Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Transpl Infect Dis ; 22(1): e13238, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31880047

RESUMO

Adenovirus is an infrequent but challenging viral complication of transplantation that is rarely reported after autologous stem cell transplant. We present a case of disseminated adenovirus infection in a woman who received an autologous stem cell transplant for treatment of multiple sclerosis. After presenting with post-transplant episodic diarrhea and viremia, endoscopic biopsies and immunohistochemical staining confirmed the diagnosis of disseminated adenovirus infection. Her symptoms and viremia resolved after treatment with cidofovir. This case demonstrates that a high index of suspicion, a systematic clinical approach, and immunohistochemical tissue staining are necessary to diagnose disseminated adenovirus infection in an unexpected host.


Assuntos
Infecções por Adenovirus Humanos/sangue , Infecções por Adenovirus Humanos/etiologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Esclerose Múltipla/terapia , Viremia/etiologia , Adenoviridae , Infecções por Adenovirus Humanos/tratamento farmacológico , Antivirais/uso terapêutico , Cidofovir/uso terapêutico , Colo/virologia , Diarreia/virologia , Duodeno/virologia , Endoscopia , Feminino , Humanos , Pessoa de Meia-Idade , Esclerose Múltipla/complicações , Transplante Autólogo/efeitos adversos , Carga Viral/efeitos dos fármacos
2.
Biol Blood Marrow Transplant ; 24(7): 1386-1391, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29481870

RESUMO

Circulating plasma cells (CPCs) have been detected in patients with multiple myeloma (MM) at various stages of disease and associated with worse outcomes. Little data exist regarding the impact of CPCs at the time of autologous peripheral blood stem cell (PBSC) collection on outcomes, and the impact of maintenance therapy after autologous stem cell transplantation (ASCT) on prognosis in patients with CPC-containing collections. All patients with MM who underwent first ASCT at Fred Hutchinson Cancer Research Center from 2012 to 2015 and had evaluation for CPCs at the time of PBSC collection were included in our analysis. Seven-color flow cytometry was used to detect the presence of CPCs. Kaplan-Meier estimates were used to generate overall survival (OS) and progression-free survival (PFS) rates from the time of ASCT. A multivariate analysis, including receipt of maintenance therapy post-ASCT, high-risk cytogenetics, and international staging system (ISS) stage, was included in a Cox proportional hazards regression model for associations with OS and PFS. We identified 227 patients with MM who underwent ASCT; of these, 144 (63.4%) patients had routine assessment of CPCs at the time of PBSC collection. One hundred seventeen (81.3%) patients did not have CPCs and 27 (18.8%) did have CPCs. The presence of CPCs was highly associated with poorer PFS (P = .031 by log-rank analysis), but did not affect OS. The median PFS for those patients without CPCs was 39.4 months (95% confidence interval [CI], 31.1 to not reached), while the median PFS for those patients with CPCs was 16.5 months (95% CI, 13.7 to not reached). A subgroup analysis of patients achieving very good partial response (VGPR) or better at time of collection, showed the median PFS for patients without CPCs was 38.3 months (95% CI, 29 to not reached), as compared with those patients with CPCs, where it was only 16.5 months (95% CI, 12 months to not reached; P = .02). There was no statistically significant difference in PFS or OS among those patients achieving partial response at the time of collection. In a Cox proportional hazards model, adjusting for post-ASCT maintenance therapy, high-risk cytogenetics, and ISS stage at time of initial diagnosis, there was a 43% higher risk of progression or death among the patients with CPCs (P = .04). The presence of CPCs at the time of autologous PBSC collection is a negative prognostic factor for risk of early relapse or death despite the advent of novel agents and maintenance strategies. The impact of CPCs was most significant among patients achieving a VGPR or better at time of collection. The presence of CPCs denotes a unique group of high-risk MM patients for whom alternative treatment strategies are needed to overcome resistance to current standard therapies.


Assuntos
Transplante de Células-Tronco Hematopoéticas/métodos , Mieloma Múltiplo/terapia , Células-Tronco de Sangue Periférico/metabolismo , Condicionamento Pré-Transplante/métodos , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/patologia , Prognóstico
3.
Cancer Cell ; 3(5): 471-82, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12781365

RESUMO

The clathrin-associated protein, Huntingtin Interacting Protein 1 (HIP1), is overexpressed in multiple human epithelial tumors. Here, we report that HIP1 is a novel oncoprotein that transforms cells. HIP1-transformed cells, in contrast to RasV12-transformed cells, have dysregulation of multiple receptors involved in clathrin trafficking. Examples include upregulation of the epidermal growth factor receptor (EGFR) and the transferrin receptor. Furthermore, accumulation of transferrin and EGF in the HIP1-transformed cells was increased, and breast tumors that had EGFR expressed also had HIP1 upregulated. Thus, HIP1 overexpression promotes tumor formation and is associated with a general alteration in receptor trafficking. HIP1 is the first endocytic protein to be directly implicated in tumor formation.


Assuntos
Proteínas de Transporte/genética , Proteínas de Ligação a DNA , Animais , Apoptose , Western Blotting , Ciclo Celular , Divisão Celular , Linhagem Celular Transformada , Transformação Celular Neoplásica , DNA Complementar/metabolismo , Endocitose , Receptores ErbB/metabolismo , Citometria de Fluxo , Humanos , Camundongos , Camundongos Nus , Microscopia Confocal , Modelos Biológicos , Células NIH 3T3 , Fosforilação , Regiões Promotoras Genéticas , Transporte Proteico , Fatores de Tempo , Transferrina/metabolismo
4.
Acta Cytol ; 56(5): 527-32, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23075894

RESUMO

OBJECTIVE: To evaluate the utility of the lymphatic endothelial marker D2-40, along with calretinin, CK5/6, desmin and MOC-31, in differentiating mesothelioma and adenocarcinoma in pleural effusion cytology. STUDY DESIGN: Forty-five pleural effusion cases representing confirmed reactive effusions (13), mesotheliomas (11) and metastatic adenocarcinomas (21) were immunostained with antibodies against D2-40, calretinin, CK5/6, desmin and MOC-31. RESULTS: D2-40 showed membranous staining in 82% of mesotheliomas and 77% of reactive effusions. Calretinin and CK5/6 were positive in 100 and 64% of mesotheliomas, and 92 and 31% of reactive effusions, respectively. All adenocarcinomas showed lack of staining with these markers. Desmin was negative in all malignant cases and positive in 85% of reactive effusions. All adenocarcinomas were positive for MOC-31 and negative for the remaining markers. CONCLUSION: Calretinin was the most sensitive in detecting mesothelial differentiation, followed by D2-40. Although useful, D2-40 necessitated cautious interpretation due to occasional focal/weak positivity, particularly in limited cellularity samples. The muscle marker desmin was useful in differentiating benign from malignant effusions but not in distinguishing mesotheliomas from adenocarcinomas. MOC-31 was both highly sensitive and specific for detecting adenocarcinoma and was useful as part of a panel of stains in differentiating cells of mesothelial origin from adenocarcinoma.


Assuntos
Adenocarcinoma/diagnóstico , Biomarcadores Tumorais/análise , Mesotelioma/diagnóstico , Derrame Pleural Maligno/diagnóstico , Adenocarcinoma/metabolismo , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais/análise , Anticorpos Monoclonais Murinos/análise , Calbindina 2 , Citodiagnóstico/métodos , Desmina/análise , Diagnóstico Diferencial , Humanos , Imuno-Histoquímica , Queratina-5/análise , Queratina-6/análise , Masculino , Mesotelioma/metabolismo , Pessoa de Meia-Idade , Derrame Pleural Maligno/metabolismo , Reprodutibilidade dos Testes , Proteína G de Ligação ao Cálcio S100/análise , Sensibilidade e Especificidade
5.
Clin Lymphoma Myeloma Leuk ; 20(9): 602-609, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32457024

RESUMO

BACKGROUND: Bortezomib has been incorporated into thalidomide and dexamethasone provided with cisplatin, doxorubicin, cyclophosphamide, and etoposide (PACE) as an intensive regimen before autologous stem-cell transplantation for multiple myeloma (MM). We examined MM patients at our center who received chemomobilization with a regimen that substituted carfilzomib and lenalidomide for bortezomib and thalidomide (KRD-PACE). PATIENTS AND METHODS: This was a retrospective study of 27 MM patients who received KRD-PACE for chemomobilization. Our analysis included patients who had circulating plasma cells (CPCs) by flow cytometry, ≥ 10% bone marrow plasma cells (BMPC), a monoclonal protein ≥ 1 g/dL, or an involved serum free light chain ≥ 10 mg/dL. RESULTS: The most common indication for KRD-PACE was BMPC ≥ 10% in 16 patients (60%), followed by CPCs in 11 (41%). The median (range) age was 61 (35-69) years, and the median (range) BMPC before treatment was 10% (5%-47%). The overall response rate was 43%, and a median (range) of 20.24 (8.08-69.88) × 106 CD34+ cells/kg were collected. CPC clearance rate was 50%, and the median reduction in BMPC was 75%. Two patients had sinus bradycardia and 5 (19%) had neutropenic fever. CONCLUSION: KRD-PACE is an effective therapy to mobilize peripheral blood stem cells in MM patients with residual disease burden. This regimen was successful at clearing CPCs and reducing BMPC burden, with an overall response rate of 43%. Despite theoretical concern regarding the combination of 3 cardiotoxic agents, we observed a low frequency of cardiac issues.


Assuntos
Transplante de Células-Tronco Hematopoéticas/métodos , Mieloma Múltiplo/terapia , Neoplasia Residual/terapia , Condicionamento Pré-Transplante/métodos , Transplante Autólogo/métodos , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/complicações , Estudos Retrospectivos
6.
Cancer Res ; 67(8): 3609-15, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17440072

RESUMO

Huntingtin interacting protein 1 (HIP1) is a multidomain oncoprotein whose expression correlates with increased epidermal growth factor receptor (EGFR) levels in certain tumors. For example, HIP1-transformed fibroblasts and HIP1-positive breast cancers have elevated EGFR protein levels. The combined association of HIP1 with huntingtin, the protein that is mutated in Huntington's disease, and the known overexpression of EGFR in glial brain tumors prompted us to explore HIP1 expression in a group of patients with different types of brain cancer. We report here that HIP1 is overexpressed with high frequency in brain cancers and that this overexpression correlates with EGFR and platelet-derived growth factor beta receptor expression. Furthermore, serum samples from patients with brain cancer contained anti-HIP1 antibodies more frequently than age-matched brain cancer-free controls. Finally, we report that HIP1 physically associates with EGFR and that this association is independent of the lipid, clathrin, and actin interacting domains of HIP1. These findings suggest that HIP1 may up-regulate or maintain EGFR overexpression in primary brain tumors by directly interacting with the receptor. This novel HIP1-EGFR interaction may work with or independent of HIP1 modulation of EGFR degradation via clathrin-mediated membrane trafficking pathways. Further investigation of HIP1 function in brain cancer biology and validation of its use as a prognostic or predictive brain tumor marker are now warranted.


Assuntos
Neoplasias Encefálicas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Receptores ErbB/metabolismo , Actinas/metabolismo , Anticorpos Antineoplásicos/sangue , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/metabolismo , Encéfalo/metabolismo , Neoplasias Encefálicas/imunologia , Clatrina/metabolismo , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/imunologia , Receptores ErbB/biossíntese , Humanos , Imunoprecipitação , Metabolismo dos Lipídeos , Ligação Proteica
7.
Cancer Res ; 67(18): 8923-31, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17875735

RESUMO

Huntingtin interacting protein 1 (HIP1) is an inositol lipid, clathrin, and actin binding protein that is overexpressed in a variety of epithelial malignancies. Here, we report for the first time that HIP1 is elevated in non-Hodgkin's and Hodgkin's lymphomas and that patients with lymphoid malignancies frequently had anti-HIP1 antibodies in their serum. Moreover, p53-deficient mice with B-cell lymphomas were 13 times more likely to have anti-HIP1 antibodies in their serum than control mice. Furthermore, transgenic overexpression of HIP1 was associated with the development of lymphoid neoplasms. The HIP1 protein was induced by activation of the nuclear factor-kappaB pathway, which is frequently activated in lymphoid malignancies. These data identify HIP1 as a new marker of lymphoid malignancies that contributes to the transformation of lymphoid cells in vivo.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Linfoma/metabolismo , Animais , Anticorpos Antineoplásicos/sangue , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Modelos Animais de Doenças , Doença de Hodgkin/genética , Doença de Hodgkin/metabolismo , Humanos , Linfoma/genética , Linfoma/patologia , Linfoma de Célula do Manto/genética , Linfoma de Célula do Manto/metabolismo , Linfoma de Célula do Manto/patologia , Linfoma não Hodgkin/genética , Linfoma não Hodgkin/imunologia , Linfoma não Hodgkin/metabolismo , Linfoma não Hodgkin/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/metabolismo , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
8.
Commun Biol ; 1: 174, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30374464

RESUMO

While the incidence of esophageal adenocarcinoma (EAC) has risen drastically in Western countries over the last 40 years, a similar trend has not been observed for EAC in China. Here, we analyzed mutational spectrum, copy number alterations, and structural variants from whole-genome sequencing of 10 Chinese EAC tumor samples and their matched normal samples, and compared them to previously reported EAC tumor specimens from Western countries. The mutational burden in Chinese EAC was significantly lower than that found in EAC from Western countries. The hallmark A>C mutational signature observed at high frequency in EAC from Western countries, which has been linked to acid reflux, is completely absent in Chinese samples. Furthermore, none of the Chinese samples showed evidence of chromothripsis and genome doubling that are often found in EAC from Western countries. In summary, Chinese EAC tumor samples had distinct genomic profiles and signatures, suggesting that EAC in Chinese individuals may arise from a different etiological pathway.

9.
J Clin Invest ; 110(3): 351-60, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12163454

RESUMO

Huntingtin-interacting protein 1 (HIP1) is a cofactor in clathrin-mediated vesicle trafficking. It was first implicated in cancer biology as part of a chromosomal translocation in leukemia. Here we report that HIP1 is expressed in prostate and colon tumor cells, but not in corresponding benign epithelia. The relationship between HIP1 expression in primary prostate cancer and clinical outcomes was evaluated with tissue microarrays. HIP1 expression was significantly associated with prostate cancer progression and metastasis. Conversely, primary prostate cancers lacking HIP1 expression consistently showed no progression after radical prostatectomy. In addition, the expression of HIP1 was elevated in prostate tumors from the transgenic mouse model of prostate cancer (TRAMP). At the molecular level, expression of a dominant negative mutant of HIP1 led to caspase-9-dependent apoptosis, suggesting that HIP1 is a cellular survival factor. Thus, HIP1 may play a role in tumorigenesis by allowing the survival of precancerous or cancerous cells. HIP1 might accomplish this via regulation of clathrin-mediated trafficking, a fundamental cellular pathway that has not previously been associated with tumorigenesis. HIP1 represents a putative prognostic factor for prostate cancer and a potential therapy target in prostate as well as colon cancers.


Assuntos
Proteínas de Transporte/genética , Neoplasias do Colo/genética , Proteínas de Ligação a DNA , Expressão Gênica , Neoplasias da Próstata/genética , Animais , Apoptose , Biomarcadores Tumorais/genética , Proteínas de Transporte/biossíntese , Caspase 9 , Caspases/metabolismo , Sobrevivência Celular , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Humanos , Masculino , Camundongos , Neoplasias da Próstata/patologia , Neoplasias da Próstata/fisiopatologia , Células Tumorais Cultivadas
10.
Mol Cell Biol ; 24(10): 4329-40, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15121852

RESUMO

In mice and humans, there are two known members of the Huntingtin interacting protein 1 (HIP1) family, HIP1 and HIP1-related (HIP1r). Based on structural and functional data, these proteins participate in the clathrin trafficking network. The inactivation of Hip1 in mice leads to spinal, hematopoietic, and testicular defects. To investigate the biological function of HIP1r, we generated a Hip1r mutant allele in mice. Hip1r homozygous mutant mice are viable and fertile without obvious morphological abnormalities. In addition, embryonic fibroblasts derived from these mice do not have gross abnormalities in survival, proliferation, or clathrin trafficking pathways. Altogether, this demonstrates that HIP1r is not necessary for normal development of the embryo or for normal adulthood and suggests that HIP1 or other functionally related members of the clathrin trafficking network can compensate for HIP1r absence. To test the latter, we generated mice deficient in both HIP1 and HIP1r. These mice have accelerated development of abnormalities seen in Hip1 -deficient mice, including kypholordosis and growth defects. The severity of the Hip1r/Hip1 double-knockout phenotype compared to the Hip1 knockout indicates that HIP1r partially compensates for HIP1 function in the absence of HIP1 expression, providing strong evidence that HIP1 and HIP1r have overlapping roles in vivo.


Assuntos
Proteínas de Transporte/genética , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Nanismo/genética , Proteínas dos Microfilamentos/genética , Coluna Vertebral/anormalidades , Proteínas Adaptadoras de Transdução de Sinal , Animais , Sequência de Bases , Mapeamento Cromossômico , DNA/genética , Nanismo/patologia , Endocitose/genética , Receptores ErbB/metabolismo , Feminino , Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Gravidez , Distribuição Tecidual , Proteínas de Transporte Vesicular
11.
Leuk Res ; 55: 23-32, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28119225

RESUMO

Dexamethasone (dex) induces apoptosis in multiple myeloma (MM) cells and is a frontline treatment for this disease. However resistance to dex remains a major challenge and novel treatment approaches are needed. We hypothesized that dex utilizes translational pathways to promote apoptosis in MM and that specific targeting of these pathways could overcome dex-resistance. Global unbiased profiling of mRNA translational profiles in MM cells treated with or without dex revealed that dex significantly repressed eIF2 signaling, an important pathway for regulating ternary complex formation and protein synthesis. We demonstrate that dex induces the phosphorylation of eIF2α resulting in the translational upregulation of ATF4, a known eIF2 regulated mRNA. Pharmacologic induction of eIF2α phosphorylation via activation of the heme-regulated eIF2α kinase (HRI) induced apoptosis in MM cell lines and in primary MM cells from patients with dex-resistant disease. In addition, co-culture with marrow stroma failed to protect MM cells from apoptosis induced by targeting the eIF2 pathway. Combination therapy with rapamycin, an mTOR inhibitor, and BTdCPU, an activator of HRI, demonstrated additive effects on apoptosis in dex-resistant cells. Thus, specific activation of the eIF2α kinase HRI is a novel therapeutic target in MM that can augment current treatment strategies.


Assuntos
Terapia de Alvo Molecular/métodos , Mieloma Múltiplo/tratamento farmacológico , eIF-2 Quinase/metabolismo , Apoptose/efeitos dos fármacos , Dexametasona/farmacologia , Resistencia a Medicamentos Antineoplásicos , Humanos , Fosforilação , Biossíntese de Proteínas , Células Tumorais Cultivadas , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/efeitos dos fármacos
12.
Amyloid ; 23(4): 254-259, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27879147

RESUMO

INTRODUCTION: High-dose melphalan and autologous stem cell transplantation (HDM/SCT) is an effective treatment modality for immunoglobulin light-chain (AL) amyloidosis; however, its application remains restricted to patients with good performance status and limited organ involvement. In recent years, the paradigm for AL amyloidosis has changed with the introduction of novel agents such as immunomodulatory drugs (IMiDs) and proteasome inhibitors (PIs). We hypothesized that use of novel agent induction regimens has improved outcomes for patients with AL amyloidosis undergoing HDM/SCT at our center. METHODS: All patients with AL amyloidosis, age ≥18 years who underwent HDM/SCT between 2001 and 2014 at the Fred Hutchinson Cancer Research Center and University of Washington Medical Center were included in this study. Any regimen administered within 6 months prior to HDM/SCT including an IMiD or a PI was considered a novel induction regimen. Use of induction regimen was evaluated in a Cox proportional hazard model for association with progression-free survival (PFS) and overall survival (OS). RESULTS: Forty-five patients with AL amyloidosis underwent HDM/SCT. The median age was 57.2 years (range 39-74.4), 15 (33.3%) were women. The median number of organs involved was 2 (range 1-5), with 20 patients having only 1 (44.4%), 10 patients having 2 (22.2%), and 15 patients (33.3%) having ≥ 3 organs involved. Novel agent induction regimens were used prior to HDM/SCT in 21 patients (46.7%); these comprised PI in 13/21 (57.1%), IMiD alone in 6/21 (28.6%), PI and cyclophosphamide (CyBorD) in 3/21 (14.3%), and IMiD and PI in 3/21 (14.3%). Use of a novel agent induction regimen was associated with improved, but not OS. The 3-year PFS for patients who received a novel agent induction was 79%, while for those who did not was 53% (hazard ratio [HR] = 0.317, p = 0.048). The 3-year OS for patients who received novel agent induction regimens was 95%, while for those who did not was 71% (HR = 0.454, p = 0.247). DISCUSSION: Our data suggest that use of a novel agent induction regimen including an IMiD or PI prior to HDM/SCT for patients with AL amyloidosis could improve outcomes, with improvement in PFS. Although these results are limited by sample size and lack of randomization, these results support possible further investigation of novel agent induction regimens in the context of a prospective clinical trial.


Assuntos
Amiloidose/diagnóstico , Antineoplásicos Alquilantes/uso terapêutico , Transplante de Células-Tronco Hematopoéticas , Cadeias Leves de Imunoglobulina/sangue , Quimioterapia de Indução/métodos , Melfalan/uso terapêutico , Adulto , Idoso , Amiloidose/sangue , Amiloidose/mortalidade , Amiloidose/patologia , Ciclofosfamida/uso terapêutico , Intervalo Livre de Doença , Quimioterapia Combinada , Feminino , Expressão Gênica , Humanos , Cadeias Leves de Imunoglobulina/genética , Fatores Imunológicos/uso terapêutico , Masculino , Pessoa de Meia-Idade , Prognóstico , Inibidores de Proteassoma/uso terapêutico , Transplante Autólogo
13.
Clin Cancer Res ; 22(2): 319-27, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26378033

RESUMO

PURPOSE: Cutaneous sclerosis occurs in 20% of patients with chronic graft-versus-host disease (GVHD) and can compromise mobility and quality of life. EXPERIMENTAL DESIGN: We conducted a prospective, multicenter, randomized, two-arm phase II crossover trial of imatinib (200 mg daily) or rituximab (375 mg/m(2) i.v. weekly × 4 doses, repeatable after 3 months) for treatment of cutaneous sclerosis diagnosed within 18 months (NCT01309997). The primary endpoint was significant clinical response (SCR) at 6 months, defined as quantitative improvement in skin sclerosis or joint range of motion. Treatment success was defined as SCR at 6 months without crossover, recurrent malignancy or death. Secondary endpoints included changes of B-cell profiles in blood (BAFF levels and cellular subsets), patient-reported outcomes, and histopathology between responders and nonresponders with each therapy. RESULTS: SCR was observed in 9 of 35 [26%; 95% confidence interval (CI); 13%-43%] participants randomized to imatinib and 10 of 37 (27%; 95% CI, 14%-44%) randomized to rituximab. Six (17%; 95% CI, 7%-34%) patients in the imatinib arm and 5 (14%; 95% CI, 5%-29%) in the rituximab arm had treatment success. Higher percentages of activated B cells (CD27(+)) were seen at enrollment in rituximab-treated patients who had treatment success (P = 0.01), but not in imatinib-treated patients. CONCLUSIONS: These results support the need for more effective therapies for cutaneous sclerosis and suggest that activated B cells define a subgroup of patients with cutaneous sclerosis who are more likely to respond to rituximab.


Assuntos
Doença Enxerto-Hospedeiro/tratamento farmacológico , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Mesilato de Imatinib/uso terapêutico , Rituximab/uso terapêutico , Esclerose/tratamento farmacológico , Dermatopatias/tratamento farmacológico , Adulto , Idoso , Linfócitos B/efeitos dos fármacos , Estudos Cross-Over , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Adulto Jovem
14.
Trends Mol Med ; 10(4): 194-9, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15059611

RESUMO

During recent years, alterations in proteins of the endocytic pathway have been associated with tumors. Disrupted regulation of the endocytic pathway is a relatively unstudied mechanism of tumorigenesis, which can concomitantly disrupt several different signaling pathways to affect growth, differentiation and survival. Several endocytic proteins have been identified, either as part of tumor-associated translocations or to have the ability to transform cells. Here, we summarize the information known about huntingtin interacting protein 1 (HIP1), an endocytic protein with transforming properties that is involved in a cancer-causing translocation and which is overexpressed in a variety of human cancers. We describe the known normal functions of HIP1 in endocytosis and receptor trafficking, the evidence for its role as an oncoprotein and how HIP1 might be altered to promote tumorigenesis.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Regulação Neoplásica da Expressão Gênica , Neoplasias/metabolismo , Alelos , Transporte Biológico , Sobrevivência Celular , Transformação Celular Neoplásica , Proteínas de Ligação a DNA/metabolismo , Endocitose , Humanos , Modelos Biológicos , Estrutura Terciária de Proteína , Transporte Proteico
15.
Hum Mol Genet ; 16(11): 1279-92, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17452370

RESUMO

The members of the huntingtin-interacting protein-1 (HIP1) family, HIP1 and HIP1-related (HIP1r), are multi-domain proteins that interact with inositol lipids, clathrin and actin. HIP1 is over-expressed in a variety of cancers and both HIP1 and HIP1r prolong the half-life of multiple growth factor receptors. To better understand the physiological importance of the HIP1 family in vivo, we have analyzed a large cohort of double Hip1/Hip1r knockout (DKO) mice. All DKO mice were dwarfed, afflicted with severe vertebral defects and died in early adulthood. These phenotypes were not observed during early adulthood in the single Hip1 or Hip1r knockouts, indicating that HIP1 and HIP1r compensate for one another. Despite the ability of HIP1 and HIP1r to modulate growth factor receptor levels when over-expressed, studies herein using DKO fibroblasts indicate that the HIP1 family is not necessary for endocytosis but is necessary for the maintenance of diverse adult tissues in vivo. To test if human HIP1 can function similar to mouse HIP1, transgenic mice with 'ubiquitous' expression of the human HIP1 cDNA were generated and crossed with DKO mice. Strikingly, the compound human HIP1 transgenic DKO mice were completely free from dwarfism and spinal defects. This successful rescue demonstrates that the human HIP1 protein shares some interchangeable functions with both HIP1 and HIP1r in vivo. In addition, we conclude that the degenerative phenotypes seen in the DKO mice are due mainly to HIP1 and HIP1r protein deficiency rather than altered expression of neighboring genes or disrupted intronic elements.


Assuntos
Proteínas de Ligação a DNA/genética , Lordose/genética , Redução de Peso/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/fisiologia , Genes Letais , Humanos , Íntrons , Cifose/genética , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos , Fenótipo
16.
J Biol Chem ; 279(14): 14294-306, 2004 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-14732715

RESUMO

Huntingtin-interacting protein 1-related (HIP1r) is the only known mammalian relative of huntingtin-interacting protein 1 (HIP1), a protein that transforms fibroblasts via undefined mechanisms. Here we demonstrate that both HIP1r and HIP1 bind inositol lipids via their epsin N-terminal homology (ENTH) domains. In contrast to other ENTH domain-containing proteins, lipid binding is preferential to the 3-phosphate-containing inositol lipids, phosphatidylinositol 3,4-bisphosphate and phosphatidylinositol 3,5-bisphosphate. Furthermore, the HIP1r ENTH domain, like that of HIP1, is necessary for lipid binding, and expression of an ENTH domain-deletion mutant, HIP1r/deltaE, induces apoptosis. Consistent with the ability of HIP1r and HIP1 to affect cell survival, full-length HIP1 and HIP1r stabilize pools of growth factor receptors by prolonging their half-life following ligand-induced endocytosis. Although HIP1r and HIP1 display only a partially overlapping pattern of protein interactions, these data suggest that both proteins share a functional homology by binding 3-phosphorylated inositol lipids and stabilizing receptor tyrosine kinases in a fashion that may contribute to their ability to alter cell growth and survival.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA , Endocitose/fisiologia , Neuropeptídeos/genética , Fosfatidilinositóis/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas de Transporte Vesicular , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Animais , Anticorpos Monoclonais , Sítios de Ligação , Células COS , Proteínas de Transporte/química , Proteínas de Transporte/imunologia , Morte Celular/fisiologia , Clatrina/metabolismo , Receptores ErbB/metabolismo , Células HeLa , Humanos , Inositol/metabolismo , Cinética , Proteínas dos Microfilamentos , Mutagênese , Estrutura Terciária de Proteína , Transporte Proteico/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa