Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Immunology ; 170(1): 60-82, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37185810

RESUMEN

Nutritional availability during fasting and refeeding affects the temporal redistribution of lymphoid and myeloid immune cells among the circulating and tissue-resident pools. Conversely, nutritional imbalance and impaired glucose metabolism are associated with chronic inflammation, aberrant immunity and anomalous leukocyte trafficking. Despite being exposed to periodic alterations in blood insulin levels upon fasting and feeding, studies exploring the physiological effects of these hormonal changes on quiescent immune cell function and trafficking are scanty. Here, we report that oral glucose load in mice and healthy men enhances the adherence of circulating peripheral blood mononuclear cells (PBMCs) and lymphocytes to fibronectin. Adherence to fibronectin is also observed upon regular intake of breakfast following overnight fasting in healthy subjects. This glucose load-induced phenomenon is abrogated in streptozotocin-injected mice that lack insulin. Intra-vital microscopy in mice demonstrated that oral glucose feeding enhances the homing of PBMCs to injured blood vessels in vivo. Furthermore, employing flow cytometry, Western blotting and adhesion assays for PBMCs and Jurkat-T cells, we elucidate that insulin enhances fibronectin adherence of quiescent lymphocytes through non-canonical signalling involving insulin-like growth factor-1 receptor (IGF-1R) autophosphorylation, phospholipase C gamma-1 (PLCγ-1) Tyr783 phosphorylation and inside-out activation of ß-integrins respectively. Our findings uncover the physiological relevance of post-prandial insulin spikes in regulating the adherence and trafficking of circulating quiescent T-cells through fibronectin-integrin interaction.

2.
Chemistry ; 29(10): e202202887, 2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36399427

RESUMEN

Artificial channels capable of facilitating the transport of Cl- ions across cell membranes while being nontoxic to the cells are rare. Such synthetic ion channels can mimic the functions of membrane transport proteins and, therefore, have the potential to treat channelopathies by replacing defective ion channels. Here we report isophthalic acid-based structurally simple molecules 1 a and 2 a, which self-assemble to render supramolecular nanochannels that allow selective transport of Cl- ions. As evident from the single-crystal X-ray diffraction analysis, the self-assembly is governed by intermolecular hydrogen bonding and π-π stacking interactions. The MD simulation studies for both 1 a and 2 a confirmed the formation of stable Cl- channel assembly in the lipid membrane and Cl- transport through them. The MQAE assay showed the efficacy of the compounds in delivering Cl- ions into cells, and the MTT assays proved that the compounds are nontoxic to cells even at a concentration of 100 µM.


Asunto(s)
Canales de Cloruro , Ácidos Ftálicos , Canales Iónicos/química , Células Epiteliales
3.
Inorg Chem ; 62(18): 7003-7013, 2023 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-37097171

RESUMEN

Herein, we present an unprecedented formation of a heterodinuclear complex [{(ppy)2IrIII}(µ-phpy){RuII(tpy)}](ClO4)2 {[1](ClO4)2} using terpyridyl/phenylpyridine as ancillary ligands and asymmetric phpy as a bridging ligand. The asymmetric binding mode (N∧N-∩-N∧N∧C-) of the phpy ligand in {[1](ClO4)2} is confirmed by 1H, 13C, 1H-1H correlated spectroscopy (COSY), high-resolution mass spectrum (HRMS), single-crystal X-ray crystallography techniques, and solution conductivity measurements. Theoretical investigation suggests that the highest occupied molecular orbital (HOMO) and the least unoccupied molecular orbital (LUMO) of [1]2+ are located on iridium/ppy and phpy, respectively. The complex displays a broad low energy charge transfer (CT) band within 450-575 nm. The time-dependent density functional theory (TDDFT) analysis suggests this as a mixture of metal-to-ligand charge transfer (MLCT) and ligand-to-ligand charge transfer (LLCT), where both ruthenium, iridium, and ligands are involved. Complex {[1](ClO4)2} exhibits RuIIIrIII/RuIIIIrIII- and RuIIIIrIII/RuIIIIrIV-based oxidative couples at 0.83 and 1.39 V, respectively. The complex shows anticancer activity and selectivity toward human breast cancer cells (IC50; MCF-7: 9.3 ± 1.2 µM, and MDA-MB-231: 8.6 ± 1.2 µM) over normal breast cells (MCF 10A: IC50 ≈ 21 ± 1.3 µM). The Western blot analysis and fluorescence microscopy images suggest that combined apoptosis and autophagy are responsible for cancer cell death.


Asunto(s)
Compuestos Organometálicos , Humanos , Estructura Molecular , Compuestos Organometálicos/química , Ligandos , Iridio/farmacología , Iridio/química , Análisis Espectral
4.
Subcell Biochem ; 100: 427-472, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36301502

RESUMEN

Cancer begins due to uncontrolled cell division. Cancer cells are insensitive to the signals that control normal cell proliferation. This uncontrolled cell division is due to the accumulation of abnormalities in different factors associated with the cell division, including different cyclins, cell cycle checkpoint inhibitors, and cellular signaling. Cellular signaling pathways are aberrantly activated in cancer mainly due to epigenetic regulation and post-translational regulation. In this chapter, the role of epigenetic regulation in aberrant activation of PI3K/AKT, Ras, Wnt, Hedgehog, Notch, JAK/STAT, and mTOR signaling pathways in cancer progression is discussed. The role of epigenetic regulators in controlling the upstream regulatory proteins and downstream effector proteins responsible for abnormal cellular signaling-mediated cancer progression is covered in this chapter. Similarly, the role of signaling pathways in controlling epigenetic gene regulation-mediated cancer progression is also discussed. We have tried to ascertain the current status of potential epigenetic drugs targeting several epigenetic regulators to prevent different cancers.


Asunto(s)
Epigénesis Genética , Neoplasias , Transducción de Señal , Humanos , Neoplasias/metabolismo , Regulación Neoplásica de la Expresión Génica
5.
J Biol Chem ; 296: 100511, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33676897

RESUMEN

F-box proteins ß-TrCP1 and ß-TrCP2 are paralogs present in the human genome. They control several cellular processes including cell cycle and DNA damage signaling. Moreover, it is reported that they facilitate DNA damage-induced accumulation of p53 by directing proteasomal degradation of MDM2, a protein that promotes p53 degradation. However, the individual roles of ß-TrCP1 and ß-TrCP2 in the genotoxic stress-induced activation of cell cycle checkpoints and DNA damage repair remain largely unknown. Here, using biochemical, molecular biology, flow cytometric, and immunofluorescence techniques, we show that ß-TrCP1 and ß-TrCP2 communicate during genotoxic stress. We found that expression levels of ß-TrCP1 are significantly increased while levels of ß-TrCP2 are markedly decreased upon induction of genotoxic stress. Further, our results revealed that DNA damage-induced activation of ATM kinase plays an important role in maintaining the reciprocal expression levels of ß-TrCP1 and ß-TrCP2 via the phosphorylation of ß-TrCP1 at Ser158. Phosphorylated ß-TrCP1 potently promotes the proteasomal degradation of ß-TrCP2 and MDM2, resulting in the activation of p53. Additionally, ß-TrCP1 impedes MDM2 accumulation via abrogation of its lysine 63-linked polyubiquitination by ß-TrCP2. Thus, ß-TrCP1 helps to arrest cells at the G2/M phase of the cell cycle and promotes DNA repair upon DNA damage through attenuation of ß-TrCP2. Collectively, our findings elucidate an intriguing posttranslational regulatory mechanism of these two paralogs under genotoxic stress and revealed ß-TrCP1 as a key player in maintaining the genome integrity through the attenuation of ß-TrCP2 levels in response to genotoxic stress.


Asunto(s)
Puntos de Control del Ciclo Celular , Daño del ADN , Reparación del ADN , Proteolisis , Ubiquitinación , Proteínas con Repetición de beta-Transducina/metabolismo , Supervivencia Celular , Humanos , Fosforilación , Transducción de Señal , Proteínas con Repetición de beta-Transducina/genética
6.
J Biol Chem ; 297(4): 101253, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34587475

RESUMEN

Apoptosis is a programmed cell death that efficiently removes damaged cells to maintain tissue homeostasis. Defect in apoptotic machinery can lead to tumor development, progression, and resistance to chemotherapy. PUMA (p53 upregulated modulator of apoptosis) and BAX (BCL2-associated X protein) are among the most well-known inducers of apoptosis. It has been reported that expression levels of BAX and PUMA are controlled at the posttranslational level by phosphorylation. However, the posttranslational regulation of these proapoptotic proteins remains largely unexplored. In this study, using biochemical, molecular biology, flow cytometric, and immunohistochemistry techniques, we show that PUMA and BAX are the direct target of the F-box protein FBXL20, which restricts their cellular levels. FBXL20 directs the proteasomal degradation of PUMA and BAX in a protein kinase AKT1-dependent manner to promote cancer cell proliferation and tumor growth. Interestingly, inactivation of AKT1 results in activation of another protein kinase GSK3α/ß, which facilitates the proteasomal degradation of FBXL20 by another F-box protein, FBXO31. Thus, a switch between two signaling kinases AKT1 and GSK3α/ß modulates the functional activity of these proapoptotic regulators, thereby determining cell survival or death. RNAi-mediated ablation of FBXL20 results in increased levels of PUMA as well as BAX, which further enhances the sensitivity of cancer cells to chemotherapeutic drugs. We showed that high level expression of FBXL20 in cancer cells reduces therapeutic drug-induced apoptosis and promotes chemoresistance. Overall, this study highlights the importance of targeting FBXL20 in cancers in conjunction with chemotherapy and may represent a promising anticancer strategy to overcome chemoresistance.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Neoplasias de la Mama/metabolismo , Proteínas F-Box/metabolismo , Proteolisis , Proteínas Proto-Oncogénicas/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos/genética , Proteínas F-Box/genética , Femenino , Células HEK293 , Humanos , Células MCF-7 , Proteínas Proto-Oncogénicas/genética , Proteína X Asociada a bcl-2/genética
7.
Int J Cancer ; 150(9): 1512-1524, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-34706096

RESUMEN

FBXO31, a member of F-box protein family, has been shown to play an important role in preventing tumorigenesis by preserving genomic stability during cell proliferation as well as upon genotoxic stress. Inactivation of FBXO31 due to loss of heterozygosity is associated with various cancers, including ovarian cancer, one of the deadliest forms of gynecological cancers. However, the role and regulation of FBXO31 in ovarian cancer remained elusive. Here, using biochemical and molecular biology techniques, we show that c-Myc suppresses the mRNA levels of FBXO31 in ovarian cancer. Chromatin immunoprecipitation experiment showed that c-Myc is recruited to the promoter region of FBXO31 and prevents FBXO31 mRNA synthesis. In contrast, FBXO31 maintains the c-Myc expression at an optimum through proteasome pathway. FBXO31 interacts with and facilitates the polyubiquitination of c-Myc through the SCF complex and thereby inhibits ovarian cancer growth both in vitro and in vivo. Moreover, FBXO31-mediated proteasomal degradation of c-Myc is unique. Unlike other negative regulators, FBXO31 recognizes c-Myc in phosphorylation independent manner to direct its degradation. Further, expression levels analysis revealed that c-Myc and FBXO31 share a converse correlation of expression in ovarian cancer cell lines and patient samples. We observed an increase in the expression levels of c-Myc with a concomitant decrease in the levels of FBXO31 in higher grades of ovarian cancer patient samples. In conclusion, our study demonstrated that oncogene c-Myc impairs the tumor-suppressive functions of FBXO31 to promote ovarian cancer progression, and therefore c-Myc-FBXO31 axis can be explored to develop better cancer therapy.


Asunto(s)
Proteínas F-Box , Neoplasias Ováricas , Proteínas Supresoras de Tumor , Carcinogénesis/genética , Línea Celular Tumoral , Proteínas F-Box/genética , Retroalimentación , Femenino , Humanos , Neoplasias Ováricas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero , Proteínas Supresoras de Tumor/genética
8.
Toxicol Appl Pharmacol ; 441: 115973, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35278439

RESUMEN

Arsenic trioxide (ATO), a potent anti-neoplastic drug, is known to prevent cancer cell growth through induction of autophagic cell death. However, importance of cellular factors in ATO-mediated autophagic cell death is poorly understood. In this study, using biochemical and immunofluorescence techniques, we show that F-box protein FBXO41 plays a critical role in anti-proliferative activity of ATO. Our study reveals the importance of FBXO41 in induction of autophagic death of cancer cells by ATO. Further, we show that the autophagic cell death induced by FBXO41 is distinct and independent of apoptosis and necrosis, showing that FBXO41 may play vital role in inducing autophagic death of apoptosis resistant cancer cells. Overall, our study elucidates the importance of FBXO41 in ATO induced autophagic cell death to prevent cancer progression, which could be explored to develop promising cancer therapeutic strategy.


Asunto(s)
Antineoplásicos , Arsenicales , Muerte Celular Autofágica , Proteínas F-Box , Neoplasias , Antineoplásicos/farmacología , Apoptosis , Trióxido de Arsénico/farmacología , Arsenicales/farmacología , Línea Celular Tumoral , Humanos , Neoplasias/tratamiento farmacológico , Óxidos/farmacología
9.
Cell Biochem Funct ; 39(2): 218-234, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32975319

RESUMEN

Reactive oxygen species (ROS), formed by the partial reduction of oxygen, were for a long time considered to be a byproduct of cellular metabolism. Since, increase in cellular levels of ROS results in oxidative stress leading to damage of nucleic acids, proteins, and lipids resulting in numerous pathological conditions; ROS was considered a bane for aerobic species. Hence, the discovery of NADPH oxidases (NOX), an enzyme family that specifically generates ROS as its prime product came as a surprise to redox biologists. NOX family proteins participate in various cellular functions including cell proliferation and differentiation, regulation of genes and protein expression, apoptosis, and host defence immunological response. Balanced expression and activation of NOX with subsequent production of ROS are critically important to regulate various genes and proteins to maintain homeostasis of the cell. However, dysregulation of NOX activation leading to enhanced ROS levels is associated with various pathophysiologies including diabetes, cardiovascular diseases, neurodegenerative diseases, ageing, atherosclerosis, and cancer. Although our current knowledge on NOX signifies its importance in the normal functioning of various cellular pathways; yet the choice of ROS producing enzymes which can tip the scale from homeostasis toward damage, as mediators of biological functions remain an oddity. Though the role of NOX in maintaining normal cellular functions is now deemed essential, yet its dysregulation leading to catastrophic events cannot be denied. Hence, this review focuses on the involvement of NOX enzymes in various pathological conditions imploring them as possible targets for therapies. SIGNIFICANCE OF THE STUDY: The NOXs are multi-subunit enzymes that generate ROS as a prime product. NOX generated ROS are usually regulated by various molecular factors and play a vital role in different physiological processes. The dysregulation of NOX activity is associated with pathological consequences. Recently, the dynamic proximity of NOX enzymes with different molecular signatures of pathologies has been studied extensively. It is essential to identify the precise role of NOX machinery in its niche during the progression of pathology. Although inhibition of NOX could be a promising approach for therapeutic interventions, it is critical to expand the current understanding of NOX's dynamicity and shed light on their molecular partners and regulators.


Asunto(s)
Enfermedades Cardiovasculares/patología , NADPH Oxidasas/metabolismo , Neoplasias/patología , Acetofenonas/uso terapéutico , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/clasificación , Isoenzimas/metabolismo , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/clasificación , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo
10.
Proc Natl Acad Sci U S A ; 115(5): 998-1003, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29343641

RESUMEN

The F-box protein FBXO31 is a tumor suppressor that is encoded in 16q24.3, for which there is loss of heterozygosity in various solid tumors. FBXO31 serves as the substrate-recognition component of the SKP/Cullin/F-box protein class of E3 ubiquitin ligases and has been shown to direct degradation of pivotal cell-cycle regulatory proteins including cyclin D1 and the p53 antagonist MDM2. FBXO31 levels are normally low but increase substantially following genotoxic stress through a mechanism that remains to be determined. Here we show that the low levels of FBXO31 are maintained through proteasomal degradation by anaphase-promoting complex/cyclosome (APC/C). We find that the APC/C coactivators CDH1 and CDC20 bind to a destruction-box (D-box) motif present in FBXO31 to promote its polyubiquitination and degradation in a cell-cycle-regulated manner, which requires phosphorylation of FBXO31 on serine-33 by the prosurvival kinase AKT. Following genotoxic stress, phosphorylation of FBXO31 on serine-278 by another kinase, the DNA damage kinase ATM, results in disruption of its interaction with CDH1 and CDC20, thereby preventing FBXO31 degradation. Collectively, our results reveal how alterations in FBXO31 phosphorylation, mediated by AKT and ATM, underlie physiological regulation of FBXO31 levels in unstressed and genotoxically stressed cells.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas F-Box/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ciclosoma-Complejo Promotor de la Anafase/antagonistas & inhibidores , Ciclosoma-Complejo Promotor de la Anafase/genética , Antígenos CD , Cadherinas/antagonistas & inhibidores , Cadherinas/genética , Cadherinas/metabolismo , Proteínas Cdc20/antagonistas & inhibidores , Proteínas Cdc20/genética , Proteínas Cdc20/metabolismo , Puntos de Control del Ciclo Celular , Daño del ADN , Proteínas F-Box/química , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Modelos Biológicos , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , ARN Interferente Pequeño/genética , Proteínas Supresoras de Tumor/química , Ubiquitinación
11.
J Biol Chem ; 294(41): 14879-14895, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31413110

RESUMEN

F-box protein 31 (FBXO31) is a reported putative tumor suppressor, and its inactivation due to loss of heterozygosity is associated with cancers of different origins. An emerging body of literature has documented FBXO31's role in preserving genome integrity following DNA damage and in the cell cycle. However, knowledge regarding the role of FBXO31 during normal cell-cycle progression is restricted to its functions during the G2/M phase. Interestingly, FBXO31 levels remain high even during the early G1 phase, a crucial stage for preparing the cells for DNA replication. Therefore, we sought to investigate the functions of FBXO31 during the G1 phase of the cell cycle. Here, using flow cytometric, biochemical, and immunofluorescence techniques, we show that FBXO31 is essential for maintaining optimum expression of the cell-cycle protein cyclin A for efficient cell-cycle progression. Stable FBXO31 knockdown led to atypical accumulation of cyclin A during the G1 phase, driving premature DNA replication and compromised loading of the minichromosome maintenance complex, resulting in replication from fewer origins and DNA double-strand breaks. Because of these inherent defects in replication, FBXO31-knockdown cells were hypersensitive to replication stress-inducing agents and displayed pronounced genomic instability. Upon entering mitosis, the cells defective in DNA replication exhibited a delay in the prometaphase-to-metaphase transition and anaphase defects such as lagging and bridging chromosomes. In conclusion, our findings establish that FBXO31 plays a pivotal role in preserving genomic integrity by maintaining low cyclin A levels during the G1 phase for faithful genome duplication and segregation.


Asunto(s)
Ciclina A/metabolismo , Replicación del ADN/genética , Proteínas F-Box/metabolismo , Genoma Humano/genética , Proteínas Supresoras de Tumor/metabolismo , Ciclo Celular/genética , Cromatina/genética , Proteínas F-Box/genética , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Cinética , Células MCF-7 , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Ubiquitinación/genética
12.
J Pathol ; 248(3): 266-279, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30714168

RESUMEN

Aberrant activation of ß-catenin has been implicated in a variety of human diseases, including cancer. In spite of significant progress, the regulation of active Wnt/ß-catenin-signaling pathways is still poorly understood. In this study, we show that F-box protein 16 (FBXO16) is a putative tumor suppressor. It is a component of the SCF (SKP1-Cullin1-F-box protein) complex, which targets the nuclear ß-catenin protein to facilitate proteasomal degradation through the 26S proteasome. FBXO16 interacts physically with the C-terminal domain of ß-catenin and promotes its lysine 48-linked polyubiquitination. In addition, it inhibits epithelial-to-mesenchymal transition (EMT) by attenuating the level of ß-catenin. Therefore, depletion of FBXO16 leads to increased levels of ß-catenin, which then promotes cell invasion, tumor growth, and EMT of cancer cells. Furthermore, FBXO16 and ß-catenin share an inverse correlation of cellular expression in clinical breast cancer patient samples. In summary, we propose that FBXO16 functions as a putative tumor suppressor by forming an SCFFBXO16 complex that targets nuclear ß-catenin in a unique manner for ubiquitination and subsequent proteasomal degradation to prevent malignancy. This work suggests a novel therapeutic strategy against human cancers related to aberrant ß-catenin activation. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Proteínas F-Box/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , beta Catenina/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Transición Epitelial-Mesenquimal/genética , Genes Supresores de Tumor/fisiología , Humanos , Proteínas Nucleares/metabolismo , Vía de Señalización Wnt/fisiología
13.
J Biol Chem ; 293(42): 16291-16306, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30171069

RESUMEN

The tumor suppressor F-box protein 31 (FBXO31) is indispensable for maintaining genomic stability. Its levels drastically increase following DNA damage, leading to cyclin D1 and MDM2 degradation and G1 and G2/M arrest. Prolonged arrest in these phases leads to cellular senescence. Accordingly, FBXO31 needs to be kept at low basal levels in unstressed conditions for normal cell cycle progression during growth and development. However, the molecular mechanism maintaining these basal FBXO31 levels has remained unclear. Here, we identified the F-box family SCF-E3 ubiquitin ligase FBXO46 (SCFFBXO46) as an important proteasomal regulator of FBXO31 and found that FBXO46 helps maintain basal FBXO31 levels under unstressed conditions and thereby prevents premature senescence. Using molecular docking and mutational studies, we showed that FBXO46 recognizes an RXXR motif located at the FBXO31 C terminus to direct its polyubiquitination and thereby proteasomal degradation. Furthermore, FBXO46 depletion enhanced the basal levels of FBXO31, resulting in senescence induction. In response to genotoxic stress, ATM (ataxia telangiectasia-mutated) Ser/Thr kinase-mediated phosphorylation of FBXO31 at Ser-278 maintained FBXO31 levels. In contrast, activated ATM phosphorylated FBXO46 at Ser-21/Ser-67, leading to its degradation via FBXO31. Thus, ATM-catalyzed phosphorylation after DNA damage governs FBXO31 levels and FBXO46 degradation via a negative feedback loop. Collectively, our findings reveal that FBXO46 is a crucial proteasomal regulator of FBXO31 and thereby prevents senescence in normal growth conditions. They further indicate that FBXO46-mediated regulation of FBXO31 is abrogated following genotoxic stress to promote increased FBXO31 levels for maintenance of genomic stability.


Asunto(s)
Senescencia Celular , Proteínas F-Box/metabolismo , Proteínas Ligasas SKP Cullina F-box/fisiología , Proteínas Supresoras de Tumor/metabolismo , Inestabilidad Genómica , Humanos , Simulación del Acoplamiento Molecular , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Ubiquitinación
14.
J Fluoresc ; 29(6): 1381-1392, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31748964

RESUMEN

In this study, one step hydrothermal synthetic strategy was adopted for preparing carbon dots (C. dots) from jeera (Cumin: Cuminum cyminum), a naturally abundant and cost effective carbon source. The physical, optical and surface functional properties of C. dots were extensively studied by different techniques such as Transmission electron microscopy (TEM), Scanning electron microscopy (SEM), spectrophotometry, fluorescence spectroscopy, Fourier transform infrared spectroscopy (FTIR) and X-ray diffraction (XRD). The obtained C. dots were highly water dispersible and photostable with a quantum yield of 5.33%. The antioxidant property of C. dots was investigated by 2, 2-diphenyl-1-picrylhydrazyl (DPPH) assay. The C. dots were then capped with cystamine using 1-(3-dimethylaminopropyl)-3-ethyl carbodiimide (EDC) and N-Hydroxysuccinimide (NHS) coupling chemistry to design a selective sensing system for chromium (VI) (Cr (VI)). The minimum detection limit of Cr (VI) was found to be 1.57 µM. Biocompatibility and low toxicity of C. dots obtained from jeera made it a potential tool for bioimaging application. The internalisation of C. dots by MCF-7 breast cancer cells and Multi Drug Resistant (MDR) pathogens such as Staphylococcus aureus and Pseudomonas aeruginosa were proved by the bioimaging of respective cells.


Asunto(s)
Antibacterianos/química , Materiales Biocompatibles/química , Cromo/análisis , Contaminantes Químicos del Agua/análisis , Antibacterianos/síntesis química , Antibacterianos/farmacología , Materiales Biocompatibles/síntesis química , Materiales Biocompatibles/farmacología , Carbono/química , Carbono/farmacología , Supervivencia Celular/efectos de los fármacos , Cuminum/química , Cistamina/química , Cistamina/farmacología , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Humanos , Células MCF-7 , Pruebas de Sensibilidad Microbiana , Imagen Óptica , Tamaño de la Partícula , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/crecimiento & desarrollo , Puntos Cuánticos/química , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/crecimiento & desarrollo , Propiedades de Superficie
15.
J Biol Chem ; 292(13): 5349-5363, 2017 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-28184007

RESUMEN

Cyclin F protein, also known as FBXO1, is the largest among all cyclins and oscillates in the cell cycle like other cyclins. Apart from being a G2/M cyclin, cyclin F functions as the substrate-binding subunit of SCFcyclin F E3 ubiquitin ligase. In a gene expression analysis performed to identify novel gene modulations associated with cell cycle dysregulation during HIV-1 infection in CD4+ T cells, we observed down-regulation of the cyclin F gene (CCNF). Later, using gene overexpression and knockdown studies, we identified cyclin F as negatively influencing HIV-1 viral infectivity without any significant impact on virus production. Subsequently, we found that cyclin F negatively regulates the expression of viral protein Vif (viral infectivity factor) at the protein level. We also identified a novel host-pathogen interaction between cyclin F and Vif protein in T cells during HIV-1 infection. Mutational analysis of a cyclin F-specific amino acid motif in the C-terminal region of Vif indicated rescue of the protein from cyclin F-mediated down-regulation. Subsequently, we showed that Vif is a novel substrate of the SCFcyclin F E3 ligase, where cyclin F mediates the ubiquitination and proteasomal degradation of Vif through physical interaction. Finally, we showed that cyclin F augments APOBEC3G expression through degradation of Vif to regulate infectivity of progeny virions. Taken together, our results demonstrate that cyclin F is a novel F-box protein that functions as an intrinsic cellular regulator of HIV-1 Vif and has a negative regulatory effect on the maintenance of viral infectivity by restoring APOBEC3G expression.


Asunto(s)
Ciclinas/fisiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Virión/patogenicidad , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo , Desaminasa APOBEC-3G/metabolismo , Linfocitos T CD4-Positivos , Células Cultivadas , Ciclinas/genética , Ciclinas/metabolismo , Proteínas F-Box , Perfilación de la Expresión Génica , Regulación Viral de la Expresión Génica , Humanos , Procesamiento Proteico-Postraduccional
16.
Proc Natl Acad Sci U S A ; 112(28): 8632-7, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26124108

RESUMEN

The tumor suppressor p53 plays a critical role in maintaining genomic stability. In response to genotoxic stress, p53 levels increase and induce cell-cycle arrest, senescence, or apoptosis, thereby preventing replication of damaged DNA. In unstressed cells, p53 is maintained at a low level. The major negative regulator of p53 is MDM2, an E3 ubiquitin ligase that directly interacts with p53 and promotes its polyubiquitination, leading to the subsequent destruction of p53 by the 26S proteasome. Following DNA damage, MDM2 is degraded rapidly, resulting in increased p53 stability. Because of the important role of MDM2 in modulating p53 function, it is critical to understand how MDM2 levels are regulated. Here we show that the F-box protein FBXO31, a candidate tumor suppressor encoded in 16q24.3 for which there is loss of heterozygosity in various solid tumors, is responsible for promoting MDM2 degradation. Following genotoxic stress, FBXO31 is phosphorylated by the DNA damage serine/threonine kinase ATM, resulting in increased levels of FBXO31. FBXO31 then interacts with and directs the degradation of MDM2, which is dependent on phosphorylation of MDM2 by ATM. FBXO31-mediated loss of MDM2 leads to elevated levels of p53, resulting in growth arrest. In cells depleted of FBXO31, MDM2 is not degraded and p53 levels do not increase following genotoxic stress. Thus, FBXO31 is essential for the classic robust increase in p53 levels following DNA damage.


Asunto(s)
División Celular/fisiología , Proteínas F-Box/fisiología , Mutágenos/toxicidad , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/fisiología , Proteínas Supresoras de Tumor/fisiología , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular , Daño del ADN , Humanos , Fosforilación , Procesamiento Proteico-Postraduccional , Proteolisis
17.
Proc Natl Acad Sci U S A ; 112(26): E3374-83, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26080397

RESUMEN

Pre-mRNA splicing is a complex regulatory nexus modulated by various trans-factors and their posttranslational modifications to create a dynamic transcriptome through alternative splicing. Signal-induced phosphorylation and dephosphorylation of trans-factors are known to regulate alternative splicing. However, the role of other posttranslational modifications, such as deacetylation/acetylation, methylation, and ubiquitination, that could modulate alternative splicing in either a signal-dependent or -independent manner remain enigmatic. Here, we demonstrate that Scaffold/matrix-associated region-binding protein 1 (SMAR1) negatively regulates alternative splicing through histone deacetylase 6 (HDAC6)-mediated deacetylation of RNA-binding protein Sam68 (Src-associated substrate during mitosis of 68 kDa). SMAR1 is enriched in nuclear splicing speckles and associates with the snRNAs that are involved in splice site recognition. ERK-MAPK pathway that regulates alternative splicing facilitates ERK-1/2-mediated phosphorylation of SMAR1 at threonines 345 and 360 and localizes SMAR1 to the cytoplasm, preventing its interaction with Sam68. We showed that endogenously, SMAR1 through HDAC6 maintains Sam68 in a deacetylated state. However, knockdown or ERK-mediated phosphorylation of SMAR1 releases the inhibitory SMAR1-HDAC6-Sam68 complex, facilitating Sam68 acetylation and alternative splicing. Furthermore, loss of heterozygosity at the Chr.16q24.3 locus in breast cancer cells, wherein the human homolog of SMAR1 (BANP) has been mapped, enhances Sam68 acetylation and CD44 variant exon inclusion. In addition, tail-vein injections in mice with human breast cancer MCF-7 cells depleted for SMAR1 showed increased CD44 variant exon inclusion and concomitant metastatic propensity, confirming the functional role of SMAR1 in regulation of alternative splicing. Thus, our results reveal the complex molecular mechanism underlying SMAR1-mediated signal-dependent and -independent regulation of alternative splicing via Sam68 deacetylation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Empalme Alternativo/fisiología , Proteínas de Ciclo Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Histona Desacetilasas/metabolismo , Matriz Nuclear/metabolismo , Proteínas Nucleares/fisiología , Proteínas de Unión al ARN/metabolismo , Acetilación , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Citoplasma/metabolismo , Proteínas de Unión al ADN/genética , Técnicas de Silenciamiento del Gen , Histona Desacetilasa 6 , Humanos , Receptores de Hialuranos/genética , Sistema de Señalización de MAP Quinasas , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transporte de Proteínas
18.
Arch Biochem Biophys ; 612: 78-90, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27789205

RESUMEN

Although the cure of amyloid related neurodegenerative diseases, non-neuropathic amyloidogenic diseases and non-neuropathic systemic amyloidosis are appealing energetic research attempts, beneficial medication is still to be discovered. There is a need to explore intensely stable therapeutic compounds, potent enough to restrict, disrupt or wipe out such toxic aggregates. We had performed a comprehensive biophysical, computational and cell based assay, that shows Nordihydroguaiaretic acid (NA) not only significantly inhibits heat induced hen egg white lysozyme (HEWL) fibrillation but also disaggregates preformed HEWL fibrils and reduces the cytoxicity of amyloid fibrils as well as disaggregated fibrillar species. The inhibitory potency of NA was determined by an IC50 of 26.3 µM. NA was also found to effectively inhibit human lysozyme (HL) fibrillation. NA interferes in the amyloid fibrillogenesis process by interacting hydrophobically with the amino acid residues found in highly prone amyloid fibril forming region of HEWL as explicated by molecular docking results. The results recommend NA as a probable neuroprotective and promising inhibitor for the therapeutic advancement prospective against amyloid related diseases.


Asunto(s)
Amiloidosis/metabolismo , Masoprocol/química , Amiloide/química , Amiloidosis/tratamiento farmacológico , Benzotiazoles , Línea Celular Tumoral , Calor , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Amiloidosis de Cadenas Ligeras de las Inmunoglobulinas , Concentración 50 Inhibidora , Cinética , Luz , Microscopía Fluorescente , Simulación del Acoplamiento Molecular , Muramidasa/química , Nefelometría y Turbidimetría , Agregado de Proteínas , Unión Proteica , Conformación Proteica , Dispersión de Radiación , Espectrometría de Fluorescencia , Tiazoles/química
19.
Biochim Biophys Acta Gene Regul Mech ; 1867(3): 195044, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38763317

RESUMEN

Maintenance of genome integrity is a precise but tedious and complex job for the cell. Several post-translational modifications (PTMs) play vital roles in maintaining the genome integrity. Although ubiquitination is one of the most crucial PTMs, which regulates the localization and stability of the nonhistone proteins in various cellular and developmental processes, ubiquitination of the histones is a pivotal epigenetic event critically regulating chromatin architecture. In addition to genome integrity, importance of ubiquitination of core histones (H2A, H2A, H3, and H4) and linker histone (H1) have been reported in several cellular processes. However, the complex interplay of histone ubiquitination and other PTMs, as well as the intricate chromatin architecture and dynamics, pose a significant challenge to unravel how histone ubiquitination safeguards genome stability. Therefore, further studies are needed to elucidate the interactions between histone ubiquitination and other PTMs, and their role in preserving genome integrity. Here, we review all types of histone ubiquitinations known till date in maintaining genomic integrity during transcription, replication, cell cycle, and DNA damage response processes. In addition, we have also discussed the role of histone ubiquitination in regulating other histone PTMs emphasizing methylation and acetylation as well as their potential implications in chromatin architecture. Further, we have also discussed the involvement of deubiquitination enzymes (DUBs) in controlling histone ubiquitination in modulating cellular processes.

20.
Cell Death Dis ; 14(9): 623, 2023 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-37736741

RESUMEN

Oncogene Moesin plays critical role in initiation, progression, and metastasis of multiple cancers. It exerts oncogenic activity due to its high-level expression as well as posttranslational modification in cancer. However, factors responsible for its high-level expression remain elusive. In this study, we identified positive as well as negative regulators of Moesin. Our study reveals that Moesin is a cellular target of F-box protein FBXW2. We showed that FBXW2 suppresses breast cancer progression through directing proteasomal degradation of Moesin. In contrast, AKT kinase plays an important role in oncogenic function of Moesin by protecting it from FBXW2-mediated proteasomal degradation. Mechanistically, AKT phosphorylates Moesin at Thr-558 and thereby prevents its degradation by FBXW2 via weakening the association between FBXW2 and Moesin. Further, accumulated Moesin prevents FBXW2-mediated degradation of oncogene SKP2, showing that Moesin functions as an upstream regulator of oncogene SKP2. In turn, SKP2 stabilizes Moesin by directing its non-degradable form of polyubiquitination and therefore AKT-Moesin-SKP2 oncogenic axis plays crucial role in breast cancer progression. Collectively, our study reveals that FBXW2 functions as a tumor suppressor in breast cancer by restricting AKT-Moesin-SKP2 axis. Thus, AKT-Moesin-SKP2 axis may be explored for the development of therapeutics for cancer treatment.


Asunto(s)
Neoplasias de la Mama , Proteínas F-Box , Proteínas Proto-Oncogénicas c-akt , Humanos , Transformación Celular Neoplásica , Proteínas F-Box/genética , Proteínas de Microfilamentos , Oncogenes , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA