Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Chemistry ; 29(38): e202300997, 2023 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-37097418

RESUMEN

Photoactivatable fluorophores (PAFs) are powerful tools for biological imaging applications because they provide spatiotemporal control of fluorescence distribution. Many of the existing PAFs can only be activated by UV irradiation. In our study, we present a blue light (1P) and NIR light (2P) activatable rhodamine fluorophore. Next to the description of the synthesis and the investigation of the photoreaction, we demonstrate the use of our PAF in the context of laser scanning microscopy. By immobilization of our PAF in a hydrogel, we were able to write and read spatially resolved illumination patterns with excellent contrast after both one-photon and two-photon excitation.


Asunto(s)
Luz , Fotones , Rodaminas , Colorantes Fluorescentes , Rayos Ultravioleta
2.
Chemistry ; 29(25): e202300149, 2023 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-36785982

RESUMEN

Two-photon (2P) activatable probes are of high value in biological and medical chemistry since near infrared (NIR) light can penetrate deeply even in blood-perfused tissue and due to the intrinsic three-dimensional activation properties. Designing two-photon chromophores is challenging. However, the two-photon absorption qualities of a photocage can be improved with an intramolecular sensitizer, which transfers the absorbed light onto the cage. We herein present the synthesis and photophysical characterization of a 2P-sensitive uncaging dyad based on rhodamine 101 as donor fluorophore and a redshifted BODIPY as acceptor photocage. Liberation of p-nitroaniline (PNA) upon one-photon photolysis was confirmed by HPLC analysis. The photoreaction was found to be accompanied by a considerable change of the fluorescence properties of the chromophores. The possibility of a fluorescent read-out enabled the detection of two-photon induced uncaging by confocal fluorescence microscopy.

3.
Phys Chem Chem Phys ; 24(3): 1795-1802, 2022 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-34985062

RESUMEN

In view of the demand for photoactivatable probes that operate in the visible (VIS) to near infrared (NIR) region of the spectrum, we designed a bichromophoric system based on a rhodamine fluorophore and a BODIPY photocage. Two-photon excited fluorescence (TPEF) measurements and quantum chemical calculations reveal excellent two-photon properties of the employed rhodamine derivative. Excitation of the rhodamine unit via a one- or two-photon process leads to excitation energy transfer (EET) onto the BODIPY part, which is followed by the liberation of the leaving group. Ultrafast transient absorption spectroscopy provides evidence for a highly efficient EET dynamics on a sub-500 femtosecond scale. Complementary quantum dynamical calculations using the multi-layer multiconfiguration time-dependent Hartree (ML-MCTDH) approach highlight the quantum coherent character of the EET transfer. Photorelease of p-nitroaniline (PNA) was investigated by UV/vis absorption spectroscopy by either excitation of the rhodamine or the BODIPY moiety. Even though a quantitative assessment of the PNA yield could not be achieved for this particular BODIPY cage, the present study provides a design principle for a class of photocages that can be broadly activated between 500 and 900 nm.

4.
Int J Cancer ; 149(9): 1628-1638, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34224592

RESUMEN

Myeloid-derived suppressor cells (MDSC) represent a highly immunosuppressive population that expands in tumor bearing hosts and inhibits both T and NK cell antitumor effector functions. Among MDSC subpopulations, the polymorphonuclear (PMN) one is gaining increasing interest since it is a predominant MDSC subset in most cancer entities and inherits unique properties to facilitate metastatic spread. In addition, further improvement in distinguishing PMN-MDSC from neutrophils has contributed to the design of novel therapeutic approaches. In this review, we summarize the current view on the origin of PMN-MDSC and their relation to classical neutrophils. Furthermore, we outline the metastasis promoting features of these cells and promising strategies of their targeting to improve the efficacy of cancer immunotherapy.


Asunto(s)
Carcinogénesis/inmunología , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/inmunología , Neutrófilos/inmunología , Microambiente Tumoral/inmunología , Animales , Humanos , Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Neoplasias/patología , Neoplasias/terapia , Linfocitos T/inmunología
5.
Cancers (Basel) ; 13(4)2021 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-33578808

RESUMEN

BACKGROUND: Despite recent improvement in the treatment of malignant melanoma by immune-checkpoint inhibitors, the disease can progress due to an immunosuppressive tumor microenvironment (TME) mainly represented by myeloid-derived suppressor cells (MDSC). However, the relative contribution of the polymorphonuclear (PMN) and monocytic (M) MDSC subsets to melanoma progression is not clear. Here, we compared both subsets regarding their immunosuppressive capacity and recruitment mechanisms. Furthermore, we inhibited PMN-MDSC migration in vivo to determine its effect on tumor progression. METHODS: Using the RET transgenic melanoma mouse model, we investigated the immunosuppressive function of MDSC subsets and chemokine receptor expression on these cells. The effect of CXCR2 inhibition on PMN-MDSC migration and tumor progression was studied in RET transgenic mice and in C57BL/6 mice after surgical resection of primary melanomas. RESULTS: Immunosuppressive capacity of intratumoral M- and PMN-MDSC was comparable in melanoma bearing mice. Anti-CXCR2 therapy prolonged survival of these mice and decreased the occurrence of distant metastasis. Furthermore, this therapy reduced the infiltration of melanoma lesions and pre-metastatic sites with PMN-MDSC that was associated with the accumulation of natural killer (NK) cells. CONCLUSIONS: We provide evidence for the tumor-promoting properties of PMN-MDSC as well as for the anti-tumor effects upon their targeting in melanoma bearing mice.

6.
Cell Immunol ; 360: 104274, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33383383

RESUMEN

The low dose application of chemotherapeutic agents such as paclitaxel was previously shown to initiate anti-tumor activity by neutralizing myeloid-derived suppressor cells (MDSCs) in melanoma mouse models. Here, we investigated immunomodulating effects of low-dose paclitaxel in 9 metastatic melanoma patients resistant to prior treatments. Three patients showed response to therapy (two partial responses and one stable disease). In responding patients, paclitaxel decreased the frequency and immunosuppressive pattern of MDSCs in the peripheral blood and skin metastases. Furthermore, paclitaxel modulated levels of inflammatory mediators in the serum. In addition, responders displayed enhanced frequencies of tumor-infiltrating CD8+ T cells and their activity indicated by the upregulation of CD25 and TCR ζ-chain expression. Our study suggests that low-dose paclitaxel treatment could improve clinical outcome of some advanced melanoma patients by enhancing anti-tumor immunity and might be proposed for combined melanoma immunotherapy.


Asunto(s)
Melanoma/tratamiento farmacológico , Paclitaxel/uso terapéutico , Anciano , Linfocitos T CD8-positivos/inmunología , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia/métodos , Masculino , Melanoma/inmunología , Melanoma/fisiopatología , Persona de Mediana Edad , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/metabolismo , Paclitaxel/administración & dosificación , Paclitaxel/metabolismo , Proyectos Piloto , Neoplasias Cutáneas/inmunología , Microambiente Tumoral/inmunología
7.
Methods Mol Biol ; 2236: 67-75, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33237541

RESUMEN

Myeloid-derived suppressor cells (MDSC) are known to inhibit functions of T and NK cells. MDSC have been shown to be generated and to accumulate under chronic inflammatory conditions that are typical for cancer. Therefore, it would be highly beneficial to find ways to diminish the number and immunosuppressive functions of these cells in tumor-bearing hosts. Here we describe current protocols to deplete MDSC or induce their maturation in preclinical tumor models that could lead to the attenuation of their immunosuppressive functions.


Asunto(s)
Diferenciación Celular , Células Supresoras de Origen Mieloide/patología , Neoplasias/patología , Animales , Anticuerpos Monoclonales/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Supresoras de Origen Mieloide/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Tretinoina/farmacología
8.
Cell Immunol ; 359: 104254, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33296753

RESUMEN

Myeloid-derived suppressor cells (MDSC) are generated during tumor progression and suppress the anti-tumor functions of T and natural killer (NK) cells. Their enrichment is associated with a bad prognosis and a worse outcome of immunotherapy in cancer patients. The cytokine interleukin (IL)-6 was found to be a crucial regulator of MDSC accumulation and activation as well as a factor, stimulating tumor cell proliferation, survival, invasiveness and metastasis. Accordingly, IL-6 can serve as a negative prognostic marker in cancer. On the other hand, this cytokine is also involved in T cell activation. This review discusses the pleiotropic effects of IL-6 on immune cell populations that are critical for tumor development, such as MDSC and T cells, and summarizes the data on targeting IL-6 or IL-6 receptor (IL-6R) for tumor immunotherapy to block MDSC-mediated immunosuppression in cancer patients.


Asunto(s)
Inmunoterapia/métodos , Interleucina-6/metabolismo , Células Supresoras de Origen Mieloide/inmunología , Proliferación Celular/efectos de los fármacos , Citocinas/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Terapia de Inmunosupresión/métodos , Inmunoterapia/tendencias , Interleucina-6/inmunología , Interleucina-6/uso terapéutico , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Pronóstico , Receptores de Interleucina-6/inmunología , Receptores de Interleucina-6/metabolismo , Linfocitos T/inmunología
9.
Int J Mol Sci ; 21(17)2020 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-32878277

RESUMEN

Extracellular vesicles (EV) can carry proteins, RNA and DNA, thus serving as communication tools between cells. Tumor cells secrete EV, which can be taken up by surrounding cells in the tumor microenvironment as well as by cells in distant organs. Tumor-derived EV (TEV) contain factors induced by tumor-associated hypoxia such as heat shock proteins or a variety of microRNA (miRNA). The interaction of TEV with tumor and host cells can promote cancer angiogenesis, invasion and metastasis. Myeloid cells are widely presented in tissues, comprise the majority of immune cells and play an essential role in immune reactions and tissue remodeling. However, in cancer, the differentiation of myeloid cells and their functions are impaired, resulting in tumor promotion. Such alterations are due to chronic inflammatory conditions associated with cancer and are mediated by the tumor secretome, including TEV. A high capacity of myeloid cells to clear EV from circulation put them in the central position in EV-mediated formation of pre-metastatic niches. The exposure of myeloid cells to TEV could trigger numerous signaling pathways. Progenitors of myeloid cells alter their differentiation upon the contact with TEV, resulting in the generation of myeloid-derived suppressor cells (MDSC), inhibiting anti-tumor function of T and natural killer (NK) cells and promoting thereby tumor progression. Furthermore, TEV can augment MDSC immunosuppressive capacity. Different subsets of mature myeloid cells such as monocytes, macrophages, dendritic cells (DC) and granulocytes take up TEV and acquire a protumorigenic phenotype. However, the delivery of tumor antigens to DC by TEV was shown to enhance their immunostimulatory capacity. The present review will discuss a diverse and complex EV-mediated crosstalk between tumor and myeloid cells in the context of the tumor type, TEV-associated cargo molecules and type of recipient cells.


Asunto(s)
Vesículas Extracelulares/inmunología , Tolerancia Inmunológica/inmunología , Células Asesinas Naturales/inmunología , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/inmunología , Microambiente Tumoral/inmunología , Animales , Diferenciación Celular , Vesículas Extracelulares/patología , Humanos , Células Asesinas Naturales/patología , Células Supresoras de Origen Mieloide/patología , Neoplasias/patología
10.
J Immunother Cancer ; 8(2)2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32788238

RESUMEN

BACKGROUND: Myeloid-derived suppressor cells (MDSC) play a major role in the immunosuppressive melanoma microenvironment. They are generated under chronic inflammatory conditions characterized by the constant production of inflammatory cytokines, chemokines and growth factors, including IL-6. Recruitment of MDSC to the tumor is mediated by the interaction between chemokines and chemokine receptors, in particular C-C chemokine receptor (CCR)5. Here, we studied the mechanisms of CCR5 upregulation and increased immunosuppressive function of CCR5+ MDSC. METHODS: The immortalized myeloid suppressor cell line MSC-2, primary immature myeloid cells and in vitro differentiated MDSC were used to determine factors and molecular mechanisms regulating CCR5 expression and immunosuppressive markers at the mRNA and protein levels. The relevance of the identified pathways was validated on the RET transgenic mouse melanoma model, which was also used to target the identified pathways in vivo. RESULTS: IL-6 upregulated the expression of CCR5 and arginase 1 in MDSC by a STAT3-dependent mechanism. MDSC differentiated in the presence of IL-6 strongly inhibited CD8+ T cell functions compared with MDSC differentiated without IL-6. A correlation between IL-6 levels, phosphorylated STAT3 and CCR5 expression in tumor-infiltrating MDSC was demonstrated in the RET transgenic melanoma mouse model. Surprisingly, IL-6 overexpressing tumors grew significantly slower in mice accompanied by CD8+ T cell activation. Moreover, transgenic melanoma-bearing mice treated with IL-6 blocking antibodies showed significantly accelerated tumor development. CONCLUSION: Our in vitro and ex vivo findings demonstrated that IL-6 induced CCR5 expression and a strong immunosuppressive activity of MDSC, highlighting this cytokine as a promising target for melanoma immunotherapy. However, IL-6 blocking therapy did not prove to be effective in RET transgenic melanoma-bearing mice but rather aggravated tumor progression. Further studies are needed to identify particular combination therapies, cancer entities or patient subsets to benefit from the anti-IL-6 treatment.


Asunto(s)
Interleucina-6/metabolismo , Melanoma Experimental/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Receptores CCR5/metabolismo , Animales , Humanos , Ratones
11.
Int J Mol Sci ; 21(7)2020 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-32235439

RESUMEN

Although melanoma is one of the most immunogenic tumors, it has an ability to evade anti-tumor immune responses by exploiting tolerance mechanisms, including negative immune checkpoint molecules. The most extensively studied checkpoints represent cytotoxic T lymphocyte-associated protein-4 (CTLA-4) and programmed cell death protein 1 (PD-1). Immune checkpoint inhibitors (ICI), which were broadly applied for melanoma treatment in the past decade, can unleash anti-tumor immune responses and result in melanoma regression. Patients responding to the ICI treatment showed long-lasting remission or disease control status. However, a large group of patients failed to respond to this therapy, indicating the development of resistance mechanisms. Among them are intrinsic tumor properties, the dysfunction of effector cells, and the generation of immunosuppressive tumor microenvironment (TME). This review discusses achievements of ICI treatment in melanoma, reasons for its failure, and promising approaches for overcoming the resistance. These methods include combinations of different ICI with each other, strategies for neutralizing the immunosuppressive TME and combining ICI with other anti-cancer therapies such as radiation, oncolytic viral, or targeted therapy. New therapeutic approaches targeting other immune checkpoint molecules are also discussed.


Asunto(s)
Inmunoterapia/métodos , Melanoma/terapia , Animales , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/inmunología , Humanos , Tolerancia Inmunológica , Melanoma/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Escape del Tumor , Microambiente Tumoral
12.
Oncoimmunology ; 9(1): 1727116, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32117594

RESUMEN

Eosinophils have been identified as a prognostic marker in immunotherapy of melanoma and suggested to contribute to anti-tumor host defense. However, the influence of immune checkpoint inhibitors (ICI) on the eosinophil population is poorly studied. Here, we applied routine laboratory tests, multicolor flow cytometry, RNA microarray analysis, and bio-plex assay to analyze circulating eosinophils and related serum inflammatory factors in 32 patients treated with pembrolizumab or the combination of nivolumab and ipilimumab. We demonstrated that clinical responses to ICI treatment were associated with an eosinophil accumulation in the peripheral blood. Moreover, immunotherapy led to the alteration of the eosinophil genetic and activation profile. Elevated serum concentrations of IL-16 during ICI treatment were found to be associated with increased frequencies of eosinophils in the peripheral blood. Using immunohistochemistry, we observed an enhanced eosinophil degranulation and a positive correlation between eosinophil and CD8+ T cell infiltration of tumor tissues from melanoma patients treated with ICI. Our findings highlight additional mechanisms of ICI effects and suggest the level of eosinophils as a novel predictive marker for melanoma patients who may benefit from this immunotherapy.


Asunto(s)
Eosinófilos , Melanoma , Humanos , Inhibidores de Puntos de Control Inmunológico , Ipilimumab/uso terapéutico , Melanoma/tratamiento farmacológico , Nivolumab/uso terapéutico
13.
J Clin Invest ; 129(10): 4080-4082, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31483288

RESUMEN

Recent work demonstrated a role for myeloid-derived suppressor cells (MDSCs) in the antimicrobial response in newborns, but the signals guiding their differentiation remained unknown. In this issue of the JCI, Liu et al. demonstrate that lactoferrin (LF) converts newborn neutrophils and monocytes to MDSCs via the low-density lipoprotein receptor-related protein-2 (LRP2) receptor and NF-κB activation. Due to their strong antimicrobial activity, adoptive transfer of MDSCs generated by in vitro culture with LF prolonged the survival of newborn mice with necrotizing enterocolitis, a severe pathology in preterm infants. These findings indicate a surprising protective role of MDSCs in newborns and demonstrate the potential of MDSC therapy for the treatment of infants with diseases associated with deregulated inflammation.


Asunto(s)
Infecciones , Células Supresoras de Origen Mieloide , Animales , Animales Recién Nacidos , Humanos , Recién Nacido , Recien Nacido Prematuro , Lactoferrina , Ratones
14.
Cancer Res ; 79(18): 4715-4728, 2019 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-31337655

RESUMEN

Tumor cell-derived extracellular vesicles (EV) convert normal myeloid cells into myeloid-derived suppressor cells (MDSC), inhibiting antitumor immune responses. Here, we show that EV from Ret mouse melanoma cells upregulate the expression of programmed cell death ligand 1 (PD-L1) on mouse immature myeloid cells (IMC), leading to suppression of T-cell activation. PD-L1 expression and the immunosuppressive potential of EV-generated MDSC were dependent on the expression of Toll-like receptors (TLR). IMC from Tlr4-/- mice failed to increase T-cell PD-L1 expression and immunosuppression with Ret-EV treatment, and this effect was dependent on heat-shock protein 86 (HSP86) as HSP86-deficient Ret cells could not stimulate PD-L1 expression on normal IMC; IMC from Tlr2-/- and Tlr7-/- mice demonstrated similar results, although to a lesser extent. HSP86-deficient Ret cells slowed tumor progression in vivo associated with decreased frequency of tumor-infiltrating PD-L1+CD11b+Gr1+ MDSC. EV from human melanoma cells upregulated PD-L1 and immunosuppression of normal monocytes dependent on HSP86. These findings highlight a novel EV-mediated mechanism of MDSC generation from normal myeloid cells, suggesting the importance of EV targeting for tumor therapy. SIGNIFICANCE: These findings validate the importance of TLR4 signaling in reprogramming normal myeloid cells into functional myeloid-derived suppressor cells.


Asunto(s)
Antígeno B7-H1/metabolismo , Vesículas Extracelulares/patología , Inmunosupresores/inmunología , Melanoma/patología , Células Supresoras de Origen Mieloide/inmunología , Receptor Toll-Like 4/fisiología , Receptor Toll-Like 7/fisiología , Animales , Células Cultivadas , Vesículas Extracelulares/metabolismo , Humanos , Tolerancia Inmunológica , Inmunosupresores/metabolismo , Activación de Linfocitos , Melanoma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/patología
15.
Br J Cancer ; 120(1): 16-25, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30413826

RESUMEN

Under steady-state conditions, bone marrow-derived immature myeloid cells (IMC) differentiate into granulocytes, macrophages and dendritic cells (DCs). This differentiation is impaired under chronic inflammatory conditions, which are typical for tumour progression, leading to the accumulation of IMCs. These cells are capable of inducing strong immunosuppressive effects through the expression of various cytokines and immune regulatory molecules, inhibition of lymphocyte homing, stimulation of other immunosuppressive cells, depletion of metabolites critical for T cell functions, expression of ectoenzymes regulating adenosine metabolism, and the production of reactive species. IMCs are therefore designated as myeloid-derived suppressor cells (MDSCs), and have been shown to accumulate in tumour-bearing mice and cancer patients. MDSCs are considered to be a strong contributor to the immunosuppressive tumour microenvironment and thus an obstacle for many cancer immunotherapies. Consequently, numerous studies are focused on the characterisation of MDSC origin and their relationship to other myeloid cell populations, their immunosuppressive capacity, and possible ways to inhibit MDSC function with different approaches being evaluated in clinical trials. This review analyses the current state of knowledge on the origin and function of MDSCs in cancer, with a special emphasis on the immunosuppressive pathways pursued by MDSCs to inhibit T cell functions, resulting in tumour progression. In addition, we describe therapeutic strategies and clinical benefits of MDSC targeting in cancer.


Asunto(s)
Inmunoterapia , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/terapia , Microambiente Tumoral/inmunología , Animales , Diferenciación Celular/genética , Humanos , Tolerancia Inmunológica , Terapia de Inmunosupresión/métodos , Macrófagos/inmunología , Macrófagos/patología , Ratones , Neoplasias/inmunología , Linfocitos T/inmunología
16.
PLoS Biol ; 16(7): e2005345, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30011270

RESUMEN

Cell motility is essential for protozoan and metazoan organisms and typically relies on the dynamic turnover of actin filaments. In metazoans, monomeric actin polymerises into usually long and stable filaments, while some protozoans form only short and highly dynamic actin filaments. These different dynamics are partly due to the different sets of actin regulatory proteins and partly due to the sequence of actin itself. Here we probe the interactions of actin subunits within divergent actin filaments using a comparative dynamic molecular model and explore their functions using Plasmodium, the protozoan causing malaria, and mouse melanoma derived B16-F1 cells as model systems. Parasite actin tagged to a fluorescent protein (FP) did not incorporate into mammalian actin filaments, and rabbit actin-FP did not incorporate into parasite actin filaments. However, exchanging the most divergent region of actin subdomain 3 allowed such reciprocal incorporation. The exchange of a single amino acid residue in subdomain 2 (N41H) of Plasmodium actin markedly improved incorporation into mammalian filaments. In the parasite, modification of most subunit-subunit interaction sites was lethal, whereas changes in actin subdomains 1 and 4 reduced efficient parasite motility and hence mosquito organ penetration. The strong penetration defects could be rescued by overexpression of the actin filament regulator coronin. Through these comparative approaches we identified an essential and common contributor, subdomain 3, which drives the differential dynamic behaviour of two highly divergent eukaryotic actins in motile cells.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Mamíferos/metabolismo , Plasmodium falciparum/metabolismo , Subunidades de Proteína/metabolismo , Citoesqueleto de Actina/química , Actinas/química , Actinas/metabolismo , Alelos , Animales , Femenino , Estadios del Ciclo de Vida , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Mutación/genética , Parásitos/crecimiento & desarrollo , Fenotipo , Plasmodium falciparum/crecimiento & desarrollo , Unión Proteica , Dominios Proteicos , Subunidades de Proteína/química , Conejos , Especificidad de la Especie , Esporozoítos/metabolismo
17.
Front Immunol ; 9: 1310, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29942309

RESUMEN

Immune checkpoint inhibitors (ICI) used for cancer immunotherapy were shown to boost the existing anti-tumor immune response by preventing the inhibition of T cells by tumor cells. Antibodies targeting two negative immune checkpoint pathways, namely cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), and programmed cell death-ligand 1 (PD-L1), have been approved first for patients with melanoma, squamous non-small cell lung cancer (NSCLC), and renal cell carcinoma. Clinical trials are ongoing to verify the efficiency of these antibodies for other cancer types and to evaluate strategies to block other checkpoint molecules. However, a number of patients do not respond to this treatment possibly due to profound immunosuppression, which is mediated partly by myeloid-derived suppressor cells (MDSC). This heterogeneous population of immature myeloid cells can strongly inhibit anti-tumor activities of T and NK cells and stimulate regulatory T cells (Treg), leading to tumor progression. Moreover, MDSC can contribute to patient resistance to immune checkpoint inhibition. Accumulating evidence demonstrates that the frequency and immunosuppressive function of MDSC in cancer patients can be used as a predictive marker for therapy response. This review focuses on the role of MDSC in immune checkpoint inhibition and provides an analysis of combination strategies for MDSC targeting together with ICI to improve their therapeutic efficiency in cancer patients.

18.
Front Immunol ; 9: 398, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29552012

RESUMEN

The immune system has many sophisticated mechanisms to balance an extensive immune response. Distinct immunosuppressive cells could protect from excessive tissue damage and autoimmune disorders. Tumor cells take an advantage of those immunosuppressive mechanisms and establish a strongly immunosuppressive tumor microenvironment (TME), which inhibits antitumor immune responses, supporting the disease progression. Myeloid-derived suppressor cells (MDSC) play a crucial role in this immunosuppressive TME. Those cells represent a heterogeneous population of immature myeloid cells with a strong immunosuppressive potential. They inhibit an antitumor reactivity of T cells and NK cells. Furthermore, they promote angiogenesis, establish pre-metastatic niches, and recruit other immunosuppressive cells such as regulatory T cells. Accumulating evidences demonstrated that the enrichment and activation of MDSC correlated with tumor progression, recurrence, and negative clinical outcome. In the last few years, various preclinical studies and clinical trials targeting MDSC showed promising results. In this review, we discuss different therapeutic approaches on MDSC targeting to overcome immunosuppressive TME and enhance the efficiency of current tumor immunotherapies.


Asunto(s)
Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/terapia , Linfocitos T/inmunología , Animales , Ensayos Clínicos como Asunto , Humanos , Tolerancia Inmunológica , Activación de Linfocitos , Neoplasias/inmunología , Escape del Tumor , Microambiente Tumoral
19.
Cancer Res ; 78(1): 157-167, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29089297

RESUMEN

Accumulation of myeloid-derived suppressor cells (MDSC) in melanoma microenvironment is supported by chemokine receptor/chemokine signaling. Although different chemokines were suggested to be involved in this process, the role of CCR5 and its ligands is not established. Using a Ret transgenic mouse melanoma model, we found an accumulation of CCR5+ MDSCs in melanoma lesions associated with both increased concentrations of CCR5 ligands and tumor progression. Tumor-infiltrating CCR5+ MDSCs displayed higher immunosuppressive activity than their CCR5- counterparts. Upregulation of CCR5 expression on CD11b+Gr1+ myeloid cells was induced in vitro by CCR5 ligands and other inflammatory factors. In melanoma patients, CCR5+ MDSCs were enriched at the tumor site and correlated with enhanced production of CCR5 ligands. Moreover, they exhibited a stronger immunosuppressive pattern compared with CCR5- MDSCs. Blocking CCR5/CCR5 ligand interactions increased survival of tumor-bearing mice and was associated with reduced migration and immunosuppressive potential of MDSCs in tumor lesions. Our findings define a critical role for CCR5 in recruitment and activation of MDSCs, suggesting a novel strategy for melanoma treatment.Significance: These findings validate the importance of the CCR5/CCR5 ligand axis not only for MDSC recruitment but also for further activation of their immunosuppressive functions in the tumor microenvironment, with potentially broad therapeutic implications, given existing clinically available inhibitors of this axis. Cancer Res; 78(1); 157-67. ©2017 AACR.


Asunto(s)
Melanoma/inmunología , Células Supresoras de Origen Mieloide/patología , Receptores CCR5/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Movimiento Celular , Femenino , Humanos , Masculino , Melanoma/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Proteínas Proto-Oncogénicas c-ret/genética , Microambiente Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...