Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Nat Immunol ; 19(1): 76-84, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29180808

RESUMEN

Exciting progress in the field of cancer immunotherapy has renewed the urgency of the need for basic studies of immunoregulation in both adaptive cell lineages and innate cell lineages. Here we found a central role for major histocompatibility complex (MHC) class I in controlling the phagocytic function of macrophages. Our results demonstrated that expression of the common MHC class I component ß2-microglobulin (ß2M) by cancer cells directly protected them from phagocytosis. We further showed that this protection was mediated by the inhibitory receptor LILRB1, whose expression was upregulated on the surface of macrophages, including tumor-associated macrophages. Disruption of either MHC class I or LILRB1 potentiated phagocytosis of tumor cells both in vitro and in vivo, which defines the MHC class I-LILRB1 signaling axis as an important regulator of the effector function of innate immune cells, a potential biomarker for therapeutic response to agents directed against the signal-regulatory protein CD47 and a potential target of anti-cancer immunotherapy.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Receptor Leucocitario Tipo Inmunoglobulina B1/inmunología , Macrófagos/inmunología , Neoplasias/inmunología , Fagocitosis/inmunología , Animales , Línea Celular Tumoral , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inmunoterapia/métodos , Receptor Leucocitario Tipo Inmunoglobulina B1/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neoplasias/metabolismo , Neoplasias/terapia , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/terapia
2.
J Immunol ; 209(3): 606-620, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35817516

RESUMEN

Despite recent therapeutic progress, advanced melanoma remains lethal for many patients. The composition of the immune tumor microenvironment (TME) has decisive impacts on therapy response and disease outcome, and high-dimensional analyses of patient samples reveal the heterogeneity of the immune TME. Macrophages infiltrate TMEs and generally associate with tumor progression, but the underlying mechanisms are incompletely understood. Because experimental systems are needed to elucidate the functional properties of these cells, we developed a humanized mouse model reconstituted with human immune cells and human melanoma. We used two strains of recipient mice, supporting or not supporting the development of human myeloid cells. We found that human myeloid cells favored metastatic spread of the primary tumor, thereby recapitulating the cancer-supportive role of macrophages. We next analyzed the transcriptome of human immune cells infiltrating tumors versus other tissues. This analysis identified a cluster of myeloid cells present in the TME, but not in other tissues, which do not correspond to canonical M2 cells. The transcriptome of these cells is characterized by high expression of glycolytic enzymes and multiple chemokines and by low expression of gene sets associated with inflammation and adaptive immunity. Compared with humanized mouse results, we found transcriptionally similar myeloid cells in patient-derived samples of melanoma and other cancer types. The humanized mouse model described here thus complements patient sample analyses, enabling further elucidation of fundamental principles in melanoma biology beyond M1/M2 macrophage polarization. The model can also support the development and evaluation of candidate antitumor therapies.


Asunto(s)
Macrófagos , Melanoma , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Activación de Macrófagos , Melanoma/patología , Ratones , Microambiente Tumoral
3.
Proc Natl Acad Sci U S A ; 114(49): E10578-E10585, 2017 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-29158380

RESUMEN

Cancer immunotherapy has emerged as a promising therapeutic intervention. However, complete and durable responses are only seen in a fraction of patients who have cancer. A key factor that limits therapeutic success is the infiltration of tumors by cells of the myeloid lineage. The inhibitory receptor signal regulatory protein-α (SIRPα) is a myeloid-specific immune checkpoint that engages the "don't eat me" signal CD47 expressed on tumors and normal tissues. We therefore developed the monoclonal antibody KWAR23, which binds human SIRPα with high affinity and disrupts its binding to CD47. Administered by itself, KWAR23 is inert, but given in combination with tumor-opsonizing monoclonal antibodies, KWAR23 greatly augments myeloid cell-dependent killing of a collection of hematopoietic and nonhematopoietic human tumor-derived cell lines. Following KWAR23 antibody treatment in a human SIRPA knockin mouse model, both neutrophils and macrophages infiltrate a human Burkitt's lymphoma xenograft and inhibit tumor growth, generating complete responses in the majority of treated animals. We further demonstrate that a bispecific anti-CD70/SIRPα antibody outperforms individually delivered antibodies in specific types of cancers. These studies demonstrate that SIRPα blockade induces potent antitumor activity by targeting multiple myeloid cell subsets that frequently infiltrate tumors. Thus, KWAR23 represents a promising candidate for combination therapy.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales/farmacología , Anticuerpos Antineoplásicos/farmacología , Antígenos de Diferenciación/inmunología , Linfoma de Burkitt/terapia , Fagocitosis/efectos de los fármacos , Receptores Inmunológicos/inmunología , Animales , Antígenos de Diferenciación/genética , Linfoma de Burkitt/genética , Linfoma de Burkitt/inmunología , Linfoma de Burkitt/patología , Ligando CD27/genética , Ligando CD27/inmunología , Antígeno CD47/genética , Antígeno CD47/inmunología , Línea Celular Tumoral , Terapia Combinada/métodos , Expresión Génica , Técnicas de Sustitución del Gen , Humanos , Inmunoterapia/métodos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Transgénicos , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Unión Proteica , Receptores Inmunológicos/genética , Transgenes , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Proc Natl Acad Sci U S A ; 112(7): 2145-50, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25646432

RESUMEN

Macrophage-mediated programmed cell removal (PrCR) is an important mechanism of eliminating diseased and damaged cells before programmed cell death. The induction of PrCR by eat-me signals on tumor cells is countered by don't-eat-me signals such as CD47, which binds macrophage signal-regulatory protein α to inhibit phagocytosis. Blockade of CD47 on tumor cells leads to phagocytosis by macrophages. Here we demonstrate that the activation of Toll-like receptor (TLR) signaling pathways in macrophages synergizes with blocking CD47 on tumor cells to enhance PrCR. Bruton's tyrosine kinase (Btk) mediates TLR signaling in macrophages. Calreticulin, previously shown to be an eat-me signal on cancer cells, is activated in macrophages for secretion and cell-surface exposure by TLR and Btk to target cancer cells for phagocytosis, even if the cancer cells themselves do not express calreticulin.


Asunto(s)
Calreticulina/fisiología , Macrófagos/inmunología , Neoplasias/patología , Proteínas Tirosina Quinasas/metabolismo , Receptores Toll-Like/fisiología , Agammaglobulinemia Tirosina Quinasa , Humanos , Neoplasias/enzimología , Neoplasias/metabolismo
5.
PLoS One ; 18(6): e0287052, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37379309

RESUMEN

Human CD34+ hematopoietic stem and progenitor cells (HSPCs) are a standard source of cells for clinical HSC transplantations as well as experimental xenotransplantation to generate "humanized mice". To further extend the range of applications of these humanized mice, we developed a protocol to efficiently edit the genomes of human CD34+ HSPCs before transplantation. In the past, manipulating HSPCs has been complicated by the fact that they are inherently difficult to transduce with lentivectors, and rapidly lose their stemness and engraftment potential during in vitro culture. However, with optimized nucleofection of sgRNA:Cas9 ribonucleoprotein complexes, we are now able to edit a candidate gene in CD34+ HSPCs with almost 100% efficiency, and transplant these modified cells in immunodeficient mice with high engraftment levels and multilineage hematopoietic differentiation. The result is a humanized mouse from which we knocked out a gene of interest from their human immune system.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Humanos , Ratones , Animales , Antígenos CD34 , Técnicas de Inactivación de Genes , Sistema Inmunológico , Trasplante de Células Madre Hematopoyéticas/métodos , Ratones SCID
6.
Front Immunol ; 12: 643852, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33692812

RESUMEN

Since the late 1980s, mice have been repopulated with human hematopoietic cells to study the fundamental biology of human hematopoiesis and immunity, as well as a broad range of human diseases in vivo. Multiple mouse recipient strains have been developed and protocols optimized to efficiently generate these "humanized" mice. Here, we review three guiding principles that have been applied to the development of the currently available models: (1) establishing tolerance of the mouse host for the human graft; (2) opening hematopoietic niches so that they can be occupied by human cells; and (3) providing necessary support for human hematopoiesis. We then discuss four remaining challenges: (1) human hematopoietic lineages that poorly develop in mice; (2) limited antigen-specific adaptive immunity; (3) absent tolerance of the human immune system for its mouse host; and (4) sub-functional interactions between human immune effectors and target mouse tissues. While major advances are still needed, the current models can already be used to answer specific, clinically-relevant questions and hopefully inform the development of new, life-saving therapies.


Asunto(s)
Inmunidad Adaptativa , Modelos Animales de Enfermedad , Hematopoyesis/inmunología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/inmunología , Animales , Xenoinjertos , Humanos , Ratones
7.
Sci Transl Med ; 10(469)2018 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-30487249

RESUMEN

Peritoneal adhesions are fibrous tissues that tether organs to one another or to the peritoneal wall and are a major cause of postsurgical and infectious morbidity. The primary molecular chain of events leading to the initiation of adhesions has been elusive, chiefly due to the lack of an identifiable cell of origin. Using clonal analysis and lineage tracing, we have identified injured surface mesothelium expressing podoplanin (PDPN) and mesothelin (MSLN) as a primary instigator of peritoneal adhesions after surgery in mice. We demonstrate that an anti-MSLN antibody diminished adhesion formation in a mouse model where adhesions were induced by surgical ligation to form ischemic buttons and subsequent surgical abrasion of the peritoneum. RNA sequencing and bioinformatics analyses of mouse mesothelial cells from injured mesothelium revealed aspects of the pathological mechanism of adhesion development and yielded several potential regulators of this process. Specifically, we show that PDPN+MSLN+ mesothelium responded to hypoxia by early up-regulation of hypoxia-inducible factor 1 alpha (HIF1α) that preceded adhesion development. Inhibition of HIF1α with small molecules ameliorated the injury program in damaged mesothelium and was sufficient to diminish adhesion severity in a mouse model. Analyses of human adhesion tissue suggested that similar surface markers and signaling pathways may contribute to surgical adhesions in human patients.


Asunto(s)
Anticuerpos/farmacología , Biomarcadores/metabolismo , Epitelio/patología , Adherencias Tisulares/patología , Animales , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/ultraestructura , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mesotelina , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Peritoneo/efectos de los fármacos , Peritoneo/lesiones , Peritoneo/patología , Adherencias Tisulares/genética , Transcripción Genética
8.
Cancer Immunol Res ; 4(12): 1072-1087, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27856424

RESUMEN

Cancer immunotherapies hold much promise, but their potential in veterinary settings has not yet been fully appreciated. Canine lymphomas are among the most common tumors of dogs and bear remarkable similarity to human disease. In this study, we examined the combination of CD47 blockade with anti-CD20 passive immunotherapy for canine lymphoma. The CD47/SIRPα axis is an immune checkpoint that regulates macrophage activation. In humans, CD47 is expressed on cancer cells and enables evasion from phagocytosis. CD47-blocking therapies are now under investigation in clinical trials for a variety of human cancers. We found the canine CD47/SIRPα axis to be conserved biochemically and functionally. We identified high-affinity SIRPα variants that antagonize canine CD47 and stimulate phagocytosis of canine cancer cells in vitro When tested as Fc fusion proteins, these therapeutic agents exhibited single-agent efficacy in a mouse xenograft model of canine lymphoma. As robust synergy between CD47 blockade and tumor-specific antibodies has been demonstrated for human cancer, we evaluated the combination of CD47 blockade with 1E4-cIgGB, a canine-specific antibody to CD20. 1E4-cIgGB could elicit a therapeutic response against canine lymphoma in vivo as a single agent. However, augmented responses were observed when combined with CD47-blocking therapies, resulting in synergy in vitro and in vivo and eliciting cures in 100% of mice bearing canine lymphoma. Our findings support further testing of CD47-blocking therapies alone and in combination with CD20 antibodies in the veterinary setting. Cancer Immunol Res; 4(12); 1072-87. ©2016 AACR.


Asunto(s)
Antígenos CD20/inmunología , Antígeno CD47/inmunología , Inmunoterapia , Linfoma de Células B Grandes Difuso/terapia , Animales , Línea Celular Tumoral , Perros , Femenino , Inmunoglobulina G/uso terapéutico , Linfoma de Células B Grandes Difuso/inmunología , Linfoma de Células B Grandes Difuso/veterinaria , Macrófagos/inmunología , Masculino , Ratones , Fagocitosis , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA