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













Base de datos
Intervalo de año de publicación
1.
FASEB J ; 37(6): e22995, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37219526

RESUMEN

Immuno-oncology (IO)-based therapies such as checkpoint inhibitors, bi-specific antibodies, and CAR-T-cell therapies have shown significant success in the treatment of several cancer indications. However, these therapies can result in the development of severe adverse events, including cytokine release syndrome (CRS). Currently, there is a paucity of in vivo models that can evaluate dose-response relationships for both tumor control and CRS-related safety issues. We tested an in vivo PBMC humanized mouse model to assess both treatment efficacy against specific tumors and the concurrent cytokine release profiles for individual human donors after treatment with a CD19xCD3 bispecific T-cell engager (BiTE). Using this model, we evaluated tumor burden, T-cell activation, and cytokine release in response to bispecific T-cell-engaging antibody in humanized mice generated with different PBMC donors. The results show that PBMC engrafted NOD-scid Il2rgnull mice lacking expression of mouse MHC class I and II (NSG-MHC-DKO mice) and implanted with a tumor xenograft predict both efficacy for tumor control by CD19xCD3 BiTE and stimulated cytokine release. Moreover, our findings indicate that this PBMC-engrafted model captures variability among donors for tumor control and cytokine release following treatment. Tumor control and cytokine release were reproducible for the same PBMC donor in separate experiments. The PBMC humanized mouse model described here is a sensitive and reproducible platform that identifies specific patient/cancer/therapy combinations for treatment efficacy and development of complications.


Asunto(s)
Leucocitos Mononucleares , Linfocitos T , Humanos , Animales , Ratones , Ratones Endogámicos NOD , Resultado del Tratamiento , Síndrome de Liberación de Citoquinas , Citocinas , Modelos Animales de Enfermedad , Ratones Noqueados , Ratones SCID
2.
FASEB J ; 36(9): e22476, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35959876

RESUMEN

Human innate immunity plays a critical role in tumor surveillance and in immunoregulation within the tumor microenvironment. Natural killer (NK) cells are innate lymphoid cells that have opposing roles in the tumor microenvironment, including NK cell subsets that mediate tumor cell cytotoxicity and subsets with regulatory function that contribute to the tumor immune suppressive environment. The balance between effector and regulatory NK cell subsets has been studied extensively in murine models of cancer, but there is a paucity of models to study human NK cell function in tumorigenesis. Humanized mice are a powerful alternative to syngeneic mouse tumor models for the study of human immuno-oncology and have proven effective tools to test immunotherapies targeting T cells. However, human NK cell development and survival in humanized NOD-scid-IL2rgnull (NSG) mice are severely limited. To enhance NK cell development, we have developed NSG mice that constitutively expresses human Interleukin 15 (IL15), NSG-Tg(Hu-IL15). Following hematopoietic stem cell engraftment of NSG-Tg(Hu-IL15) mice, significantly higher levels of functional human CD56+ NK cells are detectable in blood and spleen, as compared to NSG mice. Hematopoietic stem cell (HSC)-engrafted NSG-Tg(Hu-IL15) mice also supported the development of human CD3+ T cells, CD20+ B cells, and CD33+ myeloid cells. Moreover, the growth kinetics of a patient-derived xenograft (PDX) melanoma were significantly delayed in HSC-engrafted NSG-Tg(Hu-IL15) mice as compared to HSC-engrafted NSG mice demonstrating that human NK cells have a key role in limiting the tumor growth. Together, these data demonstrate that HSC-engrafted NSG-Tg(Hu-IL15) mice support enhanced development of functional human NK cells, which limit the growth of PDX tumors.


Asunto(s)
Inmunidad Innata , Interleucina-15 , Animales , Modelos Animales de Enfermedad , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Interleucina-15/genética , Células Asesinas Naturales , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID
3.
EMBO Mol Med ; 12(7): e8662, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32578942

RESUMEN

Mice xenotransplanted with human cells and/or expressing human gene products (also known as "humanized mice") recapitulate the human evolutionary specialization and diversity of genotypic and phenotypic traits. These models can provide a relevant in vivo context for understanding of human-specific physiology and pathologies. Humanized mice have advanced toward mainstream preclinical models and are now at the forefront of biomedical research. Here, we considered innovations and challenges regarding the reconstitution of human immunity and human tissues, modeling of human infections and cancer, and the use of humanized mice for testing drugs or regenerative therapy products. As the number of publications exploring different facets of humanized mouse models has steadily increased in past years, it is becoming evident that standardized reporting is needed in the field. Therefore, an international community-driven resource called "Minimal Information for Standardization of Humanized Mice" (MISHUM) has been created for the purpose of enhancing rigor and reproducibility of studies in the field. Within MISHUM, we propose comprehensive guidelines for reporting critical information generated using humanized mice.


Asunto(s)
Modelos Animales de Enfermedad , Guías como Asunto , Xenoinjertos/normas , Animales , Humanos , Ratones , Ratones SCID , Neoplasias , Reproducibilidad de los Resultados
4.
Methods Mol Biol ; 1953: 241-252, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30912026

RESUMEN

A significant obstacle to the study of human cancer biology and the testing of human specific immunotherapeutics is the paucity of translational models that recapitulate both the growth of human tumors and the functionality of human immune systems. Humanized mice engrafted with human hematopoietic stem cells (HSC) and patient-derived xenografts (PDX) enable preclinical investigation of the interactions between the human immune system and human cancer. We use immunodeficient non-obese diabetic (NOD, scid, gamma) NSG™ or NSG™-SGM3 mice as hosts for establishment of human immunity following HSC injection and for engraftment of human tumors. Here we describe a refined protocol for the subcutaneous implant of solid PDX tumors into humanized mice. Protocols to recover infiltrating immune cells from growing tumors and to evaluate the immune cell subsets by flow cytometry are also described.


Asunto(s)
Trasplante de Neoplasias/métodos , Neoplasias/inmunología , Trasplante Heterólogo/métodos , Animales , Citometría de Flujo/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Inmunidad , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Ratones Endogámicos NOD , Ratones SCID , Neoplasias/patología
5.
Mol Cancer Ther ; 17(12): 2530-2542, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30232146

RESUMEN

Tumors use indoleamine 2,3-dioxygenase-1 (IDO1) as a major mechanism to induce an immunosuppressive microenvironment. IDO1 expression is upregulated in many cancers and considered to be a resistance mechanism to immune checkpoint therapies. IDO1 is induced in response to inflammatory stimuli such as IFNγ and promotes immune tolerance by depleting tryptophan and producing tryptophan catabolites, including kynurenine, in the tumor microenvironment. This leads to effector T-cell anergy and enhanced Treg function through upregulation of FoxP3. As a nexus for the induction of key immunosuppressive mechanisms, IDO1 represents an important immunotherapeutic target in oncology. Here, we report the identification and characterization of the novel selective, orally bioavailable IDO1 inhibitor EOS200271/PF-06840003. It reversed IDO1-induced T-cell anergy in vitro In mice carrying syngeneic tumor grafts, PF-06840003 reduced intratumoral kynurenine levels by over 80% and inhibited tumor growth both in monotherapy and, with an increased efficacy, in combination with antibodies blocking the immune checkpoint ligand PD-L1. We demonstrate that anti-PD-L1 therapy results in increased IDO1 metabolic activity thereby providing additional mechanistic rationale for combining PD-(L)1 blockade with IDO1 inhibition in cancer immunotherapies. Supported by these preclinical data and favorable predicted human pharmacokinetic properties of PF-06840003, a phase I open-label, multicenter clinical study (NCT02764151) has been initiated.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Biocatálisis , Inhibidores Enzimáticos/farmacología , Inmunoterapia , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Indoles/farmacología , Succinimidas/farmacología , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Antineoplásicos/farmacología , Antígeno B7-H1/metabolismo , Antígeno CTLA-4/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón gamma/metabolismo , Quinurenina/sangre , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Estereoisomerismo , Especificidad por Sustrato/efectos de los fármacos , Linfocitos T/citología , Linfocitos T/efectos de los fármacos
6.
FASEB J ; 32(3): 1537-1549, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29146734

RESUMEN

Establishment of an in vivo small animal model of human tumor and human immune system interaction would enable preclinical investigations into the mechanisms underlying cancer immunotherapy. To this end, nonobese diabetic (NOD).Cg- PrkdcscidIL2rgtm1Wjl/Sz (null; NSG) mice were transplanted with human (h)CD34+ hematopoietic progenitor and stem cells, which leads to the development of human hematopoietic and immune systems [humanized NSG (HuNSG)]. HuNSG mice received human leukocyte antigen partially matched tumor implants from patient-derived xenografts [PDX; non-small cell lung cancer (NSCLC), sarcoma, bladder cancer, and triple-negative breast cancer (TNBC)] or from a TNBC cell line-derived xenograft (CDX). Tumor growth curves were similar in HuNSG compared with nonhuman immune-engrafted NSG mice. Treatment with pembrolizumab, which targets programmed cell death protein 1, produced significant growth inhibition in both CDX and PDX tumors in HuNSG but not in NSG mice. Finally, inhibition of tumor growth was dependent on hCD8+ T cells, as demonstrated by antibody-mediated depletion. Thus, tumor-bearing HuNSG mice may represent an important, new model for preclinical immunotherapy research.-Wang, M., Yao, L.-C., Cheng, M., Cai, D., Martinek, J., Pan, C.-X., Shi, W., Ma, A.-H., De Vere White, R. W., Airhart, S., Liu, E. T., Banchereau, J., Brehm, M. A., Greiner, D. L., Shultz, L. D., Palucka, K., Keck, J. G. Humanized mice in studying efficacy and mechanisms of PD-1-targeted cancer immunotherapy.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Linfocitos T CD8-positivos/inmunología , Inmunidad Celular/efectos de los fármacos , Inmunoterapia , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/inmunología , Animales , Linfocitos T CD8-positivos/patología , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Neoplasias/inmunología , Neoplasias/patología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
JCI Insight ; 2(16)2017 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-28814666

RESUMEN

Lymphatic malformations are serious but poorly understood conditions that present therapeutic challenges. The goal of this study was to compare strategies for inducing regression of abnormal lymphatics and explore underlying mechanisms. CCSP-rtTA/tetO-VEGF-C mice, in which doxycycline regulates VEGF-C expression in the airway epithelium, were used as a model of pulmonary lymphangiectasia. After doxycycline was stopped, VEGF-C expression returned to normal, but lymphangiectasia persisted for at least 9 months. Inhibition of VEGFR-2/VEGFR-3 signaling, Notch, ß-adrenergic receptors, or autophagy and antiinflammatory steroids had no noticeable effect on the amount or severity of lymphangiectasia. However, rapamycin inhibition of mTOR reduced lymphangiectasia by 76% within 7 days without affecting normal lymphatics. Efficacy of rapamycin was not increased by coadministration with the other agents. In prevention trials, rapamycin suppressed VEGF-C-driven mTOR phosphorylation and lymphatic endothelial cell sprouting and proliferation. However, in reversal trials, no lymphatic endothelial cell proliferation was present to block in established lymphangiectasia, and rapamycin did not increase caspase-dependent apoptosis. However, rapamycin potently suppressed Prox1 and VEGFR-3. These experiments revealed that lymphangiectasia is remarkably resistant to regression but is responsive to rapamycin, which rapidly reduces and normalizes the abnormal lymphatics without affecting normal lymphatics.

8.
Am J Pathol ; 184(6): 1877-89, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24726646

RESUMEN

Vascular remodeling is a feature of sustained inflammation in which capillaries enlarge and acquire the phenotype of venules specialized for plasma leakage and leukocyte recruitment. We sought to determine whether neutrophils are required for vascular remodeling in the respiratory tract by using Mycoplasma pulmonis infection as a model of sustained inflammation in mice. The time course of vascular remodeling coincided with the influx of neutrophils during the first few days after infection and peaked at day 5. Depletion of neutrophils with antibody RB6-8C5 or 1A8 reduced neutrophil influx and vascular remodeling after infection by about 90%. Similarly, vascular remodeling after infection was suppressed in Cxcr2(-/-) mice, in which neutrophils adhered to the endothelium of venules but did not extravasate into the tissue. Expression of the venular adhesion molecule P-selectin increased in endothelial cells from day 1 to day 3 after infection, as did expression of the Cxcr2-receptor ligands Cxcl1 and Cxcl2. Tumor necrosis factor α (TNFα) expression increased more than sixfold in the trachea of wild-type and Cxcr2(-/-) mice, but intratracheal administration of TNFα did not induce vascular remodeling similar to that seen in infection. We conclude that neutrophil influx is required for remodeling of capillaries into venules in the airways of mice with Mycoplasma infection and that TNFα signaling is necessary but not sufficient for vascular remodeling.


Asunto(s)
Endotelio Vascular/metabolismo , Infecciones por Mycoplasma/metabolismo , Mycoplasma pulmonis , Neutrófilos/metabolismo , Sistema Respiratorio/metabolismo , Remodelación Vascular , Animales , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/metabolismo , Endotelio Vascular/patología , Femenino , Ratones , Ratones Noqueados , Infecciones por Mycoplasma/genética , Infecciones por Mycoplasma/patología , Neutrófilos/patología , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo , Sistema Respiratorio/patología
9.
Circ Res ; 114(5): 806-22, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24429550

RESUMEN

RATIONALE: Lymphatic vessels in the respiratory tract normally mature into a functional network during the neonatal period, but under some pathological conditions they can grow as enlarged, dilated sacs that result in the potentially lethal condition of pulmonary lymphangiectasia. OBJECTIVE: We sought to determine whether overexpression of the lymphangiogenic growth factor (vascular endothelial growth factor-C [VEGF-C]) can promote lymphatic growth and maturation in the respiratory tract. Unexpectedly, perinatal overexpression of VEGF-C in the respiratory epithelium led to a condition resembling human pulmonary lymphangiectasia, a life-threatening disorder of the newborn characterized by respiratory distress and the presence of widely dilated lymphatics. METHODS AND RESULTS: Administration of doxycycline to Clara cell secretory protein-reverse tetracycline-controlled transactivator/tetracycline operator-VEGF-C double-transgenic mice during a critical period from embryonic day 15.5 to postnatal day 14 was accompanied by respiratory distress, chylothorax, pulmonary lymphangiectasia, and high mortality. Enlarged sac-like lymphatics were abundant near major airways, pulmonary vessels, and visceral pleura. Side-by-side comparison revealed morphological features similar to pulmonary lymphangiectasia in humans. The condition was milder in mice given doxycycline after age postnatal day 14 and did not develop after postnatal day 35. Mechanistic studies revealed that VEGF recptor (VEGFR)-3 alone drove lymphatic growth in adult mice, but both VEGFR-2 and VEGFR-3 were required for the development of lymphangiectasia in neonates. VEGFR-2/VEGFR-3 heterodimers were more abundant in the dilated lymphatics, consistent with the involvement of both receptors. Despite the dependence of lymphangiectasia on VEGFR-2 and VEGFR-3, the condition was not reversed by blocking both receptors together or by withdrawing VEGF-C. CONCLUSIONS: The findings indicate that VEGF-C overexpression can induce pulmonary lymphangiectasia during a critical period in perinatal development.


Asunto(s)
Enfermedades Pulmonares/congénito , Linfangiectasia/congénito , Factor C de Crecimiento Endotelial Vascular/genética , Animales , Femenino , Humanos , Lactante , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Linfangiectasia/genética , Linfangiectasia/metabolismo , Linfangiectasia/patología , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Masculino , Ratones , Ratones Endogámicos , Ratones Transgénicos , Embarazo , Edema Pulmonar/genética , Edema Pulmonar/metabolismo , Edema Pulmonar/patología , Transducción de Señal/fisiología , Tráquea/metabolismo , Tráquea/patología , Uteroglobina/genética , Uteroglobina/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
10.
Adv Anat Embryol Cell Biol ; 214: 41-54, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24276885

RESUMEN

The dynamic nature of lymphatic vessels is reflected by structural and functional modifications that coincide with changes in their environment. Lymphatics in the respiratory tract undergo rapid changes around birth, during adaptation to air breathing, when lymphatic endothelial cells develop button-like intercellular junctions specialized for efficient fluid uptake and transport. In inflammatory conditions, lymphatic vessels proliferate and undergo remodeling to accommodate greater plasma leakage and immune cell trafficking. However, the newly formed lymphatics are abnormal, and resolution of inflammation is not accompanied by complete reversal of the lymphatic vessel changes back to the baseline. As the understanding of lymphatic plasticity advances, approaches for eliminating the abnormal vessels and improving the functionality of those that remain move closer to reality. This chapter provides an overview of what is known about lymphatic vessel growth, remodeling, and other forms of plasticity that occur during development or inflammation, with an emphasis on the respiratory tract. Also addressed is the limited reversibility of changes in lymphatics during the resolution of inflammation.


Asunto(s)
Linfangiogénesis , Vasos Linfáticos/embriología , Vasos Linfáticos/fisiología , Sistema Respiratorio/embriología , Enfermedades Respiratorias/fisiopatología , Animales , Humanos , Sistema Respiratorio/fisiopatología
11.
Am J Pathol ; 180(6): 2561-75, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22538088

RESUMEN

Endothelial cells of initial lymphatics have discontinuous button-like junctions (buttons), unlike continuous zipper-like junctions (zippers) of collecting lymphatics and blood vessels. Buttons are thought to act as primary valves for fluid and cell entry into lymphatics. To learn when and how buttons form during development and whether they change in disease, we examined the appearance of buttons in mouse embryos and their plasticity in sustained inflammation. We found that endothelial cells of lymph sacs at embryonic day (E)12.5 and tracheal lymphatics at E16.5 were joined by zippers, not buttons. However, zippers in initial lymphatics decreased rapidly just before birth, as buttons appeared. The proportion of buttons increased from only 6% at E17.5 and 12% at E18.5 to 35% at birth, 50% at postnatal day (P)7, 90% at P28, and 100% at P70. In inflammation, zippers replaced buttons in airway lymphatics at 14 and 28 days after Mycoplasma pulmonis infection of the respiratory tract. The change in lymphatic junctions was reversed by dexamethasone but not by inhibition of vascular endothelial growth factor receptor-3 signaling by antibody mF4-31C1. Dexamethasone also promoted button formation during early postnatal development through a direct effect involving glucocorticoid receptor phosphorylation in lymphatic endothelial cells. These findings demonstrate the plasticity of intercellular junctions in lymphatics during development and inflammation and show that button formation can be promoted by glucocorticoid receptor signaling in lymphatic endothelial cells.


Asunto(s)
Endotelio Linfático/anatomía & histología , Infecciones por Mycoplasma/patología , Mycoplasma pulmonis , Envejecimiento/patología , Animales , Animales Recién Nacidos , Dexametasona/farmacología , Dexametasona/uso terapéutico , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio Linfático/efectos de los fármacos , Endotelio Linfático/embriología , Endotelio Linfático/crecimiento & desarrollo , Femenino , Glucocorticoides/farmacología , Glucocorticoides/uso terapéutico , Uniones Intercelulares/fisiología , Uniones Intercelulares/ultraestructura , Pulmón/anatomía & histología , Pulmón/embriología , Pulmón/crecimiento & desarrollo , Linfangiogénesis/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Infecciones por Mycoplasma/tratamiento farmacológico , Receptores de Glucocorticoides/metabolismo , Uniones Estrechas/metabolismo , Tráquea/anatomía & histología , Tráquea/efectos de los fármacos , Tráquea/embriología , Tráquea/crecimiento & desarrollo
12.
Proc Am Thorac Soc ; 8(6): 504-7, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22052927

RESUMEN

Blood vessels and lymphatic vessels in the respiratory tract play key roles in inflammation. By undergoing adaptive remodeling and growth, blood vessels undergo changes that enable the extravasation of plasma and leukocytes into inflamed tissues, and lymphatic vessels adjust to the increased fluid clearance and cell traffic involved in immune responses. Blood vessels and lymphatics in adult airways are strikingly different from those of late-stage embryos. Before birth, blood vessels in mouse airways make up a primitive plexus similar to that of the yolk sac. This plexus undergoes rapid and extensive remodeling at birth. In the early neonatal period, parts of the plexus regress. Capillaries then rapidly regrow, and with arterioles and venules form the characteristic adult vascular pattern. Lymphatic vessels of the airways also undergo rapid changes around birth, when lymphatic endothelial cells develop button-like intercellular junctions specialized for efficient fluid uptake. Among the mechanisms that underlie the onset of rapid vascular remodeling at birth, changes in tissue oxygen tension and mechanical forces associated with breathing are likely to be involved, along with growth factors that promote the growth and maturation of blood vessels and lymphatics. Whatever the mechanisms, the dynamic nature of airway blood vessels and lymphatics during perinatal development foretells the extraordinary vascular plasticity found in many diseases.


Asunto(s)
Inflamación/fisiopatología , Sistema Respiratorio/irrigación sanguínea , Enfermedades Respiratorias/fisiopatología , Animales , Capilares/fisiología , Humanos , Vasos Linfáticos/fisiología , Ratones , Modelos Animales
13.
Cancer Res ; 71(14): 4758-68, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21613405

RESUMEN

Angiogenesis inhibitors that block VEGF receptor (VEGFR) signaling slow the growth of many types of tumors, but eventually the disease progresses. Multiple strategies are being explored to improve efficacy by concurrent inhibition of other functionally relevant receptor tyrosine kinases (RTK). XL880 (foretinib, GSK1363089) and XL184 (cabozantinib) are small-molecule inhibitors that potently block multiple RTKs, including VEGFR and the receptor of hepatocyte growth factor c-Met, which can drive tumor invasion and metastasis. This study compared the cellular effects of XL880 and XL184 with those of an RTK inhibitor (XL999) that blocks VEGFR but not c-Met. Treatment of RIP-Tag2 mice with XL999 resulted in 43% reduction in vascularity of spontaneous pancreatic islet tumors over 7 days, but treatment with XL880 or XL184 eliminated approximately 80% of the tumor vasculature, reduced pericytes and empty basement membrane sleeves, caused widespread intratumoral hypoxia and tumor cell apoptosis, and slowed regrowth of the tumor vasculature after drug withdrawal. Importantly, XL880 and XL184 also decreased invasiveness of primary tumors and reduced metastasis. Overall, these findings indicate that inhibition of c-Met and functionally related kinases amplifies the effects of VEGFR blockade and leads to rapid, robust, and progressive regression of tumor vasculature, increased intratumoral hypoxia and apoptosis, and reduced tumor invasiveness and metastasis.


Asunto(s)
Adenoma de Células de los Islotes Pancreáticos/irrigación sanguínea , Adenoma de Células de los Islotes Pancreáticos/tratamiento farmacológico , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Adenoma de Células de los Islotes Pancreáticos/patología , Anilidas/farmacología , Animales , Apoptosis/efectos de los fármacos , Membrana Basal/efectos de los fármacos , Membrana Basal/metabolismo , Membrana Basal/patología , Hipoxia de la Célula/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas c-met/metabolismo , Piridinas/farmacología , Quinolinas/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Receptor 3 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 3 de Factores de Crecimiento Endotelial Vascular/biosíntesis
14.
Dev Dyn ; 239(9): 2354-66, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20730909

RESUMEN

Recent advances have documented the development of lung vasculature before and after birth, but less is known of the growth and maturation of airway vasculature. We sought to determine whether airway vasculature changes during the perinatal period and when the typical adult pattern develops. On embryonic day 16.5 mouse tracheas had a primitive vascular plexus unlike the adult airway vasculature, but instead resembling the yolk sac vasculature. Soon after birth (P0), the primitive vascular plexus underwent abrupt and extensive remodeling. Blood vessels overlying tracheal cartilage rings regressed from P1 to P3 but regrew from P4 to P7 to form the hierarchical, segmented, ladder-like adult pattern. Hypoxia and HIF-1α were present in tracheal epithelium over vessels that survived but not where they regressed. These findings reveal the plasticity of airway vasculature after birth and show that these vessels can be used to elucidate factors that promote postnatal vascular remodeling and maturation.


Asunto(s)
Vasos Sanguíneos , Pulmón , Neovascularización Fisiológica/fisiología , Animales , Animales Recién Nacidos , Apoptosis/fisiología , Vasos Sanguíneos/anatomía & histología , Vasos Sanguíneos/embriología , Vasos Sanguíneos/crecimiento & desarrollo , Proliferación Celular , Células Endoteliales/citología , Células Endoteliales/fisiología , Femenino , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Pulmón/irrigación sanguínea , Pulmón/embriología , Pulmón/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Embarazo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
15.
Am J Pathol ; 176(3): 1525-41, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20093490

RESUMEN

Angiogenesis and lymphangiogenesis participate in many inflammatory diseases, and their reversal is thought to be beneficial. However, the extent of reversibility of vessel remodeling is poorly understood. We exploited the potent anti-inflammatory effects of the corticosteroid dexamethasone to test the preventability and reversibility of vessel remodeling in Mycoplasma pulmonis-infected mice using immunohistochemistry and quantitative RT-PCR. In this model robust immune responses drive rapid and sustained changes in blood vessels and lymphatics. In infected mice not treated with dexamethasone, capillaries enlarged into venules expressing leukocyte adhesion molecules, sprouting angiogenesis and lymphangiogenesis occurred, and the inflammatory cytokines tumor necrosis factor and interleukin-1 increased. Concurrent dexamethasone treatment largely prevented the remodeling of blood vessels and lymphatics. Dexamethasone also significantly reduced cytokine expression, bacterial burden, and leukocyte influx into airways and lungs over 4 weeks of infection. In contrast, when infection was allowed to proceed untreated for 2 weeks and then was treated with dexamethasone for 4 weeks, most blood vessel changes reversed but lymphangiogenesis did not, suggesting that different survival mechanisms apply. Furthermore, dexamethasone significantly reduced the bacterial burden and influx of lymphocytes but not of neutrophils or macrophages or cytokine expression. These findings show that lymphatic remodeling is more resistant than blood vessel remodeling to corticosteroid-induced reversal. We suggest that lymphatic remodeling that persists after the initial inflammatory response has resolved may influence subsequent inflammatory episodes in clinical situations.


Asunto(s)
Dexametasona/farmacología , Inflamación/patología , Vasos Linfáticos/efectos de los fármacos , Vasos Linfáticos/patología , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/patología , Animales , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/microbiología , Vasos Sanguíneos/patología , Movimiento Celular/efectos de los fármacos , Enfermedad Crónica , Citocinas/metabolismo , Femenino , Inflamación/complicaciones , Inflamación/microbiología , Mediadores de Inflamación/metabolismo , Leucocitos/efectos de los fármacos , Leucocitos/patología , Vasos Linfáticos/microbiología , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Infecciones por Mycoplasma/complicaciones , Infecciones por Mycoplasma/microbiología , Infecciones por Mycoplasma/patología , Sistema Respiratorio/microbiología , Factores de Tiempo
16.
J Clin Invest ; 119(10): 2954-64, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19759514

RESUMEN

Inflammation is associated with blood vessel and lymphatic vessel proliferation and remodeling. The microvasculature of the mouse trachea provides an ideal opportunity to study this process, as Mycoplasma pulmonis infection of mouse airways induces widespread and sustained vessel remodeling, including enlargement of capillaries into venules and lymphangiogenesis. Although the mediators responsible for these vascular changes in mice have not been identified, VEGF-A is known not to be involved. Here, we sought to determine whether TNF-alpha drives the changes in blood vessels and lymphatics in M. pulmonis-infected mice. The endothelial cells, but not pericytes, of blood vessels, but not lymphatics, were immunoreactive for TNF receptor 1 (TNF-R1) and lymphotoxin B receptors. Most TNF-R2 immunoreactivity was on leukocytes. Infection resulted in a large and sustained increase in TNF-alpha expression, as measured by real-time quantitative RT-PCR, and smaller increases in lymphotoxins and TNF receptors that preceded vessel remodeling. Substantially less vessel remodeling and lymphangiogenesis occurred when TNF-alpha signaling was inhibited by a blocking antibody or was silenced in Tnfr1-/- mice. When administered after infection was established, the TNF-alpha-specific antibody slowed but did not reverse blood vessel remodeling and lymphangiogenesis. The action of TNF-alpha on blood vessels is probably mediated through direct effects on endothelial cells, but its effects on lymphangiogenesis may require inflammatory mediators from recruited leukocytes. We conclude that TNF-alpha is a strong candidate for a mediator that drives blood vessel remodeling and lymphangiogenesis in inflammation.


Asunto(s)
Vasos Sanguíneos , Inflamación/inmunología , Linfangiogénesis/inmunología , Vasos Linfáticos , Sistema Respiratorio , Factor de Necrosis Tumoral alfa/inmunología , Animales , Vasos Sanguíneos/anatomía & histología , Vasos Sanguíneos/fisiología , Perfilación de la Expresión Génica , Glicoproteínas/metabolismo , Vasos Linfáticos/anatomía & histología , Vasos Linfáticos/fisiología , Proteínas de Transporte de Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Mycoplasma/inmunología , Infecciones por Mycoplasma/patología , Mycoplasma pulmonis , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Sistema Respiratorio/anatomía & histología , Sistema Respiratorio/inmunología , Transducción de Señal/fisiología
17.
Am J Pathol ; 174(6): 2378-87, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19443705

RESUMEN

The integrin alpha5beta1 has been previously implicated in tumor angiogenesis, but its role in the remodeling of both blood vessels and lymphatics during inflammation is at an early stage of understanding. We examined this issue using a selective, small-molecule inhibitor of alpha5beta1 integrin, 2-aroylamino-3-{4-[(pyridin-2-ylaminomethyl)heterocyclyl]phenyl}propionic acid (JSM8757), in a model of sustained airway inflammation in mice with Mycoplasma pulmonis infection, which is known to be accompanied by robust blood vessel remodeling and lymphangiogenesis. The inhibitor significantly decreased the proliferation of lymphatic endothelial cells in culture and the number of lymphatic sprouts and new lymphatics in airways of mice infected for 2 weeks but did not reduce remodeling of blood vessels in the same airways. In inflamed airways, alpha5 integrin immunoreactivity was present on lymphatic sprouts, but not on collecting lymphatics or blood vessels, and was not found on any lymphatics of normal airways. Macrophages, potential targets of the inhibitor, did not have alpha5 integrin immunoreactivity in inflamed airways. In addition, macrophage recruitment, assessed in infected airways by quantitative reverse transcription-polymerase chain reaction measurements of expression of the marker protein ionized calcium-binding adapter molecule 1 (Iba1), was not reduced by JSM8757. We conclude that inhibition of the alpha5beta1 integrin reduces lymphangiogenesis in inflamed airways after M. pulmonis infection because expression of the integrin is selectively increased on lymphatic sprouts and plays an essential role in lymphatic growth.


Asunto(s)
Linfangiogénesis/fisiología , Neumonía/metabolismo , Animales , Femenino , Inmunohistoquímica , Inflamación/metabolismo , Inflamación/microbiología , Linfangiogénesis/efectos de los fármacos , Vasos Linfáticos/efectos de los fármacos , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Infecciones por Mycoplasma/metabolismo , Infecciones por Mycoplasma/fisiopatología , Mycoplasma pulmonis , Neumonía/microbiología , Propionatos/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Development ; 135(12): 2183-92, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18506030

RESUMEN

Morphogen gradients play fundamental roles in patterning and cell specification during development by eliciting differential transcriptional responses in target cells. In Drosophila, Decapentaplegic (Dpp), the BMP2/4 homolog, downregulates transcription of the nuclear repressor brinker (brk) in a concentration-dependent manner to generate an inverse graded distribution. Both Dpp and Brk are crucial for directing Dpp target gene expression in defined domains and the consequent execution of distinct developmental programs. Thus, determining the mechanism by which the brk promoter interprets the Dpp activity gradient is essential for understanding both Dpp-dependent patterning and how graded signaling activity can generate different responses through transcriptional repression. We have uncovered key features of the brk promoter that suggest it uses a complex enhancer logic not represented in current models. First, we find that the regulatory region contains multiple compact modules that can independently drive brk-like expression patterns. Second, each module contains binding sites for the Schnurri/Mad/Medea (SMM) complex, which mediates Dpp-dependent repression, linked to regions that direct activation. Third, the SMM repression complex acts through a distance-dependent mechanism that probably uses the canonical co-repressor C-terminal Binding Protein (CtBP). Finally, our data suggest that inputs from multiple regulatory modules are integrated to generate the final pattern. This unusual promoter organization may be necessary for brk to respond to the Dpp gradient in a precise and robust fashion.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas de Insectos/metabolismo , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Drosophila/embriología , Drosophila/genética , Proteínas de Drosophila/genética , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Proteínas de Insectos/genética , Proteínas Represoras/genética , Factores de Transcripción/genética
19.
Development ; 133(20): 4025-34, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17008448

RESUMEN

Bone Morphogenetic Proteins (Bmps) are secreted growth factors that play crucial roles in animal development across the phylogenetic spectrum. Bmp signaling results in the phosphorylation and nuclear translocation of Smads, downstream signal transducers that bind DNA. In Drosophila, the zinc finger protein Schnurri (Shn) plays a key role in signaling by the Bmp2/Bmp4 homolog Decapentaplegic (Dpp), by forming a Shn/Smad repression complex on defined promoter elements in the brinker (brk) gene. Brk is a transcriptional repressor that downregulates Dpp target genes. Thus, brk inhibition by Shn results in the upregulation of Dpp-responsive genes. We present evidence that vertebrate Shn homologs can also mediate Bmp responsiveness through a mechanism similar to Drosophila Shn. We find that a Bmp response element (BRE) from the Xenopus Vent2 promoter drives Dpp-dependent expression in Drosophila. However, in sharp contrast to its activating role in vertebrates, the frog BRE mediates repression in Drosophila. Remarkably, despite these opposite transcriptional polarities, sequence changes that abolish cis-element activity in Drosophila also affect BRE function in Xenopus. These similar cis requirements reflect conservation of trans-acting factors, as human Shn1 (hShn1; HIVEP1) can interact with Smad1/Smad4 and assemble an hShn1/Smad complex on the BRE. Furthermore, both Shn and hShn1 activate the BRE in Xenopus embryos, and both repress brk and rescue embryonic patterning defects in shn mutants. Our results suggest that vertebrate Shn proteins function in Bmp signal transduction, and that Shn proteins recruit coactivators and co-repressors in a context-dependent manner, rather than acting as dedicated activators or repressors.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Proteínas de Unión al ADN/clasificación , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/clasificación , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Elementos de Respuesta/genética , Factores de Transcripción/clasificación , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Secuencia Conservada/genética , Proteínas de Unión al ADN/genética , Drosophila/embriología , Drosophila/genética , Embrión no Mamífero/metabolismo , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Filogenia , Transducción de Señal , Proteínas Smad/metabolismo , Factores de Transcripción/genética , Vertebrados/genética , Vertebrados/metabolismo , Xenopus/genética , Xenopus/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
20.
Development ; 130(13): 2939-51, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12756177

RESUMEN

We report the identification of a Drosophila Pax gene, eye gone (eyg), which is required for eye development. Loss-of-function eyg mutations cause reduction or absence of the eye. Similar to the Pax6 eyeless (ey) gene, ectopic expression of eyg induces extra eye formation, but at sites different from those induced by ey. Several lines of evidence suggest that eyg and ey act cooperatively: (1) eyg expression is not regulated by ey, nor does it regulate ey expression, (2) eyg-induced ectopic morphogenetic furrow formation does not require ey, nor does ey-induced ectopic eye production require eyg, (3) eyg and ey can partially substitute for the function of the other, and (4) coexpression of eyg and ey has a synergistic enhancement of ectopic eye formation. Our results also show that eyg has two major functions: to promote cell proliferation in the eye disc and to promote eye development through suppression of wg transcription.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Estructuras Embrionarias/crecimiento & desarrollo , Animales , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Estructuras Embrionarias/anatomía & histología , Regulación del Desarrollo de la Expresión Génica , Genes de Insecto , Hibridación in Situ , Fenotipo , Células Fotorreceptoras de Invertebrados/anatomía & histología , Células Fotorreceptoras de Invertebrados/embriología , Células Fotorreceptoras de Invertebrados/crecimiento & desarrollo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Transcripción Genética , Transgenes , Proteína Wnt1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA