Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cancers (Basel) ; 15(9)2023 May 06.
Article in English | MEDLINE | ID: mdl-37174100

ABSTRACT

Although the roles of erythroblastic leukemia viral oncogene homolog 2 (ERBB2), neuregulin 4 (NRG4), and mitogen-inducible gene 6 (MIG6) in epidermal growth factor receptor signaling in hepatocellular carcinoma (HCC) and other malignancies have been previously investigated, the prognostic value of their serum levels in HCC remains undetermined. In the present study, correlations between serum levels and tumor characteristics, overall survival, and tumor recurrence were analyzed. Furthermore, the prognostic potential of the serum levels of these biomarkers was evaluated relative to that of alpha-fetoprotein. Both ERBB2 and NRG4 correlated with the Barcelona Clinic Liver Cancer stage, ERBB2 correlated with the tumor-maximal diameter, and NRG4 correlated with a tumor number. Cox proportional hazards regression analysis revealed that ERBB2 (hazard ratio [HR], 2.719; p = 0.007) was an independent prognostic factor for overall survival. Furthermore, ERBB2 (HR, 2.338; p = 0.002) and NRG4 (HR, 431.763; p = 0.001) were independent prognostic factors for tumor recurrence. The products of ERBB2 and NRG4 had a better area under the curve than alpha-fetoprotein for predicting 6-month, 1-year, 3-year, and 5-year mortality. Therefore, these factors could be used to evaluate prognosis and monitor treatment response in patients with HCC.

4.
Am J Physiol Renal Physiol ; 314(5): F773-F787, 2018 05 01.
Article in English | MEDLINE | ID: mdl-28724608

ABSTRACT

Tubulointerstitial fibrosis (TIF) is a prominent factor in the progression of chronic kidney disease regardless of etiology. Avian erythroblastic leukemia viral oncogene homolog 4 (ErbB4) expression levels were inversely correlated to renal fibrosis in human fibrotic kidneys. In both unilateral ureteral obstruction (UUO) and ischemia-reperfusion injury followed by uninephrectomy (IRI/UNx) mouse models, expression levels of ErbB4 were elevated in the early stage of renal injury. Using mice with global ErbB4 deletion except for transgenic rescue in cardiac tissue ( ErbB4-/-ht+), we determined that UUO induced similar injury in proximal tubules compared with wild-type mice but more severe injury in distal nephrons. TIF was apparent earlier and was more pronounced following UUO in ErbB4-/-ht+ mice. With ErbB4 deletion, UUO injury inhibited protein kinase B phosphorylation and increased the percentage of cells in G2/M arrest. There was also increased nuclear immunostaining of yes-associated protein and increased expression of phospho-Mothers against decapentaplegic homolog 3, snail1, and vimentin. These results indicate that ErbB4 deletion accelerates the development and progression of renal fibrosis in obstructive nephropathy. Similar results were found in a mouse IRI/UNx model. In conclusion, increased expression of ErbB4 in the early stages of renal injury may reflect a compensatory effect to lessen tubulointerstitial injury.


Subject(s)
Acute Kidney Injury/etiology , Gene Deletion , Kidney/metabolism , Receptor, ErbB-4/deficiency , Renal Insufficiency, Chronic/etiology , Reperfusion Injury/etiology , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Case-Control Studies , Cell Cycle Proteins , Cell Dedifferentiation , Disease Models, Animal , Disease Progression , Fibrosis , G2 Phase Cell Cycle Checkpoints , Genetic Predisposition to Disease , Kidney/pathology , Mice, Knockout , Nephrectomy , Phenotype , Phosphoproteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Severity of Illness Index , Signal Transduction , Smad3 Protein/metabolism , Snail Family Transcription Factors/metabolism , Time Factors , Ureteral Obstruction/complications , Vimentin/metabolism , YAP-Signaling Proteins
5.
Cancer ; 121(10): 1645-53, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25641763

ABSTRACT

BACKGROUND: Acquired resistance to antiepidermal growth factor receptor (anti-EGFR) therapy may be caused by EGFR-v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2 (ErbB2) heterodimerization and pathway reactivation. In preclinical studies, inhibiting ErbB2 blocked this resistance mechanism and resensitized cells to anti-EGFR therapy. Cetuximab targets EGFR, whereas lapatinib inhibits both EGFR and ErbB2. The objective of this phase 1 trial was to assess the safety, dose-limiting toxicities (DLTs), and maximum tolerated doses (MTDs) of cetuximab and lapatinib in patients with solid tumors. METHODS: Patients received standard weekly cetuximab with escalating lapatinib doses of 750 mg, 1000 mg, or 1250 mg daily in 3-week cycles. DLTs were monitored through the end of cycle 2. Pretreatment and post-treatment tumor biopsies and germline DNA samples were obtained for correlative studies. RESULTS: Twenty-two patients were enrolled, and 18 patients each were evaluable for toxicity and response. Fifty-nine percent of patients had received prior anti-EGFR therapy. Common toxicities included rash and diarrhea. No patient experienced a DLT at the highest dose level, and no grade 4 toxicity was observed. Response included no complete responses, 3 partial responses, 9 patients with stable disease, and 6 patients with disease progression, for an overall response rate of 17% and a clinical benefit rate of 67%. The clinical benefit rate in patients who had previously received anti-EGFR therapy was 70%. The mean treatment duration was 4.7 cycles (range, 1-14 cycles). Decreased expression of EGFR/ErbB2 pathway components after treatment was correlated with response, whereas increased expression in the PI3K, Jak/Stat, and MAPK pathways occurred in nonresponders. CONCLUSIONS: The combination of cetuximab and lapatinib was well tolerated, had the expected toxicities, and exhibited notable clinical activity, including in patients who had received previous anti-EGFR therapy. Further clinical study of this combination is warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Anus Neoplasms/drug therapy , Biopsy , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Squamous Cell/drug therapy , Cetuximab , Colorectal Neoplasms/drug therapy , Diarrhea/chemically induced , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Eruptions/etiology , ErbB Receptors/genetics , Female , Genetic Variation , Genotype , Head and Neck Neoplasms/drug therapy , Humans , Lapatinib , Lung Neoplasms/drug therapy , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/genetics , Pharmacogenetics , Quinazolines/administration & dosage , Quinazolines/adverse effects , Quinazolines/pharmacokinetics , Receptor, ErbB-2/genetics , Signal Transduction/drug effects , Treatment Outcome
6.
Autophagy ; 10(11): 2036-52, 2014.
Article in English | MEDLINE | ID: mdl-25483966

ABSTRACT

Earlier studies reported allelic deletion of the essential autophagy regulator BECN1 in breast cancers implicating BECN1 loss, and likely defective autophagy, in tumorigenesis. Recent studies have questioned the tumor suppressive role of autophagy, as autophagy-related gene (Atg) defects generally suppress tumorigenesis in well-characterized mouse tumor models. We now report that, while it delays or does not alter mammary tumorigenesis driven by Palb2 loss or ERBB2 and PyMT overexpression, monoallelic Becn1 loss promotes mammary tumor development in 2 specific contexts, namely following parity and in association with wingless-type MMTV integration site family, member 1 (WNT1) activation. Our studies demonstrate that Becn1 heterozygosity, which results in immature mammary epithelial cell expansion and aberrant TNFRSF11A/TNR11/RANK (tumor necrosis factor receptor superfamily, member 11a, NFKB activator) signaling, promotes mammary tumorigenesis in multiparous FVB/N mice and in cooperation with the progenitor cell-transforming WNT1 oncogene. Similar to our Becn1(+/-);MMTV-Wnt1 mouse model, low BECN1 expression and an activated WNT pathway gene signature correlate with the triple-negative subtype, TNFRSF11A axis activation and poor prognosis in human breast cancers. Our results suggest that BECN1 may have nonautophagy-related roles in mammary development, provide insight in the seemingly paradoxical roles of BECN1 in tumorigenesis, and constitute the basis for further studies on the pathophysiology and treatment of clinically aggressive triple negative breast cancers (TNBCs).


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Mammary Neoplasms, Animal/metabolism , Wnt1 Protein/metabolism , Alleles , Animals , Apoptosis , Autophagy , Beclin-1 , Breast Neoplasms/metabolism , Cell Proliferation , Epithelial Cells/cytology , Female , Gene Expression Regulation, Neoplastic , Heterozygote , Humans , Mammary Neoplasms, Experimental/metabolism , Membrane Proteins/metabolism , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis , Signal Transduction , Stem Cells/cytology , Triple Negative Breast Neoplasms/metabolism
7.
Autophagy ; 10(12): 2122-42, 2014.
Article in English | MEDLINE | ID: mdl-25427136

ABSTRACT

The triple-negative breast cancer (TN BC) subtype is the most aggressive form of invasive BC. Despite intensive efforts to improve BC treatments, patients with TN BC continue to exhibit poor survival, with half developing resistance to chemotherapy. Here we identify autophagy as a key mechanism in the progression and chemoresistance of a subset of TN tumors. We demonstrate that LC3B, a protein involved in autophagosome formation, is a reliable marker of poor prognosis in TN BC, validating this prognostic value at both the mRNA and protein levels in several independent cohorts. We also show that LC3B has no prognostic value for other BC subtypes (Luminal or HER2 BC), thus revealing a specific impact of autophagy on TN tumors. Autophagy is essential for the proliferative and invasive properties in 3D of TN BC cells characterized by high LC3B levels. Interestingly, the activity of the transcriptional co-activator YAP1 (Yes-associated protein 1) is regulated by the autophagy process and we identify YAP1 as a new actor in the autophagy-dependent proliferative and invasive properties of high-LC3B TN BC. Finally, inhibiting autophagy by silencing ATG5 or ATG7 significantly impaired high-LC3B TN tumor growth in vivo. Moreover, using a patient-derived TN tumor transplanted into mice, we show that an autophagy inhibitor, chloroquine, potentiates the effects of chemotherapeutic agents. Overall, our data identify LC3B as a new prognostic marker for TN BC and the inhibition of autophagy as a promising therapeutic strategy for TN BC patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Biomarkers, Tumor/metabolism , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/therapy , Animals , Female , Humans , Mice , Microtubule-Associated Proteins/metabolism , Prognosis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Treatment Outcome
8.
J Bone Oncol ; 3(1): 18-24, 2014 Mar.
Article in English | MEDLINE | ID: mdl-26909290

ABSTRACT

BACKGROUND: Breast cancer often metastasizes into bone and leads to osteolytic lesions. The underlying mechanisms, however, are complex and not fully understood. Syndecan-1 is a proteoglycan that has various functions relevant for tumor progression including cell-cell communication and cell-matrix interactions. Moreover, its two glycosaminoglycan-binding sites suggest that it may interfere with glycoproteins such as osteoprotegerin, a potent inhibitor of osteoclastogenesis. Thus, we hypothesize that tumor-derived syndecan-1 alters osteoclast biology by modulating osteoprotegerin. METHODS: Syndecan-1 expression was down-regulated via siRNA and the cell fate of the breast cancer cell lines MCF-7, T-47D, and MDA-MB-231 was investigated. Furthermore, we determined the regulation of syndecan-1 by dexamethasone, a commonly used antiemetic in breast cancer therapy. Additionally, we analyzed the genesis and activity of osteoclasts in indirect co-culture experiments using supernatants from MCF-7 cells with deficient and sufficient levels of syndecan-1. RESULTS: Dexamethasone time- and dose-dependently increased syndecan-1 expression up to 4-fold but did not alter cell behavior. Syndecan-1 up-regulation did not affect the survival or migration of breast cancer cells. Depletion of syndecan-1 using siRNA led to decreased vitality of progesterone receptor-positive cell lines. In MCF-7 cells osteoprotegerin production was up-regulated 2.5-fold after syndecan-1 knock-down. The culture of osteoclast precursors with the supernatant of MCF-7 cells with reduced syndecan-1 levels suppressed osteoclast formation and activity by 21% and 23%, respectively. Adding neutralizing antibodies to osteoprotegerin to the breast cancer supernatants reversed osteoclastogenesis. CONCLUSION: Thus, we identified tumor-derived syndecan-1 as a novel positive regulator of osteoclastogenesis and new player in the tumor-bone dialog.

9.
Oncoimmunology ; 3(8): e952202, 2014.
Article in English | MEDLINE | ID: mdl-25960931

ABSTRACT

Whereas the presence of autoantibodies in cancer patients has been acknowledged, their diagnostic or therapeutic significance has yet to be established. This is due, at least in part, to the lack of robust screening techniques to detect and characterize such antibodies for further assessment. In this study, we screened colorectal cancer (CRC) patient sera for antibodies specifically targeting the key cell cycle inhibitory factor p21 encoded by the cyclin-dependent kinase inhibitor 1A (CDKN1A). Anti-p21 antibody titers were higher in CRC patient samples versus controls, correlating with a more advanced disease stage and lymph node involvement. Further, we isolated for the first time a specific human antibody fragment against p21, which could potentially be useful as a tool to study tumorigenicity in CRC patients.

10.
Oncoimmunology ; 3(11): e956005, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25941587

ABSTRACT

The insulin-like growth factor-1 receptor (IGF-1R) plays a crucial role in cellular growth, proliferation, transformation, and inhibition of apoptosis. A myriad of human cancer types have been shown to overexpress IGF-1R, including breast and pancreatic adenocarcinoma. IGF-1R signaling interferes with numerous receptor pathways, rendering tumor cells resistant to chemotherapy, anti-hormonal therapy, and epidermal growth factor receptor (EGFR, also known as HER-1) and v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2, (ERBB2, best known as HER-2) -targeted therapies. Targeting the IGF:IGF-1R axis with innovative peptide inhibitors and vaccine antibodies thus represents a promising therapeutic strategy to overcome drug resistance and to provide new avenues for individualized and combinatorial treatment strategies. In this study, we designed, synthesized, and characterized several B-cell epitopes from the IGF-1:IGF-1R axis. The chimeric peptide epitopes were highly immunogenic in outbred rabbits, eliciting high levels of peptide vaccine antibodies. The IGF-1R peptide antibodies and peptide mimics inhibited cell proliferation and receptor phosphorylation, induced apoptosis and antibody-dependent cellular cytotoxicity (ADCC), and significantly inhibited tumor growth in the transplantable BxPC-3 pancreatic and JIMT-1 breast cancer models. Our results showed that the peptides and antibodies targeting residues 56-81 and 233-251 are potential therapeutic and vaccine candidates for the treatment of IGF-1R-expressing cancers, including those that are resistant to the HER-2-targeted antibody, trastuzumab. Additionally, we found additive antitumor effects for the combination treatment of the IGF-1R 56-81 epitope with HER-1-418 and HER-2-597 epitopes. Treatment with the IGF-1R/HER-1 or IGF-1R/HER-2 combination inhibited proliferation, invasion, and receptor phosphorylation, and induced apoptosis and ADCC, to a greater degree than single agents.

11.
Semin Diagn Pathol ; 30(4): 298-312, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24342286

ABSTRACT

Non-small-cell lung cancer (NSCLC) subtyping has recently been a key factor in determining patient management with novel drugs. In addition, the identification of distinct oncogenic driver mutations frequently associated with NSCLC histotype and coupled to the clinical responses to targeted therapies have revolutionized the impact of histologic type and molecular biomarkers in lung cancer. Several molecular alterations involving different genes (EGFR, KRAS, ALK, BRAF, and HER2) seem to have a remarkable predilection for adenocarcinoma and specific inhibitors of EGFR and ALK are now available for patients with adenocarcinoma harboring the relevant gene alterations. The efficacy of histology-based and molecular-targeted therapies had a deep impact in (1) re-defining classification of lung cancer (particularly adenocarcinomas) and (2) routine clinical practice of pathologists involved in optimization of handling of tissue samples in order to guarantee NSCLC subtyping with the help of immunohistochemistry and adequately preserve tumor cells for molecular analysis. In agreement with the modern multidisciplinary approach to lung cancer, we reviewed here the diagnostic and predictive value of molecular biomarkers according to the clinical, pathologic, and molecular biologist viewpoints.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/diagnosis , Lung Neoplasms/diagnosis , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Humans , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Precision Medicine/methods , Precision Medicine/trends
12.
Gastroenterology ; 145(6): 1215-29, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24140396

ABSTRACT

Cholangiocarcinomas (CCAs) are hepatobiliary cancers with features of cholangiocyte differentiation; they can be classified anatomically as intrahepatic CCA (iCCA), perihilar CCA (pCCA), or distal CCA. These subtypes differ not only in their anatomic location, but in epidemiology, origin, etiology, pathogenesis, and treatment. The incidence and mortality of iCCA has been increasing over the past 3 decades, and only a low percentage of patients survive until 5 years after diagnosis. Geographic variations in the incidence of CCA are related to variations in risk factors. Changes in oncogene and inflammatory signaling pathways, as well as genetic and epigenetic alterations and chromosome aberrations, have been shown to contribute to the development of CCA. Furthermore, CCAs are surrounded by a dense stroma that contains many cancer-associated fibroblasts, which promotes their progression. We have gained a better understanding of the imaging characteristics of iCCAs and have developed advanced cytologic techniques to detect pCCAs. Patients with iCCAs usually are treated surgically, whereas liver transplantation after neoadjuvant chemoradiation is an option for a subset of patients with pCCAs. We review recent developments in our understanding of the epidemiology and pathogenesis of CCA, along with advances in classification, diagnosis, and treatment.


Subject(s)
Bile Duct Neoplasms/diagnosis , Bile Duct Neoplasms/etiology , Bile Ducts, Intrahepatic , Cholangiocarcinoma/diagnosis , Cholangiocarcinoma/etiology , Disease Management , Animals , Bile Duct Neoplasms/therapy , Chemoradiotherapy , Cholangiocarcinoma/therapy , Combined Modality Therapy , Disease Models, Animal , Humans , Incidence , Liver Transplantation , Mesothelin , Proto-Oncogene Mas , Risk Factors , Survival Rate
13.
J Pharm Biomed Anal ; 86: 49-55, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23973791

ABSTRACT

Allitinib, also known as AST1306, is a novel irreversible inhibitor of the epidermal growth factor receptors 1 and 2. Allitinib is currently used in clinical trial to treat solid tumors. A previous study showed that allitinib is extensively metabolized in humans. Amide hydrolysis metabolite (M6) and 29,30-dihydrodiol allitinib (M10) are the major metabolites in circulation. To study the pharmacokinetics of allitinib and its two major metabolites in cancer patients, a rapid, sensitive and reliable LC-MS/MS method was developed and validated for the simultaneous determination of allitinib, M6 and M10 in human plasma. After simple protein precipitation, the analytes and the combined internal standards (lapatinib and NB-2, an analog of allitinib) were separated on a Zorbax Eclipase XDB C18 column (50 mm × 4.6 mm, 1.8 µm, Agilent) using a mobile phase of 5 mM ammonium acetate with 0.1% formic acid (phase A) and 50% (v/v) methanol in acetonitrile (phase B) with gradient elution. Mass spectrometric detection was conducted by atmospheric-pressure chemical ionization in positive ion multiple reaction monitoring modes using AB Sciex Triple Quad 6500 system. Linear calibration curves were obtained for the following concentration range: 0.300-200 ng/ml for allitinib; 0.030-20.0 ng/ml for M6; and 0.075-50.0 ng/ml for M10. Intra-day and inter-day accuracy and precision were within the acceptable limits of ±15% at all of the concentrations. The method was successfully applied to a preliminary clinical pharmacokinetic study following oral administration of allitinib tosylate tablets in cancer patients.


Subject(s)
Acrylamides/analysis , Acrylamides/blood , Quinazolines/analysis , Quinazolines/blood , Tandem Mass Spectrometry/standards , Chromatography, Liquid/standards , Chromatography, Liquid/trends , Humans , Mass Spectrometry/standards , Mass Spectrometry/trends , Tandem Mass Spectrometry/trends
14.
Med Hypotheses ; 81(3): 439-42, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23830593

ABSTRACT

VEGFR and HER2 are both important transmembrane proteins associated with several types of cancer. Overexpression of these 2 proteins had long been thought to contribute to cancer progression and poor outcomes, thus, therapies targeting HER-2 and VEGFA signaling pathways have been applied in recent years. Herceptin is a HER-2 targeted antibody that being widely used for the management of HER-2 positive breast cancer, which demonstrate significant benefits in both the metastatic and adjuvant settings. However, acquired resistance develops in most treated patients despite treatment in as early as 10 months. Identification of subpopulations best suited for and most likely to respond to Herceptin is of utmost importance. We analyzed the signaling pathways of HER-2 and found that HER-2 shares a very similar downstream network with VEGFA, while LGR5 lies in the upstream of VEGFA and could promotes its expression through CTNNB1. This discovery suggests that the LGR5 directed VEGFA overexpression may activate downstream signals of HER-2 despite Herceptin treatment. Here, we hypothesized that in LGR5 overexpressing breast cancer cases, activation of VEGFA-VRGFR bypass may account for the resistance to HER-2 directed therapies. Concurrent inhibition of VEGFR might enhance Herceptin sensitivity and moreover reverse the resistant phenotype in HER-2 positive breast cancer. Thus, we proposed alternate regimens to increase the efficacy of Herceptin-based therapy. Nevertheless, wet lab experiments and clinical trials are still required.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Breast Neoplasms/drug therapy , Receptor, ErbB-2/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Biomarkers, Tumor , Breast Neoplasms/metabolism , Female , Humans , Models, Biological , Precision Medicine/methods , Trastuzumab
15.
Eur J Obstet Gynecol Reprod Biol ; 170(1): 96-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23810060

ABSTRACT

OBJECTIVE: In this study, we compared human placental gene expression patterns of epidermal growth factor (EGF) in pregnancies with intrauterine growth restriction (IUGR) vs. normal pregnancies as control. STUDY DESIGN: Gene expression of EGF was determined from human placental samples collected from all pregnancies presenting with IUGR at our institution during the study period January 1, 2010-January 1, 2011. Multiple clinical variables were also assessed including maternal age, gestational weight gain, increase of BMI during pregnancy and fetal gender. RESULTS: A total of 241 samples were obtained (101 in the IUGR pregnancy group, 140 in the normal pregnancy group). EGF was found to be underexpressed in the IUGR group compared to normal pregnancy (Ln2(α): -1.54; p<0.04). Within the IUGR group no fetal gender-dependent difference was seen in EGF gene expression (Ln2(α): 0.44; p<0.06). Similarly, no significant difference in EGF expression was noted in cases with more vs. less severe forms of IUGR (Ln2(α): -0.08; p=0.05). IUGR pregnancies were significantly more common in the maternal age group 35-44 years compared to other age groups. Gestational weight gain and gestational BMI increase were significantly lower in IUGR pregnancies compared to controls. CONCLUSIONS: Placental expression of EGF was found to be reduced in IUGR pregnancies vs. normal pregnancies. This may partly explain the smaller placental size and placental dysfunction commonly seen with IUGR. An increased incidence of IUGR was observed with maternal age exceeding 35 years. The probability of IUGR correlated with lower gestational weight gain and lower BMI increase during pregnancy.


Subject(s)
Epidermal Growth Factor/metabolism , Fetal Growth Retardation/metabolism , Placenta/metabolism , Adolescent , Adult , Case-Control Studies , Female , Gene Expression , Humans , Infant, Newborn , Male , Pregnancy , Sex Characteristics , Young Adult
16.
Clin Chim Acta ; 424: 53-65, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-23669186

ABSTRACT

Gastric cancer is one of the most common malignancies and remains the second leading cause of cancer-related death worldwide. Over 70% of new cases and deaths occur in developing countries. In the early years of the molecular biology revolution, cancer research mainly focuses on genetic alterations, including gastric cancer. Epigenetic mechanisms are essential for normal development and maintenance of tissue-specific gene expression patterns in mammals. Disruption of epigenetic processes can lead to altered gene function and malignant cellular transformation. Recent advancements in the rapidly evolving field of cancer epigenetics have shown extensive reprogramming of every component of the epigenetic machinery in cancer, including DNA methylation, histone modifications, nucleosome positioning, noncoding RNAs, and microRNAs. Aberrant DNA methylation in the promoter regions of gene, which leads to inactivation of tumor suppressor and other cancer-related genes in cancer cells, is the most well-defined epigenetic hallmark in gastric cancer. The advantages of gene methylation as a target for detection and diagnosis of cancer in biopsy specimens and non-invasive body fluids such as serum and gastric washes have led to many studies of application in gastric cancer. This review focuses on the most common and important phenomenon of epigenetics, DNA methylation, in gastric cancer and illustrates the impact epigenetics has had on this field.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA Methylation , Epigenesis, Genetic , Neoplasm Proteins/genetics , Stomach Neoplasms/genetics , Cell Transformation, Neoplastic/pathology , CpG Islands , Histones/genetics , Histones/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic , Proto-Oncogene Mas , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Signal Transduction , Stomach Neoplasms/pathology , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
17.
Gene ; 532(1): 1-12, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-23246696

ABSTRACT

MicroRNAs (miRNAs), a family of small nonprotein-coding RNAs, play a critical role in posttranscriptional gene regulation by acting as adaptors for the miRNA-induced silencing complex to inhibit gene expression by targeting mRNAs for translational repression and/or cleavage. miR-155-5p and miR-155-3p are processed from the B-cell Integration Cluster (BIC) gene (now designated, MIR155 host gene or MIR155HG). MiR-155-5p is highly expressed in both activated B- and T-cells and in monocytes/macrophages. MiR-155-5p is one of the best characterized miRNAs and recent data indicate that miR-155-5p plays a critical role in various physiological and pathological processes such as hematopoietic lineage differentiation, immunity, inflammation, viral infections, cancer, cardiovascular disease, and Down syndrome. In this review we summarize the mechanisms by which MIR155HG expression can be regulated. Given that the pathologies mediated by miR-155-5p result from the over-expression of this miRNA it may be possible to therapeutically attenuate miR-155-5p levels in the treatment of several pathological processes.


Subject(s)
Cardiovascular Diseases/genetics , Gene Expression Regulation , Inflammation/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasms/genetics , Animals , B-Lymphocytes/physiology , Cell Differentiation/genetics , Down Syndrome/genetics , Female , Humans , Macrophages/physiology , Multigene Family , NF-kappa B/genetics , NF-kappa B/metabolism , T-Lymphocytes/physiology , Transcription Factor AP-1/metabolism
18.
Int J Mol Sci ; 13(3): 2783-2800, 2012.
Article in English | MEDLINE | ID: mdl-22489125

ABSTRACT

The human ERBB2 proto-oncogene is widely considered a key gene involved in human breast cancer onset and progression. Among spontaneous tumors, mammary tumors are the most frequent cause of cancer death in cats and second most frequent in humans. In fact, naturally occurring tumors in domestic animals, more particularly cat mammary tumors, have been proposed as a good model for human breast cancer, but critical genetic and molecular information is still scarce. The aims of this study include the analysis of the cat ERBB2 gene partial sequences (between exon 17 and 20) in order to characterize a normal and a mammary lesion heterogeneous populations. Cat genomic DNA was extracted from normal frozen samples (n = 16) and from frozen and formalin-fixed paraffin-embedded mammary lesion samples (n = 41). We amplified and sequenced two cat ERBB2 DNA fragments comprising exons 17 to 20. It was possible to identify five sequence variants and six haplotypes in the total population. Two sequence variants and two haplotypes show to be specific for cat mammary tumor samples. Bioinformatics analysis predicts that four of the sequence variants can produce alternative transcripts or activate cryptic splicing sites. Also, a possible association was identified between clinicopathological traits and the variant haplotypes. As far as we know, this is the first attempt to examine ERBB2 genetic variations in cat mammary genome and its possible association with the onset and progression of cat mammary tumors. The demonstration of a possible association between primary tumor size (one of the two most important prognostic factors) and the number of masses with the cat ERBB2 variant haplotypes reveal the importance of the analysis of this gene in veterinary medicine.


Subject(s)
Genes, erbB-2 , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Receptor, ErbB-2/genetics , Alternative Splicing/genetics , Animals , Base Sequence , Cats , Female , Genotype , Haplotypes/genetics , Humans , Molecular Sequence Data , Proto-Oncogene Mas , Sequence Analysis, DNA
19.
Article in Vietnamese | WPRIM (Western Pacific) | ID: wpr-2558

ABSTRACT

An analysis of the morphologic characteristics was performed in 21 patients diagnosed acute erythroblastic leukemia (M6). The result showed that 95.2% of them had nucleated red cell and blast in the peripheral blood. The percentage of nucleated red cell in the peripheral blood was 17.571+/-4.04. Reticulocytes was 0.5+/-0.11% in peripheral blood. (proerythroblast + basophilic erythroblast) was 32+/-8.4%. (Polychromatic erythroblast + orthochromatic erythroblast) was 30.238+/-8.86%.


Subject(s)
Leukemia, Erythroblastic, Acute , Bone Marrow , Diagnosis
20.
Leuk Lymphoma ; 1(2): 147-51, 1990.
Article in English | MEDLINE | ID: mdl-27463821

ABSTRACT

Early erythroblastic leukemia is a newly defined type of leukemia in which the blasts have the same characteristics as erythroid precursors at the level of CFU-E. The blasts are characterized by the presence of carbonic anhydrase I, CD 36 antigen, platelet peroxidase (PPO)-like activity, and ferritin-containing granules. Early erythroblastic leukemia appears to have characteristic clinical features; in the original report of nine cases, only one patient had typical de novo acute leukemia. We report here a case of early erythroblastic leukemia that presented! as de novo acute leukemia. The blasts from this patient had almost the same ultrastructural and phenotypical characteristics as those of the originally reported cases, even though our case was not examined for anti-carbonic anhydrase I antibodies. As a single marker, PPOl activity can no longer be considered specific for the megakaryocyte-platelet lineage, even though the significance of transient expression of PPO-like activity in immature erythroblastic cells at the level of CFU-E still remains to be clarified. When leukemic blasts show positivity for CD36 and negativity for megakaryocytic or monocytic markers, the diagnosis of early erythroblastic leukemia should be suspected and electron microscopical characteristics should be studied.

SELECTION OF CITATIONS
SEARCH DETAIL
...