Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Med Virol ; 95(1): e28108, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36042555

RESUMO

The VG161 represents the first recombinant oncolytic herpes simplex virus type 1 carrying multiple synergistic antitumor immuno-modulating factors. Here, we report its antitumor mechanisms and thus provide firm theoretical foundation for the upcoming clinical application in pancreatic cancer. Generally, the VG161-mediated antitumor outcomes were analyzed by a collaboration of techniques, namely the single-cell sequencing, airflow-assisted desorption electrospray ionization-mass spectrometry imaging (AFADSI-MSI) and nanostring techniques. In vitro, the efficacy of VG161 together with immune checkpoint inhibitors (ICIs) has been successfully shown to grant a long-term antitumor effect by altering tumor immunity and remodeling tumor microenvironment (TME) metabolisms. Cellular functional pathways and cell subtypes detected from patient samples before and after the treatment had undergone distinctive changes including upregulated CD8+ T and natural killer cells. More importantly, significant antitumor signals have emerged since the administration of VG161 injection. In conclusion, VG161 can systematically activate acquired and innate immunity in pancreatic models, as well as improve the tumor immune microenvironment, indicative of strong antitumor potential. The more robusting antitumor outcome for VG161 monotherapy or in combination with other therapies on pancreatic cancer is worth of being explored in further clinical trials.


Assuntos
Herpesvirus Humano 1 , Terapia Viral Oncolítica , Neoplasias Pancreáticas , Humanos , Terapia Viral Oncolítica/métodos , Herpesvirus Humano 1/genética , Imunomodulação , Neoplasias Pancreáticas/terapia , Transgenes , Linhagem Celular Tumoral , Microambiente Tumoral , Neoplasias Pancreáticas
2.
Mol Ther ; 30(2): 855-867, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-34547467

RESUMO

Cell-penetrating peptides (CPPs) hold great promise for intracellular delivery of therapeutic proteins. However, endosomal entrapment of transduced cargo is a major bottleneck hampering their successful application. While developing a transducible zinc finger protein-based artificial transcription factor targeting the expression of endothelin receptor A, we identified interaction between the CPP and the endosomal membrane or endosomal entanglement as a main culprit for endosomal entrapment. To achieve endosomal disentanglement, we utilized endosome-resident proteases to sever the artificial transcription factor from its CPP upon arrival inside the endosome. Using this approach, we greatly enhanced the correct subcellular localization of the disentangled artificial transcription factor, significantly increasing its biological activity and distribution in vivo. With rational engineering of proteolytic sensitivity, we propose a new design principle for transducible therapeutic proteins, helping CPPs attain their full potential as delivery vectors for therapeutic proteins.


Assuntos
Peptídeos Penetradores de Células , Receptores de Endotelina , Peptídeos Penetradores de Células/metabolismo , Endossomos/metabolismo , Receptores de Endotelina/metabolismo , Fatores de Transcrição/metabolismo
3.
J Virol ; 95(15): e0012721, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34011542

RESUMO

Small-molecule drugs inhibiting BK polyomavirus (BKPyV) represent a significant unmet clinical need in view of polyomavirus-associated nephropathy or hemorrhagic cystitis, which complicate 5% to 25% of kidney and hematopoietic cell transplantations. We characterized the inhibitory activity of acitretin on BKPyV replication in primary human renal proximal tubular epithelial cells (RPTECs). Effective inhibitory concentrations of 50% (EC50) and 90% (EC90) were determined in dilution series measuring BKPyV loads, transcripts, and protein expression, using cell proliferation, metabolic activity, and viability to estimate cytotoxic concentrations and selectivity indices (SI). The acitretin EC50 and EC90 in RPTECs were 0.64 (SI50, 250) and 3.25 µM (SI90, 49.2), respectively. Acitretin effectively inhibited BKPyV replication until 72 h postinfection when added 24 h before infection until 12 h after infection, but decreased to <50% at later time points. Acitretin did not interfere with nuclear delivery of BKPyV genomes, but it decreased large T-antigen transcription and protein expression. Acitretin did not inhibit the initial round of BKPyV replication following transfection of full-length viral genomes, but it affected subsequent rounds of reinfection. Acitretin also inhibited BKPyV replication in human urothelial cells and in Vero cells, but not in COS-7 cells constitutively expressing Simian virus 40 (SV40) large T antigen. Retinoic acid agonists (all-trans retinoic acid, 9-cis retinoic acid [9-cis-RA], 13-cis-RA, bexarotene, and tamibarotene) and the RAR/RXR antagonist RO41-5253 also inhibited BKPyV replication, pointing to an as-yet-undefined mechanism. IMPORTANCE Acitretin selectively inhibits BKPyV replication in primary human cell culture models of nephropathy and hemorrhagic cystitis. Since acitretin is an approved drug in clinical use reaching BKPyV-inhibiting concentrations in systemically treated patients, further studies are warranted to provide data for clinical repurposing of retinoids for treatment and prevention of replicative BKPyV-diseases.


Assuntos
Acitretina/farmacologia , Antivirais/farmacologia , Vírus BK/crescimento & desenvolvimento , Retinoides/farmacologia , Tretinoína/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Antígenos Virais de Tumores/biossíntese , Antígenos Virais de Tumores/genética , Células COS , Linhagem Celular , Chlorocebus aethiops , Cistite/tratamento farmacológico , Cistite/virologia , Genoma Viral/genética , Células HEK293 , Humanos , Nefropatias/tratamento farmacológico , Nefropatias/virologia , Testes de Sensibilidade Microbiana , Infecções por Polyomavirus/tratamento farmacológico , Tretinoína/análogos & derivados , Infecções Tumorais por Vírus/tratamento farmacológico , Células Vero
4.
Rev Med Virol ; 31(6): e2220, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33729628

RESUMO

Human polyomaviruses (HPyVs) encompass more than 10 species infecting 30%-90% of the human population without significant illness. Proven HPyV diseases with documented histopathology affect primarily immunocompromised hosts with manifestations in brain, skin and renourinary tract such as polyomavirus-associated nephropathy (PyVAN), polyomavirus-associated haemorrhagic cystitis (PyVHC), polyomavirus-associated urothelial cancer (PyVUC), progressive multifocal leukoencephalopathy (PML), Merkel cell carcinoma (MCC), Trichodysplasia spinulosa (TS) and pruritic hyperproliferative keratinopathy. Although virus-specific immune control is the eventual goal of therapy and lasting cure, antiviral treatments are urgently needed in order to reduce or prevent HPyV diseases and thereby bridging the time needed to establish virus-specific immunity. However, the small dsDNA genome of only 5 kb of the non-enveloped HPyVs only encodes 5-7 viral proteins. Thus, HPyV replication relies heavily on host cell factors, thereby limiting both, number and type of specific virus-encoded antiviral targets. Lack of cost-effective high-throughput screening systems and relevant small animal models complicates the preclinical development. Current clinical studies are limited by small case numbers, poorly efficacious compounds and absence of proper randomized trial design. Here, we review preclinical and clinical studies that evaluated small molecules with presumed antiviral activity against HPyVs and provide an outlook regarding potential new antiviral strategies.


Assuntos
Antivirais/uso terapêutico , Polyomavirus/efeitos dos fármacos , Vírus de DNA , Humanos
5.
J Virol ; 92(7)2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29343574

RESUMO

Human polyomavirus (HPyV) DNA genomes contain three regions denoted the early viral gene region (EVGR), encoding the regulatory T-antigens and one microRNA, the late viral gene region (LVGR), encoding the structural Vp capsid proteins, and the noncoding control region (NCCR). The NCCR harbors the origin of viral genome replication and bidirectional promoter/enhancer functions governing EVGR and LVGR expression on opposite DNA strands. Despite principal similarities, HPyV NCCRs differ in length, sequence, and architecture. To functionally compare HPyV NCCRs, sequences from human isolates were inserted into a bidirectional reporter vector using dsRed2 for EVGR expression and green fluorescent protein (GFP) for LVGR expression. Transfecting HPyV NCCR reporter vectors into human embryonic kidney 293 (HEK293) cells and flow cytometry normalized to archetype BKPyV NCCR revealed a hierarchy of EVGR expression levels with MCPyV, HPyV12, and STLPyV NCCRs conferring stronger levels and HPyV6, HPyV9, and HPyV10 NCCRs weaker levels, while LVGR expression was less variable and showed comparable activity levels. Transfection of HEK293T cells expressing simian virus 40 (SV40) large T antigen (LTag) increased EVGR expression for most HPyV NCCRs, which correlated with the number of LTag-binding sites (Spearman's r, 0.625; P < 0.05) and decreased following SV40 LTag small interfering RNA (siRNA) knockdown. LTag-dependent activation was specifically confirmed for two different MCPyV NCCRs in 293MCT cells expressing the cognate MCPyV LTag. HPyV NCCR expression in different cell lines derived from skin (A375), cervix (HeLaNT), lung (A549), brain (Hs683), and colon (SW480) demonstrated that host cell properties significantly modulate the baseline HPyV NCCR activity, which partly synergized with SV40 LTag expression. Clinically occurring NCCR sequence rearrangements of HPyV7 PITT-1 and -2 and HPyV9 UF1 were found to increase EVGR expression compared to the respective HPyV archetype, but this was partly host cell type specific.IMPORTANCE HPyV NCCRs integrate essential viral functions with respect to host cell specificity, persistence, viral replication, and disease. Here, we show that HPyV NCCRs not only differ in sequence length, number, and position of LTag- and common transcription factor-binding sites but also confer differences in bidirectional viral gene expression. Importantly, EVGR reporter expression was significantly modulated by LTag expression and by host cell properties. Clinical sequence variants of HPyV7 and HPyV9 NCCRs containing deletions and insertions were associated with increased EVGR expression, similar to BKPyV and JCPyV rearrangements, emphasizing that HPyV NCCR sequences are major determinants not only of host cell tropism but also of pathogenicity. These results will help to define secondary HPyV cell tropism beyond HPyV surface receptors, to identify key viral and host factors shaping the viral life cycle, and to develop preclinical models of HPyV persistence and replication and suitable antiviral targets.


Assuntos
Antígenos Virais de Tumores , Regulação Viral da Expressão Gênica , Rearranjo Gênico , Genoma Viral , Modelos Genéticos , Polyomaviridae , Antígenos Virais de Tumores/genética , Antígenos Virais de Tumores/metabolismo , Células HEK293 , Humanos , MicroRNAs/biossíntese , MicroRNAs/genética , Polyomaviridae/genética , Polyomaviridae/metabolismo , RNA Viral/biossíntese , RNA Viral/genética
6.
Circ J ; 76(8): 2015-22, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22572461

RESUMO

BACKGROUND: Hexosamine biosynthetic pathway (HBP) is implicated in increased plasminogen activator inhibitor-1 (PAI-1), and endothelial nitric oxide synthase (eNOS) dysfunction in diabetes. Glucosamine (GlcN) that directly activates HBP is a dietary supplement and is clinically used to treat osteoarthritis despite uncertain efficacy and adverse cardiovascular effects observed in animal models. p38 mitogen-activated protein kinase (p38mapk) has been shown to be involved in HBP-mediated biological processes. The aim of the present study was to investigate the role of p38mapk in GlcN-induced endothelial PAI-1 expression and eNOS dysfunction. METHODS AND RESULTS: In cultured human endothelial cells, GlcN time- and concentration-dependently increased PAI-1 protein level that was further enhanced by tumor necrosis factor (TNF)-α, which was accompanied by a transient synergistic activation of p38mapk. The stimulation of PAI-1 by GlcN alone or by GlcN and TNF-α in combination was inhibited by the specific inhibitor of p38mapk, but not that of JNK or ERK1/2. Moreover, in isolated mouse aortas, GlcN caused eNOS uncoupling resulting in enhanced superoxide and decreased NO production, as well as impaired endothelium-dependent relaxations, which were also fully prevented by the p38mapk inhibitor. CONCLUSIONS: HBP activated by GlcN increases PAI-1 expression and eNOS uncoupling depending on p38mapk, which not only explains hyperglycemic vascular complications, but also may bring into question the clinical use of GlcN. The present results, support currently ongoing clinical application of p38mapk inhibitor in patients with cardiovascular disease.


Assuntos
Aorta/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glucosamina/efeitos adversos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Inibidor 1 de Ativador de Plasminogênio/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Aorta/patologia , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Glucosamina/farmacologia , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Masculino , Camundongos , Óxido Nítrico/biossíntese , Superóxidos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
7.
Mol Biol Rep ; 38(7): 4777-86, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21152985

RESUMO

MicroRNAs (miRNAs) are endogenously expressed RNAs consisting of 20-24 nucleotides. These molecules are thought to repress protein translation by binding to target mRNAs. However, biological functions have not been assigned to most of the 175 porcine miRNAs registered in miRBase (release 15.0). In an effort to uncover miR-103 important in pigs, we examined the integrative tissue expression profile and gene ontology (GO) term enrichment of predicted target genes to determine the global biological functions of miR-103. Our results demonstrated that miR-103 is involved in various biological processes including brain development, lipid metabolism, adipocyte differentiation, hematopoiesis, and immunity. Moreover, we also experimentally verified effects of miR-103 in porcine preadipocytes. miR-103 levels increased in differentiating adipocytes, and inhibition of miR-103 effectively inhibited preadipocyte differentiation. In addition, mRNA levels of the putative miR-103 target RAI14 were higher in miR-103 inhibitor-treated adipocytes. These results demonstrate that miR-103 is involved in porcine preadipocyte differentiation and may act through the putative target gene RAI14. In a word, our data provide new insights into the global biological role of miR-103.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica , MicroRNAs/genética , Sus scrofa/genética , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Imunidade/genética , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Masculino , MicroRNAs/metabolismo , Sistema Nervoso/efeitos dos fármacos , Sistema Nervoso/crescimento & desenvolvimento , Especificidade de Órgãos/efeitos dos fármacos , Especificidade de Órgãos/genética , RNA Antissenso/farmacologia , Fatores de Tempo
8.
Viruses ; 10(9)2018 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-30200237

RESUMO

In immunosuppressed patients, BKPyV-variants emerge carrying rearranged non-coding control-regions (rr-NCCRs) that increase early viral gene region (EVGR) expression and replication capacity. BKPyV also encodes microRNAs, which have been reported to downregulate EVGR-encoded large T-antigen transcripts, to decrease viral replication in infected cells and to be secreted in exosomes. To investigate the interplay of NCCR and microRNAs, we compared archetype- and rr-NCCR-BKPyV infection in cell culture. We found that laboratory and clinical rr-NCCR-BKPyV-strains show higher replication rates but significantly lower microRNA levels than archetype virus intracellularly and in exosomes. To investigate whether rr-NCCR or increased EVGR activity modulated microRNA levels, we examined the (sp1-4)NCCR-BKPyV, which has an archetype NCCR-architecture but shows increased EVGR expression due to point mutations inactivating one Sp1 binding site. We found that microRNA levels following (sp1-4)NCCR-BKPyV infection were as low as in rr-NCCR-variants. Thus, NCCR rearrangements are not required for lower miRNA levels. Accordingly, Sp1 siRNA knock-down decreased microRNA levels in archetype BKPyV infection but had no effect on (sp1-4)- or rr-NCCR-BKPyV. However, rr-NCCR-BKPyV replication was downregulated by exosome preparations carrying BKPyV-microRNA prior to infection. To explore the potential relevance in humans, urine samples from 12 natalizumab-treated multiple sclerosis patients were analysed. In 7 patients, rr-NCCR-BKPyV were detected showing high urine BKPyV loads but low microRNAs levels, whereas the opposite was seen in 5 patients with archetype BKPyV. We discuss the results in a dynamic model of BKPyV replication according to NCCR activity and exosome regulation, which integrates immune selection pressure, spread to new host cells and rr-NCCR emergence.


Assuntos
Antivirais/metabolismo , Vírus BK/crescimento & desenvolvimento , Exossomos/química , MicroRNAs/metabolismo , RNA Viral/metabolismo , Replicação Viral , Animais , Antivirais/análise , Vírus BK/isolamento & purificação , Células COS , Chlorocebus aethiops , Humanos , Hospedeiro Imunocomprometido , MicroRNAs/análise , RNA Viral/análise , Urina/virologia , Carga Viral
9.
Aging (Albany NY) ; 7(1): 70-81, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25635535

RESUMO

p38 mitogen-activated protein kinase (p38) regulates cellular senescence and senescence-associated secretory phenotype (SASP), i.e., secretion of cytokines and/or chemokines. Previous work showed that augmented arginase-II (Arg-II) and S6K1 interact with each other to promote endothelial senescence through uncoupling of endothelial nitric oxide synthase (eNOS). Here we demonstrate eNOS-uncoupling, augmented expression/secretion of IL-6 and IL-8, elevation of p38 activation and Arg-II levels in senescent endothelial cells. Silencing Arg-II or p38α in senescent cells recouples eNOS and inhibits IL-6 and IL-8 secretion. Overexpression of Arg-II in young endothelial cells causes eNOS-uncoupling and enhances IL-6 and IL-8 expression/secretion, which is prevented by p38 inhibition or by antioxidant. Moreover, p38 activation and expression of IL-6 and KC (the murine IL-8 homologue) are increased in the heart and/or aortas of wild type (WT) old mice, which is abolished in mice with Arg-II gene deficiency (Arg-II-/-). In addition, inhibition of p38 in the old WT mice recouples eNOS function and reduces IL-6 and KC expression in the aortas and heart. Silencing Arg-II or p38a or S6K1 inhibits each other in senescence endothelial cells. Thus, Arg-II, p38, and S6K1 form a positive circuit which regulates endothelial senescence and cardiovascular aging.


Assuntos
Arginase/metabolismo , Senescência Celular , Células Endoteliais/enzimologia , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Antioxidantes/farmacologia , Arginase/genética , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Feminino , Células Endoteliais da Veia Umbilical Humana/enzimologia , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Proteínas Quinases S6 Ribossômicas 70-kDa/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Transdução de Sinais/efeitos dos fármacos , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética
10.
J Am Heart Assoc ; 1(4): e000992, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23130157

RESUMO

BACKGROUND: Macrophage-mediated chronic inflammation is mechanistically linked to insulin resistance and atherosclerosis. Although arginase I is considered antiinflammatory, the role of arginase II (Arg-II) in macrophage function remains elusive. This study characterizes the role of Arg-II in macrophage inflammatory responses and its impact on obesity-linked type II diabetes mellitus and atherosclerosis. METHODS AND RESULTS: In human monocytes, silencing Arg-II decreases the monocytes' adhesion to endothelial cells and their production of proinflammatory mediators stimulated by oxidized low-density lipoprotein or lipopolysaccharides, as evaluated by real-time quantitative reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay. Macrophages differentiated from bone marrow cells of Arg-II-deficient (Arg-II(-/-)) mice express lower levels of lipopolysaccharide-induced proinflammatory mediators than do macrophages of wild-type mice. Importantly, reintroducing Arg-II cDNA into Arg-II(-/-) macrophages restores the inflammatory responses, with concomitant enhancement of mitochondrial reactive oxygen species. Scavenging of reactive oxygen species by N-acetylcysteine prevents the Arg-II-mediated inflammatory responses. Moreover, high-fat diet-induced infiltration of macrophages in various organs and expression of proinflammatory cytokines in adipose tissue are blunted in Arg-II(-/-) mice. Accordingly, Arg-II(-/-) mice reveal lower fasting blood glucose and improved glucose tolerance and insulin sensitivity. Furthermore, apolipoprotein E (ApoE)-deficient mice with Arg-II deficiency (ApoE(-/-)Arg-II(-/-)) display reduced lesion size with characteristics of stable plaques, such as decreased macrophage inflammation and necrotic core. In vivo adoptive transfer experiments reveal that fewer donor ApoE(-/-)Arg-II(-/-) than ApoE(-/-)Arg-II(+/+) monocytes infiltrate into the plaque of ApoE(-/-)Arg-II(+/+) mice. Conversely, recipient ApoE(-/-)Arg-II(-/-) mice accumulate fewer donor monocytes than do recipient ApoE(-/-)Arg-II(+/+) animals. CONCLUSIONS: Arg-II promotes macrophage proinflammatory responses through mitochondrial reactive oxygen species, contributing to insulin resistance and atherogenesis. Targeting Arg-II represents a potential therapeutic strategy in type II diabetes mellitus and atherosclerosis. (J Am Heart Assoc. 2012;1:e000992 doi: 10.1161/JAHA.112.000992.).

11.
BMB Rep ; 42(6): 338-43, 2009 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-19558791

RESUMO

The Wnt/beta-catenin signaling pathway alters adipocyte differentiation by inhibiting adipogenic gene expression. beta-catenin plays a central role in the Wnt/beta-catenin signaling pathway. In this study, we revealed that tumour necrosis factor-alpha (TNF-alpha), a potential negative regulator of adipocyte differentiation, inhibits porcine adipogenesis through activation of the Wnt/beta-catenin signaling pathway. Under the optimal concentration of TNF-alpha, the intracellular beta-catenin protein was stabilized. Thus, the intracellular lipid accumulation of porcine preadipocyte was suppressed and the expression of important adipocyte marker genes, including peroxisome proliferator-activated receptor-gamma (PPARgamma) and CCAAT/enhancer binding protein-alpha (C/EBPalpha), were inhibited. However, a loss of beta-catenin in porcine preadipocytes enhanced the adipogenic differentiation and attenuated TNF-alpha induced anti-adipogenesis. Taken together, this study indicated that TNF-alpha inhibits adipogenesis through stabilization of beta-catenin protein in porcine preadipocytes.


Assuntos
Adipócitos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , beta Catenina/metabolismo , Adipócitos/metabolismo , Adipócitos/fisiologia , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Animais , Diferenciação Celular/genética , Células Cultivadas , Regulação para Baixo/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Estabilidade Proteica/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Suínos , Fator de Necrose Tumoral alfa/metabolismo , beta Catenina/antagonistas & inibidores , beta Catenina/genética , beta Catenina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA