Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Thorac Oncol ; 12(2): 293-301, 2017 02.
Article in English | MEDLINE | ID: mdl-27743888

ABSTRACT

INTRODUCTION: In the randomized trial of standard- versus high-dose chemoradiotherapy for locally advanced (LA) NSCLC (Radiation Therapy Oncology Group 0617), overall survival (OS) was worse in the high-dose arm. Although heart dose was suggested as a contributing factor, actionable parameters have not been established. We present an analysis of clinical and dosimetric parameters affecting OS in this patient population, focusing on heart dose. METHODS: Clinical data were collected on 416 patients with LA NSCLC treated at a single institution, with a subset of 333 available treatment plans recontoured using Radiation Therapy Oncology Group 0617 normal tissue guidelines. Toxicity and dosimetry data were analyzed for 322 patients; multivariate analysis was performed on 251 patients. Dosimetric parameters of radiation to tumor and organs at risk were analyzed with clinical data pertaining to OS, disease-free survival, and toxicity. RESULTS: Patients were treated with radiation therapy to prescribed doses of 50.0 to 84.9 Gy (median 66.0 Gy). Median follow-up was 14.5 months. Median OS was 16.8 months. The 1- and 2-year OS rates were 61.4% and 38.8%, respectively. On multivariate analysis, factors independently associated with worse OS were increasing heart V50 (volume receiving ≥50 Gy), heart volume, lung V5 (proportion of the lung structure [excluding the target volume]) receiving at least 5 Gy), bilateral mediastinal lymph node involvement, and lack of concurrent chemotherapy. When stratified by heart V50 less than 25% versus 25% or greater, the 1-year OS rates were 70.2% versus 46.8% and the 2-year OS rates were 45.9% versus 26.7% (p < 0.0001). Median heart V50 was significantly higher (20.8% versus 13.9%, p < 0.0001) for patients with cardiac toxicity with a Common Terminology Criteria for Adverse Events grade of 1 or higher. CONCLUSIONS: Heart dose is associated with OS and cardiac toxicity for patients with LA NSCLC treated with chemoradiotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Squamous Cell/mortality , Chemoradiotherapy/mortality , Heart/physiopathology , Lung Neoplasms/mortality , Neoplasm Recurrence, Local/mortality , Organs at Risk/physiopathology , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Female , Follow-Up Studies , Heart/radiation effects , Humans , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/therapy , Neoplasm Staging , Organs at Risk/radiation effects , Prognosis , Radiometry , Radiotherapy Dosage , Radiotherapy, Intensity-Modulated/methods , Retrospective Studies , Survival Rate
2.
Cardiovasc Intervent Radiol ; 39(6): 855-64, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26721589

ABSTRACT

PURPOSE: The purpose of our study is to determine if there is a relationship between dose deposition measured by PET/MRI and individual lesion response to yttrium-90 ((90)Y) microsphere radioembolization. MATERIALS AND METHODS: 26 patients undergoing lobar treatment with (90)Y microspheres underwent PET/MRI within 66 h of treatment and had follow-up imaging available. Adequate visualization of tumor was available in 24 patients, and contours were drawn on simultaneously acquired PET/MRI data. Dose volume histograms (DVHs) were extracted from dose maps, which were generated using a voxelized dose kernel. Similar contours to capture dimensional and volumetric change of tumors were drawn on follow-up imaging. Response was analyzed using both RECIST and volumetric RECIST (vRECIST) criteria. RESULTS: A total of 8 hepatocellular carcinoma (HCC), 4 neuroendocrine tumor (NET), 9 colorectal metastases (CRC) patients, and 3 patients with other metastatic disease met inclusion criteria. Average dose was useful in predicting response between responders and non-responders for all lesion types and for CRC lesions alone using both response criteria (p < 0.05). D70 (minimum dose to 70 % of volume) was also useful in predicting response when using vRECIST. No significant trend was seen in the other tumor types. For CRC lesions, an average dose of 29.8 Gy offered 76.9 % sensitivity and 75.9 % specificity for response. CONCLUSIONS: PET/MRI of (90)Y microsphere distribution showed significantly higher DVH values for responders than non-responders in patients with CRC. DVH analysis of (90)Y microsphere distribution following treatment may be an important predictor of response and could be used to guide future adaptive therapy trials.


Subject(s)
Brachytherapy/methods , Liver Neoplasms/radiotherapy , Magnetic Resonance Imaging , Microspheres , Positron-Emission Tomography , Yttrium Radioisotopes/therapeutic use , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Multimodal Imaging , Treatment Outcome
3.
Int J Radiat Oncol Biol Phys ; 92(5): 1000-1007, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26025775

ABSTRACT

PURPOSE: Acute severe lymphopenia (ASL) frequently develops during radiation therapy (RT) and concurrent temozolomide (TMZ) for high-grade glioma (HGG) and is associated with decreased survival. The current study was designed to identify potential predictors of ASL, with a focus on actionable RT-specific dosimetric parameters. METHODS AND MATERIALS: From January 2007 to December 2012, 183 patients with HGG were treated with RT+TMZ and had available data including total lymphocyte count (TLC) and radiation dose-volume histogram parameters. ASL was defined as TLC of <500/µL within the first 3 months from the start of RT. Stepwise logistic regression analysis was used to determine the most important predictors of ASL. RESULTS: Fifty-three patients (29%) developed ASL. Patients with ASL had significantly worse overall survival than those without (median: 12.5 vs 20.2 months, respectively, P<.001). Stepwise logistic regression analysis identified female sex (odds ratio [OR]: 5.30; 95% confidence interval [CI]: 2.46-11.41), older age (OR: 1.05; 95% CI: 1.02-1.09), lower baseline TLC (OR: 0.92; 95% CI: 0.87-0.98), and higher brain volume receiving 25 Gy (V25Gy) (OR: 1.03; 95% CI: 1.003-1.05) as the most significant predictors for ASL. Brain V25Gy <56% appeared to be the optimal threshold (OR: 2.36; 95% CI: 1.11-5.01), with an ASL rate of 38% versus 20% above and below this threshold, respectively (P=.006). CONCLUSIONS: Female sex, older age, lower baseline TLC, and higher brain V25Gy are significant predictors of ASL during RT+TMZ therapy for HGG. Maintaining the V25Gy of brain below 56% may reduce the risk of ASL.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , Brain Neoplasms/radiotherapy , Cranial Irradiation/adverse effects , Dacarbazine/analogs & derivatives , Glioma/radiotherapy , Lymphopenia/etiology , Acute Disease , Adult , Age Factors , Aged , Aged, 80 and over , Brain/radiation effects , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Chemotherapy, Adjuvant , Cranial Irradiation/methods , Dacarbazine/adverse effects , Female , Glioma/drug therapy , Glioma/mortality , Glioma/pathology , Humans , Lymphocyte Count , Lymphopenia/mortality , Male , Middle Aged , Odds Ratio , Radiotherapy Dosage , Regression Analysis , Sex Factors , Temozolomide , Young Adult
4.
Int J Radiat Oncol Biol Phys ; 91(2): 268-76, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25636755

ABSTRACT

PURPOSE: Anaplastic gliomas represent a heterogeneous group of primary high-grade brain tumors, and the optimal postoperative treatment remains controversial. In this report, we present our institutional data on the clinical outcomes of radiation therapy (RT) plus temozolomide (RT + TMZ) for anaplastic gliomas, stratified by histology and 1p/19q codeletion. METHODS AND MATERIALS: A single-institution retrospective review was conducted of patients with supratentorial anaplastic oligodendroglioma (AO), mixed anaplastic oligoastrocytoma (AOA), and anaplastic astrocytoma (AA). After surgery, RT was delivered at a median total dose of 60 Gy (range, 31.6-63 Gy) in daily fractions. All patients received standard concurrent TMZ, with or without adjuvant TMZ. Histological/molecular subtypes were defined as codeleted AO/AOA, non-codeleted AO/AOA, and AA. RESULTS: From 2000 to 2012, 111 cases met study criteria and were evaluable. Codeleted AO/AOA had superior overall survival (OS) to non-codeleted AO/AOA (91% vs 68% at 5 years, respectively, P=.02), whereas progression-free survival (PFS) was not significantly different (70% vs 46% at 5 years, respectively, P=.10). AA had inferior OS to non-codeleted AO/AOA (37% vs 68% at 5 years, respectively, P=.007) and inferior PFS (27% vs 46%, respectively, P=.03). On multivariate analysis, age, performance status, and histological or molecular subtype were independent predictors for both PFS and OS. Compared to historical controls, RT + TMZ provided comparable OS to RT with procarbazine, lomustine, and vincristine (RT + PCV) for codeleted AO/AOA, superior OS to RT alone for non-codeleted AO/AOA, and similar OS to RT alone for AA. CONCLUSIONS: RT + TMZ may be a promising treatment for both codeleted and non-codeleted AO/AOA, but its role for AA remains unclear.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/therapy , Chromosome Deletion , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 1/genetics , Glioma/genetics , Glioma/therapy , Adult , Aged , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Chemoradiotherapy , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Female , Gene Deletion , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Glioma/mortality , Glioma/pathology , Humans , Male , Middle Aged , Missouri/epidemiology , Prevalence , Radiotherapy Dosage , Retrospective Studies , Risk Factors , Survival Rate , Temozolomide , Treatment Outcome , Young Adult
5.
PET Clin ; 10(1): 27-44, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25455878

ABSTRACT

In this review, we review the literature on the use of PET in radiation treatment planning, with an emphasis on describing our institutional methodology (where applicable). This discussion is intended to provide other radiation oncologists with methodological details on the use of PET imaging for treatment planning in radiation oncology, or other oncologists with an introduction to the use of PET in planning radiation therapy.


Subject(s)
Fluorodeoxyglucose F18 , Neoplasms/diagnostic imaging , Neoplasms/radiotherapy , Positron-Emission Tomography/methods , Radiometry/methods , Radiotherapy Planning, Computer-Assisted/methods , Radiotherapy, Image-Guided/methods , Humans , Radiopharmaceuticals , Radiotherapy Dosage
6.
Int J Radiat Oncol Biol Phys ; 88(4): 801-5, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24462385

ABSTRACT

PURPOSE: To assess the impact on local tumor control of intraoperative ultrasonographic plaque visualization and selective application of transpupillary thermotherapy (TTT) in the treatment of posterior uveal melanoma with iodine-125 (I-125) episcleral plaque brachytherapy (EPB). METHODS AND MATERIALS: Retrospective analysis of 526 patients treated with I-125 EPB for posterior uveal melanoma. Clinical features, dosimetric parameters, TTT treatments, and local tumor control outcomes were recorded. Statistical analysis was performed using Cox proportional hazards and Kaplan-Meier life table method. RESULTS: The study included 270 men (51%) and 256 women (49%), with a median age of 63 years (mean, 62 years; range, 16-91 years). Median dose to the tumor apex was 94.4 Gy (mean, 97.8; range, 43.9-183.9) and to the tumor base was 257.9 Gy (mean, 275.6; range, 124.2-729.8). Plaque tilt >1 mm away from the sclera at plaque removal was detected in 142 cases (27%). Supplemental TTT was performed in 72 patients (13.7%). One or 2 TTT sessions were required in 71 TTT cases (98.6%). After a median follow-up of 45.9 months (mean, 53.4 months; range, 6-175 months), local tumor recurrence was detected in 19 patients (3.6%). Local tumor recurrence was associated with lower dose to the tumor base (P=.02). CONCLUSIONS: Ultrasound-guided plaque localization of I-125 EPB is associated with excellent local tumor control. Detection of plaque tilt by ultrasonography at plaque removal allows supplemental TTT to be used in patients at potentially higher risk for local recurrence while sparing the majority of patients who are at low risk. Most patients require only 1 or 2 TTT sessions.


Subject(s)
Brachytherapy/methods , Hyperthermia, Induced/methods , Iodine Radioisotopes/therapeutic use , Melanoma/radiotherapy , Ultrasonography, Interventional/methods , Uveal Neoplasms/radiotherapy , Adolescent , Adult , Aged , Aged, 80 and over , Combined Modality Therapy/methods , Female , Humans , Kaplan-Meier Estimate , Male , Melanoma/diagnostic imaging , Melanoma/pathology , Melanoma/prevention & control , Middle Aged , Neoplasm Recurrence, Local/prevention & control , Proportional Hazards Models , Radiotherapy Dosage , Retrospective Studies , Tumor Burden , Uveal Neoplasms/diagnostic imaging , Uveal Neoplasms/pathology , Uveal Neoplasms/prevention & control , Young Adult
7.
Int J Radiat Oncol Biol Phys ; 82(3): e563-72, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22197230

ABSTRACT

PURPOSE: The development of drug-resistant phenotypes has been a major obstacle to cisplatin use in non-small-cell lung cancer. We aimed to identify some of the molecular mechanisms that underlie cisplatin resistance using microarray expression analysis. METHODS AND MATERIALS: H460 cells were treated with cisplatin. The differences between cisplatin-resistant lung cancer cells and parental H460 cells were studied using Western blot, MTS, and clonogenic assays, in vivo tumor implantation, and microarray analysis. The cisplatin-R cells were treated with human recombinant insulin-like growth factor (IGF) binding protein-3 and siRNA targeting IGF-1 receptor. RESULTS: Cisplatin-R cells illustrated greater expression of the markers CD133 and aldehyde dehydrogenase, more rapid in vivo tumor growth, more resistance to cisplatin- and etoposide-induced apoptosis, and greater survival after treatment with cisplatin or radiation than the parental H460 cells. Also, cisplatin-R demonstrated decreased expression of insulin-like growth factor binding protein-3 and increased activation of IGF-1 receptor signaling compared with parental H460 cells in the presence of IGF-1. Human recombinant IGF binding protein-3 reversed cisplatin resistance in cisplatin-R cells and targeting of IGF-1 receptor using siRNA resulted in sensitization of cisplatin-R-cells to cisplatin and radiation. CONCLUSIONS: The IGF-1 signaling pathway contributes to cisplatin-R to cisplatin and radiation. Thus, this pathway represents a potential target for improved lung cancer response to treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Cisplatin/pharmacology , Drug Resistance, Neoplasm/physiology , Insulin-Like Growth Factor Binding Protein 3/physiology , Insulin-Like Growth Factor I/physiology , Lung Neoplasms/therapy , Radiation-Sensitizing Agents/pharmacology , AC133 Antigen , Aldehyde Dehydrogenase/metabolism , Animals , Antigens, CD/metabolism , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cell Survival/radiation effects , Chemoradiotherapy/methods , Drug Resistance, Neoplasm/drug effects , Female , Glycoproteins/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Microarray Analysis , Peptides/metabolism , RNA, Small Interfering/pharmacology , Radiation Tolerance/drug effects , Radiation Tolerance/physiology , Radiation Tolerance/radiation effects , Receptor, IGF Type 1/antagonists & inhibitors , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Signal Transduction/radiation effects , Xenograft Model Antitumor Assays/methods
8.
Am J Cancer Res ; 1(1): 43-61, 2011.
Article in English | MEDLINE | ID: mdl-21969218

ABSTRACT

Genotoxic agents have long targeted apoptotic cell death as a primary means of treating cancer. However, the presence of cellular defects in many cancers has contributed to an acquired resistance to apoptotic cell death, lowering the effectiveness of chemo- and radiotherapies. The mechanisms by which cells achieve this resistance to treatment are still being investigated, but an alternative approach is the study of cell death pathways that are mechanistically distinct from apoptosis. These pathways, including autophagy and necrosis, have arisen as attractive targets for cancer therapy. This review will discuss apoptosis, autophagy, and necrosis in the context of tumorigenesis and drug resistance, as well as provide an up-to-date preclinical and clinical review of inhibitors targeting these cell death pathways for multiple cancer types. The goal of these studies is to identify molecular targets that will enhance the efficacy and specificity of current cancer therapies.

9.
J Thorac Oncol ; 6(9): 1542-52, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21642866

ABSTRACT

INTRODUCTION: A major focus of cancer research is to identify compounds that sensitize resistant cancer cells to radiation treatment. Lung cancer cells, in particular, have high rates of radioresistance that lead to treatment failure. We have previously shown that the autophagy induced in the context of decreased apoptosis confers radiosensitivity to prostate and lung cancer cells. Zinc supplementation has antiapoptotic effects in cell culture. In addition, the accumulation of zinc in response to oxidative stress has been associated with increased autophagy in astrocyte and breast cancer cells. METHODS: In this study, we hypothesized that the zinc ionophore PCI-5002 radiosensitizes lung cancer cells by inducing autophagic cell death. To test this hypothesis, we used a combination of in vitro and in vivo approaches, including clonogenic assays to test for radiosensitivity, biochemical analyses of apoptosis and autophagy, and a xenograft mouse model of tumor growth. RESULTS: We found that PCI-5002 reduced clonogenic survival in treated cells compared with untreated cells (0.03% versus 0.1% surviving fraction, p < 0.001). The increased radiosensitive fraction of PCI-5002-treated cells was accompanied by increased autophagy. PCI-5002 treatment also reduced caspase-3 cleavage. In an irradiated xenograft mouse model, the tumor growth of irradiated, PCI-5002-treated mice was slower than untreated, irradiated mice (25 days versus 22 days to reach a 1.0 cm tumor size). CONCLUSIONS: PCI-5002 treatment sensitizes lung cancer cells to radiation, both in vitro and in vivo. This data suggest that PCI-5002 could potentially treat radioresistant/locally advanced lung cancer by amplifying the effects of radiotherapy.


Subject(s)
Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Coordination Complexes/pharmacology , Ionophores/pharmacology , Radiation-Sensitizing Agents/pharmacology , Zinc/metabolism , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Autophagy/radiation effects , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cells, Cultured , Cesium Radioisotopes , Cytochromes c/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/radiation effects , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Gamma Rays , Humans , Immunoblotting , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Mice , Mice, Knockout , Mice, Nude , Xenograft Model Antitumor Assays , bcl-2 Homologous Antagonist-Killer Protein/physiology , bcl-2-Associated X Protein/physiology
10.
J Thorac Oncol ; 6(4): 699-706, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21325979

ABSTRACT

INTRODUCTION: Persistent STAT3 activation contributes to lung carcinogenesis. Survivin, one of STAT3-regulated genes, is antiapoptotic and confers cancer radioresistance. METHODS: We tested whether TG101209, a small-molecule inhibitor of JAK2 (a STAT3-activating tyrosine kinase), affected survivin expression and sensitized lung cancer to radiation. We investigated whether inhibition of JAK2 signaling with TG101209 can be used to reduce survivin expression and enhance radiosensitivity of lung cancer cells in vitro and tumor growth delay in vivo. JAK2 downstream signaling, including PI3-K/Akt and Ras/MAPK/ERK pathways, was also explored. RESULTS: TG101209 inhibited STAT3 activation and survivin expression and sensitized HCC2429 (dose enhancement ratio = 1.34, p = 0.002) and H460 (dose enhancement ratio = 1.09, p = 0.006) cells to radiation in clonogenic assays. Radiation promoted phospho-Akt and phospho-ERK in H460 cells, while their levels were unchanged in HCC2429. After treatment with TG101209, phospho-ERK protein levels were reduced in both HCC2429 and H460 cells. HCC2429 cells transfected with KRAS-12V mutant were more resistant to radiation- and TG101209-induced apoptosis than wild-type control cells. In vivo, addition of TG101209 to radiation in lung xenografts produced a significant tumor growth delay (>10 days) compared with radiation alone and was well tolerated. Immunohistochemistry staining of tumor sections showed that TG101209 increased apoptosis and decreased cell proliferation and vascular density, suggesting that TG101209 also has antiangiogenic effects. CONCLUSIONS: TG101209 enhanced the effects of radiation in lung cancer in vitro and in vivo. This study suggests the potential utility of selecting lung cancer patients according to KRAS mutation status for future clinical trials testing combination of TG101209 and radiotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Janus Kinase 2/antagonists & inhibitors , Lung Neoplasms/radiotherapy , Pyrimidines/pharmacology , Radiation-Sensitizing Agents/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology , Animals , Blotting, Western , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cesium Radioisotopes , Female , Humans , Inhibitor of Apoptosis Proteins/metabolism , Janus Kinase 2/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Mice, Nude , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , Survivin , Tumor Cells, Cultured , Tumor Stem Cell Assay , X-Rays
11.
Dis Model Mech ; 4(2): 255-67, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20959633

ABSTRACT

Tuberous sclerosis complex (TSC) is an autosomal dominant disease caused by mutations in either the TSC1 (encodes hamartin) or TSC2 (encodes tuberin) genes. Patients with TSC have hamartomas in various organs throughout the whole body, most notably in the brain, skin, eye, heart, kidney and lung. To study the development of hamartomas, we generated a zebrafish model of TSC featuring a nonsense mutation (vu242) in the tsc2 gene. This tsc2(vu242) allele encodes a truncated Tuberin protein lacking the GAP domain, which is required for inhibition of Rheb and of the TOR kinase within TORC1. We show that tsc2(vu242) is a recessive larval-lethal mutation that causes increased cell size in the brain and liver. Greatly elevated TORC1 signaling is observed in tsc2(vu242/vu242) homozygous zebrafish, and is moderately increased in tsc2(vu242/+) heterozygotes. Forebrain neurons are poorly organized in tsc2(vu242/vu242) homozygous mutants, which have extensive gray and white matter disorganization and ectopically positioned cells. Genetic mosaic analyses demonstrate that tsc2 limits TORC1 signaling in a cell-autonomous manner. However, in chimeric animals, tsc2(vu242/vu242) mutant cells also mislocalize wild-type host cells in the forebrain in a non-cell-autonomous manner. These results demonstrate a highly conserved role of tsc2 in zebrafish and establish a new animal model for studies of TSC. The finding of a non-cell-autonomous function of mutant cells might help explain the formation of brain hamartomas and cortical malformations in human TSC.


Subject(s)
Codon, Nonsense/genetics , Tuberous Sclerosis/pathology , Tumor Suppressor Proteins/genetics , Zebrafish/genetics , Animals , Brain/drug effects , Brain/embryology , Brain/pathology , Cell Size/drug effects , Disease Models, Animal , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Gene Expression Regulation, Developmental/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Mutant Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neurons/transplantation , Sirolimus/pharmacology , Transcription Factors/metabolism , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism , Zebrafish/embryology
12.
Development ; 137(8): 1327-37, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20332150

ABSTRACT

Gastrulation movements form the germ layers and shape them into the vertebrate body. Gastrulation entails a variety of cell behaviors, including directed cell migration and cell delamination, which are also involved in other physiological and pathological processes, such as cancer metastasis. Decreased Prostaglandin E(2) (PGE(2)) synthesis due to interference with the Cyclooxygenase (Cox) and Prostaglandin E synthase (Ptges) enzymes halts gastrulation and limits cancer cell invasiveness, but how PGE(2) regulates cell motility remains unclear. Here we show that PGE(2)-deficient zebrafish embryos, impaired in the epiboly, internalization, convergence and extension gastrulation movements, exhibit markedly increased cell-cell adhesion, which contributes to defective cell movements in the gastrula. Our analyses reveal that PGE(2) promotes cell protrusive activity and limits cell adhesion by modulating E-cadherin transcript and protein, in part through stabilization of the Snai1a (also known as Snail1) transcriptional repressor, an evolutionarily conserved regulator of cell delamination and directed migration. We delineate a pathway whereby PGE(2) potentiates interaction between the receptor-coupled G protein betagamma subunits and Gsk3beta to inhibit proteasomal degradation of Snai1a. However, overexpression of beta-catenin cannot stabilize Snai1a in PGE(2)-deficient gastrulae. Thus, the Gsk3beta-mediated and beta-catenin-independent inhibition of cell adhesion by Prostaglandins provides an additional mechanism for the functional interactions between the PGE(2) and Wnt signaling pathways during development and disease. We propose that ubiquitously expressed PGE(2) synthesizing enzymes, by promoting the stability of Snai1a, enable the precise and rapid regulation of cell adhesion that is required for the dynamic cell behaviors that drive various gastrulation movements.


Subject(s)
Gastrula/physiology , Prostaglandins G/physiology , Zebrafish/genetics , Animals , Cell Adhesion/physiology , DNA Primers , Dinoprostone/deficiency , Dinoprostone/metabolism , Embryo, Nonmammalian/physiology , Gastrulation/physiology , In Situ Hybridization , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Snail Family Transcription Factors , Transcription Factors/genetics , Zebrafish/embryology , Zebrafish/physiology , Zebrafish Proteins/genetics
13.
Nature ; 419(6905): 395-9, 2002 Sep 26.
Article in English | MEDLINE | ID: mdl-12353035

ABSTRACT

Apoptosis is a form of programmed cell death that is controlled by aspartate-specific cysteine proteases called caspases. In the immune system, apoptosis counters the proliferation of lymphocytes to achieve a homeostatic balance, which allows potent responses to pathogens but avoids autoimmunity. The CD95 (Fas, Apo-1) receptor triggers lymphocyte apoptosis by recruiting Fas-associated death domain (FADD), caspase-8 and caspase-10 proteins into a death-inducing signalling complex. Heterozygous mutations in CD95, CD95 ligand or caspase-10 underlie most cases of autoimmune lymphoproliferative syndrome (ALPS), a human disorder that is characterized by defective lymphocyte apoptosis, lymphadenopathy, splenomegaly and autoimmunity. Mutations in caspase-8 have not been described in ALPS, and homozygous caspase-8 deficiency causes embryonic lethality in mice. Here we describe a human kindred with an inherited genetic deficiency of caspase-8. Homozygous individuals manifest defective lymphocyte apoptosis and homeostasis but, unlike individuals affected with ALPS, also have defects in their activation of T lymphocytes, B lymphocytes and natural killer cells, which leads to immunodeficiency. Thus, caspase-8 deficiency in humans is compatible with normal development and shows that caspase-8 has a postnatal role in immune activation of naive lymphocytes.


Subject(s)
Caspases/genetics , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Lymphocyte Activation , Lymphocytes/immunology , Lymphocytes/pathology , Mutation/genetics , Apoptosis , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD28 Antigens/immunology , CD3 Complex/immunology , Calcium/metabolism , Caspase 8 , Caspase 9 , Cell Division , Cytokines/analysis , Female , Homozygote , Humans , Immunologic Deficiency Syndromes/enzymology , Immunologic Deficiency Syndromes/pathology , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Male , Pedigree , Phenotype , T-Lymphocytes/immunology , T-Lymphocytes/pathology
14.
J Virol ; 76(10): 5082-93, 2002 May.
Article in English | MEDLINE | ID: mdl-11967324

ABSTRACT

An important unresolved issue of AIDS pathogenesis is the mechanism of human immunodeficiency virus (HIV)-induced CD4(+) T-lymphocyte destruction. We show here that HIV type 1 (HIV-1) exerts a profound cytopathic effect upon peripheral blood CD4(+) T lymphocytes that resembles necrosis rather than apoptosis. Necrotic cytopathology was found with both laboratory-adapted strains and primary isolates of HIV-1. We carefully investigated the role of env, which has been previously implicated in HIV cytopathicity. HIV-1 stocks with equivalent infectivity were prepared from constructs with either an intact or mutated env coding region and pseudotyped with the glycoprotein of vesicular stomatitis virus (VSV-G) so that the HIV envelope was not rate-limiting for infection. Infected Jurkat T cells died whether or not env was intact; however, the expression of env accelerated death significantly. The accelerated death was blocked by protease inhibitors, indicating that it was due to reinfection by newly produced virus in env(+) cultures. Accordingly, we found no disparity in kinetics in CD4(lo) Jurkat cells. In highly infected peripheral blood T cells, profound necrosis occurred equivalently with both env(+) and env(-) stocks of HIV-1. We also found that HIV-1 cytopathicity was undiminished by the absence of nef. However, viral stocks made by complementation or packaging of HIV-1 genomes with the natural protein-coding sequences replaced by the green fluorescent protein were highly infectious but not cytopathic. Thus, env can accelerate cell death chiefly as an entry function, but one or more viral functions other than env or nef is essential for necrosis of CD4(+) T cells induced by HIV-1.


Subject(s)
Apoptosis , CD4-Positive T-Lymphocytes/virology , Gene Products, env/physiology , HIV-1/physiology , Membrane Glycoproteins , Necrosis , CD4-Positive T-Lymphocytes/pathology , Cytopathogenic Effect, Viral , Gene Products, env/genetics , Gene Products, nef/physiology , HIV-1/genetics , Humans , Jurkat Cells , Mutation , Recombination, Genetic , Vesicular stomatitis Indiana virus/genetics , Viral Envelope Proteins/genetics , nef Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...