Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Front Endocrinol (Lausanne) ; 15: 1384603, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38660513

RESUMEN

Background: Sex hormones play a critical role in sex differences and cardiovascular disease risk associated with metabolic syndrome (MS) and inflammation. However, the associations of sex hormone ratios with metabolic and inflammatory markers are unclear according to sex and age differences. We evaluated the associations of sex hormone ratios with MS and inflammation among males and females. Methods: A retrospective cross-sectional study was conducted by including all adults from the National Health and Nutrition Examination Survey cycles 2013-2016 and excluding any pregnant women, heart disease, diabetes, and those currently taking insulin. MS was defined using the National Cholesterol Education Program criteria and a high-sensitivity C-reactive protein (CRP) level>3 mg/L was defined as a high CRP. Measures of MS components and CRP concentrations were also analyzed. The primary exposures were testosterone to estradiol (excess androgen index), testosterone to sex hormone-binding globulin (free androgen index), and estradiol to sex hormone-binding globulin (free estradiol index). The adjusted associations were summarized with a relative risk (RR) and 95% confidence interval (CI). Results: This study included 9167 subjects with 4360 males and 4807 females. Increases in free estradiol index were positively associated with MS (RR=1.48; 95%CI: 1.39, 1.58; RR=1.31; 95%CI: 1.22, 1.40) and high CRP (RR=1.49; 95%CI: 1.25, 1.77; RR=1.26; 95%CI: 1.06, 1.50) in men with age<50 years and age≥50 years, respectively. Similarly, higher free estradiol index was also robustly associated with increased prevalence of MS (RR=1.22; 95%CI: 1.15, 1.28) and high CRP (RR=1.68; 95%CI: 1.48, 1.90) in women with age ≥50 years. Among women with age<50 years, a higher free androgen index was associated with MS (RR=1.34; 95%CI: 1.25, 1.42) and high CRP (RR=1.13; 95%CI: 1.02, 1.25). These associations were unchanged even after adjusting for all sex hormones. Conclusion: Free estradiol index was consistently and positively associated with MS and high CRP in males of all ages and older females. Free androgen index was positively associated with MS and high CRP in females with age<50 years.


Asunto(s)
Hormonas Esteroides Gonadales , Inflamación , Síndrome Metabólico , Encuestas Nutricionales , Humanos , Síndrome Metabólico/sangre , Síndrome Metabólico/epidemiología , Masculino , Femenino , Estudios Transversales , Adulto , Persona de Mediana Edad , Estudios Retrospectivos , Inflamación/sangre , Inflamación/epidemiología , Hormonas Esteroides Gonadales/sangre , Estados Unidos/epidemiología , Globulina de Unión a Hormona Sexual/metabolismo , Globulina de Unión a Hormona Sexual/análisis , Estradiol/sangre , Testosterona/sangre , Proteína C-Reactiva/metabolismo , Proteína C-Reactiva/análisis , Anciano , Biomarcadores/sangre
2.
Antioxidants (Basel) ; 12(10)2023 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-37891871

RESUMEN

Reactive oxygen species are frequently associated with various cancers including pancreatic ductal adenocarcinomas (PDACs). Superoxide dismutase 2 (SOD2) is an enzyme that plays an important role in reactive oxygen species (ROS) signaling. Investigating the molecular function and biological functions of SOD2 can help us develop new therapeutic options and uncover new biomarkers for PDAC diagnosis and prognosis. Here, we show that nimbolide (NB), a triterpene limonoid, effectively blocks the growth and metastasis of PDACs by suppressing the expression and activity of SOD2. To identify the role of SOD2 in NB-induced anticancer activity, we used RNA interference to silence and plasmid transfection to overexpress it. Silencing SOD2 significantly reduced the growth and metastatic characteristics like epithelial-to-mesenchymal transition, invasion, migration, and colony-forming capabilities of PDACs, and NB treatment further reduced these characteristics. Conversely, the overexpression of SOD2 enhanced these metastatic characteristics. ROS signaling has a strong feedback mechanism with the PI3K/Akt signaling pathway, which could be mediated through SOD2. Finally, NB treatment to SOD2-overexpressing PDAC xenografts resulted in significant inhibition of tumor growth and metastasis. Overall, this work suggests that NB, a natural and safe phytochemical that silences SOD2 to induce high levels of ROS generation, results in increased apoptosis and reduced growth and progression of PDACs. The role of SOD2 in regulating NB-induced ROS generation presents itself as a therapeutic option for PDACs.

3.
Vaccines (Basel) ; 11(9)2023 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-37766141

RESUMEN

While cancer immunotherapies have become central to treatment, challenges associated with the ability of tumors to evade the immune system remain significant obstacles. At the heart of this issue is the tumor immune microenvironment, the complex interplay of the tumor microenvironment and the immune response. Recent advances in mRNA cancer vaccines represent major progress towards overcoming some of the challenges posed by deleterious components of the tumor immune microenvironment. Indeed, major breakthroughs in mRNA vaccine technology, such as the use of replacement nucleotides and lipid nanoparticle delivery, led to the vital success of mRNA vaccine technology in fighting COVID-19. This has in turn generated massive additional interest and investment in the platform. In this review, we detail recent research in the nature of the tumor immune microenvironment and in mRNA cancer vaccines and discuss applications by which mRNA cancer vaccines, often in combination with various adjuvants, represent major areas of potential in overcoming tumor immune microenvironment-imposed obstacles. To this end, we also review current mRNA cancer vaccine clinical trials.

4.
Metabolites ; 12(7)2022 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-35888767

RESUMEN

Breast cancer is the most commonly diagnosed cancer in women worldwide. Major advances have been made towards breast cancer prevention and treatment. Unfortunately, the incidence of breast cancer is still increasing globally. Metabolomics is the field of science which studies all the metabolites in a cell, tissue, system, or organism. Metabolomics can provide information on dynamic changes occurring during cancer development and progression. The metabolites identified using cutting-edge metabolomics techniques will result in the identification of biomarkers for the early detection, diagnosis, and treatment of cancers. This review briefly introduces the metabolic changes in cancer with particular focus on breast cancer.

5.
Front Oncol ; 11: 710337, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34350123

RESUMEN

BACKGROUND: Increased expression of the progesterone receptor membrane component 1, a heme and progesterone binding protein, is frequently found in triple negative breast cancer tissue. The basis for the expression of PGRMC1 and its regulation on cellular signaling mechanisms remain largely unknown. Therefore, we aim to study microRNAs that target selective genes and mechanisms that are regulated by PGRMC1 in TNBCs. METHODS: To identify altered miRNAs, whole human miRNome profiling was performed following AG-205 treatment and PGRMC1 silencing. Network analysis identified miRNA target genes while KEGG, REACTOME and Gene ontology were used to explore altered signaling pathways, biological processes, and molecular functions. RESULTS: KEGG term pathway analysis revealed that upregulated miRNAs target specific genes that are involved in signaling pathways that play a major role in carcinogenesis. While multiple downregulated miRNAs are known oncogenes and have been previously demonstrated to be overexpressed in a variety of cancers. Overlapping miRNA target genes associated with KEGG term pathways were identified and overexpression/amplification of these genes was observed in invasive breast carcinoma tissue from TCGA. Further, the top two genes (CCND1 and YWHAZ) which are highly genetically altered are also associated with poorer overall survival. CONCLUSIONS: Thus, our data demonstrates that therapeutic targeting of PGRMC1 in aggressive breast cancers leads to the activation of miRNAs that target overexpressed genes and deactivation of miRNAs that have oncogenic potential.

6.
Cancers (Basel) ; 13(13)2021 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-34206988

RESUMEN

Early full-term pregnancy is known to reduce the lifetime risk of breast cancer. Although the phenomenon of parity-induced protection is well-established, the physiological mechanisms involved in this protection are not clear. Earlier reports have shown that pregnancy results in alterations of hormone levels. How pregnancy affects hypothalamic hormones and how the change, if any, influences breast cancer is not well understood. Seven-week-old female Lewis rats were given N-methyl-N-nitrosourea. Two weeks post carcinogen exposure, a set of females were housed with males to generate the parous rats and another set of rats served as the nulliparous controls. Mammary tumorigenesis was assessed for 9 months. Hypothalamic and pituitary levels of hormones were measured at various timepoints. Further, animals were also challenged with growth hormone and prolactin secretagogues to test the effect of pregnancy on the hypothalamic-pituitary hormonal axis. Persistent alterations in the level of growth hormone-releasing hormone, thyrotropin releasing hormone, dopamine, and somatostatin in the hypothalamus of parous animals was observed. Further, we also observed that pregnancy had a significant effect on the pituitary gland and its response to growth hormone and prolactin secretagogues. Our studies using the rodent model system demonstrate that pregnancy could be reducing the risk of breast cancer by persistently altering the hypothalamic-pituitary axis, which could have implications for breast cancers in humans as well.

7.
Lab Invest ; 101(6): 733-744, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33903732

RESUMEN

Progesterone (P4) and estradiol (E2) have been shown to stimulate and regulate breast cancer proliferation via classical nuclear receptor signaling through progesterone receptor (PR) and estrogen receptor α (ERα), respectively. However, the basis of communication between PR/ERα and membrane receptors remains largely unknown. Here, we aim to identify classical and nonclassical endocrine signaling mechanisms that can alter cell proliferation through a possible crosstalk between PR, ERα, and progesterone receptor membrane component 1 (PGRMC1), a membrane receptor frequently observed in breast cancer cells. While P4 and E2 treatment increased cell proliferation of ER+/PR+/PGRMC1 overexpressing breast cancer cells, silencing ERα and PR or treatment with selective estrogen receptor modulator (SERM) tamoxifen, or (PR-antagonist) RU-486 decreased cell proliferation. All four treatments rapidly altered PGRMC1 mRNA levels and protein expression. Furthermore, P4 and E2 treatments rapidly activated EGFR a known interacting partner of PGRMC1 and its downstream signaling. Interestingly, downregulation of ERα by tamoxifen and ERα silencing decreased the expression levels of PGRMC1 with no repercussions to PR expression. Strikingly PGRMC1 silencing decreased ERα expression irrespective of PR. METABRIC and TCGA datasets further demonstrated that PGRMC1 expression was comparable to that of ERα in Luminal A and B breast cancers. Targeting of PR, ERα, and PGRMC1 confirmed that a crosstalk between classical and nonclassical signaling mechanisms exists in ER+ breast cancer cells that could enhance the growth of ER+/PR+/PGRMC1 overexpressing tumors.


Asunto(s)
Neoplasias de la Mama/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrógeno/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de Progesterona/metabolismo , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos
8.
Mol Ther Oncolytics ; 20: 596-606, 2021 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-33768141

RESUMEN

Triple-negative breast cancers (TNBCs) are aggressive cancers, which currently do not have effective treatment options. Migration and establishment of metastatic colonies require dynamic cytoskeletal modifications characterized by polymerization and depolymerization of actin. Studies have demonstrated a direct molecular link between the integrin-focal adhesion kinase (FAK) pathway and cytoskeletal modifications. Nimbolide, a major bioactive compound present in neem leaves, shows promising anti-cancer effect on various cancers. In this study, we have demonstrated the growth and metastasis inhibitory potential of nimbolide on TNBC cells. Nimbolide inhibited cell proliferation, migratory, and invasive abilities of TNBC cells and also changed the shape of MDA-MB-231 cells, which is correlated with cytoskeletal changes including actin depolymerization. Furthermore, analysis revealed that integrins αV and ß3, ILK, FAK, and PAK levels were downregulated by nimbolide. Even in cells where Rac1/Cdc42 was constitutively activated, nimbolide inhibited the formation of filopodial structures. Immunofluorescence analysis of phosphorylated p21 activated kinase (pPAK) showed reduced expression in nimbolide-treated cells. Nimbolide significantly reduced the metastatic colony formation in lung, liver, and brain of athymic nude mice. In conclusion, our data demonstrate that nimbolide inhibits TNBC by altering the integrin and FAK signaling pathway.

9.
Cell Death Dis ; 11(12): 1073, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33323928

RESUMEN

The major challenge in chemotherapy lies in the gain of therapeutic resistance properties of cancer cells. The relatively small fraction of chemo-resistant cancer cells outgrows and are responsible for tumor relapse, with acquired invasiveness and stemness. We demonstrate that zinc-finger MYND type-8 (ZMYND8), a putative chromatin reader, suppresses stemness, drug resistance, and tumor-promoting genes, which are hallmarks of cancer. Reinstating ZMYND8 suppresses chemotherapeutic drug doxorubicin-induced tumorigenic potential (at a sublethal dose) and drug resistance, thereby resetting the transcriptional program of cells to the epithelial state. The ability of ZMYND8 to chemo-sensitize doxorubicin-treated metastatic breast cancer cells by downregulating tumor-associated genes was further confirmed by transcriptome analysis. Interestingly, we observed that ZMYND8 overexpression in doxorubicin-treated cells stimulated those involved in a good prognosis in breast cancer. Consistently, sensitizing the cancer cells with ZMYND8 followed by doxorubicin treatment led to tumor regression in vivo and revert back the phenotypes associated with drug resistance and stemness. Intriguingly, ZMYND8 modulates the bivalent or poised oncogenes through its association with KDM5C and EZH2, thereby chemo-sensitizing the cells to chemotherapy for better disease-free survival. Collectively, our findings indicate that poised chromatin is instrumental for the acquisition of chemo-resistance by cancer cells and propose ZMYND8 as a suitable epigenetic tool that can re-sensitize the chemo-refractory breast carcinoma.


Asunto(s)
Oncogenes , Proteínas Supresoras de Tumor/metabolismo , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Epigénesis Genética/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genoma Humano , Histona Demetilasas/metabolismo , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Resultado del Tratamiento , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
10.
Sci Rep ; 10(1): 20225, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33214606

RESUMEN

Hepatocyte nuclear factor 1 homeobox alpha (HNF1α) is a transcription factor involved in endodermal organogenesis and pancreatic precursor cell differentiation and development. Earlier studies have reported a role for HNF1α in pancreatic ductal adenocarcinoma (PDAC) but it is controversial. The mechanism by which it impacts PDAC is yet to be explored in depth. In this study, using the online databases we observed that HNF1α is upregulated in PDAC, which was also confirmed by our immunohistochemical analysis of PDAC tissue microarray. Silencing HNF1α reduced the proliferative, migratory, invasive and colony forming capabilities of pancreatic cancer cells. Key markers involved in these processes (pPI3K, pAKT, pERK, Bcl2, Zeb, Snail, Slug) were significantly changed in response to alterations in HNF1α expression. On the other hand, overexpression of HNF1α did not induce any significant change in the aggressiveness of pancreatic cancer cells. Our results demonstrate that reduced expression of HNF1α leads to inhibition of pancreatic cancer growth and progression, which indicates that it could be a potential oncogene and target for PDAC.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Metástasis de la Neoplasia/patología , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Bases de Datos Factuales , Progresión de la Enfermedad , Humanos , Neoplasias Pancreáticas/patología
11.
Cancer Control ; 27(1): 1073274820951780, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32959673

RESUMEN

Introduction : Hispanic women residing along the US-Mexico border have the highest cervical cancer incidence rates in the US. Genital human papillomavirus (HPV) is the major causative agent, but more information is needed about the prevalence and distribution of genital HPV subtypes in this high-risk population. Methods : A population-based cross-sectional study of uninsured Hispanic women along the US-Mexico border was conducted and participants had their cervical specimens undergo DNA extraction followed by HPV genotype testing using the Linear Assay from Roche® Diagnostics, to identify 37 genital HPV subtypes. Results : Among the 585 women aged 21-65 years, 584 self-identfied as Hispanic. Any HPV subtype prevalence was 53.2% (95% CI: 49.0%-57.3%) and of these 52% (i.e. 27.5% of the total) had single infections and 48% (i.e. 25.6% of the total) had multiple infections. High-risk HPV prevalence was 15.6% (95% CI: 24-31.3%). The mean number of subtypes among those testing positive was 2.1 (SD 1.6). The prevalence of any HPV and high-risk HPV showed a U shaped pattern with age; and prevalence of 16/18 and non-16/18 high-risk subtypes (e.g. 31, 33, 35, 39, 45, 51, 52, 58); also varied with age. Forty-one percent of high-risk HPV occurrences were of a subtype not covered by the current nonavalent HPV vaccine. Discussion : Our findings suggest a different high-risk HPV subtype pattern and age distribution among Hispanic women in the USA, which could have implications for future cervical cancer prevention strategies.


Asunto(s)
Detección Precoz del Cáncer/métodos , Hispánicos o Latinos/estadística & datos numéricos , Papillomaviridae/clasificación , Papillomaviridae/genética , Infecciones por Papillomavirus/complicaciones , Neoplasias del Cuello Uterino/diagnóstico , Cuello del Útero , Estudios de Cohortes , Estudios Transversales , Femenino , Estudios de Seguimiento , Genotipo , Humanos , México/epidemiología , Persona de Mediana Edad , Infecciones por Papillomavirus/virología , Prevalencia , Pronóstico , Neoplasias del Cuello Uterino/epidemiología , Neoplasias del Cuello Uterino/virología , Frotis Vaginal
12.
Cancers (Basel) ; 12(9)2020 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-32867363

RESUMEN

Much emphasis is placed on estrogen (E2) and estrogen receptor (ER) signaling as most research is focused on understanding E2 and ER's ability to enhance proliferative signals in breast cancers. Progesterone (P4) is important for normal mammary gland development, function and menstrual control. However, P4 and its receptors (PRs) in breast cancer etiology continue to be understudied and its role in breast cancer remains controversial. The Women's Health Initiative (WHI) clinical trial clearly demonstrated the importance of progestogens in breast cancer development. P4 has historically been associated with classical-signaling through nuclear receptors, however non-classical P4 signaling via membrane receptors has been described. Progestogens have the ability to bind to nuclear and membrane receptors and studies have demonstrated that both can promote breast cancer cell proliferation and breast tumor growth. In this review, we attempt to understand the classical and non-classical signaling role of P4 in breast cancers because both nuclear and membrane receptors could become viable therapeutic options for breast cancer patients.

13.
Br J Cancer ; 123(8): 1326-1335, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32704174

RESUMEN

BACKGROUND: Increased expression of the progesterone receptor membrane component 1 (PGRMC1) has been linked to multiple cancers, including breast cancer. Despite being a regulatory receptor and a potential therapeutic target, the oncogenic potential of PGRMC1 has not been studied. METHODS: The impact of PGRMC1 on breast cancer growth and progression was studied following chemical inhibition and alteration of PGRMC1 expression, and evaluated by using online-based gene expression datasets of human breast cancer tissue. MTS, flow cytometry, qPCR, Western blotting, confocal microscopy and phosphoproteome analysis were performed. RESULTS: We observed higher PGRMC1 levels in both ER-positive ZR-75-1 and TNBC MDA-MB-468 cells. Both chemical inhibition and silencing decreased cell proliferation, induced cell-cycle arrest, promoted apoptosis and reduced the migratory and invasive capabilities of ZR-75-1 and MDA-MB-468 cells. Further, phosphoproteome analysis demonstrated an overall decrease in activation of proteins involved in PI3K/AKT/mTOR and EGFR signalling pathways. In contrast, overexpression of PGRMC1 in non-malignant MCF10A cells resulted in increased cell proliferation, and enhanced activity of PI3K/AKT/mTOR and EGFR signalling pathways. CONCLUSIONS: Our data demonstrate that PGRMC1 plays a prominent role in regulating the growth of cancer cells by altering the PI3K/AKT/mTOR and EGFR signalling mechanisms in both ER-positive and TNBC cells.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas de la Membrana/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Fosfoproteínas/metabolismo , Proteoma , Proteínas Proto-Oncogénicas c-akt/fisiología , Receptores de Progesterona/fisiología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular , Receptores ErbB/fisiología , Femenino , Humanos , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/fisiología
14.
Cancers (Basel) ; 12(6)2020 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-32532008

RESUMEN

Hypoxanthine phosphoribosyl transferase 1 (HPRT1) is traditionally believed to be a housekeeping gene; however, recent reports suggest that it is upregulated in several cancers and is associated with clinical outcomes. HPRT1 is located on chromosome X and encodes the HPRT enzyme, which functions in recycling nucleotides to supply for DNA and RNA synthesis in actively dividing cells. Here, we used transcriptomic analyses to interrogate its expression across all known cancer types and elucidated its role in regulating gene expression in breast cancer. We observed elevated HPRT1 RNA levels in malignant tissues when compared to normal controls, indicating its potential as a diagnostic and prognostic marker. Further, in breast cancer, the subtype-specific analysis showed that its expression was highest in basal and triple-negative breast cancer, and HPRT1 knockdown in breast cancer cells suggested that HPRT1 positively regulates genes related to cancer pathways. Collectively, our results essentially highlight the importance of and change the way in which HPRT1's function is studied in biology, warranting careful examination of its role in cancer.

15.
Cancer Causes Control ; 31(6): 569-581, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32300943

RESUMEN

PURPOSE: Hepatitis C virus (HCV) infection is the prevalent risk factor for chronic hepatitis, cirrhosis, and hepatocellular carcinoma (HCC) worldwide. The association between metabolic syndrome (MetS) and HCV infection has not been studied effectively, particularly among different ethnic/racial groups in the US. METHODS: A retrospective cross-sectional study was conducted using data from the National Health and Nutrition Examination Survey (1999-2014). Unadjusted and adjusted associations were summarized using the prevalence ratio (PR) and 95% confidence interval (CI) after exploring possible interactions. RESULTS: In the overall population, MetS was significantly associated with HCV infection with an interaction of age. After adjusting for all potential confounders, MetS was found to be significantly associated with HCV among non-obese and younger adults of age less than 60 years (PR 1.67, 95% CI 1.21-2.30, p = 0.002). MetS was also associated with an increased prevalence of HCV in each racial/ethnic group, while the association was strongly modified by age and obesity status of the subjects in different ethnic/racial groups. CONCLUSIONS: MetS or its components are associated with an increased prevalence of HCV in some sub-populations of all ethnic/racial groups in the US. A better understanding of the pathophysiology of MetS associated with HCV is important as MetS may have a role in HCV infection treatment outcomes.


Asunto(s)
Hepatitis C , Síndrome Metabólico , Hepacivirus , Hepatitis C/complicaciones , Hepatitis C/epidemiología , Humanos , Síndrome Metabólico/complicaciones , Síndrome Metabólico/epidemiología , Persona de Mediana Edad , Obesidad , Grupos Raciales/estadística & datos numéricos , Estados Unidos/epidemiología
16.
Adv Clin Chem ; 95: 105-147, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32122521

RESUMEN

Long noncoding RNAs (lncRNAs) have recently gained considerable attention as key players in biological regulation; however, the mechanisms by which lncRNAs govern various disease processes remain mysterious and are just beginning to be understood. The ease of next-generation sequencing technologies has led to an explosion of genomic information, especially for the lncRNA class of noncoding RNAs. LncRNAs exhibit the characteristics of mRNAs, such as polyadenylation, 5' methyl capping, RNA polymerase II-dependent transcription, and splicing. These transcripts comprise more than 200 nucleotides (nt) and are not translated into proteins. Directed interrogation of annotated lncRNAs from RNA-Seq datasets has revealed dramatic differences in their expression, largely driven by alterations in transcription, the cell cycle, and RNA metabolism. The fact that lncRNAs are expressed cell- and tissue-specifically makes them excellent biomarkers for ongoing biological events. Notably, lncRNAs are differentially expressed in several cancers and show a distinct association with clinical outcomes. Novel methods and strategies are being developed to study lncRNA function and will provide researchers with the tools and opportunities to develop lncRNA-based therapeutics for cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , ARN Largo no Codificante/antagonistas & inhibidores , Animales , Humanos , Neoplasias/genética , Neoplasias/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
17.
Radiology ; 292(3): 552-561, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31237494

RESUMEN

BackgroundThe higher level of background parenchymal enhancement (BPE) at breast MRI has the potential for early detection and prediction of the risk of breast cancer. However, conflicting findings have been reported about the association between the level of BPE at breast MRI and the presence of breast cancer.PurposeTo evaluate the association between qualitative and quantitative BPE at dynamic contrast material-enhanced MRI and breast cancer among populations with average risk and high risk separately.Materials and MethodsA retrospective meta-analysis of observational studies comparing either qualitative or quantitative assessments of BPE in women with and women without breast cancer was performed for studies published through July 2018. Pooled odds ratios (ORs) or standardized mean differences and 95% confidence intervals (CIs) were estimated by using DerSimonian-Laird random-effects models. The heterogeneity across the studies was measured by using the statistic I 2. Sensitivity analyses were conducted to test this association according to different study characteristics. P values less than or equal to 5% were considered to indicate statistically significant results.ResultsEighteen studies comprising 1910 women with breast cancer and 2541 control participants were included in the analysis. Among women with high risk, at least moderate BPE (OR, 1.6; 95% CI: 1.0, 2.6; P = .04) or at least mild BPE (OR, 2.1; 95% CI: 1.5, 3.0; P < .001) was associated with higher odds of breast cancer. Furthermore, women with breast cancer showed a higher average BPE percentage compared with control participants with high risk (standardized mean difference, 0.5; 95% CI: 0.2, 0.9; P = .001). No association was observed between at least mild BPE level (P = .15) or at least moderate BPE level (P = .38) and the presence of breast cancer among the population with average risk.ConclusionA higher level of background parenchymal enhancement measured at breast MRI is associated with the presence of breast cancer in women with high risk, but not in women with average risk.© RSNA, 2019Online supplemental material is available for this article.See also the editorial by Mann and Pinker in this issue.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Medios de Contraste , Aumento de la Imagen/métodos , Imagen por Resonancia Magnética/métodos , Adulto , Mama/diagnóstico por imagen , Femenino , Humanos , Estudios Observacionales como Asunto , Reproducibilidad de los Resultados
18.
Exp Mol Med ; 51(1): 1-13, 2019 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-30617282

RESUMEN

Growth hormone receptor (GHR) plays a vital role in breast cancer chemoresistance and metastasis but the mechanism is not fully understood. We determined if GHR could be a potential therapeutic target for estrogen receptor negative (ER-ve) breast cancer, which are highly chemoresistant and metastatic. GHR was stably knocked down in ER-ve breast cancer cells and its effect on cell proliferation, metastatic behavior, and chemosensitivity to docetaxel (DT) was assessed. Microarray analysis was performed to identify potential GHR downstream targets involved in chemoresistance. GHR and ATP-binding cassette sub-family G member 2 (ABCG2) overexpression and knockdown studies were performed to investigate the mechanism of GHR-induced chemoresistance. Patient-derived xenografts was used to study the effect of GHR and ABCG2. Immunohistochemical data was used to determine the correlation between GHR, pAKT, pmTOR, and ABCG2 expressions. GHR silencing drastically reduced the chemoresistant and metastatic behavior of ER-ve breast cancer cells and also inhibited AKT/mTOR pathway. In contrast, activation, or overexpression of GHR increased chemoresistance and metastasis by increasing the expression and promoter activity, of ABCG2. Inhibition of JAK2/STAT5 signaling repressed GHR-induced ABCG2 promoter activity and expression. Further, ABCG2 knockdown significantly increased the chemosensitivity. Finally, patient-derived xenograft studies revealed the role of GHR in chemoresistance. Overall, these findings demonstrate that targeting GHR could be a novel therapeutic approach to overcome chemoresistance and associated metastasis in aggressive ER-ve breast cancers.


Asunto(s)
Silenciador del Gen , Neoplasias Mamarias Experimentales/terapia , Tratamiento con ARN de Interferencia/métodos , Receptores de Somatotropina/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Animales , Línea Celular , Células Cultivadas , Femenino , Humanos , Janus Quinasa 2/metabolismo , Células MCF-7 , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Desnudos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Estrógenos/genética , Receptores de Somatotropina/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
19.
Oncogenesis ; 7(7): 52, 2018 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-29976975

RESUMEN

IGF-1R signaling controls various vital cellular functions and this signaling is deregulated in many cancers, including pancreatic cancer. Several efforts have mainly focused on inhibiting the IGF-1R signaling cascade. The outcomes of these focused preclinical studies have been positive, whereas clinical trials of IGF-1R inhibitors in pancreatic cancer have failed, raising the questions about this therapeutic approach. This necessitates a better understanding of the role of IGF-1R signaling in pancreatic cancer. We investigated the impact of IGF-1R signaling on crucial transcription factors and identified the FOXC1 as one of the crucial regulator of IGF-1R signaling. We employed genetic approaches to overexpress and silence FOXC1 in pancreatic cancer cells. Our results demonstrate that IGF-1R and FOXC1 seem to positively regulate each other. Further, FOXC1 increased the metastatic abilities of pancreatic cancer cells by enhancing cell proliferation, migration, invasion, epithelial-to-mesenchymal transition, and angiogenesis. The data from xenograft experiments further established the importance of FOXC1 in pancreatic tumorigenesis. In conclusion, FOXC1 is a potent oncogenic transcription factor, which promotes pancreatic cancer growth and metastasis. Thus, targeting FOXC1 could be a potential therapeutic strategy against pancreatic cancer.

20.
Prog Mol Biol Transl Sci ; 151: 137-176, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29096892

RESUMEN

Decades of cancer research have led to substantial progress in the treatment of primary breast cancers. Despite of the advancement in this field, treating a metastatic disease has remained a mammoth task. One of the possible theories explaining metastatic disease involves the cancer stem cells (CSCs). CSCs have been shown to be an integral part of solid tumors. The process of metastasis involves the fine orchestration between CSCs and other microenvironmental factors. This chapter will provide an overview about the process of metastasis and the interactive role of CSCs with the components of the microenvironment in each phase of the metastatic cascade.


Asunto(s)
Metástasis de la Neoplasia/patología , Células Madre Neoplásicas/patología , Transición Epitelial-Mesenquimal , Humanos , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...