Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Br J Cancer ; 130(6): 1046-1058, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38278978

RESUMO

BACKGROUND: The repurposing of FDA-approved drugs for anti-cancer therapies is appealing due to their established safety profiles and pharmacokinetic properties and can be quickly moved into clinical trials. Cancer progression and resistance to conventional chemotherapy remain the key hurdles in improving the clinical management of colon cancer patients and associated mortality. METHODS: High-throughput screening (HTS) was performed using an annotated library of 1,600 FDA-approved drugs to identify drugs with strong anti-CRC properties. The candidate drug exhibiting most promising inhibitory effects in in-vitro studies was tested for its efficacy using in-vivo models of CRC progression and chemoresistance and patient derived organoids (PTDOs). RESULTS: Albendazole, an anti-helminth drug, demonstrated the strongest inhibitory effects on the tumorigenic potentials of CRC cells, xenograft tumor growth and organoids from mice. Also, albendazole sensitized the chemoresistant CRC cells to 5-fluorouracil (5-FU) and oxaliplatin suggesting potential to treat chemoresistant CRC. Mechanistically, Albendazole treatment modulated the expression of RNF20, to promote apoptosis in CRC cells by delaying the G2/M phase and suppressing anti-apoptotic-Bcl2 family transcription. CONCLUSIONS: Albendazole, an FDA approved drug, carries strong therapeutic potential to treat colon cancers which are aggressive and potentially resistant to conventional chemotherapeutic agents. Our findings also lay the groundwork for further clinical testing.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Humanos , Animais , Camundongos , Albendazol/farmacologia , Albendazol/uso terapêutico , Neoplasias Colorretais/patologia , Ubiquitina/farmacologia , Ubiquitina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Fluoruracila/uso terapêutico , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Ubiquitina-Proteína Ligases
2.
Med Oncol ; 40(11): 325, 2023 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-37805625

RESUMO

Cytarabine-resistant acute myeloid leukemia (AML) is a common phenomenon, necessitating the search for new chemotherapeutics. WEE1 participates in cell cycle checkpoint signaling and inhibitors targeting WEE1 (WEE1i) constitute a potential novel strategy for AML treatment. HDAC (histone deacetylase) inhibitors have been shown to enhance the anti-tumor effects of WEE1i but molecular mechanisms of HDAC remain poorly characterized. In this study, the WEE1 inhibitor PD0166285 showed a relatively good anti-leukemia effect. Notably, PD0166285 can arise the expression of HDAC11 which was negatively correlated with survival of AML patients. Moreover, HDAC11 can reduced the anti-tumor effect of PD0166285 through an effect on p53 stability and the changes in phosphorylation levels of MAPK pathways. Overall, the cell cycle inhibitor, PD0166285, is a potential chemotherapeutic drug for AML. These fundings contribute to a functional understanding of HDAC11 in AML.


Assuntos
Proteínas de Ciclo Celular , Leucemia Mieloide Aguda , Humanos , Proteínas de Ciclo Celular/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina/metabolismo , Ubiquitina/farmacologia , Ubiquitina/uso terapêutico , Proteínas Nucleares/metabolismo , Leucemia Mieloide Aguda/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Histona Desacetilases/metabolismo , Apoptose , Linhagem Celular Tumoral
3.
Am J Physiol Endocrinol Metab ; 325(5): E500-E512, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37672249

RESUMO

The ubiquitin-proteasome system (UPS) and the autophagy-lysosome pathway are the primary means of degradation in mammalian tissues. We sought to determine the individual contribution of the UPS and autophagy to tissue catabolism during fasting. Mice were overnight fasted for 15 h before regaining food access ("Fed" group, n = 6) or continuing to fast ("Fast" group, n = 7) for 3 h. In addition, to investigate the effects of autophagy on systemic metabolism and tissue degradation, one group of mice was fasted for 18 h and treated with chloroquine ("Fast + CLQ" group, n = 7) and a fourth group of mice was treated with bortezomib ("Fast + Bort" group, n = 7) to assess the contribution of the UPS. Body weight, tissue weight, circulating hormones and metabolites, intracellular signaling pathways, and protein synthesis were investigated. Fasting induced the loss of body weight, liver mass, and white adipose tissue in the Fast and the Fast + CLQ group, whereas the Fast + Bort group maintained tissue and body weight. Fasting reduced glucose and increased ß hydroxybutyrate in the circulation of all mice. Both changes were most profound in the Fast + Bort group compared with the other fasting conditions. Molecular signaling indicated a successful inhibition of hepatic UPS with bortezomib and an upregulation of the PI3K/AKT/mTOR pathway. The latter was further supported by an increase in hepatic protein synthesis with bortezomib. Inhibition of the UPS through bortezomib blocks body weight loss and tissue catabolism during an acute overnight fast in mice. The effects were likely mediated through a combined effect of the drug on biomolecule degradation and synthesis.NEW & NOTEWORTHY Bortezomib treatment prevents tissue and body weight loss during fasting. The loss of proteasome activity with bortezomib exacerbates fasting-induced ketogenesis. During fasting, bortezomib increases AMPK and PI3K/AKT signaling in the liver, which promotes protein synthesis.


Assuntos
Fosfatidilinositol 3-Quinases , Complexo de Endopeptidases do Proteassoma , Camundongos , Animais , Complexo de Endopeptidases do Proteassoma/metabolismo , Bortezomib/farmacologia , Proteólise , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ubiquitina/metabolismo , Ubiquitina/farmacologia , Jejum/metabolismo , Nutrientes , Redução de Peso , Peso Corporal , Autofagia , Mamíferos/metabolismo
4.
Int Immunopharmacol ; 124(Pt B): 110896, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37729796

RESUMO

Elevated evidence has reported the important role of oxidative stress injury and inflammatory response in the progression of colitis. Tumor Suppressor TSBF1, TRIM59, is a ubiquitin E3 ligase and mediates immune response. However, the underlying molecular function of TRIM59 on regulation of colitis is still not understood. In the current study, we identify the TRIM59 as a critical and novel endogenous suppressor of kelch-like ECH-associated protein 1 (KEAP1), and we also determine that TRIM59 is a KEAP1-interacting partner protein that catalyses its ubiquitination and degradation in intestinal epithelial cells (IEC). Moreover, IEC-specific loss of the Trim59 disrupts colon metabolic homeostasis, accompanied by intestinal oxidative stress injury, elevated endogenous reactive oxygen species (ROS) production and pro-inflammatory cytokines release, significantly promotes acute or chronic colitis progression. Conversely, transgenic mice with Trim59 overexpression by adeno-associated virus (AAV)-induced Trim59 gene therapeutics mitigates colitis in acute or chronic colitis rodent models and in vitro experiments. Mechanistically, in response to onset of colitis, TRIM59 directly interacts with KEAP1 and promotes ubiquitin-proteasome degradation, thus results in NRF2 activation and its downstream cascade anti-oxidative stress-related pathway activation, which facilitates anti-oxidant defense and reduces tissue damage. All the findings elucidated the potential role of TRIM59 in colitis progression by mediating KEAP1 deactivation and degradation, and could be considered as a therapeutic target for the treatment of such disease.


Assuntos
Colite , Fator 2 Relacionado a NF-E2 , Animais , Camundongos , Doença Crônica , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Camundongos Transgênicos , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Transdução de Sinais , Ubiquitina/metabolismo , Ubiquitina/farmacologia
5.
Brain Pathol ; 33(5): e13175, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37259617

RESUMO

Alpha-synuclein (αsyn) aggregates are pathological features of several neurodegenerative conditions including Parkinson disease (PD), dementia with Lewy bodies, and multiple system atrophy (MSA). Accumulating evidence suggests that mitochondrial dysfunction and impairments of the autophagic-lysosomal system can contribute to the deposition of αsyn, which in turn may interfere with health and function of these organelles in a potentially vicious cycle. Here we investigated a potential convergence of αsyn with the PINK1-PRKN-mediated mitochondrial autophagy pathway in cell models, αsyn transgenic mice, and human autopsy brain. PINK1 and PRKN identify and selectively label damaged mitochondria with phosphorylated ubiquitin (pS65-Ub) to mark them for degradation (mitophagy). We found that disease-causing multiplications of αsyn resulted in accumulation of the ubiquitin ligase PRKN in cells. This effect could be normalized by starvation-induced autophagy activation and by CRISPR/Cas9-mediated αsyn knockout. Upon acute mitochondrial damage, the increased levels of PRKN protein contributed to an enhanced pS65-Ub response. We further confirmed increased pS65-Ub-immunopositive signals in mouse brain with αsyn overexpression and in postmortem human disease brain. Of note, increased pS65-Ub was associated with neuronal Lewy body-type αsyn pathology, but not glial cytoplasmic inclusions of αsyn as seen in MSA. While our results add another layer of complexity to the crosstalk between αsyn and the PINK1-PRKN pathway, distinct mechanisms may underlie in cells and brain tissue despite similar outcomes. Notwithstanding, our finding suggests that pS65-Ub may be useful as a biomarker to discriminate different synucleinopathies and may serve as a potential therapeutic target for Lewy body disease.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Animais , Humanos , Camundongos , alfa-Sinucleína/metabolismo , Camundongos Transgênicos , Mitofagia , Doença de Parkinson/metabolismo , Proteínas Quinases/metabolismo , Proteínas Quinases/farmacologia , Ubiquitina/metabolismo , Ubiquitina/farmacologia , Ubiquitina-Proteína Ligases/genética
6.
J Innate Immun ; 15(1): 428-441, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36944318

RESUMO

The correlation between ubiquitin-editing enzyme A20 and E3 ubiquitin ligase ring finger protein (RNF) 168 has been reported to be critical for repair of DNA damage. This study aimed to evaluate the potential role of this regulatory interaction in the pathogenesis of lupus nephritis (LN). The expression of RNF168 and A20 was measured in the podocytes derived from MRL/lpr murine lupus as well as patients with LN. Cell-based studies using renal podocytes bearing silenced RNF168, over-expressed A20, autophagy-related gene (Atg) 5 (a ubiquitin-like modifier), or silenced Atg5 were used to assess the effect of RNF168, A20, and Atg5 on DNA damage repair and nuclear factor kappa-B (NF-κB) activation in LN. It was found that podocyte autophagy was over-activated in LN and the abnormal podocyte autophagy led to down-regulation of A20, up-regulation of RNF168, and activation of the NF-κB. RNF168 silencing or A20 restoration inhibited activation of NF-κB pathway and promoted repair of DNA damage, where the level of autophagy was not changed. Activated A20 in podocytes weakened the promoting action of cell autophagy on RNF168. The current results suggest that RNF168 dysfunction may be involved in the pathogenesis of LN via down-regulation of A20 expression. Autophagy and RNF168 may be therapeutic targets for the prevention and treatment of LN.


Assuntos
Nefrite Lúpica , NF-kappa B , Animais , Humanos , Camundongos , Autofagia , Rim , Nefrite Lúpica/metabolismo , NF-kappa B/metabolismo , Ubiquitina/metabolismo , Ubiquitina/farmacologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
7.
Am J Physiol Lung Cell Mol Physiol ; 324(5): L666-L676, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36852930

RESUMO

Respiratory viruses, such as influenza, decrease airway cilia function and expression, which leads to reduced mucociliary clearance and inhibited overall immune defense. Ubiquitination is a posttranslational modification using E3 ligases, which plays a role in the assembly and disassembly of cilia. We examined the role of membrane-associated RING-CH (MARCH) family of E3 ligases during influenza infection and determined that MARCH10, specifically expressed in ciliated epithelial cells, is significantly decreased during influenza infection in mice, human lung epithelial cells, and human lung tissue. Cellular depletion of MARCH10 in differentiated human bronchial epithelial cells (HBECs) using CRISPR/Cas9 showed a decrease in ciliary beat frequency. Furthermore, MARCH10 cellular knockdown in combination with influenza infection selectively decreased immunoreactive levels of the ciliary component, dynein axonemal intermediate chain 1. Cellular overexpression of MARCH10 significantly decreased influenza hemagglutinin protein levels in the differentiated HBECs and knockdown of MARCH10 increased IL-1ß cytokine expression, whereas overexpression had the reciprocal effect. These findings suggest that MARCH10 may have a protective role in airway pulmonary host defense and innate immunity during influenza infection.


Assuntos
Influenza Humana , Orthomyxoviridae , Camundongos , Humanos , Animais , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/farmacologia , Influenza Humana/metabolismo , Ubiquitina/metabolismo , Ubiquitina/farmacologia , Pulmão , Cílios/metabolismo
8.
Brain Behav ; 13(3): e2922, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36793204

RESUMO

AIMS: Opioid addiction is a major public health issue, yet its underlying mechanism is still unknown. The aim of this study was to explore the roles of ubiquitin-proteasome system (UPS) and regulator of G protein signaling 4 (RGS4) in morphine-induced behavioral sensitization, a well-recognized animal model of opioid addiction. METHODS: We explored the characteristics of RGS4 protein expression and polyubiquitination in the development of behavioral sensitization induced by a single morphine exposure in rats, and the effect of a selective proteasome inhibitor, lactacystin (LAC), on behavioral sensitization. RESULTS: Polyubiquitination expression was increased in time-dependent and dose-related fashions during the development of behavioral sensitization, while RGS4 protein expression was not significantly changed during this phase. Stereotaxic administration of LAC into nucleus accumbens (NAc) core inhibited the establishment of behavioral sensitization. CONCLUSION: UPS in NAc core is positively involved in behavioral sensitization induced by a single morphine exposure in rats. Polyubiquitination was observed during the development phase of behavioral sensitization, while RGS4 protein expression was not significantly changed, indicating that other members of RGS family might be substrate proteins in UPS-mediated behavioral sensitization.


Assuntos
Morfina , Transtornos Relacionados ao Uso de Opioides , Animais , Ratos , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/farmacologia , Morfina/farmacologia , Morfina/metabolismo , Núcleo Accumbens/metabolismo , Transtornos Relacionados ao Uso de Opioides/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Complexo de Endopeptidases do Proteassoma/farmacologia , Ubiquitina/metabolismo , Ubiquitina/farmacologia
9.
Am J Physiol Renal Physiol ; 324(3): F315-F328, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36727946

RESUMO

The Na+/K+/2Cl- cotransporter (NKCC2) in the thick ascending limb of the loop of Henle (TAL) mediates NaCl reabsorption. cGMP, the second messenger of nitric oxide and atrial natriuretic peptide, inhibits NKCC2 activity by stimulating NKCC2 ubiquitination and decreasing surface NKCC2 levels. Among the E3 ubiquitin ligase families, the cullin-RING E3 ubiquitin ligase (CRL) family is the largest. Cullins are molecular scaffold proteins that recruit multiple subunits to form the CRL complex. We hypothesized that a CRL complex mediates the cGMP-dependent increase in NKCC2 ubiquitination in TALs. Cullin-1, cullin-2, cullin-3, cullin-4A, and cullin-5 were expressed at the protein level, whereas the other members of the cullin family were expressed at the mRNA level, in rat TALs. CRL complex activity is regulated by neuronal precursor cell-expressed developmentally downregulated protein 8 (Nedd8) to cullins, a process called neddylation. Inhibition of cullin neddylation blunted the cGMP-dependent increase in ubiquitinated NKCC2 while increasing the expression of cullin-1 by threefold, but this effect was not seen with other cullins. CRL complex activity is also regulated by cullin-associated Nedd8-dissociated 1 (CAND1). CAND1 binds to cullins and promotes the exchange of substrate-recognition proteins to target different proteins for ubiquitination. CAND1 inhibition exacerbated the cGMP-dependent increase in NKCC2 ubiquitination and decreased surface NKCC2 expression. Finally, cGMP increased neddylation of cullins. We conclude that the cGMP-dependent increase in NKCC2 ubiquitination is mediated by a CRL complex. To the best of our knowledge, this is the first evidence that a CRL complex mediates NKCC2 ubiquitination in native TALs.NEW & NOTEWORTHY The Na+/K+/2Cl- cotransporter (NKCC2) reabsorbs NaCl by the thick ascending limb. Nitric oxide and atrial natriuretic peptide decrease NaCl reabsorption in thick ascending limbs by increasing the second messenger cGMP. The present findings indicate that cGMP increases NKCC2 ubiquitination via a cullin-RING ligase complex and regulates in part surface NKCC2 levels. Identifying the E3 ubiquitin ligases that regulate NKCC2 expression and activity may provide new targets for the development of specific loop diuretics.


Assuntos
Ubiquitina-Proteína Ligases , Ubiquitina , Animais , Ratos , Fator Natriurético Atrial/metabolismo , Proteínas Culina/metabolismo , GMP Cíclico/metabolismo , Alça do Néfron/metabolismo , Óxido Nítrico/metabolismo , Cloreto de Sódio/metabolismo , Ubiquitina/metabolismo , Ubiquitina/farmacologia , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
10.
Hematol Oncol ; 41(3): 474-486, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36422297

RESUMO

Primary myelofibrosis (PMF) is characterized by immature megakaryocytic hyperplasia, splenomegaly, extramedullary hematopoiesis and bone marrow fibrosis. Our preclinical study had demonstrated that aurora kinase A (AURKA) inhibitor MLN8237 reduced the mutation burden of PMF by inducing differentiation of immature megakaryocytes. However, it only slightly alleviated splenomegaly, reduced tissue fibrosis, and normalized megakaryocytes in PMF patients of the preliminary clinical study. So enhancing therapeutic efficacy of PMF is needed. In this study, we found that AURKA directly interacted with heat shock protein 90 (HSP90) and HSP90 inhibitors promoted the ubiquitin-dependent AURKA degradation. We demonstrated that HSP90 inhibitors 17-allylamino-17-demethoxygeldanamycin (17-AAG) and 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG), normalized peripheral blood counts, improved splenomegaly, attenuated extramedullary hematopoiesis, decreased tissue fibrosis and reduced mutant burden in a MPLW515L mouse model of PMF. Importantly, both 17-AAG and 17-DMAG treatment at effective doses in vivo did not influence on hematopoiesis in healthy mice. Collectively, the study demonstrates that HSP90 inhibitors induce cell differentiation via the ubiquitin-dependent AURKA and also are safe and effective for the treatment of a MPLW515L mouse model of PMF, which may provide a new strategy for PMF therapy. Further, we demonstrate that combined therapy shows superior activity in acute megakaryocytic leukemia mouse model than single therapy.


Assuntos
Antineoplásicos , Mielofibrose Primária , Camundongos , Humanos , Animais , Mielofibrose Primária/tratamento farmacológico , Mielofibrose Primária/genética , Aurora Quinase A , Esplenomegalia/tratamento farmacológico , Ubiquitina/farmacologia , Ubiquitina/uso terapêutico , Diferenciação Celular/genética , Antineoplásicos/uso terapêutico , Fibrose , Proteínas de Choque Térmico/farmacologia , Proteínas de Choque Térmico/uso terapêutico
11.
eNeuro ; 9(2)2022.
Artigo em Inglês | MEDLINE | ID: mdl-35131865

RESUMO

The antidiabetic drug metformin has been shown to reduce pain hypersensitivity in preclinical models of chronic pain and in neuropathic pain in humans. Multiple intracellular pathways have been described as metformin targets. Among them, metformin is an activator of the adenosine 5'-monophosphate protein kinase that can in turn modulate the activity of the E3 ubiquitin ligase NEDD4-2 and thus post-translational expression of voltage-gated sodium channels (NaVs). In this study, we found that the bulk of the effect of metformin on Na1.7 is dependent on NEDD4-2. In HEK cells, the expression of NaV1.7 at the membrane fraction, obtained by a biotinylation approach, is only reduced by metformin when cotransfected with NEDD4-2. Similarly, in voltage-clamp recordings, metformin significantly reduced NaV1.7 current density when cotransfected with NEDD4-2. In mouse dorsal root ganglion (DRG) neurons, without changing the biophysical properties of NaV1.7, metformin significantly decreased NaV1.7 current densities, but not in Nedd4L knock-out mice (SNS-Nedd4L-/-). In addition, metformin induced a significant reduction in NEDD4-2 phosphorylation at the serine-328 residue in DRG neurons, an inhibitory phosphorylation site of NEDD4-2. In current-clamp recordings, metformin reduced the number of action potentials elicited by DRG neurons from Nedd4Lfl/fl , with a partial decrease also present in SNS-Nedd4L-/- mice, suggesting that metformin can also change neuronal excitability in an NEDD4-2-independent manner. We suggest that NEDD4-2 is a critical player for the effect of metformin on the excitability of nociceptive neurons; this action may contribute to the relief of neuropathic pain.


Assuntos
Metformina , Canais de Sódio Disparados por Voltagem , Animais , Gânglios Espinais/metabolismo , Hipoglicemiantes/farmacologia , Metformina/metabolismo , Metformina/farmacologia , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Ubiquitina/metabolismo , Ubiquitina/farmacologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Canais de Sódio Disparados por Voltagem/metabolismo
12.
Drug Chem Toxicol ; 45(4): 1536-1543, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33198515

RESUMO

Acrylamide is a harmful chemical, and its metabolism occurs mainly in the liver. Acrylamide can form adducts on proteins. Protein homeostasis is vital for metabolic and secretory functions of the liver. No study has investigated the effect of acrylamide on the ubiquitin-proteasome system (UPS). Also, the effect of acrylamide on autophagy and its regulation is not fully known. We aimed to investigate the effects of acrylamide on the UPS, autophagy, mammalian target of rapamycin (mTOR), and heat shock protein 70 (HSP70) in HepG2 cells as well as to examine the effects of N-acetylcysteine and curcumin on these parameters in acrylamide-treated cells. HepG2 cells were initially treated with variable concentrations of acrylamide (0.01-0.1-1-10 mM) for 24 hours. Then, HepG2 cells were treated with 5 mM N-acetylcysteine and 6.79 µM curcumin in the presence of 10 mM acrylamide for 24 hours. Cell viability was evaluated by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide assay. Ubiquitinated protein, mTOR, microtubule-associated proteins 1 A/1B light chain 3B-II (LC3B-II), and HSP70 levels were measured by immunoblotting. Acrylamide at 10 mM concentration, without any significant change at lower concentrations, caused an increase in ubiquitinated protein, LC3B-II, and HSP70 levels and a decrease in mTOR phosphorylation. Furthermore, 5 mM N-acetylcysteine caused a decrease in ubiquitinated protein and HSP70 levels; however, 6.79 µM curcumin did not affect 10 mM in acrylamide-treated cells. Our study showed that acrylamide at high concentration inhibits UPS and mTOR, activates autophagy, and increases HSP70 levels in HepG2 cells, and N-acetylcysteine reduces UPS inhibition and HSP70 levels in acrylamide-treated cells.


Assuntos
Curcumina , Acetilcisteína/farmacologia , Acrilamida/toxicidade , Autofagia , Curcumina/farmacologia , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Fígado/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Complexo de Endopeptidases do Proteassoma/farmacologia , Proteólise , Serina-Treonina Quinases TOR/metabolismo , Serina-Treonina Quinases TOR/farmacologia , Ubiquitina/metabolismo , Ubiquitina/farmacologia , Proteínas Ubiquitinadas/metabolismo , Proteínas Ubiquitinadas/farmacologia
13.
J Invertebr Pathol ; 183: 107602, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33964303

RESUMO

In the present study, a ubiquitin (designated as RpUbi) was identified and characterized from clam Ruditapes philippinarum. Phylogenetic analysis strongly suggested that RpUbi was a member of the ubiquitin family. In non-stimulated clams, RpUbi transcripts were constitutively expressed in all examined tissues, especially in the gills and hemocytes. After Vibrio anguillarum challenge, expression of RpUbi mRNA in hemocytes was significantly up-regulated. Recombinant RpUbi (rRpUbi) showed high antibacterial activity against Gram-positive and Gram-negative bacteria. Notably, membrane integrity and electrochemical assay indicated that rRpUbi could invade the inner layer. Moreover, DNA migration could be inhibited by rRpUbi in a concentration-dependent manner. In general, our results suggested that RpUbi played an important role in host defense against invading bacteria, perhaps through a DNA-binding process.


Assuntos
Antibacterianos/farmacologia , Bivalves/química , Ubiquitina/farmacologia , Animais , Antibacterianos/química , Bivalves/microbiologia , Ubiquitina/química
14.
Angew Chem Int Ed Engl ; 60(13): 7018-7023, 2021 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-33326152

RESUMO

Development of modulators targeting specific interactions of ubiquitin-based conjugates with their partners is a formidable task since it requires a suitable screening assay and homogeneous ubiquitin conjugates. We developed a novel high-throughput strategy for screening ligands for Lys48-linked tetraubiquitin chain in a relatively simple, fast, and affordable manner. This approach combined with a state-of-the-art toolbox of chemical protein synthesis and a specially optimized Cys deprotection protocol enabled us to design highly potent, Lys48-linked tetraubiquitin chain selective "next generation" dimeric peptide modulators. The dimeric peptide exhibited cancer cell permeability and induced cell death with higher efficiency compared to its monocyclic analogue. These features make our dimeric peptide a promising candidate for further studies using in vivo models. Our assay can be adopted for other various ubiquitin chains in their free or anchored forms as well as conjugates for Ub-like modifiers.


Assuntos
Desenvolvimento de Medicamentos , Fluorescência , Peptídeos Cíclicos/química , Ubiquitina/química , Morte Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células HeLa , Ensaios de Triagem em Larga Escala , Humanos , Ligantes , Peptídeos Cíclicos/síntese química , Peptídeos Cíclicos/farmacologia , Ubiquitina/farmacologia
15.
Cardiovasc Drugs Ther ; 35(6): 1227-1232, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-32910339

RESUMO

Ischemic heart disease (IHD) accounts for the majority of heart disease-related deaths worldwide. Ubiquitin (UB), found in all eukaryotic cells, is a highly conserved low molecular weight (~8.5 kDa) protein. A well-known intracellular function of UB is to regulate protein turnover via the UB-proteasome system. UB is a normal constituent of plasma, and elevated levels of UB are observed in the serum of patients under a variety of pathological conditions. Recent studies provide evidence for cardioprotective potential of exogenous UB in the remodeling process of the heart in IHD, including effects on cardiac myocyte apoptosis, inflammatory response, and reorganization of the vasculature and extracellular matrix. This review summarizes functions of UB with an emphasis on the role of exogenous UB in myocardial remodeling in IHD.


Assuntos
Cardiotônicos/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Ubiquitina/farmacologia , Apoptose/efeitos dos fármacos , Células Cultivadas , Fibrose/patologia , Mediadores da Inflamação/metabolismo , Receptores CXCR4/metabolismo
16.
Georgian Med News ; (292-293): 87-92, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31560670

RESUMO

Hematological disorders caused by radiation remain the most challenging problem of the last decades. Environmental radiation, as well as medical application of radiation causes serious problems especially from the point of view of blood formation and passage of blood functional cells. Bone marrow emptying followed by the rise of immature cells in the bloodstream is the main pathology that must be eliminated. The importance of the issue is well recognized by all investigators. Opening of agents for regulation of spontaneous regeneration of hematopoietic cell lines is of prime importance in cancer treatment. Ubiquitin is a globular protein of eukaryotic cells. It is involved in regulation of cell cycle. Recently we studied the influence of extracellular ubiquitin on regeneration of leukopoiesis. Interestingly what is its effect on erythropoietic cell lines? Erythrocytes are more resistant to irradiation, than nucleic cells. Their depletion-elevation picks during blood regeneration clearly reveal low sharpness. Nevertheless, depletion of erythropoietic cells if not treated, may cause short- and long-term side effects. We studied the influence of intraperitoneally injected ubiquitin on the process of spontaneous regeneration of erythropoietic cell lines of bone marrow (BM) and peripheral blood (PB) after irradiation in mice. The source of radiation was 137Cs with dose rate 1Gy/min., the duration of exposure 3 min and 5 min. Nonlinear white mice of 22±2 gr. were used for experiment. Animals were divided into five groups (6 animals in each group): the first control group of intact mice; the second test group of mice irradiated by the sublethal dose of 3Gy; the third test group of mice irradiated by 3Gy intraperitoneally injected by ubiquitin by the dose of 100µg/ml at the 72 hour point after irradiation; the fourth test group of mice irradiated by the dose of LD50 5Gy; the fifth test group of mice irradiated by 5Gy intraperitoneally injected by ubiquitin at the 72 hour point after irradiation. PB and BM samples from the control group and the test groups of mice have been taken every 24 hours after irradiation during 7 days. Microscopy and statistical methods have been implicated for calculation of cell count of PB and BM. Analyzing the results we concluded that Ubiquitin prevents depletion-elevation picks of erythrocytes and erythroblasts regardless of the severity of damage caused by different doses of radiation indicating normalization of the regeneration process after irradiation. The study is important for opening new therapeutic agents for normalization of regeneration hematopoiesis after irradiation.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Radioisótopos de Césio , Hematopoese/efeitos dos fármacos , Lesões Experimentais por Radiação , Regeneração/efeitos dos fármacos , Ubiquitina/farmacologia , Animais , Ciclo Celular/efeitos da radiação , Camundongos , Ubiquitina/administração & dosagem
17.
Org Lett ; 21(17): 6790-6794, 2019 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-31398045

RESUMO

Active-site directed probes are powerful tools for studying the ubiquitin conjugation and deconjugation machinery. Branched ubiquitin chains have emerged as important proteasome-targeting signals for aggregation-prone proteins and cell cycle regulators. By implementing a new synthetic strategy for the electrophilic warhead, we herein report on the generation and reactivity of a series of branched triubiquitin active-site directed probes. These new tools can be used to dissect the molecular basis of branched chain assembly and disassembly.


Assuntos
Sondas Moleculares/síntese química , Ubiquitina/síntese química , Domínio Catalítico/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Modelos Moleculares , Sondas Moleculares/química , Sondas Moleculares/farmacologia , Estrutura Molecular , Agregados Proteicos/efeitos dos fármacos , Proteínas/antagonistas & inibidores , Ubiquitina/química , Ubiquitina/farmacologia
18.
mBio ; 10(4)2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31337717

RESUMO

Crimean-Congo hemorrhagic fever virus (CCHFV) infection can result in a severe hemorrhagic syndrome for which there are no antiviral interventions available to date. Certain RNA viruses, such as CCHFV, encode cysteine proteases of the ovarian tumor (OTU) family that antagonize interferon (IFN) production by deconjugating ubiquitin (Ub). The OTU of CCHFV, a negative-strand RNA virus, is dispensable for replication of the viral genome, despite being part of the large viral RNA polymerase. Here, we show that mutations that prevent binding of the OTU to cellular ubiquitin are required for the generation of recombinant CCHFV containing a mutated catalytic cysteine. Similarly, the high-affinity binding of a synthetic ubiquitin variant (UbV-CC4) to CCHFV OTU strongly inhibits viral growth. UbV-CC4 inhibits CCHFV infection even in the absence of intact IFN signaling, suggesting that its antiviral activity is not due to blocking the OTU's immunosuppressive function. Instead, the prolonged occupancy of the OTU with UbV-CC4 directly targets viral replication by interfering with CCHFV RNA synthesis. Together, our data provide mechanistic details supporting the development of antivirals targeting viral OTUs.IMPORTANCE Crimean-Congo hemorrhagic fever virus is an important human pathogen with a wide global distribution for which no therapeutic interventions are available. CCHFV encodes a cysteine protease belonging to the ovarian tumor (OTU) family which is involved in host immune suppression. Here we demonstrate that artificially prolonged binding of the OTU to a substrate inhibits virus infection. This provides novel insights into CCHFV OTU function during the viral replicative cycle and highlights the OTU as a potential antiviral target.


Assuntos
Cisteína Proteases/metabolismo , Enzimas Desubiquitinantes/metabolismo , Vírus da Febre Hemorrágica da Crimeia-Congo/enzimologia , Ubiquitina/farmacologia , Replicação Viral , Animais , Linhagem Celular Tumoral , Cisteína Proteases/genética , Citocinas/genética , Citocinas/metabolismo , Enzimas Desubiquitinantes/genética , Feminino , Vírus da Febre Hemorrágica da Crimeia-Congo/fisiologia , Humanos , Camundongos , Mutação , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Ubiquitinação , Ubiquitinas/genética , Ubiquitinas/metabolismo
19.
Physiol Res ; 68(4): 675-679, 2019 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-31177801

RESUMO

Chemokine (C-X-C motif) receptor 4 (CXCR4) agonists have been shown to protect lung endothelial barrier function in vitro. In vivo effects of CXCR4 modulation on lung endothelial permeability are unknown. Here we tested the effects of the CXCR4 agonist ubiquitin and the antagonist AMD3100 on lung vascular permeability and cytokine concentrations in a rat hemorrhage model. Animals were hemorrhaged (mean arterial blood pressure 30 mmHg for 30 min), treated with vehicle, ubiquitin (0.7 and 3.5 µmol/kg) or AMD3100 (3.5 µmol/kg), and resuscitated with crystalloids. Evans blue extravasation was employed to quantify lung vascular permeability. Ubiquitin dose-dependently reduced Evans blue extravasation into the lung. AMD3100 increased Evans blue extravasation. With AMD3100, TNFalpha levels in lung homogenates were increased; while TNFalpha levels were lower with ubiquitin, these differences did not reach statistical significance. Our findings suggest that CXCR4 regulates lung vascular permeability and further point towards CXCR4 as a drug target to confer lung protection during resuscitation from traumatic-hemorrhagic shock.


Assuntos
Permeabilidade Capilar/fisiologia , Receptores CXCR4/fisiologia , Mucosa Respiratória/metabolismo , Ressuscitação , Choque Hemorrágico/metabolismo , Choque Hemorrágico/terapia , Animais , Benzilaminas , Permeabilidade Capilar/efeitos dos fármacos , Ciclamos , Relação Dose-Resposta a Droga , Compostos Heterocíclicos/farmacologia , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Receptores CXCR4/agonistas , Receptores CXCR4/antagonistas & inibidores , Mucosa Respiratória/efeitos dos fármacos , Ressuscitação/tendências , Ubiquitina/farmacologia
20.
Bioessays ; 41(6): e1900031, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31087674

RESUMO

The confluence of protein engineering techniques and delivery protocols are providing new opportunities in cell biology. In particular, techniques that render the membrane of cells transiently permeable make the introduction of nongenetically encodable macromolecular probes into cells possible. This, in turn, can enable the monitoring of intracellular processes in ways that can be both precise and quantitative, ushering an area that one may envision as cellular biochemistry. Herein, the author reviews pioneering examples of such new cell-based assays, provides evidence that challenges the paradigm that cell penetration is a necessarily damaging and stressful event for cells, and highlights some of the challenges that should be addressed to fully unlock the potential of this nascent field.


Assuntos
Bioensaio/métodos , Membrana Celular/efeitos dos fármacos , Sistemas de Liberação de Medicamentos/métodos , Engenharia de Proteínas/métodos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Técnicas Citológicas/métodos , Meia-Vida , Humanos , Ligação Proteica , Processamento de Proteína Pós-Traducional , Proteólise , Ubiquitina/metabolismo , Ubiquitina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...