Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
BMJ Case Rep ; 20182018 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-30065054

RESUMO

We describe a dramatic clinical presentation of extramedullary multiple myeloma (MM) in an elderly patient with known monoclonal gammopathy of unknown significance (MGUS). Gastrointestinal symptoms and a gastric mass on imaging studies suggested an advanced solid gastric malignancy. Pathological workup of gastric biopsies first suspected a lymphoma, a second opinion finally confirmed an extramedullary MM. Treatment with bortezomib, cyclophosphamide and dexamethasone induced rapid relief of symptoms and normalisation of renal function as well as serum MM markers. Our case highlights the diagnostic difficulties when MM presents with signs and symptoms of respective end-organ involvement rather than typical 'CRAB' criteria. It underlines the importance of actively considering MM in a patient with MGUS, regardless of the clinical presentation of a specific medical problem. Our report also impressively illustrates the rapid response of MM and its gastric extramedullary manifestation to guideline-adherent chemotherapy.


Assuntos
Mieloma Múltiplo/diagnóstico , Pênfigo/diagnóstico , Insuficiência Renal/diagnóstico , Estenose Espinal/diagnóstico , Neoplasias Gástricas/diagnóstico , Dor Abdominal , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica , Ascite/diagnóstico por imagem , Tosse , Feminino , Humanos , Mieloma Múltiplo/tratamento farmacológico , Derrame Pleural/diagnóstico por imagem , Diálise Renal , Insuficiência Renal/fisiopatologia , Insuficiência Renal/terapia , Estenose Espinal/fisiopatologia , Estenose Espinal/terapia , Neoplasias Gástricas/tratamento farmacológico , Tomografia Computadorizada por Raios X , Resultado do Tratamento
2.
Angiogenesis ; 19(3): 339-58, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27038485

RESUMO

UNLABELLED: Viral VEGF-E (ovVEGF-E), a homolog of VEGF-A, was discovered in the genome of Orf virus. Together with VEGF-A, B, C, D, placental growth factor (PlGF) and snake venom VEGF (svVEGF), ovVEGF-E is a member of the VEGF family of potent angiogenesis factors with a bioactivity similar to VEGF-A: it induces proliferation, migration and sprouting of cultured vascular endothelial cells and proliferative lesions in the skin of sheep, goat and man that are characterized by massive capillary proliferation and dilation. These biological functions are mediated exclusively via its interaction with VEGF receptor-2 (VEGFR-2). Here, we have generated transgenic mice specifically expressing ovVEGF-E in ß-cells of the endocrine pancreas (Rip1VEGF-E; RVE). RVE mice show an increase in number and size of the islets of Langerhans and a distorted organization of insulin and glucagon-expressing cells. Islet endothelial cells of RVE mice hyper-proliferate and form increased numbers of functional blood vessels. In addition, the formation of disorganized lymphatic vessels and increased immune cell infiltration is observed. Upon crossing RVE single-transgenic mice with Rip1Tag2 (RT2) transgenic mice, a well-studied model of pancreatic ß-cell carcinogenesis, double-transgenic mice (RT2;RVE) display hyper-proliferation of endothelial cells resulting in the formation of hemangioma-like lesions. In addition, RT2;RVE mice exhibit activated lymphangiogenesis at the tumor periphery and increased neutrophil and macrophage tumor infiltration and micro-metastasis to lymph nodes and lungs. These phenotypes markedly differ from the phenotypes observed with the transgenic expression of the other VEGF family members in ß-cells of normal mice and of RT2 mice.


Assuntos
Hemangioma/etiologia , Neoplasias Pancreáticas/etiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Virais/metabolismo , Animais , Proliferação de Células , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Cabras , Hemangioma/metabolismo , Hemangioma/patologia , Humanos , Células Secretoras de Insulina/metabolismo , Linfangiogênese/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ovinos , Transdução de Sinais , Proteínas Virais/genética
3.
Cancer Res ; 71(17): 5717-27, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21778249

RESUMO

Members of the Angiopoietin family regulate various aspects of physiologic and pathologic angiogenesis. Although Angiopoietin-1 (Ang-1) decreases endothelial cell permeability and increases vascular stabilization via recruitment of pericytes and smooth muscle cells to growing blood vessels, Angiopoietin-2 (Ang-2) mediates angiogenic sprouting and vascular regression. In this study, we used the Rip1Tag2 transgenic mouse model of pancreatic ß-cell carcinogenesis to investigate the roles of Ang-1 and Ang-2 in tumor angiogenesis and tumor progression. On their own, transgenic expression of human Ang-1 or Ang-2 in pancreatic ß cells caused formation of peri-insular lymphatic vessels in the absence of effects on blood vessel density, islet morphology, or physiology. When crossed to Rip1Tag2 mice, both Ang-1-and Ang-2-expressing ß-cell tumors showed increased peritumoral lymphangiogenesis in the absence of metastasis to local lymph nodes or distant organs. There was no alteration in tumor outgrowth, blood vessel density, or vessel maturation in Ang-1-expressing tumors. In contrast, Ang-2-expressing tumors exhibited diminished pericyte recruitment to blood vessels that were dilated, nonfunctional, and highly permeable. These tumors were hemorrhagic, highly infiltrated by leukocytes, and impaired in outgrowth. Together, our findings establish that Ang-2 antagonizes Ang-1 function, leading to excessive vessel sprouting with impaired pericyte recruitment and vessel stabilization. The poor perfusion of immature blood vessels results in retarded tumor growth, defining an important pathophysiologic pathway required for efficient tumorigenesis.


Assuntos
Angiopoietina-1/antagonistas & inibidores , Angiopoietina-2/metabolismo , Linfangiogênese , Neovascularização Patológica/metabolismo , Neoplasias Pancreáticas/irrigação sanguínea , Angiopoietina-1/genética , Angiopoietina-1/metabolismo , Angiopoietina-2/genética , Animais , Células Cultivadas , Humanos , Camundongos , Camundongos Transgênicos , Neovascularização Patológica/genética , Neoplasias Pancreáticas/patologia , Regiões Promotoras Genéticas , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
PLoS One ; 5(11): e14109, 2010 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21124841

RESUMO

BACKGROUND: The family of vascular endothelial growth factors (VEGF) contains key regulators of blood and lymph vessel development, including VEGF-A, -B, -C, -D, and placental growth factor. The role of VEGF-B during physiological or pathological angiogenesis has not yet been conclusively delineated. Herein, we investigate the function of VEGF-B by the generation of mouse models of cancer with transgenic expression of VEGF-B or homozygous deletion of Vegfb. METHODOLOGY/PRINCIPAL FINDINGS: Ectopic expression of VEGF-B in the insulin-producing ß-cells of the pancreas did not alter the abundance or architecture of the islets of Langerhans. The vasculature from transgenic mice exhibited a dilated morphology, but was of similar density as that of wildtype mice. Unexpectedly, we found that transgenic expression of VEGF-B in the RIP1-Tag2 mouse model of pancreatic neuroendocrine tumorigenesis retarded tumor growth. Conversely, RIP1-Tag2 mice deficient for Vegfb presented with larger tumors. No differences in vascular density, perfusion or immune cell infiltration upon altered Vegfb gene dosage were noted. However, VEGF-B acted to increase blood vessel diameter both in normal pancreatic islets and in RIP1-Tag2 tumors. CONCLUSIONS/SIGNIFICANCE: Taken together, our results illustrate the differences in biological function between members of the VEGF family, and highlight the necessity of in-depth functional studies of VEGF-B to fully understand the effects of VEGFR-1 inhibitors currently used in the clinic.


Assuntos
Células Secretoras de Insulina/metabolismo , Tumores Neuroendócrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Fator B de Crescimento do Endotélio Vascular/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Humanos , Immunoblotting , Imuno-Histoquímica , Células Secretoras de Insulina/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Tumores Neuroendócrinos/genética , Tumores Neuroendócrinos/patologia , Pâncreas/irrigação sanguínea , Pâncreas/metabolismo , Pâncreas/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Carga Tumoral , Fator B de Crescimento do Endotélio Vascular/genética
5.
Cancer Res ; 67(22): 10840-8, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18006829

RESUMO

Members of the vascular endothelial growth factor (VEGF) family are critical players in angiogenesis and lymphangiogenesis. Although VEGF-A has been shown to exert fundamental functions in physiologic and pathologic angiogenesis, the exact role of the VEGF family member placental growth factor (PlGF) in tumor angiogenesis has remained controversial. To gain insight into PlGF function during tumor angiogenesis, we have generated transgenic mouse lines expressing human PlGF-1 in the beta cells of the pancreatic islets of Langerhans (Rip1PlGF-1). In single-transgenic Rip1PlGF-1 mice, intra-insular blood vessels are found highly dilated, whereas islet physiology is unaffected. Upon crossing of these mice with the Rip1Tag2 transgenic mouse model of pancreatic beta cell carcinogenesis, tumors of double-transgenic Rip1Tag2;Rip1PlGF-1 mice display reduced growth due to attenuated tumor angiogenesis. The coexpression of transgenic PlGF-1 and endogenous VEGF-A in the beta tumor cells of double-transgenic animals causes the formation of low-angiogenic hPlGF-1/mVEGF-A heterodimers at the expense of highly angiogenic mVEGF-A homodimers resulting in diminished tumor angiogenesis and reduced tumor infiltration by neutrophils, known to contribute to the angiogenic switch in Rip1Tag2 mice. The results indicate that the ratio between the expression levels of two members of the VEGF family of angiogenic factors, PlGF-1 and VEGF-A, determines the overall angiogenic activity and, thus, the extent of tumor angiogenesis and tumor growth.


Assuntos
Células Secretoras de Insulina/metabolismo , Neovascularização Patológica , Neoplasias Pancreáticas/patologia , Proteínas da Gravidez/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Colágeno/química , Dimerização , Ensaio de Imunoadsorção Enzimática , Humanos , Ilhotas Pancreáticas/metabolismo , Metilmetacrilato/química , Camundongos , Camundongos Transgênicos , Invasividade Neoplásica , Neoplasias Pancreáticas/metabolismo , Fator de Crescimento Placentário , Proteínas da Gravidez/metabolismo
6.
Am J Pathol ; 170(4): 1348-61, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17392173

RESUMO

In many human carcinomas, expression of the lymphangiogenic factor vascular endothelial growth factor-D (VEGF-D) correlates with up-regulated lymphangiogenesis and regional lymph node metastasis. Here, we have used the Rip1Tag2 transgenic mouse model of pancreatic beta-cell carcinogenesis to investigate the functional role of VEGF-D in the induction of lymphangiogenesis and tumor progression. Expression of VEGF-D in beta cells of single-transgenic Rip1VEGF-D mice resulted in the formation of peri-insular lymphatic lacunae, often containing leukocyte accumulations and blood hemorrhages. When these mice were crossed to Rip1Tag2 mice, VEGF-D-expressing tumors also exhibited peritumoral lymphangiogenesis with lymphocyte accumulations and hemorrhages, and they frequently developed lymph node and lung metastases. Notably, tumor outgrowth and blood microvessel density were significantly reduced in VEGF-D-expressing tumors. Our results demonstrate that VEGF-D induces lymphangiogenesis, promotes metastasis to lymph nodes and lungs, and yet represses hemangiogenesis and tumor outgrowth. Because a comparable transgenic expression of vascular endothelial growth factor-C (VEGF-C) in Rip1Tag2 has been shown previously to provoke lymphangiogenesis and lymph node metastasis in the absence of any distant metastasis, leukocyte infiltration, or angiogenesis-suppressing effects, these results reveal further functional differences between VEGF-D and VEGF-C.


Assuntos
Linfangiogênese/fisiologia , Metástase Neoplásica/fisiopatologia , Fator D de Crescimento do Endotélio Vascular/fisiologia , Animais , Células Cultivadas , Feminino , Genótipo , Humanos , Immunoblotting , Leucócitos/metabolismo , Leucócitos/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/fisiopatologia , Neoplasias Pulmonares/secundário , Linfonodos/metabolismo , Linfonodos/patologia , Linfangiogênese/genética , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patologia , Vasos Linfáticos/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Metástase Neoplásica/genética , Neovascularização Patológica/genética , Neovascularização Patológica/fisiopatologia , Neoplasias Pancreáticas/irrigação sanguínea , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/ultraestrutura , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/fisiologia , Fator D de Crescimento do Endotélio Vascular/genética , Fator D de Crescimento do Endotélio Vascular/metabolismo
7.
Cancer Res ; 64(23): 8630-8, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15574770

RESUMO

Reduced expression of neural cell adhesion molecule (NCAM) has been implicated in the progression to tumor malignancy in cancer patients. Previously, we have shown that the loss of NCAM function causes the formation of lymph node metastasis in a transgenic mouse model of pancreatic beta cell carcinogenesis (Rip1Tag2). Here we show that tumors of NCAM-deficient Rip1Tag2 transgenic mice exhibit up-regulated expression of the lymphangiogenic factors vascular endothelial growth factor (VEGF)-C and -D (17% in wild-type versus 60% in NCAM-deficient Rip1Tag2 mice) and, with it, increased lymphangiogenesis (0% in wild-type versus 19% in NCAM-deficient Rip1Tag2 mice). Repression of VEGF-C and -D function by adenoviral expression of a soluble form of their cognate receptor, VEGF receptor-3, results in reduced tumor lymphangiogenesis (56% versus 28% in control versus treated mice) and lymph node metastasis (36% versus 8% in control versus treated mice). The results indicate that the loss of NCAM function causes lymph node metastasis via VEGF-C- and VEGF-D-mediated lymphangiogenesis. These results also establish Rip1Tag2;NCAM-deficient mice as a unique model for stochastic, endogenous tumor lymphangiogenesis and lymph node metastasis in immunocompetent mice.


Assuntos
Ilhotas Pancreáticas/patologia , Linfangiogênese/fisiologia , Moléculas de Adesão de Célula Nervosa/deficiência , Neoplasias Pancreáticas/patologia , Animais , Metástase Linfática , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patologia , Camundongos , Camundongos Transgênicos , Neovascularização Patológica/patologia , Moléculas de Adesão de Célula Nervosa/biossíntese , Neoplasias Pancreáticas/irrigação sanguínea , Neoplasias Pancreáticas/metabolismo , Regulação para Cima , Fator C de Crescimento do Endotélio Vascular/biossíntese , Fator D de Crescimento do Endotélio Vascular/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA