Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Immunol ; : e2451020, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39288297

RESUMO

Ultraviolet (UV) irradiation of the skin causes mutations that can promote the development of melanoma and nonmelanoma skin cancer. High-dose UVB exposure triggers a vigorous skin reaction characterized by inflammation resulting in acute sunburn. This response includes the formation of sunburn cells and keratinocytes (KC) undergoing programmed cell death (apoptosis) when repair mechanisms of DNA damage are inadequate. The primary objective of this research was to clarify the involvement of Langerhans cells (LC) in the development of acute sunburn following intense UVB skin irradiation. To address this, we subjected the dorsal skin of mice to a single high-dose UVB exposure and analyzed the immediate immune response occurring within the skin tissue. Acute sunburn triggered an activation of LC, coinciding with a rapid influx of neutrophils that produced TNF-α. Furthermore, our investigation unveiled a marked increase in DNA-damaged KC and the subsequent induction of apoptosis in these cells. Importantly, we demonstrate a crucial link between the inflammatory cascade, the initiation of apoptosis in DNA-damaged KC, and the presence of LC in the skin. LC were observed to modulate the chemokine response in the skin following exposure to UVB, thereby affecting the trafficking of neutrophils. Skin lacking LC revealed diminished inflammation, contained fewer TNF-α-producing neutrophils, and due to the prevention of apoptosis induction, a lingering population of DNA-damaged KC, presumably carrying the risk of enduring genomic alterations. In summary, our results underscore the pivotal role of LC in preserving the homeostasis of UVB-irradiated skin. These findings contribute to a deeper understanding of the intricate mechanisms underlying acute sunburn responses and their implications for UV-induced skin cancer.

2.
Eur J Immunol ; 53(11): e2249819, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-36512638

RESUMO

This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs and various nonlymphoid tissues. DC are sentinels of the immune system present in almost every mammalian organ. Since they represent a rare cell population, DC need to be extracted from organs with protocols that are specifically developed for each tissue. This article provides detailed protocols for the preparation of single-cell suspensions from various mouse nonlymphoid tissues, including skin, intestine, lung, kidney, mammary glands, oral mucosa and transplantable tumors. Furthermore, our guidelines include comprehensive protocols for multiplex flow cytometry analysis of DC subsets and feature top tricks for their proper discrimination from other myeloid cells. With this collection, we provide guidelines for in-depth analysis of DC subsets that will advance our understanding of their respective roles in healthy and diseased tissues. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all coauthors, making it an essential resource for basic and clinical DC immunologists.


Assuntos
Células Dendríticas , Pele , Animais , Humanos , Citometria de Fluxo , Células Mieloides , Rim , Mamíferos
3.
Eur J Immunol ; 52(12): 2006-2009, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35944142

RESUMO

This novel 26-color flow cytometry panel allows the detailed immune phenotyping of the complex network of myeloid cells in murine lymph nodes and skin. With the optimized panel the different murine DC-subsets and other myeloid cell types can be identified and further characterized for co-stimulatory and inhibitory surface molecules.


Assuntos
Células Dendríticas , Camundongos , Animais , Citometria de Fluxo
4.
Eur J Immunol ; 52(11): 1829-1841, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-34932821

RESUMO

Human skin is a preferred vaccination site as it harbors multiple dendritic cell (DC) subsets, which display distinct C-type lectin receptors (CLR) that recognize pathogens. Antigens can be delivered to CLR by antibodies or ligands to boost antigen-specific immune responses. This concept has been established in mouse models but detailed insights into the functional consequences of antigen delivery to human skin DC in situ are sparse. In this study, we cloned and produced an anti-human Langerin antibody conjugated to the EBV nuclear antigen 1 (EBNA1). We confirmed specific binding of anti-Langerin-EBNA1 to Langerhans cells (LC). This novel LC-based vaccine was then compared to an existing anti-DEC-205-EBNA1 fusion protein by loading LC in epidermal cell suspensions before coculturing them with autologous T cells. After restimulation with EBNA1-peptides, we detected elevated levels of IFN-γ- and TNF-α-positive CD4+ T cells with both vaccines. When we injected the fusion proteins intradermally into human skin explants, emigrated skin DC targeted via DEC-205-induced cytokine production by T cells, whereas the Langerin-based vaccine failed to do so. In summary, we demonstrate that antibody-targeting approaches via the skin are promising vaccination strategies, however, further optimizations of vaccines are required to induce potent immune responses.


Assuntos
Células Dendríticas , Células de Langerhans , Lectinas Tipo C , Vacinas , Animais , Humanos , Camundongos , Antígenos/metabolismo , Células de Langerhans/metabolismo , Lectinas Tipo C/metabolismo , Lectinas de Ligação a Manose , Pele
5.
Exp Dermatol ; 30(9): 1279-1289, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33797121

RESUMO

Dendritic cells (DC) are promising targets for immunotherapy of cancer. Clinically, immunization against cancer antigens by means of the most potent antigen-presenting cells, that is DC, remains an important treatment option in combination with the modern immune checkpoint approaches. Instead of adoptively transferring in vitro monocyte-derived DC, they can also be loaded in situ by antibody-mediated targeting of antigen. Conventionally, these vaccines are delivered by classical intradermal injections. Here, we tested an alternative approach, namely laser-assisted epicutaneous immunization. With an infrared laser ("Precise Laser Epidermal System"/P.L.E.A.S.E.® Laser System), we created micropores in human skin and applied monoclonal antibodies (mAbs) against C-type lectins, for example DEC-205/CD205 and Langerin/CD207. Optimal parameters for formation of pores in epidermis and dermis were determined. We could induce pores of defined depths without enhanced apoptosis around them. Antibodies applied epicutaneously to the laser-porated skin could be detected both in Langerhans cells (LC) in situ in the epidermis and in migratory skin DC subsets from short term human skin explant culture, demonstrating uptake and transport of Langerin and DEC-205 mAbs. Efficacy of targeting was similar between the different laser treatments and pore depths. Thus, laser-assisted epicutaneous immunization may be a valuable alternative to intradermal injection, yet the loading efficacy of DC needs to be further improved.


Assuntos
Administração Cutânea , Anticorpos/imunologia , Antígenos CD/imunologia , Células Dendríticas/imunologia , Imunização/métodos , Células de Langerhans/imunologia , Lasers , Lectinas Tipo C/imunologia , Antígenos de Histocompatibilidade Menor/imunologia , Receptores de Superfície Celular/imunologia , Adulto , Idoso , Feminino , Humanos , Masculino , Adulto Jovem
6.
Immunity ; 37(5): 766-8, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-23159223

RESUMO

It is unclear how the Langerhans cell (LC) network is maintained in adult epidermis. In this issue of Immunity, Seré et al. (2012) show that LCs are replenished in two waves. Monocyte-derived, short-lived LCs come first. A second wave follows, and these LCs of nonmonocytic origin are long-lived.

7.
Int J Cancer ; 146(5): 1409-1420, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31702822

RESUMO

Therapeutic success of targeted therapy with BRAF inhibitors (BRAFi) for melanoma is limited by resistance development. Observations from preclinical mouse models and recent insights into the immunological effects caused by BRAFi give promise for future development of combination therapy for human melanoma. In our study, we used the transplantable D4M melanoma mouse model with the BRAFV600E mutation and concomitant PTEN loss in order to characterize alterations in tumor-infiltrating effector immune cells when tumors become resistant to BRAFi. We found that BRAFi-sensitive tumors displayed a pronounced inflammatory milieu characterized by high levels of cytokines and chemokines accompanied by an infiltration of T and NK cells. The tumor-infiltrating effector cells were activated and produced high levels of IFN-γ, TNF-α and granzyme B. When tumors became resistant and progressively grew, they reverted to a low immunogenic state similar to untreated tumors as reflected by low mRNA levels of proinflammatory cytokines and chemokines and fewer tumor-infiltrating T and NK cells. Moreover, these T and NK cells were functionally impaired in comparison to their counterparts in BRAFi-sensitive tumors. Their effector cell function could be restored by additional peritumoral treatment with the TLR7 agonist imiquimod, a clinically approved agent for nonmelanoma skin cancer. Indeed, resistance to BRAFi therapy was delayed and accompanied by high numbers of activated T and NK cells in tumors. Thus, combining BRAFi with an immune stimulating agent such as a TLR ligand could be a promising alternative approach for the treatment of melanoma.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Melanoma Experimental/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linhagem Celular Tumoral/transplante , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Feminino , Humanos , Imiquimode/farmacologia , Imiquimode/uso terapêutico , Indóis/farmacologia , Indóis/uso terapêutico , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Linfócitos do Interstício Tumoral/imunologia , Masculino , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/metabolismo , Camundongos , Mutação , Células T Matadoras Naturais , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas B-raf/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/metabolismo
8.
J Dtsch Dermatol Ges ; 18(11): 1270-1277, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33197129

RESUMO

BACKGROUND AND OBJECTIVES: We examined retrospectively whether the combination of standard dacarbazine (DTIC) and/or fotemustine chemotherapy and autologous peptide-loaded dendritic cell (DC) vaccination may improve survival of stage IV melanoma patients. Furthermore, a small cohort of long-term survivors was studied in more detail. PATIENTS AND METHODS: Between 1998 and 2008, 41 patients were vaccinated at least three times with DCs while receiving chemotherapy and compared to all other 168 patients in our database who only received chemotherapy (1993-2008). RESULTS: Median life expectancy of patients receiving additional DC-vaccination was 18 months, compared to eleven months for patients under standard chemotherapy alone. In contrast to patients with other haplotypes, the HLA-A1/A1 subset of DC-treated patients showed significantly lower median survival (12 vs. 25 months). Autoantibodies were frequently detected in serum of both vaccinated and non-vaccinated patients, and there was no correlation between titers, loss or appearance of autoantibodies and survival. Additionally, phenotyping of DCs and PBMCs also did not reveal any conspicuous correlation with survival. CONCLUSIONS: Combining standard chemotherapy and DC vaccination appears superior to chemotherapy alone. The impact of HLA haplotypes on survival emphasizes the importance of a careful selection of patients with specific, well-defined HLA haplotypes for future vaccination trials using peptide-pulsed DCs, possibly combined with checkpoint inhibitors.


Assuntos
Células Dendríticas , Melanoma , Feminino , Humanos , Masculino , Peptídeos , Estudos Retrospectivos
9.
Int J Cancer ; 145(7): 1958-1969, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30972741

RESUMO

The efficacy of cancer vaccines has been limited by the immunosuppressive tumor microenvironment, which can be alleviated by immune checkpoint inhibitor (ICI) therapy. Here, we tested if oncolytic viruses (OVs), similar to ICI, can also synergize with cancer vaccines by modulating the tumor microenvironment. VSV-GP, a chimeric vesicular stomatitis virus (VSV) pseudotyped with the glycoprotein (GP) of the lymphocytic choriomeningitis virus, is a promising new OV candidate. Here, we show that in mouse B16-OVA melanoma, combination treatment of VSV-GP with an ovalbumin (OVA) peptide-loaded dendritic cell (DC) vaccine (DCVacc) significantly enhanced survival over the single agent therapies, although both DCVacc and DCVacc/VSV-GP treatments induced comparable levels of OVA-specific CD8 T cell responses. Virus replication was minimal so that direct viral oncolysis in B16-OVA did not contribute to this synergism. The strong therapeutic effect of the DCVacc/VSV-GP combination treatment was associated with high numbers of tumor-infiltrating, highly activated T cells and the relative reduction of regulatory T cells in treated and contra-lateral nontreated tumors. Accordingly, depletion of CD8 T cells but not natural killer cells abrogated the therapeutic effect of DCVacc/VSV-GP supporting the crucial role of CD8 T cells. In addition, a drastic increase in several proinflammatory cytokines was observed in VSV-GP-treated tumors. Taken together, OVs, similar to ICI, have the potential to markedly increase the efficacy of cancer vaccines by alleviating local immune suppression in the tumor microenvironment.


Assuntos
Vacinas Anticâncer/administração & dosagem , Glicoproteínas/metabolismo , Melanoma Experimental/terapia , Terapia Viral Oncolítica/métodos , Vírus da Estomatite Vesicular Indiana/fisiologia , Animais , Vacinas Anticâncer/farmacologia , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Glicoproteínas/genética , Humanos , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/metabolismo , Melanoma Experimental/imunologia , Camundongos , Vírus Oncolíticos/fisiologia , Ovalbumina/imunologia , Resultado do Tratamento , Microambiente Tumoral/efeitos dos fármacos , Vírus da Estomatite Vesicular Indiana/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J Cell Mol Med ; 20(5): 930-8, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26931745

RESUMO

Atopic dermatitis (AD) is a widespread inflammatory skin disease with an early onset, characterized by pruritus, eczematous lesions and skin dryness. This chronic relapsing disease is believed to be primarily a result of a defective epidermal barrier function associated with genetic susceptibility, immune hyper-responsiveness of the skin and environmental factors. Although the important role of abnormal immune reactivity in the pathogenesis of AD is widely accepted, the role of regulatory T cells (Tregs) remains elusive. We found that the Treg population is expanded in a mouse model of AD, i.e. mice topically treated with vitamin D3 (VitD). Moreover, mice with AD-like symptoms exhibit increased inducible T-cell costimulator (ICOS)-, cytotoxic T-lymphocyte antigen-4 (CTLA-4)- and Glycoprotein-A repetitions predominant receptor (GARP)-expressing Tregs in skin-draining lymph nodes. Importantly, the differentiation of Tregs into thymus-derived Tregs is favoured in our mouse model of AD. Emigrated skin-derived dendritic cells are required for Treg induction and Langerhans cells are responsible for the biased expansion of thymus-derived Tregs . Intriguingly, thymus-derived Tregs isolated from mice with AD-like symptoms exhibit a Th2 cytokine profile. Thus, AD might favour the expansion of pathogenic Tregs able to produce Th2 cytokines and to promote the disease instead of alleviating symptoms.


Assuntos
Citocinas/imunologia , Dermatite Atópica/patologia , Células de Langerhans/patologia , Linfócitos T Citotóxicos/patologia , Linfócitos T Reguladores/patologia , Animais , Antígeno CTLA-4/genética , Antígeno CTLA-4/imunologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colecalciferol/farmacologia , Citocinas/genética , Dermatite Atópica/tratamento farmacológico , Dermatite Atópica/genética , Dermatite Atópica/imunologia , Modelos Animais de Doenças , Epiderme/efeitos dos fármacos , Epiderme/imunologia , Epiderme/patologia , Regulação da Expressão Gênica , Humanos , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Células de Langerhans/efeitos dos fármacos , Células de Langerhans/imunologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Linfonodos/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Equilíbrio Th1-Th2/efeitos dos fármacos , Timo/efeitos dos fármacos , Timo/imunologia , Timo/patologia
11.
Blood ; 123(2): 217-27, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-24092934

RESUMO

Langerhans cells (LCs) are dendritic cells (DCs) residing in epithelia, where they critically regulate immunity and tolerance. The p14 adaptor molecule is part of the late endosomal/LAMTOR (lysosomal adaptor and mitogen-activated protein kinase and mammalian target of rapamycin [mTOR] activator/regulator) complex, thereby contributing to the signal transduction of the extracellular signaling-regulated kinase (ERK) and the mTOR cascade. Furthermore, p14 represents an important regulator for endosomal sorting processes within the cell. Mutated, dysfunctional p14 leads to a human immunodeficiency disorder with endosomal/lysosomal defects in immune cells. Because p14 participates in the regulation of endosomal trafficking, growth factor signaling, and cell proliferation, we investigated the role of p14 in mouse DCs/LCs using a conditional knockout mouse model. p14-deficient animals displayed a virtually complete loss of LCs in the epidermis early after birth due to impaired proliferation and increased apoptosis of LCs. Repopulation analysis after application of contact sensitizer leads to the recruitment of a transient LC population, predominantly consisting of short-term LCs. The underlying molecular mechanism involves the p14-mediated disruption of the LAMTOR complex which results in the malfunction of both ERK and mTOR signal pathways. Hence, we conclude that p14 acts as a novel and essential regulator of LC homeostasis in vivo.


Assuntos
Endossomos/metabolismo , Homeostase , Células de Langerhans/metabolismo , Proteínas/genética , Proteínas/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/genética , Antígeno CD11c/genética , Antígeno CD11c/metabolismo , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Mitose/genética , Complexos Multiproteicos/metabolismo , Transdução de Sinais , Pele/imunologia , Pele/metabolismo , Pele/patologia , Serina-Treonina Quinases TOR/metabolismo
13.
Eur J Immunol ; 43(10): 2718-29, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23843024

RESUMO

The dual erbB1/2 tyrosine kinase inhibitor lapatinib as well as the anthracycline doxorubicin are both used in the therapy of HER2-positive breast cancer. Using MMTV-neu mice as an animal model for HER2-positive breast cancer, we observed enhanced tumor infiltration by IFN-γ-secreting T cells after treatment with doxorubicin and/or lapatinib. Antibody depletion experiments revealed a contribution of CD8⁺ but not CD4⁺ T cells to the antitumor effect of these drugs. Doxorubicin treatment additionally decreased the content of immunosuppressive tumor-associated macrophages (TAMs) in the tumor bed. In contrast, Stat1-deficient mice were resistant to tumor growth inhibition by lapatinib and/or doxorubicin and exhibited impaired T-cell activation and reduced T-cell infiltration of the tumor in response to drug treatment. Furthermore, Stat1-deficiency resulted in reduced expression of the T-cell chemotactic factors CXCL9, CXCL10, and CXCL11 in the tumor epithelium. The inhibition of TAM infiltration of the tumor by doxorubicin and the immunosuppressive function of TAMs were found to be Stat1 independent. Taken together, the results point to an important contribution toward enhancing T-cell and IFN-γ-based immunity by lapatinib as well as doxorubicin and emphasize the role of Stat1 in building an effective antitumor immune response.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Linfócitos T CD8-Positivos/efeitos dos fármacos , Doxorrubicina/administração & dosagem , Inibidores de Proteínas Quinases/administração & dosagem , Quinazolinas/administração & dosagem , Fator de Transcrição STAT1/metabolismo , Animais , Antígenos de Neoplasias/imunologia , Neoplasias da Mama/imunologia , Linfócitos T CD8-Positivos/imunologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Modelos Animais de Doenças , Receptores ErbB/antagonistas & inibidores , Feminino , Humanos , Técnicas In Vitro , Interferon gama/metabolismo , Lapatinib , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ratos , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia
14.
Exp Dermatol ; 23(12): 909-15, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25346475

RESUMO

Skin dendritic cells (DC) express C-type lectin receptors for the recognition of pathogens. Langerhans cells (LC) express the receptor Langerin/CD207, whereas DEC-205/CD205 is mainly expressed by dermal DC, but can also be detected at low levels on LC. In this study, we tested an ex vivo approach for targeting DC in situ with monoclonal antibodies (mAb) against Langerin and DEC-205. The targeting mAb was injected intradermally into human skin biopsies or added to the medium during skin explant culture. Corresponding to the expression patterns of these lectin receptors on skin DC, Langerin mAb was detected merely in LC in the epidermis and DEC-205 mainly in dermal DC in human skin explants, regardless of the application route. Migratory skin DC bound and carried targeting mAb from skin explants according to their lectin receptor expression profiles. In contrast to the very selective transport of Langerin mAb by LC, DEC-205 mAb was more widely distributed on all CD1a(+) skin DC subsets but almost absent in CD14(+) dermal DC. As effective vaccination requires the addition of adjuvant, we co-administered the toll-like receptor (TLR)-3 ligand poly I:C with the mAb. This adjuvant enhanced binding of DEC-205 mAb to all skin DC subsets, whereas Langerin targeting efficacy remained unchanged. Our findings demonstrate that LC can be preferentially targeted by Langerin mAb. In contrast, DEC-205 mAb can be bound by all CD1a(+) skin DC subsets. The efficacy of DEC-205 mAb targeting strategy can be boosted by addition of poly I:C underlining the potential of this combination for immunotherapeutical interventions.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Células de Langerhans/imunologia , Receptores Mitogênicos/antagonistas & inibidores , Adjuvantes Imunológicos/administração & dosagem , Anticorpos Monoclonais/metabolismo , Complexo Antígeno-Anticorpo/metabolismo , Antígenos CD/imunologia , Células Cultivadas , Humanos , Imunização , Injeções Intradérmicas , Lectinas Tipo C/antagonistas & inibidores , Lectinas Tipo C/imunologia , Lectinas de Ligação a Manose/antagonistas & inibidores , Lectinas de Ligação a Manose/imunologia , Antígenos de Histocompatibilidade Menor , Poli I-C/administração & dosagem , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/imunologia , Receptores Mitogênicos/imunologia , Receptor 3 Toll-Like/metabolismo
15.
J Immunol ; 188(5): 2146-55, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22291181

RESUMO

Incorporation of Ags by dendritic cells (DCs) increases when Ags are targeted to endocytic receptors by mAbs. We have previously demonstrated in the mouse that mAbs against C-type lectins administered intradermally are taken up by epidermal Langerhans cells (LCs), dermal Langerin(neg) DCs, and dermal Langerin(+) DCs in situ. However, the relative contribution of these skin DC subsets to the induction of immune responses after Ag targeting has not been addressed in vivo. We show in this study that murine epidermal LCs and dermal DCs transport intradermally injected mAbs against the lectin receptor DEC-205/CD205 in vivo. Skin DCs targeted in situ with mAbs migrated through lymphatic vessels in steady state and inflammation. In the skin-draining lymph nodes, targeting mAbs were found in resident CD8α(+) DCs and in migrating skin DCs. More than 70% of targeted DCs expressed Langerin, including dermal Langerin(+) DCs and LCs. Numbers of targeted skin DCs in the nodes increased 2-3-fold when skin was topically inflamed by the TLR7 agonist imiquimod. Complete removal of the site where OVA-coupled anti-DEC-205 had been injected decreased endogenous cytotoxic responses against OVA peptide-loaded target cells by 40-50%. Surprisingly, selective ablation of all Langerin(+) skin DCs in Langerin-DTR knock-in mice did not affect such responses independently of the adjuvant chosen. Thus, in cutaneous immunization strategies where Ag is targeted to DCs, Langerin(+) skin DCs play a major role in transport of anti-DEC-205 mAb, although Langerin(neg) dermal DCs and CD8α(+) DCs are sufficient to subsequent CD8(+) T cell responses.


Assuntos
Anticorpos/administração & dosagem , Antígenos CD/imunologia , Antígenos de Superfície/biossíntese , Células Dendríticas/imunologia , Mediadores da Inflamação/fisiologia , Lectinas Tipo C/biossíntese , Lectinas Tipo C/imunologia , Lectinas de Ligação a Manose/biossíntese , Receptores de Superfície Celular/imunologia , Pele/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Antígenos de Superfície/fisiologia , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Técnicas de Introdução de Genes , Mediadores da Inflamação/metabolismo , Injeções Intradérmicas , Células de Langerhans/imunologia , Células de Langerhans/metabolismo , Lectinas Tipo C/fisiologia , Linfonodos/imunologia , Linfonodos/metabolismo , Linfonodos/patologia , Lectinas de Ligação a Manose/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Antígenos de Histocompatibilidade Menor , Técnicas de Cultura de Órgãos , Transporte Proteico/genética , Transporte Proteico/imunologia , Ratos , Pele/metabolismo , Pele/patologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/patologia
16.
Front Immunol ; 15: 1342895, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38566997

RESUMO

Excessive fibrous capsule formation around silicone mammary implants (SMI) involves immune reactions to silicone. Capsular fibrosis, a common SMI complication linked to host responses, worsens with specific implant topographies. Our study with 10 patients investigated intra- and inter-individually, reduced surface roughness effects on disease progression, wound responses, chronic inflammation, and capsular composition. The results illuminate the significant impact of surface roughness on acute inflammatory responses, fibrinogen accumulation, and the subsequent fibrotic cascade. The reduction of surface roughness to an average roughness of 4 µm emerges as a promising approach for mitigating detrimental immune reactions, promoting healthy wound healing, and curbing excessive fibrosis. The identified proteins adhering to rougher surfaces shed light on potential mediators of pro-inflammatory and pro-fibrotic processes, further emphasizing the need for meticulous consideration of surface design. The composition of the implant capsule and the discovery of intracapsular HSP60 expression highlight the intricate web of stress responses and immune activation that can impact long-term tissue outcomes.


Assuntos
Inflamação , Próteses e Implantes , Humanos , Silicones , Fibrose , Cicatrização
17.
J Immunother Cancer ; 12(4)2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38631706

RESUMO

BACKGROUND: Tumor-targeted therapy causes impressive tumor regression, but the emergence of resistance limits long-term survival benefits in patients. Little information is available on the role of the myeloid cell network, especially dendritic cells (DC) during tumor-targeted therapy. METHODS: Here, we investigated therapy-mediated immunological alterations in the tumor microenvironment (TME) and tumor-draining lymph nodes (LN) in the D4M.3A preclinical melanoma mouse model (harboring the V-Raf murine sarcoma viral oncogene homolog B (BRAF)V600E mutation) by using high-dimensional multicolor flow cytometry in combination with multiplex immunohistochemistry. This was complemented with RNA sequencing and cytokine quantification to characterize the immune status of the tumors. The importance of T cells during tumor-targeted therapy was investigated by depleting CD4+ or CD8+ T cells in tumor-bearing mice. Tumor antigen-specific T-cell responses were characterized by performing in vivo T-cell proliferation assays and the contribution of conventional type 1 DC (cDC1) to T-cell immunity during tumor-targeted therapy was assessed using Batf3-/- mice lacking cDC1. RESULTS: Our findings reveal that BRAF-inhibitor therapy increased tumor immunogenicity, reflected by an upregulation of genes associated with immune activation. The T cell-inflamed TME contained higher numbers of activated cDC1 and cDC2 but also inflammatory CCR2-expressing monocytes. At the same time, tumor-targeted therapy enhanced the frequency of migratory, activated DC subsets in tumor-draining LN. Even more, we identified a cDC2 population expressing the Fc gamma receptor I (FcγRI)/CD64 in tumors and LN that displayed high levels of CD40 and CCR7 indicating involvement in T cell-mediated tumor immunity. The importance of cDC2 is underlined by just a partial loss of therapy response in a cDC1-deficient mouse model. Both CD4+ and CD8+ T cells were essential for therapy response as their respective depletion impaired therapy success. On resistance development, the tumors reverted to an immunologically inert state with a loss of DC and inflammatory monocytes together with the accumulation of regulatory T cells. Moreover, tumor antigen-specific CD8+ T cells were compromised in proliferation and interferon-γ-production. CONCLUSION: Our results give novel insights into the remodeling of the myeloid landscape by tumor-targeted therapy. We demonstrate that the transient immunogenic tumor milieu contains more activated DC. This knowledge has important implications for the development of future combinatorial therapies.


Assuntos
Melanoma , Humanos , Animais , Camundongos , Melanoma/metabolismo , Linfócitos T CD8-Positivos , Proteínas Proto-Oncogênicas B-raf/genética , Células Dendríticas , Antígenos de Neoplasias , Microambiente Tumoral
18.
Eur J Immunol ; 42(3): 589-97, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22488362

RESUMO

Cytohesin-interacting protein (Cytip) is induced during dendritic cell (DC) maturation and in T cells upon activation. It has also been shown to be involved in the regulation of immune responses. Here, we evaluated the functional consequences of Cytip deficiency in DCs using Cytip knockout (KO) mice. No difference in DC subpopulations in the skin draining lymph nodes (LNs) was found between Cytip KO mice and their wild-type counterparts, excluding a role in DC development. To investigate the function of Cytip in DCs in vivo, we used 2,4,6-trinitrochlorobenzene (TNCB)-induced contact hypersensitivity (CHS) as a model system. In the sensitization as well as in the elicitation phase, DCs derived from Cytip KO mice induced an increased inflammatory reaction indicated by more pronounced ear swelling. Furthermore, IL-12 production was increased in Cytip KO bone marrow-derived DCs (BMDCs) after CpG stimulation. Additionally, Cytip-deficient DCs loaded with ovalbumin induced stronger proliferation of antigen-specific CD4(+) and CD8(+) T cells in vitro. Finally, migration of skin DCs was not altered after TNCB application due to Cytip deficiency. Taken together, these data suggest a suppressive function for Cytip in mouse DCs in limiting immune responses.


Assuntos
Proteínas de Transporte/imunologia , Células Dendríticas/imunologia , Dermatite de Contato/imunologia , Proteínas de Membrana/imunologia , Linfócitos T/imunologia , Animais , Proteínas de Transporte/genética , Processos de Crescimento Celular/imunologia , Células Dendríticas/citologia , Dermatite de Contato/patologia , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe II/imunologia , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/patologia , Interleucina-12/genética , Interleucina-12/imunologia , Ativação Linfocitária/imunologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Cloreto de Picrila/administração & dosagem , RNA Mensageiro/química , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Pele/imunologia , Pele/patologia , Linfócitos T/patologia
19.
Inflamm Regen ; 43(1): 53, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37904253

RESUMO

BACKGROUND: Chronic non-healing wounds pose a global health challenge. Under optimized conditions, skin wounds heal by the formation of scar tissue. However, deregulated cell activation leads to persistent inflammation and the formation of granulation tissue, a type of premature scar tissue without epithelialization. Regenerative cells from the wound periphery contribute to the healing process, but little is known about their cellular fate in an inflammatory, macrophage-dominated wound microenvironment. METHODS: We examined CD45-/CD31-/CD34+ preadipocytes and CD68+ macrophages in human granulation tissue from pressure ulcers (n=6) using immunofluorescence, immunohistochemistry, and flow cytometry. In vitro, we studied macrophage-preadipocyte interactions using primary human adipose-derived stem cells (ASCs) exposed to conditioned medium harvested from IFNG/LPS (M1)- or IL4/IL13 (M2)-activated macrophages. Macrophages were derived from THP1 cells or CD14+ monocytes. In addition to confocal microscopy and flow cytometry, ASCs were analyzed for metabolic (OXPHOS, glycolysis), morphological (cytoskeleton), and mitochondrial (ATP production, membrane potential) changes. Angiogenic properties of ASCs were determined by HUVEC-based angiogenesis assay. Protein and mRNA levels were assessed by immunoblotting and quantitative RT-PCR. RESULTS: CD45-/CD31-/CD34+ preadipocytes were observed with a prevalence of up to 1.5% of total viable cells in human granulation tissue. Immunofluorescence staining suggested a spatial proximity of these cells to CD68+ macrophages in vivo. In vitro, ASCs exposed to M1, but not to M2 macrophage secretome showed a pro-fibrotic response characterized by stress fiber formation, elevated alpha smooth muscle actin (SMA), and increased expression of integrins ITGA5 and ITGAV. Macrophage-secreted IL1B and TGFB1 mediated this response via the PI3K/AKT and p38-MAPK pathways. In addition, ASCs exposed to M1-inflammatory stress demonstrated reduced migration, switched to a glycolysis-dominated metabolism with reduced ATP production, and increased levels of inflammatory cytokines such as IL1B, IL8, and MCP1. Notably, M1 but not M2 macrophages enhanced the angiogenic potential of ASCs. CONCLUSION: Preadipocyte fate in wound tissue is influenced by macrophage polarization. Pro-inflammatory M1 macrophages induce a pro-fibrotic response in ASCs through IL1B and TGFB1 signaling, while anti-inflammatory M2 macrophages have limited effects. These findings shed light on cellular interactions in chronic wounds and provide important information for the potential therapeutic use of ASCs in human wound healing.

20.
J Immunol ; 182(12): 7644-54, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19494288

RESUMO

APCs, such as dendritic cells (DC), can present glycolipid Ags on CD1d molecules to NKT cells. This interaction activates DC and NKT cells, leading to release of cytokines and enhanced T cell responses. Thus, glycolipid Ags are currently being tested as adjuvants for immunotherapy. We were interested in the interaction of murine skin DC with NKT cells in skin-draining lymph nodes. We observed that all skin DC subsets expressed CD1d upon migration to the lymph nodes. Moreover, skin DC were able to present the synthetic glycolipid Ag alpha-galactosylceramide (alpha-GalCer) to the NKT cell hybridoma DN32.D3. Intradermally injected alpha-GalCer was presented by migratory skin DC and lymph node DC to NKT hybridoma cells in vitro. When we injected alpha-GalCer intradermally into the skin, the numbers of various leukocyte subsets in the draining lymph nodes did not change significantly. However, T and B cells as well as NKT cells up-regulated the activation marker CD69. Coapplication of alpha-GalCer with the tumor model Ag OVA induced strong cytolytic CD8(+) T cell function that could inhibit the growth of B16 melanoma cells expressing OVA. However, mice that were devoid of migratory skin DC developed similar cytotoxic immune responses after intradermal immunization, indicating that skin DC are not required for the adjuvant properties of NKT cell activation and Ag presentation by this immunization route. In conclusion, migratory skin DC are able to interact with NKT cells; however, intradermally applied glycolipids are presented predominantly by lymph node DC to NKT cells.


Assuntos
Glicolipídeos/imunologia , Linfonodos/imunologia , Células T Matadoras Naturais/imunologia , Pele/imunologia , Animais , Antígenos CD1d/imunologia , Linhagem Celular , Movimento Celular/imunologia , Modelos Animais de Doenças , Progressão da Doença , Galactosilceramidas/química , Glicolipídeos/química , Células de Langerhans/citologia , Células de Langerhans/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neoplasias/imunologia , Neoplasias/patologia , Pele/citologia , Linfócitos T Citotóxicos/imunologia , Técnicas de Cultura de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA