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










Base de dados
Intervalo de ano de publicação
1.
Nat Cancer ; 5(1): 66-84, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38151625

RESUMO

Chromosomal instability (CIN) is a hallmark of cancer, caused by persistent errors in chromosome segregation during mitosis. Aggressive cancers like high-grade serous ovarian cancer (HGSOC) and triple-negative breast cancer (TNBC) have a high frequency of CIN and TP53 mutations. Here, we show that inhibitors of the KIF18A motor protein activate the mitotic checkpoint and selectively kill chromosomally unstable cancer cells. Sensitivity to KIF18A inhibition is enriched in TP53-mutant HGSOC and TNBC cell lines with CIN features, including in a subset of CCNE1-amplified, CDK4-CDK6-inhibitor-resistant and BRCA1-altered cell line models. Our KIF18A inhibitors have minimal detrimental effects on human bone marrow cells in culture, distinct from other anti-mitotic agents. In mice, inhibition of KIF18A leads to robust anti-cancer effects with tumor regression observed in human HGSOC and TNBC models at well-tolerated doses. Collectively, our results provide a rational therapeutic strategy for selective targeting of CIN cancers via KIF18A inhibition.


Assuntos
Cinesinas , Neoplasias de Mama Triplo Negativas , Humanos , Animais , Camundongos , Cinesinas/genética , Cinesinas/metabolismo , Mitose/genética , Linhagem Celular , Pontos de Checagem da Fase M do Ciclo Celular
2.
J Med Chem ; 65(6): 4972-4990, 2022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-35286090

RESUMO

Chromosomal instability (CIN) is a hallmark of cancer that results from errors in chromosome segregation during mitosis. Targeting of CIN-associated vulnerabilities is an emerging therapeutic strategy in drug development. KIF18A, a mitotic kinesin, has been shown to play a role in maintaining bipolar spindle integrity and promotes viability of CIN cancer cells. To explore the potential of KIF18A, a series of inhibitors was identified. Optimization of an initial hit led to the discovery of analogues that could be used as chemical probes to interrogate the role of KIF18A inhibition. Compounds 23 and 24 caused significant mitotic arrest in vivo, which was sustained for 24 h. This would be followed by cell death either in mitosis or in the subsequent interphase. Furthermore, photoaffinity labeling experiments reveal that this series of inhibitors binds at the interface of KIF18A and tubulin. This study represents the first disclosure of KIF18A inhibitors with in vivo activity.


Assuntos
Cinesinas , Neoplasias , Morte Celular , Humanos , Mitose , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Fuso Acromático/metabolismo , Tubulina (Proteína)/metabolismo
3.
Cancer Discov ; 8(12): 1582-1597, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30254093

RESUMO

The prosurvival BCL2 family member MCL1 is frequently dysregulated in cancer. To overcome the significant challenges associated with inhibition of MCL1 protein-protein interactions, we rigorously applied small-molecule conformational restriction, which culminated in the discovery of AMG 176, the first selective MCL1 inhibitor to be studied in humans. We demonstrate that MCL1 inhibition induces a rapid and committed step toward apoptosis in subsets of hematologic cancer cell lines, tumor xenograft models, and primary patient samples. With the use of a human MCL1 knock-in mouse, we demonstrate that MCL1 inhibition at active doses of AMG 176 is tolerated and correlates with clear pharmacodynamic effects, demonstrated by reductions in B cells, monocytes, and neutrophils. Furthermore, the combination of AMG 176 and venetoclax is synergistic in acute myeloid leukemia (AML) tumor models and in primary patient samples at tolerated doses. These results highlight the therapeutic promise of AMG 176 and the potential for combinations with other BH3 mimetics. SIGNIFICANCE: AMG 176 is a potent, selective, and orally bioavailable MCL1 inhibitor that induces a rapid commitment to apoptosis in models of hematologic malignancies. The synergistic combination of AMG 176 and venetoclax demonstrates robust activity in models of AML at tolerated doses, highlighting the promise of BH3-mimetic combinations in hematologic cancers.See related commentary by Leber et al., p. 1511.This article is highlighted in the In This Issue feature, p. 1494.

4.
Cancer Cell ; 32(6): 731-747.e6, 2017 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-29232552

RESUMO

Bone metastasis is a major health threat to breast cancer patients. Tumor-derived Jagged1 represents a central node in mediating tumor-stromal interactions that promote osteolytic bone metastasis. Here, we report the development of a highly effective fully human monoclonal antibody against Jagged1 (clone 15D11). In addition to its inhibitory effect on bone metastasis of Jagged1-expressing tumor cells, 15D11 dramatically sensitizes bone metastasis to chemotherapy, which induces Jagged1 expression in osteoblasts to provide a survival niche for cancer cells. We further confirm the bone metastasis-promoting function of osteoblast-derived Jagged1 using osteoblast-specific Jagged1 transgenic mouse model. These findings establish 15D11 as a potential therapeutic agent for the prevention or treatment of bone metastasis.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Proteína Jagged-1/antagonistas & inibidores , Animais , Humanos , Camundongos , Camundongos Transgênicos , Osteoblastos/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Eur Respir J ; 48(4): 1137-1149, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27471204

RESUMO

Pulmonary arterial hypertension (PAH) is characterised by excessive pulmonary vascular remodelling involving deregulated proliferation of cells in intima, media as well as adventitia. Pulmonary arterial endothelial cell (PAEC) hyperproliferation and survival underlies the endothelial pathobiology of the disease.The indispensable involvement of Notch1 in the arterial endothelial phenotype and angiogenesis provides intriguing prospects for its involvement in the pathogenesis of PAH.We observed an increased expression of Notch1 in lungs of idiopathic PAH (IPAH) patients and hypoxia/SU5416 (SUHx) rats compared with healthy subjects. In vitro loss- and gain-of-function studies demonstrated that Notch1 increased proliferation of human PAECs (hPAECs) via downregulation of p21 and inhibited apoptosis via Bcl-2 and Survivin. Inhibition of Notch signalling using the γ-secretase inhibitor dibenzazepine dose-dependently decreased proliferation and migration of hPAECs. Notably, Notch1 expression and transcriptional activity were increased under hypoxia in hPAECs and knockdown of Notch1 inhibited hypoxia-induced proliferation of the cells. Furthermore, in vivo treatment with a γ-secretase inhibitor (AMG2008827) significantly reduced the right ventricular systolic pressure and right heart hypertrophy in SUHx rats.Here, we conclude that Notch1 plays a critical role in PAH and Notch inhibitors may be a promising therapeutic option for PAH.


Assuntos
Apoptose , Endotélio Vascular/patologia , Receptor Notch1/metabolismo , Animais , Estudos de Casos e Controles , Hipóxia Celular , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Regulação para Baixo , Ecocardiografia , Hipertensão Pulmonar Primária Familiar , Células HEK293 , Humanos , Hipertrofia , Proteínas Inibidoras de Apoptose/metabolismo , Neovascularização Patológica , Fenótipo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Receptores Notch/metabolismo , Transdução de Sinais , Survivina , Transcrição Gênica
6.
Infect Dis (Lond) ; 48(4): 303-309, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26585858

RESUMO

Background Alcohol intake has been suggested to have an impact on the development of many chronic diseases. How alcohol intake may modulate risk of Helicobacter pylori (H. pylori) infection, however, remains a subject open for investigation. A dose-response meta-analysis was performed of epidemiological studies to better quantify this relationship. Materials and methods Twelve observational articles were identified. The summary odds ratio (OR) and confidence intervals (CI) were calculated for alcohol drinkers vs non-drinkers. The summary OR estimates were obtained using the random-effects model and dose-response meta-analysis. Sub-group and sensitivity analysis were also conducted. Results The summary OR was 0.78 (95% CI = 0.69-0.89). The dose-response analysis demonstrated that for drinkers of 10, 15, 30, 60 and 96 g/day alcohol intake, the estimated ORs were 0.80 (95% CI = 0.76-0.85), 0.79 (95% CI = 0.75-0.84), 0.83 (95% CI = 0.78-0.87), 0.85 (95% CI = 0.78-0.93) and 0.87 (95% CI = 0.70-1.06), respectively, compared to non-drinkers. The inverse relationship between alcohol intake and H. pylori infection was consistent, regardless of sex, age, geographic areas, detection methods or beverage types. CONCLUSION: Evidence from these observational studies suggests that moderate alcohol intake is associated with a reduction in H. pylori infection of ∼ 22% and may facilitate elimination of H. pylori.


Assuntos
Consumo de Bebidas Alcoólicas/epidemiologia , Infecções por Helicobacter/epidemiologia , Helicobacter pylori/isolamento & purificação , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Observacionais como Assunto , Razão de Chances , Fatores de Risco
7.
Mol Cancer Ther ; 9(2): 400-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20124448

RESUMO

AMG 102 is a fully human monoclonal antibody that selectively targets and neutralizes hepatocyte growth factor/scatter factor (HGF/SF). A detailed biochemical and functional characterization of AMG 102 was done to support its clinical development for the treatment of cancers dependent on signaling through the HGF/SF:c-Met pathway. In competitive equilibrium binding experiments, AMG 102 bound to human and cynomolgus monkey HGF with affinities of approximately 19 pmol/L and 41 pmol/L, respectively. However, AMG 102 did not detect mouse or rabbit HGF on immunoblots. Immunoprecipitation experiments showed that AMG 102 preferentially bound to the mature, active form of HGF, and incubation of AMG 102/HGF complexes with kallikrein protease indicated that AMG 102 had no apparent effect on proteolytic processing of the inactive HGF precursor. AMG 102 inhibited human and cynomolgus monkey HGF-induced c-Met autophosphorylation in PC3 cells with IC(50) values of 0.12 nmol/L and 0.24 nmol/L, respectively. AMG 102 also inhibited cynomolgus monkey HGF-induced migration of human MDA-MB-435 cells but not rat HGF-induced migration of mouse 4T1 cells. Epitope-mapping studies of recombinant HGF molecules comprising human/mouse chimeras and human-to-mouse amino acid substitutions showed that amino acid residues near the NH(2)-terminus of the beta-chain are critical for AMG 102 binding. Bound AMG 102 protected one trypsin protease cleavage site near the NH(2)-terminus of the beta-chain of human HGF, further substantiating the importance of this region for AMG 102 binding. Currently, AMG 102 is in phase II clinical trials in a variety of solid tumor indications. Mol Cancer Ther; 9(2); 400-9.


Assuntos
Anticorpos Monoclonais/química , Fator de Crescimento de Hepatócito/química , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Mapeamento de Epitopos , Humanos , Immunoblotting , Concentração Inibidora 50 , Macaca fascicularis , Camundongos , Biblioteca de Peptídeos , Fosforilação , Primatas , Coelhos , Proteínas Recombinantes/química
8.
Mol Cancer Ther ; 8(10): 2803-10, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19825800

RESUMO

Hepatocyte growth factor/scatter factor (HGF/SF) and its receptor, c-Met, have been implicated in the growth and progression of a variety of solid human tumors. Thus, inhibiting HGF/SF:c-Met signaling may provide a novel therapeutic approach for treating human tumors. We have generated and characterized fully human monoclonal antibodies that bind to and neutralize human HGF/SF. In this study, we tested the effects of the investigational, human anti-human HGF/SF monoclonal antibody, AMG 102, and a mixture of mouse anti-human HGF/SF monoclonal antibodies (Amix) on HGF/SF-mediated cell migration, proliferation, and invasion in vitro. Both agents had high HGF/SF-neutralizing activity in these cell-based assays. The HGF/SF:c-Met pathway has been implicated in the growth of sarcomas; thus, we also investigated the effect of AMG 102 on the growth of human leiomyosarcoma (SK-LMS-1) in HGF/SF transgenic C3H severe combined immunodeficient mice engineered to express high levels of human HGF/SF, as well as tumor growth of an autocrine variant of the SK-LMS-1 cell line (SK-LMS-1TO) in nude mice. The results indicate that interrupting autocrine and/or paracrine HGF/SF:c-Met signaling with AMG 102 has profound antitumor effects. These findings suggest that blocking HGF/SF:c-Met signaling may provide a potent intervention strategy to treat patients with HGF/SF:c-Met-dependent tumors.


Assuntos
Anticorpos Neutralizantes/uso terapêutico , Comunicação Autócrina , Fator de Crescimento de Hepatócito/imunologia , Leiomiossarcoma/tratamento farmacológico , Leiomiossarcoma/patologia , Comunicação Parácrina , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Anticorpos Neutralizantes/farmacologia , Comunicação Autócrina/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Fibrinolisina/metabolismo , Humanos , Leiomiossarcoma/imunologia , Ligantes , Camundongos , Camundongos Transgênicos , Modelos Imunológicos , Invasividade Neoplásica , Comunicação Parácrina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Clin Cancer Res ; 13(22 Pt 1): 6735-42, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18006775

RESUMO

PURPOSE: Hepatocyte growth factor (HGF/SF) and its receptor c-Met have previously been shown to be up-regulated in multiple human cancers, including glioblastoma multiforme. To better understand if AMG 102, a fully human, anti-HGF/SF-neutralizing antibody, could be incorporated into current clinical practice, AMG 102 was tested preclinically in combination with temozolomide or docetaxel to determine if enhanced efficacy was observed compared with AMG 102 alone. EXPERIMENTAL DESIGN: The effects of AMG 102 were tested for antiproliferative activity in combination with temozolomide or docetaxel on U-87 MG cells in vitro and for antitumor activity in a U-87 MG xenograft model in vivo. Apoptotic activity was also measured for AMG 102 and docetaxel combined in vitro. RESULTS: Treatment with temozolomide combined with AMG 102 resulted in increased inhibition of cell growth in vitro compared with treatment with either single agent alone. In U-87 MG xenografts in vivo, AMG 102 combined with temozolomide or docetaxel significantly increased the inhibitory effect on tumor growth when compared with treatment with either agent alone (P < 0.0001 and P < 0.015, respectively). In vitro, docetaxel alone induced both caspase-3/7 activity as well as poly(ADP)ribose polymerase and caspase-7 cleavage in U-87 MG cells; these events were enhanced when used in combination with AMG 102. Importantly, there was no evidence of interference between AMG 102 and either temozolomide or docetaxel in vitro or in vivo. CONCLUSION: These studies support testing of AMG 102 in combination with temozolomide or docetaxel. Such combinations may represent promising, novel clinical therapeutic strategies for cancers that are dependent on the HGF/SF/SF:c-Met pathway in the oncology setting.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Dacarbazina/análogos & derivados , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Taxoides/uso terapêutico , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dacarbazina/farmacologia , Dacarbazina/uso terapêutico , Docetaxel , Humanos , Camundongos , Camundongos Endogâmicos , Taxoides/farmacologia , Temozolomida , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Clin Cancer Res ; 12(14 Pt 1): 4154-62, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16857786

RESUMO

PURPOSE: Many proteins are proteolytically released from the cell surface by a process known as ectodomain shedding. Shedding occurs under normal physiologic conditions and can be increased in certain pathologies. Among the many receptors for which ectodomain shedding has been shown is c-Met, the hepatocyte growth factor (HGF) receptor tyrosine kinase. HGF stimulates mitogenesis, motogenesis, and morphogenesis in a variety of cellular targets during development, homeostasis, and tissue regeneration. Inappropriate HGF signaling resulting in unregulated cell proliferation, motility, and invasion occurs in several human malignancies. This can occur through paracrine signaling, autocrine loop formation, receptor mutation, gene amplification, or gene rearrangement, accompanied frequently with overexpression of ligand and/or receptor proteins. We hypothesized that c-Met overexpression in cancer might result in increased ectodomain shedding, and that its measure could be a useful biomarker of tumor progression. EXPERIMENTAL DESIGN: We developed a sensitive electrochemiluminescent immunoassay to quantitate c-Met protein in cell lysates, culture supernatants, and biological samples. RESULTS: A survey of cultured cell models of oncogenic transformation revealed significant direct correlations (P < 0.001, t test or ANOVA) between malignant potential and the rate of c-Met ectodomain shedding that was independent of steady-state receptor expression level. Moreover, weekly plasma and urine samples from mice harboring s.c. human tumor xenografts (n = 4 per group) displayed soluble human c-Met levels that were measurable before tumors became palpable and that correlated directly with tumor volume (R2 > 0.92, linear regression). CONCLUSIONS: For a variety of human cancers, c-Met ectodomain shedding may provide a reliable and practical indicator of malignant potential and overall tumor burden.


Assuntos
Proteínas Proto-Oncogênicas c-met/química , Animais , Biomarcadores Tumorais/química , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Progressão da Doença , Relação Dose-Resposta a Droga , Eletroquímica , Humanos , Luminescência , Camundongos , Metástase Neoplásica , Transplante de Neoplasias , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-met/metabolismo , Proteínas Recombinantes/química
11.
Cancer Res ; 66(3): 1721-9, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16452232

RESUMO

c-Met is a well-characterized receptor tyrosine kinase for hepatocyte growth factor (HGF). Compelling evidence from studies in human tumors and both cellular and animal tumor models indicates that signaling through the HGF/c-Met pathway mediates a plethora of normal cellular activities, including proliferation, survival, migration, and invasion, that are at the root of cancer cell dysregulation, tumorigenesis, and tumor metastasis. Inhibiting HGF-mediated signaling may provide a novel therapeutic approach for treating patients with a broad spectrum of human tumors. Toward this goal, we generated and characterized five different fully human monoclonal antibodies that bound to and neutralized human HGF. Antibodies with subnanomolar affinities for HGF blocked binding of human HGF to c-Met and inhibited HGF-mediated c-Met phosphorylation, cell proliferation, survival, and invasion. Using a series of human-mouse chimeric HGF proteins, we showed that the neutralizing antibodies bind to a unique epitope in the beta-chain of human HGF. Importantly, these antibodies inhibited HGF-dependent autocrine-driven tumor growth and caused significant regression of established U-87 MG tumor xenografts. Treatment with anti-HGF antibody rapidly inhibited tumor cell proliferation and significantly increased the proportion of apoptotic U-87 MG tumor cells in vivo. These results suggest that an antibody to an epitope in the beta-chain of HGF has potential as a novel therapeutic agent for treating patients with HGF-dependent tumors.


Assuntos
Anticorpos Monoclonais/farmacologia , Glioblastoma/terapia , Fator de Crescimento de Hepatócito/imunologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/imunologia , Linhagem Celular Tumoral , Epitopos/imunologia , Feminino , Glioblastoma/imunologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Imunoglobulina G/biossíntese , Imunoglobulina G/química , Imunoglobulina G/imunologia , Camundongos , Camundongos Nus , Fosforilação , Ensaios Antitumorais Modelo de Xenoenxerto
12.
J Biomed Opt ; 10(3): 034006, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16229650

RESUMO

Multiphoton microscopy has evolved into a powerful bioimaging tool in three dimensions. However, the ability to image biological specimens in-depth can be hindered by sample spherical aberration and scattering. These two phenomena can result in the degradation of image resolution and the loss of detected multiphoton signal. In this work, we use the correction collar (for cover glass thickness) associated with a water immersion objective in an attempt to improve multiphoton imaging. In the two samples we examined (human skin and rat tail tendon), we found that while the improvement in image resolution was not visible qualitatively, the measured axial fluorescence or second harmonic generation signal profiles indicate that the use of the correction collar can help to improve the detected multiphoton signals. The maximum increases are 36% and 57% for the skin (sulforhodamine B fluorescence) and tendon (second harmonic generation) specimens, respectively. Our result shows that for in-depth multiphoton imaging, the correction collar may be used to correct for spherical aberration. However, each tissue type needs to be examined to determine the optimal correction collar setting to be used.


Assuntos
Artefatos , Aumento da Imagem/instrumentação , Imageamento Tridimensional/instrumentação , Microscopia de Fluorescência por Excitação Multifotônica/instrumentação , Pele/citologia , Tendões/citologia , Animais , Desenho de Equipamento , Análise de Falha de Equipamento , Humanos , Imageamento Tridimensional/métodos , Técnicas In Vitro , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Ratos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
13.
Eur Biophys J ; 33(7): 572-9, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15045473

RESUMO

Ricin is a promising candidate for the treatment of cancer because it can be selectively targeted to tumor cells via linkage to monoclonal antibodies. Biochemical evidence suggests that escape of ricin or its ribosome-inactivating subunit from an intracellular compartment is mediated by retrograde transport to the endoplasmic reticulum and subsequent direction into the ER-associated degradation pathway. Alternatively, lipase activity of ricin may facilitate leakage from endocytic vesicles. We have observed ricin-mediated release of macromolecular dyes from lipid vesicles that mimic the composition of endosomal membranes. Release of small molecules occurs to the same extent, suggesting an all-or-none mechanism due to bilayer destabilization. The level of accompanying membrane fusion depends on vesicle composition. Since it takes 24 h of incubation before the first traces of lysolipids are detectable by matrix-assisted laser desorption/ionization mass spectrometry, membrane destabilization is not due to the lipase activity of ricin.


Assuntos
Endossomos/química , Gangliosídeo G(M1)/química , Lipossomos/química , Fluidez de Membrana , Fosfatidilcolinas/química , Ricina/química , Difusão , Cinética , Membranas Artificiais , Porosidade , Soluções
14.
Opt Express ; 11(25): 3377-84, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19471468

RESUMO

In this work, we demonstrate the application of multiphoton polarization imaging in resolving the structures in surface stratum corneum and dermal layers of ex-vivo human skin. By varying the excitation and emission polarizations, we characterized the structural features in both Laurdan labeled stratum corneum and dermal fibers. The results presented here have important consequences in bioimaging applications of the skin. Both the mechanics of transdermal drug delivery across the skin and physiological significance of the structural changes of the dermis can be monitored. Our results show that the transition dipoles of Laurdan molecules are preferentially oriented normal to the membrane surface. Furthermore, polarization imaging shows that fibrous structures in the dermis generate emission aligned strongly along the excitation polarization. This work shows that multiphoton polarization imaging can be a powerful method in identifying structural orientations in the skin and other biological structures.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA