Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomater Sci ; 8(13): 3678-3684, 2020 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-32469353

RESUMO

Porphyrin-based periodic mesoporous organosilica nanoparticles (PMO) synthesized from a large functional octatriethoxysilylated porphyrin precursor and allowing two-photon excitation photodynamic therapy (TPE-PDT) and NIR imaging were synthesized. These PMO were grafted with polyethylene glycol (PEG) moieties and an analogue of mannose 6-phosphate functionalized at the anomeric position (AMFA). AMFAs are known to efficiently target mannose 6-phosphate receptors (M6PRs) which are over-expressed in various cancers. Here, we demonstrated for the first time that M6PRs were over-expressed in rhabdomyosarcoma (RMS) cells and could be efficiently targeted with PMO-AMFA allowing TPE imaging and TPE-PDT of RMS cells. The comparison with healthy myoblasts demonstrated an absence of biological effects, suggesting a cancer cell specificity in the biomedical action observed.


Assuntos
Antineoplásicos/farmacologia , Materiais Biocompatíveis/farmacologia , Compostos de Organossilício/farmacologia , Receptor IGF Tipo 2/antagonistas & inibidores , Rabdomiossarcoma/tratamento farmacológico , Nanomedicina Teranóstica , Antineoplásicos/síntese química , Antineoplásicos/química , Materiais Biocompatíveis/síntese química , Materiais Biocompatíveis/química , Humanos , Nanopartículas/química , Imagem Óptica , Compostos de Organossilício/síntese química , Compostos de Organossilício/química , Tamanho da Partícula , Fotoquimioterapia , Porosidade , Porfirinas/química , Porfirinas/farmacologia , Proteômica , Receptor IGF Tipo 2/genética , Rabdomiossarcoma/diagnóstico por imagem , Rabdomiossarcoma/genética , Propriedades de Superfície , Células Tumorais Cultivadas
2.
Hypertension ; 75(5): 1242-1250, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32200675

RESUMO

Megalin is an endocytic receptor contributing to protein reabsorption. Impaired expression or trafficking of megalin increases urinary renin and allowed the detection of prorenin, which normally is absent in urine. Here, we investigated (pro)renin uptake by megalin, using both conditionally immortalized proximal tubule epithelial cells and Brown Norway Rat yolk sac cells (BN16). To distinguish binding and internalization, cells were incubated with recombinant human (pro)renin at 4°C and 37°C, respectively. (Pro)renin levels were assessed by immunoradiometric assay. At 4°C, BN16 cells bound 3× more prorenin than renin, suggestive for a higher affinity of prorenin. Similarly, at 37°C, prorenin accumulated at 3- to 4-fold higher levels than renin in BN16 cells. Consequently, depletion of medium prorenin (but not renin) content occurred after 24 hours. No such differences were observed in conditionally immortalized proximal tubule epithelial cells, and M6P (mannose-6-phosphate) greatly reduced conditionally immortalized proximal tubule epithelial cells (pro)renin uptake, suggesting that these cells accumulate (pro)renin largely via M6P receptors. M6P did not affect (pro)renin uptake in BN16 cells. Yet, inhibiting megalin expression with siRNA greatly reduced (pro)renin binding and internalization by BN16 cells. Furthermore, treating BN16 cells with albumin, an endogenous ligand of megalin, also decreased binding and internalization of (pro)renin, while deleting the (pro)renin receptor affected the latter only. Exposing prorenin's prosegment with the renin inhibitor aliskiren dramatically increased prorenin binding, while after prosegment cleavage with trypsin prorenin binding was identical to that of renin. In conclusion, megalin might function as an endocytic receptor for (pro)renin and displays a preference for prorenin. Megalin-mediated endocytosis requires the (pro)renin receptor.


Assuntos
Endocitose/fisiologia , Precursores Enzimáticos/metabolismo , Túbulos Renais Proximais/metabolismo , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Renina/metabolismo , Amidas/farmacologia , Animais , Linhagem Celular Transformada , Células Epiteliais/metabolismo , Fumaratos/farmacologia , Humanos , Túbulos Renais Proximais/citologia , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/antagonistas & inibidores , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Endogâmicos BN , Ratos Sprague-Dawley , Receptor IGF Tipo 2/antagonistas & inibidores , Receptor IGF Tipo 2/genética , Receptor IGF Tipo 2/metabolismo , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes/metabolismo , Renina/química , Renina/efeitos dos fármacos , Soroalbumina Bovina/metabolismo , Soroalbumina Bovina/farmacologia , Especificidade por Substrato , Temperatura , Tripsina/metabolismo , Saco Vitelino/citologia , Receptor de Pró-Renina
3.
EMBO Mol Med ; 12(1): e11019, 2020 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-31793167

RESUMO

Duchenne muscular dystrophy (DMD) is a debilitating fatal X-linked muscle disorder. Recent findings indicate that IGFs play a central role in skeletal muscle regeneration and development. Among IGFs, insulinlike growth factor 2 (IGF2) is a key regulator of cell growth, survival, migration and differentiation. The type 2 IGF receptor (IGF2R) modulates circulating and tissue levels of IGF2 by targeting it to lysosomes for degradation. We found that IGF2R and the store-operated Ca2+ channel CD20 share a common hydrophobic binding motif that stabilizes their association. Silencing CD20 decreased myoblast differentiation, whereas blockade of IGF2R increased proliferation and differentiation in myoblasts via the calmodulin/calcineurin/NFAT pathway. Remarkably, anti-IGF2R induced CD20 phosphorylation, leading to the activation of sarcoplasmic/endoplasmic reticulum Ca2+ -ATPase (SERCA) and removal of intracellular Ca2+ . Interestingly, we found that IGF2R expression was increased in dystrophic skeletal muscle of human DMD patients and mdx mice. Blockade of IGF2R by neutralizing antibodies stimulated muscle regeneration, induced force recovery and normalized capillary architecture in dystrophic mdx mice representing an encouraging starting point for the development of new biological therapies for DMD.


Assuntos
Músculo Esquelético/crescimento & desenvolvimento , Distrofia Muscular de Duchenne/tratamento farmacológico , Receptor IGF Tipo 2/antagonistas & inibidores , Regeneração , Animais , Sítios de Ligação , Criança , Humanos , Camundongos , Camundongos Endogâmicos mdx , Mioblastos , Adulto Jovem
4.
Sci Rep ; 9(1): 11476, 2019 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-31391495

RESUMO

Osteosarcoma (OS) represents 3.4% of all childhood cancers with overall survival of 70% not improving in 30 years. The consistent surface overexpression of insulin-like growth factor-2 receptor (IGF2R) has been reported in commercial and patient-derived xenograft (PDX) OS cell lines. We aimed to assess efficacy and safety of treating PDX and commercial OS tumors in mice with radiolabeled antibody to IGF2R and to investigate IGF2R expression on canine OS tumors. IGF2R expression on human commercial lines 143B and SaOS2 and PDX lines OS-17, OS-33 and OS-31 was evaluated by FACS. The biodistribution and microSPECT/CT imaging with 111Indium-2G11 mAb was performed in 143B and OS-17 tumor-bearing SCID mice and followed by radioimmunotherapy (RIT) with 177Lutetium-2G11 and safety evaluation. IGF2R expression in randomly selected canine OS tumors was measured by immunohistochemistry. All OS cell lines expressed IGF2R. Biodistribution and microSPECT/CT revealed selective uptake of 2G11 mAb in 143B and OS-17 xenografts. RIT significantly slowed down the growth of OS-17 and 143B tumors without local and systemic toxicity. Canine OS tumors expressed IGF2R. This study demonstrates the feasibility of targeting IGF2R on OS in PDX and spontaneous canine tumors and sets the stage for further development of RIT of OS using comparative oncology.


Assuntos
Doenças do Cão/terapia , Imunoconjugados/administração & dosagem , Osteossarcoma/terapia , Radioimunoterapia/métodos , Receptor IGF Tipo 2/metabolismo , Animais , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/patologia , Osso e Ossos/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células , Doenças do Cão/diagnóstico por imagem , Doenças do Cão/patologia , Cães , Estudos de Viabilidade , Feminino , Humanos , Camundongos , Osteossarcoma/diagnóstico por imagem , Osteossarcoma/patologia , Receptor IGF Tipo 2/antagonistas & inibidores , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único , Microtomografia por Raio-X , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Virus Res ; 245: 44-51, 2018 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-29275103

RESUMO

Cathepsins, endosomal acid proteases, are transported from the trans-Golgi network to late endosomes by the mannose-6-phosphate receptor (M6PR). We have previously demonstrated that some rotavirus strains, like UK, Wa, WI61, DS-1, and YM, require the cation-dependent (CD-) M6PR and cathepsins to enter from late endosomes to the cytoplasm in MA104 cells, while other strains, like the simian strain RRV, which enter cells from maturing endosomes, do not. However, the role of other trans-Golgi network-late endosome transporters, such as the cation-independent (CI-) M6PR and sortillin-1, has not been evaluated. In this work, we found that several rotavirus strains that require the CD-M6PR for cell entry are also dependent on CI-M6PR and sortilin-1. Furthermore, we showed that the infectivity of all these rotavirus strains also requires cathepsins to enter not only MA104 cells, but also human intestinal Caco-2 cells. This study identifies sortilin-1 as a novel cell factor necessary for the infectivity of a virus; in addition, our results strongly suggest that cathepsins could be common cell factors needed for the infectivity of most rotavirus strains.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Catepsinas/genética , Interações Hospedeiro-Patógeno , Receptor IGF Tipo 2/genética , Rotavirus/metabolismo , Internalização do Vírus , Proteínas Adaptadoras de Transporte Vesicular/antagonistas & inibidores , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Transporte Biológico , Células CACO-2 , Catepsinas/antagonistas & inibidores , Catepsinas/metabolismo , Linhagem Celular , Endossomos/metabolismo , Endossomos/virologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Regulação da Expressão Gênica , Genótipo , Humanos , Macaca mulatta , Camundongos , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptor IGF Tipo 2/antagonistas & inibidores , Receptor IGF Tipo 2/metabolismo , Rotavirus/classificação , Rotavirus/genética , Rotavirus/crescimento & desenvolvimento , Especificidade da Espécie , Rede trans-Golgi/metabolismo , Rede trans-Golgi/virologia
6.
Peptides ; 91: 49-57, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28363795

RESUMO

Glucagon-like peptide (GLP)-2 stimulates intestinal epithelial proliferation by acting, in part, via IGF release from sub-epithelial myofibroblasts. The response of myofibroblasts to GLP-2 remains incompletely understood. We studied the action of GLP-2 on myofibroblasts from colon cancer and adjacent tissue, and the effects of conditioned medium from these cells on epithelial cell proliferation, migration and invasion. GLP-2 stimulated proliferation, migration and invasion of myofibroblasts and the proliferative and invasive responses of cancer-associated myofibroblasts were greater than those of myofibroblasts from adjacent tissue. The responses were inhibited by an IGF receptor inhibitor, AG1024. Conditioned medium from GLP-2 treated myofibroblasts increased proliferation, migration and invasion of SW480, HT29, LoVo epithelial cells and these responses were inhibited by AG1024; GLP-2 alone had no effect on these cells. In addition, when myofibroblasts and epithelial cells were co-cultured in Ibidi chambers there was mutual stimulation of migration in response to GLP-2. The latter increased both IGF-1 and IGF-2 transcript abundance in myofibroblasts. Moreover, a number of IGF binding proteins (IGFBP-4, -5, -7) were identified in myofibroblast medium; in the presence of GLP-2 there was increased abundance of the cleavage products of IGBBP-4 and IGFBP-5 suggesting activation of a degradation mechanism that might increase IGF bioavailability. The data suggest that GLP-2 stimulates cancer myofibroblast proliferation, migration and invasion; GLP-2 acts indirectly on epithelial cells partly via increased IGF expression in myofibroblasts and partly, perhaps, by increased bioavailability through degradation of IGFBPs.


Assuntos
Movimento Celular , Neoplasias do Colo/patologia , Peptídeo 2 Semelhante ao Glucagon/fisiologia , Mucosa Intestinal/patologia , Miofibroblastos/patologia , Idoso de 80 Anos ou mais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Neoplasias do Colo/metabolismo , Meios de Cultivo Condicionados/farmacologia , Células Epiteliais/efeitos dos fármacos , Feminino , Peptídeo 2 Semelhante ao Glucagon/farmacologia , Células HT29 , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Redes e Vias Metabólicas/efeitos dos fármacos , Miofibroblastos/efeitos dos fármacos , Invasividade Neoplásica , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 2/antagonistas & inibidores , Receptor IGF Tipo 2/metabolismo , Células Tumorais Cultivadas , Tirfostinas/farmacologia
7.
Biochem J ; 474(5): 771-780, 2017 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-28096202

RESUMO

We have recently shown that hypoxia and Akt-induced stem cell factor (HASF) protects the heart from ischemia-induced damage and promotes cardiomyocyte proliferation. While we have identified certain signaling pathways responsible for these protective effects, the receptor mediating these effects was unknown. Here, we undertook studies to identify the HASF receptor. A yeast two-hybrid screen identified a partial fragment of insulin-like growth factor 1 receptor (IGF1R) as a binding partner of HASF. Subsequent co-immunoprecipitation experiments showed that HASF bound to full-length IGF1R. Binding assays revealed a high affinity of HASF for IGF1R. The treatment of neonatal ventricular cardiomyocytes with HASF resulted in the phosphorylation of IGF1R and other proteins known to be involved in IGF1R-mediated signaling pathways. HASF-mediated ERK activation was abrogated by IGF1R pharmacological inhibitors and siRNAs that targeted IGF1R. However, siRNA-mediated knockdown of either IGF2R or the insulin receptor had no effect on HASF-induced cell signaling. Additionally, pharmacologic inhibition of IGF1R impeded HASF's ability to induce cardiomyocyte proliferation. Finally, we documented that in vivo deletion of the IGF1R completely abolished the ability of HASF to promote cardiomyocyte proliferation in an overexpression mouse model providing further evidence in vivo that the IGF1R is the functional receptor for HASF.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Ventrículos do Coração/metabolismo , Proteínas de Membrana/genética , Miócitos Cardíacos/metabolismo , Receptor IGF Tipo 1/genética , Proteínas Adaptadoras de Transporte Vesicular/antagonistas & inibidores , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Animais Recém-Nascidos , Sítios de Ligação , Proliferação de Células/efeitos dos fármacos , Regulação da Expressão Gênica , Células HEK293 , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Humanos , Ligantes , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Fosforilação , Cultura Primária de Células , Ligação Proteica , Pirimidinas/farmacologia , Pirróis/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 2/antagonistas & inibidores , Receptor IGF Tipo 2/genética , Receptor IGF Tipo 2/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transdução de Sinais , Técnicas do Sistema de Duplo-Híbrido
8.
Proc Natl Acad Sci U S A ; 113(20): E2766-75, 2016 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-27140600

RESUMO

Among the 15 extracellular domains of the mannose 6-phosphate/insulin-like growth factor-2 receptor (M6P/IGF2R), domain 11 has evolved a binding site for IGF2 to negatively regulate ligand bioavailability and mammalian growth. Despite the highly evolved structural loops of the IGF2:domain 11 binding site, affinity-enhancing AB loop mutations suggest that binding is modifiable. Here we examine the extent to which IGF2:domain 11 affinity, and its specificity over IGF1, can be enhanced, and we examine the structural basis of the mechanistic and functional consequences. Domain 11 binding loop mutants were selected by yeast surface display combined with high-resolution structure-based predictions, and validated by surface plasmon resonance. We discovered previously unidentified mutations in the ligand-interacting surface binding loops (AB, CD, FG, and HI). Five combined mutations increased rigidity of the AB loop, as confirmed by NMR. When added to three independently identified CD and FG loop mutations that reduced the koff value by twofold, these mutations resulted in an overall selective 100-fold improvement in affinity. The structural basis of the evolved affinity was improved shape complementarity established by interloop (AB-CD) and intraloop (FG-FG) side chain interactions. The high affinity of the combinatorial domain 11 Fc fusion proteins functioned as ligand-soluble antagonists or traps that depleted pathological IGF2 isoforms from serum and abrogated IGF2-dependent signaling in vivo. An evolved and reengineered high-specificity M6P/IGF2R domain 11 binding site for IGF2 may improve therapeutic targeting of the frequent IGF2 gain of function observed in human cancer.


Assuntos
Fator de Crescimento Insulin-Like II/metabolismo , Receptor IGF Tipo 2/metabolismo , Adulto , Sequência de Aminoácidos , Substituição de Aminoácidos , Linhagem Celular Tumoral , Cristalografia por Raios X , Evolução Molecular Direcionada , Humanos , Fator de Crescimento Insulin-Like II/química , Fator de Crescimento Insulin-Like II/genética , Modelos Moleculares , Pichia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Receptor IGF Tipo 2/antagonistas & inibidores , Receptor IGF Tipo 2/química , Receptor IGF Tipo 2/genética
9.
Cell Signal ; 27(7): 1297-304, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25817573

RESUMO

Neuropeptide Y binds to G-protein coupled receptors whose action results in inhibition of adenylyl cyclase activity. Using HEK293 cells stably expressing the native neuropeptide Y Y1 receptors, we found that the NPY agonist elicits a transient phosphorylation of the extracellular signal-regulated kinases (ERK1/2). We first show that ERK1/2 activation following Y1 receptor stimulation is dependent on heterotrimeric Gi/o since it is completely inhibited by pre-treatment with pertussis toxin. In addition, ERK1/2 activation is internalization-independent since mutant Y1 receptors unable to recruit ß-arrestins, can still activate ERK signaling to the same extent as wild-type receptors. We next show that this activation of the MAPK pathway is inhibited by the MEK inhibitor U0126, is not dependent on calcium signaling at the Y1 receptor (no effect upon inhibition of phospholipase C, protein kinase C or protein kinase D) but instead dependent on Gß/γ and associated signaling pathways that activate PI3-kinase. Although inhibition of the epidermal-growth factor receptor tyrosine kinase did not influence NPY-induced ERK1/2 activation, we show that the inhibition of insulin growth factor receptor IGFR by AG1024 completely blocks activation of ERK1/2 by the Y1 receptor. This Gß/γ-PI3K-AG1024-sensitive pathway does not involve activation of IGFR through the release of a soluble ligand by metalloproteinases since it is not affected by the metalloproteinase inhibitor marimastat. Finally, we found that a similar pathway, sensitive to wortmannin-AG1024 but insensitive to marimastat, is implicated in activation of ERK signaling in HEK293 cells by endogenously expressed GPCRs coupled to Gq-protein (muscarinic M3 receptors) or coupled to Gs-protein (endothelin ETB receptors). Our analysis is the first to show that ß-arrestin recruitment to the NPY Y1 receptor is not necessary for MAPK activation by this receptor but that transactivation of the IGFR receptor is required.


Assuntos
Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 2/metabolismo , Receptores de Neuropeptídeo Y/metabolismo , Butadienos/farmacologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Neuropeptídeo Y/farmacologia , Nitrilas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Receptor IGF Tipo 1/agonistas , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 2/antagonistas & inibidores , Receptor IGF Tipo 2/genética , Receptores de Neuropeptídeo Y/genética , Transdução de Sinais/efeitos dos fármacos , Ativação Transcricional , Tirfostinas/farmacologia
10.
Angew Chem Int Ed Engl ; 54(20): 5952-6, 2015 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-25802144

RESUMO

The development of personalized and non-invasive cancer therapies based on new targets combined with nanodevices is a major challenge in nanomedicine. In this work, the over-expression of a membrane lectin, the cation-independent mannose 6-phosphate receptor (M6PR), was specifically demonstrated in prostate cancer cell lines and tissues. To efficiently target this lectin a mannose-6-phosphate analogue was synthesized in six steps and grafted onto the surface of functionalized mesoporous silica nanoparticles (MSNs). These MSNs were used for in vitro and ex vivo photodynamic therapy to treat prostate cancer cell lines and primary cell cultures prepared from patient biopsies. The results demonstrated the efficiency of M6PR targeting for prostate cancer theranostic.


Assuntos
Biomarcadores Tumorais/antagonistas & inibidores , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/tratamento farmacológico , Receptor IGF Tipo 2/antagonistas & inibidores , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Humanos , Masculino , Manosefosfatos/síntese química , Manosefosfatos/química , Nanopartículas/química , Nanopartículas/uso terapêutico , Tamanho da Partícula , Fotoquimioterapia , Porosidade , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Receptor IGF Tipo 2/genética , Dióxido de Silício/química , Propriedades de Superfície
11.
Int J Oncol ; 45(3): 1241-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24968760

RESUMO

Insulin-like growth factor-II (IGF-II)/IGF2R signaling plays a pivotal role in cell growth, migration and differentiation in many malignancies. An individual with high IGF-II expression levels has a high risk of developing cancer, but IGF2R is often considered to be a tumor suppressor. To date, little has been reported about the role of IGF-II/IGF2R signaling in hemangiomas (HAs). Thus, uncovering the mechanisms of IGF-II/IGF2R signaling is very important to understanding the development of HAs. In the present study, the expression of IGF-II and IGF2R was investigated in 27 cases of HAs of different phases by immunohistochemistry. Through lentivirus-mediated IGF2R siRNA (Lv-siIGF2R) in HA-derived endothelial cells (HDECs), we observed the effects of IGF2R knockdown on the biological behavior of HA cells. We found that the expression of IGF-II and IGF2R was significantly increased in proliferating phase HAs, but decreased in involuting phase HAs. Furthermore, knockdown of IGF2R in vitro significantly diminished the proliferative activity and induced apoptosis and cycle arrest with decreased expression of PCNA, Ki-67, Bcl-2, Cyclin D1 and E and increased the expression of Bax in the proliferative phase HAs (HDEC and CRL-2586 EOMA cells). In addition, the tumor volumes in a subcutaneous HDEC nude mouse model treated with Lv-siIGF2R were significantly smaller than those of the control group. Taken together, our findings indicate that the expression of IGF-II and IGF2R is increased in proliferating phase HAs, and knockdown of IGF2R suppresses proliferation and induces apoptosis in HA cells in vitro and in vivo, suggesting that IGF2R may represent a novel therapeutic target for the treatment of human HAs.


Assuntos
Apoptose , Técnicas de Silenciamento de Genes/métodos , Hemangioma/terapia , Fator de Crescimento Insulin-Like II/metabolismo , RNA Interferente Pequeno/metabolismo , Receptor IGF Tipo 2/antagonistas & inibidores , Animais , Estudos de Casos e Controles , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Terapia Genética , Hemangioma/patologia , Humanos , Técnicas In Vitro , Lentivirus/genética , Camundongos , Camundongos Nus , Neoplasias Experimentais , RNA Interferente Pequeno/genética , Receptor IGF Tipo 2/genética
12.
Curr Pharm Des ; 20(17): 2899-911, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23944362

RESUMO

Insulin-like growth factors (IGFs), along with their receptors and binding proteins, play key roles in human cell proliferation, differentiation and apoptosis. There is now substantial evidence suggesting that the IGF system is involved in the pathogenesis and progression of various malignancies. Recent studies have shown that targeting of the IGF-1 receptor (IGF-1R) signaling pathway might be a novel approach for the treatment of cancer. Presently numerous agents featuring different mechanisms of IGF targeting methods such as IGF-1R monoclonal antibodies, IGF-1R tyrosine kinase inhibitors and IGF ligand specific antibodies are being investigated in more than 170 clinical trials and appear to have potential therapeutic efficacy. However, advanced trials reiterate the importance of predictive biomarkers to guide the clinical efforts of these agents. As a result, current research strategies are emerging to identify the most suitable subpopulations of patients that might benefit from these treatments. Furthermore, newly presented toxicity and growth hormone response and implication of hybrid receptors in IGF signaling pathway pose unprecedented challenges in the design and application of anti-IGF agents. On the other hand, cross-talk in downstream signaling between IGF-1R and other tumor promoting pathways and the development of multi-target agents might encourage the IGF-1R-targeted therapies further into comprehensive treatments of cancer. With both challenges and prospects ahead, this paper reviewed the progress in this particular field.


Assuntos
Antineoplásicos/uso terapêutico , Terapia de Alvo Molecular/tendências , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 2/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Somatomedinas/efeitos dos fármacos , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/farmacologia , Humanos , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/uso terapêutico , Modelos Biológicos , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 2/metabolismo , Somatomedinas/metabolismo
13.
Int J Cancer ; 133(11): 2542-50, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23686499

RESUMO

Although loss of the mannose 6-phosphate/insulin-like growth factor-II receptor (M6P/IGF-IIR) in breast cancer is believed to play a role in tumorigenesis, it has not been demonstrated that M6P/IGF-IIR loss is sufficient to confer a malignant phenotype in an untransformed cell. We investigated the impact of M6P/IGF-IIR silencing using phenotypically normal (MCF-10A) and oncogenically transformed (MCF-10T, the c-Ha-ras transformed derivative of MCF-10A) human breast epithelial cell lines as model systems. In both cell lines, silencing of M6P/IGF-IIR increased cell proliferation and motility, with the effects being more pronounced in MCF-10A cells. Although anchorage-independent growth was increased by M6P/IGF-IIR silencing in MCF-10T cells, MCF-10A cells did not acquire the ability to grow in soft agar. Conversely, reduced M6P/IGF-IIR expression increased the invasive potential of MCF-10A cells, but did not enhance the already high rate of invasion of MCF-10T cells. M6P/IGF-IIR silencing had no effect on basal or IGF-II-stimulated IGF-I receptor (IGF-IR) or AKT phosphorylation in either cell line, but both were abrogated by IGF-IR kinase inhibition, which also reduced the stimulatory effect of M6P/IGF-IIR silencing on proliferation under basal and IGF-II-stimulated conditions in both cell lines. However, cell motility was neither stimulated by IGF-II nor reduced by IGF-IR inhibition, suggesting that potentiation of specific tumorigenic features in response to M6P/IGF-IIR silencing involves IGF-II- dependent and -independent mechanisms. Collectively, these data suggest that M6P/IGF-IIR silencing alone is insufficient to confer a tumorigenic phenotype, but can enhance tumorigenicity in an already transformed cell.


Assuntos
Neoplasias da Mama/genética , Transformação Celular Neoplásica/genética , Células Epiteliais/metabolismo , Receptor IGF Tipo 2/metabolismo , Mama/citologia , Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular , Células Epiteliais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Receptor IGF Tipo 2/antagonistas & inibidores , Receptor IGF Tipo 2/genética
14.
Eur J Pharmacol ; 683(1-3): 276-84, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22429571

RESUMO

We examined the effects of L-ascorbic acid and its analogues on DNA synthesis and cell proliferation. We also investigated the signal transduction pathways involved in the induction of mitogenesis by L-ascorbic acid and its analogues using primary cultures of adult rat hepatocytes. Following a 4-h serum-free cultivation, both L-ascorbic acid and its stable analogue, L-ascorbic acid 2-glucoside, time- and dose-dependently stimulated hepatocyte DNA synthesis and cell proliferation, with EC50 values of 6.46×10⁻8 M and 3.34×10⁻8 M, respectively. Dehydroascorbic acid (10⁻6 M-10⁻5 M) weakly stimulated hepatocyte mitogenesis, whereas isoascorbic acid (10⁻9 M-10⁻5 M) had no effect. Hepatocyte mitogenesis induced by L-ascorbic acid or L-ascorbic acid 2-glucoside was dose-dependently abolished by treatment with monoclonal antibodies against insulin-like growth factor (IGF)-I receptor, but not by treatment with monoclonal antibodies against insulin receptor or IGF-II receptor. Western blot analysis showed that both L-ascorbic acid and L-ascorbic acid 2-glucoside significantly stimulated IFG-I receptor tyrosine kinase activity within 3 min, and mitogen-activated protein (MAP) kinase activity within 5 min. These results demonstrate that both L-ascorbic acid and L-ascorbic acid 2-glucoside induce DNA synthesis and cell proliferation in primary cultures of adult rat hepatocytes by interacting with the IGF-I receptor site and by activating the receptor tyrosine kinase/MAP kinase pathway.


Assuntos
Ácido Ascórbico/análogos & derivados , Ácido Ascórbico/metabolismo , Proliferação de Células , DNA/biossíntese , Hepatócitos/metabolismo , Mitógenos/farmacologia , Transdução de Sinais , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Ácido Ascórbico/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Glucosídeos/farmacologia , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Concentração Osmolar , Ratos , Ratos Wistar , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 2/antagonistas & inibidores , Receptor IGF Tipo 2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
15.
PLoS One ; 7(2): e32191, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22363814

RESUMO

The ability of embryonic stem cells to differentiate into endothelium and form functional blood vessels has been well established and can potentially be harnessed for therapeutic angiogenesis. However, after almost two decades of investigation in this field, limited knowledge exists for directing endothelial differentiation. A better understanding of the cellular mechanisms regulating vasculogenesis is required for the development of embryonic stem cell-based models and therapies. In this study, we elucidated the mechanistic role of insulin-like growth factors (IGF1 and 2) and IGF receptors (IGFR1 and 2) in endothelial differentiation using an embryonic stem cell embryoid body model. Both IGF1 or IGF2 predisposed embryonic stem to differentiate towards a mesodermal lineage, the endothelial precursor germ layer, as well as increased the generation of significantly more endothelial cells at later stages. Inhibition of IGFR1 signaling using neutralizing antibody or a pharmacological inhibitor, picropodophyllin, significantly reduced IGF-induced mesoderm and endothelial precursor cell formation. We confirmed that IGF-IGFR1 signaling stabilizes HIF1α and leads to up-regulation of VEGF during vasculogenesis in embryoid bodies. Understanding the mechanisms that are critical for vasculogenesis in various models will bring us one step closer to enabling cell based therapies for neovascularization.


Assuntos
Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/fisiologia , Fator de Crescimento Insulin-Like II/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Proteínas de Transporte/metabolismo , Diferenciação Celular/efeitos dos fármacos , Corpos Embrioides/citologia , Corpos Embrioides/efeitos dos fármacos , Corpos Embrioides/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/enzimologia , Endotélio/citologia , Endotélio/efeitos dos fármacos , Endotélio/embriologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Mesoderma/citologia , Mesoderma/efeitos dos fármacos , Mesoderma/embriologia , Camundongos , Modelos Biológicos , Podofilotoxina/análogos & derivados , Podofilotoxina/farmacologia , Estabilidade Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 2/antagonistas & inibidores , Receptor IGF Tipo 2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Biol Reprod ; 84(3): 440-6, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20980691

RESUMO

Insulin-like growth factor 2 (IGF2) enhances proliferation and survival of human first-trimester cytotrophoblasts (CTB) by signaling through the insulin-like growth factor 1 receptor (IGF1R). However, the role of the IGF2 receptor (IGF2R) in regulating trophoblast kinetics is unclear: It could act as a clearance receptor for trafficking excess ligand to lysosomes for degradation and/or directly mediate IGF2 signaling. We used an IGF2R knockdown strategy in BeWo cells and placental villous explants to investigate trophoblast proliferation and survival in response to stimulation by IGF. Both IGF1 and IGF2 significantly (P < 0.001) increased mitosis and reduced apoptosis in serum-starved BeWo cells. Small interfering RNA (siRNA)-mediated knockdown of IGF2R further enhanced IGF2-stimulated mitosis (P < 0.01), and IGF2-mediated rescue of apoptosis (P < 0.001) in these cells. Leu(27)IGF2, an IGF2 analogue that binds to IGF2R but not IGF1R, also protected IGF2R-expressing BeWo cells from apoptosis but did not increase mitosis. IGF treatment of term placental villous explants with reduced syncytial expression of IGF2R increased CTB proliferation (P < 0.001) and decreased apoptosis (P < 0.01) compared to untreated controls. Moreover, IGF2-mediated rescue of CTB apoptosis was significantly greater than that in tissue with normal IGF2R expression. Leu(27)IGF2 promoted mitogenesis and survival only in explants with intact IGF2R expression. Given that altered CTB turnover is observed in pregnancies complicated by fetal growth restriction, the development of strategies to manipulate the IGF2R signaling axis in the syncytiotrophoblast may provide a therapeutic avenue for treating this condition.


Assuntos
Fator de Crescimento Insulin-Like II/farmacologia , Fator de Crescimento Insulin-Like II/farmacocinética , Placenta/efeitos dos fármacos , Receptor IGF Tipo 2/fisiologia , Trofoblastos/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Feminino , Técnicas de Silenciamento de Genes , Humanos , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like II/fisiologia , Taxa de Depuração Metabólica , Mitose/efeitos dos fármacos , Mitose/genética , Modelos Biológicos , Placenta/metabolismo , Placenta/fisiologia , Gravidez , Processamento de Proteína Pós-Traducional/fisiologia , RNA Interferente Pequeno/farmacologia , Receptor IGF Tipo 2/antagonistas & inibidores , Receptor IGF Tipo 2/genética , Receptor IGF Tipo 2/metabolismo , Transfecção , Trofoblastos/citologia , Trofoblastos/metabolismo , Trofoblastos/fisiologia
17.
Anticancer Res ; 29(4): 1383-8, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19414391

RESUMO

BACKGROUND: The M6P/IGF-II receptor belongs to the IGF system which plays a crucial role in tumorigenicity. While the role of the IGF-I receptor in signal transduction is well documented, previous experiments failed to uncover a clear signalling function for the M6P/IGF-II receptor. However, more recent studies have shown the capability of M6P/IGF-II receptor to initiate transmembrane signalling. MATERIALS AND METHODS: Human melanoma cells were used to detect the cell surface expression of the M6P/IGF-II receptor and its modulation by different effectors and monoclonal anti-receptor antibodies. RESULTS: M6P (5 mM) caused an increase of the luminescent receptor signal of about 50% . Pre-incubation of cells with Act-D (5 microg/mL) or CHI (10 microg/mL) following M6P stimulation in the presence of the inhibitors caused a reduction of receptor cell surface expression of 27% or 31%, respectively. The monoclonal antibody (mAb) 2G11 was able to mimic the M6P effect on the receptor up-regulation but the mAb MEM-238 did not. The synergistic effect detected with the combination of M6P and the mAb 2G11 and the failure of 2G11 to compete with the M6P action suggests that both effectors have different binding sites on the receptor. Unlike 2G11 the mAb MEM-238 prevented the M6P effect on receptor up-regulation confirming partially overlapping binding epitopes of both effectors. Brefeldin A was shown to have an inhibiting effect on the vesicular transport of the receptor protein to the plasma membrane and forskolin had an activating effect on the receptor exocytosis with the following enhanced integration into the plasma membrane. CONCLUSION: Up-regulation of the tumour suppressor M6P/IGF-II receptor might represent an approach for anticancer therapy. In addition, results support recent data on the receptor's capability of signal transduction.


Assuntos
Anticorpos Monoclonais/imunologia , Manosefosfatos/metabolismo , Melanoma/metabolismo , Receptor IGF Tipo 2/metabolismo , Anticorpos Monoclonais/farmacologia , Brefeldina A/farmacologia , Colforsina/farmacologia , Humanos , Ligantes , Luminescência , Melanoma/imunologia , Melanoma/patologia , Inibidores da Síntese de Proteínas/farmacologia , Receptor IGF Tipo 2/antagonistas & inibidores , Receptor IGF Tipo 2/imunologia , Células Tumorais Cultivadas
18.
Biochem Biophys Res Commun ; 374(1): 101-5, 2008 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-18602895

RESUMO

The biological mechanism of a recent discovered association of type 2 diabetes with the ACAA-insertion/deletion polymorphism at the 3'UTR of the IGF2R gene has remained unclear. A very recently emerging novel polymorphic control layer by microRNAs (miRNAs) makes it possible to elucidate this issue. In this study, a prediction by web tools MicroInspector and miRanda demonstrated that DNA sequence polymorphism (DSPs) ACAA-insertion/deletion in IGF2R 3'UTR is located within the hsa-miR-657 and hsa-miR-453 binding sites. And luciferase reporter assay revealed that hsa-miR-657 acts directly at the 3'UTR of the IGF2R. Furthermore, ACAA-deletion exerted a further repression compared with ACAA-insertion, indicating that hsa-miR-657 regulates IGF2R gene expression in a polymorphic control manner. Importantly, we also demonstrated that hsa-miR-657 can translationally regulate the IGF2R expression levels in Hep G2 cells. Thus, our findings testify the possibility that the ACAA-insertion/deletion polymorphism may result in the change of IGF2R expression levels at least in part by hsa-miR-657-mediated regulation, contributing to the elucidation for the pathogenesis of type 2 diabetes and raise the possibility that miRNAs or in combination with functional DNA sequence polymorphism may be valuable in the treatment of human type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/genética , Regulação da Expressão Gênica , MicroRNAs/metabolismo , Polimorfismo Genético , Receptor IGF Tipo 2/genética , Regiões 3' não Traduzidas/genética , Regiões 3' não Traduzidas/metabolismo , Alelos , Sítios de Ligação/genética , Linhagem Celular , Biologia Computacional , Humanos , Mutagênese Insercional , Receptor IGF Tipo 2/antagonistas & inibidores , Deleção de Sequência
19.
Cancer Res ; 68(6): 1997-2005, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18339882

RESUMO

We previously showed that transgenic enhancement of histamine production in B16-F10 melanomas strongly supports tumor growth in C57BL/6 mice. In the present study, gene expression profiles of transgenic mouse melanomas, secreting different amounts of histamine, were compared by whole genome microarrays. Array results were validated by real-time PCR, and genes showing histamine-affected behavior were further analyzed by immunohistochemistry. Regulation of histamine-coupled genes was investigated by checking the presence and functional integrity of all four known histamine receptors in experimental melanomas and by administering histamine H1 receptor (H1R) and H2 receptor (H2R) antagonists to tumor-bearing mice. Finally, an attempt was made to integrate histamine-affected genes in known gene regulatory circuits by in silico pathway analysis. Our results show that histamine enhances melanoma growth via H1R rather than through H2R. We show that H1R activation suppresses RNA-level expression of the tumor suppressor insulin-like growth factor II receptor (IGF-IIR) and the antiangiogenic matrix protein fibulin-5 (FBLN5), decreases their intracellular protein levels, and also reduces their availability in the plasma membrane and extracellular matrix, respectively. Pathway analysis suggests that because plasma membrane-bound IGF-IIR is required to activate matrix-bound, latent transforming growth factor-beta1, a factor suggested to sustain FBLN5 expression, the data can be integrated in a known antineoplastic regulatory pathway that is suppressed by H1R. On the other hand, we show that engagement of H2R also reduces intracellular protein pools of IGF-IIR and FBLN5, but being a downstream acting posttranslational effect with minimal consequences on exported IGF-IIR and FBLN5 protein levels, H2R is rather irrelevant compared with H1R in melanoma.


Assuntos
Proteínas da Matriz Extracelular/biossíntese , Liberação de Histamina/fisiologia , Melanoma Experimental/metabolismo , Receptor IGF Tipo 2/biossíntese , Proteínas Recombinantes/biossíntese , Animais , Western Blotting , Proteínas da Matriz Extracelular/antagonistas & inibidores , Proteínas da Matriz Extracelular/genética , Feminino , Perfilação da Expressão Gênica , Histamina/biossíntese , Histamina/genética , Antagonistas dos Receptores Histamínicos/farmacologia , Liberação de Histamina/genética , Imuno-Histoquímica , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Família Multigênica , Análise de Sequência com Séries de Oligonucleotídeos , Receptor IGF Tipo 2/antagonistas & inibidores , Receptor IGF Tipo 2/genética , Receptores Histamínicos/fisiologia , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética
20.
Microb Pathog ; 44(5): 410-6, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18162363

RESUMO

Chlamydia pneumoniae is a human respiratory pathogen that has also been associated with cardiovascular disease. C. pneumoniae infection accelerates atherosclerotic plaque development in hyperlipidemic animals and promotes oxidation of low density lipoprotein in vitro. All-trans-retinoic acid (ATRA), an antioxidant, has been shown to inhibit C. pneumoniae infectivity for endothelial cells by preventing binding of the organism to the M6P/IGF2 receptor on the cell surface. This current study investigates whether ATRA similarly affects C. pneumoniae infectivity of epithelial cells, which are the primary site of infection in the respiratory tract, and the effects on intracellular growth in both endothelial and epithelial cells. Because ATRA binds to both the nuclear retinoid acid receptor (RAR) and the M6P/IGF2 receptor, 4-[(E)-2-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)-1-propenyl]benzoic acid (TTNPB), an ATRA analog, which binds to the RAR but not the M6P/IGF2 receptor was used to differentiate the receptor mediating the effects of ATRA. The results of this study showed two separate effects of ATRA. The first effect is through interaction with the M6P/IGF2 receptor on the cell surface preventing attachment of the organism (inhibition by ATRA but not TTNPB) in endothelial cells and the second is through the nuclear receptor (inhibition by both ATRA and TTNPB) which inhibits growth in both epithelial and endothelial cells.


Assuntos
Chlamydophila pneumoniae/imunologia , Células Endoteliais/microbiologia , Receptor IGF Tipo 2/antagonistas & inibidores , Receptores do Ácido Retinoico/antagonistas & inibidores , Tretinoína/imunologia , Aderência Bacteriana/efeitos dos fármacos , Linhagem Celular , Chlamydophila pneumoniae/crescimento & desenvolvimento , Citoplasma/microbiologia , Citoplasma/ultraestrutura , Humanos , Corpos de Inclusão/microbiologia , Corpos de Inclusão/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...