Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Int J Mol Sci ; 22(15)2021 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-34360875

RESUMEN

Single prostate stem cells can generate stem and progenitor cells to form prostaspheres in 3D culture. Using a prostasphere-based label retention assay, we recently identified keratin 13 (KRT13)-enriched prostate stem cells at single-cell resolution, distinguishing them from daughter progenitors. Herein, we characterized the epithelial cell lineage hierarchy in prostaspheres using single-cell RNA-seq analysis. Keratin profiling revealed three clusters of label-retaining prostate stem cells; cluster I represents quiescent stem cells (PSCA, CD36, SPINK1, and KRT13/23/80/78/4 enriched), while clusters II and III represent active stem and bipotent progenitor cells (KRT16/17/6 enriched). Gene set enrichment analysis revealed enrichment of stem and cancer-related pathways in cluster I. In non-label-retaining daughter progenitor cells, three clusters were identified; cluster IV represents basal progenitors (KRT5/14/6/16 enriched), while clusters V and VI represent early and late-stage luminal progenitors, respectively (KRT8/18/10 enriched). Furthermore, MetaCore analysis showed enrichment of the "cytoskeleton remodeling-keratin filaments" pathway in cancer stem-like cells from human prostate cancer specimens. Along with common keratins (KRT13/23/80/78/4) in normal stem cells, unique keratins (KRT10/19/6C/16) were enriched in cancer stem-like cells. Clarification of these keratin profiles in human prostate stem cell lineage hierarchy and cancer stem-like cells can facilitate the identification and therapeutic targeting of prostate cancer stem-like cells.


Asunto(s)
Queratinas/metabolismo , Células Madre Neoplásicas , Neoplasias de la Próstata , ARN/metabolismo , Adulto , Células Cultivadas , Humanos , Masculino , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Cultivo Primario de Células , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Análisis de la Célula Individual , Adulto Joven
2.
Prostate ; 79(10): 1166-1179, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31135075

RESUMEN

BACKGROUND: The progression of castration-resistant prostate cancer (CRPC) still relies on the function of androgen receptor (AR), achieved by evolving mechanisms to reactivate AR signaling under hormonal therapy. Histone deacetylase inhibitors (HDACis) disrupt cytoplasmic AR chaperone heat shock protein 90 (Hsp90) via HDAC6 inhibition, leading to AR degradation and growth suppression of prostate cancer (PCa) cells. However, current HDACis are not effective in clinical trials treating CRPC. METHODS: We designed hybrid molecules containing partial chemical scaffolds of AR antagonist enzalutamide (Enz) and HDACi suberoylanilide hydroxamic acid (SAHA) as new anti-PCa agents. We previously demonstrated that Enz-HDACi hybrid drug 2-75 targets both AR and Hsp90, which inhibits the growth of Enz-resistant C4-2 cells. In the current study, we further investigate the molecular and cellular actions of 2-75 and test its anti-PCa effects in vivo. RESULTS: Compared with Enz, 2-75 had greater AR antagonistic effects by decreasing the stability, transcriptional activity, and nuclear translocation of intracellular AR. In addition to inhibition of full-length AR (FL AR), 2-75 downregulated the AR-V7 variant in multiple PCa cell lines. Mechanistic studies indicated that the AR affinity of 2-75 retains the drug in the cytoplasm of AR + PCa cells and further directs 2-75 to the AR-associated protein complex, which permits localized effects on AR-associated Hsp90. Further, unlike pan-HDACi SAHA, the cytoplasm-retaining property allows 2-75 to significantly inhibit cytoplasmic HDAC6 with limited impact on nuclear HDACs. These selective cytoplasmic actions of 2-75 overcome the unfavorable resistance and toxicity properties associated with classical AR antagonists, HDACis, and Hsp90 inhibitors. Finally, 2-75 showed greater antitumor activities than Enz in vivo on SQ xenografts derived from LNCaP cells. CONCLUSIONS: Novel therapeutic strategy using newly designed 2-75 and related AR antagonist-HDACi hybrid drugs has great potential for effective treatment of CRPC.


Asunto(s)
Antineoplásicos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/uso terapéutico , Feniltiohidantoína/análogos & derivados , Próstata/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Antineoplásicos/farmacología , Benzamidas , Línea Celular Tumoral , Regulación hacia Abajo , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Nitrilos , Feniltiohidantoína/farmacología , Feniltiohidantoína/uso terapéutico , Próstata/patología , Neoplasias de la Próstata/patología
3.
Exp Eye Res ; 121: 102-13, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24509257

RESUMEN

Mouse retinal degeneration models have been investigated for many years in the hope of understanding the mechanism of photoreceptor cell death. N -methyl- N -nitrosourea (MNU) has been previously shown to induce outer retinal degeneration in mice. After MNU was intraperitoneally injected in C57/BL mice, we observed a gradual decrease in the outer nuclear layer (ONL) thickness associated with photoreceptor outer segment loss, bipolar cell dendritic retraction and reactive gliosis. Reactive gliosis was confirmed by increased GFAP protein levels. More serious damage to the central retina as opposed to the peripheral retina was found in the MNU-induced retinal degeneration model. Retinal ganglion cells (RGC) appear to be spared for at least two months after MNU treatment. Following retinal vessel labelling, we observed vascular complexes in the distal vessels, indicating retinal vessel damage. In the remnant retinal photoreceptor of the MNU-treated mouse, concentrated colouring nuclei were detected by electron microscopy, together with the loss of mitochondria and displaced remnant synaptic ribbons in the photoreceptor. We also observed decreased mitochondrial protein levels and increased amounts of nitrosylation/nitration in the photoreceptors. The mechanism of MNU-induced apoptosis may result from oxidative stress or the loss of retinal blood supply. MNU-induced mouse retinal degeneration in the outer retina is a useful animal model for photoreceptor degeneration diseases, such as age-related macular degeneration (AMD) and retinitis pigmentosa (RP).


Asunto(s)
Alquilantes/toxicidad , Modelos Animales de Enfermedad , Metilnitrosourea/toxicidad , Degeneración Retiniana/inducido químicamente , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Western Blotting , Proteínas del Ojo/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Mitocondrias/efectos de los fármacos , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Células Fotorreceptoras de Vertebrados/ultraestructura , Degeneración Retiniana/patología , Neuronas Retinianas/efectos de los fármacos , Neuronas Retinianas/ultraestructura
4.
Am J Clin Exp Urol ; 10(6): 377-389, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36636689

RESUMEN

Cancer stem cells (CSCs) are resistant to conventional cancer therapies, permitting the repopulation of new tumor growth and driving disease progression. Models for testing prostate CSC-propagated tumor growth are presently limited yet necessary for therapeutic advancement. Utilizing the congenic nontumorigenic NRP152 and tumorigenic NRP154 rat prostate epithelial cell lines, the present study investigated the self-renewal, differentiation, and regenerative abilities of prostate stem/progenitor cells and developed a CSC-based PCa model. NRP154 cells expressed reduced levels of tumor suppressor caveolin-1 and increased p-Src as compared to NRP152 cells. Gene knockdown of caveolin-1 in NRP152 cells upregulated p-Src, implicating their role as potential oncogenic mediators in NRP154 cells. A FACS-based Hoechst exclusion assay revealed a side population of stem-like cells (0.1%) in both NRP152 and NRP154 cell lines. Using a 3D Matrigel culture system, stem cells from both cell lines established prostaspheres at a 0.1% efficiency through asymmetric self-renewal and rapid proliferation of daughter progenitor cells. Spheres derived from both cell lines contained CD117+ and CD133+ stem cell subpopulations and basal progenitor cell subpopulations (p63+ and CK5+) but were negative for luminal cell CK8 markers at day 7. While some NRP152 sphere cells were androgen receptor (AR) positive at this timepoint, NRP154 cells were AR- up to 30 days of 3D culture. The regenerative capacity of the stem/progenitor cells was demonstrated by in vivo tissue recombination with urogenital sinus mesenchyme (UGM) and renal grafting in nude mice. While stem/progenitor cells from NRP152 spheroids generated normal prostate structures, CSCs and progeny cells from NRP154 tumoroids generated tumor tissues that were characterized by immunohistochemistry. Atypical hyperplasia and prostatic intraepithelial neoplasia (PIN) lesions progressed to adenocarcinoma with kidney invasion over 4 months. This provides clear evidence that prostate CSCs can repopulate new tumor growth outside the prostate gland that rapidly progresses to poorly differentiated adenocarcinoma with invasive capabilities. The dual in vitro/in vivo CSC model system presented herein provides a novel platform for screening therapeutic agents that target prostate CSCs for effective combined treatment protocols for local and advanced disease stages.

5.
Biochem Pharmacol ; 197: 114902, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34968493

RESUMEN

Per- and polyfluorinated alkyl substances (PFAS) are a large family of widely used synthetic chemicals that are environmentally and biologically persistent and present in most individuals. Chronic PFAS exposure have been linked to increased prostate cancer risk in occupational settings, however, underlying mechanisms have not been interrogated. Herein we examined exposure of normal human prostate stem-progenitor cells (SPCs) to 10 nM PFOA or PFOS using serial passage of prostasphere cultures. Exposure to either PFAS for 3-4 weeks increased spheroid numbers and size indicative of elevated stem cell self-renewal and progenitor cell proliferation. Transcriptome analysis using single-cell RNA sequencing (scRNA-seq) showed 1) SPC expression of PPARs and RXRs able to mediate PFAS effects, 2) the emergence of a new cell cluster of aberrantly differentiated luminal progenitor cells upon PFOS/PFOA exposure, and 3) enrichment of cancer-associated signaling pathways. Metabolomic analysis of PFAS-exposed prostaspheres revealed increased glycolytic pathways including the Warburg effect as well as strong enrichment of serine and glycine metabolism which may promote a pre-malignant SPC fate. Finally, growth of in vivo xenografts of tumorigenic RWPE-2 human prostate cells, shown to contain cancer stem-like cells, was markedly enhanced by daily PFOS feeding to nude mice hosts. Together, these findings are the first to identify human prostate SPCs as direct PFAS targets with resultant reprogrammed transcriptomes and metabolomes that augment a preneoplastic state and may contribute to an elevated prostate cancer risk with chronic exposures.


Asunto(s)
Contaminantes Ambientales/toxicidad , Fluorocarburos/toxicidad , Próstata/efectos de los fármacos , Próstata/patología , Células Madre/efectos de los fármacos , Células Madre/patología , Animales , Humanos , Masculino , Ratones , Ratones Desnudos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Adulto Joven
6.
Chem Biol Drug Des ; 97(5): 1059-1078, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33470049

RESUMEN

Sustained androgen receptor (AR) signaling and apoptosis evasion are among the main hurdles of castration-resistant prostate cancer (CRPC) treatment. We designed and synthesized isothiocyanate (ITC)-containing hybrid AR antagonist (ITC-ARi) and rationally combined ITC-ARi with GSH synthesis inhibitor buthionine sulfoximine (BSO) to efficiently downregulate AR/AR splice variant and induce ferroptosis in CRPC cells. The representative ITC-ARi 13 is an AR ligand that contains an N-acetyl cysteine-masked ITC moiety and gradually releases parental unconjugated ITC 12b in aqueous solution. The in vitro anti-PCa activities of 13, such as growth inhibition and AR downregulation, are significantly enhanced when combined with BSO. The drug combination caused notable lipid peroxidation and the cell viability was effectively rescued by iron chelator, antioxidants or the inhibitor of heme oxygenase-1, supporting the induction of ferroptosis. 13 and BSO cooperatively downregulate AR and induce ferroptosis likely through increasing the accessibility of 13/12b to cellular targets, escalating free intracellular ferrous iron and attenuating GSH-centered cellular defense and adaptation. Further studies on the combination of ITC-ARi and GSH synthesis inhibitor could result in a new modality against CRPC.


Asunto(s)
Antagonistas de Receptores Androgénicos/farmacología , Regulación hacia Abajo/efectos de los fármacos , Diseño de Fármacos , Ferroptosis/efectos de los fármacos , Isotiocianatos/química , Antagonistas de Receptores Androgénicos/síntesis química , Antagonistas de Receptores Androgénicos/metabolismo , Antagonistas de Receptores Androgénicos/uso terapéutico , Sitios de Unión , Butionina Sulfoximina/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Glutatión/metabolismo , Humanos , Masculino , Simulación del Acoplamiento Molecular , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/patología , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores Androgénicos/química , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Activación Transcripcional/efectos de los fármacos
7.
Environ Health Perspect ; 128(6): 67008, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32525701

RESUMEN

BACKGROUND: Inorganic arsenic (iAs) is an environmental toxicant associated with an increased risk of prostate cancer in chronically exposed populations worldwide. However, the biological mechanisms underlying iAs-induced prostate carcinogenesis remain unclear. OBJECTIVES: We studied how iAs affects normal human prostate stem-progenitor cells (PrSPCs) and drives transformation and interrogated the molecular mechanisms involved. METHODS: PrSPCs were enriched by spheroid culture from normal human primary or immortalized prostate epithelial cells, and their differentiation capability was evaluated by organoid culture. Microarray analysis was conducted to identify iAs-dysregulated genes, and lentiviral infection was used for stable manipulation of identified genes. Soft agar colony growth assays were applied to examine iAs-induced transformation. For in vivo study, PrSPCs mixed with rat urogenital sinus mesenchyme were grafted under the renal capsule of nude mice to generate prostatelike tissues, and mice were exposed to 5 ppm (∼65µM) iAs in drinking water for 3 months. RESULTS: Low-dose iAs (1µM) disturbed PrSPC homeostasis in vitro, leading to increased self-renewal and suppressed differentiation. Transcriptomic analysis indicated that iAs activated oncogenic pathways in PrSPCs, including the KEAP1-NRF2 pathway. Further, iAs-exposed proliferative progenitor cells exhibited NRF2 pathway activation that was sustained in their progeny cells. Knockdown of NRF2 inhibited spheroid formation by driving PrSPC differentiation, whereas its activation enhanced spheroid growth. Importantly, iAs-induced transformation was suppressed by NRF2 knockdown. Mechanistically, iAs suppressed Vacuolar ATPase subunit VMA5 expression, impairing lysosome acidification and inhibiting autophagic protein degradation including p62, which further activated NRF2. In vivo, chronic iAs exposure activated NRF2 in both epithelial and stroma cells of chimeric human prostate grafts and induced premalignant events. CONCLUSIONS: Low-dose iAs increased self-renewal and decreased differentiation of human PrSPCs by activating the p62-NRF2 axis, resulting in epithelial cell transformation. NRF2 is activated by iAs through specific autophagic flux blockade in progenitor cells, which may have potential therapeutic implications. https://doi.org/10.1289/EHP6471.


Asunto(s)
Arsénico/toxicidad , Sustancias Peligrosas/toxicidad , Animales , Línea Celular , Transformación Celular Neoplásica/inducido químicamente , Humanos , Masculino , Ratones , Ratones Desnudos , Factor 2 Relacionado con NF-E2 , Próstata , Ratas , Células Madre
8.
J Vis Exp ; (154)2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31885380

RESUMEN

Despite advances in adult stem cell research, identification and isolation of stem cells from tissue specimens remains a major challenge. While resident stem cells are relatively quiescent with niche restraints in adult tissues, they enter the cell cycle in anchor-free three-dimensional (3D) culture and undergo both symmetric and asymmetric cell division, giving rise to both stem and progenitor cells. The latter proliferate rapidly and are the major cell population at various stages of lineage commitment, forming heterogeneous spheroids. Using primary normal human prostate epithelial cells (HPrEC), a spheroid-based, label-retention assay was developed that permits the identification and functional isolation of the spheroid-initiating stem cells at a single cell resolution. HPrEC or cell lines are two-dimensionally (2D) cultured with BrdU for 10 days to permit its incorporation into the DNA of all dividing cells, including self-renewing stem cells. Wash out commences upon transfer to the 3D culture for 5 days, during which stem cells self-renew through asymmetric division and initiate spheroid formation. While relatively quiescent daughter stem cells retain BrdU-labeled parental DNA, the daughter progenitors rapidly proliferate, losing the BrdU label. BrdU can be substituted with CFSE or Far Red pro-dyes, which permit live stem cell isolation by FACS. Stem cell characteristics are confirmed by in vitro spheroid formation, in vivo tissue regeneration assays, and by documenting their symmetric/asymmetric cell divisions. The isolated label-retaining stem cells can be rigorously interrogated by downstream molecular and biologic studies, including RNA-seq, ChIP-seq, single cell capture, metabolic activity, proteome profiling, immunocytochemistry, organoid formation, and in vivo tissue regeneration. Importantly, this marker-free functional stem cell isolation approach identifies stem-like cells from fresh cancer specimens and cancer cell lines from multiple organs, suggesting wide applicability. It can be used to identify cancer stem-like cell biomarkers, screen pharmaceuticals targeting cancer stem-like cells, and discover novel therapeutic targets in cancers.


Asunto(s)
Separación Celular/métodos , Esferoides Celulares , Células Madre/citología , Bromodesoxiuridina , Recuento de Células , Ciclo Celular/fisiología , División Celular , Células Cultivadas , Citometría de Flujo , Humanos , Masculino , Próstata/citología
9.
Endocrinology ; 160(11): 2692-2708, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31433456

RESUMEN

Human prostate stem and progenitor cells express estrogen receptor (ER)α and ERß and exhibit proliferative responses to estrogens. In this study, membrane-initiated estrogen signaling was interrogated in human prostate stem/progenitor cells enriched from primary epithelial cultures and stem-like cell lines from benign and cancerous prostates. Subcellular fractionation and proximity ligation assays localized ERα and ERß to the cell membrane with caveolin-1 interactions. Exposure to 17ß-estradiol (E2) for 15 to 60 minutes led to sequential phosphorylation of signaling molecules in MAPK and AKT pathways, IGF1 receptor, epidermal growth factor receptor, and ERα, thus documenting an intact membrane signalosome that activates diverse downstream cascades. Treatment with an E2-dendrimer conjugate or ICI 182,870 validated E2-mediated actions through membrane ERs. Overexpression and knockdown of ERα or ERß in stem/progenitor cells identified pathway selectivity; ERα preferentially activated AKT, whereas ERß selectively activated MAPK cascades. Furthermore, prostate cancer stem-like cells expressed only ERß, and brief E2 exposure activated MAPK but not AKT cascades. A gene subset selectively regulated by nongenomic E2 signaling was identified in normal prostate progenitor cells that includes BGN, FOSB, FOXQ1, and MAF. Membrane-initiated E2 signaling rapidly modified histone methyltransferases, with MLL1 cleavage observed downstream of phosphorylated AKT and EZH2 phosphorylation downstream of MAPK signaling, which may jointly modify histones to permit rapid gene transcription. Taken together, the present findings document ERα and ERß membrane-initiated signaling in normal and cancerous human prostate stem/progenitor cells with differential engagement of downstream effectors. These signaling pathways influence normal prostate stem/progenitor cell homeostasis and provide novel therapeutic sites to target the elusive prostate cancer stem cell population.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Células Madre Neoplásicas/metabolismo , Próstata/metabolismo , Caveolina 1/metabolismo , Células Cultivadas , Histona Metiltransferasas/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Fosforilación , Fosfotransferasas/metabolismo , Próstata/citología
10.
Environ Health Perspect ; 126(11): 117001, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30387366

RESUMEN

BACKGROUND: Previous work determined that early life exposure to low-dose Bisphenol A (BPA) increased rat prostate cancer risk with aging. Herein, we report on prostate-specific results from CLARITY-BPA (Consortium Linking Academic and Regulatory Insights on BPA Toxicity), which aims to resolve uncertainties regarding BPA toxicity. OBJECTIVES: We sought to a) reassess whether a range of BPA exposures drives prostate pathology and/or alters prostatic susceptibility to hormonal carcinogenesis, and b) test whether chronic low-dose BPA targets prostate epithelial stem and progenitor cells. METHODS: Sprague-Dawley rats were gavaged daily with vehicle, ethinyl estradiol (EE) or [Formula: see text] BPA/kg-BW during development or chronically, and prostate pathology was assessed at one year. One developmentally exposed cohort was given testosterone plus estradiol ([Formula: see text]) implants at day 90 to promote carcinogenesis with aging. Epithelial stem and progenitor cells were isolated by prostasphere (PS) culture from dorsolateral prostates (DLP) of rats continuously exposed for six months to [Formula: see text] BPA/kg-BW. Gene expression was analyzed by quantitative real time reverse transcription polymerase chain reaction (qRT-PCR). RESULTS: Exposure to BPA alone at any dose did not drive prostate pathology. However, rats treated with EE, 2.5, 250, or [Formula: see text] BPA/kg-BW plus [Formula: see text] showed greater severity of lateral prostate intraepithelial neoplasia (PIN), and DLP ductal adenocarcinoma multiplicity was markedly elevated in tumor-bearing rats exposed to [Formula: see text]-BW. DLP stem cells, assessed by PS number, doubled with chronic EE and [Formula: see text] exposures. PS size, reflecting progenitor cell proliferation, was greater at 25 and [Formula: see text] BPA doses, which also shifted lineage commitment toward basal progenitors while reducing luminal progenitor cells. CONCLUSIONS: Together, these results confirm and extend previous evidence using a rat model and human prostate epithelial cells that low-dose BPA augments prostate cancer susceptibility and alters adult prostate stem cell homeostasis. Therefore, we propose that BPA exposures may contribute to the increased carcinogenic risk in humans that occurs with aging. https://doi.org/10.1289/EHP3953.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Fenoles/toxicidad , Neoplasias de la Próstata/inducido químicamente , Células Madre/efectos de los fármacos , Adenocarcinoma/inducido químicamente , Envejecimiento , Animales , Estradiol/farmacología , Expresión Génica , Homeostasis , Masculino , Próstata/citología , Neoplasia Intraepitelial Prostática/inducido químicamente , Ratas Sprague-Dawley , Testosterona/farmacología
11.
Minerva Med ; 108(3): 268-276, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27973467

RESUMEN

INTRODUCTION: The aim of this study was to evaluate the correlation of High-mobility group box 1 (HMGB1) expression in the serum with chronic hepatitis B (CHB) related liver fibrosis, severe hepatitis B and acute-on-chronic liver failure (ACLF). EVIDENCE ACQUISITION: We made a literature search in PubMed, Embase, Web of Science, Medline, Google Scholar, China National Knowledge Infrastructure, WanFang with no language restriction. Pooled data were analyzed and mean difference with corresponding 95% confidence intervals were calculated. EVIDENCE SYNTHESIS: A total of 16 relevant studies were identified. HMGB1 serum levels were higher in severe hepatitis B or ACLF patients than those in CHB patients. Pooled mean differences of HMGB1 in severe hepatitis B and ACLF patients compared with CHB patients were 4.32 (95% CI: 0.34-8.29, Z=2.13, I2=59%, P=0.03) and 15.96 (95% CI: -0.37-32.28, Z=1.92, P=0.06). Four studies showed there was a different HMGB1 expression in mild, moderate and severe CHB patients (P values were <0.05, <0.05, <0.05 and <0.01, respectively). Pooled mean difference of HMGB1 in low liver fibrosis patients compared with high liver fibrosis was -125.38 (95% CI: -539.44-288.68, Z=0.59, I2=98%, P=0.55). CONCLUSIONS: The results suggested that HMGB1 levels in the serum were statistically higher in severe hepatitis B and ACLF patients. Therefore, HMGB1 may be a useful therapeutic target for severe hepatitis B and ACLF diagnosis.


Asunto(s)
Insuficiencia Hepática Crónica Agudizada/sangre , Proteína HMGB1/sangre , Hepatitis B Crónica/sangre , Cirrosis Hepática/sangre , Hepatitis B Crónica/complicaciones , Humanos , Cirrosis Hepática/etiología , Índice de Severidad de la Enfermedad
12.
Mol Med Rep ; 16(4): 4327-4333, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28731181

RESUMEN

The aim of the present study was to investigate the role of microRNA (miR)­21 in regulating collagen I and Smad7 expression in activated rat hepatic stellate cells (HSCs). Rat HSCs were isolated by single­step density gradient centrifugation with Nycodenz. Cellular content of miR­21, SMAD7, α­smooth muscle actin (α­SMA), collagen type I alpha 1 (COLLA1) and COLL alpha 2 (A2) mRNA was examined by reverse transcription­quantitative polymerase chain reaction (RT­qPCR), and cellular content of Smad7 and α­SMA protein was detected by western blotting. Binding of miR­21 to the 3'­untranslated region (UTR) of Smad7 was verified by dual­luciferase assay. The authors observed that, in activated HSCs, expression of miR­21 was significantly increased in a time­dependent manner, while expression of Smad7 mRNA and protein was significantly reduced. In addition, miR­21 mimics significantly enhanced cellular α­SMA mRNA and protein content, while miR­21 inhibitor significantly reduced α­SMA mRNA and protein levels. Similarly, cellular content of COLLA1 and COLLA2 mRNA was significantly elevated by miR­21 mimics, but reduced by miR­21 inhibitor, in activated HSCs. Moreover, cellular content of Smad7 mRNA and protein was significantly reduced by miR­21 mimics, but significantly increased by miR­21 inhibitor. Furthermore, miR­21 mimics activated firefly luciferase in HEK293 cells transfected with the wild type 3'­UTR of Smad7. miR­21 regulates expression of α­SMA and collagen I in activated rat HSCs by directly targeting Smad7.


Asunto(s)
Actinas/metabolismo , Colágeno Tipo I/metabolismo , Células Estrelladas Hepáticas/metabolismo , MicroARNs/metabolismo , Transducción de Señal , Proteína smad7/metabolismo , Animales , Cadena alfa 1 del Colágeno Tipo I , Células HEK293 , Células Estrelladas Hepáticas/efectos de los fármacos , Humanos , Masculino , MicroARNs/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Proteína smad7/genética , Factor de Crecimiento Transformador beta/farmacología , Regulación hacia Arriba/efectos de los fármacos
13.
Stem Cell Res ; 23: 1-12, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28651114

RESUMEN

Using primary cultures of normal human prostate epithelial cells, we developed a novel prostasphere-based, label-retention assay that permits identification and isolation of stem cells at a single cell level. Their bona fide stem cell nature was corroborated using in vitro and in vivo regenerative assays and documentation of symmetric/asymmetric division. Robust WNT10B and KRT13 levels without E-cadherin or KRT14 staining distinguished individual stem cells from daughter progenitors in spheroids. Following FACS to isolate label-retaining stem cells from label-free progenitors, RNA-seq identified unique gene signatures for the separate populations which may serve as useful biomarkers. Knockdown of KRT13 or PRAC1 reduced sphere formation and symmetric self-renewal highlighting their role in stem cell maintenance. Pathways analysis identified ribosome biogenesis and membrane estrogen-receptor signaling enriched in stem cells with NF-ĸB signaling enriched in progenitors; activities that were biologically confirmed. Further, bioassays identified heightened autophagy flux and reduced metabolism in stem cells relative to progenitors. These approaches similarly identified stem-like cells from prostate cancer specimens and prostate, breast and colon cancer cell lines suggesting wide applicability. Together, the present studies isolate and identify unique characteristics of normal human prostate stem cells and uncover processes that maintain stem cell homeostasis in the prostate gland.


Asunto(s)
Separación Celular/métodos , Células Epiteliales/citología , Próstata/citología , Análisis de la Célula Individual/métodos , Adulto , División Celular Asimétrica , Autofagia , Autorrenovación de las Células , Células Cultivadas , Fluoresceínas/metabolismo , Técnicas de Silenciamiento del Gen , Ontología de Genes , Humanos , Queratinas/metabolismo , Masculino , Mitocondrias/metabolismo , Células Madre Neoplásicas/metabolismo , Regeneración , Análisis de Secuencia de ARN , Esferoides Celulares/citología , Coloración y Etiquetado , Succinimidas/metabolismo , Transcriptoma/genética , Adulto Joven
14.
Int J Clin Exp Pathol ; 8(8): 9062-71, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26464648

RESUMEN

High mobility group box 1 (HMGB1) has been widely reported to mediate damage caused by inflammatory responses. The aim of our study is to investigate the role of HMGB1 in endotoxin tolerance (ET) alleviating inflammation of acute liver failure (ALF) rats and its possible signaling mechanism. To mimic ET, male Sprague-Dawley rats were pretreated with low dose of lipopolysaccharide (LPS) (0.1 mg/kg once a day intraperitoneally for consecutive five days) before subsequent ALF induction. ALF was induced by intraperitoneal administration of D-GalN/LPS. ET induced by LPS pretreatment significantly improved the survival rate of ALF rats. Moreover, after ALF induction, ET+ALF rats exhibited lower serum enzyme (ALT, AST and TBiL) levels, lower production of inflammatory cytokines (IL-6, TNF-a and HMGB1) and more minor liver histopathological damage than ALF rats. ET+ALF rats showed enhanced expression levels of HMGB1, decreased levels of STAT1 and p-STAT1, augmented expression of SOCS1 in liver tissues than ALF rats. These results indicated that ET induced by low-dose LPS pretreatment may alleviate inflammation and liver injury in experimental acute liver failure rats mainly through inhibition of hepatic HMGB1 translocation and release.


Asunto(s)
Tolerancia a Medicamentos , Proteína HMGB1/metabolismo , Lipopolisacáridos , Fallo Hepático Agudo/metabolismo , Hígado/metabolismo , Animales , Modelos Animales de Enfermedad , Interleucina-6/metabolismo , Hígado/patología , Fallo Hepático Agudo/inducido químicamente , Fallo Hepático Agudo/patología , Masculino , Fosforilación , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
15.
Endocrinology ; 155(3): 805-17, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24424067

RESUMEN

Previous studies in rodent models have shown that early-life exposure to bisphenol A (BPA) reprograms the prostate and enhances its susceptibility to hormonal carcinogenesis with aging. To determine whether the human prostate is similarly sensitive to BPA, the current study used human prostate epithelial stem-like cells cultured from prostates of young, disease-free donors. Similar to estradiol-17ß (E2), BPA increased stem-progenitor cell self-renewal and expression of stem-related genes in a dose-dependent manner. Further, 10 nM BPA and E2 possessed equimolar membrane-initiated signaling with robust induction of p-Akt and p-Erk at 15 minutes. To assess in vivo carcinogenicity, human prostate stem-progenitor cells combined with rat mesenchyme were grown as renal grafts in nude mice, forming normal human prostate epithelium at 1 month. Developmental BPA exposure was achieved through oral administration of 100 or 250 µg BPA/kg body weight to hosts for 2 weeks after grafting, producing free BPA levels of 0.39 and 1.35 ng/mL serum, respectively. Carcinogenesis was driven by testosterone plus E2 treatment for 2 to 4 months to model rising E2 levels in aging men. The incidence of high-grade prostate intraepithelial neoplasia and adenocarcinoma markedly increased from 13% in oil-fed controls to 33% to 36% in grafts exposed in vivo to BPA (P < .05). Continuous developmental BPA exposure through in vitro (200 nM) plus in vivo (250 µg/kg body weight) treatments increased high-grade prostate intraepithelial neoplasia/cancer incidence to 45% (P < .01). Together, the present findings demonstrate that human prostate stem-progenitor cells are direct BPA targets and that developmental exposure to BPA at low doses increases hormone-dependent cancer risk in the human prostate epithelium.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Epitelio/patología , Estrógenos no Esteroides/toxicidad , Fenoles/toxicidad , Próstata/citología , Neoplasias de la Próstata/patología , Células Madre/efectos de los fármacos , Animales , Carcinogénesis , Células Cultivadas , Cromatografía Líquida de Alta Presión , Epitelio/efectos de los fármacos , Estrógenos/metabolismo , Citometría de Flujo , Humanos , Masculino , Ratones , Ratones Desnudos , Próstata/efectos de los fármacos , Neoplasias de la Próstata/inducido químicamente , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Células Madre/citología , Espectrometría de Masas en Tándem , Factores de Tiempo , Adulto Joven
16.
Mol Cell Endocrinol ; 354(1-2): 63-73, 2012 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-21914459

RESUMEN

Estrogen reprogramming of the prostate gland as a function of developmental exposures (aka developmental estrogenization) results in permanent alterations in structure and gene expression that lead to an increased incidence of prostatic lesions with aging. Endocrine disrupting chemicals (EDCs) with estrogenic activity have been similarly linked to an increased prostate cancer risk. Since it has been suggested that stem cells and cancer stem cells are potential targets of cancer initiation and disease management, it is highly possible that estrogens and EDCs influence the development and progression of prostate cancer through reprogramming and transforming the prostate stem and early stage progenitor cells. In this article, we review recent literature highlighting the effects of estrogens and EDCs on prostate cancer risk and discuss recent advances in prostate stem/progenitor cell research. Our laboratory has recently developed a novel prostasphere model using normal human prostate stem/progenitor cells and established that these cells express estrogen receptors (ERs) and are direct targets of estrogen action. Further, using a chimeric in vivo prostate model derived from these normal human prostate progenitor cells, we demonstrated for the first time that estrogens initiate and promote prostatic carcinogenesis in an androgen-supported environment. We herein discuss these findings and highlight new evidence using our in vitro human prostasphere assay for perturbations in human prostate stem cell self-renewal and differentiation by natural steroids as well as EDCs. These findings support the hypothesis that tissue stem cells may be direct EDC targets which may underlie life-long reprogramming as a consequence of developmental and/or transient adult exposures.


Asunto(s)
Disruptores Endocrinos/toxicidad , Estrógenos/toxicidad , Neoplasias de la Próstata/patología , Células Madre/efectos de los fármacos , Animales , Transformación Celular Neoplásica , Exposición a Riesgos Ambientales , Contaminantes Ambientales/toxicidad , Humanos , Masculino , Factores de Riesgo
17.
Endocrinology ; 152(6): 2150-63, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21427218

RESUMEN

The present study sought to determine whether estrogens with testosterone support are sufficient to transform the normal human prostate epithelium and promote progression to invasive adenocarcinoma using a novel chimeric prostate model. Adult prostate stem/early progenitor cells were isolated from normal human prostates through prostasphere formation in three-dimensional culture. The stem/early progenitor cell status and clonality of prostasphere cells was confirmed by immunocytochemistry and Hoechst staining. Normal prostate progenitor cells were found to express estrogen receptor α, estrogen receptor ß, and G protein-coupled receptor 30 mRNA and protein and were responsive to 1 nm estradiol-17ß with increased numbers and prostasphere size, implicating them as direct estrogen targets. Recombinants of human prostate progenitor cells with rat urogenital sinus mesenchyme formed chimeric prostate tissue in vivo under the renal capsule of nude mice. Cytodifferentiation of human prostate progenitor cells in chimeric tissues was confirmed by immunohistochemistry using epithelial cell markers (p63, cytokeratin 8/18, and androgen receptor), whereas human origin and functional differentiation were confirmed by expression of human nuclear antigen and prostate-specific antigen, respectively. Once mature tissues formed, the hosts were exposed to elevated testosterone and estradiol-17ß for 1-4 months, and prostate pathology was longitudinally monitored. Induction of prostate cancer in the human stem/progenitor cell-generated prostatic tissue was observed over time, progressing from normal histology to epithelial hyperplasia, prostate intraepithelial neoplasia, and prostate cancer with local renal invasion. These findings provide the first direct evidence that human prostate progenitor cells are estrogen targets and that estradiol in an androgen-supported milieu is a carcinogen for human prostate epithelium.


Asunto(s)
Células Madre Adultas/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Epitelio/efectos de los fármacos , Estrógenos/efectos adversos , Neoplasias de la Próstata/etiología , Adulto , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Epitelio/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Próstata/citología , Próstata/efectos de los fármacos , Próstata/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA