Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Ann Neurol ; 2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-39073169

RESUMEN

OBJECTIVE: Intellectual disability is often the outcome of neurodevelopmental disorders and is characterized by significant impairments in intellectual and adaptive functioning. X-linked intellectual disability (XLID) is a subset of these disorders caused by genetic defects on the X chromosome, affecting about 2 out of 1,000 males. In syndromic form, it leads to a broad range of cognitive, behavioral, ocular, and physical disabilities. METHODS: Employing exome or genome sequencing, here we identified 4 missense variants (c.475C > G; p.H159D, c.1373C > A; p.T458N, and c.1585G > A; p.E529K, c.953C > T; p.S318L) and a putative truncating variant (c.1413_1414del; p.Y471*) in the SRPK3 gene in 9 XLID patients from 5 unrelated families. To validate SRPK3 as a novel XLID gene, we established a knockout (KO) model of the SRPK3 orthologue in zebrafish. RESULTS: The 8 patients ascertained postnatally shared common clinical features including intellectual disability, agenesis of the corpus callosum, abnormal eye movement, and ataxia. A ninth case, ascertained prenatally, had a complex structural brain phenotype. Together, these data indicate a pathological role of SRPK3 in neurodevelopmental disorders. In post-fertilization day 5 larvae (free swimming stage), KO zebrafish exhibited severe deficits in eye movement and swim bladder inflation, mimicking uncontrolled ocular movement and physical clumsiness observed in human patients. In adult KO zebrafish, cerebellar agenesis and behavioral abnormalities were observed, recapitulating human phenotypes of cerebellar atrophy and intellectual disability. INTERPRETATION: Overall, these results suggest a crucial role of SRPK3 in the pathogenesis of syndromic X-linked intellectual disability and provide new insights into brain development, cognitive and ocular dysfunction in both humans and zebrafish. ANN NEUROL 2024.

2.
Int J Biol Macromol ; 273(Pt 1): 133005, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38866268

RESUMEN

Atopic dermatitis (AD) is a chronic cutaneous disease with a complex underlying mechanism, and it cannot be completely cured. Thus, most treatment strategies for AD aim at relieving the symptoms. Although corticosteroids are topically applied to alleviate AD, adverse side effects frequently lead to the withdrawal of AD therapy. Tacrolimus (TAC), a calcineurin inhibitor, has been used to treat AD, but its high molecular weight and insolubility in water hinder its skin permeability. Herein, we developed and optimized TAC-loaded chitosan-based nanoparticles (TAC@CNPs) to improve the skin permeability of TAC by breaking the tight junctions in the skin. The prepared nanoparticles were highly loadable and efficient and exhibited appropriate characteristics for percutaneous drug delivery. TAC@CNP was stable for 4 weeks under physiological conditions. CNP released TAC in a controlled manner, with enhanced skin penetration observed. In vitro experiments showed that CNP was non-toxic to keratinocyte (HaCaT) cells, and TAC@CNP dispersed in an aqueous solution was as anti-proliferative as TAC solubilized in a good organic solvent. Importantly, an in vivo AD mouse model revealed that topical TAC@CNP containing ~1/10 of the dose of TAC found in commercially used Protopic® Ointment exhibited similar anti-inflammatory activity to that of the commercial product. TAC@CNP represents a potential therapeutic strategy for the management of AD.


Asunto(s)
Quitosano , Dermatitis Atópica , Nanopartículas , Tacrolimus , Dermatitis Atópica/tratamiento farmacológico , Dermatitis Atópica/patología , Tacrolimus/química , Tacrolimus/farmacología , Tacrolimus/administración & dosificación , Tacrolimus/farmacocinética , Tacrolimus/uso terapéutico , Quitosano/química , Animales , Nanopartículas/química , Ratones , Humanos , Portadores de Fármacos/química , Piel/efectos de los fármacos , Piel/patología , Piel/metabolismo , Administración Tópica , Absorción Cutánea/efectos de los fármacos , Liberación de Fármacos , Modelos Animales de Enfermedad , Células HaCaT
3.
Exp Mol Med ; 56(3): 656-673, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38443596

RESUMEN

ISG15 is an interferon-stimulated ubiquitin-like protein (UBL) with multifaceted roles as a posttranslational modifier in ISG15 conjugation (ISGylation). However, the mechanistic consequences of ISGylation in cancer have not been fully elucidated, largely due to a lack of knowledge on the ISG15 target repertoire. Here, we identified SIRT1, a nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase, as a new target for ISGylation. SIRT1 ISGylation impairs the association of SIRT1 with its negative regulator, deleted in breast cancer 1 (DBC1), which unleashes SIRT1 from its inactive state and leads to an increase in its deacetylase activity. Importantly, SIRT1 ISGylation promoted lung cancer progression and limited lung cancer cell sensitivity to DNA damage-based therapeutics in vivo and in vitro models. The levels of ISG15 mRNA and protein were significantly higher in lung cancer tissues than in adjacent normal tissues. Accordingly, elevated expression of SIRT1 and ISG15 was associated with poor prognosis in lung cancer patients, a finding that could be translated for lung cancer patient stratification and disease outcome evaluation. Taken together, our findings provide a mechanistic understanding of the regulatory effect of SIRT1 ISGylation on tumor progression and therapeutic efficacy in lung cancer.


Asunto(s)
Neoplasias Pulmonares , Humanos , Interferones/metabolismo , Neoplasias Pulmonares/genética , Sirtuina 1/genética
4.
Cells ; 12(20)2023 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-37887348

RESUMEN

The mTOR signaling pathway integrates signaling inputs from nutrients, including glucose and amino acids, which are precisely regulated by transporters depending on nutrient levels. The L-type amino acid transporter 1 (LAT1) affects the activity of mTORC1 through upstream regulators that sense intracellular amino acid levels. While mTORC1 activation by LAT1 has been thoroughly investigated in cultured cells, the effects of LAT1 expression on the activity of mTORC2 has scarcely been studied. Here, we provide evidence that LAT1 recruits and activates mTORC2 on the lysosome for PMA-induced cell migration. LAT1 is translocated to the lysosomes in cells treated with PMA in a dose- and time-dependent manner. Lysosomal LAT1 interacted with mTORC2 through a direct interaction with Rictor, leading to the lysosomal localization of mTORC2. Furthermore, the depletion of LAT1 reduced PMA-induced cell migration in a wound-healing assay. Consistent with these results, the LAT1 N3KR mutant, which is defective in PMA-induced endocytosis and lysosomal localization, did not induce mTORC2 recruitment to the lysosome, with the activation of mTORC2 determined via Akt phosphorylation or the LAT1-mediated promotion of cell migration. Taken together, lysosomal LAT1 recruits and activates the mTORC2 complex and downstream Akt for PMA-mediated cell migration. These results provide insights into the development of therapeutic drugs targeting the LAT1 amino acid transporter to block metastasis, as well as disease progression in various types of cancer.


Asunto(s)
Transportador de Aminoácidos Neutros Grandes 1 , Lisosomas , Proteínas Proto-Oncogénicas c-akt , Movimiento Celular/fisiología , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/metabolismo
5.
Int J Biol Macromol ; 194: 347-357, 2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-34801583

RESUMEN

Site-specific ubiquitination can regulate the functions of Rab proteins in membrane trafficking. Previously we showed that site-specific monoubiquitination on Rab5 downregulates its function. Rab7 acts in the downstream of Rab5. Although site-specific ubiquitination of Rab7 can affect its function, it remains elusive how the ubiquitination is involved in modulation of the function of Rab7 at molecular level. Here, we report molecular basis for the regulation of Rab7 by site-specific monoubiquitination. Rab7 was predominantly monoubiquitinated at multiple sites in the membrane fraction of cultured cells. Two major ubiquitination sites (K191 and K194), identified by mutational analysis with single K mutants, were responsible for membrane localization of monoubiquitinated Rab7. Using small-angle X-ray scattering, we derived structural models of site-specifically monoubiquitinated Rab7 in solution. Structural analysis combined with molecular dynamics simulation corroborated that the ubiquitin moieties on K191 and K194 are key determinants for exclusion of Rab7 from the endosomal membrane. Ubiquitination on the two major sites apparently mitigated colocalization of Rab7 with ORF3a of SARS-CoV-2, potentially deterring the egression of SARS-CoV-2. Our results establish that the regulatory effects of a Rab protein through site-specific monoubiquitination are commonly observed among Rab GTPases while the ubiquitination sites differ in each Rab protein.


Asunto(s)
SARS-CoV-2/metabolismo , Proteínas Virales/metabolismo , Proteínas de Unión a GTP rab7/metabolismo , Células HEK293 , Células HeLa , Humanos , Unión Proteica , Ubiquitinación
6.
J Cell Mol Med ; 25(1): 473-483, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33230972

RESUMEN

Osteoporosis is a disease in which bone mineral density decreases due to abnormal activity of osteoclasts, and is commonly found in post-menopausal women who have decreased levels of female hormones. Sphingosylphosphorylcholine (SPC) is an important biological lipid that can be converted to sphingosine-1-phosphate (S1P) by autotaxin. S1P is known to be involved in osteoclast activation by stimulating osteoblasts, but bone regulation by SPC is not well understood. In this study, we found that SPC strongly inhibits RANKL-induced osteoclast differentiation. SPC-induced inhibitory effects on osteoclast differentiation were not affected by several antagonists of S1P receptors or pertussis toxin, suggesting cell surface receptor independency. However, SPC inhibited RANKL-induced calcineurin activation and subsequent NFATc1 activity, leading to decrease of the expression of Trap and Ctsk. Moreover, we found that bone loss in an experimental osteoporosis mouse model was recovered by SPC injection. SPC also blocked ovariectomy-induced body weight increase and Nfatc1 gene expression in mice. We also found that SPC inhibits RANKL-induced osteoclast differentiation in human macrophages. Since currently available treatments for osteoporosis, such as administration of female hormones or hormone receptor modulators, show serious side effects, SPC has potential as a new agent for osteoporosis treatment.


Asunto(s)
Calcio/metabolismo , Calmodulina/metabolismo , Osteoclastos/metabolismo , Osteoporosis/metabolismo , Ovariectomía/efectos adversos , Fosforilcolina/análogos & derivados , Esfingosina/análogos & derivados , Animales , Western Blotting , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/metabolismo , Supervivencia Celular/efectos de los fármacos , Femenino , Ratones , Ratones Endogámicos C57BL , Osteoclastos/efectos de los fármacos , Osteoporosis/tratamiento farmacológico , Fosforilcolina/uso terapéutico , Reacción en Cadena en Tiempo Real de la Polimerasa , Esfingosina/uso terapéutico , Microtomografía por Rayos X
7.
Int J Mol Sci ; 21(18)2020 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-32971846

RESUMEN

Interleukin 33 (IL-33) is an IL-1 family cytokine that plays a central role in immune system by regulating and initiating inflammatory responses. The binding of IL-33 to the suppressor of tumorigenicity 2 (ST2) receptor induces mitogen-activated protein kinases (MAPK) and nuclear factor κB (NF-κB) pathways, thereby leading to inflammatory cytokines production in type 2 helper T cells and type 2 innate lymphoid cells. To develop an antibody specific to IL-33 with a defined epitope, we characterized a single-chain antibody variable fragments (scFvs) clone specific to IL-33, C2_2E12, which was selected from a human synthetic library of scFvs using phage display. Affinity (Kd) of C2_2E12 was determined to be 38 nM using enzyme-linked immunosorbent assay. C2_2E12 did not show cross-reactivity toward other interleukin cytokines, including closely related IL-1 family cytokines and unrelated proteins. Mutational scanning analysis revealed that the epitope of IL-33 consisted of residues 149-158 with key residues being L150 and K151 of IL-33. Structural modeling suggested that L150 and K151 residues are important for the interaction of IL-33 with C2_2E12, implicating that C2_2E12 could block the binding of ST2 to IL-33. Pull-down and in-cell assays supported that C2_2E12 can inhibit the IL-33/ST2 signaling axis. These results suggest that the scFv clone characterized here can function as a neutralizing antibody.


Asunto(s)
Epítopos , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33 , Sistema de Señalización de MAP Quinasas/inmunología , Anticuerpos de Cadena Única , Línea Celular , Epítopos/química , Epítopos/inmunología , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/química , Proteína 1 Similar al Receptor de Interleucina-1/inmunología , Interleucina-33/antagonistas & inhibidores , Interleucina-33/química , Interleucina-33/inmunología , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/inmunología
8.
Cancer Immunol Res ; 8(11): 1393-1406, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32878747

RESUMEN

Regulatory T cells (Treg) are enriched in the tumor microenvironment (TME) and suppress antitumor immunity; however, the molecular mechanism underlying the accumulation of Tregs in the TME is poorly understood. In various tumor models, tumor-infiltrating Tregs were highly enriched in the TME and had significantly higher expression of immune checkpoint molecules. To characterize tumor-infiltrating Tregs, we performed bulk RNA sequencing (RNA-seq) and found that proliferation-related genes, immune suppression-related genes, and cytokine/chemokine receptor genes were upregulated in tumor-infiltrating Tregs compared with tumor-infiltrating CD4+Foxp3- conventional T cells or splenic Tregs from the same tumor-bearing mice. Single-cell RNA-seq and T-cell receptor sequencing also revealed active proliferation of tumor infiltrating Tregs by clonal expansion. One of these genes, ST2, an IL33 receptor, was identified as a potential factor driving Treg accumulation in the TME. Indeed, IL33-directed ST2 signaling induced the preferential proliferation of tumor-infiltrating Tregs and enhanced tumor progression, whereas genetic deletion of ST2 in Tregs limited their TME accumulation and delayed tumor growth. These data demonstrated the IL33/ST2 axis in Tregs as one of the critical pathways for the preferential accumulation of Tregs in the TME and suggests that the IL33/ST2 axis may be a potential therapeutic target for cancer immunotherapy.


Asunto(s)
Inmunoterapia/métodos , Interleucina-33/metabolismo , Linfocitos T Reguladores/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Transducción de Señal , Microambiente Tumoral
9.
Int J Mol Sci ; 21(18)2020 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-32906784

RESUMEN

The root bark of Morus has long been appreciated as an antiphlogistic, diuretic and expectorant drug in Chinese herbal medicine, albeit with barely known targets and mechanisms of action. In the 1970s, the development of analytic chemistry allowed for the discovery of morusin as one of 7 different isoprene flavonoid derivatives in the root bark of Morus. However, the remarkable antioxidant capacity of morusin with the unexpected potential for health benefits over the other flavonoid derivatives has recently sparked scientific interest in the biochemical identification of target proteins and signaling pathways and further clinical relevance. In this review, we discuss recent advances in the understanding of the functional roles of morusin in multiple biological processes such as inflammation, apoptosis, metabolism and autophagy. We also highlight recent in vivo and in vitro evidence on the clinical potential of morusin treatment for multiple human pathologies including inflammatory diseases, neurological disorders, diabetes, cancer and the underlying mechanisms.


Asunto(s)
Flavonoides/metabolismo , Flavonoides/farmacología , Morus/metabolismo , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Butadienos/química , Flavonoides/química , Hemiterpenos/química , Humanos , Inflamación/tratamiento farmacológico , Corteza de la Planta/metabolismo , Extractos Vegetales/farmacología , Raíces de Plantas/metabolismo , Transducción de Señal/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos
10.
Int J Mol Sci ; 21(15)2020 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-32731602

RESUMEN

Stress granules are membraneless organelles composed of numerous components including ribonucleoproteins. The stress granules are characterized by a dynamic complex assembly in response to various environmental stressors, which has been implicated in the coordinated regulation of diverse biological pathways, to exert a protective role against stress-induced cell death. Here, we show that stress granule formation is induced by morusin, a novel phytochemical displaying antitumor capacity through barely known mechanisms. Morusin-mediated induction of stress granules requires activation of protein kinase R (PKR) and subsequent eIF2α phosphorylation. Notably, genetic inactivation of stress granule formation mediated by G3BP1 knockout sensitized cancer cells to morusin treatment. This protective function against morusin-mediated cell death can be attributed at least in part to the sequestration of receptors for activated C kinase-1 (RACK1) within the stress granules, which reduces caspase-3 activation. Collectively, our study provides biochemical evidence for the role of stress granules in suppressing the antitumor capacity of morusin, proposing that morusin treatment, together with pharmacological inhibition of stress granules, could be an efficient strategy for targeting cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Gránulos Citoplasmáticos/metabolismo , Flavonoides/farmacología , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Receptores de Cinasa C Activada/metabolismo , eIF-2 Quinasa/metabolismo , Gránulos Citoplasmáticos/patología , Células HCT116 , Células HeLa , Humanos , Células PC-3
11.
Mol Cell ; 75(5): 1058-1072.e9, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31375263

RESUMEN

The endoplasmic reticulum (ER) is susceptible to wear-and-tear and proteotoxic stress, necessitating its turnover. Here, we show that the N-degron pathway mediates ER-phagy. This autophagic degradation initiates when the transmembrane E3 ligase TRIM13 (also known as RFP2) is ubiquitinated via the lysine 63 (K63) linkage. K63-ubiquitinated TRIM13 recruits p62 (also known as sequestosome-1), whose complex undergoes oligomerization. The oligomerization is induced when the ZZ domain of p62 is bound by the N-terminal arginine (Nt-Arg) of arginylated substrates. Upon activation by the Nt-Arg, oligomerized TRIM13-p62 complexes are separated along with the ER compartments and targeted to autophagosomes, leading to lysosomal degradation. When protein aggregates accumulate within the ER lumen, degradation-resistant autophagic cargoes are co-segregated by ER membranes for lysosomal degradation. We developed synthetic ligands to the p62 ZZ domain that enhance ER-phagy for ER protein quality control and alleviate ER stresses. Our results elucidate the biochemical mechanisms and pharmaceutical means that regulate ER homeostasis.


Asunto(s)
Proteínas Portadoras/metabolismo , Retículo Endoplásmico/metabolismo , Proteolisis , Proteína Sequestosoma-1/metabolismo , Animales , Proteínas Portadoras/genética , Retículo Endoplásmico/genética , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Noqueados , Proteína Sequestosoma-1/genética , Ubiquitinación
12.
Am J Cancer Res ; 9(1): 94-107, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30755814

RESUMEN

Covalent conjugations of the SUMO-1 moiety on a target protein play important roles in the regulation of cellular protein function. SUMO-conjugation of PML is a regulatory step for PML nuclear body (PML-NB) formation, and HIPK2 is SUMO-conjugated and recruited into the PML-NBs. Although HIPK2 mutations (R861W and N951I) were found in acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) patients, little is known about the underlying mechanisms by which HIPK2 mutations are associated with the pathogenesis of leukemia. Here we show that HIPK2 mutants found in AML and MDS patients are defective in SUMO-interacting motif (SIM) function. Due to defective SIM function, the HIPK2 mutants were not modified with SUMO-1, and not recruited to the PML-NBs. However, the HIPK2 mutants can normally bind to and phosphorylate AML1b. Therefore, the HIPK2 mutants can sequestrate the AML1 complex out of the PML-NBs, resulting in the disruption of AML1-mediated activation of target genes for myeloid differentiation. In addition, the differentiation of K562 blast cells was impaired by the expression of the HIPK2 SIM-defective mutants. These results suggest that HIPK2 targeting into the PML-NBs via the SIMs is crucial for HIPK2-mediated induction of myeloid differentiation, and is associated with AML pathogenesis.

13.
Plant Cell Physiol ; 60(4): 826-834, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-30605532

RESUMEN

Plant cells have a remarkable plasticity that allows cellular reprogramming from differentiated cells and subsequent tissue regeneration. Callus formation occurs from pericycle-like cells through a lateral root developmental pathway, and even aerial parts can also undergo the cell fate transition. Pluripotent calli are then subjected primarily to shoot regeneration in in vitro tissue culture. Successful completion of plant regeneration from aerial explants thus entails a two-step conversion of tissue identity. Here we show that a single chromatin modifier, ARABIDOPSIS TRITHORAX 4 (ATX4)/SET DOMAIN GROUP 16, is dynamically regulated during plant regeneration to address proper callus formation and shoot regeneration. The ATX4 protein massively activates shoot identity genes by conferring H3K4me3 deposition at the loci. ATX4-deficient mutants display strong silencing of shoot identity and thus enhanced callus formation. Subsequently, de novo shoot organogenesis from calli is impaired in atx4 mutants. These results indicate that a series of epigenetic reprogramming of tissue identity underlies plant regeneration, and molecular components defining tissue identity can be used as invaluable genetic sources for improving crop transformation efficiency.


Asunto(s)
Proteínas de Arabidopsis/genética , Arabidopsis/genética , Epigénesis Genética/genética , Regulación de la Expresión Génica de las Plantas/genética , Histonas/genética , Histonas/metabolismo , Plantas Modificadas Genéticamente/genética
14.
EMBO Rep ; 19(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29487085

RESUMEN

Autophagy begins with the formation of autophagosomes, a process that depends on the activity of the serine/threonine kinase ULK1 (hATG1). Although earlier studies indicated that ULK1 activity is regulated by dynamic polyubiquitination, the deubiquitinase involved in the regulation of ULK1 remained unknown. In this study, we demonstrate that ubiquitin-specific protease 20 (USP20) acts as a positive regulator of autophagy initiation through stabilizing ULK1. At basal state, USP20 binds to and stabilizes ULK1 by removing the ubiquitin moiety, thereby interfering with the lysosomal degradation of ULK1. The stabilization of basal ULK1 protein levels is required for the initiation of starvation-induced autophagy, since the depletion of USP20 by RNA interference inhibits LC3 puncta formation, a marker of autophagic flux. At later stages of autophagy, USP20 dissociates from ULK1, resulting in enhanced ULK1 degradation and apoptosis. Taken together, our findings provide the first evidence that USP20 plays a crucial role in autophagy initiation by maintaining the basal expression level of ULK1.


Asunto(s)
Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Autofagia , Ubiquitina Tiolesterasa/metabolismo , Animales , Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Línea Celular , Supervivencia Celular , Expresión Génica , Células HEK293 , Humanos , Lisosomas/metabolismo , Ratones , Unión Proteica , Estabilidad Proteica , Proteolisis , Interferencia de ARN , ARN Interferente Pequeño/genética , Ubiquitina Tiolesterasa/genética , Ubiquitinación
15.
Sci Rep ; 7(1): 16136, 2017 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-29170424

RESUMEN

Homeodomain-interacting protein kinase 2 (HIPK2) is a nuclear serine/threonine kinase that functions in development and tumor suppression. One of the prominent features of this kinase is that it is tightly regulated by proteasomal degradation. In the present study, we present evidence suggesting that the protein stability of HIPK2 can be regulated by p300-mediated acetylation. p300 increased the protein level of HIPK2 via its acetyltransferase activity. p300 increased the acetylation of HIPK2 while decreased polyubiquitination and its proteasomal degradation. We also observed that DNA damage induced acetylation of HIPK2 along with an increase in the protein amount, which was inhibited by p300 RNAi. Importantly, p300 promoted p53 activation and the HIPK2-mediated suppression of cell proliferation, suggesting acetylation-induced HIPK2 stabilization contributed to the enhanced activation of HIPK2. Overexpression of p300 promoted the HIPK2-mediated suppression of tumor growth in mouse xenograft model as well. Taken together, our data suggest that p300-mediated acetylation of HIPK2 increases the protein stability of HIPK2 and enhances its tumor suppressor function.


Asunto(s)
Proteínas Portadoras/metabolismo , Supervivencia Celular/fisiología , Proteína p300 Asociada a E1A/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Acetilación , Animales , Western Blotting , Proteínas Portadoras/genética , Supervivencia Celular/genética , Proteína p300 Asociada a E1A/genética , Células HEK293 , Humanos , Inmunoprecipitación , Ratones , Ratones Endogámicos BALB C , Proteínas Serina-Treonina Quinasas/genética , Estabilidad Proteica , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Elife ; 62017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28968219

RESUMEN

Rab GTPases, which are involved in intracellular trafficking pathways, have recently been reported to be ubiquitinated. However, the functions of ubiquitinated Rab proteins remain unexplored. Here we show that Rab5 is monoubiquitinated on K116, K140, and K165. Upon co-transfection with ubiquitin, Rab5 exhibited abnormalities in endosomal localization and EGF-induced EGF receptor degradation. Rab5 K140R and K165R mutants restored these abnormalities, whereas K116R did not. We derived structural models of individual monoubiquitinated Rab5 proteins (mUbRab5s) by solution scattering and observed different conformational flexibilities in a site-specific manner. Structural analysis combined with biochemical data revealed that interactions with downstream effectors were impeded in mUbRab5K140, whereas GDP release and GTP loading activities were altered in mUbRab5K165. By contrast, mUbRab5K116 apparently had no effect. We propose a regulatory mechanism of Rab5 where monoubiquitination downregulates effector recruitment and GDP/GTP conversion in a site-specific manner.


Asunto(s)
Regulación hacia Abajo , Nucleótidos de Guanina/metabolismo , Ubiquitinación , Proteínas de Unión al GTP rab5/metabolismo , Línea Celular , Análisis Mutacional de ADN , Humanos , Hidrólisis , Unión Proteica , Conformación Proteica , Dispersión del Ángulo Pequeño , Proteínas de Unión al GTP rab5/química , Proteínas de Unión al GTP rab5/genética
17.
Am J Cancer Res ; 7(3): 518-530, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28401008

RESUMEN

Autophagy is a cellular process by which damaged organelles and dysfunctional proteins are degraded. Morusin is an anti-cancer drug isolated from the root bark of Morus alba. Morusin induces apoptosis in human prostate cancer cells by reducing STAT3 activity. In this study, we examined whether morusin induces autophagy and also examined the effects of autophagy on the morusin-induced apoptosis. Morusin induces LC3-II accumulation and ULK1 activation in HeLa cells. In addition, we found that induction of ULK1 Ser317 phosphorylation and reduction of ULK1 Ser757 phosphorylation occurred simultaneously during morusin-induced autophagy. Consistently, morusin induces autophagy by activation of AMPK and inhibition of mTOR activity. Next, we investigated the role of autophagy in morusin-induced apoptosis. Inhibition of autophagy by treating cells with the 3-methyladenine (3-MA) autophagic inhibitor induces high levels of morusin-mediated apoptosis, while treatment of cells with morusin alone induces moderate levels of apoptosis. Cell survival was greatly reduced when cells were treated with morusin and 3-MA. Taken together, morusin induces autophagy, which is an impediment for morusin-induced apoptosis, suggesting combined treatment of morusin with an autophagic inhibitor would increase the efficacy of morusin as an anti-cancer drug.

18.
Cell Death Dis ; 7(10): e2438, 2016 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-27787517

RESUMEN

Homeodomain-interacting protein kinase 2 (HIPK2) is a nuclear serine/threonine kinase that functions in DNA damage response and development. In the present study, we propose that the protein stability and proapoptotic function of HIPK2 are regulated by poly(ADP-ribose) polymerase 1 (PARP1). We present evidence indicating that PARP1 promotes the proteasomal degradation of HIPK2. The tryptophan-glycine-arginine (WGR) domain of PARP1 was necessary and sufficient for the promotion of HIPK2 degradation independently of the PARP1 enzymatic activity. The WGR domain mediated the interaction between HIPK2 and C-terminus of HSP70-interacting protein (CHIP) via HSP70. We found that CHIP can function as a ubiquitin ligase for HIPK2. The interaction between PAPR1 and HIPK2 was weakened following DNA damage. Importantly, PARP1 reduced the HIPK2-mediated p53 phosphorylation, proapoptotic transcriptional activity and cell death. These results suggest that PARP1 can modulate the tumor-suppressing function of HIPK2 by regulating the protein stability of HIPK2.


Asunto(s)
Apoptosis , Proteínas Portadoras/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Daño del ADN , Células HCT116 , Células HEK293 , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Poli(ADP-Ribosa) Polimerasas/química , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Proteolisis , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
19.
Electrophoresis ; 37(22): 3002-3009, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27510307

RESUMEN

DNA profiling of short tandem repeats (STR) has been successfully used for the identification of individuals in forensic samples, accidents and natural disasters. However, STR profiling of DNA isolated from old crime scenes and damaged biological samples is difficult due to DNA degradation and fragmentation. Here, we show that pre-amplification of STR loci using biotinylated primers for the STR loci is an efficient strategy to obtain STR profiling results from fragmented forensic samples. Analysis of STR loci with longer amplicon sizes is generally hampered, since these relatively long loci are vulnerable to DNA fragmentation. This problem was overcome by using reduced or increased primer concentrations for loci with shorter or longer amplicon sizes, respectively, in our pre-amplification strategy. In addition, pre-amplification of STR loci into two groups of short or long amplicon size increases the efficiency of STR profiling from highly fragmented forensic DNA samples. Therefore, differential pre-amplification of STR loci is an effective way to obtain DNA profiling results from fragmented forensic samples.


Asunto(s)
Dermatoglifia del ADN/métodos , ADN/análisis , Genética Forense/métodos , Repeticiones de Microsatélite/genética , Reacción en Cadena de la Polimerasa/métodos , ADN/química , Células HeLa , Humanos
20.
Cancer Res ; 76(16): 4728-40, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27335110

RESUMEN

The receptor Notch1 plays an important role in malignant progression of many cancers, but its regulation is not fully understood. In this study, we report that the kinase HIPK2 is responsible for facilitating the Fbw7-dependent proteasomal degradation of Notch1 by phosphorylating its intracellular domain (Notch1-IC) within the Cdc4 phosphodegron motif. Notch1-IC expression was higher in cancer cells than normal cells. Under genotoxic stress, Notch1-IC was phosphorylated constitutively by HIPK2 and was maintained at a low level through proteasomal degradation. HIPK2 phosphorylated the residue T2512 in Notch1-IC. Somatic mutations near this residue rendered Notch1-IC resistant to degradation, as induced either by HIPK2 overexpression or adriamycin treatment. In revealing an important mechanism of Notch1 stability, the results of this study could offer a therapeutic strategy to block Notch1-dependent progression in many types of cancer. Cancer Res; 76(16); 4728-40. ©2016 AACR.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas Portadoras/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Notch1/metabolismo , Animales , Western Blotting , Neoplasias de la Mama/metabolismo , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Femenino , Técnica del Anticuerpo Fluorescente , Xenoinjertos , Humanos , Immunoblotting , Inmunoprecipitación , Ratones , Mutación , Invasividad Neoplásica/patología , Fosforilación , Reacción en Cadena de la Polimerasa , Estabilidad Proteica , Receptor Notch1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA