Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
EMBO Rep ; 16(6): 753-68, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25916856

RESUMO

T-cell-dependent antigenic stimulation drives the differentiation of B cells into antibody-secreting plasma cells and memory B cells, but how B cells regulate this process is unclear. We show that LKB1 expression in B cells maintains B-cell quiescence and prevents the premature formation of germinal centers (GCs). Lkb1-deficient B cells (BKO) undergo spontaneous B-cell activation and secretion of multiple inflammatory cytokines, which leads to splenomegaly caused by an unexpected expansion of T cells. Within this cytokine response, increased IL-6 production results from heightened activation of NF-κB, which is suppressed by active LKB1. Secreted IL-6 drives T-cell activation and IL-21 production, promoting T follicular helper (TFH ) cell differentiation and expansion to support a ~100-fold increase in steady-state GC B cells. Blockade of IL-6 secretion by BKO B cells inhibits IL-21 expression, a known inducer of TFH -cell differentiation and expansion. Together, these data reveal cell intrinsic and surprising cell extrinsic roles for LKB1 in B cells that control TFH -cell differentiation and GC formation, and place LKB1 as a central regulator of T-cell-dependent humoral immunity.


Assuntos
Linfócitos B/imunologia , Linfócitos B/metabolismo , Centro Germinativo/fisiologia , Ativação Linfocitária , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/genética , Linfócitos T Auxiliares-Indutores/imunologia , Proteínas Quinases Ativadas por AMP , Animais , Diferenciação Celular , Interleucina-6/imunologia , Interleucina-6/metabolismo , Interleucinas/imunologia , Camundongos , NF-kappa B/genética , Linfócitos T Auxiliares-Indutores/fisiologia
2.
Blood ; 118(22): 5872-82, 2011 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21908434

RESUMO

The contributions of the host microenvironment to the pathogenesis of multiple myeloma, including progression from the non-malignant disorder monoclonal gammopathy of undetermined significance, are poorly understood. In the present study, microarray analysis of a murine model requiring a unique host microenvironment for myeloma development identified decreased host-derived adiponectin compared with normal mice. In support, clinical analysis revealed decreased serum adiponectin concentrations in monoclonal gammopathy of undetermined significance patients who subsequently progressed to myeloma. We investigated the role of adiponectin in myeloma pathogenesis and as a treatment approach, using both mice deficient in adiponectin and pharmacologic enhancement of circulating adiponectin. Increased tumor burden and bone disease were observed in myeloma-bearing adiponectin-deficient mice, and adiponectin was found to induce myeloma cell apoptosis. The apolipoprotein peptide mimetic L-4F was used for pharmacologic enhancement of adiponectin. L-4F reduced tumor burden, increased survival of myeloma-bearing mice, and prevented myeloma bone disease. Collectively, our studies have identified a novel mechanism whereby decreased host-derived adiponectin promotes myeloma tumor growth and osteolysis. Furthermore, we have established the potential therapeutic benefit of increasing adiponectin for the treatment of myeloma and the associated bone disease.


Assuntos
Neoplasias Ósseas/terapia , Terapia de Alvo Molecular , Mieloma Múltiplo/terapia , Adiponectina/genética , Adiponectina/fisiologia , Animais , Doenças Ósseas/etiologia , Doenças Ósseas/genética , Neoplasias Ósseas/genética , Neoplasias Ósseas/secundário , Linhagem Celular Tumoral , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Transplante de Neoplasias , Peptídeos/administração & dosagem , Peptídeos/uso terapêutico , Células Tumorais Cultivadas , Microambiente Tumoral , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Plast Reconstr Surg ; 147(5): 1229-1233, 2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-33890911

RESUMO

SUMMARY: The persistence of health care disparities along racial and ethnic lines highlights the complex and multifactorial nature of this national concern. The paucity of physicians ethnically underrepresented in medicine to treat an ever-growing heterogeneous population inherently contributes to these ongoing disparities. The authors proposed an approach to improve the representation of physicians underrepresented in medicine in their plastic surgery residency program. With a renewed commitment to ethnic diversity and inclusion, a multifaceted recruitment and retention approach was implemented at the University of Pennsylvania plastic and reconstructive surgery residency program from 2015 to 2020 (5 academic years). A retrospective review of the demographics of the program's residents was then assessed over the past 9 academic years for comparison (2011 to 2020). The representation of underrepresented-in-medicine residents within the plastic and reconstructive surgery residency program steadily increased with the implementation of this multifaceted approach, reaching an unprecedented high. Currently, 29 percent of all residents are underrepresented in medicine and 29 percent are female, some of whom are also underrepresented in medicine. Although the female representation is on par with the national average, the underrepresented-in-medicine representation is far greater than the national average. As a result of this multifaceted approach, the representation of African American and Latino plastic surgery residents at the University of Pennsylvania now far exceeds current national averages. Unfortunately, the representation of Native American and Alaskan Natives is still lacking, despite the program's broadened recruitment efforts. The success of this experience describes a successful strategy that institutions can implement to enhance underrepresented-in-medicine representation among its plastic surgery trainees.


Assuntos
Etnicidade , Mão de Obra em Saúde , Internato e Residência , Grupos Minoritários , Seleção de Pessoal/métodos , Grupos Raciais , Cirurgia Plástica , Feminino , Humanos , Masculino , Pennsylvania , Estudos Retrospectivos , Cirurgia Plástica/educação
4.
Clin Cancer Res ; 26(7): 1541-1554, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31672768

RESUMO

The progression of multiple myeloma, a hematologic malignancy characterized by unregulated plasma cell growth, is associated with increasing innate and adaptive immune system dysfunction, notably in the T-cell repertoire. Although treatment advances in multiple myeloma have led to deeper and more durable clinical responses, the disease remains incurable for most patients. Therapeutic strategies aimed at overcoming the immunosuppressive tumor microenvironment and activating the host immune system have recently shown promise in multiple myeloma, particularly in the relapsed and/or refractory disease setting. As the efficacy of T-cell-dependent immuno-oncology therapy is likely affected by the health of the endogenous T-cell repertoire, these therapies may also provide benefit in alternate treatment settings (e.g., precursor disease; after stem cell transplantation). This review describes T-cell-associated changes during the evolution of multiple myeloma and provides an overview of T-cell-dependent immuno-oncology approaches under investigation. Vaccine and checkpoint inhibitor interventions are being explored across the multiple myeloma disease continuum; treatment modalities that redirect patient T cells to elicit an anti-multiple myeloma response, namely, chimeric antigen receptor (CAR) T cells and bispecific antibodies [including BiTE (bispecific T-cell engager) molecules], have been primarily evaluated to date in the relapsed and/or refractory disease setting. CAR T cells and bispecific antibodies/antibody constructs directed against B-cell maturation antigen have generated excitement, with clinical data demonstrating deep responses. An increased understanding of the complex interplay between the immune system and multiple myeloma throughout the disease course will aid in maximizing the potential for T-cell-dependent immuno-oncology strategies in multiple myeloma.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Imunoterapia/métodos , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/terapia , Plasmócitos/patologia , Linfócitos T/imunologia , Humanos , Mieloma Múltiplo/patologia , Plasmócitos/imunologia
5.
Am J Hematol ; 84(5): 268-72, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19296472

RESUMO

The proteasome inhibitor bortezomib has a striking clinical benefit in patients with multiple myeloma. It is unknown whether the bone marrow microenvironment directly contributes to the dramatic response of myeloma cells to proteasome inhibition in vivo. We have used the well-characterized 5TGM1 murine model of myeloma to investigate myeloma growth within bone and response to the proteasome inhibitor bortezomib in vivo. Myeloma cells freshly isolated from the bone marrow of myeloma-bearing mice were found to have an increase in proteasome activity and an enhanced response to in vitro proteasome inhibition, as compared with pre-inoculation myeloma cells. Treatment of myeloma-bearing mice with bortezomib resulted in a greater reduction in tumor burden when the myeloma cells were located within the bone marrow when compared with extra-osseous sites. Our results demonstrate that myeloma cells exhibit an increase in proteasome activity and an enhanced response to bortezomib treatment when located within the bone marrow microenvironment in vivo.


Assuntos
Ácidos Borônicos/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/enzimologia , Inibidores de Proteases/farmacologia , Inibidores de Proteassoma , Pirazinas/farmacologia , Animais , Medula Óssea/patologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Bortezomib , Comunicação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ativação Enzimática , Imunoglobulina G/sangue , Camundongos , Mieloma Múltiplo/patologia , Transplante de Neoplasias , Complexo de Endopeptidases do Proteassoma/metabolismo , Carga Tumoral/efeitos dos fármacos
6.
Invest Ophthalmol Vis Sci ; 48(2): 907-15, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17251494

RESUMO

PURPOSE: The efficacy of three matrix metalloproteinase (MMP) inhibitors with various selectivities (Ro-31-9790, AG3340, and DPC-A37668) was investigated in a rat model of retinopathy of prematurity, to examine the roles of MMP-2 and -9 in retinal neovascularization. The susceptibilities of MMP-2(-/-) and -9(-/-) mice to preretinal neovascularization were investigated in a mouse model of oxygen-induced retinopathy. METHODS: Sprague-Dawley newborn rats were exposed to alternating episodes of 50% and 10% oxygen (variable oxygen exposure) to induce retinal neovascularization. Three MMP inhibitors with various selectivity profiles were administered to variable oxygen-exposed rats via local or systemic routes. Antineovascular efficacy was determined in drug-treated versus vehicle-treated rat pups by computerized imaging of adenosine diphosphatase (ADPase)-stained retinal flatmounts. Wild-type C57BL/6J and isogenic MMP-2(-/-) and -9(-/-) mice were exposed to 75% oxygen followed by normoxia. The mice were killed immediately before or after the normoxic exposure, and eyes were either harvested for retinal dissection and flatmounting or were paraffin embedded and sectioned. Retinal vascular area and retinal neovascularization were assessed by adenosine diphosphatase staining of retinal flatmounts and by counting preretinal nuclei of hematoxylin and eosin-stained retinal sections, respectively. RESULTS: Ro-31-9790, AG3340, and DPC-A37668 had no effect on normal development of the rat retinal vasculature, regardless of dose or route of administration. Intravitreal injection of Ro-31-9790 (broad-spectrum) immediately after variable-oxygen exposure and 2 days after exposure resulted in 78% and 82% inhibition of retinal neovascularization, respectively. AG3340 (MMP-2- and -9-selective inhibitor) and DPC-A37668 (MMP-2-selective inhibitor) resulted in 65% and 52% inhibition, respectively, when administered by intravitreal injection immediately after variable-oxygen exposure. Intraperitoneal injection of 5, 15, and 50 mg/mL AG3340 or DPC-A37668 for 6 days after variable oxygen exposure resulted in 22% to 39% and 0% to 31% inhibition of neovascularization, respectively. AG3340 and DPC-A37668 administered by oral gavage at doses of 3, 10, or 30 mg/mL provided up to 42% and 86% inhibition of neovascularization, respectively. The average vascular areas of retinas from MMP-2(-/-) or -9(-/-) mice at postnatal day 12 were not significantly different from the wild-type control. There was a 75% (P < 0.001) and 44% (P < 0.01) reduction in preretinal neovascularization in oxygen-exposed MMP-2(-/-) and -9(-/-) mice at postnatal day 19, respectively, compared with wild-type control mice. CONCLUSIONS: The results of this study suggest that MMP-2 plays a predominant role in retinal angiogenesis in both the mouse and rat models of oxygen-induced retinopathy. Furthermore, MMP-2 inhibition may be a viable therapeutic approach for ocular diseases characterized by retinal neovascularization.


Assuntos
Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Inativação Gênica/fisiologia , Metaloproteinase 2 da Matriz/fisiologia , Metaloproteinase 9 da Matriz/fisiologia , Neovascularização Retiniana/enzimologia , Retinopatia da Prematuridade/enzimologia , Animais , Animais Recém-Nascidos , Apirase/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Recém-Nascido , Injeções , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos Endogâmicos C57BL , Compostos Orgânicos/farmacologia , Oxigênio/toxicidade , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Retina/enzimologia , Neovascularização Retiniana/tratamento farmacológico , Neovascularização Retiniana/patologia , Retinopatia da Prematuridade/tratamento farmacológico , Retinopatia da Prematuridade/patologia , Corpo Vítreo
7.
Immunol Lett ; 95(1): 37-44, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15325796

RESUMO

Interleukin-3 (IL-3), which is derived from T cells and other sources, can promote the differentiation, proliferation, and migration of mast cells, basophils, and eosinophils. However, little is known about the ability of IL-3 to regulate the function of these cells in IgE-dependent and -independent allergic responses in vivo. Therefore, we sought to investigate the extent to which endogenously produced IL-3 can influence mast cell secretory function, the expression of local and systemic anaphylactic responses, and ragweed-induced eosinophilic peritonitis. We found that peritoneal mast cells from IL-3 deficient (IL-3 -/-) mice released less serotonin following challenge with low doses of anti-IgE antibody or antigen ex vivo than do cells isolated from corresponding wild-type (IL-3 +/+) mice. Both IL-3 -/- and +/+ mice expressed equivalent IgE-dependent passive cutaneous anaphylaxis responses following challenge with specific antigen and exhibited equivalent active systemic anaphylaxis responses to ovalbumin as assessed by changes in body temperature, death rates, total IgE production, and histamine release. In contrast, ragweed allergen immunization and peritoneal allergen challenge resulted in eosinophil recruitment that was greater in IL-3 -/- mice than in IL-3 +/+ mice. Our data demonstrates that IL-3 does not appear to be essential for local or systemic anaphylaxis. However, IL-3 production in vivo was found to enhance the mediator release from freshly isolated peritoneal mast cells stimulated ex vivo, and, unexpectedly, to inhibit the accumulation of eosinophils associated with a ragweed-induced allergic peritonitis model.


Assuntos
Anafilaxia/imunologia , Eosinófilos/imunologia , Interleucina-3/fisiologia , Anafilaxia Cutânea Passiva/imunologia , Peritonite/imunologia , Rinite Alérgica Sazonal/imunologia , Alérgenos/imunologia , Ambrosia/imunologia , Anafilaxia/etiologia , Animais , Modelos Animais de Doenças , Mastócitos/citologia , Mastócitos/metabolismo , Camundongos , Cavidade Peritoneal/citologia , Serotonina/biossíntese
8.
Cancer Res ; 72(9): 2183-9, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22374979

RESUMO

The rapid progression of multiple myeloma is dependent upon cellular interactions within the bone marrow microenvironment. In vitro studies suggest that bone marrow stromal cells (BMSC) can promote myeloma growth and survival and osteolytic bone disease. However, it is not possible to recreate all cellular aspects of the bone marrow microenvironment in an in vitro system, and the contributions of BMSCs to myeloma pathogenesis in an intact, immune competent, in vivo system are unknown. To investigate this, we used a murine myeloma model that replicates many features of the human disease. Coinoculation of myeloma cells and a BMSC line, isolated from myeloma-permissive mice, into otherwise nonpermissive mice resulted in myeloma development, associated with tumor growth within bone marrow and osteolytic bone disease. In contrast, inoculation of myeloma cells alone did not result in myeloma. BMSCs inoculated alone induced osteoblast suppression, associated with an increase in serum concentrations of the Wnt signaling inhibitor, Dkk1. Dkk1 was highly expressed in BMSCs and in myeloma-permissive bone marrow. Knockdown of Dkk1 expression in BMSCs decreased their ability to promote myeloma and the associated bone disease in mice. Collectively, our results show novel roles of BMSCs and BMSC-derived Dkk1 in the pathogenesis of multiple myeloma in vivo.


Assuntos
Células da Medula Óssea/patologia , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Mieloma Múltiplo/patologia , Células Estromais/patologia , Animais , Células da Medula Óssea/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Células Estromais/metabolismo , Microambiente Tumoral , Proteínas Wnt/antagonistas & inibidores
9.
Bone ; 48(1): 121-8, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20615487

RESUMO

Multiple myeloma is a hematological malignancy that is associated with the development of a destructive osteolytic bone disease, which is a major cause of morbidity for patients with myeloma. Interactions between myeloma cells and cells of the bone marrow microenvironment promote both tumor growth and survival and bone destruction, and the osteolytic bone disease is now recognized as a contributing component to tumor progression. Since myeloma bone disease is associated with both an increase in osteoclastic bone resorption and a suppression of osteoblastic bone formation, research to date has largely focused upon the role of the osteoclast and osteoblast. However, it is now clear that other cell types within the bone marrow, including cells of the immune system, mesenchymal stem cells and bone marrow stromal cells, can contribute to the development of myeloma bone disease. This review discusses the cellular mechanisms and potential therapeutic targets that have been implicated in myeloma bone disease.


Assuntos
Doenças Ósseas/patologia , Comunicação Celular , Mieloma Múltiplo/complicações , Mieloma Múltiplo/patologia , Doenças Ósseas/complicações , Doenças Ósseas/etiologia , Medula Óssea/metabolismo , Medula Óssea/patologia , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Humanos , Mieloma Múltiplo/tratamento farmacológico , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteogênese
10.
Dis Model Mech ; 2(11-12): 604-11, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19779066

RESUMO

Multiple myeloma, and the associated osteolytic bone disease, is highly dependent upon cellular interactions within the bone marrow microenvironment. A major limitation of existing myeloma models is the requirement for a specific host strain of mouse, preventing molecular examination of the bone marrow microenvironment. The aim of the current study was to develop a model of myeloma in which the host microenvironment could be modified genetically. The Radl 5T murine model of myeloma is well characterized and closely mimics human myeloma. In the current study, we demonstrate 5T myeloma establishment in recombination activating gene 2 (RAG-2)-deficient mice, which have improper B- and T-cell development. Importantly, these mice can be easily bred with genetically modified mice to generate double knockout mice, allowing manipulation of the host microenvironment at a molecular level. Inoculation of 5TGM1 myeloma cells into RAG-2(-/-) mice resulted in myeloma development, which was associated with tumor growth within bone and an osteolytic bone disease, as assessed by microcomputed tomography (microCT), histology and histomorphometry. Myeloma-bearing RAG-2(-/-) mice displayed many features that were similar to both human myeloma and the original Radl 5T model. To demonstrate the use of this model, we have examined the effect of host-derived matrix metalloproteinase 9 (MMP-9) in the development of myeloma in vivo. Inoculation of 5TGM1 myeloma cells into mice that are deficient in RAG-2 and MMP-9 resulted in a reduction in both tumor burden and osteolytic bone disease when compared with RAG-2-deficient wild-type myeloma-bearing mice. The establishment of myeloma in RAG-2(-/-) mice permits molecular examination of the host contribution to myeloma pathogenesis in vivo.


Assuntos
Doenças Ósseas/genética , Modelos Animais de Doenças , Mieloma Múltiplo/genética , Mieloma Múltiplo/fisiopatologia , Animais , Doenças Ósseas/fisiopatologia , Proteínas de Ligação a DNA/genética , Feminino , Fêmur/patologia , Técnicas Genéticas , Imuno-Histoquímica/métodos , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Tíbia/patologia , Tomografia Computadorizada por Raios X/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA