Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 608(7922): 421-428, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35922508

RESUMEN

Glucose uptake is essential for cancer glycolysis and is involved in non-shivering thermogenesis of adipose tissues1-6. Most cancers use glycolysis to harness energy for their infinite growth, invasion and metastasis2,7,8. Activation of thermogenic metabolism in brown adipose tissue (BAT) by cold and drugs instigates blood glucose uptake in adipocytes4,5,9. However, the functional effects of the global metabolic changes associated with BAT activation on tumour growth are unclear. Here we show that exposure of tumour-bearing mice to cold conditions markedly inhibits the growth of various types of solid tumours, including clinically untreatable cancers such as pancreatic cancers. Mechanistically, cold-induced BAT activation substantially decreases blood glucose and impedes the glycolysis-based metabolism in cancer cells. The removal of BAT and feeding on a high-glucose diet under cold exposure restore tumour growth, and genetic deletion of Ucp1-the key mediator for BAT-thermogenesis-ablates the cold-triggered anticancer effect. In a pilot human study, mild cold exposure activates a substantial amount of BAT in both healthy humans and a patient with cancer with mitigated glucose uptake in the tumour tissue. These findings provide a previously undescribed concept and paradigm for cancer therapy that uses a simple and effective approach. We anticipate that cold exposure and activation of BAT through any other approach, such as drugs and devices either alone or in combination with other anticancer therapeutics, will provide a general approach for the effective treatment of various cancers.


Asunto(s)
Tejido Adiposo Pardo , Frío , Metabolismo Energético , Neoplasias , Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Glucemia/metabolismo , Terapia Combinada , Glucólisis , Humanos , Ratones , Neoplasias/metabolismo , Neoplasias/prevención & control , Neoplasias/terapia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/prevención & control , Neoplasias Pancreáticas/terapia , Termogénesis/genética , Proteína Desacopladora 1/metabolismo
2.
Proc Natl Acad Sci U S A ; 120(29): e2303740120, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37428914

RESUMEN

Defining reliable surrogate markers and overcoming drug resistance are the most challenging issues for improving therapeutic outcomes of antiangiogenic drugs (AADs) in cancer patients. At the time of this writing, no biomarkers are clinically available to predict AAD therapeutic benefits and drug resistance. Here, we uncovered a unique mechanism of AAD resistance in epithelial carcinomas with KRAS mutations that targeted angiopoietin 2 (ANG2) to circumvent antivascular endothelial growth factor (anti-VEGF) responses. Mechanistically, KRAS mutations up-regulated the FOXC2 transcription factor that directly elevated ANG2 expression at the transcriptional level. ANG2 bestowed anti-VEGF resistance as an alternative pathway to augment VEGF-independent tumor angiogenesis. Most colorectal and pancreatic cancers with KRAS mutations were intrinsically resistant to monotherapies of anti-VEGF or anti-ANG2 drugs. However, combination therapy with anti-VEGF and anti-ANG2 drugs produced synergistic and potent anticancer effects in KRAS-mutated cancers. Together, these data demonstrate that KRAS mutations in tumors serve as a predictive marker for anti-VEGF resistance and are susceptible to combination therapy with anti-VEGF and anti-ANG2 drugs.


Asunto(s)
Carcinoma , Factores de Crecimiento Endotelial , Humanos , Factores de Crecimiento Endotelial/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Angiopoyetina 2/genética , Angiopoyetina 2/metabolismo , Angiopoyetina 1/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(40): e2203307119, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-36161914

RESUMEN

Brown adipose tissue (BAT) is a highly specialized adipose tissue in its immobile location and size during the entire adulthood. In response to cold exposure and other ß3-adrenoreceptor stimuli, BAT commits energy consumption by nonshivering thermogenesis (NST). However, the molecular machinery in controlling the BAT mass in adults is unknown. Here, we show our surprising findings that the BAT mass and functions can be manipulated in adult animals by controlling BAT adipocyte differentiation in vivo. Platelet-derived growth factor receptor α (PDGFα) expressed in BAT progenitor cells served a signaling function to avert adipose progenitor differentiation. Genetic and pharmacological loss-of-function of PDGFRα eliminated the differentiation barrier and permitted progenitor cell differentiation to mature and functional BAT adipocytes. Consequently, an enlarged BAT mass (megaBAT) was created by PDGFRα inhibition owing to increases of brown adipocyte numbers. Under cold exposure, a microRNA-485 (miR-485) was identified as a master suppressor of the PDGFRα signaling, and delivery of miR-485 also produced megaBAT in adult animals. Noticeably, megaBAT markedly improved global metabolism, insulin sensitivity, high-fat-diet (HFD)-induced obesity, and diabetes by enhancing NST. Together, our findings demonstrate that the adult BAT mass can be increased by blocking the previously unprecedented inhibitory signaling for BAT progenitor cell differentiation. Thus, blocking the PDGFRα for the generation of megaBAT provides an attractive strategy for treating obesity and type 2 diabetes mellitus (T2DM).


Asunto(s)
Adipocitos Marrones , Adipocitos , Adipogénesis , Tejido Adiposo Pardo , MicroARNs , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas , Adipocitos/citología , Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Animales , Diabetes Mellitus Tipo 2/terapia , Metabolismo Energético , MicroARNs/genética , MicroARNs/metabolismo , Obesidad/terapia , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Termogénesis/genética
4.
Proc Natl Acad Sci U S A ; 117(37): 22910-22919, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32859758

RESUMEN

Lymphocyte-based immunotherapy has emerged as a breakthrough in cancer therapy for both hematologic and solid malignancies. In a subpopulation of cancer patients, this powerful therapeutic modality converts malignancy to clinically manageable disease. However, the T cell- and chimeric antigen receptor T (CAR-T) cell-mediated antimetastatic activity, especially their impacts on microscopic metastatic lesions, has not yet been investigated. Here we report a living zebrafish model that allows us to visualize the metastatic cancer cell killing effect by tumor- infiltrating lymphocytes (TILs) and CAR-T cells in vivo at the single-cell level. In a freshly isolated primary human melanoma, specific TILs effectively eliminated metastatic cancer cells in the living body. This potent metastasis-eradicating effect was validated using a human lymphoma model with CAR-T cells. Furthermore, cancer-associated fibroblasts protected metastatic cancer cells from T cell-mediated killing. Our data provide an in vivo platform to validate antimetastatic effects by human T cell-mediated immunotherapy. This unique technology may serve as a precision medicine platform for assessing anticancer effects of cellular immunotherapy in vivo before administration to human cancer patients.


Asunto(s)
Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/metabolismo , Análisis de la Célula Individual/métodos , Animales , Línea Celular Tumoral , Citotoxicidad Inmunológica/inmunología , Inmunoterapia Adoptiva/métodos , Activación de Linfocitos/fisiología , Modelos Animales , Metástasis de la Neoplasia/patología , Neoplasias/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Pez Cebra
5.
Gut ; 71(1): 129-147, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33568427

RESUMEN

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDAC) is the most lethal malignancy and lacks effective treatment. We aimed to understand molecular mechanisms of the intertwined interactions between tumour stromal components in metastasis and to provide a new paradigm for PDAC therapy. DESIGN: Two unselected cohorts of 154 and 20 patients with PDAC were subjected to correlation between interleukin (IL)-33 and CXCL3 levels and survivals. Unbiased expression profiling, and genetic and pharmacological gain-of-function and loss-of-function approaches were employed to identify molecular signalling in tumour-associated macrophages (TAMs) and myofibroblastic cancer-associated fibroblasts (myoCAFs). The role of the IL-33-ST2-CXCL3-CXCR2 axis in PDAC metastasis was evaluated in three clinically relevant mouse PDAC models. RESULTS: IL-33 was specifically elevated in human PDACs and positively correlated with tumour inflammation in human patients with PDAC. CXCL3 was highly upregulated in IL-33-stimulated macrophages that were the primary source of CXCL3. CXCL3 was correlated with poor survival in human patients with PDAC. Mechanistically, activation of the IL-33-ST2-MYC pathway attributed to high CXCL3 production. The highest level of CXCL3 was found in PDAC relative to other cancer types and its receptor CXCR2 was almost exclusively expressed in CAFs. Activation of CXCR2 by CXCL3 induced a CAF-to-myoCAF transition and α-smooth muscle actin (α-SMA) was uniquely upregulated by the CXCL3-CXCR2 signalling. Type III collagen was identified as the CXCL3-CXCR2-targeted adhesive molecule responsible for myoCAF-driven PDAC metastasis. CONCLUSIONS: Our work provides novel mechanistic insights into understanding PDAC metastasis by the TAM-CAF interaction and targeting each of these signalling components would provide an attractive and new paradigm for treating pancreatic cancer.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Carcinoma Ductal Pancreático/patología , Quimiocinas CXC/metabolismo , Neoplasias Pancreáticas/patología , Macrófagos Asociados a Tumores/metabolismo , Animales , Carcinoma Ductal Pancreático/mortalidad , Estudios de Cohortes , Humanos , Interleucina-33/metabolismo , Ratones Noqueados , Metástasis de la Neoplasia , Neoplasias Pancreáticas/mortalidad , Regulación hacia Arriba
6.
Proc Natl Acad Sci U S A ; 114(45): E9635-E9644, 2017 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-29078273

RESUMEN

Anti-VEGF drugs are commonly used for treatment of a variety of cancers in human patients, and they often develop resistance. The mechanisms underlying anti-VEGF resistance in human cancer patients are largely unknown. Here, we show that in mouse tumor models and in human cancer patients, the anti-VEGF drug-induced kidney hypoxia augments circulating levels of erythropoietin (EPO). Gain-of-function studies show that EPO protects tumor vessels from anti-VEGF treatment and compromises its antitumor effects. Loss of function by blocking EPO function using a pharmacological approach markedly increases antitumor activity of anti-VEGF drugs through inhibition of tumor angiogenesis. Similarly, genetic loss-of-function data shows that deletion of EpoR in nonerythroid cells significantly increases antiangiogenic and antitumor effects of anti-VEGF therapy. Finally, in a relatively large cohort study, we show that treatment of human colorectal cancer patients with bevacizumab augments circulating EPO levels. These findings uncover a mechanism of desensitizing antiangiogenic and anticancer effects by kidney-produced EPO. Our work presents conceptual advances of our understanding of mechanisms underlying antiangiogenic drug resistance.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Eritropoyetina/metabolismo , Riñón/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Animales , Bevacizumab/farmacología , Estudios de Cohortes , Neoplasias Colorrectales/metabolismo , Humanos , Riñón/metabolismo , Ratones , Neovascularización Patológica/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
J Pathol ; 246(4): 447-458, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30101525

RESUMEN

Aggressive tumor cells can adopt an endothelial cell-like phenotype and contribute to the formation of a tumor vasculature, independent of tumor angiogenesis. This adoptive mechanism is referred to as vascular mimicry and it is associated with poor survival in cancer patients. To what extent tumor cells capable of vascular mimicry phenocopy the angiogenic cascade is still poorly explored. Here, we identify pericytes as important players in vascular mimicry. We found that pericytes are recruited by vascular mimicry-positive tumor cells in order to facilitate sprouting and to provide structural support of the vascular-like networks. The pericyte recruitment is mediated through platelet-derived growth factor (PDGF)-B. Consequently, preventing PDGF-B signaling by blocking the PDGF receptors with either the small tyrosine kinase inhibitor imatinib or blocking antibodies inhibits vascular mimicry and tumor growth. Collectively, the current study identifies an important role for pericytes in the formation of vascular-like structures by tumor cells. Moreover, the mechanism that controls the pericyte recruitment provides therapeutic opportunities for patients with aggressive vascular mimicry-positive cancer types. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Mimetismo Biológico/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Mesilato de Imatinib/farmacología , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/tratamiento farmacológico , Neovascularización Patológica , Pericitos/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Animales , Comunicación Celular/efectos de los fármacos , Línea Celular Tumoral , Técnicas de Cocultivo , Humanos , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Desnudos , Pericitos/metabolismo , Pericitos/patología , Factor de Crecimiento Derivado de Plaquetas/inmunología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Proc Natl Acad Sci U S A ; 113(15): 4158-63, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-27035988

RESUMEN

Anti-VEGF-based antiangiogenic drugs are designed to block tumor angiogenesis for treatment of cancer patients. However, anti-VEGF drugs produce off-tumor target effects on multiple tissues and organs and cause broad adverse effects. Here, we show that vasculatures in endocrine organs were more sensitive to anti-VEGF treatment than tumor vasculatures. In thyroid, adrenal glands, and pancreatic islets, systemic treatment with low doses of an anti-VEGF neutralizing antibody caused marked vascular regression, whereas tumor vessels remained unaffected. Additionally, a low dose of VEGF blockade significantly inhibited the formation of thyroid vascular fenestrae, leaving tumor vascular structures unchanged. Along with vascular structural changes, the low dose of VEGF blockade inhibited vascular perfusion and permeability in thyroid, but not in tumors. Prolonged treatment with the low-dose VEGF blockade caused hypertension and significantly decreased circulating levels of thyroid hormone free-T3 and -T4, leading to functional impairment of thyroid. These findings show that the fenestrated microvasculatures in endocrine organs are more sensitive than tumor vasculatures in response to systemic anti-VEGF drugs. Thus, our data support the notion that clinically nonbeneficial treatments with anti-VEGF drugs could potentially cause adverse effects.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Glándulas Endocrinas/irrigación sanguínea , Neoplasias/irrigación sanguínea , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Glándulas Endocrinas/fisiopatología , Humanos , Ratones , Ratones Endogámicos C57BL , Neoplasias/tratamiento farmacológico
9.
Proc Natl Acad Sci U S A ; 113(38): E5618-27, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27608497

RESUMEN

Vascular pericytes, an important cellular component in the tumor microenvironment, are often associated with tumor vasculatures, and their functions in cancer invasion and metastasis are poorly understood. Here we show that PDGF-BB induces pericyte-fibroblast transition (PFT), which significantly contributes to tumor invasion and metastasis. Gain- and loss-of-function experiments demonstrate that PDGF-BB-PDGFRß signaling promotes PFT both in vitro and in in vivo tumors. Genome-wide expression analysis indicates that PDGF-BB-activated pericytes acquire mesenchymal progenitor features. Pharmacological inhibition and genetic deletion of PDGFRß ablate the PDGF-BB-induced PFT. Genetic tracing of pericytes with two independent mouse strains, TN-AP-CreERT2:R26R-tdTomato and NG2-CreERT2:R26R-tdTomato, shows that PFT cells gain stromal fibroblast and myofibroblast markers in tumors. Importantly, coimplantation of PFT cells with less-invasive tumor cells in mice markedly promotes tumor dissemination and invasion, leading to an increased number of circulating tumor cells and metastasis. Our findings reveal a mechanism of vascular pericytes in PDGF-BB-promoted cancer invasion and metastasis by inducing PFT, and thus targeting PFT may offer a new treatment option of cancer metastasis.


Asunto(s)
Carcinoma de Células Renales/genética , Pericitos/metabolismo , Proteínas Proto-Oncogénicas c-sis/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Animales , Becaplermina , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Pericitos/patología , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Microambiente Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Proc Natl Acad Sci U S A ; 112(22): E2900-9, 2015 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-25991856

RESUMEN

The biological functions of VEGF-B in cancer progression remain poorly understood. Here, we report that VEGF-B promotes cancer metastasis through the remodeling of tumor microvasculature. Knockdown of VEGF-B in tumors resulted in increased perivascular cell coverage and impaired pulmonary metastasis of human melanomas. In contrast, the gain of VEGF-B function in tumors led to pseudonormalized tumor vasculatures that were highly leaky and poorly perfused. Tumors expressing high levels of VEGF-B were more metastatic, although primary tumor growth was largely impaired. Similarly, VEGF-B in a VEGF-A-null tumor resulted in attenuated primary tumor growth but substantial pulmonary metastases. VEGF-B also led to highly metastatic phenotypes in Vegfr1 tk(-/-) mice and mice treated with anti-VEGF-A. These data indicate that VEGF-B promotes cancer metastasis through a VEGF-A-independent mechanism. High expression levels of VEGF-B in two large-cohort studies of human patients with lung squamous cell carcinoma and melanoma correlated with poor survival. Taken together, our findings demonstrate that VEGF-B is a vascular remodeling factor promoting cancer metastasis and that targeting VEGF-B may be an important therapeutic approach for cancer metastasis.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Microvasos/efectos de los fármacos , Metástasis de la Neoplasia/fisiopatología , Neoplasias/irrigación sanguínea , Factor B de Crecimiento Endotelial Vascular/metabolismo , Factor B de Crecimiento Endotelial Vascular/farmacología , Animales , Western Blotting , Hipoxia de la Célula , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Inyecciones Subcutáneas , Estimación de Kaplan-Meier , Ratones , Reacción en Cadena de la Polimerasa , Factor B de Crecimiento Endotelial Vascular/administración & dosificación , Pez Cebra
11.
Proc Natl Acad Sci U S A ; 110(2): 654-9, 2013 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-23267058

RESUMEN

The role of placental growth factor (PlGF) in modulation of tumor angiogenesis and tumor growth remains an enigma. Furthermore, anti-PlGF therapy in tumor angiogenesis and tumor growth remains controversial in preclinical tumor models. Here we show that in both human and mouse tumors, PlGF induced the formation of dilated and normalized vascular networks that were hypersensitive to anti-VEGF and anti-VEGFR-2 therapy, leading to dormancy of a substantial number of avascular tumors. Loss-of-function using plgf shRNA in a human choriocarcinoma significantly accelerated tumor growth rates and acquired resistance to anti-VEGF drugs, whereas gain-of-function of PlGF in a mouse tumor increased anti-VEGF sensitivity. Further, we show that VEGFR-2 and VEGFR-1 blocking antibodies displayed opposing effects on tumor angiogenesis. VEGFR-1 blockade and genetic deletion of the tyrosine kinase domain of VEGFR-1 resulted in enhanced tumor angiogenesis. These findings demonstrate that tumor-derived PlGF negatively modulates tumor angiogenesis and tumor growth and may potentially serve as a predictive marker of anti-VEGF cancer therapy.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Antineoplásicos/metabolismo , Coriocarcinoma/genética , Neovascularización Patológica/inducido químicamente , Proteínas Gestacionales/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Anticuerpos Bloqueadores/farmacología , Línea Celular Tumoral , Coriocarcinoma/metabolismo , Cartilla de ADN/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/metabolismo , Factor de Crecimiento Placentario , Proteínas Gestacionales/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
12.
Adv Sci (Weinh) ; 10(24): e2301505, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37330661

RESUMEN

The circadian clock in animals and humans plays crucial roles in multiple physiological processes. Disruption of circadian homeostasis causes detrimental effects. Here, it is demonstrated that the disruption of the circadian rhythm by genetic deletion of mouse brain and muscle ARNT-like 1 (Bmal1) gene, coding for the key clock transcription factor, augments an exacerbated fibrotic phenotype in various tumors. Accretion of cancer-associated fibroblasts (CAFs), especially the alpha smooth muscle actin positive myoCAFs, accelerates tumor growth rates and metastatic potentials. Mechanistically, deletion of Bmal1 abrogates expression of its transcriptionally targeted plasminogen activator inhibitor-1 (PAI-1). Consequently, decreased levels of PAI-1 in the tumor microenvironment instigate plasmin activation through upregulation of tissue plasminogen activator and urokinase plasminogen activator. The activated plasmin converts latent TGF-ß into its activated form, which potently induces tumor fibrosis and the transition of CAFs into myoCAFs, the latter promoting cancer metastasis. Pharmacological inhibition of the TGF-ß signaling largely ablates the metastatic potentials of colorectal cancer, pancreatic ductal adenocarcinoma, and hepatocellular carcinoma. Together, these data provide novel mechanistic insights into disruption of the circadian clock in tumor growth and metastasis. It is reasonably speculated that normalization of the circadian rhythm in patients provides a novel paradigm for cancer therapy.


Asunto(s)
Neoplasias Hepáticas , Factor de Crecimiento Transformador beta , Ratones , Humanos , Animales , Factor de Crecimiento Transformador beta/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Fibrinolisina/metabolismo , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Músculos , Encéfalo/metabolismo , Microambiente Tumoral
13.
Cancer Commun (Lond) ; 43(6): 637-660, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37120719

RESUMEN

BACKGROUND: Tumors possess incessant growth features, and expansion of their masses demands sufficient oxygen supply by red blood cells (RBCs). In adult mammals, the bone marrow (BM) is the main organ regulating hematopoiesis with dedicated manners. Other than BM, extramedullary hematopoiesis is discovered in various pathophysiological settings. However, whether tumors can contribute to hematopoiesis is completely unknown. Accumulating evidence shows that, in the tumor microenvironment (TME), perivascular localized cells retain progenitor cell properties and can differentiate into other cells. Here, we sought to better understand whether and how perivascular localized pericytes in tumors manipulate hematopoiesis. METHODS: To test if vascular cells can differentiate into RBCs, genome-wide expression profiling was performed using mouse-derived pericytes. Genetic tracing of perivascular localized cells employing NG2-CreERT2:R26R-tdTomato mouse strain was used to validate the findings in vivo. Fluorescence-activated cell sorting (FACS), single-cell sequencing, and colony formation assays were applied for biological studies. The production of erythroid differentiation-specific cytokine, erythropoietin (EPO), in TME was checked using quantitative polymerase chain reaction (qPCR), enzyme-linked immunosorbent assay (ELISA, magnetic-activated cell sorting and immunohistochemistry. To investigate BM function in tumor erythropoiesis, BM transplantation mouse models were employed. RESULTS: Genome-wide expression profiling showed that in response to platelet-derived growth factor subunit B (PDGF-B), neural/glial antigen 2 (NG2)+ perivascular localized cells exhibited hematopoietic stem and progenitor-like features and underwent differentiation towards the erythroid lineage. PDGF-B simultaneously targeted cancer-associated fibroblasts to produce high levels of EPO, a crucial hormone that necessitates erythropoiesis. FACS analysis using genetic tracing of NG2+ cells in tumors defined the perivascular localized cell-derived subpopulation of hematopoietic cells. Single-cell sequencing and colony formation assays validated the fact that, upon PDGF-B stimulation, NG2+ cells isolated from tumors acted as erythroblast progenitor cells, which were distinctive from the canonical BM hematopoietic stem cells. CONCLUSIONS: Our data provide a new concept of hematopoiesis within tumor tissues and novel mechanistic insights into perivascular localized cell-derived erythroid cells within TME. Targeting tumor hematopoiesis is a novel therapeutic concept for treating various cancers that may have profound impacts on cancer therapy.


Asunto(s)
Eritropoyesis , Neoplasias , Animales , Ratones , Médula Ósea/fisiología , Diferenciación Celular , Mamíferos , Neoplasias/metabolismo , Pericitos , Microambiente Tumoral
14.
Proc Natl Acad Sci U S A ; 106(41): 17505-10, 2009 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-19805167

RESUMEN

Vascular functions of PlGF remain poorly understood and controversial. Here, we show that tumor cell-derived PlGF-1 and PlGF-2 displayed significant remodeling effects on the tumor vasculature, leading to a normalized vascular phenotype and improved functions against leakage. In two murine tumor models, that is, T241 fibrosarcoma and Lewis lung carcinoma, stable expression of PlGF-1 and PlGF-2 in tumor cells resulted in significant reduction of tumor microvascular density and branch formation. Markedly, the vasculature in PlGF-expressing tumors consisted of relatively large-diameter microvessels with substantial improvement of pericyte coverage. Similarly, PlGF-induced vascular normalization and remodeling were also observed in a spontaneous human choriocarcinoma that expressed endogenous PlGF. Our findings shed light on functions of PlGF as a vascular remodeling factor that normalizes the tumor vasculature and thus may have conceptual implications of cancer therapy.


Asunto(s)
Neoplasias Pulmonares/irrigación sanguínea , Proteínas Gestacionales/uso terapéutico , Animales , Fibrosarcoma/irrigación sanguínea , Fibrosarcoma/tratamiento farmacológico , Humanos , Ratones , Neovascularización Patológica/patología , Pericitos/citología , Pericitos/patología , Pericitos/fisiología , Factor de Crecimiento Placentario , Factor A de Crecimiento Endotelial Vascular/farmacología , Factor C de Crecimiento Endotelial Vascular/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/uso terapéutico
15.
JCI Insight ; 7(10)2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35439170

RESUMEN

Molecular signaling in the tumor microenvironment (TME) is complex, and crosstalk among various cell compartments in supporting metastasis remains poorly understood. In particular, the role of vascular pericytes, a critical cellular component in the TME, in cancer invasion and metastasis warrants further investigation. Here, we report that an elevation of FGF-2 signaling in samples from patients with nasopharyngeal carcinoma (NPC) and xenograft mouse models promoted NPC metastasis. Mechanistically, tumor cell-derived FGF-2 strongly promoted pericyte proliferation and pericyte-specific expression of an orphan chemokine (C-X-C motif) ligand 14 (CXCL14) via FGFR1/AHR signaling. Gain- and loss-of-function experiments validated that pericyte-derived CXCL14 promoted macrophage recruitment and polarization toward an M2-like phenotype. Genetic knockdown of FGF2 or genetic depletion of tumoral pericytes blocked CXCL14 expression and tumor-associated macrophage (TAM) infiltration. Pharmacological inhibition of TAMs by clodronate liposome treatment resulted in a reduction of FGF-2-induced pulmonary metastasis. Together, these findings shed light on the inflammatory role of tumoral pericytes in promoting TAM-mediated metastasis. We provide mechanistic insight into an FGF-2/FGFR1/pericyte/CXCL14/TAM stromal communication axis in NPC and propose an effective antimetastasis therapy concept by targeting a pericyte-derived inflammation for NPC or FGF-2hi tumors.


Asunto(s)
Neoplasias Nasofaríngeas , Pericitos , Animales , Línea Celular Tumoral , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Macrófagos/metabolismo , Ratones , Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Pericitos/metabolismo , Microambiente Tumoral
16.
Cell Death Dis ; 13(8): 724, 2022 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-35985991

RESUMEN

Nasopharyngeal carcinoma (NPC) clinical trials show that antiangiogenic drugs (AADs) fail to achieve the expected efficacy, and combining AAD with chemoradiotherapy does not show superiority over chemoradiotherapy alone. Accumulating evidence suggests the intrinsic AAD resistance in NPC patients with poorly understood molecular mechanisms. Here, we describe NPC-specific FGF-2 expression-triggered, VEGF-independent angiogenesis as a mechanism of AAD resistance. Angiogenic factors screening between AAD-sensitive cancer type and AAD-resistant NPC showed high FGF-2 expression in NPC in both xenograft models and clinical samples. Mechanistically, the FGF-2-FGFR1-MYC axis drove endothelial cell survival and proliferation as an alternative to VEGF-VEGFR2-MYC signaling. Genetic knockdown of FGF-2 in NPC tumor cells reduced tumor angiogenesis, enhanced AAD sensitivity, and reduced pulmonary metastasis. Moreover, lenvatinib, an FDA recently approved multi-kinase inhibitor targeting both VEGFR2 and FGFR1, effectively inhibits the tumor vasculature, and exhibited robust anti-tumor effects in NPC-bearing nude mice and humanized mice compared with an agent equivalent to bevacizumab. These findings provide mechanistic insights on FGF-2 signaling in the modulation of VEGF pathway activation in the NPC microenvironment and propose an effective NPC-targeted therapy by using a clinically available drug.


Asunto(s)
Inhibidores de la Angiogénesis , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas , Compuestos de Fenilurea , Quinolinas , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Ratones , Ratones Desnudos , Carcinoma Nasofaríngeo/tratamiento farmacológico , Neoplasias Nasofaríngeas/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Compuestos de Fenilurea/farmacología , Compuestos de Fenilurea/uso terapéutico , Proteínas Proto-Oncogénicas c-myc/metabolismo , Quinolinas/farmacología , Quinolinas/uso terapéutico , Transducción de Señal , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Nat Metab ; 4(12): 1674-1683, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36482111

RESUMEN

Patients with COVID-19 frequently manifest adipose atrophy, weight loss and cachexia, which significantly contribute to poor quality of life and mortality1,2. Browning of white adipose tissue and activation of brown adipose tissue are effective processes for energy expenditure3-7; however, mechanistic and functional links between SARS-CoV-2 infection and adipose thermogenesis have not been studied. In this study, we provide experimental evidence that SARS-CoV-2 infection augments adipose browning and non-shivering thermogenesis (NST), which contributes to adipose atrophy and body weight loss. In mouse and hamster models, SARS-CoV-2 infection activates brown adipose tissue and instigates a browning or beige phenotype of white adipose tissues, including augmented NST. This browning phenotype was also observed in post-mortem adipose tissue of four patients who died of COVID-19. Mechanistically, high levels of vascular endothelial growth factor (VEGF) in the adipose tissue induces adipose browning through vasculature-adipocyte interaction. Inhibition of VEGF blocks COVID-19-induced adipose tissue browning and NST and partially prevents infection-induced body weight loss. Our data suggest that the browning of adipose tissues induced by COVID-19 can contribute to adipose tissue atrophy and weight loss observed during infection. Inhibition of VEGF signaling may represent an effective approach for preventing and treating COVID-19-associated weight loss.


Asunto(s)
COVID-19 , Factor A de Crecimiento Endotelial Vascular , Ratones , Animales , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Calidad de Vida , COVID-19/metabolismo , SARS-CoV-2 , Tejido Adiposo Pardo/metabolismo , Obesidad/metabolismo , Pérdida de Peso , Mamíferos
18.
Nat Commun ; 11(1): 3704, 2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32709869

RESUMEN

FGF-2 displays multifarious functions in regulation of angiogenesis and vascular remodeling. However, effective drugs for treating FGF-2+ tumors are unavailable. Here we show that FGF-2 modulates tumor vessels by recruiting NG2+ pricytes onto tumor microvessels through a PDGFRß-dependent mechanism. FGF-2+ tumors are intrinsically resistant to clinically available drugs targeting VEGF and PDGF. Surprisingly, dual targeting the VEGF and PDGF signaling produces a superior antitumor effect in FGF-2+ breast cancer and fibrosarcoma models. Mechanistically, inhibition of PDGFRß ablates FGF-2-recruited perivascular coverage, exposing anti-VEGF agents to inhibit vascular sprouting. These findings show that the off-target FGF-2 is a resistant biomarker for anti-VEGF and anti-PDGF monotherapy, but a highly beneficial marker for combination therapy. Our data shed light on mechanistic interactions between various angiogenic and remodeling factors in tumor neovascularization. Optimization of antiangiogenic drugs with different principles could produce therapeutic benefits for treating their resistant off-target cancers.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Factor 2 de Crecimiento de Fibroblastos/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Neoplasias/tratamiento farmacológico , Factor de Crecimiento Derivado de Plaquetas/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Animales , Biomarcadores de Tumor , Presión Sanguínea , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Permeabilidad Capilar , Proliferación Celular , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Quimioterapia Combinada , Humanos , Ratones , Ratones Endogámicos C57BL , Neoplasias/patología , Neovascularización Patológica/tratamiento farmacológico , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas , Transducción de Señal/efectos de los fármacos , Hipoxia Tumoral , Microambiente Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
Lymphat Res Biol ; 6(1): 15-27, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18361767

RESUMEN

The immunohistochemical properties of selective lymph vessel markers, and NO synthase (NOS) and cyclo-oxygenase (COX) activities, were examined in two kinds of human lymphatic endothelial cells isolated from collecting (macro-) and initial (micro-) lymph vessels. The constitutively expressed genes in the two kinds of lymphatic endothelial cells were also evaluated by using oligonucleotide microarray analysis and RT-PCR. We also investigated the effects of oxygen concentration in culture conditions or growth factors such as basic fibroblast growth factor (bFGF), VEGF-A, and VEGF-C on proliferation activities of the two kinds of human lymphatic endothelial cells. Immunoreactivity to LYVE-1 and the RT-PCR expression level of LYVE-1 mRNA in endothelial cells of micro-lymph vessels were stronger than those of macro-lymph vessels. Immunoreactivity to VEGF R1 was also observed as significantly stronger in the micro-lymph vessels. In contrast, the immunoreactivity to Prox-1 and the RT-PCR expression level of Prox-1 mRNA in endothelial cells of macro-lymph vessels were stronger than those of micro-lymph vessels. Similarly, immunoreactivity to ecNOS, iNOS, COX1, and COX2 was also found as significantly higher than in macro-lymph vessels. In contrast, the increase of O(2) concentration ranging from 5% to 21% caused a significant reduction of the proliferation activity of endothelial cells in macro-lymph vessels. In conclusion, these findings suggest marked heterogeneity in the immunohistochemical, genomic, and proliferation activity of human lymphatic endothelial cells between micro-(initial) and macro-(collecting) lymph vessels.


Asunto(s)
Células Endoteliales/metabolismo , Vasos Linfáticos/anatomía & histología , Vasos Linfáticos/metabolismo , Proliferación Celular , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Expresión Génica , Humanos , Inmunohistoquímica , Óxido Nítrico Sintasa/biosíntesis , Análisis de Secuencia por Matrices de Oligonucleótidos , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Transporte Vesicular/biosíntesis
20.
JCI Insight ; 3(20)2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30333314

RESUMEN

Molecular mechanisms underlying the cancer stroma in metastasis need further exploration. Here, we discovered that cancer-associated fibroblasts (CAFs) produced high levels of IL-33 that acted on tumor-associated macrophages (TAMs), causing them to undergo the M1 to M2 transition. Genomic profiling of metastasis-related genes in the IL-33-stimulated TAMs showed a >200-fold increase of MMP9. Signaling analysis demonstrated the IL-33-ST2-NF-κB-MMP9-laminin pathway that governed tumor stroma-mediated metastasis. In mouse and human fibroblast-rich pancreatic cancers, genetic deletion of IL-33, ST2, or MMP9 markedly blocked metastasis. Pharmacological inhibition of NF-κB and MMP9 also blocked cancer metastasis. Deletion of IL-33, ST2, or MMP9 restored laminin, a key basement membrane component associated with tumor microvessels. Together, our data provide mechanistic insights on the IL-33-NF-κB-MMP9-laminin axis that mediates the CAF-TAM-committed cancer metastasis. Thus, targeting the CAF-TAM-vessel axis provides an outstanding therapeutic opportunity for cancer treatment.


Asunto(s)
Interleucina-33/metabolismo , Neoplasias/inmunología , Microambiente Tumoral/inmunología , Animales , Fibroblastos Asociados al Cáncer/inmunología , Fibroblastos Asociados al Cáncer/metabolismo , Comunicación Celular/inmunología , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/genética , Interleucina-33/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Ratones , Ratones Noqueados , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Neoplasias/patología , Transducción de Señal/inmunología , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA