Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nano Lett ; 23(15): 7092-7099, 2023 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-37498114

RESUMO

Venous malformations (VMs) consist of hugely enlarged and dysmorphic veins. These lesions cause significant disfigurement, pain, and complications such as bleeding and coagulopathy. Pharmacotherapy for the treatment of VMs has limited efficacy and potentially limiting toxicity. Current treatment for patients with VMs entails life-long pharmacotherapy or surgical procedures. Here we explored whether intravenously administered agents can be used to destroy VMs by photothermal therapy (PTT), using gold nanoshells (AuNSs) that generated heat following irradiation with near-infrared (NIR) light. In a murine model of VMs, intravenous AuNSs accumulated within the VMs. Irradiation of the VMs induced marked regression and even elimination. Nanoparticle-based photothermal therapy can provide effective therapy for VMs, which are otherwise relatively refractory to treatment.


Assuntos
Hipertermia Induzida , Nanoconchas , Humanos , Camundongos , Animais , Terapia Fototérmica , Ouro/uso terapêutico , Nanoconchas/uso terapêutico , Hipertermia Induzida/métodos , Fototerapia
2.
Angiogenesis ; 25(3): 331-342, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35391614

RESUMO

Somatic mutations in NRAS drive the pathogenesis of melanoma and other cancers but their role in vascular anomalies and specifically human endothelial cells is unclear. The goals of this study were to determine whether the somatic-activating NRASQ61R mutation in human endothelial cells induces abnormal angiogenesis and to develop in vitro and in vivo models to identify disease-causing pathways and test inhibitors. Here, we used mutant NRASQ61R and wild-type NRAS (NRASWT) expressing human endothelial cells in in vitro and in vivo angiogenesis models. These studies demonstrated that expression of NRASQ61R in human endothelial cells caused a shift to an abnormal spindle-shaped morphology, increased proliferation, and migration. NRASQ61R endothelial cells had increased phosphorylation of ERK compared to NRASWT cells indicating hyperactivation of MAPK/ERK pathways. NRASQ61R mutant endothelial cells generated abnormal enlarged vascular channels in a 3D fibrin gel model and in vivo, in xenografts in nude mice. These studies demonstrate that NRASQ61R can drive abnormal angiogenesis in human endothelial cells. Treatment with MAP kinase inhibitor U0126 prevented the change to a spindle-shaped morphology in NRASQ61R endothelial cells, whereas mTOR inhibitor rapamycin did not.


Assuntos
GTP Fosfo-Hidrolases , Proteínas de Membrana , Malformações Vasculares , Animais , Células Endoteliais/metabolismo , GTP Fosfo-Hidrolases/genética , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Nus , Mutação , Malformações Vasculares/genética
3.
Angiogenesis ; 23(3): 425-442, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32350708

RESUMO

Capillary lymphatic venous malformations (CLVM) are complex vascular anomalies characterized by aberrant and enlarged lymphatic and blood vessels. CLVM appear during fetal development and enlarge after birth, causing life-long complications such as coagulopathy, pulmonary embolism, chronic pain, and disfigurement. Treatment includes surgical debulking, amputation, and recurrent sclerotherapy. Somatic, mosaic mutations in the 110-kD catalytic α-subunit of phosphoinositide-3-kinase (PIK3CA) gene have been previously identified in affected tissues from CLVM patients; however, the cell population harboring the mutation is still unknown. In this study, we hypothesized that endothelial cells (EC) carry the PIK3CA mutations and play a major role in the cellular origin of CLVM. We isolated EC from the lesions of seven patients with CLVM and identified PIK3CA hotspot mutations. The CLVM EC exhibited constitutive phosphorylation of the PI3K effector AKT as well as hyperproliferation and increased resistance to cell death compared to normal EC. Inhibitors of PIK3CA (BYL719) and AKT (ARQ092) attenuated the proliferation of CLVM EC in a dose-dependent manner. A xenograft model of CLVM was developed by injecting patient-derived EC into the flanks of immunocompromised mice. CLVM EC formed lesions with enlarged lymphatic and vascular channels, recapitulating the patient histology. EC subpopulations were further obtained by both immunomagnetic separation into lymphatic EC (LEC) and vascular EC (VEC) and generation of clonal populations. By sequencing these subpopulations, we determined that both LEC and VEC from the same patient express the PIK3CA mutation, exhibit increased AKT activation and can form lymphatic or vascular lesions in mouse.


Assuntos
Capilares/anormalidades , Classe I de Fosfatidilinositol 3-Quinases , Células Endoteliais da Veia Umbilical Humana , Vasos Linfáticos , Mutação , Malformações Vasculares , Adulto , Animais , Capilares/enzimologia , Capilares/patologia , Pré-Escolar , Classe I de Fosfatidilinositol 3-Quinases/genética , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Feminino , Células Endoteliais da Veia Umbilical Humana/enzimologia , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Lactente , Vasos Linfáticos/anormalidades , Vasos Linfáticos/enzimologia , Vasos Linfáticos/patologia , Masculino , Camundongos , Camundongos Nus , Malformações Vasculares/enzimologia , Malformações Vasculares/genética , Malformações Vasculares/patologia
4.
Arterioscler Thromb Vasc Biol ; 39(3): 496-512, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30626204

RESUMO

Objective- Venous malformations (VMs) arise from developmental defects of the vasculature and are characterized by massively enlarged and tortuous venous channels. VMs grow commensurately leading to deformity, obstruction of vital structures, bleeding, and pain. Most VMs are associated with the activating mutation L914F in the endothelial cell (EC) tyrosine kinase receptor TIE2. Therapeutic options for VM are limited and ineffective while therapy with the mammalian target of rapamycin inhibitor rapamycin shows moderate efficacy. Here, we investigated novel therapeutic targets promoting VM regression. Approach and Results- We performed an unbiased screen of Food and Drug Administration-approved drugs in human umbilical vein ECs expressing the TIE2-L914F mutation (HUVEC-TIE2-L914F). Three ABL (Abelson) kinase inhibitors prevented cell proliferation of HUVEC-TIE2-L914F. Moreover, c-ABL, common target of these inhibitors, was highly phosphorylated in HUVEC-TIE2-L914F and VM patient-derived ECs with activating TIE2 mutations. Knockdown of c-ABL/ARG in HUVEC-TIE2-L914F reduced cell proliferation and vascularity of murine VM. Combination treatment with the ABL kinase inhibitor ponatinib and rapamycin caused VM regression in a xenograft model based on injection of HUVEC-TIE2-L914F. A reduced dose of this drug combination was effective in this VM murine model with minimal side effects. The drug combination was antiproliferative, enhanced cell apoptosis and vascular channel regression both in vivo and in a 3-dimensional fibrin gel assay. Conclusions- This is the first report of a combination therapy with ponatinib and rapamycin promoting regression of VM. Mechanistically, the drug combination enhanced AKT inhibition compared with single drug treatment and reduced PLCγ (phospholipase C) and ERK (extracellular signal-regulated kinase) activity.


Assuntos
Imidazóis/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Piridazinas/uso terapêutico , Sirolimo/uso terapêutico , Malformações Vasculares/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Quimiotaxia , Avaliação Pré-Clínica de Medicamentos , Quimioterapia Combinada , Xenoenxertos , Células Endoteliais da Veia Umbilical Humana/transplante , Humanos , Imidazóis/administração & dosagem , Imidazóis/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Nus , Mutação de Sentido Incorreto , Fosfolipase C gama/antagonistas & inibidores , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Piridazinas/administração & dosagem , Piridazinas/farmacologia , Receptor TIE-2/genética , Transdução de Sinais/efeitos dos fármacos , Sirolimo/administração & dosagem , Sirolimo/farmacologia , Malformações Vasculares/patologia
5.
Pediatr Blood Cancer ; 67(9): e28529, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32634277

RESUMO

Kaposiform lymphangiomatosis (KLA) is a rare, life-threatening congenital lymphatic malformation. Diagnosis is often delayed due to complex indistinct symptoms. Blood angiopoietin-2 (ANG2) levels are elevated in KLA and may be useful as a biomarker to monitor disease status. We report a 7-year-old male child with easy bruising, inguinal swelling, and consumptive coagulopathy, diagnosed with KLA. A multimodal treatment regimen of prednisone, sirolimus, vincristine, and adjunctive zoledronate was used. Plasma ANG2 levels were highly elevated at diagnosis but decreased during treatment. The patient showed significant clinical improvement over a 38-month period and normalization of ANG2 levels correlated with resolution of the coagulopathy.


Assuntos
Angiopoietina-2/sangue , Hemangioendotelioma/terapia , Síndrome de Kasabach-Merritt/terapia , Sarcoma de Kaposi/terapia , Trombose/prevenção & controle , Criança , Terapia Combinada , Hemangioendotelioma/sangue , Hemangioendotelioma/patologia , Humanos , Síndrome de Kasabach-Merritt/sangue , Síndrome de Kasabach-Merritt/patologia , Masculino , Prognóstico , Sarcoma de Kaposi/sangue , Sarcoma de Kaposi/patologia , Trombose/sangue , Trombose/patologia
6.
Pediatr Blood Cancer ; 66(8): e27790, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31045327

RESUMO

BACKGROUND: Kaposiform lymphangiomatosis (KLA) is a rare lymphatic anomaly with significant morbidity and mortality. KLA is characterized by diffuse multifocal lesions comprised of focal areas of "kaposiform" spindled cells accompanying malformed lymphatic channels. The goal of this study was to identify activated signaling pathways in cells isolated from three KLA patients for the purpose of testing new therapies. PROCEDURE: Cells were obtained from the lungs of one patient isolated at autopsy and the spleen of two patients removed in surgery due to disease complications. A protein kinase array was performed on the KLA cell lysates and normal lymphatic endothelial cells. RESULTS: Higher activation of key signaling pathways in the KLA cells, including PRAS40, AKT1/2/3, and ERK-1/2, was identified by protein kinase array and confirmed by Western blot analysis. This indicated a role for highly activated PI3K-AKT and MAPK-ERK-1/2 signaling pathways in KLA cells. Cell proliferation studies assessed PI3K inhibitors (LY294002; BYL719), AKT inhibitor ARQ092, mTOR inhibitor rapamycin, and MAPK inhibitor U0126. These studies demonstrated that PI3K-AKT-mTOR and MAPK signaling are important mediators of KLA cell proliferation. BYL719 and rapamycin were more effective at inhibiting KLA cell proliferation than U0126. CONCLUSIONS: Our studies using cells from KLA patient lesions demonstrate that these cells are highly proliferative and the PI3K-AKT-mTOR and MAPK pathways are promising therapeutic targets. Development and clinical trials of PI3K, AKT, and MAPK inhibitors for cancer treatment and the data in this study lend support for early clinical trials assessing the efficacy of these inhibitors in KLA patients.


Assuntos
Antineoplásicos/farmacologia , Hemangioendotelioma/patologia , Síndrome de Kasabach-Merritt/patologia , Linfangioma/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/química , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Sarcoma de Kaposi/patologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Adolescente , Proliferação de Células/efeitos dos fármacos , Pré-Escolar , Feminino , Seguimentos , Hemangioendotelioma/tratamento farmacológico , Hemangioendotelioma/metabolismo , Humanos , Lactente , Síndrome de Kasabach-Merritt/tratamento farmacológico , Síndrome de Kasabach-Merritt/metabolismo , Linfangioma/tratamento farmacológico , Linfangioma/metabolismo , Masculino , Inibidores de Proteínas Quinases/farmacologia , Estudos Retrospectivos , Sarcoma de Kaposi/tratamento farmacológico , Sarcoma de Kaposi/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
7.
Angiogenesis ; 21(4): 725-735, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29786783

RESUMO

Vascular malformations are defects caused by the abnormal growth of the vasculature. Among them, venous malformation (VM) is an anomaly characterized by slow-flow vascular lesions with abnormally shaped veins, typically in sponge-like configuration. VMs can expand over years causing disfigurement, obstruction of vital structures, thrombosis, bleeding, and pain. Treatments have been very limited and primarily based on supportive care, compression garments, sclerotherapy, and/or surgical resection. Sirolimus treatment has recently shown efficacy in some patients with complicated vascular anomalies, including VMs. Activating somatic TIE2 gene mutations have been identified in up to 60% of VMs and PIK3CA mutations have been found in another 25%. Here, we report a xenograft model of VM that reflects the patients' mutation heterogeneity. First, we established a protocol to isolate and expand in culture endothelial cells (VM-EC) from VM tissue or VM blood of nine patients. In these cells, we identified somatic mutations of TIE2, PIK3CA, or a combination of both. Both TIE2 and PIK3CA mutations induced constitutive AKT activation, while TIE2 mutations also showed high MAPK-ERK signaling. Finally, VM-EC implanted into immune-deficient mice generated lesions with ectatic blood-filled channels with scarce smooth muscle cell coverage, similar to patients' VM. This VM xenograft model could be instrumental to test the therapeutic efficacy of Sirolimus in the presence of the different TIE2 or PIK3CA mutations or to test for efficacy of additional compounds in targeting the specific mutated protein(s), thus enabling development of personalized treatment options for VM patients.


Assuntos
Modelos Animais de Doenças , Células Endoteliais da Veia Umbilical Humana , Malformações Vasculares , Animais , Classe I de Fosfatidilinositol 3-Quinases/genética , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Xenoenxertos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Células Endoteliais da Veia Umbilical Humana/transplante , Humanos , Masculino , Camundongos , Camundongos Nus , Mutação , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Malformações Vasculares/genética , Malformações Vasculares/metabolismo , Malformações Vasculares/patologia
8.
Cell Mol Life Sci ; 73(8): 1715-39, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26646071

RESUMO

The circulatory system is walled off by different cell types, including vascular mural cells and podocytes. The interaction and interplay between endothelial cells (ECs) and mural cells, such as vascular smooth muscle cells or pericytes, play a pivotal role in vascular biology. Endoglin is an RGD-containing counter-receptor for ß1 integrins and is highly expressed by ECs during angiogenesis. We find that the adhesion between vascular ECs and mural cells is enhanced by integrin activators and inhibited upon suppression of membrane endoglin or ß1-integrin, as well as by addition of soluble endoglin (SolEng), anti-integrin α5ß1 antibody or an RGD peptide. Analysis of different endoglin mutants, allowed the mapping of the endoglin RGD motif as involved in the adhesion process. In Eng (+/-) mice, a model for hereditary hemorrhagic telangectasia type 1, endoglin haploinsufficiency induces a pericyte-dependent increase in vascular permeability. Also, transgenic mice overexpressing SolEng, an animal model for preeclampsia, show podocyturia, suggesting that SolEng is responsible for podocytes detachment from glomerular capillaries. These results suggest a critical role for endoglin in integrin-mediated adhesion of mural cells and provide a better understanding on the mechanisms of vessel maturation in normal physiology as well as in pathologies such as preeclampsia or hereditary hemorrhagic telangiectasia.


Assuntos
Antígenos CD/metabolismo , Adesão Celular/fisiologia , Endotélio Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Podócitos/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Antígenos CD/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Endoglina , Feminino , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Integrina beta1/genética , Células Jurkat , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Neovascularização Patológica/metabolismo , Pericitos/metabolismo , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/patologia , Gravidez , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno , Receptores de Superfície Celular/genética , Retina/metabolismo , Telangiectasia Hemorrágica Hereditária/genética , Telangiectasia Hemorrágica Hereditária/patologia
9.
Angiogenesis ; 18(2): 151-62, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25424831

RESUMO

Lymphatic malformations (LM) are characterized by abnormal formation of lymphatic vessels and tissue overgrowth. The lymphatic vessels present in LM lesions may become blocked and enlarged as lymphatic fluid collects, forming a mass or cyst. Lesions are typically diagnosed during childhood and are often disfiguring and life threatening. Available treatments consist of sclerotherapy, surgical removal and therapies to diminish complications. We isolated lymphatic endothelial cells (LM-LEC) from a surgically removed microcystic LM lesion. LM-LEC and normal human dermal-LEC (HD-LEC) expressed endothelial (CD31, VE-Cadherin) as well as lymphatic endothelial (Podoplanin, PROX1, LYVE1)-specific markers. Targeted gene sequencing analysis in patient-derived LM-LEC revealed the presence of two mutations in class I phosphoinositide 3-kinases (PI3K) genes. One is an inherited, premature stop codon in the PI3K regulatory subunit PIK3R3. The second is a somatic missense mutation in the PI3K catalytic subunit PIK3CA; this mutation has been found in association with overgrowth syndromes and cancer growth. LM-LEC exhibited angiogenic properties: both cellular proliferation and sprouting in collagen were significantly increased compared with HD-LEC. AKT-Thr308 was constitutively hyper-phosphorylated in LM-LEC. Treatment of LM-LEC with PI3-Kinase inhibitors Wortmannin and LY294 decreased cellular proliferation and prevented the phosphorylation of AKT-Thr308 in both HD-LEC and LM-LEC. Treatment with the mTOR inhibitor rapamycin also diminished cellular proliferation, sprouting and AKT phosphorylation, but only in LM-LEC. Our results implicate disrupted PI3K-AKT signaling in LEC isolated from a human lymphatic malformation lesion.


Assuntos
Endotélio/enzimologia , Vasos Linfáticos/anormalidades , Mutação , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Endotélio/patologia , Feminino , Humanos , Masculino , Fosforilação , Sirolimo/farmacologia
10.
Stem Cells ; 32(3): 684-93, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24022922

RESUMO

Infantile hemangioma (IH) is the most common tumor of infancy. Hemangioma stem cells (HemSC) are a mesenchymal subpopulation isolated from IH CD133+ cells. HemSC can differentiate into endothelial and pericyte/smooth muscle cells and form vascular networks when injected in immune-deficient mice. α6-Integrin subunit has been implicated in the tumorgenicity of glioblastoma stem cells and the homing properties of hematopoietic, endothelial, and mesenchymal progenitor cells. Therefore, we investigated the possible function(s) of α6-integrin in HemSC. We documented α6-integrin expression in IH tumor specimens and HemSC by RT-qPCR and flow cytometry. We examined the effect of blocking or silencing α6-integrin on the adhesive and proliferative properties of HemSC in vitro and the vasculogenic and homing properties of HemSC in vivo. Targeting α6-integrin in cultured HemSC inhibited adhesion to laminin but had no effect on proliferation. Vessel-forming ability in Matrigel implants and hepatic homing after i.v. delivery were significantly decreased in α6-integrin siRNA-transfected HemSC. In conclusion, α6-integrin is required for HemSC adherence to laminin, vessel formation in vivo, and for homing to the liver. Thus, we uncovered an important role for α6 integrin in the vasculogenic properties of HemSC. Our results suggest that α6-integrin expression on HemSC could be a new target for antihemangioma therapy.


Assuntos
Hemangioma/metabolismo , Hemangioma/patologia , Integrina alfa6/metabolismo , Neovascularização Fisiológica , Células-Tronco/metabolismo , Células-Tronco/patologia , Animais , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células , Pré-Escolar , Feminino , Técnicas de Silenciamento de Genes , Humanos , Lactente , Laminina/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Neovascularização Fisiológica/efeitos dos fármacos
11.
Blood ; 120(14): 2925-9, 2012 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-22791286

RESUMO

Inflammatory cytokines and growth factors drive angiogenesis independently; however, their integrated role in pathologic and physiologic angiogenesis is not fully understood. Suppressor of cytokine signaling-3 (SOCS3) is an inducible negative feedback regulator of inflammation and growth factor signaling. In the present study, we show that SOCS3 curbs pathologic angiogenesis. Using a Cre/Lox system, we deleted SOCS3 in vessels and studied developmental and pathologic angiogenesis in murine models of oxygen-induced retinopathy and cancer. Conditional loss of SOCS3 leads to increased pathologic neovascularization, resulting in pronounced retinopathy and increased tumor size. In contrast, physiologic vascularization is not regulated by SOCS3. In vitro, SOCS3 knockdown increases proliferation and sprouting of endothelial cells costimulated with IGF-1 and TNFα via reduced feedback inhibition of the STAT3 and mTOR pathways. These results identify SOCS3 as a pivotal endogenous feedback inhibitor of pathologic angiogenesis and a potential therapeutic target acting at the converging crossroads of growth factor- and cytokine-induced vessel growth.


Assuntos
Carcinoma Pulmonar de Lewis/prevenção & controle , Hipóxia/patologia , Melanoma Experimental/prevenção & controle , Neovascularização Patológica/prevenção & controle , Síndromes Paraneoplásicas Oculares/prevenção & controle , Proteínas Supressoras da Sinalização de Citocina/fisiologia , Animais , Western Blotting , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/patologia , Proliferação de Células , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Integrases/metabolismo , Masculino , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/etiologia , Síndromes Paraneoplásicas Oculares/patologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Proteína 3 Supressora da Sinalização de Citocinas , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 33(3): 501-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23288163

RESUMO

OBJECTIVE: Infantile hemangioma (IH) is a rapidly growing vascular tumor affecting newborns. It is composed of immature endothelial cells and pericytes that proliferate into a disorganized mass of blood vessels. We isolated pericytes from IH (Hem-pericytes) to test our hypothesis that Hem-pericytes are unable to stabilize blood vessels. METHODS AND RESULTS: We injected pericytes in vivo, in combination with endothelial cells, and found that Hem-pericytes formed more microvessels compared with control retinal pericytes. We, thereby, analyzed proangiogenic properties of the Hem-pericytes. They grew fast in vitro, and were unable to stabilize endothelial cell growth and migration, and expressed high levels of vascular endothelial growth factor-A compared with retinal pericytes. Hem-pericytes from proliferating phase IH showed lower contractility in vitro, compared with Hem-pericytes from the involuting phase and retinal pericytes. Consistent with a diminished ability to stabilize endothelium, angiopoietin 1 was reduced in Hem-pericytes compared with retinal pericytes. Normal retinal pericytes in which angiopoietin 1 was silenced produced conditioned medium that stimulated endothelial cell proliferation and migration. CONCLUSIONS: We report the first successful isolation of patient-derived pericytes from IH tissue. Hem-pericytes exhibited proangiogenic properties and low levels of angiopoietin 1, consistent with a diminished ability to stabilize blood vessels in IH.


Assuntos
Angiopoietina-1/metabolismo , Hemangioma/metabolismo , Neovascularização Patológica , Pericitos/metabolismo , Angiopoietina-1/genética , Animais , Biomarcadores/metabolismo , Comunicação Celular , Movimento Celular , Proliferação de Células , Separação Celular/métodos , Células Cultivadas , Meios de Cultivo Condicionados/metabolismo , Regulação para Baixo , Hemangioma/patologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/transplante , Humanos , Lactente , Recém-Nascido , Masculino , Camundongos , Camundongos Nus , Pericitos/patologia , Pericitos/transplante , Interferência de RNA , RNA Mensageiro/metabolismo , Vasos Retinianos/metabolismo , Fatores de Tempo , Transfecção , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
J Clin Invest ; 134(8)2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38618955

RESUMO

Capillary malformation (CM), or port wine birthmark, is a cutaneous congenital vascular anomaly that occurs in 0.1%-2% of newborns. Patients with a CM localized on the forehead have an increased risk of developing a neurocutaneous disorder called encephalotrigeminal angiomatosis or Sturge-Weber syndrome (SWS), with complications including seizure, developmental delay, glaucoma, and vision loss. In 2013, a groundbreaking study revealed causative activating somatic mutations in the gene (GNAQ) encoding guanine nucleotide-binding protein Q subunit α (Gαq) in CM and SWS patient tissues. In this Review, we discuss the disease phenotype, the causative GNAQ mutations, and their cellular origin. We also present the endothelial Gαq-related signaling pathways, the current animal models to study CM and its complications, and future options for therapeutic treatment. Further work remains to fully elucidate the cellular and molecular mechanisms underlying the formation and maintenance of the abnormal vessels.


Assuntos
Capilares/anormalidades , Glaucoma , Malformações Vasculares , Recém-Nascido , Animais , Humanos , Modelos Animais , Mutação
14.
N Engl J Med ; 362(11): 1005-13, 2010 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-20237346

RESUMO

BACKGROUND: Corticosteroids are commonly used to treat infantile hemangioma, but the mechanism of action of this therapy is unknown. We investigated the effect of corticosteroids in a previously described in vivo model of infantile hemangioma and in cultured hemangioma-derived cells. METHODS: We tested hemangioma-derived stem cells for vasculogenic activity in vivo after implantation into immune-deficient (nude) mice. We studied dexamethasone treatment of both the cells before implantation and the mice after implantation. We also tested hemangioma-derived stem cells for expression of vascular endothelial growth factor A (VEGF-A) in vitro and studied the inhibition of VEGF-A expression, using short hairpin RNA (shRNA) in vivo and in vitro. RESULTS: Systemic treatment with dexamethasone led to dose-dependent inhibition of tumor vasculogenesis in the murine model. Pretreatment of hemangioma-derived stem cells in vitro before implantation also inhibited vasculogenesis. Dexamethasone suppressed VEGF-A production by hemangioma-derived stem cells in vitro but not by hemangioma-derived endothelial cells or human umbilical-vein endothelial cells. Silencing VEGF-A in hemangioma-derived stem cells reduced vasculogenesis in vivo. VEGF-A was detected in hemangioma specimens in the proliferating phase but not in the involuting phase and was shown by immunostaining to reside outside of vessels. Corticosteroid treatment suppressed other proangiogenic factors in hemangioma-derived stem cells, including urokinase plasminogen activator receptor, interleukin-6, monocyte chemoattractant protein 1, and matrix metalloproteinase 1. CONCLUSIONS: In a murine model, dexamethasone inhibited the vasculogenic potential of stem cells derived from human infantile hemangioma. The corticosteroid also inhibited the expression of VEGF-A by hemangioma-derived stem cells, and silencing of VEGF-A expression in these cells inhibited vasculogenesis in vivo.


Assuntos
Inibidores da Angiogênese/farmacologia , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Hemangioma/metabolismo , Células-Tronco Multipotentes/efeitos dos fármacos , Neovascularização Patológica/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular/metabolismo , Inibidores da Angiogênese/uso terapêutico , Animais , Dexametasona/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação para Baixo , Glucocorticoides/uso terapêutico , Hemangioma/tratamento farmacológico , Hemangioma/patologia , Humanos , Lactente , Camundongos , Camundongos Nus , Células-Tronco Multipotentes/metabolismo , Neovascularização Patológica/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/genética
15.
Nat Commun ; 14(1): 1929, 2023 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-37024491

RESUMO

Activating non-inherited mutations in the guanine nucleotide-binding protein G(q) subunit alpha (GNAQ) gene family have been identified in childhood vascular tumors. Patients experience extensive disfigurement, chronic pain and severe complications including a potentially lethal coagulopathy termed Kasabach-Merritt phenomenon. Animal models for this class of vascular tumors do not exist. This has severely hindered the discovery of the molecular consequences of GNAQ mutations in the vasculature and, in turn, the preclinical development of effective targeted therapies. Here we report a mouse model expressing hyperactive mutant GNAQ in endothelial cells. Mutant mice develop vascular and coagulopathy phenotypes similar to those seen in patients. Mechanistically, by transcriptomic analysis we demonstrate increased mitogen activated protein kinase signaling in the mutant endothelial cells. Targeting of this pathway with Trametinib suppresses the tumor growth by reducing vascular cell proliferation and permeability. Trametinib also prevents the development of coagulopathy and improves mouse survival.


Assuntos
Melanoma , Neoplasias Uveais , Neoplasias Vasculares , Animais , Camundongos , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Células Endoteliais/metabolismo , Apoptose , Melanoma/genética , Neoplasias Uveais/genética , Mutação , Modelos Animais de Doenças , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Linhagem Celular Tumoral
16.
Am J Pathol ; 179(5): 2266-77, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21945324

RESUMO

Vascular endothelial growth factor receptor-1 (VEGFR-1) is a member of the VEGFR family, and binds to VEGF-A, VEGF-B, and placental growth factor. VEGFR-1 contributes to tumor growth and metastasis, but its role in the pathological formation of blood vessels is still poorly understood. Herein, we used infantile hemangioma (IH), the most common tumor of infancy, as a means to study VEGFR-1 activation in pathological vasculogenesis. IH arises from stem cells (HemSCs) that can form the three most prominent cell types in the tumor: endothelial cells, pericytes, and adipocytes. HemSCs can recapitulate the IH life cycle when injected in immuncompromised mice, and are targeted by corticosteroids, the traditional treatment for IH. We report here that VEGF-A or VEGF-B induces VEGFR-1-mediated ERK1/2 phosphorylation in HemSCs and promotes differentiation of HemSCs to endothelial cells. Studies of VEGFR-2 phosphorylation status and down-regulation of neuropilin-1 in the HemSCs demonstrate that VEGFR-2 and NRP1 are not needed for VEGF-A- or VEGF-B-induced ERK1/2 activation. U0216-mediated blockade of ERK1/2 phosphorylation or shRNA-mediated suppression of VEGFR-1 prevents HemSC-to-EC differentiation. Furthermore, the down-regulation of VEGFR-1 in the HemSCs results in decreased formation of blood vessels in vivo and reduced ERK1/2 activation. Thus, our study reveals a critical role for VEGFR-1 in the HemSC-to-EC differentiation that underpins pathological vasculogenesis in IH.


Assuntos
Células Endoteliais/citologia , Hemangioma/irrigação sanguínea , Neovascularização Patológica/fisiopatologia , Neoplasias Cutâneas/irrigação sanguínea , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Animais , Proliferação de Células , Transformação Celular Neoplásica/patologia , Inibidores Enzimáticos/farmacologia , Hemangioma/patologia , Humanos , Lactente , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Camundongos Nus , Microvasos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Transplante de Neoplasias , Células-Tronco Neoplásicas/patologia , Neuropilina-1/metabolismo , Fosforilação , Neoplasias Cutâneas/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator B de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
17.
Arterioscler Thromb Vasc Biol ; 31(10): 2181-92, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21757656

RESUMO

OBJECTIVE: The aim of our study is to determine the cellular and molecular origin for the pericytes in infantile hemangioma (IH) and their functional role in the formation of pathological blood vessels. METHODS AND RESULTS: Here we show that IH-derived stem cells (HemSCs) form pericyte-like cells. With in vitro studies, we demonstrate that HemSC-to-pericyte differentiation depends on direct contact with endothelial cells. JAGGED1 expressed ectopically in fibroblasts was sufficient to induce HemSCs to acquire a pericyte-like phenotype, indicating a critical role for JAGGED1. In vivo, we blocked pericyte differentiation with recombinant JAGGED1, and we observed reduced formation of blood vessels, with an evident lack of pericytes. Silencing JAGGED1 in the endothelial cells reduced blood vessel formation and resulted in a paucity of pericytes. CONCLUSIONS: Our data show that endothelial JAGGED1 controls HemSC-to-pericyte differentiation in a murine model of IH and suggests that pericytes have a fundamental role in formation of blood vessels in IH.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Transdiferenciação Celular , Hemangioma/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Células-Tronco Neoplásicas/metabolismo , Neovascularização Patológica/metabolismo , Pericitos/metabolismo , Transdução de Sinais , Animais , Proteínas de Ligação ao Cálcio/genética , Comunicação Celular , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/metabolismo , Hemangioma/irrigação sanguínea , Hemangioma/genética , Hemangioma/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteína Jagged-1 , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Nus , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Miócitos de Músculo Liso/transplante , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/transplante , Neovascularização Patológica/genética , Pericitos/patologia , Pericitos/transplante , Interferência de RNA , Proteínas Recombinantes/metabolismo , Proteínas Serrate-Jagged , Fatores de Tempo , Transfecção
18.
J Clin Invest ; 132(3)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35104803

RESUMO

Propranolol is a nonselective ß-adrenergic receptor (AR) blocker that has been the first-line therapy for problematic infantile hemangioma (IH), the most frequent childhood vascular tumor. Although IHs are benign and eventually regress spontaneously, at least 15% of patients require treatment. Despite the extensive use of propranolol for IH treatment, its mode of action remains unclear. In this issue of the JCI, Seebauer et al. investigated the cellular and molecular consequences of propranolol treatment on IH vascular tumor formation in a murine model of IH. The efficacy of propranolol was independent of its ß-AR blocker activity and was attributable to the direct targeting of the transcription factor SOX18, which, in turn, reduced hemangioma blood vessel formation. We believe these results will guide clinical translation for the use of more efficient and safer therapies for IH and possibly for other vascular anomalies in which SOX18 plays a role.


Assuntos
Hemangioma , Malformações Vasculares , Antagonistas Adrenérgicos beta/administração & dosagem , Animais , Criança , Hemangioma/tratamento farmacológico , Hemangioma/genética , Hemangioma/patologia , Humanos , Camundongos , Propranolol/farmacologia , Fatores de Transcrição SOXF , Fatores de Transcrição
19.
J Clin Invest ; 118(7): 2592-9, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18535669

RESUMO

Infantile hemangioma is a benign endothelial tumor composed of disorganized blood vessels. It exhibits a unique life cycle of rapid postnatal growth followed by slow regression to a fibrofatty residuum. Here, we have reported the isolation of multipotential stem cells from hemangioma tissue that give rise to hemangioma-like lesions in immunodeficient mice. Cells were isolated based on expression of the stem cell marker CD133 and expanded from single cells as clonal populations. The CD133-selected cells generated human blood vessels 7 days after implantation in immunodeficient mice. Cell retrieval experiments showed the cells could again form vessels when transplanted into secondary recipients. The human vessels expressed GLUT-1 and merosin, immunodiagnostic markers for infantile hemangioma. Two months after implantation, the number of blood vessels diminished and human adipocytes became evident. Lentiviral expression of GFP was used to confirm that the hemangioma-derived cells formed the blood vessels and adipocytes in the immunodeficient mice. Thus, when transplanted into immunodeficient mice, hemangioma-derived cells recapitulated the unique evolution of infantile hemangioma--the formation of blood vessels followed by involution to fatty tissue. In summary, this study identifies a stem cell as the cellular origin of infantile hemangioma and describes for what we believe is the first time an animal model for this common tumor of infancy.


Assuntos
Modelos Animais de Doenças , Hemangioma/patologia , Células-Tronco Multipotentes/transplante , Células-Tronco Neoplásicas/transplante , Adipócitos/química , Adipócitos/patologia , Animais , Antígenos Nucleares/análise , Antígenos de Superfície/análise , Vasos Sanguíneos/química , Vasos Sanguíneos/patologia , Proteínas de Transporte , Diferenciação Celular/efeitos dos fármacos , Separação Celular , Transplante de Células/efeitos adversos , Transplante de Células/métodos , Células Endoteliais/química , Células Endoteliais/patologia , Transportador de Glucose Tipo 1/análise , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Hemangioma/etiologia , Hemangioma/metabolismo , Humanos , Lactente , Laminina/análise , Masculino , Camundongos , Camundongos Nus , Células-Tronco Multipotentes/química , Células-Tronco Multipotentes/patologia , Células-Tronco Neoplásicas/química , Células-Tronco Neoplásicas/patologia , Perilipina-1 , Fosfoproteínas/análise , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Receptores de Fatores de Crescimento do Endotélio Vascular/análise , Transfecção
20.
Methods Mol Biol ; 2206: 179-192, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32754818

RESUMO

Xenograft models allow for an in vivo approach to monitor cellular functions within the context of a host microenvironment. Here we describe a protocol to generate a xenograft model of venous malformation (VM) based on the use of human umbilical vein endothelial cells (HUVEC) expressing a constitutive active form of the endothelial tyrosine kinase receptor TEK (TIE2 p.L914F) or patient-derived EC containing TIE2 and/or PIK3CA gene mutations. Hyperactive somatic TIE2 and PIK3CA mutations are a common hallmark of VM in patient lesions. The EC are injected subcutaneously on the back of athymic nude mice to generate ectatic vascular channels and recapitulate histopathological features of VM patient tissue histology. Lesion plugs with TIE2/PIK3CA-mutant EC are visibly vascularized within 7-9 days of subcutaneous injection, making this a great tool to study venous malformation.


Assuntos
Xenoenxertos/patologia , Células Endoteliais da Veia Umbilical Humana/patologia , Malformações Vasculares/patologia , Veias/patologia , Animais , Células Cultivadas , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Modelos Animais de Doenças , Xenoenxertos/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Camundongos Nus , Receptor TIE-2/metabolismo , Malformações Vasculares/metabolismo , Veias/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA