Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Wound Care ; 33(Sup8a): clxxxii-cxciii, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39163155

RESUMO

Alternative 3-dimensional (3D) skin models that replicate in vivo human skin are required to investigate important events during wound healing, such as collective cell migration, epidermal layer formation, dermal substrate formation, re-epithelialisation and collagen production. In this study, a matched human 3D skin equivalent model (3D-SEM) was developed from human skin cells (fibroblast and keratinocytes), characterised using haematoxylin and eosin, immunofluorescence staining and microRNA profiling. The 3D-SEM was then functionally tested for its use in wound healing studies. Mesenchymal stem cells (MSCs) were isolated and characterised according to the criteria stipulated by the International Society for Cell Therapy. Cytokine and growth factor secretions were analysed by enzyme-linked immunosorbent assay. MSC-conditioned medium (MSC-CM) was then tested for wound healing capacity using the developed 3D-SEM at different timepoints i.e., at one, two and four weeks. The constructed 3D-SEM showed consistent development of skin-like structures composed of dermal layers and epidermal layers, with the ability to express epidermal differentiation markers and full stratification. They also showed prolonged longevity in culture media, retaining full differentiation and stratification within the four weeks. MicroRNA profiling revealed a strong correlation in microRNA expression between the developed 3D-SEM and the original native skin (p<0.001; R=0.64). Additionally, MSC-CM significantly enhanced migration, proliferation and differentiation of epidermal cells in the wounded models compared to control models at the different timepoints. In conclusion, in this study, the developed 3D-SEM mimicked native skin at the cellular and molecular levels, and clearly showed the important stages of skin regeneration during the healing process. MSC secretome contains growth factors that play a pivotal role in the healing process and could be used as a therapeutic option to accelerate skin healing.


Assuntos
Células-Tronco Mesenquimais , Cicatrização , Humanos , Meios de Cultivo Condicionados/farmacologia , Cicatrização/efeitos dos fármacos , Queratinócitos/efeitos dos fármacos , Pele/lesões , Pele/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas
2.
Wound Repair Regen ; 32(4): 475-486, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38572659

RESUMO

Chronic non-healing cutaneous wounds represent a major burden to patients and healthcare providers worldwide, emphasising the continued unmet need for credible and efficacious therapeutic approaches for wound healing. We have recently shown the potential for collagen peptides to promote proliferation and migration during cutaneous wound healing. In the present study, we demonstrate that the application of porcine-derived collagen peptides significantly increases keratinocyte and dermal fibroblast expression of integrin α2ß1 and activation of an extracellular signal-related kinase (ERK)-focal adhesion kinase (FAK) signalling cascade during wound closure in vitro. SiRNA-mediated knockdown of integrin ß1 impaired porcine-derived collagen peptide-induced wound closure and activation of ERK-FAK signalling in keratinocytes but did not impair ERK or FAK signalling in dermal fibroblasts, implying the activation of differing downstream signalling pathways. Studies in ex vivo human 3D skin equivalents subjected to punch biopsy-induced wounding confirmed the ability of porcine-derived collagen peptides to promote wound closure by enhancing re-epithelialisation. Collectively, these data highlight the translational and clinical potential for porcine-derived collagen peptides as a viable therapeutic approach to promote re-epithelialisation of superficial cutaneous wounds.


Assuntos
Colágeno , Fibroblastos , Queratinócitos , Reepitelização , Transdução de Sinais , Cicatrização , Animais , Humanos , Suínos , Colágeno/metabolismo , Colágeno/farmacologia , Queratinócitos/metabolismo , Reepitelização/efeitos dos fármacos , Fibroblastos/metabolismo , Cicatrização/efeitos dos fármacos , Cicatrização/fisiologia , Integrina alfa2beta1/metabolismo , Proliferação de Células , Células Cultivadas , Movimento Celular , Pele/lesões , Pele/metabolismo , Peptídeos/farmacologia
3.
Br J Dermatol ; 190(4): 549-558, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38006317

RESUMO

BACKGROUND: Combined expression of the autophagy-regulatory protein AMBRA1 (activating molecule in Beclin1-regulated autophagy) and the terminal differentiation marker loricrin in the peritumoral epidermis of stage I melanomas can identify tumour subsets at low risk of -metastasis. OBJECTIVES: To validate the combined expression of peritumoral AMBRA1 and loricrin (AMBLor) as a prognostic biomarker able to identify both stage I and II melanomas at low risk of tumour recurrence. METHODS: Automated immunohistochemistry was used to analyse peritumoral AMBRA1 and loricrin expression in geographically distinct discovery (n = 540) and validation (n = 300) cohorts of nonulcerated American Joint Committee on Cancer (AJCC) stage I and II melanomas. AMBLor status was correlated with clinical outcomes in the discovery and validation cohorts separately and combined. RESULTS: Analysis of AMBLor in the discovery cohort revealed a recurrence-free survival (RFS) rate of 95.5% in the AMBLor low-risk group vs. 81.7% in the AMBLor at-risk group (multivariate log-rank, P < 0.001) and a negative predictive value (NPV) of 96.0%. In the validation cohort, AMBLor analysis revealed a RFS rate of 97.6% in the AMBLor low-risk group vs. 78.3% in the at-risk group (multivariate log-rank, P < 0.001) and a NPV of 97.6%. In a multivariate model considering AMBLor, Breslow thickness, age and sex, analysis of the combined discovery and validation cohorts showed that the estimated effect of AMBLor was statistically significant, with a hazard ratio of 3.469 (95% confidence interval 1.403-8.580, P = 0.007) and an overall NPV of 96.5%. CONCLUSIONS: These data provide further evidence validating AMBLor as a prognostic biomarker to identify nonulcerated AJCC stage I and II melanoma tumours at low risk of disease recurrence.


Assuntos
Melanoma , Proteínas de Membrana , Neoplasias Cutâneas , Humanos , Estados Unidos , Melanoma/patologia , Prognóstico , Recidiva Local de Neoplasia/patologia , Epiderme/metabolismo , Biomarcadores , Estadiamento de Neoplasias , Proteínas Adaptadoras de Transdução de Sinal/metabolismo
4.
Proc Natl Acad Sci U S A ; 120(18): e2216587120, 2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-37098069

RESUMO

Innate lymphoid cells (ILCs) play a key role in tissue-mediated immunity and can be controlled by coreceptor signaling. Here, we define a subset of ILCs that are Tbet+NK1.1- and are present within the tumor microenvironment (TME). We show programmed death-1 receptor (PD-1) expression on ILCs within TME is found in Tbet+NK1.1- ILCs. PD-1 significantly controlled the proliferation and function of Tbet+NK1.1- ILCs in multiple murine and human tumors. We found tumor-derived lactate enhanced PD-1 expression on Tbet+NK1.1- ILCs within the TME, which resulted in dampened the mammalian target of rapamycin (mTOR) signaling along with increased fatty acid uptake. In line with these metabolic changes, PD-1-deficient Tbet+NK1.1- ILCs expressed significantly increased IFNγ and granzyme B and K. Furthermore, PD-1-deficient Tbet+NK1.1- ILCs contributed toward diminished tumor growth in an experimental murine model of melanoma. These data demonstrate that PD-1 can regulate antitumor responses of Tbet+NK1.1- ILCs within the TME.


Assuntos
Linfócitos , Neoplasias , Camundongos , Animais , Humanos , Imunidade Inata , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Microambiente Tumoral , Neoplasias/metabolismo , Apoptose , Mamíferos/metabolismo
5.
PLoS One ; 17(4): e0265048, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35377887

RESUMO

OBJECTIVES: Cutaneous melanoma rates are steadily increasing. Up to 20% of patients diagnosed with AJCC Stage I/II melanomas will develop metastatic disease. To date there are no consistently reliable means to accurately identify truly high versus low-risk patient subpopulations. There is hence an urgent need for more accurate prediction of prognosis to determine appropriate clinical management. Validation of a novel prognostic test based on the immunohistochemical expression of two protein biomarkers in the epidermal microenvironment of primary melanomas was undertaken; loss of these biomarkers had previously been shown to be associated with a higher risk of recurrence or metastasis. A parallel qualitative study exploring secondary care health professional and patient views of the test was undertaken and this paper reports the perceived barriers and enablers to its implementation into the melanoma care pathway. METHODS: Qualitative methods were employed drawing upon the Theoretical Domains Framework (TDF) in the exploration and analysis. An inductive-deductive analysis was performed, with all data coded using a thematic then TDF framework. FINDINGS: 20 dermatologists, plastic surgeons, cancer nurse specialists, oncologists and histopathologists participated. Nine TDF domains were relevant to all health professional groups and the 'Skills' and 'Beliefs about Capabilities' domains were relevant only to histopathologists. 'Optimism' and 'Beliefs about consequences' were strong enablers particularly for clinicians. 'Environmental context and resources' (impact on pathology services) and 'Knowledge' (the need for robust evidence about the test reliability) were the main perceived barriers. 19 patients and one carer were interviewed. For the patients eight domains were relevant. ('Knowledge', 'Emotions', 'Beliefs about consequences', 'Social Role and identity', 'Behavioural regulation', 'Memory, attention and decision processes', 'Reinforcement' and 'Skills'). The consequences of the implementation of the test were reassurance about future risk, changes to the follow-up pathway on which there were mixed views, and the need to ensure they maintained self-surveillance (Beliefs about consequences). The test was acceptable to all patient interviewees but the resultant changes to management would need to be supported by mechanisms for fast-track back into the clinic, further information on self-surveillance and clear management plans at the time the result is conveyed (Behavioural regulation). CONCLUSIONS: Health professionals and patients perceived positive consequences-for patients and for health services-of adopting the test. However, its implementation would require exploration of the resource implications for pathology services, psychological support for patients with a high-risk test result and mechanisms to reassure and support patients should the test lead to reduced frequency or duration of follow-up. Exploring implementation at an early stage with health professionals presented challenges related to the provision of specific details of the test and its validation.


Assuntos
Melanoma , Neoplasias Cutâneas , Humanos , Melanoma/diagnóstico , Papel Profissional , Prognóstico , Pesquisa Qualitativa , Reprodutibilidade dos Testes , Neoplasias Cutâneas/diagnóstico , Microambiente Tumoral , Melanoma Maligno Cutâneo
6.
Autophagy ; 18(4): 799-815, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34338148

RESUMO

Macroautophagy/autophagy is critical for the regulation of pancreatic ß-cell mass and its deregulation has been implicated in the pathogenesis of type 2 diabetes (T2D). We have previously shown that treatment of pancreatic ß-cells with the GLP1R (glucagon like peptide 1 receptor) agonist exendin-4 stimulates autophagic flux in a setting of chronic nutrient excess. The aim of this study was to identify the underlying pathways contributing to enhanced autophagic flux.Pancreatic ß-cells (INS-1E),mouse and human islets were treated with glucolipotoxic stress (0.5 mM palmitate and 25 mM glucose) in the presence of exendin-4. Consistent with our previous work, exendin-4 stimulated autophagic flux. Using chemical inhibitors and siRNA knockdown, we identified RAPGEF4/EPAC2 (Rap guanine nucleotide exchange factor 4) and downstream calcium signaling to be essential for regulation of autophagic flux by exendin-4. This pathway was independent of AMPK and MTOR signaling. Further analysis identified PPP3/calcineurin and its downstream regulator TFEB (transcription factor EB) as key proteins mediating exendin-4 induced autophagy. Importantly, inhibition of this pathway prevented exendin-4-mediated cell survival and overexpression of TFEB mimicked the cell protective effects of exendin-4 in INS-1E and human islets. Moreover, treatment of db/db mice with exendin-4 for 21 days increased the expression of lysosomal markers within the pancreatic islets. Collectively our data identify the RAPGEF4/EPAC2-calcium-PPP3/calcineurin-TFEB axis as a key mediator of autophagic flux, lysosomal function and cell survival in pancreatic ß-cells. Pharmacological modulation of this axis may offer a novel therapeutic target for the treatment of T2D.Abbreviations: AKT1/protein kinase B: AKT serine/threonine kinase 1; AMPK: 5' AMP-activated protein kinase; CAMKK: calcium/calmodulin-dependent protein kinase kinase; cAMP: cyclic adenosine monophosphate; CASP3: caspase 3; CREB: cAMP response element-binding protein; CTSD: cathepsin D; Ex4: exendin-4(1-39); GLP-1: glucagon like peptide 1; GLP1R: glucagon like peptide 1 receptor; GLT: glucolipotoxicity; INS: insulin; MTOR: mechanistic target of rapamycin kinase; NFAT: nuclear factor of activated T-cells; PPP3/calcineurin: protein phosphatase 3; PRKA/PKA: protein kinase cAMP activated; RAPGEF3/EPAC1: Rap guanine nucleotide exchange factor 3; RAPGEF4/EPAC2: Rap guanine nucleotide exchange factor 4; SQSTM1/p62: sequestosome 1; T2D: type 2 diabetes; TFEB: transcription factor EB.


Assuntos
Calcineurina , Diabetes Mellitus Tipo 2 , Proteínas Quinases Ativadas por AMP , Animais , Autofagia , Calcineurina/metabolismo , Cálcio/metabolismo , Exenatida/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1 , Fatores de Troca do Nucleotídeo Guanina , Camundongos , Serina-Treonina Quinases TOR/metabolismo
7.
Diabetes ; 66(5): 1272-1285, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28232493

RESUMO

Studies in animal models of type 2 diabetes have shown that glucagon-like peptide 1 (GLP-1) receptor agonists prevent ß-cell loss. Whether GLP-1 mediates ß-cell survival via the key lysosomal-mediated process of autophagy is unknown. In this study, we report that treatment of INS-1E ß-cells and primary islets with glucolipotoxicity (0.5 mmol/L palmitate and 25 mmol/L glucose) increases LC3 II, a marker of autophagy. Further analysis indicates a blockage in autophagic flux associated with lysosomal dysfunction. Accumulation of defective lysosomes leads to lysosomal membrane permeabilization and release of cathepsin D, which contributes to cell death. Our data further demonstrated defects in autophagic flux and lysosomal staining in human samples of type 2 diabetes. Cotreatment with the GLP-1 receptor agonist exendin-4 reversed the lysosomal dysfunction, relieving the impairment in autophagic flux and further stimulated autophagy. Small interfering RNA knockdown showed the restoration of autophagic flux is also essential for the protective effects of exendin-4. Collectively, our data highlight lysosomal dysfunction as a critical mediator of ß-cell loss and shows that exendin-4 improves cell survival via restoration of lysosomal function and autophagic flux. Modulation of autophagy/lysosomal homeostasis may thus define a novel therapeutic strategy for type 2 diabetes, with the GLP-1 signaling pathway as a potential focus.


Assuntos
Autofagia/efeitos dos fármacos , Diabetes Mellitus Tipo 2/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Glucose/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Palmitatos/farmacologia , Adulto , Animais , Apoptose , Western Blotting , Estudos de Casos e Controles , Catepsina D/efeitos dos fármacos , Catepsina D/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Exenatida , Humanos , Imuno-Histoquímica , Incretinas/farmacologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Lisossomos/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/metabolismo , Pessoa de Meia-Idade , Peptídeos/farmacologia , RNA Interferente Pequeno , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Peçonhas/farmacologia
8.
Front Oncol ; 6: 236, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27882308

RESUMO

Prognosis and survival for malignant melanoma is highly dependent on early diagnosis and treatment. While the American Joint Committee on Cancer (AJCC) criterion provides a means of staging melanomas and guiding treatment approaches, it is unable to identify the risk of disease progression of early stage tumors or provide reliable stratification for novel adjuvant therapies. The demand for credible prognostic/companion biomarkers able to identify high-risk melanoma subgroups as well as guide more effective personalized/precision-based therapy is therefore of paramount importance. Autophagy, the principle lysosomal-mediated process for the degradation/recycling of cellular debris, is a hot topic in cancer medicine, and observations of its deregulation in melanoma have brought its potential as a prognostic biomarker to the forefront of current research. Key regulatory proteins, including Atg8/microtubule-associated light chain 3 (LC3) and BECN1 (Beclin 1), have been proposed as potential prognostic biomarkers. However, given the dynamic nature of autophagy, their expression in vitro does not translate to their use as a prognostic biomarker for melanoma in vivo. We have recently identified the expression levels of Sequestosome1/SQSTM1 (p62) and activating molecule in Beclin 1-regulated autophagy protein 1 (AMBRA1) as novel independent prognostic biomarkers for early stage melanomas. While increasing followed by subsequent decreasing levels of p62 expression reflects the paradoxical role of autophagy in melanoma, expression levels additionally define a novel prognostic biomarker for AJCC stage II tumors. Conversely, loss of AMBRA1 in the epidermis overlying primary melanomas defines a novel prognostic biomarker for AJCC stage I tumors. Collectively, the definition of AMBRA1 and p62 as prognostic biomarkers for early stage melanomas provides novel and accurate means through which to identify tumors at risk of disease progression, facilitating earlier patient therapeutic intervention and stratification tools for novel personalized therapeutic approaches to improve clinical outcome.

9.
Mol Cancer Ther ; 14(11): 2665-73, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26330548

RESUMO

Metastatic melanoma remains incurable, emphasizing the acute need for improved research models to investigate the underlying biologic mechanisms mediating tumor invasion and metastasis, and to develop more effective targeted therapies to improve clinical outcome. Available animal models of melanoma do not accurately reflect human disease and current in vitro human skin equivalent models incorporating melanoma cells are not fully representative of the human skin microenvironment. We have developed a robust and reproducible, fully humanized three-dimensional (3D) skin equivalent comprising a stratified, terminally differentiated epidermis and a dermal compartment consisting of fibroblast-generated extracellular matrix. Melanoma cells incorporated into the epidermis were able to invade through the basement membrane and into the dermis, mirroring early tumor invasion in vivo. Comparison of our novel 3D melanoma skin equivalent with melanoma in situ and metastatic melanoma indicates that this model accurately recreates features of disease pathology, making it a physiologically representative model of early radial and vertical growth-phase melanoma invasion.


Assuntos
Melanoma/patologia , Neoplasias Cutâneas/patologia , Pele Artificial , Pele/patologia , Células 3T3 , Animais , Biomarcadores Tumorais/metabolismo , Técnicas de Cultura de Células/métodos , Linhagem Celular Tumoral , Células Cultivadas , Derme/metabolismo , Derme/patologia , Epiderme/metabolismo , Epiderme/patologia , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Antígeno MART-1/metabolismo , Melanoma/metabolismo , Camundongos , Microscopia Eletrônica de Varredura , Microscopia de Fluorescência , Modelos Biológicos , Invasividade Neoplásica , Metástase Neoplásica , Pele/metabolismo , Pele/ultraestrutura , Neoplasias Cutâneas/metabolismo , Microambiente Tumoral
10.
J Invest Dermatol ; 135(6): 1629-1637, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25674907

RESUMO

Although the global incidence of cutaneous melanoma is increasing, survival rates for patients with metastatic disease remain <10%. Novel treatment strategies are therefore urgently required, particularly for patients bearing BRAF/NRAS wild-type tumors. Targeting autophagy is a means to promote cancer cell death in chemotherapy-resistant tumors, and the aim of this study was to test the hypothesis that cannabinoids promote autophagy-dependent apoptosis in melanoma. Treatment with Δ(9)-Tetrahydrocannabinol (THC) resulted in the activation of autophagy, loss of cell viability, and activation of apoptosis, whereas cotreatment with chloroquine or knockdown of Atg7, but not Beclin-1 or Ambra1, prevented THC-induced autophagy and cell death in vitro. Administration of Sativex-like (a laboratory preparation comprising equal amounts of THC and cannabidiol (CBD)) to mice bearing BRAF wild-type melanoma xenografts substantially inhibited melanoma viability, proliferation, and tumor growth paralleled by an increase in autophagy and apoptosis compared with standard single-agent temozolomide. Collectively, our findings suggest that THC activates noncanonical autophagy-mediated apoptosis of melanoma cells, suggesting that cytotoxic autophagy induction with Sativex warrants clinical evaluation for metastatic disease.


Assuntos
Autofagia , Canabinoides/química , Melanoma/patologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Canabidiol , Canabinol/química , Morte Celular , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Dacarbazina/análogos & derivados , Dacarbazina/química , Dronabinol/química , Combinação de Medicamentos , Humanos , Masculino , Melanoma/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Microscopia Confocal , Metástase Neoplásica , Transplante de Neoplasias , Neoplasias/metabolismo , Extratos Vegetais/química , Proteínas Proto-Oncogênicas B-raf/metabolismo , Neoplasias Cutâneas/metabolismo , Temozolomida , Proteínas ras/metabolismo
11.
Mol Cell Oncol ; 2(3): e995016, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-27308477

RESUMO

Autophagy and endoplasmic reticulum (ER) stress are involved in the development, progression, and chemoresistance of melanoma. We recently reported that oncogenic serine/threonine-protein kinase BRAF induces chronic ER stress, hence increasing baseline autophagy and promoting chemoresistance. The attenuation of ER stress restores basal autophagic activity and resensitizes melanoma cells to apoptosis.

12.
J Cell Biochem ; 116(3): 438-49, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25336069

RESUMO

Glucose-regulated protein 78 (GRP78) is a stress sensor which interacts with unfolded protein response (UPR) activators in the endoplasmic reticulum (ER). The aim of this study was to test the hypothesis that GRP78 has distinct functional roles in mediating the effects of ER stress in neuroblastoma compared to other neuroectodermal cancer types. GRP78 was knocked down or overexpressed in neuroectodermal tumor cell lines. Protein and transcript expression were measured using Western blotting, confocal microscopy, and real-time polymerase chain reaction; cell stress was assessed by measurement of oxidative stress and accumulation of ubiquitinated proteins and cell response by measurement of apoptosis and cell viability. Neuroblastoma cells were more sensitive to ER stress than melanoma and glioblastoma cells. GRP78 knockdown increased stress sensitivity of melanoma and glioblastoma cells, but not neuroblastoma cells. Over-expression of GRP78 decreased the stress sensitivity of melanoma and glioblastoma cells but, in contrast, increased the stress sensitivity of neuroblastoma cells by activation of caspase-3-independent cell death and substantially increased the expression of UPR activators, particularly inositol-requiring element 1 (IRE1). The results from this study suggest that cell-type specific differences in the relationships between GRP78 and the UPR activators, particularly IRE1, may determine differential sensitivity to ER stress.


Assuntos
Proteínas de Choque Térmico/metabolismo , Placa Neural/citologia , Placa Neural/metabolismo , Estresse Fisiológico , Biomarcadores/metabolismo , Ácidos Borônicos/farmacologia , Bortezomib , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Forma Celular/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fenretinida/farmacologia , Técnicas de Silenciamento de Genes , Humanos , Pirazinas/farmacologia , RNA Interferente Pequeno/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Resposta a Proteínas não Dobradas/efeitos dos fármacos
14.
Healthcare (Basel) ; 2(1): 60-73, 2014 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-27429260

RESUMO

In an era of personalized medicine, disease specific biomarkers play an increasing role in the stratification of high-risk patient groups. Cutaneous malignant melanoma is the most deadly form of skin cancer with an ever-increasing global incidence, especially in patients under 35-years of age. Despite the excellent prognosis for patients diagnosed with early stage disease, metastatic disease still carries significant overall mortality. Biomarkers aim not only to identify high-risk patients, but also to provide potential therapeutic targets for differing patient subgroups. Furthermore, accessibility to tissue samples from a range of disease stages in malignant melanoma, unlike most other solid tissue tumours, provides the unique opportunity to explore the biology of tumour progression that may be relevant in the biology of cancer as a whole. Over the past decade, there have been major advances in targeted therapies, providing new avenues and hope to patients with this devastating disease. This review will focus on most up to date histological, serological and molecular biomarkers in malignant melanoma.

15.
Melanoma Manag ; 1(2): 127-137, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30190818

RESUMO

Melanoma cells employ several survival strategies, including induction of the unfolded protein response, which mediates resistance to endoplasmic reticulum (ER) stress-induced apoptosis. Activation of oncogenes specifically suppresses ER stress-induced apoptosis, while upregulation of ER chaperone proteins and antiapoptotic BCL-2 family members increases the protein folding capacity of the cell and the threshold for the induction of ER stress-induced apoptosis, respectively. Modulation of unfolded protein response signaling, inhibition of the protein folding machinery and/or active induction of ER stress may thus represent potential strategies for the therapeutic management of melanoma. To this aim, the present article focuses on the current understanding of how melanoma cells avoid or overcome ER stress-induced apoptosis, as well as therapeutic strategies through which to harness ER stress for therapeutic benefit.

16.
Exp Dermatol ; 22(11): 767-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24118207

RESUMO

The Bcl-2 family member Mcl-1 is essential for melanoma survival; however, the influence of oncogenic BRAF signalling remains elusive. In this study, Mcl-1 splice variant expression was determined in a panel of melanoma cell lines in relation to BRAF mutational status. Mcl-1L mRNA expression was increased in melanoma cells compared with primary melanocytes with significantly increased mRNA and protein expression observed in BRAF(V600E) mutant melanoma cells. Although no change in Mcl-1S mRNA was observed, Mcl-1S protein expression also increased in BRAF mutant melanoma cells. Additionally, while over-expression of mutant BRAF(V600E) increased both Mcl-1L and Mcl-1S expression, inhibition of hyperactive BRAF signalling resulted in decreased Mcl-1L expression. These studies suggest that the regulation of Mcl-1 expression by BRAF signalling is increased by oncogenic activation of BRAF, revealing a mechanism of apoptotic resistance which may be overcome by the use of more specifically targeted Mcl-1 inhibitors.


Assuntos
Regulação Neoplásica da Expressão Gênica , Melanoma/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/metabolismo , Apoptose , Linhagem Celular Tumoral , Humanos , Melanócitos/metabolismo , Melanoma/genética , Mutação , Neoplasias Cutâneas/genética , Melanoma Maligno Cutâneo
17.
Oncol Rep ; 27(1): 293-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21964808

RESUMO

Despite the successful introduction of 13-cis retinoic acid (13cisRA) therapy for the treatment of neuroblastoma, approximately 50% patients do not respond or experience relapse. A retinoid analogue, fenretinide [N-(4-hydroxyphenyl) retinamide; 4-HPR] can induce apoptosis in neuroblastoma cell lines and could have clinical use after therapy with 13cisRA. However, there are important questions concerning potential retinoid drug interactions which need to be addressed. The aim of this study was to investigate the influence of retinoic acid pre-treatment on fenretinide-induced apoptosis and fenretinide metabolism in neuroblastoma cell lines. Apoptosis was measured by flow cytometry of propidium iodide-stained neuroblastoma cells and a live-cell imaging assay. Intracellular fenretinide metabolism was determined by HPLC analysis. Pre-treatment of neuroblastoma cell lines with retinoic acid (RA) resulted in a significant decrease in the apoptotic response to fenretinide in three of the four lines tested. Comparison between responsive and non-responsive cell lines suggested that RA sensitivity was required to promote fenretinide resistance, and that this was mediated by up-regulation of Bcl-2 and the inhibition of pro-apoptotic fenretinide signalling pathways. Induction of the oxidative metabolism of fenretinide after RA pre-treatment did not significantly impact on intracellular parent drug levels and is unlikely to explain the decreased apoptotic response observed. The interaction between RA and fenretinide could have important implications for the scheduling of fenretinide in therapeutic protocols for neuroblastoma.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Fenretinida/farmacologia , Neuroblastoma/metabolismo , Tretinoína/farmacologia , Western Blotting , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Interações Medicamentosas , Fenretinida/metabolismo , Citometria de Fluxo , Humanos , Neuroblastoma/patologia
18.
Clin Cancer Res ; 17(8): 2216-26, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21270111

RESUMO

PURPOSE: Metastatic melanoma is characterized by extremely poor survival rates and hence novel therapies are urgently required. The ability of many anticancer drugs to activate autophagy, a lysosomal-mediated catabolic process which usually promotes cell survival, suggests targeting the autophagy pathway may be a novel means to augment therapy. EXPERIMENTAL DESIGN: Autophagy and apoptosis were assessed in vitro in human melanoma cell lines in response to clinically achievable concentrations of the endoplasmic reticulum (ER) stress-inducing drugs fenretinide or bortezomib, and in vivo using a s.c. xenograft model. RESULTS: Autophagy was activated in response to fenretinide or bortezomib in B-RAF wild-type cells, shown by increased conversion of LC3 to the autophagic vesicle-associated form (LC3-II) and redistribution to autophagosomes and autolysosomes, increased acidic vesicular organelle formation and autophagic vacuolization. In contrast, autophagy was significantly reduced in B-RAF-mutated melanoma cells, an effect attributed partly to oncogenic B-RAF. Rapamycin treatment was unable to stimulate LC3-II accumulation or redistribution in the presence of mutated B-RAF, indicative of de-regulated mTORC1-dependent autophagy. Knockdown of Beclin-1 or ATG7 sensitized B-RAF wild-type cells to fenretinide- or bortezomib-induced cell death, demonstrating a pro-survival function of autophagy. In addition, autophagy was partially reactivated in B-RAF-mutated cells treated with the BH3 mimetic ABT737 in combination with fenretinide or bortezomib, suggesting autophagy resistance is partly mediated by abrogated Beclin-1 function. CONCLUSIONS: Our findings suggest inhibition of autophagy in combination with ER stress-inducing agents may represent a means by which to harness autophagy for the therapeutic benefit of B-RAF wild-type melanoma.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Autofagia/efeitos dos fármacos , Melanoma/tratamento farmacológico , Proteínas Proto-Oncogênicas B-raf/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Compostos de Bifenilo/administração & dosagem , Compostos de Bifenilo/farmacologia , Western Blotting , Ácidos Borônicos/administração & dosagem , Ácidos Borônicos/farmacologia , Bortezomib , Linhagem Celular Tumoral , Retículo Endoplasmático/metabolismo , Feminino , Fenretinida/administração & dosagem , Fenretinida/farmacologia , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Nitrofenóis/administração & dosagem , Nitrofenóis/farmacologia , Piperazinas/administração & dosagem , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas B-raf/genética , Pirazinas/administração & dosagem , Pirazinas/farmacologia , Interferência de RNA , Sulfonamidas/administração & dosagem , Sulfonamidas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Pigment Cell Melanoma Res ; 23(5): 675-82, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20546536

RESUMO

Targeting endoplasmic reticulum stress-induced apoptosis may offer an alternative therapeutic strategy for metastatic melanoma. Fenretinide and bortezomib induce apoptosis of melanoma cells but their efficacy may be hindered by the unfolded protein response, which promotes survival by ameliorating endoplasmic reticulum stress. The aim of this study was to test the hypothesis that inhibition of GRP78, a vital unfolded protein response mediator, increases cell death in combination with endoplasmic reticulum stress-inducing agents. Down-regulation of GRP78 by small-interfering RNA increased fenretinide- or bortezomib-induced apoptosis. Treatment of cells with a GRP78-specific subtilase toxin produced a synergistic enhancement with fenretinide or bortezomib. These data suggest that combining endoplasmic reticulum stress-inducing agents with strategies to down-regulate GRP78, or other components of the unfolded protein response, may represent a novel therapeutic approach for metastatic melanoma.


Assuntos
Antineoplásicos , Morte Celular/efeitos dos fármacos , Proteínas de Choque Térmico/antagonistas & inibidores , Melanoma/metabolismo , Melanoma/patologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Ácidos Borônicos/farmacologia , Ácidos Borônicos/uso terapêutico , Bortezomib , Linhagem Celular Tumoral , Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático , Fenretinida/farmacologia , Fenretinida/uso terapêutico , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Melanoma/tratamento farmacológico , Pirazinas/farmacologia , Pirazinas/uso terapêutico , RNA Interferente Pequeno/metabolismo , Resposta a Proteínas não Dobradas
20.
J Invest Dermatol ; 130(9): 2250-8, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20520630

RESUMO

Melanoma remains notoriously resistant to current chemotherapeutics, leaving an acute need for novel therapeutic approaches. The aim of this study was to determine the prognostic and therapeutic significance of X-linked inhibitor of apoptosis protein (XIAP) in melanoma through correlation of XIAP expression with disease stage, RAS/RAF mutational status, clinical outcome, and susceptibility to endoplasmic reticulum (ER) stress-induced cell death. XIAP expression and N-RAS/B-RAF mutational status were retrospectively determined in a cohort of 55 primary cutaneous melanocytic lesions selected and grouped according to the American Joint Committee on Cancer staging system. Short hairpin RNA interference of XIAP was used to analyze the effect of XIAP expression on ER stress-induced apoptosis in response to fenretinide or bortezomib in vitro. The results showed that XIAP positivity increased with progressive disease stage, although there was no significant correlation between XIAP positivity and combined N-RAS/B-RAF mutational status or clinical outcome. However, XIAP knockdown significantly increased ER stress-induced apoptosis of melanoma cells in a caspase-dependant manner. The correlation of XIAP expression with disease stage, as well as data showing that XIAP knockdown significantly increases fenretinide and bortezomib-induced apoptosis of metastatic melanoma cells, suggests that XIAP may prove to be an effective therapeutic target for melanoma therapy.


Assuntos
Antineoplásicos/farmacologia , Ácidos Borônicos/farmacologia , Fenretinida/farmacologia , Melanoma/tratamento farmacológico , Pirazinas/farmacologia , Neoplasias Cutâneas/tratamento farmacológico , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Bortezomib , Resistencia a Medicamentos Antineoplásicos , Retículo Endoplasmático/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica , Genes ras/fisiologia , Humanos , Técnicas In Vitro , Masculino , Melanoma/metabolismo , Melanoma/patologia , Pessoa de Meia-Idade , Mutação/genética , Nevo Pigmentado/tratamento farmacológico , Nevo Pigmentado/metabolismo , Nevo Pigmentado/patologia , Proteínas Proto-Oncogênicas B-raf/genética , RNA Interferente Pequeno , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Estresse Fisiológico/fisiologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA