Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cardiovasc Res ; 118(1): 141-155, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-33135065

RESUMO

AIMS: During atherosclerosis, smooth muscle cells (SMCs) accumulate in the intima where they switch from a contractile to a synthetic phenotype. From porcine coronary artery, we isolated spindle-shaped (S) SMCs exhibiting features of the contractile phenotype and rhomboid (R) SMCs typical of the synthetic phenotype. S100A4 was identified as a marker of R-SMCs in vitro and intimal SMCs, in pig and man. S100A4 exhibits intra- and extracellular functions. In this study, we investigated the role of extracellular S100A4 in SMC phenotypic transition. METHODS AND RESULTS: S-SMCs were treated with oligomeric recombinant S100A4 (oS100A4), which induced nuclear factor (NF)-κB activation. Treatment of S-SMCs with oS100A4 in combination with platelet-derived growth factor (PDGF)-BB induced a complete SMC transition towards a pro-inflammatory R-phenotype associated with NF-κB activation, through toll-like receptor-4. RNA sequencing of cells treated with oS100A4/PDGF-BB revealed a strong up-regulation of pro-inflammatory genes and enrichment of transcription factor binding sites essential for SMC phenotypic transition. In a mouse model of established atherosclerosis, neutralization of extracellular S100A4 decreased area of atherosclerotic lesions, necrotic core, and CD68 expression and increased α-smooth muscle actin and smooth muscle myosin heavy chain expression. CONCLUSION: We suggest that the neutralization of extracellular S100A4 promotes the stabilization of atherosclerotic plaques. Extracellular S100A4 could be a new target to influence the evolution of atherosclerotic plaques.


Assuntos
Anticorpos Neutralizantes/farmacologia , Doenças da Aorta/tratamento farmacológico , Aterosclerose/tratamento farmacológico , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Placa Aterosclerótica , Proteína A4 de Ligação a Cálcio da Família S100/antagonistas & inibidores , Actinas/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Becaplermina/farmacologia , Células Cultivadas , Modelos Animais de Doenças , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Cadeias Pesadas de Miosina/metabolismo , Fenótipo , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Proteína A4 de Ligação a Cálcio da Família S100/farmacologia , Transdução de Sinais , Miosinas de Músculo Liso/metabolismo , Sus scrofa , Receptor 4 Toll-Like/metabolismo
2.
Mol Cancer Ther ; 19(12): 2598-2611, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32999046

RESUMO

S100A4 oncoprotein plays a critical role during prostate cancer progression and induces immunosuppression in host tissues. We hypothesized that S100A4-regulated oncogenic activity in immunosuppressed prostate tumors promotes growth of neoplastic cells, which are likely to become aggressive. In the current study, we investigated whether biopsy-S100A4 gene alteration independently predicts the outcome of disease in patients and circulatory-S100A4 is druggable target for treating immunosuppressive prostate cancer. Aided by DECIPHER-genomic test, we show biopsy-S100A4 overexpression as predictive of (i) poor ADT response and (ii) high risk of mortality in 228 radical prostatectomy-treated patients. Furthermore, analysis of tumor genome data of more than 1,000 patients with prostate cancer (PRAD/SU2C/FHCRC studies) validated the association of S100A4-alteration to poor survival and metastasis. We show that increased serum-S100A4 levels are associated to the prostate cancer progression in patients. The prerequisite for metastasis is the escape of tumor cells via vascular system. We show that extracellular-S100A4 protein as a growth factor induces vascular transmigration of prostate cancer cells and bone demineralization thus forms an ideal target for therapies for treating prostate cancer. By employing surface plasmon resonance and isothermal titration calorimetry, we show that mab6B12 antibody interacts with and neutralizes S100A4 protein. When tested for therapeutic efficacy, the mab6B12 therapy reduced the (i) osteoblastic demineralization of bone-derived MSCs, (ii) S100A4-target (NFκB/MMP9/VEGF) levels in prostate cancer cells, and (iii) tumor growth in a TRAMPC2 syngeneic mouse model. The immuno-profile analysis showed that mAb6B12-therapy (i) shifted Th1/Th2 balance (increased Stat4+/T-bet+ and decreased GATA2+/CD68+/CD45+/CD206+ cells); (ii) modulated cytokine levels in CD4+ T cells; and (iii) decreased levels of IL5/6/12/13, sTNFR1, and serum-RANTES. We suggest that S100A4-antibody therapy has clinical applicability in treating immunosuppressive prostate cancer in patients.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Imunomodulação/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , Proteína A4 de Ligação a Cálcio da Família S100/antagonistas & inibidores , Antineoplásicos Imunológicos/farmacologia , Biomarcadores Tumorais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Humanos , Biópsia Líquida , Contagem de Linfócitos , Masculino , Prognóstico , Neoplasias da Próstata/sangue , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/etiologia , Proteína A4 de Ligação a Cálcio da Família S100/sangue , Proteína A4 de Ligação a Cálcio da Família S100/genética , Resultado do Tratamento
3.
Methods Mol Biol ; 1929: 339-365, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30710284

RESUMO

The metastasis-promoting S100A4 protein, a member of the S100 family, has recently been discovered as a potent factor implicated in various inflammation-associated diseases. S100A4 is involved in a range of biological functions such as angiogenesis, cell differentiation, apoptosis, motility, and invasion. Moreover, S100A4 is also a potent trigger of inflammatory processes and induces the release of cytokines and growth factors under different pathological conditions.Indeed, the release of S100A4 upon stress and mainly its pro-inflammatory role emerges as the most decisive activity in disease development, such as rheumatoid arthritis (RA), systemic sclerosis (SSc) allergy, psoriasis, and cancer. In the scope of this review, we will focus on the role of S100A4 as a mediator of pro-inflammatory pathways and its associated biological processes involved in the pathogenesis of various human noncommunicable diseases (NCDs) including cancer.


Assuntos
Inflamação/metabolismo , Neoplasias/metabolismo , Proteína A4 de Ligação a Cálcio da Família S100/genética , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Animais , Apoptose , Movimento Celular , Citocinas/genética , Citocinas/metabolismo , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Inflamação/genética , Metástase Neoplásica , Neoplasias/genética , Microambiente Tumoral
4.
Biol Cell ; 2018 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-29907957

RESUMO

BACKGROUND INFORMATION: Tumor stroma remodeling is a key feature of malignant tumors and can promote cancer progression. Laminins are major constituents of basement membranes that physically separate the epithelium from the underlying stroma. RESULTS: By employing mouse models expressing high and low levels of the laminin α1 chain (LMα1), we highlighted its implication in a tumor-stroma crosstalk, thus leading to increased colon tumor incidence, angiogenesis and tumor growth. The underlying mechanism involves attraction of carcinoma-associated fibroblasts by LMα1, VEGFA expression triggered by the complex integrin α2ß1-CXCR4 and binding of VEGFA to LM-111, which in turn promotes angiogenesis, tumor cell survival and proliferation. A gene signature comprising LAMA1, ITGB1, ITGA2, CXCR4 and VEGFA has negative predictive value in colon cancer. CONCLUSIONS: Together, we have identified VEGFA, CXCR4 and α2ß1 integrin downstream of LMα1 in colon cancer as of bad prognostic value for patient survival. SIGNIFICANCE: This information opens novel opportunities for diagnosis and treatment of colon cancer.

5.
Eur J Cancer ; 68: 114-124, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27750112

RESUMO

INTRODUCTION: S100a4 is a calcium-binding protein belonging to the family of S100-proteins, highly expressed in different stromal cell types. S100A4 has been reported as a prognostic marker in colorectal cancer in association with tumour progression and metastasis. METHODS: In this study, we analysed the in vivo role of S100a4 in intestinal tumour initiation and progression using different transgenic and knockout mouse models. RESULTS: We found that genetic ablation or overexpression of S100a4 in both Apc- and Smad4-mutant mice do not affect tumour initiation in the intestinal tract. In contrast, S100a4 epithelial overexpression in Apc1638N/+/KRASV12G mice increases the dissemination of intestinal tumour cells to the liver, in agreement with its role in tumour metastasis. Moreover, we report a novel role for S100a4 in desmoid formation where S100a4 deficiency results in a significant reduction of the tumour burden characteristic of the Apc1638N model. In agreement with these results, S100a4 appears to be co-expressed together with mesenchymal stem cell (MSC) markers in desmoid tumours from Apc1638N/+ mice, as well as from sporadic and hereditary human desmoids. CONCLUSION: Our data provide the first report on the in vivo role of S100a4 in intestinal tumourigenesis and describe a new role for S100a4 in the aetiology of desmoids formation.


Assuntos
Carcinogênese/genética , Neoplasias Colorretais/genética , Fibromatose Agressiva/genética , Proteína A4 de Ligação a Cálcio da Família S100/genética , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Neoplasias Colorretais/metabolismo , Técnicas de Introdução de Genes , Humanos , Neoplasias Intestinais/genética , Camundongos , Camundongos Knockout , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Proteína Smad4/genética
6.
BMC Cancer ; 15: 44, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25884510

RESUMO

BACKGROUND: The tumor microenvironment plays a determinative role in stimulating tumor progression and metastasis. Notably, tumor-stroma signals affect the pattern of infiltrated immune cells and the profile of tumor-released cytokines. Among the known molecules that are engaged in stimulating the metastatic spread of tumor cells is the S100A4 protein. S100A4 is known as an inducer of inflammatory processes and has been shown to attract T-cells to the primary tumor and to the pre-metastatic niche. The present study aims to examine the immunomodulatory role of S100A4 in vivo and in vitro and assess the mode of action of 6B12, a S100A4 neutralizing antibody. METHODS: The therapeutic effect of the 6B12 antibody was evaluated in two different mouse models. First, in a model of spontaneous breast cancer we assessed the dynamics of tumor growth and metastasis. Second, in a model of metastatic niche formation we determined the expression of metastatic niche markers. The levels of cytokine expression were assessed using antibody as well as PCR arrays and the results confirmed by qRT-PCR and ELISA. T-cell phenotyping and in vitro differentiation analyses were performed by flow cytometry. RESULTS: We show that the S100A4 protein alters the expression of transcription factor and signal transduction pathway genes involved in the T-cell lineage differentiation. T-cells challenged with S100A4 demonstrated reduced proportion of Th1-polarized cells shifting the Th1/Th2 balance towards the Th2 pro-tumorigenic phenotype. The 6B12 antibody restored the Th1/Th2 balance. Furthermore, we provide evidence that the 6B12 antibody deploys its anti-metastatic effect, by suppressing the attraction of T-cells to the site of primary tumor and pre-metastatic niche. This was associated with delayed primary tumor growth, decreased vessel density and inhibition of metastases. CONCLUSION: The S100A4 blocking antibody (6B12) reduces tumor growth and metastasis in a model of spontaneous breast cancer. The 6B12 antibody treatment inhibits T cell accumulation at the primary and pre-metastatic tumor sites. The 6B12 antibody acts as an immunomodulatory agent and thus supports the view that the 6B12 antibody is a promising therapeutic candidate to fight cancer.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Neoplasias/imunologia , Neoplasias/metabolismo , Proteínas S100/antagonistas & inibidores , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Feminino , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Camundongos , Camundongos Knockout , Metástase Neoplásica , Neoplasias/genética , Neoplasias/patologia , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/genética , Proteínas S100/metabolismo , Transdução de Sinais , Baço/imunologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T/metabolismo , Microambiente Tumoral/genética
7.
Cell Adh Migr ; 9(1-2): 4-13, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25611571

RESUMO

The extracellular matrix (ECM) molecule tenascin-C (TNC) promotes tumor progression. This has recently been demonstrated in the stochastic murine RIP1-Tag2 insulinoma model, engineered to either express TNC abundantly or to be devoid of TNC. However, our knowledge about organization of the TNC microenvironment is scant. Here we determined the spatial distribution of TNC together with other ECM molecules in murine RIP1-Tag2 insulinoma and human cancer tissue (insulinoma and colorectal carcinoma). We found that TNC is organized in matrix tracks together with other ECM molecules of the AngioMatrix signature, a previously described gene expression profile that characterizes the angiogenic switch. Moreover, stromal cells including endothelial cells, fibroblasts and leukocytes were enriched in the TNC tracks. Thus, TNC tracks may provide niches for stromal cells and regulate their behavior. Given similarities of TNC rich niches for stromal cells in human insulinoma and colon cancer, we propose that the RIP1-Tag2 model may be useful for providing insights into the contribution of the tumor stroma specific ECM as promoter of cancer progression.


Assuntos
Movimento Celular/fisiologia , Neoplasias Colorretais/metabolismo , Matriz Extracelular/metabolismo , Células Estromais/patologia , Tenascina/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Fibroblastos/metabolismo , Humanos , Camundongos Transgênicos
8.
Sci Transl Med ; 6(218): 218ra4, 2014 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-24401939

RESUMO

The identification of diagnostic markers and therapeutic candidate genes in common diseases is complicated by the involvement of thousands of genes. We hypothesized that genes co-regulated with a key gene in allergy, IL13, would form a module that could help to identify candidate genes. We identified a T helper 2 (TH2) cell module by small interfering RNA-mediated knockdown of 25 putative IL13-regulating transcription factors followed by expression profiling. The module contained candidate genes whose diagnostic potential was supported by clinical studies. Functional studies of human TH2 cells as well as mouse models of allergy showed that deletion of one of the genes, S100A4, resulted in decreased signs of allergy including TH2 cell activation, humoral immunity, and infiltration of effector cells. Specifically, dendritic cells required S100A4 for activating T cells. Treatment with an anti-S100A4 antibody resulted in decreased signs of allergy in the mouse model as well as in allergen-challenged T cells from allergic patients. This strategy, which may be generally applicable to complex diseases, identified and validated an important diagnostic and therapeutic candidate gene in allergy.


Assuntos
Estudos de Associação Genética , Hipersensibilidade/genética , Hipersensibilidade/imunologia , Proteínas S100/genética , Pesquisa Translacional Biomédica , Adulto , Animais , Anticorpos Bloqueadores/farmacologia , Polaridade Celular , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Dermatite/diagnóstico , Dermatite/genética , Dermatite/imunologia , Dermatite/prevenção & controle , Modelos Animais de Doenças , Epitopos/efeitos dos fármacos , Deleção de Genes , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Hipersensibilidade/diagnóstico , Hipersensibilidade/prevenção & controle , Memória Imunológica/efeitos dos fármacos , Interleucina-13/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Imunológicos , Rinite Alérgica Sazonal/diagnóstico , Rinite Alérgica Sazonal/genética , Rinite Alérgica Sazonal/imunologia , Rinite Alérgica Sazonal/patologia , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/deficiência , Células Th2/citologia , Células Th2/imunologia
9.
Genes Cancer ; 4(5-6): 224-34, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-24069509

RESUMO

S100A4, a calcium-binding protein, is known for its role in the metastatic spread of tumor cells, a late event of cancer disease. This is the first report showing that S100A4 is not merely a metastatic protein but also an oncoprotein that plays a critical role in the development of tumors. We earlier showed that S100A4 expression progressively increases in prostatic tissues with the advancement of prostate cancer (CaP) in TRAMP, an autochthonous mouse model. To study the functional significance of S100A4 in CaP, we generated a heterozygously deleted S100A4 (TRAMP/S100A4(+/-)) genotype by crossing TRAMP with S100A4(-/-) mice. TRAMP/S100A4(+/-) did not show a lethal phenotype, and transgenes were functional. As compared to age-matched TRAMP littermates, TRAMP/S100A4(+/-) mice exhibited 1) an increased tumor latency period (P < 0.001), 2) a 0% incidence of metastasis, and 3) reduced prostatic weights (P < 0.001). We generated S100A4-positive clones from S100A4-negative CaP cells and tested their potential. S100A4-positive tumors grew at a faster rate than S100A4-negative tumors in vitro and in a xenograft mouse model. The S100A4 protein exhibited growth factor-like properties in multimode (intracellular and extracellular) forms. We observed that 1) the growth-promoting effect of S100A4 is due to its activation of NFκB, 2) S100A4-deficient tumors exhibit reduced NFκB activity, 3) S100A4 regulates NFκB through the RAGE receptor, and 4) S100A4 and RAGE co-localize in prostatic tissues of mice. Keeping in view its growth-promoting role, we suggest that S100A4 qualifies as an excellent candidate to be exploited for therapeutic agents to treat CaP in humans.

10.
Biochim Biophys Acta ; 1833(12): 2653-2663, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23830916

RESUMO

OBJECTIVE: S100A4 is a Ca-binding protein that regulates cell growth, survival, and motility. The abundant expression of S100A4 in rheumatiod arthritis contributes to the invasive growth of joint tissue and to bone damage. In the present study, we analysed the role of S100A4 in bone homeostasis. METHODS: Peripheral quantitative computed tomography and histomorphometric analysis were performed in mice lacking the entire S100A4 protein (S100A4KO) and in wild-type (WT) counterparts treated with shRNA-lentiviral constructs targeting S100A4 (S100A4-shRNA). Control groups consisted of sex-matched WT counterparts and WT mice treated with a non-targeting RNA construct. RESULTS: S100A4 deficiency was associated with higher trabecular and cortical bone mass, increased number and thickness of trabeculi combined with larger periosteal circumference and higher predicted bone strength. S100A4 inhibition by shRNA led to an increase in cortical bone in WT mice. S100A4-deficieny was associated with a reduced number of functional osteoclasts. S100A4KO and S100A4-shRNA-treated bone marrow progenitors gave rise to a large number of small TRAP+ cells with few nuclei and few pseudopodial processes. Poor osteoclastogenesis and the low resorptive capacity in S100A4Ko mice may be linked to low levels of surface integrins, impaired adhesion capacity, and poor multinucleation in S100A4-deficient osteoclasts, as well as a low content of proteolytic enzymes cathepsin K and MMP3 and MMP9 to break down the organic matrix. CONCLUSION: S100A4 emerges as a negative regulator of bone metabolism potentially responsible for the excessive bone turnover in conditions marked by high levels of S100A4 protein, such as inflammation and rheumatoid arthritis.


Assuntos
Reabsorção Óssea/metabolismo , Osteoclastos/metabolismo , Osteoclastos/patologia , Proteínas S100/metabolismo , Animais , Remodelação Óssea , Reabsorção Óssea/complicações , Reabsorção Óssea/patologia , Reabsorção Óssea/fisiopatologia , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Membrana Celular/metabolismo , Forma Celular , Matriz Extracelular/metabolismo , Integrinas/metabolismo , Metaloproteinases da Matriz/metabolismo , Camundongos , Tamanho do Órgão , Osteólise/complicações , Osteólise/patologia , Osteólise/fisiopatologia , Fenótipo , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/deficiência
11.
J Mol Cell Cardiol ; 57: 72-81, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23352991

RESUMO

Metastasis-associated protein, S100A4 is suggested as a marker for fibrosis in several organs. It also modulates DNA binding of p53 and affects its function. However, the functional role of S100A4 in the myocardium has remained unclear. Therefore, we investigated the role of S100A4 and its relationship with p53 in cardiac fibrosis. In Dahl-rat hypertensive heart disease model, S100A4 was upregulated in the hypertrophic myocardium and further activated during transition to heart failure (HF). It was expressed in various cells including fibroblasts. In in vitro cardiac fibroblasts, the knockdown of S100A4 significantly suppressed both cell proliferation and collagen expressions. S100A4 co-localized and interacted with p53 in the nucleus. S100A4 knockdown increased the expression of p53-downstream genes, p21 and mdm2, and concomitant knockdown of p53 recovered cell proliferation and collagen expression. Transverse aortic constriction (TAC) was performed in S100A4 knockout (KO) mice, which showed a similar baseline-phenotype to wild type (WT) mice. Although there was no difference in hypertrophic response, KO mice showed reduced interstitial fibrosis, decreased myofibroblasts, and suppressed expressions of collagens and profibrotic cytokines in the left ventricle. Also, DNA microarray analysis showed that S100A4 knockout in vivo had a significant impact on expressions of p53-associated genes. These findings suggest that S100A4 modulates p53 function in fibroblasts and thereby mediates myocardial interstitial fibrosis through two distinct mechanisms; cell proliferation and collagen expression. Blockade of S100A4 may have therapeutic potential in cardiac hypertrophy and HF by attenuating cardiac fibrosis.


Assuntos
Insuficiência Cardíaca/metabolismo , Ventrículos do Coração/patologia , Miofibroblastos/metabolismo , Proteínas S100/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Angiotensina II/fisiologia , Animais , Proliferação de Células , Colágeno/genética , Colágeno/metabolismo , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fibrose , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Insuficiência Cardíaca/patologia , Ventrículos do Coração/metabolismo , Hipertrofia Ventricular Esquerda/metabolismo , Hipertrofia Ventricular Esquerda/patologia , Masculino , Camundongos , Camundongos Knockout , Miofibroblastos/fisiologia , Células NIH 3T3 , Peptídeo Natriurético Encefálico/sangue , Ratos , Ratos Endogâmicos Dahl , Proteína A4 de Ligação a Cálcio da Família S100 , Transcriptoma
12.
Nat Commun ; 3: 1197, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23149742

RESUMO

Identification of novel pro-survival factors in the brain is paramount for developing neuroprotective therapies. The multifunctional S100 family proteins have important roles in many human diseases and are also upregulated by brain injury. However, S100 functions in the nervous system remain unclear. Here we show that the S100A4 protein, mostly studied in cancer, is overexpressed in the damaged human and rodent brain and released from stressed astrocytes. Genetic deletion of S100A4 exacerbates neuronal loss after brain trauma or excitotoxicity, increasing oxidative cell damage and downregulating the neuroprotective protein metallothionein I+II. We identify two neurotrophic motifs in S100A4 and show that these motifs are neuroprotective in animal models of brain trauma. Finally, we find that S100A4 rescues neurons via the Janus kinase/STAT pathway and, partially, the interleukin-10 receptor. Our data introduce S100A4 as a therapeutic target in neurodegeneration, and raise the entire S100 family as a potentially important factor in central nervous system injury.


Assuntos
Citoproteção , Metástase Neoplásica/patologia , Neurônios/patologia , Proteínas S100/metabolismo , Motivos de Aminoácidos , Animais , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Morte Celular/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , Feminino , Deleção de Genes , Células HEK293 , Humanos , Janus Quinases/metabolismo , Ácido Caínico , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/metabolismo , Neurotoxinas/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Ratos , Receptores de Interleucina-10/metabolismo , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/química , Fatores de Transcrição STAT/metabolismo , Convulsões/tratamento farmacológico , Convulsões/patologia , Regulação para Cima/efeitos dos fármacos
13.
Neoplasia ; 14(12): 1260-8, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23308057

RESUMO

The small Ca-binding protein, S100A4, has a well-established metastasis-promoting activity. Moreover, its expression is tightly correlated with poor prognosis in patients with numerous types of cancer. Mechanistically, the extracellular S100A4 drives metastasis by affecting the tumor microenvironment, making it an attractive target for anti-cancer therapy. In this study, we produced a function-blocking anti-S100A4 monoclonal antibody with metastasis-suppressing activity. Antibody treatment significantly reduced metastatic burden in the lungs of experimental animals by blocking the recruitment of T cells to the site of the primary tumor. In vitro studies demonstrated that this antibody efficiently reduced the invasion of T cells in a fibroblast monolayer. Moreover, it was capable of suppressing the invasive growth of human and mouse fibroblasts. We presume therefore that the antibody exerts its activity by suppressing stroma cell recruitment to the site of the growing tumor. Our epitope mapping studies suggested that the antibody recognition site overlaps with the target binding interface of human S100A4. We conclude here that this antibody could serve as a solid basis for development of an efficient anti-metastatic therapy.


Assuntos
Adenocarcinoma/imunologia , Anticorpos Antineoplásicos , Neoplasias da Mama/imunologia , Neoplasias Pulmonares/secundário , Proteínas S100/imunologia , Microambiente Tumoral/imunologia , Adenocarcinoma/secundário , Animais , Anticorpos Monoclonais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular , Mapeamento de Epitopos , Fibroblastos , Humanos , Neoplasias Pulmonares/prevenção & controle , Camundongos , Proteína A4 de Ligação a Cálcio da Família S100 , Células Estromais , Linfócitos T/imunologia
14.
Respir Res ; 12: 159, 2011 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-22185646

RESUMO

BACKGROUND: Idiopathic and familial forms of pulmonary arterial hypertension (PAH) occur more frequently in women than men. However, the reason for this remains unknown. Both the calcium binding protein S100A4/Mts1 (Mts1) and its endogenous receptor (receptor for advanced glycosylation end products; RAGE) have been implicated in the development of PAH. We wished to investigate if the Mts1/RAGE pathway may play a role in the gender bias associated with PAH. METHODS: We investigated the effects of gender on development of PAH in mice over-expressing Mts1 (Mts1+ mice) via measurement of pulmonary arterial remodeling, systolic right ventricular pressure (sRVP) and right ventricular hypertrophy (RVH). Gender differences in pulmonary arterial Mts1 and RAGE expression were assessed by qRT-PCR and immunohistochemistry. Western blotting and cell counts were used to investigate interactions between 17ß-estradiol, Mts1 and RAGE on proliferation of human pulmonary artery smooth muscle cells (hPASMCs). Statistical analysis was by one-way analysis of variance with Dunnetts post test or two-way analysis of variance with Bonferronis post test, as appropriate. RESULTS: Female Mts1+ mice developed increased sRVP and pulmonary vascular remodeling, whereas male Mts1+ mice remained unaffected. The development of plexiform-like lesions in Mts1+ mice was specific to females. These lesions stained positive for both Mts1 and RAGE in the endothelial and adventitial layers. Expression of pulmonary arterial Mts1 was greater in female than male Mts1+ mice, and was localised to the medial and adventitial layers in non plexiform-like pulmonary arteries. RAGE gene expression and immunoreactivity were similar between male and female Mts1+ mice and RAGE staining was localised to the endothelial layer in non plexiform-like pulmonary arteries adjacent to airways. In non-plexiform like pulmonary arteries not associated with airways RAGE staining was present in the medial and adventitial layers. Physiological concentrations of 17ß-estradiol increased Mts1 expression in hPASMCs. 17ß-estradiol-induced hPASMC proliferation was inhibited by soluble RAGE, which antagonises the membrane bound form of RAGE. CONCLUSIONS: Mts1 over-expression combined with female gender is permissive to the development of experimental PAH in mice. Up-regulation of Mts1 and subsequent activation of RAGE may contribute to 17ß-estradiol-induced proliferation of hPASMCs.


Assuntos
Pressão Sanguínea , Hipertensão Pulmonar/metabolismo , Artéria Pulmonar/metabolismo , Proteínas S100/metabolismo , Análise de Variância , Animais , Pressão Sanguínea/genética , Western Blotting , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Estradiol/metabolismo , Hipertensão Pulmonar Primária Familiar , Feminino , Predisposição Genética para Doença , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/fisiopatologia , Hipertrofia Ventricular Direita/genética , Hipertrofia Ventricular Direita/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Fenótipo , Artéria Pulmonar/patologia , Artéria Pulmonar/fisiopatologia , Reação em Cadeia da Polimerase em Tempo Real , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/genética , Fatores Sexuais , Transdução de Sinais , Regulação para Cima , Função Ventricular Direita , Pressão Ventricular
15.
J Infect Dis ; 204(5): 722-30, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21844298

RESUMO

BACKGROUND: Efficient host defense mechanisms are crucial for survival in sepsis and septic arthritis. S100 proteins are reported to have proinflammatory and bactericidal properties. The aim of this study was to investigate the role of S100A4 in staphylococcal arthritis. METHODS: S100A4 knockout mice (S100A4KO) and wild-type counterparts (WT) were intravenously and intra-articularly challenged with Staphylococcus aureus strain LS-1. Clinical and morphological signs of arthritis and sepsis, phagocytosis, bone mineral density (BMD), and bone metabolism were then monitored in S100A4 and WT mice. RESULTS: S100A4KO mice had a lower bacterial load in the kidneys than WT mice (P < .05) but developed more severe clinical signs of arthritis (P < .001) and had higher levels of interleukin 6 and L-selectin (P = .002). S100A4KO mice had fewer morphological signs of synovitis and cartilage/bone destruction following intra-articular instillation of bacteria. S100A4KO mice were protected from loss of BMD and had lower levels of RANKL, MMP3, and MMP9 (P < .05). S100A4 was not bactericidal in vitro. CONCLUSIONS: In staphylococcal infection, S100A4 regulates bacterial clearance as well as systemic and local inflammatory responses.


Assuntos
Artrite Infecciosa/patologia , Doenças das Cartilagens/patologia , Articulação do Joelho/patologia , Proteínas S100/deficiência , Infecções Estafilocócicas/patologia , Sinovite/patologia , Animais , Artrite Infecciosa/metabolismo , Artrite Infecciosa/microbiologia , Carga Bacteriana , Densidade Óssea , Antígeno CD11b/metabolismo , Antígenos CD18/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Doenças das Cartilagens/microbiologia , Cartilagem Articular/microbiologia , Cartilagem Articular/patologia , Feminino , Granulócitos/metabolismo , Interleucina-6/sangue , Rim/microbiologia , Articulação do Joelho/metabolismo , Articulação do Joelho/microbiologia , Selectina L/sangue , Metaloproteinase 3 da Matriz/sangue , Metaloproteinase 9 da Matriz/sangue , Camundongos , Camundongos Knockout , Ligante RANK/sangue , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/fisiologia , Índice de Gravidade de Doença , Infecções Estafilocócicas/metabolismo , Sinovite/microbiologia
16.
Mol Cancer Res ; 9(5): 553-63, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21454378

RESUMO

The tumor microenvironment is now recognized as a major factor in determining the survival and growth of disseminated tumor cells at potential metastatic sites. Tumor cells send signals to stroma cells and stimulate them to produce factors that in turn create favorable conditions for tumor cell metastasis. Activated fibroblasts constitute an important component of the tumor-associated stroma. We have previously shown that S100A4 protein produced by stromal fibroblasts in the primary tumor stimulates metastasis formation. Here we show that activated fibroblasts also stimulate the formation of metastases independently of S100A4 expression during organ colonization. To identify genes that could potentially interfere with fibroblast-driven metastasis, we used gene expression profiling of S100A4-deficient fibroblasts treated with and without tumor cell-conditioned media. Five differentially expressed genes encoding cell surface and secreted proteins with potential metastasis-modulating activity were selected. Expression of lymphocyte antigen 6 complex (Ly6c) and matrix metalloproteinase 3 (Mmp3) was upregulated in fibroblasts in response to tumor-conditioned medium, whereas expression of cadherin-16 (Cdh16), Ccn2, and fibulin-5 (Fbln5) was downregulated. Further analysis showed that Fibulin-5 is able to suppress the metastatic colonization of lungs and liver. Additional studies suggest a mechanism in which Fibulin-5 suppresses metastasis formation by inhibiting production of matrix metalloproteinase 9 (MMP9) and reducing the invasive behavior of fibroblasts. Together our data are consistent with the notion that tumors secrete factors that downregulate expression of Fbln5 in fibroblasts at sites of metastatic colonization, in turn upregulating Mmp9 expression and stimulating metastatic organ colonization.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Metaloproteinase 9 da Matriz/metabolismo , Animais , Antígenos Ly/efeitos dos fármacos , Antígenos Ly/metabolismo , Caderinas/efeitos dos fármacos , Caderinas/metabolismo , Linhagem Celular Tumoral , Fator de Crescimento do Tecido Conjuntivo/efeitos dos fármacos , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Meios de Cultivo Condicionados/farmacologia , Regulação para Baixo , Proteínas da Matriz Extracelular/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Neoplasias Hepáticas/genética , Neoplasias Pulmonares/genética , Metaloproteinase 3 da Matriz/efeitos dos fármacos , Metaloproteinase 3 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/efeitos dos fármacos , Camundongos
17.
BMC Cancer ; 10: 444, 2010 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-20723242

RESUMO

INTRODUCTION: Tumour phenotype is regulated in a complex fashion as a result of interactions between malignant cells and the tumour stroma. Fibroblasts are the most abundant and perhaps most active part of the tumour stroma. A better understanding of the changes that occur in fibroblasts in response to the presence of malignant cells may lead to the development of new strategies for cancer treatment. We explored the effects of fibroblasts on the growth and invasion of mammary carcinoma tumour cells in vitro and in vivo. METHODS: In order to analyse secreted factors that affect invasive abilities of breast cancer cells we co-cultured human mammary fibroblasts (HMF3s) and cancer cells (MCF7S1) in three-dimensional (3D) growth conditions devoid of heterogeneous cell-cell contact. To study the possible influence of fibroblasts on MCF7S1 cancer cell growth in vivo we co-injected HMF3s and MCF7S1 cells in Balb/c nu/nu mice. RESULTS: In 3D co-culture both HMF3s and MCF7S1 cells demonstrated enhanced invasion into a Matrigel matrix. This was correlated with enhanced expression of the metastasis promoting S100A4 protein in fibroblasts, stimulation of the matrix metalloproteinase (MMP)-2 activity, and enhanced secretion of a range of different cytokines. Orthotopic injection of oestrogen-dependent MCF7S1 cancer cells together with fibroblasts showed stimulation of tumour growth in mice without an external oestrogen supply. The resulting tumours were characterized by increased development of extracellular matrix, as well as an increase of murine S100A4 concentration and activity of MMP-2 in the tumour interstitial fluid. CONCLUSION: Stimulation of the invasive phenotype of tumour cells in 3D co-cultures with fibroblasts could be correlated with increased production of S100A4 and MMP-2. We propose that enhanced development of mouse host-derived tumour stroma in a MCF7S1 co-injection xenograft model leads to oestrogen independency and is triggered by the initial presence of human fibroblasts.


Assuntos
Neoplasias da Mama/patologia , Mama/citologia , Fibroblastos/citologia , Células Estromais/patologia , Animais , Western Blotting , Mama/metabolismo , Neoplasias da Mama/metabolismo , Células Cultivadas , Técnicas de Cocultura , Ensaio de Imunoadsorção Enzimática , Feminino , Fibroblastos/metabolismo , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/metabolismo , Células Estromais/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
18.
PLoS One ; 5(4): e10374, 2010 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-20442771

RESUMO

BACKGROUND: The tumor microenvironment has been described as a critical milieu determining tumor growth and metastases. A pivotal role of metastasis-inducing S100A4 in the development of tumor stroma has been proven in animal models and verified in human breast cancer biopsies. Expression and release of S100A4 has been shown in various types of stroma composing cells, including fibroblasts and immune cells. However, the events implicated in upstream and downstream pathways regulating the activity of the extracellular S100A4 protein in the tumor milieu remain unsolved. METHODOLOGY/PRINCIPAL FINDINGS: We studied the interplay between the tumor cell-derived cytokine regulated-upon-activation, normal T-cell expressed and secreted (RANTES; CCL5) and S100A4 which were shown to be critical factors in tumor progression. We found that RANTES stimulates the externalization of S100A4 via microparticle shedding from the plasma membrane of tumor and stroma cells. Conversely, the released S100A4 protein induces the upregulation of fibronectin (FN) in fibroblasts and a number of cytokines, including RANTES in tumor cells as well as stimulates cell motility in a wound healing assay. Importantly, using wild type and S100A4-deficient mouse models, we demonstrated a substantial influence of tumor cell-derived RANTES on S100A4 release into blood circulation which ultimately increases the metastatic burden in mice. CONCLUSIONS/SIGNIFICANCE: Altogether, the data presented strongly validate the pro-metastatic function of S100A4 in the tumor microenvironment and define how the tumor cell-derived cytokine RANTES acts as a critical regulator of S100A4-dependent tumor cell dissemination. Additionally, for the first time we demonstrated the mechanism of S100A4 release associated with plasma membrane microparticle shedding from various cells types.


Assuntos
Quimiocina CCL5/fisiologia , Metástase Neoplásica/patologia , Proteínas S100/fisiologia , Animais , Linhagem Celular Tumoral , Micropartículas Derivadas de Células , Células Cultivadas , Progressão da Doença , Fibroblastos , Macrófagos , Camundongos , Camundongos Knockout , Proteína A4 de Ligação a Cálcio da Família S100
19.
Cancer Res ; 70(3): 936-47, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20103644

RESUMO

Interactions between tumor and stroma cells are essential for the progression of cancer from its initial growth at a primary site to its metastasis to distant organs. The metastasis-stimulating protein S100A4 exerts its function as a stroma cell-derived factor. Genetic depletion of S100A4 significantly reduced the metastatic burden in lungs of PyMT-induced mammary tumors. In S100A4(+/+) PyMT mice, massive leukocyte infiltration at the site of the growing tumor at the stage of malignant transition was associated with increased concentration of extracellular S100A4 in the tumor microenvironment. In contrast, in S100A4(-/-) PyMT tumors, a significant suppression of T-cell infiltration was documented at the transition period. In vitro, the S100A4 protein mediated the attraction of T cells. Moreover, S100A4(+/+), but not S100A4(-/-), fibroblasts stimulated the invasion of T lymphocytes into fibroblast monolayers. In vivo, the presence of S100A4(+/+), but not S100A4(-/-), fibroblasts significantly stimulated the attraction of T lymphocytes to the site of the growing tumor. Increased levels of T cells were also observed in the premetastatic lungs of tumor-bearing mice primed to metastasize by S100A4(+/+) fibroblasts. Treatment of T cells with the S100A4 protein stimulated production of cytokines, particularly granulocyte colony-stimulating factor and eotaxin-2. The same cytokines were detected in the fluid of S100A4(+/+) PyMT tumors at the transition period. We suggest that release of S100A4 in the primary tumor stimulates infiltration of T cells and activates secretion of cytokines, thus triggering sequential events that fuel tumor cells to metastasize. Similar processes could occur in the premetastatic lungs, facilitating generation of inflammatory milieu favorable for metastasis formation.


Assuntos
Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Proteínas S100/genética , Linfócitos T/patologia , Animais , Western Blotting , Quimiocina CCL24/metabolismo , Citocinas/metabolismo , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Fator Estimulador de Colônias de Granulócitos/metabolismo , Imuno-Histoquímica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/virologia , Vírus do Tumor Mamário do Camundongo/fisiologia , Camundongos , Camundongos Knockout , Infecções por Retroviridae/patologia , Infecções por Retroviridae/virologia , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/metabolismo , Proteínas S100/farmacologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Infecções Tumorais por Vírus/patologia , Infecções Tumorais por Vírus/virologia
20.
FEBS J ; 276(20): 5936-48, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19740107

RESUMO

The function of S100A4, a member of the calcium-binding S100 protein family, has been associated with tumor invasion and metastasis. Although an essential pro-metastatic role of extracellular S100A4 in tumor progression has been demonstrated, the identification of the precise underlying mechanisms and protein partners (receptors) has remained elusive. To identify putative targets for extracellular S100A4, we screened a phage display peptide library using S100A4 as bait. We identified three independent peptide motifs with varying affinities for the S100A4 protein. Sequence analyses indicated that the most abundant peptide mimicked the F/YCC motif present in the epidermal growth factor domain of ErbB receptor ligands. S100A4 selectively interacted with a number of epidermal growth factor receptor (EGFR) ligands, demonstrating highest affinity for amphiregulin. Importantly, we found that S100A4 stimulated EGFR/ErbB2 receptor signaling and enhanced the amphiregulin-mediated proliferation of mouse embryonic fibroblasts. S100A4-neutralizing antibodies, as well as EGFR- and ErbB2 receptor-specific tyrosine kinase inhibitors, blocked these effects. The present results suggest that extracellular S100A4 regulates tumor progression by interacting with EGFR ligands, thereby enhancing EGFR/ErbB2 receptor signaling and cell proliferation. Structured digital abstract: * MINT-7256556: EGF (uniprotkb:P01133) binds (MI:0407) to S100A4 (uniprotkb:P26447) by far western blotting (MI:0047) * MINT-7256512: BC (uniprotkb:P35070) binds (MI:0407) to S100A4 (uniprotkb:P26447) by far western blotting (MI:0047) * MINT-7256485, MINT-7256618, MINT-7256636: AR (uniprotkb:P15514) binds (MI:0407) to S100A4 (uniprotkb:P26447) by far western blotting (MI:0047) * MINT-7256494: HB-EGF (uniprotkb:Q99075) binds (MI:0407) to S100A4 (uniprotkb:P26447) by far western blotting (MI:0047) * MINT-7256502: P53 (uniprotkb:P04637) binds (MI:0407) to S100A4 (uniprotkb:P26447) by far western blotting (MI:0047) * MINT-7256654: S100A2 (uniprotkb:P29034) binds (MI:0407) to AR (uniprotkb:P15514) by far western blotting (MI:0047) * MINT-7256693: S100A5 (uniprotkb:P33763) binds (MI:0407) to AR (uniprotkb:P15514) by far western blotting (MI:0047) * MINT-7256593: S100A4 (uniprotkb:P26447) binds (MI:0407) to BC (uniprotkb:P35070) by pull down (MI:0096) * MINT-7256567: S100A4 (uniprotkb:P26447) binds (MI:0407) to AR (uniprotkb:P15514) by pull down (MI:0096).


Assuntos
Receptores ErbB/metabolismo , Peptídeos/metabolismo , Proteínas S100/química , Proteínas S100/metabolismo , Sequência de Aminoácidos , Anfirregulina , Animais , Sítios de Ligação , Far-Western Blotting , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cromatografia em Gel , Família de Proteínas EGF , Ensaio de Imunoadsorção Enzimática , Glicoproteínas/metabolismo , Glicoproteínas/farmacologia , Humanos , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Camundongos , Dados de Sequência Molecular , Biblioteca de Peptídeos , Peptídeos/química , Peptídeos/genética , Ligação Proteica , Receptor ErbB-2/metabolismo , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/genética , Proteínas S100/farmacologia , Homologia de Sequência de Aminoácidos , Ressonância de Plasmônio de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...