Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 14: 1060258, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37398649

RESUMEN

The skin is the body's largest organ. It serves as a barrier to pathogen entry and the first site of immune defense. In the event of a skin injury, a cascade of events including inflammation, new tissue formation and tissue remodeling contributes to wound repair. Skin-resident and recruited immune cells work together with non-immune cells to clear invading pathogens and debris, and guide the regeneration of damaged host tissues. Disruption to the wound repair process can lead to chronic inflammation and non-healing wounds. This, in turn, can promote skin tumorigenesis. Tumors appropriate the wound healing response as a way of enhancing their survival and growth. Here we review the role of resident and skin-infiltrating immune cells in wound repair and discuss their functions in regulating both inflammation and development of skin cancers.


Asunto(s)
Piel , Cicatrización de Heridas , Humanos , Piel/patología , Inflamación , Carcinogénesis/patología
2.
Kidney Int ; 103(6): 1105-1119, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37097268

RESUMEN

Nuclear factor κB (NF-κB) activation is a deleterious molecular mechanism that drives acute kidney injury (AKI) and manifests in transplanted kidneys as delayed graft function. The TNFAIP3 gene encodes A20, a cytoplasmic ubiquitin ligase and a master negative regulator of the NF- κB signaling pathway. Common population-specific TNFAIP3 coding variants that reduce A20's enzyme function and increase NF- κB activation have been linked to heightened protective immunity and autoimmune disease, but have not been investigated in AKI. Here, we functionally identified a series of unique human TNFAIP3 coding variants linked to the autoimmune genome-wide association studies single nucleotide polymorphisms of F127C; namely F127C;R22Q, F127C;G281E, F127C;W448C and F127C;N449K that reduce A20's anti-inflammatory function in an NF- κB reporter assay. To investigate the impact of TNFAIP3 hypomorphic coding variants in AKI we tested a mouse Tnfaip3 hypomorph in a model of ischemia reperfusion injury (IRI). The mouse Tnfaip3 coding variant I325N increases NF- κB activation without overt inflammatory disease, providing an immune boost as I325N mice exhibit enhanced innate immunity to a bacterial challenge. Surprisingly, despite exhibiting increased intra-kidney NF- κB activation with inflammation in IRI, the kidney of I325N mice was protected. The I325N variant influenced the outcome of IRI by changing the dynamic expression of multiple cytoprotective mechanisms, particularly by increasing NF- κB-dependent anti-apoptotic factors BCL-2, BCL-XL, c-FLIP and A20, altering the active redox state of the kidney with a reduction of superoxide levels and the enzyme super oxide dismutase-1, and enhancing cellular protective mechanisms including increased Foxp3+ T cells. Thus, TNFAIP3 gene variants represent a kidney and population-specific molecular factor that can dictate the course of IRI.


Asunto(s)
Lesión Renal Aguda , FN-kappa B , Humanos , Ratones , Animales , FN-kappa B/metabolismo , Factores de Transcripción/genética , Ubiquitina , Estudio de Asociación del Genoma Completo , Ligasas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Lesión Renal Aguda/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética
3.
Diabetes ; 72(6): 758-768, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36929171

RESUMEN

Intrahepatic islet transplantation for type 1 diabetes is limited by the need for multiple infusions and poor islet viability posttransplantation. The development of alternative transplantation sites is necessary to improve islet survival and facilitate monitoring and retrieval. We tested a clinically proven biodegradable temporizing matrix (BTM), a polyurethane-based scaffold, to generate a well-vascularized intracutaneous "neodermis" within the skin for islet transplantation. In murine models, BTM did not impair syngeneic islet renal-subcapsular transplant viability or function, and it facilitated diabetes cure for over 150 days. Furthermore, BTM supported functional neonatal porcine islet transplants into RAG-1-/- mice for 400 days. Hence, BTM is nontoxic for islets. Two-photon intravital imaging used to map vessel growth through time identified dense vascular networks, with significant collagen deposition and increases in vessel mass up to 30 days after BTM implantation. In a preclinical porcine skin model, BTM implants created a highly vascularized intracutaneous site by day 7 postimplantation. When syngeneic neonatal porcine islets were transplanted intracutaneously, the islets remained differentiated as insulin-producing cells, maintained normal islet architecture, secreted c-peptide, and survived for over 100 days. Here, we show that BTM facilitates formation of an islet-supportive intracutaneous neodermis in a porcine preclinical model, as an alternative islet-transplant site. ARTICLE HIGHLIGHTS: Human and porcine pancreatic islets were transplanted into a fully vascularized biodegradable temporizing matrix (Novosorb) that creates a unique intracutaneous site outside of the liver in a large-animal preclinical model. The intracutaneous prevascularized site supported pancreatic islet survival for 3 months in a syngeneic porcine-transplant model. Pancreatic (human and porcine) islet survival and function were demonstrated in an intracutaneous site outside of the liver for the first time in a large-animal preclinical model.


Asunto(s)
Diabetes Mellitus Tipo 1 , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Porcinos , Humanos , Animales , Ratones , Trasplante de Islotes Pancreáticos/métodos , Supervivencia de Injerto , Islotes Pancreáticos/irrigación sanguínea , Diabetes Mellitus Tipo 1/cirugía , Colágeno
4.
Cancer Res ; 83(8): 1315-1328, 2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-36787115

RESUMEN

The inflammatory microenvironment of solid tumors creates a protumorigenic milieu that resembles chronic inflammation akin to a subverted wound healing response. Here, we investigated the effect of converting the tumor microenvironment from a chronically inflamed state to one of acute microbial inflammation by injecting microbial bioparticles directly into tumors. Intratumoral microbial bioparticle injection led to rapid and dramatic changes in the tumor immune composition, the most striking of which was a substantial increase in the presence of activated neutrophils. In situ photoconversion and intravital microscopy indicated that tumor neutrophils transiently switched from sessile producers of VEGF to highly motile neutrophils that clustered to make neutrophil-rich domains in the tumor. The neutrophil clusters remodeled tumor tissue and repressed tumor growth. Single-cell transcriptional analysis of microbe-stimulated neutrophils showed a profound shift in gene expression towards heightened activation and antimicrobial effector function. Microbe-activated neutrophils also upregulated chemokines known to regulate neutrophil and CD8+ T-cell recruitment. Microbial therapy also boosted CD8+ T-cell function and enhanced the therapeutic benefit of checkpoint inhibitor therapy in tumor-bearing mice and provided protection in a model of tumor recurrence. These data indicate that one of the major effector mechanisms of microbial therapy is the conversion of tumor neutrophils from a wound healing to an acutely activated cytotoxic phenotype, highlighting a rationale for broader deployment of microbial therapy in the treatment of solid cancers. SIGNIFICANCE: Intratumoral injection of microbial bioparticles stimulates neutrophil antitumor functions, suggesting pathways for optimizing efficacy of microbial therapies and paving the way for their broader utilization in the clinic.


Asunto(s)
Neoplasias , Neutrófilos , Ratones , Animales , Neutrófilos/metabolismo , Neoplasias/genética , Neoplasias/terapia , Neoplasias/metabolismo , Linfocitos T CD8-positivos , Inflamación/patología , Fenotipo , Infiltración Neutrófila , Microambiente Tumoral
5.
Nature ; 608(7924): 757-765, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35948641

RESUMEN

The notion that mobile units of nucleic acid known as transposable elements can operate as genomic controlling elements was put forward over six decades ago1,2. However, it was not until the advancement of genomic sequencing technologies that the abundance and repertoire of transposable elements were revealed, and they are now known to constitute up to two-thirds of mammalian genomes3,4. The presence of DNA regulatory regions including promoters, enhancers and transcription-factor-binding sites within transposable elements5-8 has led to the hypothesis that transposable elements have been co-opted to regulate mammalian gene expression and cell phenotype8-14. Mammalian transposable elements include recent acquisitions and ancient transposable elements that have been maintained in the genome over evolutionary time. The presence of ancient conserved transposable elements correlates positively with the likelihood of a regulatory function, but functional validation remains an essential step to identify transposable element insertions that have a positive effect on fitness. Here we show that CRISPR-Cas9-mediated deletion of a transposable element-namely the LINE-1 retrotransposon Lx9c11-in mice results in an exaggerated and lethal immune response to virus infection. Lx9c11 is critical for the neogenesis of a non-coding RNA (Lx9c11-RegoS) that regulates genes of the Schlafen family, reduces the hyperinflammatory phenotype and rescues lethality in virus-infected Lx9c11-/- mice. These findings provide evidence that a transposable element can control the immune system to favour host survival during virus infection.


Asunto(s)
Elementos Transponibles de ADN , Interacciones Microbiota-Huesped , Inmunidad , Retroelementos , Virosis , Animales , Sistemas CRISPR-Cas/genética , Elementos Transponibles de ADN/genética , Elementos Transponibles de ADN/inmunología , Evolución Molecular , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Inmunidad/genética , Ratones , ARN no Traducido/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Retroelementos/genética , Retroelementos/inmunología , Virosis/genética , Virosis/inmunología
6.
Cancer Immunol Immunother ; 71(12): 3099-3106, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35624180

RESUMEN

Near-infrared photoimmunotherapy (NIR-PIT) selectively kills tumor cells to which the photo-absorber dye IR700DX-conjugated antibodies are bound and induces a systemic anti-tumor immune response. NIR-PIT induces immunogenic cell death (ICD), releases damage-associated molecular patterns (DAMPs) molecules from dying tumor cells, and activates dendritic cells (DCs). However, it is unclear whether NIR-PIT affects migration of tumor-infiltrating (Ti)-DCs to draining lymph nodes (dLNs), where a systemic anti-tumor response is induced. Here, we utilized in vivo photolabeling of Ti-DCs in tumors in photoconvertible protein Kikume Green-Red (KikGR) mice to show that NIR-PIT enhanced migration of Ti-DCs including cDC1s, cDC2s, and CD326+ DCs to dLNs. This effect was abolished by blocking adenosine triphosphate (ATP), one of the DAMPs molecules, as well as by inhibition of Gαi signaling by pertussis toxin. Thus, ICD induction by NIR-PIT stimulates Ti-DC migration to dLNs via ATP-P2X7 receptor and Gαi protein-coupled receptor signaling pathways and may augment tumor antigen presentation to induce anti-tumor T cells in dLNs.


Asunto(s)
Inmunoterapia , Receptores Purinérgicos P2X7 , Ratones , Animales , Toxina del Pertussis , Línea Celular Tumoral , Ratones Desnudos , Muerte Celular Inmunogénica , Células Dendríticas , Adenosina Trifosfato , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cells ; 10(8)2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34440875

RESUMEN

The lymphatic system is a complex network of lymphatic vessels and lymph nodes designed to balance fluid homeostasis and facilitate host immune defence. Neutrophils are rapidly recruited to sites of inflammation to provide the first line of protection against microbial infections. The traditional view of neutrophils as short-lived cells, whose role is restricted to providing sterilizing immunity at sites of infection, is rapidly evolving to include additional functions at the interface between the innate and adaptive immune systems. Neutrophils travel via the lymphatics from the site of inflammation to transport antigens to lymph nodes. They can also enter lymph nodes from the blood by crossing high endothelial venules. Neutrophil functions in draining lymph nodes include pathogen control and modulation of adaptive immunity. Another facet of neutrophil interactions with the lymphatic system is their ability to promote lymphangiogenesis in draining lymph nodes and inflamed tissues. In this review, we discuss the significance of neutrophil migration to secondary lymphoid organs and within the lymphatic vasculature and highlight emerging evidence of the neutrophils' role in lymphangiogenesis.


Asunto(s)
Sistema Linfático/inmunología , Neutrófilos/inmunología , Inmunidad Adaptativa , Animales , Movimiento Celular , Humanos , Inflamación , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Linfangiogénesis , Sistema Linfático/metabolismo , Vasos Linfáticos/inmunología , Vasos Linfáticos/metabolismo , Neutrófilos/metabolismo
8.
J Immunol ; 207(3): 771-776, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34290103

RESUMEN

Tumor-infiltrating regulatory T cells (Tregs) have been extensively studied as therapeutic targets. However, not all infiltrating T cells exert their functions equally, presumably because of their heterogeneity and substantial turnover in tissues. In this study, we hypothesized that intertissue migration underlies the functional heterogeneity of Tregs. To test this, we applied in vivo photolabeling to examine single-cell diversity of immunosuppressive molecules in mouse Tregs migrating to, remaining in, and emigrating from MC38 tumors. Neuropilin-1 (Nrp1) expression was inversely correlated with that of six other molecules associated with Treg function. Unsupervised clustering analyses revealed that clusters containing Tregs that were retained in tumors expressed high levels of the six functional molecules but not of Nrp1. However, these clusters represented only half of the Tregs migrating to the tumor, suggesting evolving heterogeneity of tumor-infiltrating Tregs. Thus, we propose progressive pathways of Treg activation and migration between tumors and draining lymph nodes.


Asunto(s)
Adenocarcinoma/inmunología , Neoplasias del Colon/inmunología , Factores de Transcripción Forkhead/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Análisis de la Célula Individual/métodos , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Línea Celular Tumoral , Movimiento Celular , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/genética , Humanos , Activación de Linfocitos , Ratones , Ratones Transgénicos , Neoplasias Experimentales , Neuropilina-1/genética , Neuropilina-1/metabolismo , Fenotipo
9.
iScience ; 24(5): 102424, 2021 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-33997702

RESUMEN

Immunogenic tumor cell death enhances anti-tumor immunity. However, the mechanisms underlying this effect are incompletely understood. We established a system to induce tumor cell death in situ and investigated its effect on dendritic cell (DC) migration and T cell responses using intravital photolabeling in mice expressing KikGR photoconvertible protein. We demonstrate that tumor cell death induces phagocytosis of tumor cells by tumor-infiltrating (Ti)-DCs, and HMGB1-TLR4 and ATP-P2X7 receptor signaling-dependent Ti-DC emigration to draining lymph nodes (dLNs). This led to an increase in anti-tumor CD8+ T cells of memory precursor effector phenotype and secondary tumor growth inhibition in a CD103+ DC-dependent manner. However, combining tumor cell death induction with lipopolysaccharide treatment stimulated Ti-DC maturation and emigration to dLNs but did not improve tumor immunity. Thus, immunogenic tumor cell death enhances tumor immunity by increasing Ti-DC migration to dLNs where they promote anti-tumor T cell responses and tumor growth inhibition.

10.
Sci Rep ; 10(1): 19085, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33154446

RESUMEN

Germline loss-of-function variation in TNFAIP3, encoding A20, has been implicated in a wide variety of autoinflammatory and autoimmune conditions, with acquired somatic missense mutations linked to cancer progression. Furthermore, human sequence data reveals that the A20 locus contains ~ 400 non-synonymous coding variants, which are largely uncharacterised. The growing number of A20 coding variants with unknown function, but potential clinical impact, poses a challenge to traditional mouse-based approaches. Here we report the development of a novel functional genomics approach that utilizes a new A20-deficient zebrafish (Danio rerio) model to investigate the impact of TNFAIP3 genetic variants in vivo. A20-deficient zebrafish are hyper-responsive to microbial immune activation and exhibit spontaneous early lethality. Ectopic addition of human A20 rescued A20-null zebrafish from lethality, while missense mutations at two conserved A20 residues, S381A and C243Y, reversed this protective effect. Ser381 represents a phosphorylation site important for enhancing A20 activity that is abrogated by its mutation to alanine, or by a causal C243Y mutation that triggers human autoimmune disease. These data reveal an evolutionarily conserved role for TNFAIP3 in limiting inflammation in the vertebrate linage and show how this function is controlled by phosphorylation. They also demonstrate how a zebrafish functional genomics pipeline can be utilized to investigate the in vivo significance of medically relevant human TNFAIP3 gene variants.


Asunto(s)
Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/fisiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología , Pez Cebra/genética , Sustitución de Aminoácidos , Animales , Animales Modificados Genéticamente , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/genética , Secuencia Conservada , Evolución Molecular , Variación Genética , Humanos , Inflamación/etiología , Inflamación/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Modelos Animales , Modelos Genéticos , Mutación Missense , FN-kappa B/metabolismo , Fosforilación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/deficiencia , Pez Cebra/fisiología , Proteínas de Pez Cebra/deficiencia
11.
Cell Immunol ; 350: 103898, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-30712753

RESUMEN

Neutrophils are the first cellular responders of the immune system. They employ their impressive arsenal of microbicidal molecules to provide rapid and efficient defense against pathogens. However, the role of neutrophils extends far beyond microbial destruction to include tissue repair and remodeling, provision of signals to the adaptive immune system and body homeostasis. Intravital imaging has allowed the visualization of neutrophils in their native environment in both health and disease and provided crucial insights into their mechanisms of action. In the last few years the power of intravital imaging has been considerably extended by the introduction of photoconvertible proteins and intracellular signaling reporter mice. This review will highlight recent advances in our understanding of neutrophil biology based on the use of intravital microscopy to visualize their modus operandi in vivo including migration in and out of inflamed tissues, host-pathogen interactions and cell fate.


Asunto(s)
Microscopía Intravital/métodos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Animales , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Sistema Inmunológico/inmunología , Inmunidad Innata/inmunología , Inflamación/patología , Ratones , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Infiltración Neutrófila/inmunología
12.
Front Immunol ; 10: 1168, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31191539

RESUMEN

Lymphatic vessels collect interstitial fluid that has extravasated from blood vessels and return it to the circulatory system. Another important function of the lymphatic network is to facilitate immune cell migration and antigen transport from the periphery to draining lymph nodes. This migration plays a crucial role in immune surveillance, initiation of immune responses and tolerance. Here we discuss the significance and mechanisms of lymphatic migration of innate and adaptive immune cells in homeostasis, inflammation and cancer.


Asunto(s)
Movimiento Celular , Sistema Inmunológico/citología , Sistema Inmunológico/fisiología , Vasos Linfáticos , Inmunidad Adaptativa , Animales , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Susceptibilidad a Enfermedades , Humanos , Inmunidad Innata , Neutrófilos/citología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
13.
Front Immunol ; 10: 386, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30899263

RESUMEN

Recent studies of the patterns of chemokine-mediated immune cell recruitment into solid tumors have enhanced our understanding of the role played by various immune cell subsets both in amplifying and inhibiting tumor cell growth and spread. Here we discuss how the chemokine/chemokine receptor networks bring together immune cells within the microenvironment of skin tumors, particularly melanomas, including their effect on disease progression, prognosis and therapeutic options.


Asunto(s)
Quimiocinas/metabolismo , Células Dendríticas/inmunología , Leucocitos/inmunología , Macrófagos/inmunología , Melanoma/inmunología , Neoplasias Cutáneas/inmunología , Animales , Movimiento Celular/inmunología , Humanos , Ratones , Receptores de Quimiocina/metabolismo , Escape del Tumor/inmunología
14.
Cell Rep ; 21(1): 274-288, 2017 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-28978480

RESUMEN

The small GTPase RhoA is involved in a variety of fundamental processes in normal tissue. Spatiotemporal control of RhoA is thought to govern mechanosensing, growth, and motility of cells, while its deregulation is associated with disease development. Here, we describe the generation of a RhoA-fluorescence resonance energy transfer (FRET) biosensor mouse and its utility for monitoring real-time activity of RhoA in a variety of native tissues in vivo. We assess changes in RhoA activity during mechanosensing of osteocytes within the bone and during neutrophil migration. We also demonstrate spatiotemporal order of RhoA activity within crypt cells of the small intestine and during different stages of mammary gestation. Subsequently, we reveal co-option of RhoA activity in both invasive breast and pancreatic cancers, and we assess drug targeting in these disease settings, illustrating the potential for utilizing this mouse to study RhoA activity in vivo in real time.


Asunto(s)
Técnicas Biosensibles , Transferencia Resonante de Energía de Fluorescencia/métodos , Microscopía Intravital/métodos , Imagen de Lapso de Tiempo/métodos , Proteínas de Unión al GTP rho/genética , Animales , Antineoplásicos/farmacología , Huesos/citología , Huesos/metabolismo , Movimiento Celular/efectos de los fármacos , Dasatinib/farmacología , Clorhidrato de Erlotinib/farmacología , Femenino , Transferencia Resonante de Energía de Fluorescencia/instrumentación , Regulación de la Expresión Génica , Intestino Delgado/metabolismo , Intestino Delgado/ultraestructura , Microscopía Intravital/instrumentación , Glándulas Mamarias Animales/irrigación sanguínea , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/ultraestructura , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/ultraestructura , Mecanotransducción Celular , Ratones , Ratones Transgénicos , Neutrófilos/metabolismo , Neutrófilos/ultraestructura , Osteocitos/metabolismo , Osteocitos/ultraestructura , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/ultraestructura , Imagen de Lapso de Tiempo/instrumentación , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
15.
Proc Natl Acad Sci U S A ; 114(22): 5677-5682, 2017 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-28507145

RESUMEN

Immune therapy is rapidly gaining prominence in the clinic as a major weapon against cancer. Whereas much attention has been focused on the infiltration of tumors by immune cells, the subsequent fate of these infiltrates remains largely unexplored. We therefore established a photoconversion-based model that allowed us to label tumor-infiltrating immune cells and follow their migration. Using this system, we identified a population of tumor-experienced cells that emigrate from primary tumors to draining lymph nodes via afferent lymphatic vessels. Although the majority of tumor-infiltrating cells were myeloid, T cells made up the largest population of tumor-egressing leukocytes. Strikingly, the subset composition of tumor-egressing T cells was greatly skewed compared with those that had infiltrated the tumor and those resident in the draining lymph node. Some T-cell subsets such as CD8+ T cells emigrated more readily; others including CD4-CD8- T cells were preferentially retained, suggesting that specific mechanisms guide immune cell egress from tumors. Furthermore, tumor-egressing T cells were more activated and displayed enhanced effector function in comparison with their lymph node counterparts. Finally, we demonstrated that tumor-infiltrating T cells migrate to distant secondary tumors and draining lymph nodes, highlighting a mechanism whereby tumor-experienced effector T cells may mediate antitumor immunity at metastatic sites. Thus, our results provide insights into migration and function of tumor-infiltrating immune cells and the role of these cells in tumor immunity outside of primary tumor deposits.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Carcinoma Pulmonar de Lewis/inmunología , Movimiento Celular/inmunología , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos T Reguladores/inmunología , Animales , Línea Celular Tumoral , Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología
16.
Front Immunol ; 8: 309, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28382036

RESUMEN

There is ample evidence for both beneficial and harmful involvement of the immune system in tumor development and spread. Immune cell recruitment to tumors is essential not only for the success of anticancer immune therapies but also for tumor-induced immune suppression. Now that immune-based therapies are playing an increasingly important role in treatment of solid tumors such as metastatic melanomas, precise analysis of the in vivo contributions of different leukocyte subsets in tumor immunity has become an even greater priority. Recently, this goal has been markedly facilitated by the use of intravital microscopy, which has enabled us to visualize the dynamic interactions between cells of the immune system and tumor targets in the context of the tumor microenvironment. For example, intravital imaging techniques have shed new light on T cell infiltration of tumors, the mechanisms of cancer cell killing, and how myeloid cells contribute to tumor tolerance and spread. This mini-review summarizes the recent advances made to our understanding of the roles of innate and adaptive immune cells in cancer based on the use of these in vivo imaging approaches.

17.
PLoS Comput Biol ; 12(9): e1005082, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27589606

RESUMEN

The advent of two-photon microscopy now reveals unprecedented, detailed spatio-temporal data on cellular motility and interactions in vivo. Understanding cellular motility patterns is key to gaining insight into the development and possible manipulation of the immune response. Computational simulation has become an established technique for understanding immune processes and evaluating hypotheses in the context of experimental data, and there is clear scope to integrate microscopy-informed motility dynamics. However, determining which motility model best reflects in vivo motility is non-trivial: 3D motility is an intricate process requiring several metrics to characterize. This complicates model selection and parameterization, which must be performed against several metrics simultaneously. Here we evaluate Brownian motion, Lévy walk and several correlated random walks (CRWs) against the motility dynamics of neutrophils and lymph node T cells under inflammatory conditions by simultaneously considering cellular translational and turn speeds, and meandering indices. Heterogeneous cells exhibiting a continuum of inherent translational speeds and directionalities comprise both datasets, a feature significantly improving capture of in vivo motility when simulated as a CRW. Furthermore, translational and turn speeds are inversely correlated, and the corresponding CRW simulation again improves capture of our in vivo data, albeit to a lesser extent. In contrast, Brownian motion poorly reflects our data. Lévy walk is competitive in capturing some aspects of neutrophil motility, but T cell directional persistence only, therein highlighting the importance of evaluating models against several motility metrics simultaneously. This we achieve through novel application of multi-objective optimization, wherein each model is independently implemented and then parameterized to identify optimal trade-offs in performance against each metric. The resultant Pareto fronts of optimal solutions are directly contrasted to identify models best capturing in vivo dynamics, a technique that can aid model selection more generally. Our technique robustly determines our cell populations' motility strategies, and paves the way for simulations that incorporate accurate immune cell motility dynamics.


Asunto(s)
Movimiento Celular/fisiología , Leucocitos/citología , Microscopía/métodos , Modelos Biológicos , Animales , Biología Computacional , Simulación por Computador , Ratones , Ratones Endogámicos C57BL
18.
Semin Immunol ; 28(2): 129-36, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-27025975

RESUMEN

Secondary lymphoid organs provide a specialized microenvironment tailored to foster communication between cells of the innate and adaptive immune systems. These interactions allow immune cells to coordinate multilayered defense against pathogens. Until recently dendritic cells and macrophages were thought to comprise the main innate immune cell subsets responsible for delivering signals that drive the adaptive immune response, while the function of neutrophils was largely confined to the innate immune system. However, the discovery of neutrophils in lymph nodes has raised the question of whether neutrophils might play a more extensive role not only in innate immunity per se, but also in coordinating the interactions between innate and adaptive immune responses. In this review we discuss the mechanisms and consequences of neutrophil recruitment to lymph nodes and how this recruitment influences subsequent immune responses both in situ and at distant sites.


Asunto(s)
Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Inmunidad Adaptativa , Animales , Comunicación Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Susceptibilidad a Enfermedades , Humanos , Inmunidad Innata , Ganglios Linfáticos/patología , Activación Neutrófila/genética , Activación Neutrófila/inmunología , Infiltración Neutrófila/inmunología , Especificidad de Órganos/inmunología
19.
PLoS Biol ; 13(12): e1002330, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26717410

RESUMEN

During pregnancy, the ETS transcription factor ELF5 establishes the milk-secreting alveolar cell lineage by driving a cell fate decision of the mammary luminal progenitor cell. In breast cancer, ELF5 is a key transcriptional determinant of tumor subtype and has been implicated in the development of insensitivity to anti-estrogen therapy. In the mouse mammary tumor virus-Polyoma Middle T (MMTV-PyMT) model of luminal breast cancer, induction of ELF5 levels increased leukocyte infiltration, angiogenesis, and blood vessel permeability in primary tumors and greatly increased the size and number of lung metastasis. Myeloid-derived suppressor cells, a group of immature neutrophils recently identified as mediators of vasculogenesis and metastasis, were recruited to the tumor in response to ELF5. Depletion of these cells using specific Ly6G antibodies prevented ELF5 from driving vasculogenesis and metastasis. Expression signatures in luminal A breast cancers indicated that increased myeloid cell invasion and inflammation were correlated with ELF5 expression, and increased ELF5 immunohistochemical staining predicted much shorter metastasis-free and overall survival of luminal A patients, defining a group who experienced unexpectedly early disease progression. Thus, in the MMTV-PyMT mouse mammary model, increased ELF5 levels drive metastasis by co-opting the innate immune system. As ELF5 has been previously implicated in the development of antiestrogen resistance, this finding implicates ELF5 as a defining factor in the acquisition of the key aspects of the lethal phenotype in luminal A breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias Pulmonares/secundario , Pulmón/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/fisiopatología , Neoplasias de la Mama/virología , Permeabilidad Capilar , Proliferación Celular , Proteínas de Unión al ADN , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hemorragia/etiología , Hemorragia/prevención & control , Humanos , Leucocitos/inmunología , Leucocitos/patología , Pulmón/irrigación sanguínea , Pulmón/inmunología , Pulmón/patología , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/prevención & control , Depleción Linfocítica , Ratones Transgénicos , Células Mieloides/inmunología , Células Mieloides/patología , Proteínas de Neoplasias/genética , Neovascularización Patológica/etiología , Neovascularización Patológica/prevención & control , Infiltración Neutrófila , Poliomavirus/patogenicidad , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Recombinantes de Fusión/metabolismo , Análisis de Supervivencia , Factores de Transcripción , Carga Tumoral
20.
Nat Commun ; 6: 7139, 2015 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-25972253

RESUMEN

Neutrophil recruitment to the site of injury is an essential first step of an anti-bacterial response. However, little is known about the basis for and relevance of neutrophil migration from inflamed tissue into lymphoid organs. We established a photoconversion-based system to monitor the fate of neutrophils recruited to inflamed skin. While neutrophils are efficiently recruited to sites of both microbial and sterile lesions, subsequent re-localization to draining lymph nodes happens only when bacteria are present in the primary lesion. Skin egress of neutrophils occurs via lymphatic vessels and is dependent on CD11b and CXCR4 but not CCR7. Neutrophils are the predominant immune cell to migrate from inflamed skin into lymph nodes where they augment lymphocyte proliferation. Furthermore, inhibition of neutrophil migration from skin reduces T-cell proliferation in draining lymph nodes. Thus neutrophils mediate rapid cellular communication between the initial injury site and secondary lymphoid organs and modulate immune responsiveness.


Asunto(s)
Ganglios Linfáticos/patología , Linfocitos/microbiología , Infiltración Neutrófila , Neutrófilos/microbiología , Animales , Infecciones Bacterianas/sangre , Antígeno CD11b/biosíntesis , Movimiento Celular , Femenino , Sistema Inmunológico , Inflamación , Vasos Linfáticos , Activación de Linfocitos/inmunología , Linfocitos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/citología , Fenotipo , Receptores CCR7/biosíntesis , Receptores CXCR4/biosíntesis , Piel/microbiología , Staphylococcus aureus/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...