Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Front Immunol ; 10: 2547, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31749806

RESUMO

Background: Ultrasound guided sampling of human lymph node (LN) combined with advanced flow cytometry allows phenotypic analysis of multiple immune cell subsets. These may provide insights into immune processes and responses to immunotherapies not apparent from analysis of the blood. Methods: Ultrasound guided inguinal LN samples were obtained by both fine needle aspiration (FNA) and core needle biopsy in 10 adults within 8 weeks of diagnosis of type 1 diabetes (T1D) and 12 age-matched healthy controls at two study centers. Peripheral blood mononuclear cells (PBMC) were obtained on the same occasion. Samples were transported same day to the central laboratory and analyzed by multicolour flow cytometry. Results: LN sampling was well-tolerated and yielded sufficient cells for analysis in 95% of cases. We confirmed the segregation of CD69+ cells into LN and the predominance of CD8+ Temra cells in blood previously reported. In addition, we demonstrated clear enrichment of CD8+ naïve, FOXP3+ Treg, class-switched B cells, CD56bright NK cells and plasmacytoid dendritic cells (DC) in LNs as well as CD4+ T cells of the Th2 phenotype and those expressing Helios and Ki67. Conventional NK cells were virtually absent from LNs as were Th22 and Th1Th17 cells. Paired correlation analysis of blood and LN in the same individuals indicated that for many cell subsets, especially those associated with activation: such as CD25+ and proliferating (Ki67+) T cells, activated follicular helper T cells and class-switched B cells, levels in the LN compartment could not be predicted by analysis of blood. We also observed an increase in Th1-like Treg and less proliferating (Ki67+) CD4+ T cells in LN from T1D compared to control LNs, changes which were not reflected in the blood. Conclusions: LN sampling in humans is well-tolerated. We provide the first detailed "roadmap" comparing immune subsets in LN vs. blood emphasizing a role for differentiated effector T cells in the blood and T cell regulation, B cell activation and memory in the LN. For many subsets, frequencies in blood, did not correlate with LN, suggesting that LN sampling would be valuable for monitoring immuno-therapies where these subsets may be impacted.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Citometria de Fluxo , Linfonodos/imunologia , Linfócitos/imunologia , Adulto , Diabetes Mellitus Tipo 1/patologia , Feminino , Humanos , Linfonodos/patologia , Linfócitos/patologia , Masculino
2.
JCI Insight ; 2(16)2017 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-28814669

RESUMO

The maintenance of peripheral naive T lymphocytes in humans is dependent on their homeostatic division, not continuing emigration from the thymus, which undergoes involution with age. However, postthymic maintenance of naive T cells is still poorly understood. Previously we reported that recent thymic emigrants (RTEs) are contained in CD31+CD25- naive T cells as defined by their levels of signal joint T cell receptor rearrangement excision circles (sjTRECs). Here, by differential gene expression analysis followed by protein expression and functional studies, we define that the naive T cells having divided the least since thymic emigration express complement receptors (CR1 and CR2) known to bind complement C3b- and C3d-decorated microbial products and, following activation, produce IL-8 (CXCL8), a major chemoattractant for neutrophils in bacterial defense. We also observed an IL-8-producing memory T cell subpopulation coexpressing CR1 and CR2 and with a gene expression signature resembling that of RTEs. The functions of CR1 and CR2 on T cells remain to be determined, but we note that CR2 is the receptor for Epstein-Barr virus, which is a cause of T cell lymphomas and a candidate environmental factor in autoimmune disease.

3.
Cancer Manag Res ; 9: 65-72, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28293120

RESUMO

BACKGROUND: Glucocorticoid receptor (GR) activity plays a role in many aspects of human physiology and may play a crucial role in chemotherapy resistance in a wide variety of solid tumors. A novel immunohistochemistry (IHC) based assay has been previously developed and validated in order to assess GR immunoreactivity in triple-negative breast cancer. The current study investigates the standardized use of this validated assay to assess GR expression in a broad range of solid tumor malignancies. METHODS: Archived formalin-fixed paraffin-embedded tumor bank samples (n=236) from 20 different solid tumor types were analyzed immunohistochemically. Nuclear staining was reported based on the H-score method using differential intensity scores (0, 1+, 2+, or 3+) with the percent stained (out of at least 100 carcinoma cells) recorded at each intensity. RESULTS: GR was expressed in all tumor types that had been evaluated. Renal cell carcinoma, sarcoma, cervical cancer, and melanoma were those with the highest mean H-scores, indicating high levels of GR expression. Colon, endometrial, and gastric cancers had lower GR staining percentages and intensities, resulting in the lowest mean H-scores. CONCLUSION: A validated IHC assay revealed GR immunoreactivity in all solid tumor types studied and allowed for standardized comparison of reactivity among the different malignancies. IMPACT: Baseline expression levels of GR may be a useful biomarker when pharmaceutically targeting GR in research or clinical setting.

4.
Arch Pathol Lab Med ; 140(11): 1259-1266, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27788043

RESUMO

CONTEXT: - With the abundance of therapeutics targeted against programmed death receptor-1 and its ligand (PD-L1) that are currently approved or in clinical development, there is interest in identifying those patients most likely to respond to these drugs. Expression of PD-L1 may be an indicator of an initial and robust inflammatory response to the presence of tumor cells. Therefore, tumors that express PD-L1 may be the most likely to respond to therapies that interrupt the negative feedback mechanism that leads to PD-L1 upregulation. OBJECTIVE: - To develop a prototype immunohistochemistry assay using the anti-PD-L1 antibody clone 22C3. DESIGN: - The assay was developed and optimized using commercially available reagents and archival tumor-bank tissue. RESULTS: - The optimized immunohistochemistry method had high precision and reproducibility. Using the prototype assay in 142 non-small cell lung cancer and 79 melanoma archival tumor-bank tissue samples, PD-L1 staining was observed at the plasma membrane of nucleated tumor and nontumor cells and, in some cases, as a distinct lichenoid pattern at the tumor-stroma border. Using a preliminary scoring method, 56% (80 of 142) of non-small cell lung cancer and 53% (42 of 79) of melanoma samples were defined as PD-L1+ based on a modified H-score of 1 or more or the presence of a distinctive staining pattern at the tumor-stroma interface. CONCLUSIONS: - The immunohistochemistry assay using the anti-PD-L1 antibody 22C3 merits further investigation in clinical trials and prevalence assessments to further understand the prognostic and predictive value of PD-L1 expression in cancer.


Assuntos
Anticorpos Monoclonais/metabolismo , Antígeno B7-H1/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Animais , Especificidade de Anticorpos , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/genética , Biomarcadores Tumorais/metabolismo , Células CHO , Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Carcinoma Pulmonar de Células não Pequenas/patologia , Membrana Celular/metabolismo , Membrana Celular/patologia , Células Clonais , Cricetulus , Humanos , Imuno-Histoquímica , Pulmão/metabolismo , Pulmão/patologia , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/patologia , Melanoma/diagnóstico , Melanoma/patologia , Camundongos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Bancos de Tecidos
5.
PLoS Med ; 13(10): e1002139, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27727279

RESUMO

BACKGROUND: Interleukin-2 (IL-2) has an essential role in the expansion and function of CD4+ regulatory T cells (Tregs). Tregs reduce tissue damage by limiting the immune response following infection and regulate autoreactive CD4+ effector T cells (Teffs) to prevent autoimmune diseases, such as type 1 diabetes (T1D). Genetic susceptibility to T1D causes alterations in the IL-2 pathway, a finding that supports Tregs as a cellular therapeutic target. Aldesleukin (Proleukin; recombinant human IL-2), which is administered at high doses to activate the immune system in cancer immunotherapy, is now being repositioned to treat inflammatory and autoimmune disorders at lower doses by targeting Tregs. METHODS AND FINDINGS: To define the aldesleukin dose response for Tregs and to find doses that increase Tregs physiologically for treatment of T1D, a statistical and systematic approach was taken by analysing the pharmacokinetics and pharmacodynamics of single doses of subcutaneous aldesleukin in the Adaptive Study of IL-2 Dose on Regulatory T Cells in Type 1 Diabetes (DILT1D), a single centre, non-randomised, open label, adaptive dose-finding trial with 40 adult participants with recently diagnosed T1D. The primary endpoint was the maximum percentage increase in Tregs (defined as CD3+CD4+CD25highCD127low) from the baseline frequency in each participant measured over the 7 d following treatment. There was an initial learning phase with five pairs of participants, each pair receiving one of five pre-assigned single doses from 0.04 × 106 to 1.5 × 106 IU/m2, in order to model the dose-response curve. Results from each participant were then incorporated into interim statistical modelling to target the two doses most likely to induce 10% and 20% increases in Treg frequencies. Primary analysis of the evaluable population (n = 39) found that the optimal doses of aldesleukin to induce 10% and 20% increases in Tregs were 0.101 × 106 IU/m2 (standard error [SE] = 0.078, 95% CI = -0.052, 0.254) and 0.497 × 106 IU/m2 (SE = 0.092, 95% CI = 0.316, 0.678), respectively. On analysis of secondary outcomes, using a highly sensitive IL-2 assay, the observed plasma concentrations of the drug at 90 min exceeded the hypothetical Treg-specific therapeutic window determined in vitro (0.015-0.24 IU/ml), even at the lowest doses (0.040 × 106 and 0.045 × 106 IU/m2) administered. A rapid decrease in Treg frequency in the circulation was observed at 90 min and at day 1, which was dose dependent (mean decrease 11.6%, SE = 2.3%, range 10.0%-48.2%, n = 37), rebounding at day 2 and increasing to frequencies above baseline over 7 d. Teffs, natural killer cells, and eosinophils also responded, with their frequencies rapidly and dose-dependently decreased in the blood, then returning to, or exceeding, pretreatment levels. Furthermore, there was a dose-dependent down modulation of one of the two signalling subunits of the IL-2 receptor, the ß chain (CD122) (mean decrease = 58.0%, SE = 2.8%, range 9.8%-85.5%, n = 33), on Tregs and a reduction in their sensitivity to aldesleukin at 90 min and day 1 and 2 post-treatment. Due to blood volume requirements as well as ethical and practical considerations, the study was limited to adults and to analysis of peripheral blood only. CONCLUSIONS: The DILT1D trial results, most notably the early altered trafficking and desensitisation of Tregs induced by a single ultra-low dose of aldesleukin that resolves within 2-3 d, inform the design of the next trial to determine a repeat dosing regimen aimed at establishing a steady-state Treg frequency increase of 20%-50%, with the eventual goal of preventing T1D. TRIAL REGISTRATION: ISRCTN Registry ISRCTN27852285; ClinicalTrials.gov NCT01827735.


Assuntos
Diabetes Mellitus Tipo 1/prevenção & controle , Interleucina-2/análogos & derivados , Linfócitos T Reguladores/efeitos dos fármacos , Adolescente , Adulto , Biomarcadores , Quimiocinas/biossíntese , Relação Dose-Resposta a Droga , Eosinófilos/efeitos dos fármacos , Feminino , Humanos , Imunofenotipagem , Mediadores da Inflamação/metabolismo , Interleucina-2/efeitos adversos , Interleucina-2/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/farmacologia , Adulto Jovem
6.
Cancer Manag Res ; 7: 361-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26673410

RESUMO

BACKGROUND: Glucocorticoid receptor (GR) activity has been associated with chemotherapy resistance and poor outcomes in patients with triple negative breast cancer (TNBC). The aim of this study was to develop an immunohistochemistry (IHC) assay to assess GR expression in archival formalin-fixed, paraffin-embedded human invasive breast carcinoma samples. METHODS: An optimized GR assay protocol was developed using rabbit monoclonal antibody to GR clone D8H2. Precision and reproducibility of the GR IHC assay was determined by conducting multiple staining runs of four invasive breast carcinoma samples using replicate serial sections. Assay sensitivity was examined in 50 TNBC samples (>10 mm) obtained from a tumor bank, and 43 paired TNBC samples from a tissue microarray (TMA) (1.5 mm). GR positivity was assessed using a percent scoring approach with a ≥10% cutoff for nuclear staining of tumor cells at any intensity. Analysis of the paired TMA cores was performed by averaging the scores of the two cores for each case. RESULTS: Equivalent cellular patterns of GR reactivity were observed in all replicates from the multiple staining runs; coefficients of variation did not exceed 4.7% for average H-scores greater than 3.4, thus meeting the criteria for assay precision and reproducibility (coefficient of variation ≤20%). GR expression in TNBC single-tissue samples and TMA cores was characterized as mostly nuclear, with some concurrent cytoplasmic reactivity. Eighty-four percent of the 49 evaluable TNBC samples and 60% of the 42 evaluable paired TMA samples were positive for GR expression. CONCLUSION: A robust and reproducible GR IHC assay was successfully developed for use in invasive breast carcinoma tissues. Differences in GR expression between larger single tissues and smaller TMA cores illustrate the heterogeneity of the disease, as well as potential intra-tumoral heterogeneity. This assay is currently being utilized in clinical trials of mifepristone, a GR antagonist, in patients with TNBC.

7.
J Immunol ; 193(2): 587-96, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24943216

RESUMO

Immune-deficient mice, reconstituted with human stem cells, have been used to analyze human immune responses in vivo. Although they have been used to study immune responses to xenografts, allografts, and pathogens, there have not been models of autoimmune disease in which the mechanisms of the pathologic process can be analyzed. We have found that reconstituted "humanized" mice treated with anti-CTLA-4 Ab (ipilimumab) develop autoimmune disease characterized by hepatitis, adrenalitis, sialitis, anti-nuclear Abs, and weight loss. Induction of autoimmunity involved activation of T cells and cytokine production, and increased infiltration of APCs. When anti-CTLA-4 mAb-treated mice were cotreated with anti-CD3 mAb (teplizumab), hepatitis and anti-nuclear Abs were no longer seen and weight loss did not occur. The anti-CD3 blocked proliferation and activation of T cells, release of IFN-γ and TNF, macrophage infiltration, and release of IP-10 that was induced with anti-CTLA-4 mAb. We also found increased levels of T regulatory cells (CD25(+)CD127(-)) in the spleen and mesenteric lymph nodes in the mice treated with both Abs and greater constitutive phosphorylation of STAT5 in T regulatory cells in spleen cells compared with mice treated with anti-CTLA-4 mAb alone. We describe a model of human autoimmune disease in vivo. Humanized mice may be useful for understanding the mechanisms of biologics that are used in patients. Hepatitis, lymphadenopathy, and other inflammatory sequelae are adverse effects of ipilimumab treatment in humans, and this study may provide insights into this pathogenesis and the effects of immunologics on autoimmunity.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Doenças Autoimunes/terapia , Modelos Animais de Doenças , Transplante de Células-Tronco/métodos , Linfócitos T/imunologia , Glândulas Suprarrenais/efeitos dos fármacos , Glândulas Suprarrenais/imunologia , Glândulas Suprarrenais/metabolismo , Animais , Anticorpos Monoclonais/toxicidade , Doenças Autoimunes/induzido quimicamente , Doenças Autoimunes/imunologia , Citocinas/sangue , Citocinas/imunologia , Citocinas/metabolismo , Citometria de Fluxo , Humanos , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Ipilimumab , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Fosforilação/efeitos dos fármacos , Fosforilação/imunologia , Fator de Transcrição STAT5/imunologia , Fator de Transcrição STAT5/metabolismo , Análise de Sobrevida , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Transplante Heterólogo , Redução de Peso/efeitos dos fármacos , Redução de Peso/imunologia
8.
J Biol Chem ; 288(43): 30843-54, 2013 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-23986451

RESUMO

Molecularly engineered antibodies with fit-for-purpose properties will differentiate next generation antibody therapeutics from traditional IgG1 scaffolds. One requirement for engineering the most appropriate properties for a particular therapeutic area is an understanding of the intricacies of the target microenvironment in which the antibody is expected to function. Our group and others have demonstrated that proteases secreted by invasive tumors and pathological microorganisms are capable of cleaving human IgG1, the most commonly adopted isotype among monoclonal antibody therapeutics. Specific cleavage in the lower hinge of IgG1 results in a loss of Fc-mediated cell-killing functions without a concomitant loss of antigen binding capability or circulating antibody half-life. Proteolytic cleavage in the hinge region by tumor-associated or microbial proteases is postulated as a means of evading host immune responses, and antibodies engineered with potent cell-killing functions that are also resistant to hinge proteolysis are of interest. Mutation of the lower hinge region of an IgG1 resulted in protease resistance but also resulted in a profound loss of Fc-mediated cell-killing functions. In the present study, we demonstrate that specific mutations of the CH2 domain in conjunction with lower hinge mutations can restore and sometimes enhance cell-killing functions while still retaining protease resistance. By identifying mutations that can restore either complement- or Fcγ receptor-mediated functions on a protease-resistant scaffold, we were able to generate a novel protease-resistant platform with selective cell-killing functionality.


Assuntos
Anticorpos Monoclonais , Citotoxicidade Celular Dependente de Anticorpos , Sítios de Ligação de Anticorpos , Engenharia de Proteínas , Proteólise , Anticorpos Monoclonais/química , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Citotoxicidade Celular Dependente de Anticorpos/genética , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Sítios de Ligação de Anticorpos/genética , Sítios de Ligação de Anticorpos/imunologia , Linhagem Celular , Humanos , Imunoglobulina G , Receptores de IgG/genética , Receptores de IgG/imunologia
9.
Sci Transl Med ; 4(118): 118ra12, 2012 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-22277969

RESUMO

The development and optimization of immune therapies in patients has been hampered by the lack of preclinical models in which their effects on human immune cells can be studied. As a result, observations that have been made in preclinical studies have suggested mechanisms of drug action in murine models that have not been confirmed in clinical studies. Here, we used a humanized mouse reconstituted with human hematopoietic stem cells to study the mechanism of action of teplizumab, an Fc receptor nonbinding humanized monoclonal antibody to CD3 being tested in clinical trials for the treatment of patients with type 1 diabetes mellitus. In this model, human gut-tropic CCR6(+) T cells exited the circulation and secondary lymph organs and migrated to the small intestine. These cells then produced interleukin-10 (IL-10), a regulatory cytokine, in quantities that could be detected in the peripheral circulation. Blocking T cell migration to the small intestine with natalizumab, which prevents cellular adhesion by inhibiting α(4) integrin binding, abolished the treatment effects of teplizumab. Moreover, IL-10 expression by CD4(+)CD25(high)CCR6(+)FoxP3 cells returning to the peripheral circulation was increased in patients with type 1 diabetes treated with teplizumab. These findings demonstrate that humanized mice may be used to identify novel immunologic mechanisms that occur in patients treated with immunomodulators.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Trato Gastrointestinal/citologia , Trato Gastrointestinal/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Complexo CD3/imunologia , Movimento Celular/efeitos dos fármacos , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/imunologia , Fatores de Transcrição Forkhead/metabolismo , Trato Gastrointestinal/efeitos dos fármacos , Humanos , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Interleucina-10/metabolismo , Intestino Delgado/citologia , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/imunologia , Selectina L/metabolismo , Camundongos , Mucosa/citologia , Mucosa/efeitos dos fármacos , Mucosa/imunologia , Natalizumab , Análise de Sequência com Séries de Oligonucleotídeos , Receptores CCR6/metabolismo
10.
J Trauma ; 70(5): 1208-12, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21610434

RESUMO

BACKGROUND: Angiographic embolization (AE) has emerged as an important therapy for patients with nonvariceal upper gastrointestinal bleeding (UGIB). We hypothesized that discrete factors predictive of AE failure could be identified. METHODS: A retrospective review was performed for patients with nonvariceal UGIB who underwent AE from 1999 to 2009 at Penn State Milton S. Hershey Medical Center. AE clinical failure was defined as requirement for another intervention (surgery, endoscopic therapy, or another AE) for nonvariceal UGIB and/or death from bleeding after AE. Statistical analysis was performed using Fisher's exact test and Student's t test to explore the risk of AE failure. RESULTS: Of 48 total AE cases, 17 patients (35.4%) had clinically failed AE. Mortality rate was significantly higher in patients with AE clinical failure than in patients with AE clinical success (64.7% vs. 12.9%, p=0.001). Factors associated with AE clinical failure include anticoagulant use before admission (p=0.001), use of corticosteroids before admission (p=0.045), pre-AE vasopressor use (p=0.038), and embolization using either coils alone (p=0.05) or using coils with or without additional embolic materials (p=0.018). CONCLUSIONS: AE clinical failure portends poor prognosis. Caution should be exercised when considering AE, particularly AE using coils, in patients with a history of anticoagulant, corticosteroid, or vasopressor use.


Assuntos
Angiografia/métodos , Embolização Terapêutica/métodos , Hemorragia Gastrointestinal/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Seguimentos , Hemorragia Gastrointestinal/diagnóstico por imagem , Hemorragia Gastrointestinal/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Pennsylvania/epidemiologia , Prognóstico , Estudos Prospectivos , Fatores de Risco , Taxa de Sobrevida/tendências , Falha de Tratamento
11.
BMJ Case Rep ; 2009: bcr0220091617, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-21687037

RESUMO

A 40-year-old woman presented with a 10 day history of episodic vagueness, speech disturbance and blurred vision. Episodes typically occurred in the morning after awaking from sleep and resolved with food ingestion. She had no past medical history, did not drink alcohol and was not on any medication. Physical examination was normal with no evidence of endocrinopathy. After 10 h of fasting, she became hypoglycaemic with evidence of neuroglycopenia, which resolved with intravenous dextrose. Biochemical investigations revealed decreased glucose, insulin and C-peptide values with an increased excess insulin-like growth factor II: excess insulin-like growth factor I (IGF-II: IGF-I) ratio. Radiological examinations of the abdomen and pelvis revealed a heterogenous 10.5 cm left renal mass. The patient underwent a radical left nephrectomy. She had complete resolution of hypoglycaemic events. Histology revealed a renal sarcoma, grade 2/3. This is the first report in the literature involving a renal sarcoma causing non-islet cell tumour hypoglycaemia via excess IGF-II secretion.

13.
Mol Cancer Ther ; 6(5): 1478-82, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17483435

RESUMO

Li-Fraumeni syndrome is an autosomal dominant disorder that greatly increases the risk of developing multiple types of cancer. The majority of Li-Fraumeni syndrome families contain germ-line mutations in the p53 tumor suppressor gene. We describe treatment of a refractory, progressive Li-Fraumeni syndrome embryonal carcinoma with a p53 therapy (Advexin) targeted to the underlying molecular defect of this syndrome. p53 treatment resulted in complete and durable remission of the injected lesion by fluorodeoxyglucose-positron emission tomography scans with improvement of tumor-related symptoms. With respect to molecular markers, the patient's tumor had abnormal p53 and expressed coxsackie adenovirus receptors with a low HDM2 and bcl-2 profile conducive for adenoviral p53 activity. p53 treatment resulted in the induction of cell cycle arrest and apoptosis documented by p21 and cleaved caspase-3 detection. Increased adenoviral antibody titers after repeated therapy did not inhibit adenoviral p53 activity or result in pathologic sequelae. Relationships between these clinical, radiographic, and molecular markers may prove useful in guiding future application of p53 tumor suppressor therapy.


Assuntos
Genes p53 , Terapia Genética/métodos , Síndrome de Li-Fraumeni/terapia , Adulto , Apoptose , Caspase 3/metabolismo , Criança , Feminino , Fluordesoxiglucose F18/farmacologia , Humanos , Síndrome de Li-Fraumeni/genética , Masculino , Pessoa de Meia-Idade , Linhagem , Tomografia por Emissão de Pósitrons/métodos , Proteína Supressora de Tumor p53/metabolismo
15.
DNA Seq ; 15(1): 1-8, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15354348

RESUMO

Murine NGD5 is a gene identified from NG108-15 cells which is postulated to be involved in opioid receptor function. Here we report the cloning and characterization of a cDNA C20orf9-003 (ACI-1) encoding the human orthologue of the mouse NGD5. Analysis of the genomic structure revealed that C20orf9-003 (ACI-1) contains 13 exons and 12 introns, spanning 52.5kb of genomic DNA and is a variant of C20orf9. Chromosomal localization of human C20orf9-003 (ACI-1) assigned this gene to chromosome 20q13.12. Genes at this locus have been associated with the progression and possibly the development of various cancers. In addition several linkage studies support the possibility that one or more genes affecting obesity are located in 20q13. No function can be clearly assigned to C20orf9-003 (ACI-1), however, the protein has a cytoplasmic subcellular location and the secondary structure contains a Rossman fold like feature which is found in many nucleotide binding proteins.


Assuntos
Cromossomos Humanos Par 20/genética , Proteínas/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação , Neoplasias da Mama/genética , Mapeamento Cromossômico , Clonagem Molecular , DNA Complementar/genética , Éxons , Feminino , Marcadores Genéticos , Predisposição Genética para Doença , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Dados de Sequência Molecular , Peso Molecular , Obesidade/genética , Neoplasias da Próstata/genética , Proteínas/química , Proteínas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
16.
Br J Pharmacol ; 142(6): 921-32, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15210573

RESUMO

1 In an effort to identify endogenous, native mammalian urotensin-II (U-II) receptors (UT), a diverse range of human, primate and rodent cell lines (49 in total) were screened for the presence of detectable [125I]hU-II binding sites. 2 UT mRNA (Northern blot, PCR) and protein (immunocytochemistry) were evident in human skeletal muscle tissue and cells. 3 [(125)I]hU-II bound to a homogenous population of high-affinity, saturable (Kd 67.0+/-11.8 pm, Bmax 9687+/-843 sites cell(-1)) receptors in the skeletal muscle (rhabdomyosarcoma) cell line SJRH30. Radiolabel was characteristically slow to dissociate (< or =15% dissociation 90 min). A lower density of high-affinity U-II binding sites was also evident in the rhabdomyosarcoma cell line TE671 (1667+/-165 sites cell(-1), Kd 74+/-8 pm). 4 Consistent with the profile recorded in human recombinant UT-HEK293 cells, [125I]hU-II binding to SJRH30 cells was selectively displaced by both mammalian and fish U-II isopeptides (Kis 0.5+/-0.1-1.2+/-0.3 nm) and related analogues (hU-II[4-11]>[Cys(5,10)]Acm hU-II; Kis 0.4+/-0.1 and 864+/-193 nm, respectively). 5 U-II receptor activation was functionally coupled to phospholipase C-mediated [Ca2+]i mobilization (EC50 6.9+/-2.2 nm) in SJRH30 cells. 6 The present study is the first to identify the presence of 'endogenous' U-II receptors in SJRH30 and TE671 cells. SJRH30 cells, in particular, might prove to be of utility for (a) investigating the pharmacological properties of hU-II and related small molecule antagonists at native human UT and (b) delineating the role of this neuropeptide in the (patho)physiological regulation of mammalian neuromuscular function.


Assuntos
Receptores Acoplados a Proteínas G/metabolismo , Rabdomiossarcoma/metabolismo , Animais , Sítios de Ligação , Ligação Competitiva/efeitos dos fármacos , Northern Blotting , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Cálcio/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Feminino , Perfilação da Expressão Gênica , Células HeLa , Humanos , Hormônios Hipotalâmicos/farmacologia , Imuno-Histoquímica , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Radioisótopos do Iodo , Cinética , Masculino , Melaninas/farmacologia , Neuropeptídeo Y/farmacologia , Neurofisinas/farmacologia , Toxina Pertussis/farmacologia , Hormônios Hipofisários/farmacologia , Precursores de Proteínas/farmacologia , Ensaio Radioligante , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rabdomiossarcoma/patologia , Tapsigargina/farmacologia , Urotensinas/genética , Urotensinas/metabolismo , Urotensinas/farmacologia , Vasopressinas/farmacologia
17.
Int J Cancer ; 107(2): 209-14, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12949796

RESUMO

Expression of Fas ligand (FasL/CD95L) may help to maintain colon cancers in a state of immune privilege by inducing apoptosis of antitumor immune effector cells. Colon tumor-derived cell lines appear to be relatively insensitive to apoptosis mediated by their own or exogenous FasL in vitro, despite expression of cell surface Fas. In our present study, we sought to investigate if FasL upregulated in human colon cancers leads to any increase in apoptosis of the tumor cells in vivo. FasL and Fas receptor (APO-1/CD95) expression by tumor cells were detected immunohistochemically. Apoptotic tumor cell death was detected by immunohistochemistry for caspase-cleaved cytokeratin-18. FasL expression did not correlate with the extent of apoptosis of tumor cells. There was no significant local difference in the frequency of apoptosis of tumor cells between tumor nests that expressed FasL (mean = 2.4%) relative to those that did not (mean = 2.6%) (p = 0.625, n = 10; Wilcoxon signed rank). FasL expressed by the tumor cells appeared to be functional, since FasL expression in tumor nests correlated with diminished infiltration of tumor-infiltrating lymphocytes (TILs). TILs were detected using immunohistochemistry for CD45. Expression of FasL by tumor nests was associated with a mean 4-fold fewer TILs relative to FasL-negative nests (range 2.4-33-fold, n = 10, p < 0.003). Together, our results indicate that colon tumors are insensitive to FasL-mediated apoptosis in vivo.


Assuntos
Adenocarcinoma/patologia , Apoptose , Neoplasias do Colo/patologia , Glicoproteínas de Membrana/metabolismo , Adenocarcinoma/metabolismo , Neoplasias do Colo/metabolismo , Proteína Ligante Fas , Feminino , Humanos , Técnicas Imunoenzimáticas , Marcação In Situ das Extremidades Cortadas , Queratinas/metabolismo , Antígenos Comuns de Leucócito/imunologia , Ligantes , Depleção Linfocítica , Linfócitos do Interstício Tumoral/imunologia , Masculino , Regulação para Cima , Receptor fas/metabolismo
18.
Exp Cell Res ; 287(1): 39-46, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12799180

RESUMO

dUTP nucleotidohydrolase (dUTPase) catalyzes the hydrolysis of dUTP to dUMP and pyrophosphate and is the central regulator of cellular dUTP pools. Nuclear (DUT-N) and mitochondrial (DUT-M) isoforms of the protein have been identified in humans and arise from the same gene by the alternative use of 5' exons. Recently, it has been shown that these isoforms are aberrantly expressed in some cancers and overexpression of dUTPase in the nucleus is associated with resistance to chemotherapeutic agents that target thymidylate biosynthesis. In this study, we have examined the signals necessary for dUTPase isoform localization using green fluorescent protein fusion constructs. We report that the N-terminal 23 amino acids of DUT-N are required but not sufficient for complete nuclear localization. Within this region, we identified a small cluster of basic residues (K(14)R(15)R(17)) that resemble a classic monopartite nuclear localization signal (NLS). Mutation of these residues completely abolishes nuclear localization. In addition, phosphorylation of Ser11 near the putative NLS has no affect on DUT-N nuclear localization. Through deletion analysis we show improved sorting of DUT-N to the nucleus when most of the protein sequence is present. Therefore, we conclude that DUT-N may contain a complex NLS that is located throughout the entire protein.


Assuntos
Núcleo Celular/enzimologia , Resistencia a Medicamentos Antineoplásicos/genética , Células Eucarióticas/enzimologia , Neoplasias/enzimologia , Pirofosfatases/genética , Uridina Trifosfato/metabolismo , Células 3T3 , Processamento Alternativo/genética , Sequência de Aminoácidos/genética , Animais , Compartimento Celular/genética , Núcleo Celular/efeitos dos fármacos , Células Eucarióticas/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Células HT29 , Humanos , Camundongos , Mutação/genética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Isoformas de Proteínas/genética , Pirofosfatases/metabolismo , Proteínas Recombinantes de Fusão , Serina/genética , Transdução de Sinais/genética , Timidilato Sintase/metabolismo
19.
J Cell Biochem ; 88(3): 635-42, 2003 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-12532338

RESUMO

The Bin1/Amphiphysin II gene encodes at least seven alternately spliced adapter proteins that have been implicated in membrane dynamics and nuclear processes. Nuclear localized Bin1 polypeptides have tumor suppressor and proapoptotic activities, suggesting that Bin1 may suppress cancer in tissues where nuclear expression may occur. One question is the extent to which human tissues express nuclear Bin1 isoforms. A secondary issue has been the need for a specific antibody that can detect all the splice isoforms expressed by the human, mouse, and rat Bin1 genes. Using a novel mouse monoclonal antibody with these characteristics, we performed an immunohistochemical analysis of Bin1 expression in a panel of normal human tissues. We also compared the expression profile of Bin1 in normal or malignant tissues derived from human prostate, where Bin1 is a candidate tumor suppressor gene. In brain, a distinct nuclear staining pattern overlapped with a cytosolic staining pattern present in certain layers of the cerebral cortex and cerebellum. Bone marrow cells displayed mainly nuclear localization whereas peripheral lymphoid cells exhibited mainly cytosolic localization. In several epithelial tissues, nuclear or nucleocytosolic staining patterns were displayed by basal cells in skin, breast, or prostate, whereas cytosolic or plasma membrane-associated staining patterns were noted in gastrointestinal cells. Interestingly, a striking gradient of expression was observed in gastrointestinal epithelia, particularly in the large intestine, with the strongest staining displayed by cells destined to undergo apoptosis at the villus tip. In prostate, Bin1 staining was frequently absent in cases of primary prostate adenocarcinoma. This study used a novel reagent to document the extent of expression of nuclear Bin1 isoforms, which exhibit cancer suppression and proapoptotic activity in human cells.


Assuntos
Adenocarcinoma/metabolismo , Núcleo Celular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neoplasias da Próstata/metabolismo , Isoformas de Proteínas/metabolismo , Adenocarcinoma/patologia , Animais , Anticorpos Monoclonais/metabolismo , Linhagem Celular , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Neoplasias da Próstata/patologia , Distribuição Tecidual
20.
Cytokine ; 18(2): 61-71, 2002 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-12096920

RESUMO

We have recently reported the identification of four novel members of the interleukin-1 (IL-1) family which we designated as IL-1 homologue 1-4 (IL-1H1-4). These proteins exhibit significant sequence homology to other members of the IL-1 family. Of these homologues, only IL-1H4 (renamed IL-1F7b) was predicted to contain a propeptide domain and a caspase cleavage site. We now report that caspase-1 cleaves IL-1F7b at the predicted site to generate mature IL-1F7b. Caspase-4 was also able to process IL-1F7b, albeit inefficiently. Other caspases and Granzyme-B did not cleave IL-1F7b. Furthermore, adenovirus-mediated expression of IL-1F7b in HEK 293 cells led to in situ processing and secretion of mature IL-1F7b. In a screen to identify a potential receptor, both pro and mature IL-1F7b bound to the soluble IL-18 receptor alpha-Fc (IL-18Ralpha-Fc) but not to the soluble IL-1R-Fc or ST2R-Fc fusion proteins. Mature IL-1F7b bound to the IL-18Ralpha-Fc protein with higher affinity than the pro form, although the affinities for both proteins were significantly lower than that observed for IL-18. Consistent with this observation, only IL-18 and not IL-1F7b induced IFN-gamma production by KG1a cells. We also report that pro and mature IL-1F7b form homodimers with association constants of 4 microM and 5 nM, respectively, suggesting biological relevance to IL-1F7b processing. Finally, we have localized the expression of IL-1F7b protein in discrete cell populations including plasma cells and tumor cells. These data suggest that IL-1F7b may be involved in immune response, inflammatory diseases and/or cancer.


Assuntos
Caspase 1/metabolismo , Caspases/metabolismo , Interferon gama/biossíntese , Interleucina-18/metabolismo , Interleucina-1/genética , Interleucina-1/metabolismo , Receptores de Interleucina/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular , DNA Complementar , Humanos , Subunidade alfa de Receptor de Interleucina-18 , Receptores de Interleucina-18 , Proteínas Recombinantes de Fusão/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA