Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Blood ; 143(19): 1965-1979, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38271660

RESUMEN

ABSTRACT: Acute myeloid leukemia (AML) is an aggressive hematological malignancy originating from transformed hematopoietic stem or progenitor cells. AML prognosis remains poor owing to resistance and relapse driven by leukemia stem cells (LSCs). Targeting molecules essential for LSC function is a promising therapeutic approach. The phosphatidylinositol 3-kinase (PI3K)/AKT pathway is often dysregulated in AML. We found that although PI3Kγ is highly enriched in LSCs and critical for self-renewal, it was dispensable for normal hematopoietic stem cells. Mechanistically, PI3Kγ-AKT signaling promotes nuclear factor erythroid 2-related factor 2 (NRF2) nuclear accumulation, which induces 6-phosphogluconate dehydrogenase (PGD) and the pentose phosphate pathway, thereby maintaining LSC stemness. Importantly, genetic or pharmacological inhibition of PI3Kγ impaired expansion and stemness of murine and human AML cells in vitro and in vivo. Together, our findings reveal a key role for PI3Kγ in selectively maintaining LSC function by regulating AKT-NRF2-PGD metabolic pathway. Targeting the PI3Kγ pathway may, therefore, eliminate LSCs without damaging normal hematopoiesis, providing a promising therapeutic strategy for AML.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ib , Leucemia Mieloide Aguda , Células Madre Neoplásicas , Vía de Pentosa Fosfato , Animales , Humanos , Ratones , Autorrenovación de las Células , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Vía de Pentosa Fosfato/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal
2.
Nature ; 617(7959): 42-43, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37076711

Asunto(s)
Incendios , Neoplasias , Humanos
3.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33846242

RESUMEN

Precision medicine in oncology leverages clinical observations of exceptional response. Toward an understanding of the molecular features that define this response, we applied an integrated, multiplatform analysis of RNA profiles derived from clinically annotated glioblastoma samples. This analysis suggested that specimens from exceptional responders are characterized by decreased accumulation of microglia/macrophages in the glioblastoma microenvironment. Glioblastoma-associated microglia/macrophages secreted interleukin 11 (IL11) to activate STAT3-MYC signaling in glioblastoma cells. This signaling induced stem cell states that confer enhanced tumorigenicity and resistance to the standard-of-care chemotherapy, temozolomide (TMZ). Targeting a myeloid cell restricted an isoform of phosphoinositide-3-kinase, phosphoinositide-3-kinase gamma isoform (PI3Kγ), by pharmacologic inhibition or genetic inactivation disrupted this signaling axis by reducing microglia/macrophage-associated IL11 secretion in the tumor microenvironment. Mirroring the clinical outcomes of exceptional responders, PI3Kγ inhibition synergistically enhanced the anti-neoplastic effects of TMZ in orthotopic murine glioblastoma models. Moreover, inhibition or genetic inactivation of PI3Kγ in murine glioblastoma models recapitulated expression profiles observed in clinical specimens isolated from exceptional responders. Our results suggest key contributions from tumor-associated microglia/macrophages in exceptional responses and highlight the translational potential for PI3Kγ inhibition as a glioblastoma therapy.


Asunto(s)
Glioblastoma/metabolismo , Microglía/metabolismo , Temozolomida/farmacología , Adulto , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Resistencia a Antineoplásicos/fisiología , Femenino , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Humanos , Interleucina-11/inmunología , Interleucina-11/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Desnudos , Microglía/fisiología , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Transducción de Señal/efectos de los fármacos , Temozolomida/metabolismo , Microambiente Tumoral/efectos de los fármacos , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/fisiología
4.
Trends Immunol ; 40(4): 310-327, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30890304

RESUMEN

Macrophages are phagocytes that serve as a first line of defense against pathogenic insults to tissues. These innate immune cells mount proinflammatory responses to pathogens and repair damaged tissues. However, tumor-associated macrophages (TAMs) express cytokines and chemokines that can suppress antitumor immunity and promote tumor progression. Preclinical studies have identified crucial pathways regulating the recruitment, polarization, and metabolism of TAMs during tumor progression. Moreover, novel therapeutics targeting these pathways can indirectly stimulate cytotoxic T cell activation and recruitment, and synergize with checkpoint inhibitors, chemotherapy and/or radiation therapy in preclinical studies. Thus, clinical trials with therapeutic agents that promote phagocytosis or suppress survival, proliferation, trafficking, or polarization of TAMs are currently underway. These early results offer the promise of improved cancer outcomes.


Asunto(s)
Macrófagos/inmunología , Neoplasias/inmunología , Animales , Humanos , Neoplasias/terapia
5.
Nature ; 539(7629): 437-442, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27642729

RESUMEN

Macrophages play critical, but opposite, roles in acute and chronic inflammation and cancer. In response to pathogens or injury, inflammatory macrophages express cytokines that stimulate cytotoxic T cells, whereas macrophages in neoplastic and parasitic diseases express anti-inflammatory cytokines that induce immune suppression and may promote resistance to T cell checkpoint inhibitors. Here we show that macrophage PI 3-kinase γ controls a critical switch between immune stimulation and suppression during inflammation and cancer. PI3Kγ signalling through Akt and mTor inhibits NFκB activation while stimulating C/EBPß activation, thereby inducing a transcriptional program that promotes immune suppression during inflammation and tumour growth. By contrast, selective inactivation of macrophage PI3Kγ stimulates and prolongs NFκB activation and inhibits C/EBPß activation, thus promoting an immunostimulatory transcriptional program that restores CD8+ T cell activation and cytotoxicity. PI3Kγ synergizes with checkpoint inhibitor therapy to promote tumour regression and increased survival in mouse models of cancer. In addition, PI3Kγ-directed, anti-inflammatory gene expression can predict survival probability in cancer patients. Our work thus demonstrates that therapeutic targeting of intracellular signalling pathways that regulate the switch between macrophage polarization states can control immune suppression in cancer and other disorders.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Tolerancia Inmunológica/inmunología , Animales , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase Ib/deficiencia , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Femenino , Humanos , Inflamación/inmunología , Activación de Linfocitos , Macrófagos/enzimología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Linfocitos T/citología , Linfocitos T/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Escape del Tumor/inmunología
7.
Proc Natl Acad Sci U S A ; 110(22): 9042-7, 2013 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-23671068

RESUMEN

Lymph nodes are initial sites of tumor metastasis, yet whether the lymph node microenvironment actively promotes tumor metastasis remains unknown. We show here that VEGF-C/PI3Kα-driven remodeling of lymph nodes promotes tumor metastasis by activating integrin α4ß1 on lymph node lymphatic endothelium. Activated integrin α4ß1 promotes expansion of the lymphatic endothelium in lymph nodes and serves as an adhesive ligand that captures vascular cell adhesion molecule 1 (VCAM-1)(+) metastatic tumor cells, thereby promoting lymph node metastasis. Experimental induction of α4ß1 expression in lymph nodes is sufficient to promote tumor cell adhesion to lymphatic endothelium and lymph node metastasis in vivo, whereas genetic or pharmacological blockade of integrin α4ß1 or VCAM-1 inhibits it. As lymph node metastases accurately predict poor disease outcome, and integrin α4ß1 is a biomarker of lymphatic endothelium in tumor-draining lymph nodes from animals and patients, these results indicate that targeting integrin α4ß1 or VCAM to inhibit the interactions of tumor cells with the lymph node microenvironment may be an effective strategy to suppress tumor metastasis.


Asunto(s)
Carcinoma Ductal de Mama/patología , Endotelio Linfático/metabolismo , Integrina alfa4beta1/metabolismo , Ganglios Linfáticos/metabolismo , Metástasis de la Neoplasia/fisiopatología , Fosfatidilinositol 3-Quinasas/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Análisis de Varianza , Animales , Adhesión Celular/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Inmunohistoquímica , Linfangiogénesis/fisiología , Ratones , Metástasis de la Neoplasia/prevención & control , Molécula 1 de Adhesión Celular Vascular/fisiología
8.
Science ; 384(6702): eade8520, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38900864

RESUMEN

Unleashing antitumor T cell activity by checkpoint inhibitor immunotherapy is effective in cancer patients, but clinical responses are limited. Cytokine signaling through the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway correlates with checkpoint immunotherapy resistance. We report a phase I clinical trial of the JAK inhibitor ruxolitinib with anti-PD-1 antibody nivolumab in Hodgkin lymphoma patients relapsed or refractory following checkpoint inhibitor immunotherapy. The combination yielded a best overall response rate of 53% (10/19). Ruxolitinib significantly reduced neutrophil-to-lymphocyte ratios and percentages of myeloid suppressor cells but increased numbers of cytokine-producing T cells. Ruxolitinib rescued the function of exhausted T cells and enhanced the efficacy of immune checkpoint blockade in preclinical solid tumor and lymphoma models. This synergy was characterized by a switch from suppressive to immunostimulatory myeloid cells, which enhanced T cell division.


Asunto(s)
Enfermedad de Hodgkin , Inhibidores de Puntos de Control Inmunológico , Inhibidores de las Cinasas Janus , Nitrilos , Nivolumab , Pirazoles , Pirimidinas , Linfocitos T , Adulto , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Sinergismo Farmacológico , Enfermedad de Hodgkin/tratamiento farmacológico , Enfermedad de Hodgkin/inmunología , Enfermedad de Hodgkin/terapia , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia , Inhibidores de las Cinasas Janus/uso terapéutico , Quinasas Janus/metabolismo , Quinasas Janus/antagonistas & inhibidores , Nitrilos/uso terapéutico , Nivolumab/uso terapéutico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Linfocitos T/inmunología , Ratones Endogámicos C57BL , Ratones Endogámicos BALB C
9.
J Exp Clin Cancer Res ; 43(1): 157, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38824552

RESUMEN

Phosphoinositide-3-kinase γ (PI3Kγ) plays a critical role in pancreatic ductal adenocarcinoma (PDA) by driving the recruitment of myeloid-derived suppressor cells (MDSC) into tumor tissues, leading to tumor growth and metastasis. MDSC also impair the efficacy of immunotherapy. In this study we verify the hypothesis that MDSC targeting, via PI3Kγ inhibition, synergizes with α-enolase (ENO1) DNA vaccination in counteracting tumor growth.Mice that received ENO1 vaccination followed by PI3Kγ inhibition had significantly smaller tumors compared to those treated with ENO1 alone or the control group, and correlated with i) increased circulating anti-ENO1 specific IgG and IFNγ secretion by T cells, ii) increased tumor infiltration of CD8+ T cells and M1-like macrophages, as well as up-modulation of T cell activation and M1-like related transcripts, iii) decreased infiltration of Treg FoxP3+ T cells, endothelial cells and pericytes, and down-modulation of the stromal compartment and T cell exhaustion gene transcription, iv) reduction of mature and neo-formed vessels, v) increased follicular helper T cell activation and vi) increased "antigen spreading", as many other tumor-associated antigens were recognized by IgG2c "cytotoxic" antibodies. PDA mouse models genetically devoid of PI3Kγ showed an increased survival and a pattern of transcripts in the tumor area similar to that of pharmacologically-inhibited PI3Kγ-proficient mice. Notably, tumor reduction was abrogated in ENO1 + PI3Kγ inhibition-treated mice in which B cells were depleted.These data highlight a novel role of PI3Kγ in B cell-dependent immunity, suggesting that PI3Kγ depletion strengthens the anti-tumor response elicited by the ENO1 DNA vaccine.


Asunto(s)
Vacunas de ADN , Animales , Ratones , Vacunas de ADN/farmacología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Humanos , Línea Celular Tumoral , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacología , Modelos Animales de Enfermedad , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo
10.
Sci Transl Med ; 16(754): eadi6887, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38959328

RESUMEN

Virulent infectious agents such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and methicillin-resistant Staphylococcus aureus (MRSA) induce tissue damage that recruits neutrophils, monocyte, and macrophages, leading to T cell exhaustion, fibrosis, vascular leak, epithelial cell depletion, and fatal organ damage. Neutrophils, monocytes, and macrophages recruited to pathogen-infected lungs, including SARS-CoV-2-infected lungs, express phosphatidylinositol 3-kinase gamma (PI3Kγ), a signaling protein that coordinates both granulocyte and monocyte trafficking to diseased tissues and immune-suppressive, profibrotic transcription in myeloid cells. PI3Kγ deletion and inhibition with the clinical PI3Kγ inhibitor eganelisib promoted survival in models of infectious diseases, including SARS-CoV-2 and MRSA, by suppressing inflammation, vascular leak, organ damage, and cytokine storm. These results demonstrate essential roles for PI3Kγ in inflammatory lung disease and support the potential use of PI3Kγ inhibitors to suppress inflammation in severe infectious diseases.


Asunto(s)
COVID-19 , Fosfatidilinositol 3-Quinasa Clase Ib , Inflamación , SARS-CoV-2 , COVID-19/patología , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Animales , Inflamación/patología , Humanos , Tratamiento Farmacológico de COVID-19 , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Pulmón/patología , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Permeabilidad Capilar/efectos de los fármacos , Ratones Endogámicos C57BL , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/patología
11.
J Biol Chem ; 286(17): 14952-62, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21343305

RESUMEN

Growth and remodeling of lymphatic vasculature occur during development and during various pathologic states. A major stimulus for this process is the unique lymphatic vascular endothelial growth factor-C (VEGF-C). Other endothelial growth factors, such as fibroblast growth factor-2 (FGF-2) or VEGF-A, may also contribute. Heparan sulfate is a linear sulfated polysaccharide that facilitates binding and action of some vascular growth factors such as FGF-2 and VEGF-A. However, a direct role for heparan sulfate in lymphatic endothelial growth and sprouting responses, including those mediated by VEGF-C, remains to be examined. We demonstrate that VEGF-C binds to heparan sulfate purified from primary lymphatic endothelia, and activation of lymphatic endothelial Erk1/2 in response to VEGF-C is reduced by interference with heparin or pretreatment of cells with heparinase, which destroys heparan sulfate. Such treatment also inhibited phosphorylation of the major VEGF-C receptor VEGFR-3 upon VEGF-C stimulation. Silencing lymphatic heparan sulfate chain biosynthesis inhibited VEGF-C-mediated Erk1/2 activation and abrogated VEGFR-3 receptor-dependent binding of VEGF-C to the lymphatic endothelial surface. These findings prompted targeting of lymphatic N-deacetylase/N-sulfotransferase-1 (Ndst1), a major sulfate-modifying heparan sulfate biosynthetic enzyme. VEGF-C-mediated Erk1/2 phosphorylation was inhibited in Ndst1-silenced lymphatic endothelia, and scratch-assay responses to VEGF-C and FGF-2 were reduced in Ndst1-deficient cells. In addition, lymphatic Ndst1 deficiency abrogated cell-based growth and proliferation responses to VEGF-C. In other studies, lymphatic endothelia cultured ex vivo from Ndst1 gene-targeted mice demonstrated reduced VEGF-C- and FGF-2-mediated sprouting in collagen matrix. Lymphatic heparan sulfate may represent a novel molecular target for therapeutic intervention.


Asunto(s)
Linfangiogénesis , Factor C de Crecimiento Endotelial Vascular/fisiología , Animales , Endotelio Linfático , Heparitina Sulfato/deficiencia , Vasos Linfáticos , Ratones , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Unión Proteica , Sulfotransferasas/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular
12.
Blood ; 116(25): 5773-83, 2010 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-20826718

RESUMEN

Angiogenesis is controlled by signals that stimulate motility in endothelial cells at the tips of vascular sprouts while maintaining cell-cell adhesion in the stalks of angiogenic sprouts. We show here that Gs-linked G protein-coupled receptor activation of cAMP-dependent protein kinase (PKA) plays an important role in regulating the switch between endothelial cell adhesion and migration by activating C-terminal Src kinase, leading to inhibition of pp60Src. Activated PKA blocks pp60Src-dependent vascular endot helial-cadherin phosphorylation, thereby stimulating cell-cell adhesion while suppressing endothelial cell polarization, motility, angiogenesis, and vascular permeability. Similar to the actions of Notch and Dll4, PKA activation blocks sprouting in newly forming embryonic blood vessels, while PKA inhibition promotes excessive sprouting in these vessels. These findings demonstrate that G protein-coupled receptors and PKA regulate vascular sprouting during angiogenesis by controlling endothelial cell migration and cell-cell adhesion through their actions on pp60Src.


Asunto(s)
Adhesión Celular/fisiología , Movimiento Celular/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Neovascularización Fisiológica , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Transducción de Señal , Western Blotting , Proteína Tirosina Quinasa CSK , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Inmunoprecipitación , Fosforilación , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Venas Umbilicales/citología , Venas Umbilicales/metabolismo , Familia-src Quinasas
13.
Nat Commun ; 13(1): 1768, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35365657

RESUMEN

Myeloid cells play key roles in cancer immune suppression and tumor progression. In response to tumor derived factors, circulating monocytes and granulocytes extravasate into the tumor parenchyma where they stimulate angiogenesis, immune suppression and tumor progression. Chemokines, cytokines and interleukins stimulate PI3Kγ-mediated Rap1 activation, leading to conformational changes in integrin α4ß1 that promote myeloid cell extravasation and tumor inflammation Here we show that PI3Kγ activates a high molecular weight form of myosin light chain kinase, MLCK210, that promotes myosin-dependent Rap1 GTP loading, leading to integrin α4ß1 activation. Genetic or pharmacological inhibition of MLCK210 suppresses integrin α4ß1 activation, as well as tumor inflammation and progression. These results demonstrate a critical role for myeloid cell MLCK210 in tumor inflammation and serve as basis for the development of alternative approaches to develop immune oncology therapeutics.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Quinasa de Cadena Ligera de Miosina , Neoplasias , Adhesión Celular/fisiología , Humanos , Inflamación , Peso Molecular , Células Mieloides/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Neoplasias/genética
14.
Nat Med ; 8(9): 995-1003, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12185361

RESUMEN

Angiogenesis is a highly regulated process that results from the sequential actions of naturally occurring stimulators and inhibitors. Here, we show that parathyroid hormone-related peptide, a peptide hormone derived from normal and tumor cells that regulates bone metabolism and vascular tone, is a naturally occurring angiogenesis inhibitor. Parathyroid hormone-related peptide or a ten-amino-acid peptide from its N terminus inhibits endothelial cell migration in vitro and angiogenesis in vivo by activating endothelial cell protein kinase A. Activation of protein kinase A inhibits cell migration and angiogenesis by inhibiting the small GTPase Rac. In contrast, inhibition of protein kinase A reverses the anti-migratory and anti-angiogenic properties of parathyroid hormone-related peptide. These studies show that parathyroid hormone-related peptide is a naturally occurring angiogenesis inhibitor that functions by activation of protein kinase A.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas/metabolismo , Proteínas/farmacología , Sulfonamidas , Animales , Pruebas de Carcinogenicidad , Movimiento Celular , Embrión de Pollo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Endotelio Vascular , Inhibidores Enzimáticos/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Técnicas de Transferencia de Gen , Isoquinolinas/farmacología , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Fisiológica/efectos de los fármacos , Proteína Relacionada con la Hormona Paratiroidea , Mapeo Peptídico , Proteínas/genética , Proteínas de Unión al GTP rac/metabolismo
15.
J Clin Invest ; 116(12): 3111-3, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17143327

RESUMEN

The formation of new blood vessels, a process known as angiogenesis, is important for embryonic development and wound healing as well as the development of cancer and inflammation; therefore, angiogenesis is a valuable target for clinical intervention. Both logic and empiricism suggest that a balance of stimulatory and inhibitory switches is required for orderly formation of blood vessels. Thrombospondins 1 and 2 were among the first natural angiogenesis inhibitors to be identified. However, the cellular origins and mechanisms of action of these important proteins during angiogenesis have remained largely unknown. Studies by Kopp et al., presented in this issue of the JCI, clarify some of these issues by revealing that megakaryocytes and their "sticky" wound-healing progeny, platelets, are important sources of thrombospondins 1 and 2 and that these thrombopoietic cells play key roles in controlling blood vessel formation during hematopoiesis and ischemic wound healing (see the related article beginning on page 3277).


Asunto(s)
Neovascularización Patológica , Neovascularización Fisiológica , Trombospondinas/metabolismo , Animales , Plaquetas/citología , Plaquetas/metabolismo , Médula Ósea/irrigación sanguínea , Médula Ósea/metabolismo , Megacariocitos/citología , Megacariocitos/inmunología , Ratones , Ratones Transgénicos , Modelos Biológicos , Trombospondina 1/genética , Trombospondina 1/metabolismo , Trombospondinas/genética
16.
J Surg Res ; 151(1): 68-73, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18599080

RESUMEN

BACKGROUND: The lymphatic system is a major route for cancer cell dissemination, and a potential target for antitumor therapy. Despite ongoing interest in this area of research, the real-time behavior of cancer cells trafficking in the lymphatic system is poorly understood due to lack of appropriate tools to image this process. MATERIALS AND METHODS: We have used monoclonal-antibody and fluorescence technology to color-code lymphatic vessels and the cancer cells inside them in a living animal. Monoclonal anti-mouse LYVE-1 antibody was conjugated to a green fluorophore and delivered to the lymphatic system of a nude mouse, allowing imaging of mouse lymphatics. Tumor cells engineered to express red fluorescent protein were then imaged traveling within the labeled lymphatics in real time. RESULTS: AlexaFluor-labeled monoclonal anti-mouse LYVE-1 created a durable signal with clear delineation of lymphatic architecture. The duration of fluorescent signal after conjugated LYVE-1 delivery was far superior to that of fluorescein isothiocyanate-dextran or control fluorophore-conjugated IgG. Tumor cells engineered to express red fluorescent protein delivered to the inguinal lymph node enabled real-time tracking of tumor cell movement within the green fluorescent-labeled lymphatic vessels. CONCLUSIONS: This technology offers a powerful tool for the in vivo study of real-time trafficking of tumor cells within lymphatic vessels, for the deposition of the tumor cells in lymph nodes, as well as for screening of potential antitumor lymphatic therapies.


Asunto(s)
Diagnóstico por Imagen/métodos , Glicoproteínas , Ganglios Linfáticos/patología , Vasos Linfáticos/patología , Células Neoplásicas Circulantes , Neoplasias Pancreáticas/patología , Animales , Línea Celular Tumoral , Movimiento Celular , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Humanos , Proteínas Luminiscentes , Ganglios Linfáticos/inmunología , Vasos Linfáticos/inmunología , Proteínas de Transporte de Membrana , Ratones , Ratones Desnudos , Invasividad Neoplásica/inmunología , Invasividad Neoplásica/patología , Neoplasias Pancreáticas/inmunología , Retroviridae , Transducción Genética , Proteína Fluorescente Roja
17.
Methods Mol Biol ; 467: 139-55, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19301669

RESUMEN

Recent studies have found that bone marrow-derived cells give rise to endothelial cells during states of tissue repair and disease. We have found that one key integrin, integrin-alpha4beta1, promotes the homing of circulating endothelial progenitor cells (EPCs) to sites of ongoing tissue repair. This integrin facilitates the adhesion of EPCs to the vascular endothelium in inflamed tissue or within tumors. We demonstrate how to identify, isolate, purify, and characterize EPCs. We also demonstrate in vivo analysis of the roles of bone marrow-derived cells in tumor growth and angiogenesis by demonstrating adoptive transfer, bone marrow transplantation, tumor models, and immunohistochemistry for markers of blood and endothelial vessels. Finally, we show how to characterize cell adhesion mechanisms regulating bone marrow-derived progenitor cell trafficking.


Asunto(s)
Movimiento Celular , Células Endoteliales/citología , Células Madre/citología , Animales , Trasplante de Médula Ósea , Adhesión Celular , Citometría de Flujo , Humanos , Inmunohistoquímica , Neoplasias/metabolismo , Neoplasias/patología
18.
Adv Mater ; 31(35): e1902952, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31267590

RESUMEN

Despite the promise of ribonucleic acid interference therapeutics, the delivery of oligonucleotides selectively to diseased tissues in the body, and specifically to the cellular location in the tissues needed to provide optimal therapeutic outcome, remains a significant challenge. Here, key material properties and biological mechanisms for delivery of short interfering RNAs (siRNAs) to effectively silence target-specific cells in vivo are identified. Using porous silicon nanoparticles as the siRNA host, tumor-targeting peptides for selective tissue homing, and fusogenic lipid coatings to induce fusion with the plasma membrane, it is shown that the uptake mechanism can be engineered to be independent of common receptor-mediated endocytosis pathways. Two examples of the potential broad clinical applicability of this concept in a mouse xenograft model of ovarian cancer peritoneal carcinomatosis are provided: silencing the Rev3l subunit of polymerase Pol ζ to impair DNA repair in combination with cisplatin; and reprogramming tumor-associated macrophages into a proinflammatory state.


Asunto(s)
Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Endosomas/metabolismo , Nanopartículas/química , Péptidos/metabolismo , ARN Interferente Pequeño/química , Silicio/química , Animales , Línea Celular Tumoral , Silenciador del Gen , Humanos , Ratones , Porosidad , ARN Interferente Pequeño/genética
19.
Cancer Res ; 79(19): 5048-5059, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31416839

RESUMEN

Tumor-associated macrophages (TAM) are highly expressed within the tumor microenvironment of a wide range of cancers, where they exert a protumor phenotype by promoting tumor cell growth and suppressing antitumor immune function. Here, we show that TAM accumulation in human and mouse tumors correlates with tumor cell expression of integrin αvß3, a known driver of epithelial cancer progression and drug resistance. A monoclonal antibody targeting αvß3 (LM609) exploited the coenrichment of αvß3 and TAMs to not only eradicate highly aggressive drug-resistant human lung and pancreas cancers in mice, but also to prevent the emergence of circulating tumor cells. Importantly, this antitumor activity in mice was eliminated following macrophage depletion. Although LM609 had no direct effect on tumor cell viability, it engaged macrophages but not natural killer (NK) cells to induce antibody-dependent cellular cytotoxicity (ADCC) of αvß3-expressing tumor cells despite their expression of the CD47 "don't eat me" signal. In contrast to strategies designed to eliminate TAMs, these findings suggest that anti-αvß3 represents a promising immunotherapeutic approach to redirect TAMs to serve as tumor killers for late-stage or drug-resistant cancers. SIGNIFICANCE: Therapeutic antibodies are commonly engineered to optimize engagement of NK cells as effectors. In contrast, LM609 targets αvß3 to suppress tumor progression and enhance drug sensitivity by exploiting TAMs to trigger ADCC.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Integrina alfaVbeta3/inmunología , Macrófagos/inmunología , Neoplasias Glandulares y Epiteliales/inmunología , Animales , Antineoplásicos/farmacología , Progresión de la Enfermedad , Humanos , Ratones , Neoplasias Glandulares y Epiteliales/patología , Fagocitosis/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
20.
Oncogene ; 38(28): 5599-5611, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30967626

RESUMEN

The MST1R (RON) kinase is overexpressed in >80% of human pancreatic cancers, but its role in pancreatic carcinogenesis is unknown. In this study, we examined the relevance of Mst1r kinase to Kras driven pancreatic carcinogenesis using genetically engineered mouse models. In the setting of mutant Kras, Mst1r overexpression increased acinar-ductal metaplasia (ADM), accelerated the progression of pancreatic intraepithelial neoplasia (PanIN), and resulted in the accumulation of (mannose receptor C type 1) MRC1+, (arginase 1) Arg+ macrophages in the tumor microenvironment. Conversely, absence of a functional Mst1r kinase slowed PanIN initiation, resulted in smaller tumors, prolonged survival and a reduced tumor-associated macrophage content. Mst1r expression was associated with increased production of its ligand Mst1, and in orthotopic models, suppression of Mst1 expression resulted in reduced tumor size, changes in macrophage polarization and enhanced T cell infiltration. This study demonstrates the functional significance of Mst1r during pancreatic cancer initiation and progression. Further, it provides proof of concept that targeting Mst1r can modulate pancreatic cancer growth and the microenvironment. This study provides further rationale for targeting Mst1r as a therapeutic strategy.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Células Epiteliales/patología , Macrófagos/patología , Neoplasias Pancreáticas/patología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Carcinoma Ductal Pancreático/enzimología , Progresión de la Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias Pancreáticas/enzimología , Prueba de Estudio Conceptual , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA