Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Cancer Gene Ther ; 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38360887

RESUMO

Recent pan-cancer genomic analyses have identified numerous oncogenic driver mutations that occur in a cell-type and tissue-specific distribution. For example, oncogenic mutations in Braf and Nras genes arise predominantly in melanocytic neoplasms of the epidermis, while oncogenic mutations in Gnaq/11 genes arise mostly in melanocytic lesions of the dermis or the uvea. The mechanisms promoting cell-type and tissue-specific oncogenic events currently remain poorly understood. Here, we report that Gnaq/11 hotspot mutations occur as early oncogenic drivers during the evolution of primary melanomas in Hgf-Cdk4 mice. Additional single base substitutions in the Trp53 gene and structural chromosomal aberrations favoring amplifications of the chromosomal region containing the Met receptor gene accumulate during serial tumor transplantation and in cell lines established in vitro. Mechanistically, we found that the GnaqQ209L mutation transactivates the Met receptor. Overexpression of oncogenic GnaqQ209L in the immortalized melanocyte cell line promoted in vivo growth that was enhanced by transgenic Hgf expression in the tumor microenvironment. This cross-signaling mechanism explains the selection of oncogenic Gnaq/11 in primary Hgf-Cdk4 melanomas and provides an example of how oncogenic driver mutations, intracellular signaling cascades, and microenvironmental cues cooperate to drive cancer development in a tissue-specific fashion.

2.
Nature ; 618(7967): 1033-1040, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37316667

RESUMO

Most clinically applied cancer immunotherapies rely on the ability of CD8+ cytolytic T cells to directly recognize and kill tumour cells1-3. These strategies are limited by the emergence of major histocompatibility complex (MHC)-deficient tumour cells and the formation of an immunosuppressive tumour microenvironment4-6. The ability of CD4+ effector cells to contribute to antitumour immunity independently of CD8+ T cells is increasingly recognized, but strategies to unleash their full potential remain to be identified7-10. Here, we describe a mechanism whereby a small number of CD4+ T cells is sufficient to eradicate MHC-deficient tumours that escape direct CD8+ T cell targeting. The CD4+ effector T cells preferentially cluster at tumour invasive margins where they interact with MHC-II+CD11c+ antigen-presenting cells. We show that T helper type 1 cell-directed CD4+ T cells and innate immune stimulation reprogramme the tumour-associated myeloid cell network towards interferon-activated antigen-presenting and iNOS-expressing tumouricidal effector phenotypes. Together, CD4+ T cells and tumouricidal myeloid cells orchestrate the induction of remote inflammatory cell death that indirectly eradicates interferon-unresponsive and MHC-deficient tumours. These results warrant the clinical exploitation of this ability of CD4+ T cells and innate immune stimulators in a strategy to complement the direct cytolytic activity of CD8+ T cells and natural killer cells and advance cancer immunotherapies.


Assuntos
Linfócitos T CD4-Positivos , Morte Celular , Imunoterapia , Inflamação , Neoplasias , Microambiente Tumoral , Humanos , Células Apresentadoras de Antígenos/imunologia , Antígeno CD11c/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Morte Celular/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Imunidade Inata , Inflamação/imunologia , Interferons/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Microambiente Tumoral/imunologia , Imunoterapia/métodos , Células Matadoras Naturais/imunologia , Células Mieloides/imunologia , Células Th1/citologia , Células Th1/imunologia
4.
Dermatologie (Heidelb) ; 73(12): 937-942, 2022 Dec.
Artigo em Alemão | MEDLINE | ID: mdl-36350371

RESUMO

Neurocognitive impairments of memory, speech, and attention can occur in cancer patients as a direct result of the cancer but also in the context of therapy. With the development of modern immunotherapies and their use in combination with surgery and radiation therapy, the number of long-term survivors has significantly increased. As a result, detrimental effects on brain function and structure in cancer patients not only during treatment but also after completion of therapy have become a key issue in clinical oncology. Early diagnosis and treatment of neurocognitive disorders is of great importance for quality of life, therapy adherence, and overall survival of the affected patients. In this review, we discuss the underlying mechanisms with a special focus on metastatic melanoma. Furthermore, practice-relevant diagnostics, prophylaxis, and intervention options are discussed.


Assuntos
Disfunção Cognitiva , Melanoma , Segunda Neoplasia Primária , Humanos , Qualidade de Vida , Melanoma/complicações , Imunoterapia/efeitos adversos , Sobreviventes/psicologia , Disfunção Cognitiva/etiologia
5.
J Biophotonics ; 15(10): e202200129, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35802400

RESUMO

We introduce a new single-head multimodal optical system that integrates optical coherence tomography (OCT), 18 MHz ultrasound (US) tomography and Raman spectroscopy (RS), allowing for fast (<2 min) and noninvasive skin cancer diagnostics and lesion depth measurement. The OCT can deliver structural and depth information of smaller skin lesions (<1 mm), while the US allows to measure the penetration depth of thicker lesions (≥4 mm), and the RS analyzes the chemical composition from a small chosen spot (≤300 µm) that can be used to distinguish between benign and malignant melanoma. The RS and OCT utilize the same scanning and optical setup, allowing for co-localized measurements. The US on the other side is integrated with an acoustical reflector, which enables B-mode measurements on the same position as OCT and RS. The US B-mode scans can be translated across the sample by laterally moving the US transducer, which is made possible by the developed adapter with a flexible membrane. We present the results on custom-made liquid and agar phantoms that show the resolution and depth capabilities of the setup, as well as preliminary ex vivo measurements on mouse models with ∼4.3 mm thick melanoma.


Assuntos
Melanoma , Tomografia de Coerência Óptica , Ágar , Animais , Biópsia , Melanoma/diagnóstico por imagem , Melanoma/patologia , Camundongos , Análise Espectral Raman/métodos , Tomografia de Coerência Óptica/métodos
6.
Int J Cancer ; 150(1): 142-151, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34528710

RESUMO

Most melanoma-associated deaths result from the early development of metastasis. Toll-like receptor 4 (TLR4) expression on nontumor cells is well known to contribute to tumor development and metastatic progression. The role of TLR4 expression on tumor cells however is less well understood. Here we describe TLR4 as a driver of tumor progression and metastatic spread of melanoma cells by employing a transplantable mouse melanoma model. HCmel12 melanoma cells lacking functional TLR4 showed increased sensitivity to tumor necrosis factor α induced cell killing in vitro compared to cells with intact TLR4. Interestingly, TLR4 knockout melanoma cells also showed impaired migratory capacity in vitro and a significantly reduced ability to metastasize to the lungs after subcutaneous transplantation in vivo. Finally, we demonstrate that activation of TLR4 also promotes migration in a subset of human melanoma cell lines. Our work describes TLR4 as an important mediator of melanoma migration and metastasis and provides a rationale for therapeutic inhibition of TLR4 in melanoma.


Assuntos
Movimento Celular , Neoplasias Pulmonares/secundário , Melanoma/patologia , Receptor 4 Toll-Like/metabolismo , Animais , Apoptose , Sistemas CRISPR-Cas , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Melanoma/genética , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/genética , Células Tumorais Cultivadas
7.
Pharmaceutics ; 15(1)2022 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-36678686

RESUMO

G protein-coupled receptors (GPCRs) transfer extracellular signals across cell membranes by activating intracellular heterotrimeric G proteins. Several studies suggested G proteins as novel drug targets for the treatment of complex diseases, e.g., asthma and cancer. Recently, we developed specific radiotracers, [³H]PSB-15900-FR and [³H]PSB-16254-YM, for the Gαq family of G proteins by tritiation of the macrocyclic natural products FR900359 (FR) and YM-254890 (YM). In the present study, we utilized these potent radioligands to perform autoradiography studies in tissues of healthy mice, mouse models of disease, and human tissues. Specific binding was high, while non-specific binding was extraordinarily low, giving nearly identical results for both radioligands. High expression levels of Gαq proteins were detected in healthy mouse organs showing the following rank order of potency: kidney > liver > brain > pancreas > lung > spleen, while expression in the heart was low. Organ sub-structures, e.g., of mouse brain and lung, were clearly distinguishable. Whereas an acute asthma model in mice did not result in altered Gαq protein expressions as compared to control animals, a cutaneous melanoma model displayed significantly increased expression in comparison to healthy skin. These results suggest the future development of Gαq-protein-binding radio-tracers as novel diagnostics.

9.
Clin Transl Immunology ; 10(4): e1276, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33968406

RESUMO

OBJECTIVES: Type I interferons are evolutionally conserved cytokines, with broad antimicrobial and immunoregulatory functions. Despite well-characterised role in spontaneous cancer immunosurveillance, the function of type I IFNs in cancer immunotherapy remains incompletely understood. METHODS: We utilised genetic mouse models to explore the role of the type I IFN system in CD8+ T-cell immunotherapy targeting the melanocytic lineage antigen gp100. RESULTS: The therapeutic efficacy of adoptively transferred T cells was found to depend on a functional type I IFN system in myeloid immune cells. Compromised type I IFN signalling in myeloid immune cells did not prevent expansion, tumor infiltration or effector function of melanoma-specific Pmel-1 CD8+ T cells. However, melanomas growing in globally (Ifnar1-/-) or conditionally (Ifnar1ΔLysM) type I IFN system-deficient mice displayed increased myeloid infiltration, hypoxia and melanoma cell dedifferentiation. Mechanistically, hypoxia was found to induce dedifferentiation and loss of the gp100 target antigen in melanoma cells and type I IFN could directly inhibit the inflammatory activation of myeloid cells. Unexpectedly, the immunotherapy induced significant reduction in tumor blood vessel density and whereas host type I IFN system was not required for the vasculosculpting, it promoted vessel permeability. CONCLUSION: Our results substantiate a complex and plastic phenotypic interconnection between melanoma and myeloid cells in the context of T-cell immunotherapy. Type I IFN signalling in myeloid cells was identified as a key regulator of the balance between antitumor immunity and disease-promoting inflammation, thus supporting the development of novel combinatorial immunotherapies targeting this immune cell compartment.

10.
J Dtsch Dermatol Ges ; 18(11): 1245-1248, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32954611

RESUMO

Uveal melanoma is the most common form of eye cancer and has a poor prognosis. Although the primary tumor in most cases is treated effectively by local surgery or radiotherapy, over 50 % of patients develop systemic metastasis, especially in the liver. In contrast to cutaneous melanoma, there is no standard-of-care treatment for metastasized uveal melanoma. Recently, oncogenic driver mutations in GNAQ or GNA11 were identified in about 85 % of uveal melanomas, which lead to constitutively active signaling in the Gαq/11 pathway and its downstream effectors. Direct targeting of deregulated Gαq/11 signaling might therefore be a therapeutic option for patients with uveal melanoma. In our study we identified the cyclic depsipeptide FR-900359, which is isolated from the evergreen plant Ardisia crenata as an effective inhibitor of constitutively active Gαq/11 proteins and their downstream targets. Although our data are preliminary, they might contribute to a future treatment option for patients with metastasized uveal melanoma.


Assuntos
Melanoma/genética , Neoplasias Uveais/genética , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Humanos , Mutação , Neoplasias Cutâneas
11.
Int J Cancer ; 147(10): 2902-2913, 2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-32790916

RESUMO

The aryl hydrocarbon receptor (AHR) is a ligand binding-transcription factor of the basic helix-loop-helix family regulating multiple cellular functions such as differentiation, cell cycle, apoptosis, and inflammatory reactions. In neoplastic diseases, the AHR has been described to modulate proliferation and differentiation in dichotomous ways, either inhibiting or augmenting the growth of tumors. The precise role of AHR in melanoma is mostly unknown. Here, we report a functional effect of AHR activation on inflammation-induced melanoma cell dedifferentiation and the development of lung metastases in a mouse model. Via in silico analyses of "The Cancer Genome Atlas" human melanoma cohort, we detected a correlation between AHR expression levels and a dedifferentiated melanoma cell phenotype with an invasive gene signature, which we were able to functionally recapitulate in a panel of human melanoma cell lines. Both human and mouse melanoma cell lines upregulated AHR expression after inflammatory stimulation with tumor necrosis factor-α (TNF-α). Activation of AHR in human and mouse melanoma cell lines with the endogenous ligand formylindolo(3,2-b)carbazole (FICZ) promoted inflammation-induced dedifferentiation in vitro. Importantly, mouse melanoma cells with CRISPR/Cas9-mediated disruption of the AHR gene showed impaired in vivo tumor growth after transplantation in the skin as well as decreased numbers of spontaneous lung metastases. Taken together, our results demonstrate a functional role for AHR expression in melanoma development and metastatic progression. This provides a scientific basis for future experiments that further dissect the underlying molecular mechanisms and assess the potential for AHR inhibition as part of multimodal melanoma treatment strategies.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Melanoma/patologia , Receptores de Hidrocarboneto Arílico/genética , Fator de Necrose Tumoral alfa/farmacologia , Animais , Carbazóis/farmacologia , Desdiferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Melanoma/genética , Camundongos , Transplante de Neoplasias , Regulação para Cima
12.
J Dtsch Dermatol Ges ; 18(8): 815-824, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32717116

RESUMO

COVID-19, caused by the coronavirus SARS-CoV-2, has become pandemic. A further level of complexity opens up as soon as we look at diseases whose pathogenesis and therapy involve different immunological signaling pathways, which are potentially affected by COVID-19. Medical treatments must often be reassessed and questioned in connection with this infection. This article summarizes the current knowledge of COVID-19 in the light of major dermatological and allergological diseases. It identifies medical areas lacking sufficient data and draws conclusions for the management of our patients during the pandemic. We focus on common chronic inflammatory skin diseases with complex immunological pathogenesis: psoriasis, eczema including atopic dermatitis, type I allergies, autoimmune blistering and inflammatory connective tissue diseases, vasculitis, and skin cancers. Since several other inflammatory skin diseases display related or comparable immunological reactions, clustering of the various inflammatory dermatoses into different disease patterns may help with therapeutic decisions. Thus, following these patterns of skin inflammation, our review may supply treatment recommendations and thoughtful considerations for disease management even beyond the most frequent diseases discussed here.


Assuntos
COVID-19/complicações , Dermatopatias/etiologia , COVID-19/imunologia , Humanos , Dermatopatias/patologia
13.
J Dtsch Dermatol Ges ; 18(5): 456-471, 2020 May.
Artigo em Alemão | MEDLINE | ID: mdl-32413226
14.
J Dtsch Dermatol Ges ; 18(5): 456-469, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32311823

RESUMO

Skin lesions associated with internal malignancy may present as cutaneous metastases or as typical lesions occurring in the context of certain cancer-associated genetic syndromes. Paraneoplastic syndromes, on the other hand, are only indirectly associated with an underlying malignancy and are not malignant per se. Historically, a distinction has been made between "obligate" and "facultative" paraneoplastic disorders, depending on the likelihood with which they are potentially associated with malignancy. In addition, there are nonspecific cutaneous manifestations that are only rarely associated with an underlying malignancy. Another possible classification is based on the pathophysiological mechanisms underlying the cutaneous lesions. In everyday practice, it is essential that dermatologists recognize potentially cancer-associated dermatoses, as this will frequently contribute to the initial diagnosis of an underlying neoplasm.


Assuntos
Neoplasias/complicações , Dermatopatias/diagnóstico , Dermatopatias/etiologia , Humanos , Síndromes Paraneoplásicas/diagnóstico , Síndromes Paraneoplásicas/patologia , Neoplasias Cutâneas/secundário
15.
J Invest Dermatol ; 140(7): 1410-1417.e2, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31972251

RESUMO

Oncogenic mutations in the BRAF kinase gene represent the most frequent genomic driver in acquired melanocytic nevi and in cutaneous melanomas. It is currently thought that oncogene-induced senescence and cell cycle arrest limit the ability of oncogenic BRAF to promote melanocyte proliferation in benign nevi. The molecular and cellular mechanisms that allow an oncogenic BRAF mutation to fully transform melanocytes into invasively growing melanoma cells that are able to metastasize systemically are only partially understood. In this study, we show in a genetic mouse model that constitutively enhanced Hgf-Met signaling cooperates with oncogenic BRAF to drive tumor development and metastatic spread. Activation of oncogenic BRAF in mice with transgenic Hgf overexpression and an oncogenic CDK4 germline mutation accelerated and increased the development of primary cutaneous melanomas. Primary melanomas showed considerable phenotypic heterogeneity with frequent signs of dedifferentiation. BRAF activation in Hgf-CDK4 mice also increased the number of lung metastases. Melanoma cells showed a pronounced angiotropic growth pattern both at the invasive front in primary tumors and in metastatic lesions of the lung. Taken together, our work supports the notion that activated Hgf-Met signaling and oncogenic BRAF can cooperate in melanoma pathogenesis.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Neoplasias Pulmonares/metabolismo , Melanoma/metabolismo , Metástase Neoplásica , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Neoplasias Cutâneas/metabolismo , Alelos , Animais , Diferenciação Celular , Quinase 4 Dependente de Ciclina/metabolismo , Modelos Animais de Doenças , Feminino , Neoplasias Pulmonares/secundário , Masculino , Melanoma/patologia , Camundongos , Mutação , Fenótipo , Pigmentação , Transdução de Sinais , Neoplasias Cutâneas/patologia , Transgenes
16.
Sci Signal ; 12(573)2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30890659

RESUMO

Somatic gain-of-function mutations of GNAQ and GNA11, which encode α subunits of heterotrimeric Gαq/11 proteins, occur in about 85% of cases of uveal melanoma (UM), the most common cancer of the adult eye. Molecular therapies to directly target these oncoproteins are lacking, and current treatment options rely on radiation, surgery, or inhibition of effector molecules downstream of these G proteins. A hallmark feature of oncogenic Gαq/11 proteins is their reduced intrinsic rate of hydrolysis of guanosine triphosphate (GTP), which results in their accumulation in the GTP-bound, active state. Here, we report that the cyclic depsipeptide FR900359 (FR) directly interacted with GTPase-deficient Gαq/11 proteins and preferentially inhibited mitogenic ERK signaling rather than canonical phospholipase Cß (PLCß) signaling driven by these oncogenes. Thereby, FR suppressed the proliferation of melanoma cells in culture and inhibited the growth of Gαq-driven UM mouse xenografts in vivo. In contrast, FR did not affect tumor growth when xenografts carried mutated B-RafV600E as the oncogenic driver. Because FR enabled suppression of malignant traits in cancer cells that are driven by activating mutations at codon 209 in Gαq/11 proteins, we envision that similar approaches could be taken to blunt the signaling of non-Gαq/11 G proteins.


Assuntos
Depsipeptídeos/farmacologia , Sistemas de Liberação de Medicamentos , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Subunidades alfa de Proteínas de Ligação ao GTP , Mutação com Ganho de Função , Melanoma , Proteínas de Neoplasias , Neoplasias Uveais , Animais , Linhagem Celular Tumoral , Depsipeptídeos/química , Subunidades alfa de Proteínas de Ligação ao GTP/antagonistas & inibidores , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/antagonistas & inibidores , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Melanoma/tratamento farmacológico , Melanoma/enzimologia , Melanoma/genética , Melanoma/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Uveais/tratamento farmacológico , Neoplasias Uveais/enzimologia , Neoplasias Uveais/genética , Neoplasias Uveais/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Immunity ; 47(4): 789-802.e9, 2017 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-29045907

RESUMO

Inhibitors of the receptor tyrosine kinase c-MET are currently used in the clinic to target oncogenic signaling in tumor cells. We found that concomitant c-MET inhibition promoted adoptive T cell transfer and checkpoint immunotherapies in murine cancer models by increasing effector T cell infiltration in tumors. This therapeutic effect was independent of tumor cell-intrinsic c-MET dependence. Mechanistically, c-MET inhibition impaired the reactive mobilization and recruitment of neutrophils into tumors and draining lymph nodes in response to cytotoxic immunotherapies. In the absence of c-MET inhibition, neutrophils recruited to T cell-inflamed microenvironments rapidly acquired immunosuppressive properties, restraining T cell expansion and effector functions. In cancer patients, high serum levels of the c-MET ligand HGF correlated with increasing neutrophil counts and poor responses to checkpoint blockade therapies. Our findings reveal a role for the HGF/c-MET pathway in neutrophil recruitment and function and suggest that c-MET inhibitor co-treatment may improve responses to cancer immunotherapy in settings beyond c-MET-dependent tumors.


Assuntos
Imunoterapia/métodos , Neoplasias Experimentais/terapia , Neutrófilos/imunologia , Proteínas Proto-Oncogênicas c-met/imunologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Estimativa de Kaplan-Meier , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/metabolismo , Neutrófilos/metabolismo , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
20.
Oncoimmunology ; 5(2): e1078964, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27057457

RESUMO

Increased neutrophil counts both in tumor tissue and peripheral blood correlate with poor clinical outcome in melanoma patients suggesting a pro-tumorigenic role of neutrophils for the pathogenesis of malignant melanoma. Recently, we discovered that neutrophilic skin inflammatory responses induced by UV exposure promote metastatic spread of primary cutaneous melanomas in genetically engineered Hgf-Cdk4(R24C) mice. We hypothesized that other pro-inflammatory stimuli that induce neutrophilic inflammatory responses also promote the development and progression of melanomas. In the current study, we therefore investigated how the most potent and frequently used tumor promoter 12-O-Tetradecanoylphorbol-13-acetate (TPA) affects the development and progression of carcinogen-induced melanomas in Hgf-Cdk4(R24C) mice. Local and systemic neutrophilic inflammatory responses induced by TPA also selectively increase the metastatic spread of melanoma cells to draining lymph nodes and lungs. Using a highly metastatic Hgf-Cdk4(R24C) melanoma skin transplant we could show that TPA enhances systemic spread of melanoma cells which was depended on intact TLR4 signaling in recipient mice and on the presence of neutrophils. Altogether, our experimental results support an important mechanistic role of TLR4-driven neutrophilic inflammation for melanoma progression.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA