthesis and hormone synthesis and turnover. One of the appealing features of PGRMC1 and its associated protein complex is its affinity for steroids and drugs. Together with its biological role in promoting tumor survival, PGRMC1 is an attractive target for therapeutic intervention in cancer and related malignancies.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Receptores de Progesterona/metabolismo , Receptores de Progesterona/fisiologia , Esteroides/metabolismo , Animais , Encéfalo/metabolismo , Sobrevivência Celular , Colesterol/biossíntese , Dano ao DNA , Descoberta de Drogas , Heme/metabolismo , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/farmacologia , Receptores de Progesterona/química
15.
Breast Cancer Res ; 10(6): 113, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19090968

RESUMO

Estrogen receptor (ER) status is a critical biomarker in breast cancer, in large part because the ER is the target of tamoxifen and similar drugs. In the previous issue of Breast Cancer Research, Neubauer and colleagues used a proteomic approach to identify proteins that are differentially regulated by ER in breast tumors. The authors showed that ER-negative tumors have elevated levels of PGRMC1 (progesterone receptor membrane component-1), a hormone receptor component and binding partner for P450 proteins. In contrast, PGRMC1 was phosphorylated in ER-positive tumors. The staining patterns of ER and PGRMC1 were mutually exclusive in breast tumor sections, and PGRMC1 staining was sharply increased in hypoxic areas of the tumor. The results suggest that PGRMC1 is a candidate biomarker for ER status and hypoxia in breast cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Proteínas de Membrana/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Neoplasias da Mama/patologia , Feminino , Humanos
16.
J Biol Chem ; 282(50): 36543-51, 2007 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-17954932

RESUMO

Fungal infections arise frequently in immunocompromised patients, and sterol synthesis is a primary pathway targeted by antifungal drugs. In particular, the P450 protein Erg11/Cyp51 catalyzes a critical step in ergosterol synthesis, and the azole class of antifungal drugs inhibits Erg11. Dap1 is a heme-binding protein related to cytochrome b5 that activates Erg11, so that cells lacking Dap1 accumulate the Erg11 substrate and are hypersensitive to Erg11 inhibitors. Heme binding by Dap1 is crucial for its function, and point mutants in its heme-binding domain render Dap1 inactive for sterol biosynthesis and DNA damage resistance. Like Dap1, the human homologue, PGRMC1/Hpr6, also regulates sterol synthesis and DNA damage resistance. In the present study, we demonstrate that the Dap1 heme-1 domain is required for growth under conditions of low iron availability. Loss of Dap1 is suppressed by elevated levels of Erg11 but not by increased heme biosynthesis. Dap1 localizes to punctate cytoplasmic structures that co-fractionate with endosomes, and Dap1 contributes to the integrity of the vacuole. The results suggest that Saccharomyces cerevisiae Dap1 stimulates a P450-catalyzed step in sterol synthesis via a distinct localization from its homologues in Schizosaccharomyces pombe and mammals and that this function regulates iron metabolism.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Hemeproteínas/metabolismo , Homeostase , Ferro/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , Antifúngicos/metabolismo , Antifúngicos/farmacologia , Azóis/metabolismo , Azóis/farmacologia , Sistema Enzimático do Citocromo P-450/genética , Citocromos b5/genética , Citocromos b5/metabolismo , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/genética , Farmacorresistência Fúngica/efeitos dos fármacos , Farmacorresistência Fúngica/genética , Endossomos/enzimologia , Endossomos/genética , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Ergosterol/biossíntese , Heme/biossíntese , Heme/genética , Hemeproteínas/genética , Humanos , Proteínas de Membrana/genética , Mutação Puntual , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Schizosaccharomyces/enzimologia , Schizosaccharomyces/genética
17.
Biochim Biophys Acta ; 1772(9): 1103-11, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17689225

RESUMO

Cancer chemotherapy inhibits tumor growth, in part, by triggering apoptosis, and anti-apoptotic proteins reduce the effectiveness of chemotherapy. Clusterin, a chaperone-like protein that binds to apoptotic and DNA repair proteins, is induced by chemotherapy and promotes tumor cell survival. Histone deacetylase inhibitors (HDIs) such as sodium butyrate and suberoylanilide hydroxamic acid (SAHA) are pharmacological agents that induce differentiation and apoptosis in cancer cells by altering chromatin structure, and we have found that combinations of chemotherapeutic drugs such as doxorubicin and HDIs efficiently induce apoptosis, even though they paradoxically induce high levels of clusterin. The hyper-expressed form of clusterin localizes to mitochondria, inhibits cytochrome c release, and is inhibited by the proteasome. When HDIs are used as single agents, clusterin suppresses cytochrome c release and apoptosis. However, doxorubicin/HDI-induced apoptosis is not inhibited by clusterin, and clusterin-resistant apoptosis corresponds with markers of the extrinsic/receptor-mediated apoptotic pathway. Thus, chemotherapy-HDI combinations are capable of overcoming an innate anti-apoptotic pathway of tumor cells, suggesting that chemotherapy-HDI combinations have potential for treating advanced stage breast cancer.


Assuntos
Neoplasias da Mama/genética , Clusterina/genética , Transdução de Sinais/fisiologia , Proteínas Reguladoras de Apoptose/fisiologia , Neoplasias da Mama/patologia , Clusterina/metabolismo , Citocromos c/metabolismo , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica , Inibidores de Histona Desacetilases , Humanos , Mitocôndrias/metabolismo , Modelos Biológicos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transdução de Sinais/genética , Células Tumorais Cultivadas
18.
J Bioenerg Biomembr ; 38(3-4): 163-7, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17039395

RESUMO

NM23-H1 is a metastasis suppressor protein that exhibits 3'-5' exonuclease activity in vitro. As 3'-5' exonucleases are generally required for maintenance of genome integrity, this activity represents a plausible candidate mediator of the metastasis suppressor properties of the NM23-H1 molecule. Consistent with an antimutator function, ablation of the yeast NM23 homolog, YNK1, results in increased mutation rates following exposure to UV irradiation and exposure to the DNA damaging agents etoposide, cisplatin, and MMS. In human cells, a DNA repair function is further suggested by increased NM23-H1 expression and nuclear translocation following DNA damage. Also, forced expression of NM23-H1 in NM23-deficient and metastatic cell lines results in coordinate downregulation of multiple DNA repair genes, possibly reflecting genomic instability associated with the NM23-deficient state. To assess the relevance of the 3'-5' exonuclease activity of NM23-H1 to its antimutator and metastasis suppressor functions, a panel of mutants harboring defects in the 3'-5' exonuclease and other enzymatic activities of the molecule (NDPK, histidine kinase) have been expressed by stable transfection in the melanoma cell line, 1205Lu. Pilot in vivo metastasis assays indicate 1205Lu cells are highly responsive to the metastasis suppressor effects of NM23-H1, thus providing a valuable model for measuring the extent to which the nuclease function opposes metastasis and metastatic progression.


Assuntos
Reparo do DNA , Exonucleases/genética , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Metástase Neoplásica/genética , Núcleosídeo-Difosfato Quinase/genética , Linhagem Celular Tumoral , Exonucleases/metabolismo , Humanos , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Modelos Moleculares , Mutação/genética , Nucleosídeo NM23 Difosfato Quinases , Metástase Neoplásica/prevenção & controle , Núcleosídeo-Difosfato Quinase/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Leveduras
19.
J Pharmacol Exp Ther ; 316(1): 448-55, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16234411

RESUMO

Cancer cells have varying levels of susceptibility to chemotherapeutic agents, and the proteins that direct drug susceptibility are promising targets for intervention in cancer. Hpr6 (heme-1 domain protein)/PGRMC1 (progesterone receptor membrane component 1) is overexpressed in tumors, and Hpr6 is the human homolog of a budding yeast damage resistance gene called Dap1p. Cells lacking Dap1p are damage-sensitive, and we have found that inhibition of Hpr6 expression by RNA inhibition (RNAi) increases sensitivity of breast cancer cells to chemotherapeutic drugs. Hpr6 is composed largely of a cytochrome b(5)-related heme-1 domain, and we have found that purified Hpr6 binds to heme, similar to its yeast and rodent homologues. We generated an aspartate 120-to-glycine (D120G) mutant of Hpr6 at a highly conserved site in the heme-1 domain and demonstrated that Hpr6-D120G cannot bind to heme. The Hpr6-D120G mutant was named Hpr6(hbd) for heme binding defective. We prepared an adenovirus encoding Hpr6(hbd) and found that adenovirus Hpr6(hbd) increases susceptibility of breast cancer cells to doxorubicin and camptothecin. Our findings support a model in which Hpr6, similar to its yeast homolog, binds to heme and regulates susceptibility to damaging agents.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Membrana/fisiologia , Neoplasias/tratamento farmacológico , Receptores de Progesterona/fisiologia , Antibióticos Antineoplásicos/metabolismo , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/metabolismo , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Feminino , Citometria de Fluxo , Humanos , Proteínas de Membrana/metabolismo , Neoplasias/genética , Neoplasias/patologia , Plasmídeos/genética , Ligação Proteica , Interferência de RNA/efeitos dos fármacos , Receptores de Progesterona/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vírus/genética
20.
Mol Pharmacol ; 68(6): 1747-56, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16150928

RESUMO

Doxorubicin is an anthracycline antibiotic used for cancer chemotherapy. The utility of doxorubicin is limited by its inability to kill all of the cells within a tumor and by resistant cells emerging from the treated population. We have screened for genes that regulate doxorubicin susceptibility in highly tumorigenic breast cancer cells by cDNA microarray and RNA interference (RNAi) analysis, and we have identified genes associated with both proliferation and cell cycle arrest after doxorubicin treatment. We confirmed that MDA-MB-231 cells treated with doxorubicin induce the expression of carbonic anhydrase II (CAII), inhibitor of differentiation/DNA binding 2 (Id2), activating transcription factor 3 (Atf3), and the phosphatidylinositol 3-kinase 55-kDa regulatory subunit p55PIK. These genes were induced at different times and with varying specificities to different chemotherapeutic drugs. In addition to being induced at the transcriptional level, the CAII and clusterin proteins were elevated after doxorubicin treatment. CAII, Id2, p55PIK, and clusterin were not altered by doxorubicin in MCF-7 cells, a weakly tumorigenic cell line used in previous studies of doxorubicin-regulated gene expression. By inhibiting gene expression using RNAi, we found that CAII and clusterin increase cell survival after doxorubicin treatment, whereas Id2 increases susceptibility to doxorubicin. Our results support a model in which highly tumorigenic breast cancer cells induce a transcriptional response to doxorubicin that is distinct from less malignant cells. The induced genes regulate drug susceptibility positively and negatively and may be novel targets for therapeutic intervention.


Assuntos
Neoplasias da Mama/patologia , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Ciclo Celular/genética , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Feminino , Perfilação da Expressão Gênica , Humanos , Transcrição Gênica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...