Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Science ; 384(6701): eadk5382, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38870290

RESUMEN

Polycystic ovary syndrome (PCOS), a prevalent reproductive disorder in women of reproductive age, features androgen excess, ovulatory dysfunction, and polycystic ovaries. Despite its high prevalence, specific pharmacologic intervention for PCOS is challenging. In this study, we identified artemisinins as anti-PCOS agents. Our finding demonstrated the efficacy of artemisinin derivatives in alleviating PCOS symptoms in both rodent models and human patients, curbing hyperandrogenemia through suppression of ovarian androgen synthesis. Artemisinins promoted cytochrome P450 family 11 subfamily A member 1 (CYP11A1) protein degradation to block androgen overproduction. Mechanistically, artemisinins directly targeted lon peptidase 1 (LONP1), enhanced LONP1-CYP11A1 interaction, and facilitated LONP1-catalyzed CYP11A1 degradation. Overexpression of LONP1 replicated the androgen-lowering effect of artemisinins. Our data suggest that artemisinin application is a promising approach for treating PCOS and highlight the crucial role of the LONP1-CYP11A1 interaction in controlling hyperandrogenism and PCOS occurrence.


Asunto(s)
Proteasas ATP-Dependientes , Artemisininas , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol , Proteínas Mitocondriales , Síndrome del Ovario Poliquístico , Animales , Femenino , Humanos , Ratones , Ratas , Andrógenos/metabolismo , Artemisininas/uso terapéutico , Artemisininas/farmacología , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Modelos Animales de Enfermedad , Hiperandrogenismo/tratamiento farmacológico , Hiperandrogenismo/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , Ovario/efectos de los fármacos , Ovario/metabolismo , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Proteolisis , Ratones Endogámicos C57BL , Adulto Joven , Adulto , Ratas Sprague-Dawley , Proteasas ATP-Dependientes/genética , Proteasas ATP-Dependientes/metabolismo
2.
Acta Pharmacol Sin ; 44(10): 2103-2112, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37193754

RESUMEN

Checkpoint inhibitors such as PD-1/PD-L1 antibody therapeutics are a promising option for the treatment of multiple cancers. Due to the inherent limitations of antibodies, great efforts have been devoted to developing small-molecule PD-1/PD-L1 signaling pathway inhibitors. In this study we established a high-throughput AlphaLISA assay to discover small molecules with new skeletons that could block PD-1/PD-L1 interaction. We screened a small-molecule library of 4169 compounds including natural products, FDA approved drugs and other synthetic compounds. Among the 8 potential hits, we found that cisplatin, a first-line chemotherapeutic drug, reduced AlphaLISA signal with an EC50 of 8.3 ± 2.2 µM. Furthermore, we showed that cisplatin-DMSO adduct, but not semplice cisplatin, inhibited PD-1/PD-L1 interaction. Thus, we assessed several commercial platinum (II) compounds, and found that bis(benzonitrile) dichloroplatinum (II) disturbed PD-1/PD-L1 interaction (EC50 = 13.2 ± 3.5 µM). Its inhibitory activity on PD-1/PD-L1 interaction was confirmed in co-immunoprecipitation and PD-1/PD-L1 signaling pathway blockade bioassays. Surface plasmon resonance assay revealed that bis(benzonitrile) dichloroplatinum (II) bound to PD-1 (KD = 2.08 µM) but not PD-L1. In immune-competent wild-type mice but not in immunodeficient nude mice, bis(benzonitrile) dichloroplatinum (II) (7.5 mg/kg, i.p., every 3 days) significantly suppressed the growth of MC38 colorectal cancer xenografts with increasing tumor-infiltrating T cells. These data highlight that platinum compounds are potential immune checkpoint inhibitors for the treatment of cancers.


Asunto(s)
Cisplatino , Inhibidores de Puntos de Control Inmunológico , Neoplasias , Animales , Humanos , Ratones , Anticuerpos , Antígeno B7-H1/metabolismo , Cisplatino/farmacología , Cisplatino/uso terapéutico , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1 , Inhibidores de Puntos de Control Inmunológico/farmacología
3.
Sci China Life Sci ; 66(8): 1869-1887, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37059927

RESUMEN

Protein-biomolecule interactions play pivotal roles in almost all biological processes. For a biomolecule of interest, the identification of the interacting protein(s) is essential. For this need, although many assays are available, highly robust and reliable methods are always desired. By combining a substrate-based proximity labeling activity from the pupylation pathway of Mycobacterium tuberculosis and the streptavidin (SA)-biotin system, we developed the Specific Pupylation as IDEntity Reporter (SPIDER) method for identifying protein-biomolecule interactions. Using SPIDER, we validated the interactions between the known binding proteins of protein, DNA, RNA, and small molecule. We successfully applied SPIDER to construct the global protein interactome for m6A and mRNA, identified a variety of uncharacterized m6A binding proteins, and validated SRSF7 as a potential m6A reader. We globally identified the binding proteins for lenalidomide and CobB. Moreover, we identified SARS-CoV-2-specific receptors on the cell membrane. Overall, SPIDER is powerful and highly accessible for the study of protein-biomolecule interactions.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2 , Proteínas , Unión Proteica
4.
Acta Pharmacol Sin ; 42(10): 1556-1566, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33495516

RESUMEN

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by toxic aggregates of mutant huntingtin protein (mHTT) in the brain. Decreasing mHTT is a potential strategy for therapeutic purpose of HD. Valosin-containing protein (VCP/p97) is a crucial regulator of proteostasis, which regulates the degradation of damaged protein through proteasome and autophagy pathway. Since VCP has been implicated in pathogenesis of HD as well as other neurodegenerative diseases, small molecules that specifically regulate the activity of VCP may be of therapeutic benefits for HD patients. In this study we established a high-throughput screening biochemical assay for VCP ATPase activity measurement and identified gossypol, a clinical approved drug in China, as a novel modulator of VCP. Gossypol acetate dose-dependently inhibited the enzymatic activity of VCP in vitro with IC50 of 6.53±0.6 µM. We further demonstrated that gossypol directly bound to the interface between the N and D1 domains of VCP. Gossypol acetate treatment not only lowered mHTT levels and rescued HD-relevant phenotypes in HD patient iPS-derived Q47 striatal neurons and HD knock-in mouse striatal cells, but also improved motor function deficits in both Drosophila and mouse HD models. Taken together, gossypol acetate acted through a gain-of-function way to induce the formation of VCP-LC3-mHTT ternary complex, triggering autophagic degradation of mHTT. This study reveals a new strategy for treatment of HD and raises the possibility that an existing drug can be repurposed as a new treatment of neurodegenerative diseases.


Asunto(s)
Autofagia/efectos de los fármacos , Gosipol/uso terapéutico , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Animales , Drosophila , Inhibidores Enzimáticos/uso terapéutico , Femenino , Células HEK293 , Células HeLa , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/genética , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación , Multimerización de Proteína/efectos de los fármacos , Proteolisis/efectos de los fármacos , Proteína que Contiene Valosina/antagonistas & inhibidores , Proteína que Contiene Valosina/metabolismo
5.
Acta Pharmacol Sin ; 42(5): 801-813, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-32796956

RESUMEN

Grincamycins (GCNs) are a class of angucycline glycosides isolated from actinomycete Streptomyces strains that have potent antitumor activities, but their antitumor mechanisms remain unknown. In this study, we tried to identify the cellular target of grincamycin B (GCN B), one of most dominant and active secondary metabolites, using a combined strategy. We showed that GCN B-selective-induced apoptosis of human acute promyelocytic leukemia (APL) cell line NB4 through increase of ER stress and intracellular reactive oxygen species (ROS) accumulation. Using a strategy of combining phenotype, transcriptomics and protein microarray approaches, we identified that isocitrate dehydrogenase 1(IDH1) was the putative target of GCN B, and confirmed that GCNs were a subset of selective inhibitors targeting both wild-type and mutant IDH1 in vitro. It is well-known that IDH1 converts isocitrate to 2-oxoglutarate (2-OG), maintaining intracellular 2-OG homeostasis. IDH1 and its mutant as the target of GCN B were validated in NB4 cells and zebrafish model. Knockdown of IDH1 in NB4 cells caused the similar phenotype as GCN B treatment, and supplementation of N-acetylcysteine partially rescued the apoptosis caused by IDH1 interference in NB4 cells. In zebrafish model, GCN B effectively restored myeloid abnormality caused by overexpression of mutant IDH1(R132C). Taken together, we demonstrate that IDH1 is one of the antitumor targets of GCNs, suggesting wild-type IDH1 may be a potential target for hematological malignancies intervention in the future.


Asunto(s)
Antraquinonas/farmacología , Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Glicósidos/farmacología , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Animales , Antraquinonas/metabolismo , Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Estrés del Retículo Endoplásmico/efectos de los fármacos , Inhibidores Enzimáticos/metabolismo , Glicósidos/metabolismo , Humanos , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Ácidos Cetoglutáricos/metabolismo , Simulación del Acoplamiento Molecular , Mutación , Unión Proteica , Especies Reactivas de Oxígeno/metabolismo , Pez Cebra
6.
Acta Pharmacol Sin ; 41(9): 1246-1254, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32210356

RESUMEN

Mitophagy is a degradative pathway that mediates the degradation of the entire mitochondria, and defects in this process are implicated in many diseases including cancer. In mammals, mitophagy is mediated by BNIP3L (also known as NIX) that is a dual regulator of mitochondrial turnover and programmed cell death pathways. Acute myeloid leukemia (AML) cells with deficiency of BNIP3L are more sensitive to mitochondria-targeting drugs. But small molecular inhibitors for BNIP3L are currently not available. Some immunomodulatory drugs (IMiDs) have been proved by FDA for hematologic malignancies, however, the underlining molecular mechanisms are still elusive, which hindered the applications of BNIP3L inhibition for AML treatment. In this study we carried out MS-based quantitative proteomics analysis to identify the potential neosubstrates of a novel thalidomide derivative CC-885 in A549 cells. In total, we quantified 5029 proteins with 36 downregulated in CRBN+/+ cell after CC-885 administration. Bioinformatic analysis showed that macromitophagy pathway was enriched in the negative pathway after CC-885 treatment. We further found that CC-885 caused both dose- and time-dependent degradation of BNIP3L in CRBN+/+, but not CRBN-/- cell. Thus, our data uncover a novel role of CC-885 in the regulation of mitophagy by targeting BNIP3L for CRL4CRBN E3 ligase-dependent ubiquitination and degradation, suggesting that CC-885 could be used as a selective BNIP3L degradator for the further investigation. Furthermore, we demonstrated that CC-885 could enhance AML cell sensitivity to the mitochondria-targeting drug rotenone, suggesting that combining CC-885 and mitochondria-targeting drugs may be a therapeutic strategy for AML patients.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de la Membrana/metabolismo , Mitofagia/efectos de los fármacos , Compuestos de Fenilurea/farmacología , Proteínas Proto-Oncogénicas/metabolismo , Talidomida/análogos & derivados , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Línea Celular Tumoral , Sinergismo Farmacológico , Células HEK293 , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos , Rotenona/farmacología , Talidomida/farmacología , Ubiquitinación/efectos de los fármacos
8.
Cancer Biol Med ; 12(3): 163-74, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26487961

RESUMEN

OBJECTIVE: Human induced pluripotent stem (iPS) cells exhibit great potential for generating functional human cells for medical therapies. In this paper, we report for use of human iPS cells labeled with fluorescent magnetic nanoparticles (FMNPs) for targeted imaging and synergistic therapy of gastric cancer cells in vivo. METHODS: Human iPS cells were prepared and cultured for 72 h. The culture medium was collected, and then was co-incubated with MGC803 cells. Cell viability was analyzed by the MTT method. FMNP-labeled human iPS cells were prepared and injected into gastric cancer-bearing nude mice. The mouse model was observed using a small-animal imaging system. The nude mice were irradiated under an external alternating magnetic field and evaluated using an infrared thermal mapping instrument. Tumor sizes were measured weekly. RESULTS: iPS cells and the collected culture medium inhibited the growth of MGC803 cells. FMNP-labeled human iPS cells targeted and imaged gastric cancer cells in vivo, as well as inhibited cancer growth in vivo through the external magnetic field. CONCLUSION: FMNP-labeled human iPS cells exhibit considerable potential in applications such as targeted dual-mode imaging and synergistic therapy for early gastric cancer.

9.
Diabetes ; 64(12): 4061-74, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26324179

RESUMEN

Obesity is associated with chronic low-level inflammation, especially in fat tissues, which contributes to insulin resistance and type 2 diabetes mellitus (T2DM). Protein inhibitor of activated STAT 1 (PIAS1) modulates a variety of cellular processes such as cell proliferation and DNA damage responses. Particularly, PIAS1 functions in the innate immune system and is a key regulator of the inflammation cascade. However, whether PIAS1 is involved in the regulation of insulin sensitivity remains unknown. Here, we demonstrated that PIAS1 expression in white adipose tissue (WAT) was downregulated by c-Jun N-terminal kinase in prediabetic mice models. Overexpression of PIAS1 in inguinal WAT of prediabetic mice significantly improved systemic insulin sensitivity, whereas knockdown of PIAS1 in wild-type mice led to insulin resistance. Mechanistically, PIAS1 inhibited the activation of stress-induced kinases and the expression of nuclear factor-κB target genes in adipocytes, mainly including proinflammatory and chemotactic factors. In doing so, PIAS1 inhibited macrophage infiltration in adipose tissue, thus suppressing amplification of the inflammation cascade, which in turn improved insulin sensitivity. These results were further verified in a fat transplantation model. Our findings shed light on the critical role of PIAS1 in controlling insulin sensitivity and suggest a therapeutic potential of PIAS1 in T2DM.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Diabetes Mellitus Tipo 2/etiología , Resistencia a la Insulina , Macrófagos/metabolismo , Obesidad/metabolismo , Estado Prediabético/etiología , Proteínas Inhibidoras de STAT Activados/metabolismo , Tejido Adiposo Blanco/inmunología , Animales , Quimiotaxis , Diabetes Mellitus Tipo 2/prevención & control , Dieta Alta en Grasa/efectos adversos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas , Activación de Macrófagos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Obesidad/inmunología , Obesidad/fisiopatología , Obesidad/terapia , Estado Prediabético/prevención & control , Proteínas Inhibidoras de STAT Activados/antagonistas & inhibidores , Proteínas Inhibidoras de STAT Activados/genética , Células RAW 264.7 , Interferencia de ARN , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
10.
J Biol Chem ; 290(17): 11119-29, 2015 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-25778399

RESUMEN

The integration of signals involved in deciding the fate of mesenchymal stem cells is largely unknown. We used proteomics profiling to identify RhoGDIß, an inhibitor of the small G-protein Rho family, as a component that regulates commitment of C3H10T1/2 mesenchymal stem cells to the adipocyte or smooth muscle cell lineage in response to bone morphogenetic protein 4 (BMP4). RhoGDIß is notably down-regulated during BMP4-induced adipocytic lineage commitment of C3H10T1/2 mesenchymal stem cells, and this involves the cytoskeleton-associated protein lysyl oxidase. Excess RhoGDIß completely prevents BMP4-induced commitment to the adipocyte lineage and simultaneously stimulates smooth muscle cell commitment by suppressing the activation of Rac1. Overexpression of RhoGDIß induces stress fibers of F-actin by a process involving phosphomyosin light chain, indicating that cytoskeletal tension regulated by RhoGDIß contributes to determining adipocyte versus myocyte commitment. Furthermore, the overexpression of RacV12 (constitutively active form of Rac1) totally rescues the inhibition of adipocyte commitment by RhoGDIß, simultaneously preventing formation of the smooth muscle-like phenotype and disrupting the stress fibers in cells overexpressing RhoGDIß. Collectively, these results indicate that RhoGDIß functions as a novel BMP4 signaling target that regulates adipogenesis and myogensis.


Asunto(s)
Adipocitos/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular/fisiología , Desarrollo de Músculos/fisiología , Miocitos del Músculo Liso/metabolismo , Transducción de Señal/fisiología , Inhibidor beta de Disociación del Nucleótido Guanina rho/metabolismo , Adipocitos/citología , Animales , Proteína Morfogenética Ósea 4/genética , Línea Celular , Ratones , Miocitos del Músculo Liso/citología , Neuropéptidos/genética , Neuropéptidos/metabolismo , Fibras de Estrés/genética , Fibras de Estrés/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo , Inhibidor beta de Disociación del Nucleótido Guanina rho/genética
11.
Mol Cell Biol ; 33(22): 4606-17, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24061474

RESUMEN

It is well recognized that PIAS1, a SUMO (small ubiquitin-like modifier) E3 ligase, modulates such cellular processes as cell proliferation, DNA damage responses, and inflammation responses. Recent studies have shown that PIAS1 also plays a part in cell differentiation. However, the role of PIAS1 in adipocyte differentiation remains unknown. CCAAT/enhancer-binding protein ß (C/EBPß), a major regulator of adipogenesis, is a target of SUMOylation, but the E3 ligase responsible for the SUMOylation of C/EBPß has not been identified. The present study showed that PIAS1 functions as a SUMO E3 ligase of C/EBPß to regulate adipogenesis. PIAS1 expression was significantly and transiently induced on day 4 of 3T3-L1 adipocyte differentiation, when C/EBPß began to decline. PIAS1 was found to interact with C/EBPß through the SAP (scaffold attachment factor A/B/acinus/PIAS) domain and SUMOylate it, leading to increased ubiquitination and degradation of C/EBPß. C/EBPß became more stable when PIAS1 was silenced by RNA interference (RNAi). Moreover, adipogenesis was inhibited by overexpression of wild-type PIAS1 and promoted by knockdown of PIAS1. The mutational study indicated that the catalytic activity of SUMO E3 ligase was required for PIAS1 to restrain adipogenesis. Importantly, the inhibitory effect of PIAS1 overexpression on adipogenesis was rescued by overexpressed C/EBPß. Thus, PIAS1 could play a dynamic role in adipogenesis by promoting the SUMOylation of C/EBPß.


Asunto(s)
Adipogénesis , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Células 3T3-L1 , Animales , Proteína beta Potenciadora de Unión a CCAAT/química , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Proteínas Inhibidoras de STAT Activados/genética , Estabilidad Proteica , Proteolisis , Sumoilación , Ubiquitinación
12.
Mol Cell Biol ; 33(16): 3180-90, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23754749

RESUMEN

Autophagy is a highly conserved self-digestion pathway involved in various physiological and pathophysiological processes. Recent studies have implicated a pivotal role of autophagy in adipocyte differentiation, but the molecular mechanism for its role and how it is regulated during this process are not clear. Here, we show that CCAAT /enhancer-binding protein ß (C/EBPß), an important adipogenic factor, is required for the activation of autophagy during 3T3-L1 adipocyte differentiation. An autophagy-related gene, Atg4b, is identified as a de novo target gene of C/EBPß and is shown to play an important role in 3T3-L1 adipocyte differentiation. Furthermore, autophagy is required for the degradation of Klf2 and Klf3, two negative regulators of adipocyte differentiation, which is mediated by the adaptor protein p62/SQSTM1. Importantly, the regulation of autophagy by C/EBPß and the role of autophagy in Klf2/3 degradation and in adipogenesis are further confirmed in mouse models. Our data describe a novel function of C/EBPß in regulating autophagy and reveal the mechanism of autophagy during adipocyte differentiation. These new insights into the molecular mechanism of adipose tissue development provide a functional pathway with therapeutic potential against obesity and its related metabolic disorders.


Asunto(s)
Adipocitos/citología , Adipogénesis , Autofagia , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Cisteína Endopeptidasas/metabolismo , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Proteínas Relacionadas con la Autofagia , Cisteína Endopeptidasas/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Proteolisis , Factor de Transcripción TFIIH , Factores de Transcripción/metabolismo , Activación Transcripcional
13.
DNA Seq ; 15(3): 219-24, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15497447

RESUMEN

Present work reported the cloning and characterization of a human novel RNA binding gene Partner of NOB1 (PNO1), with a length of 1637bp and a putative open reading frame of 759 bp, isolated from human kidney. It is composed of seven exons and is localized on chromosome 2p14. Western blot showed that the molecular weight of PNO1 is about 35kDa. RT-PCR results in 16 human tissues indicated that PNO1 is expressed mainly in liver, lung, spleen and kidney, slightly in thymus, testis, ovary, respectively, but not in heart, brain, skeletal muscle, placenta, pancreas, prostate, small intestine, colon and peripheral blood leukocytes. GFP fusion expression in mammalian cells exhibited its localization in the nucleus, especially in nucleoli. Subcellular localization of thirteen GFP fusion PNO1 deletion proteins showed that the region of 92-230 aa is solely responsible for its nucleolar retention, and KH domain alone is not sufficient for nucleolar retention. The PNO1 family shows significant conservation in both eukaryotes and prokaryotes.


Asunto(s)
Cromosomas Humanos Par 2/genética , Proteínas de Unión al ARN/genética , Secuencia de Aminoácidos , Secuencia de Bases , Western Blotting , Nucléolo Celular/metabolismo , Clonación Molecular , Cartilla de ADN , Componentes del Gen , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes , Humanos , Riñón/metabolismo , Datos de Secuencia Molecular , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA