Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Front Immunol ; 12: 801281, 2021.
Article in English | MEDLINE | ID: mdl-34975916

ABSTRACT

BK virus (BKV)-hemorrhagic cystitis (HC) is a well-known and rarely fatal complication of hematopoietic stem cell transplantation (HSCT). Treatment for BKV-HC is limited, but virus-specific T-cells (VST) represent a promising therapeutic option feasible for use posttransplant. We report on the case of a 16-year-old male with dedicator of cytokinesis 8 (DOCK8) deficiency who underwent haploidentical HSCT complicated by severe BKV-HC, catastrophic renal hemorrhage, and VST-associated cytokine release syndrome (CRS). Gross hematuria refractory to multiple interventions began with initiation of posttransplant cyclophosphamide (PT/Cy). Complete left renal arterial embolization (day +43) was ultimately indicated to control intractable renal hemorrhage. Subsequent infusion of anti-BK VSTs was complicated by CRS and progressive multiorgan failure, with postmortem analysis confirming diagnosis of hepatic sinusoidal obstruction syndrome (SOS). This case illustrates opportunities for improvement in the management of severe BKV-HC posttransplant while highlighting rare and potentially life-threatening complications of BKV-HC and VST therapy.


Subject(s)
Adoptive Transfer/adverse effects , BK Virus/pathogenicity , Cystitis/therapy , Cytokine Release Syndrome/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Hemorrhage/therapy , Polyomavirus Infections/therapy , T-Lymphocytes/transplantation , Tumor Virus Infections/therapy , Adolescent , BK Virus/immunology , Cystitis/diagnosis , Cystitis/immunology , Cystitis/virology , Cytokine Release Syndrome/diagnosis , Fatal Outcome , Hemorrhage/diagnosis , Hemorrhage/immunology , Hemorrhage/virology , Humans , Male , Multiple Organ Failure/etiology , Polyomavirus Infections/diagnosis , Polyomavirus Infections/immunology , Polyomavirus Infections/virology , Severity of Illness Index , T-Lymphocytes/immunology , T-Lymphocytes/virology , Treatment Outcome , Tumor Virus Infections/diagnosis , Tumor Virus Infections/immunology , Tumor Virus Infections/virology
2.
J Cancer ; 11(14): 4106-4114, 2020.
Article in English | MEDLINE | ID: mdl-32368293

ABSTRACT

MicroRNAs (miRNAs) are small, non-coding RNA molecules with multiple roles in many biological processes. Few studies have shown the molecular characteristics in younger prostate cancer (PCa) patients. In this study, we performed miRNA profiling in young PCa (EO-PCa) cases compared with PCa arising in older men (LO-PCa). Experimental Design: Formalin-fixed, paraffin embedded tissue was used. miRNA was extracted for PCR array and NanoString methods. Relative miRNAs expression levels were obtained by comparing young vs older men, and young PCa tumor samples vs normal epithelium. Results: miRNA profiling showed a different expression pattern in PCa arising in younger men, and young PCa tumoral and its normal counterpart. Nine miRNAs (hsa-miR-140-5p, hsa-miR-146a, hsa-miR-29b, hsa-miR-9, hsa-miR-124-3p, hsa-let-7f-5p, hsa-miR-184, hsa-miR-373, hsa-miR-146b-5p) showed differences in the expression compared to LO-PCa. Fourteen miRNAs were significantly up-regulated (miR-1973, miR-663a, miR-575, miR-93-5p, miR-630, miR-600, miR-494, miR-150-5p, miR-137, miR-25-3p, miR-375, miR-489, miR-888-5p, miR-142-3p), while 9 were found down-regulated (miR-21-5p, miR-363-3p, miR-205-5p, miR-548ai, miR-3195, 145-5p, miR-143-3p, miR-222-3p, miR-221-3p) comparing young PCa tumoral tissue compared to normal counterpart. The higher expression of miR-600 and miR-137 were associated with high Gleason score, extraprostatic extension and lymphatic invasion. Conclusion: These results suggest that PCa in younger patients has a different expression profile compared to normal tissue and PCa arising in older man. Differentially expressed miRNAs provide insights of molecular mechanisms involve in this PCa subtype.

3.
JCO Clin Cancer Inform ; 4: 367-382, 2020 04.
Article in English | MEDLINE | ID: mdl-32330067

ABSTRACT

PURPOSE: To develop an artificial intelligence (AI)-based model for identifying patients with lymph node (LN) metastasis based on digital evaluation of primary tumors and train the model using cystectomy specimens available from The Cancer Genome Atlas (TCGA) Project; patients from our institution were included for validation of the leave-out test cohort. METHODS: In all, 307 patients were identified for inclusion in the study (TCGA, n = 294; in-house, n = 13). Deep learning models were trained from image patches at 2.5×, 5×, 10×, and 20× magnifications, and spatially resolved prediction maps were combined with microenvironment (lymphocyte infiltration) features to derive a final patient-level AI score (probability of LN metastasis). Training and validation included 219 patients (training, n = 146; validation, n = 73); 89 patients (TCGA, n = 75; in-house, n = 13) were reserved as an independent testing set. Multivariable logistic regression models for predicting LN status based on clinicopathologic features alone and a combined model with AI score were fit to training and validation sets. RESULTS: Several patients were determined to have positive LN metastasis in TCGA (n = 105; 35.7%) and in-house (n = 3; 23.1%) cohorts. A clinicopathologic model that considered using factors such as age, T stage, and lymphovascular invasion demonstrated an area under the curve (AUC) of 0.755 (95% CI, 0.680 to 0.831) in the training and validation cohorts compared with the cross validation of the AI score (likelihood of positive LNs), which achieved an AUC of 0.866 (95% CI, 0.812 to 0.920; P = .021). Performance in the test cohort was similar, with a clinicopathologic model AUC of 0.678 (95% CI, 0.554 to 0.802) and an AI score of 0.784 (95% CI, 0.702 to 0.896; P = .21). In addition, the AI score remained significant after adjusting for clinicopathologic variables (P = 1.08 × 10-9), and the combined model significantly outperformed clinicopathologic features alone in the test cohort with an AUC of 0.807 (95% CI, 0.702 to 0.912; P = .047). CONCLUSION: Patients who are at higher risk of having positive LNs during cystectomy can be identified on primary tumor samples using novel AI-based methodologies applied to digital hematoxylin and eosin-stained slides.


Subject(s)
Urinary Bladder Neoplasms , Artificial Intelligence , Humans , Lymph Nodes , Lymphatic Metastasis , Retrospective Studies , Tumor Microenvironment
4.
J Cancer ; 4(5): 350-7, 2013.
Article in English | MEDLINE | ID: mdl-23781281

ABSTRACT

MicroRNAs are small non-coding RNA molecules that have been shown to regulate the expression of genes linked to cancer. The relevance of microRNAs in the development, progression and prognosis of prostate cancer is not fully understood. It is also possible that these specific molecules may assist in the recognition of aggressive tumors and the development of new molecular targets. Our study investigated the importance of several microRNAs in cases of prostate cancer from 37 patients that were manually microdissected to obtain pure populations of tumor cells, normal epithelium and adjacent stroma. MicroRNA was extracted for PCR array profiling. Differentially expressed miRNAs for each case were used to compare tumor vs. normal epithelium and tumor-adjacent stroma samples. Loss of 18 miRNAs (e.g.miR-34c, miR-29b, miR-212 and miR-10b) and upregulation of miR-143 and miR-146b were significantly found in all the tumors in comparison with normal epithelium and/or stroma (p≤ 0.001). A different signature was found in the high grade tumors (Gleason score ≥ 8) when compared with tumors Gleason score 6. Upregulation of miR-122, miR-335, miR-184, miR-193, miR-34, miR-138, miR-373, miR-9, miR-198, miR-144 and miR-215 and downregulation of miR-96, miR-222, miR-148, miR-92, miR-27, miR-125, miR-126, miR-27 were found in the high grade tumors. MicroRNA profiling in prostate cancer appears to have unique expression patterns in comparison with normal tissue. These differential expressed miRNAs may provide novel diagnostic and prognostic tools that will assist in the recognition of prostate cancers with aggressive behavior.

5.
J Urol ; 189(2): 430-5, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22982371

ABSTRACT

PURPOSE: The condition hereditary leiomyomatosis and renal cell carcinoma is characterized by cutaneous leiomyomas, uterine fibroids and aggressive papillary renal cell carcinoma. A number of our patients with hereditary leiomyomatosis and renal cell carcinoma had atypical adrenal nodules, which were further evaluated to determine whether these nodules were associated with hereditary leiomyomatosis and renal cell carcinoma. MATERIALS AND METHODS: Patients with hereditary leiomyomatosis and renal cell carcinoma underwent a comprehensive clinical and genetic evaluation. We reviewed the clinical presentation, anatomical and functional imaging, endocrine evaluation, pathological examination and germline mutation testing results. RESULTS: Of 255 patients with hereditary leiomyomatosis and renal cell carcinoma 20 (7.8%) had primary adrenal lesions, including 4 with bilateral adrenal lesions and 4 with multiple nodules. Two patients had adrenocorticotropic hormone independent hypercortisolism. A total of 27 adrenal lesions were evaluated. The imaging characteristics of 5 of these lesions (18.5%) were not consistent with adenoma by noncontrast computerized tomography criteria. Positron emission tomography was positive in 7 of 10 cases (70%). A total of 12 nodules were surgically resected from 10 adrenal glands. Pathological examination revealed macronodular adrenal hyperplasia in all specimens. CONCLUSIONS: Unilateral and bilateral adrenal nodular hyperplasia was detected in a subset of patients with hereditary leiomyomatosis and renal cell carcinoma. A functional endocrine evaluation is recommended when an adrenal lesion is discovered. Imaging frequently reveals lesions that are not typical of adenomas and positron emission tomography may be positive. To date no patient has had adrenal malignancy, and active surveillance of hereditary leiomyomatosis and renal cell carcinoma adrenal nodules appears justified.


Subject(s)
Adrenal Gland Diseases/etiology , Adrenal Gland Diseases/pathology , Adrenal Glands/pathology , Kidney Neoplasms/complications , Leiomyomatosis/complications , Neoplastic Syndromes, Hereditary/complications , Adult , Aged , Female , Humans , Hyperplasia/etiology , Male , Middle Aged , Skin Neoplasms , Uterine Neoplasms , Young Adult
6.
J Nucl Med ; 53(4): 538-45, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22343504

ABSTRACT

UNLABELLED: This work characterizes the uptake of (11)C-acetate in prostate cancer (PCa), benign prostate hyperplasia, and normal prostate tissue in comparison with multiparametric MRI, whole-mount histopathology, and clinical markers to evaluate the potential utility of (11)C-acetate for delineating intraprostatic tumors in a population of patients with localized PCa. METHODS: Thirty-nine men with presumed localized PCa underwent dynamic-static abdominal-pelvic (11)C-acetate PET/CT for 30 min and 3-T multiparametric MRI before prostatectomy. PET/CT images were registered to MR images using pelvic bones for initial rotation-translation, followed by manual adjustments to account for prostate motion and deformation from the MRI endorectal coil. Whole-mount pathology specimens were sectioned using an MRI-based patient-specific mold resulting in improved registration between the MRI, PET, and pathology. (11)C-acetate PET standardized uptake values were compared with multiparametric MRI and pathology. RESULTS: (11)C-acetate uptake was rapid but reversible, peaking at 3-5 min after injection and reaching a relative plateau at approximately 10 min. The average maximum standardized uptake value (10-12 min) of tumors was significantly higher than that of normal prostate tissue (4.4 ± 2.05 [range, 1.8-9.2] vs. 2.1 ± 0.94 [range, 0.7-3.4], respectively; P < 0.001); however, it was not significantly different from that of benign prostatic hyperplasia (4.8 ± 2.01 [range, 1.8-8.8]). A sector-based comparison with histopathology, including all tumors greater than 0.5 cm, revealed a sensitivity and specificity of 61.6% and 80.0%, respectively, for (11)C-acetate PET/CT and 82.3% and 95.1%, respectively, for MRI. The (11)C-acetate accuracy was comparable to that of MRI when only tumors greater than 0.9 cm were considered. In a small cohort (n = 9), (11)C-acetate uptake was independent of fatty acid synthase expression using immunohistochemistry. CONCLUSION: (11)C-acetate PET/CT demonstrates higher uptake in tumor foci than in normal prostate tissue; however, (11)C-acetate uptake in tumors is similar to that in benign prostate hyperplasia nodules. Although (11)C-acetate PET/CT is not likely to have utility as an independent modality for evaluation of localized PCa, the high uptake in tumors may make it useful for monitoring focal therapy when tissue damage after therapy may limit anatomic imaging methods.


Subject(s)
Acetates , Carbon , Magnetic Resonance Imaging , Multimodal Imaging , Positron-Emission Tomography , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Tomography, X-Ray Computed , Acetates/metabolism , Adult , Aged , Biological Transport , Biomarkers, Tumor/metabolism , Carbon/metabolism , Humans , Male , Middle Aged , Prostatic Neoplasms/metabolism , Sensitivity and Specificity
7.
J Cancer ; 2: 515-26, 2011.
Article in English | MEDLINE | ID: mdl-22043236

ABSTRACT

BACKGROUND & AIMS: In order to understand the role of miRNAs in renal tumorigenesis, we undertook a stepwise approach that included a comprehensive differential miRNA expression analysis for the most common histological subtypes of human renal neoplasms appearing in either sporadic or hereditary forms. We also aimed to test the hypothesis that microRNAs can act as an alternative mechanism of VHL gene inactivation and therefore might be correlated with tumorigenesis in ccRCC. Finally, we wanted to explore whether the well-known hypoxic activation of ccRCC is followed by a specific pattern of miRNA expression. METHODS: Tumor and normal adjacent kidney parenchyma from patients with RCC were tested for microRNA expression. Twenty cases of different histologies were used for profiling by PCR miRNA arrays. For validation, a separate cohort of samples used to test specifically miR92a expression and its involvement in VHL gene mRNA silencing. Finally, miR210 as a marker of hypoxia was evaluated. Expression values were correlated with important clinicopathologic features from the patients. RESULTS: We identified unique miRNA expression signatures for each histologic subtype of kidney tumors. Expression values for downregulated miRNAs ranged from 0.3-fold (in VHL-clear cell RCC) up to 0.393 fold (in papillary type II (HLRCC) tumors). For the upregulated miRNAs, fold-changes ranged from 2.1 up to 290-fold. Specific patterns together with type-specific profiles were observed. Twenty-three miRNAs were found to be differentially expressed in both sporadic and VHL-dependent ccRCC. Sporadic clear cell tumors showed a unique pattern of 14-miRNA that were absent from the VHL-dependent tumors. These also showed 15 miRNAs specific to the hereditary type. Common miRNAs to both sporadic and hereditary forms included miR-92a and miR-210. For miR-92a, and a striking inverse correlation with VHL mRNA levels was found. For the hypoxia-regulated miR-210, clear cell tumors showed significantly higher expression levels when compared to tumor of non-clear cell histology (9.90-fold vs. 1.36, p<0.001). CONCLUSIONS: microRNA expression seems to be involved in every step of RCC pathogenesis: both as an element for tumor development as well as a consequence of or in response to the initial malignant transformation and part of tumor progression. Our data show consistent disregulation of miRNAs in human kidney cancer, some of which are potentially involved in critical gene silencing in RCC and others that are activated as part of the pathophysiological response in these tumors.

8.
Nat Rev Urol ; 8(6): 321-33, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21587224

ABSTRACT

Clear cell renal cell carcinoma (RCC) represents the most common histological subtype of malignant kidney tumors. Based on symptoms alone, clear cell RCC is indistinguishable from other histological classes of RCC unless the tumor is present in the context of an RCC syndrome. Histopathological examination is, therefore, important to accurately identify clear cell RCC. Clear cell RCCs have characteristic morphological criteria; these tumors can be easily identified upon typical presentation, but diagnosis can be challenging when tumor cell pattern is unusual or when availability of tissue samples is limited. In this Review, the clinical, radiological and pathological characteristics of clear cell RCCs are described, as well as the potential tumors that can be confused with clear cell RCC and need to be considered in the differential diagnoses. Finally, the importance of an accurate diagnosis is highlighted in the context of the increasing use of preoperative tissue sampling and the prevalence of clear cell tumors associated with hereditary syndromes, which could have different therapeutic and prognostic implications for patients and their families.


Subject(s)
Carcinoma, Renal Cell/diagnosis , Diagnostic Errors , Kidney Neoplasms/diagnosis , Animals , Carcinoma, Renal Cell/classification , Carcinoma, Renal Cell/epidemiology , Humans , Kidney Neoplasms/classification , Kidney Neoplasms/epidemiology
9.
J Cancer ; 2: 67-75, 2011 Feb 07.
Article in English | MEDLINE | ID: mdl-21326627

ABSTRACT

AIMS: microRNAs (miRNAs) are a class of small noncoding RNAs that can act as key modulators in tumorigenesis-related genes. Specifically, it has been suggested that miR-21 overexpression plays a role in the development and progression of breast cancer. So far, the role of miRNAs in pregnancy-associated breast cancer (PABC) has not been investigated. METHODS AND RESULTS: We evaluated miR-21 expression by quantitative RT-PCR in 35 patients, 25 with PABC and 10 control breast cancer cases not pregnancy-associated with similar clinicopathological features. We then analyzed protein expression for PTEN, BCL2 and PDCD4 as miR-21 target genes by IHC, and finally correlated the results with patients' clinicopathological features.Significant overexpression of miR-21 in PABC tumors compared to normal adjacent tissue was found. Overexpression of miR-21 was frequently found in high grade tumors with loss of hormone receptor expression and was significantly associated with positive lymph nodes (p=0.025). In PABC patients, PTEN, BCL2 and PDCD4 target protein expression was decreased in 80%, 76% and 40% respectively. CONCLUSION: Our study supports the involvement of miR-21 in breast cancer progression and metastasis formation in PABC implying a role of this miRNA as a marker for poor prognosis in PABC patients.

10.
Urol Oncol ; 29(1): 85-9, 2011.
Article in English | MEDLINE | ID: mdl-19110452

ABSTRACT

OBJECTIVES: To evaluate the immunohistochemical profile of a carcinoid (low grade neuroendocrine tumor of the kidney) from a patient with lymph node positive disease who remains disease free for 31 months after radical nephrectomy, lymph node dissection, and adjuvant therapy with sunitinib malate. METHODS: Immunohistochemical staining was performed for chromogranin, synaptophysin, CD31, VEGF, HIF-1α, HIF-2, and Glut-1. Staining was evaluated in 3 high-power fields and samples scored as strongly positive (3+), moderately positive (2+), weakly positive (1+), or negative (0). A clear cell renal cell carcinoma was used as positive control. RESULTS: Immunohistochemical staining was strongly positive VEGF, weak to moderately positive for HIF-2, and negative for HIF-1α and Glut-1. CONCLUSIONS: Our case of primary renal carcinoid stained intensely for VEGF and HIF-2, consistent with a VHL-HIF1-HIF2-Glut1 independent pathway for VEGF activation. These data suggest that like other neuroendocrine tumors, primary renal carcinoid is a potential target for anti-angiogenic therapy with sunitinib.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Angiogenic Proteins/antagonists & inhibitors , Carcinoid Tumor/drug therapy , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Adult , Angiogenic Proteins/metabolism , Carcinoid Tumor/metabolism , Carcinoid Tumor/pathology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Chemotherapy, Adjuvant , Female , Humans , Immunoenzyme Techniques , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Tomography, X-Ray Computed
11.
J Neurooncol ; 101(2): 199-206, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20617367

ABSTRACT

Chordomas are low to intermediate grade malignancies that arise from remnants of embryonic notochord. They often recur after surgery and are highly resistant to conventional adjuvant therapies. Recently, the development of effective targeted molecular therapy has been investigated in chordomas that show receptors for tyrosine kinase (RTKs) activation. Expression of specific RTKs such as Epidermal Growth Factor Receptor (EGFR) and Mesenchymal-epithelial transition factor (c-MET) in chordomas may offer valuable therapeutic options. We investigated changes in copy number of chromosome 7 and correlated it with EGFR gene status and EGFR and c-MET protein expression in 22 chordoma samples. Chromosome 7 copy number was evaluated by chromogenic in situ hybridization (CISH) and protein expression of EGFR and c-MET by immunohistochemistry. Tumors mostly showed conventional histopathologic features and were found mainly in sacral (41%) and cranial sites (54.5%). Aneusomy of chromosome 7 was seen in 73% of the samples, 62% of primary tumors and in all recurrent chordomas. EGFR and c-MET were both expressed, but only c-MET protein expression was significantly correlated with chromosome 7 aneusomy (P ≤ 0.001). c-MET overexpression may represent an early chromosome 7 alteration that could play an important role during chordoma pathogenesis. c-MET overexpression shows promise as a molecular marker of response to targeted molecular therapy in the treatment of chordomas.


Subject(s)
Bone Neoplasms , Chordoma , Chromosomes, Human, Pair 7 , Proto-Oncogene Proteins c-met/metabolism , Adolescent , Adult , Aged , Bone Neoplasms/diagnosis , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Chordoma/diagnosis , Chordoma/genetics , Chordoma/metabolism , DNA Copy Number Variations/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Humans , In Situ Hybridization/methods , Male , Middle Aged , Proto-Oncogene Proteins c-met/genetics , Statistics as Topic , Young Adult
12.
J Cancer ; 1: 184-96, 2010 Oct 14.
Article in English | MEDLINE | ID: mdl-20975849

ABSTRACT

In this study, we aimed to evaluate the protein expression profile of a spectrum of renal cell carcinomas (RCC) to find potential biomarkers for disease onset and progression and therefore, prospective therapeutic targets. A 2D-gel based proteomic analysis was used to outline differences in protein levels among different subtypes of renal cell carcinomas, including clear cell carcinomas, papillary lesions, chromophobe tumors and renal oncocytomas. Spot pattern was compared to the corresponding normal kidney from the same patients and distinctive, differentially expressed proteins were characterized by mass spectrometry. Twenty-one protein spots were found differentially expressed between clear cell RCC and normal tissue and 38 spots were found expressed in chromophobe tumors. Eleven proteins were identified, with most differentially expressed -by fold change- between clear cell tumors and the corresponding normal tissue. Two of the identified proteins, Triosephosphate isomerase 1 (TPI-1) and Heat Shock protein 27 (Hsp27), were further validated in a separate set of tumors by immunohistochemistry and expression levels were correlated with clinicopathologic features of the patients. Hsp27 was highly expressed in 82% of the tumors used for validation, and all cases showed strong immunoreactivity for TPI-1. In both Hsp27 and TPI-1, protein expression positively correlated with histologic features of the disease. Our results suggest that the subjacent cytogenetic abnormalities seen in different histological types of RCC are followed by specific changes in protein expression. From these changes, Hsp27 and TPI-1 emerged as potential candidates for the differentiation and prognosis in RCC.

13.
Mol Cancer ; 9: 160, 2010 Jun 23.
Article in English | MEDLINE | ID: mdl-20573232

ABSTRACT

BACKGROUND: Germline mutations in the FLCN gene are responsible for the development of fibrofolliculomas, lung cysts and renal neoplasia in Birt-Hogg-Dube' (BHD) syndrome. The encoded protein folliculin (FLCN) is conserved across species but contains no classic motifs or domains and its function remains unknown. Somatic mutations or loss of heterozygosity in the remaining wild type copy of the FLCN gene have been found in renal tumors from BHD patients suggesting that FLCN is a classic tumor suppressor gene. RESULTS: To examine the tumor suppressor function of FLCN, wild-type or mutant FLCN (H255R) was stably expressed in a FLCN-null renal tumor cell line, UOK257, derived from a BHD patient. When these cells were injected into nude mice, tumor development was inversely dependent upon the level of wild-type FLCN expression. We identified genes that were differentially expressed in the cell lines with or without wild-type FLCN, many of which are involved in TGF-beta signaling, including TGF-beta2 (TGFB2), inhibin beta A chain (INHBA), thrombospondin 1 (THBS1), gremlin (GREM1), and SMAD3. In support of the in vitro data, TGFB2, INHBA, THBS1 and SMAD3 expression levels were significantly lower in BHD-associated renal tumors compared with normal kidney tissue. Although receptor mediated SMAD phosphorylation was not affected, basal and maximal TGF-beta-induced levels of TGFB2, INHBA and SMAD7 were dramatically reduced in FLCN-null cells compared with FLCN-restored cells. Secreted TGF-beta2 and activin A (homo-dimer of INHBA) protein levels were also lower in FLCN-null cells compared with FLCN-restored cells. Consistent with a growth suppressive function, activin A (but not TGF-beta2) completely suppressed anchorage-independent growth of FLCN-null UOK257 cells. CONCLUSIONS: Our data demonstrate a role for FLCN in the regulation of key molecules in TGF-beta signaling and confirm deregulation of their expression in BHD-associated renal tumors. Thus, deregulation of genes involved in TGF-beta signaling by FLCN inactivation is likely to be an important step for tumorigenesis in BHD syndrome.


Subject(s)
Gene Expression Regulation/physiology , Genes, Tumor Suppressor , Kidney Neoplasms/pathology , Proto-Oncogene Proteins/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/physiology , Animals , Cell Line, Tumor , Genetic Vectors , Humans , Kidney Neoplasms/genetics , Lentivirus/genetics , Mice , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics
14.
Anal Chem ; 82(5): 1584-8, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20121140

ABSTRACT

A method that relies on subtractive tissue-directed shot-gun proteomics to identify tumor proteins in the blood of a patient newly diagnosed with cancer is described. To avoid analytical and statistical biases caused by physiologic variability of protein expression in the human population, this method was applied on clinical specimens obtained from a single patient diagnosed with nonmetastatic renal cell carcinoma (RCC). The proteomes extracted from tumor, normal adjacent tissue and preoperative plasma were analyzed using 2D-liquid chromatography-mass spectrometry (LC-MS). The lists of identified proteins were filtered to discover proteins that (i) were found in the tumor but not normal tissue, (ii) were identified in matching plasma, and (iii) whose spectral count was higher in tumor tissue than plasma. These filtering criteria resulted in identification of eight tumor proteins in the blood. Subsequent Western-blot analysis confirmed the presence of cadherin-5, cadherin-11, DEAD-box protein-23, and pyruvate kinase in the blood of the patient in the study as well as in the blood of four other patients diagnosed with RCC. These results demonstrate the utility of a combined blood/tissue analysis strategy that permits the detection of tumor proteins in the blood of a patient diagnosed with RCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/blood , Kidney Neoplasms/blood , Biomarkers, Tumor/analysis , Carcinoma, Renal Cell/diagnosis , Chromatography, Liquid , Humans , Kidney Neoplasms/diagnosis , Mass Spectrometry
15.
PLoS One ; 5(12): e15793, 2010 Dec 29.
Article in English | MEDLINE | ID: mdl-21209915

ABSTRACT

BACKGROUND: Germline mutations in a tumor suppressor gene FLCN lead to development of fibrofolliculomas, lung cysts and renal cell carcinoma (RCC) in Birt-Hogg-Dubé syndrome. TFE3 is a member of the MiTF/TFE transcription factor family and Xp11.2 translocations found in sporadic RCC involving TFE3 result in gene fusions and overexpression of chimeric fusion proteins that retain the C-terminal DNA binding domain of TFE3. We found that GPNMB expression, which is regulated by MiTF, was greatly elevated in renal cancer cells harboring either TFE3 translocations or FLCN inactivation. Since TFE3 is implicated in RCC, we hypothesized that elevated GPNMB expression was due to increased TFE3 activity resulting from the inactivation of FLCN. METHODOLOGY/PRINCIPAL FINDINGS: TFE3 knockdown reduced GPNMB expression in renal cancer cells harboring either TFE3 translocations or FLCN inactivation. Moreover, FLCN knockdown induced GPNMB expression in FLCN-restored renal cancer cells. Conversely, wildtype FLCN suppressed GPNMB expression in FLCN-null cells. FLCN inactivation was correlated with increased TFE3 transcriptional activity accompanied by its nuclear localization as revealed by elevated GPNMB mRNA and protein expression, and predominantly nuclear immunostaining of TFE3 in renal cancer cells, mouse embryo fibroblast cells, mouse kidneys and mouse and human renal tumors. Nuclear localization of TFE3 was associated with TFE3 post-translational modifications including decreased phosphorylation. CONCLUSIONS/SIGNIFICANCE: Increased TFE3 activity is a downstream event induced by FLCN inactivation and is likely to be important for renal tumor development. This study provides an important novel mechanism for induction of TFE3 activity in addition to TFE3 overexpression resulting from Xp11.2 translocations, suggesting that TFE3 may be more broadly involved in tumorigenesis.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Nucleus/metabolism , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/metabolism , Mutation , Proto-Oncogene Proteins/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/metabolism , Animals , Humans , Kidney/metabolism , Mice , Phosphorylation , Recombinant Fusion Proteins/metabolism
16.
Cancer Genet Cytogenet ; 196(1): 45-55, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19963135

ABSTRACT

Energy deregulation and abnormalities of tumor cell metabolism are critical issues in understanding cancer. Hereditary leiomyomatosis renal cell carcinoma (HLRCC) is an aggressive form of RCC characterized by germline mutation of the Krebs cycle enzyme fumarate hydratase (FH), and one known to be highly metastatic and unusually lethal. There is considerable utility in establishing preclinical cell and xenograft models for study of disorders of energy metabolism, as well as in development of new therapeutic approaches targeting of tricarboxylic acid (TCA) cycle enzyme-deficient human cancers. Here we describe a new immortalized cell line, UOK 262, derived from a patient having aggressive HLRCC-associated recurring kidney cancer. We investigated gene expression, chromosome profiles, efflux bioenergetic analysis, mitochondrial ultrastructure, FH catabolic activity, invasiveness, and optimal glucose requirements for in vitro growth. UOK 262 cells have an isochromosome 1q recurring chromosome abnormality, i(1)(q10), and exhibit compromised oxidative phosphorylation and in vitro dependence on anaerobic glycolysis consistent with the clinical manifestation of HLRCC. The cells also display glucose-dependent growth, an elevated rate of lactate efflux, and overexpression of the glucose transporter GLUT1 and of lactate dehydrogenase A (LDHA). Mutant FH protein was present primarily in edematous mitochondria, but with catalytic activity nearly undetectable. UOK 262 xenografts retain the characteristics of HLRCC histopathology. Our findings indicate that the severe compromise of oxidative phosphorylation and rapid glycolytic flux in UOK 262 are an essential feature of this TCA cycle enzyme-deficient form of kidney cancer. This tumor model is the embodiment of the Warburg effect. UOK 262 provides a unique in vitro and in vivo preclinical model for studying the bioenergetics of the Warburg effect in human cancer.


Subject(s)
Carcinoma, Renal Cell/genetics , Fumarate Hydratase/genetics , Kidney Neoplasms/genetics , Leiomyomatosis/genetics , Models, Biological , Animals , Blotting, Western , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/metabolism , Cell Line , Energy Metabolism , Fumarate Hydratase/metabolism , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , In Vitro Techniques , Kidney Neoplasms/enzymology , Kidney Neoplasms/metabolism , Leiomyomatosis/enzymology , Leiomyomatosis/metabolism , Mice , Mice, Nude
17.
Proc Natl Acad Sci U S A ; 106(44): 18722-7, 2009 Nov 03.
Article in English | MEDLINE | ID: mdl-19850877

ABSTRACT

Germline mutations in the BHD/FLCN tumor suppressor gene predispose patients to develop renal tumors in the hamartoma syndrome, Birt-Hogg-Dubé (BHD). BHD encodes folliculin, a protein with unknown function that may interact with the energy- and nutrient-sensing AMPK-mTOR signaling pathways. To clarify BHD function in the mouse, we generated a BHD knockout mouse model. BHD homozygous null (BHD(d/d)) mice displayed early embryonic lethality at E5.5-E6.5, showing defects in the visceral endoderm. BHD heterozygous knockout (BHDd(/+)) mice appeared normal at birth but developed kidney cysts and solid tumors as they aged (median kidney-lesion-free survival = 23 months, median tumor-free survival = 25 months). As observed in human BHD kidney tumors, three different histologic types of kidney tumors developed in BHD(d/+) mice including oncocytic hybrid, oncocytoma, and clear cell with concomitant loss of heterozygosity (LOH), supporting a tumor suppressor function for BHD in the mouse. The PI3K-AKT pathway was activated in both human BHD renal tumors and kidney tumors in BHD(d/+) mice. Interestingly, total AKT protein was elevated in kidney tumors compared to normal kidney tissue, but without increased levels of AKT mRNA, suggesting that AKT may be regulated by folliculin through post translational or post-transcriptional modification. Finally, BHD inactivation led to both mTORC1 and mTORC2 activation in kidney tumors from BHD(d/+) mice and human BHD patients. These data support a role for PI3K-AKT pathway activation in kidney tumor formation caused by loss of BHD and suggest that inhibitors of both mTORC1 and mTORC2 may be effective as potential therapeutic agents for BHD-associated kidney cancer.


Subject(s)
Embryo Loss/pathology , Homozygote , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics , Embryo Loss/genetics , Embryo, Mammalian/pathology , Embryonic Development , Enzyme Activation , Humans , Kidney Neoplasms/complications , Kidney Neoplasms/enzymology , Loss of Heterozygosity/genetics , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Proteins , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases
18.
Mol Cancer Ther ; 8(3): 626-35, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19276158

ABSTRACT

The genetic basis for the hereditary leiomyomatosis and renal cell cancer syndrome is germ-line inactivating mutation in the gene for the Krebs/tricarboxylic acid cycle enzyme, fumarate hydratase (FH), the enzyme that converts fumarate to malate. These individuals are predisposed to development of leiomyomas of the skin and uterus as well as highly aggressive kidney cancers. Inhibition of FH should result in significant decrease in oxidative phosphorylation necessitating that glycolysis followed by fermentation of pyruvate to lactate will be required to provide adequate ATP as well as to regenerate NAD+. Moreover, FH deficiency is known to up-regulate expression of hypoxia-inducible factor (HIF)-1alpha by enhancing the stability of HIF transcript. This leads to activation of various HIF-regulated genes including vascular endothelial growth factor and glucose transporter GLUT1 and increased expression of several glycolytic enzymes. Because lactate dehydrogenase-A (LDH-A), also a HIF-1alpha target, promotes fermentative glycolysis (conversion of pyruvate to lactate), a step essential for regenerating NAD+, we asked whether FH-deficient cells would be exquisitely sensitive to LDH-A blockade. Here, we report that hereditary leiomyomatosis and renal cell cancer tumors indeed overexpress LDH-A, that LDH-A inhibition results in increased apoptosis in a cell with FH deficiency and that this effect is reactive oxygen species mediated, and that LDH-A knockdown in the background of FH knockdown results in significant reduction in tumor growth in a xenograft mouse model.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Enzyme Inhibitors/therapeutic use , Kidney Neoplasms/drug therapy , L-Lactate Dehydrogenase/antagonists & inhibitors , Leiomyomatosis/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/complications , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Enzyme Inhibitors/pharmacology , Female , Fumarate Hydratase/genetics , Gene Knockdown Techniques , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Kidney Neoplasms/complications , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Lactate Dehydrogenase 5 , Leiomyomatosis/complications , Leiomyomatosis/genetics , Male , Mice , Mice, Nude , Models, Biological , Syndrome , Tumor Cells, Cultured , Validation Studies as Topic , Xenograft Model Antitumor Assays , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/pathology
19.
Ann Surg Oncol ; 15(9): 2472-81, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18594929

ABSTRACT

BACKGROUND: This study sought to clarify the distribution of intrahepatic micrometastases and elucidate an adequate hepatectomy margin for colorectal carcinoma liver metastases. METHODS: Intrahepatic micrometastases in resected specimens from 90 patients who underwent hepatectomy for colorectal carcinoma liver metastases were examined retrospectively. Intrahepatic micrometastases were defined as microscopic lesions spatially separated from the gross tumor. Distances from these lesions to the hepatic tumor borders were measured histologically, and the density of intrahepatic micrometastases (number of lesions/mm(2)) calculated relative to the advancing tumor border in a zone <1 cm from the border (close) or >/=1 cm away (distant). Median follow-up time was 127 months. RESULTS: A total of 294 intrahepatic micrometastases were detected in 52 (58%) patients; 95% of these occurred in the close zone. The density of intrahepatic micrometastases was significantly higher in the close zone (mean 74.8 x 10(-4 ) lesions/mm(2)) than in the distant zone (mean 7.4 x 10(-4 ) lesions/mm(2); P < 0.001). Hepatectomy margin status was positive by 0 cm in 10 patients or negative by <1 cm in 51, and by >/=1 cm in 29 patients. The median survival times were 18, 33, and 89 months in patients with hepatectomy margins 0 cm, <1 cm, and >/=1 cm, respectively. Hepatectomy margin status independently influenced survival (P < 0.001) and disease-free survival (P < 0.001). CONCLUSION: The currently recommended >/=1 cm hepatectomy margin should remain the goal for resections of colorectal carcinoma liver metastases, based on the distribution of intrahepatic micrometastases and survival risk.


Subject(s)
Adenocarcinoma/surgery , Colorectal Neoplasms/pathology , Hepatectomy , Liver Neoplasms/surgery , Neoplasm Recurrence, Local/diagnosis , Adenocarcinoma/secondary , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/surgery , Female , Follow-Up Studies , Humans , Liver Neoplasms/secondary , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Prognosis , Retrospective Studies , Survival Rate , Treatment Outcome
20.
Gene ; 415(1-2): 60-7, 2008 May 31.
Article in English | MEDLINE | ID: mdl-18403135

ABSTRACT

Birt-Hogg-Dube' syndrome characterized by increased risk for renal neoplasia is caused by germline mutations in the BHD/FLCN gene encoding a novel tumor suppressor protein, folliculin(FLCN), which interacts with FNIP1 and 5'-AMP-activated protein kinase(AMPK). Here we report the identification and characterization of a novel FNIP1 homolog FNIP2 that also interacts with FLCN and AMPK. C-terminally-deleted FLCN mutants, similar to those produced by naturally-occurring germline mutations in BHD patients, were unable to bind FNIP2. These data taken together with our previous results that demonstrated FNIP1 binding to the C-terminus of FLCN suggest that FLCN tumor suppressor function may be facilitated by interactions with both FNIP1 and FNIP2 through its C-terminus. Furthermore, we demonstrate that FNIP1 and FNIP2 are able to form homo- or heteromeric multimers suggesting that they may function independently or cooperatively with FLCN. Differential expression of FNIP1 and FNIP2 transcripts in some normal tissues may indicate tissue specificity for these homologs. Interestingly FNIP1 and FNIP2 were oppositely expressed in human clear cell renal cell carcinoma (RCC), and coordinately expressed in chromophobe RCC and oncocytoma, suggesting their differential function in different histologic variants of RCC.


Subject(s)
Carrier Proteins/genetics , Adenylate Kinase/metabolism , Amino Acid Sequence , Carcinoma, Renal Cell/genetics , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Line , Evolution, Molecular , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Kidney/metabolism , Kidney/pathology , Kidney Neoplasms/genetics , Molecular Sequence Data , Multiprotein Complexes/metabolism , Protein Binding , Protein Transport , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Nucleic Acid , Subcellular Fractions/enzymology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...