Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Eur J Haematol ; 113(3): 351-356, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38804098

RESUMO

BACKGROUND: Precursor plasma cell disorders such as monoclonal gammopathy of undetermined significance (MGUS) always precede the development of active malignancies such as multiple myeloma (MM). There is a need for novel biomarkers to identify those patients with such precursor plasma cell disorders who rapidly progress to MM. Plasma-derived extracellular vesicles (EVs) may serve as a reservoir of potential biomarkers that can shed light on the pathogenesis and disease biology of MM. METHODS: This study isolated small EVs (SEVs) and large EVs (LEVs) from the platelet-poor peripheral blood plasma of MGUS (n = 9) and MM (n = 12) patients using the size exclusion chromatography-based method and evaluated their proteome using a label-free proteomics workflow. RESULTS: In total, 2055 proteins were identified in SEVs, while 2794 proteins were identified in LEVs. The transferrin receptor (or CD71) protein was upregulated in both populations of EVs derived from MM patients compared to MGUS patients and was of prognostic significance. Similarly, three isoforms of serum amyloid A (SAA) protein, SAA1, SAA2, and SAA4, were also highly upregulated in SEVs within MM patients relative to MGUS patients. Finally, CD40 expression was also higher in the LEVs derived from MM patients than in MGUS patients. CONCLUSIONS: This study demonstrates the feasibility of successfully isolating both SEVs and LEVs from the peripheral blood of patients with plasma cell disorders and quantifying protein biomarkers within these EVs that could be of prognostic and diagnostic interest.


Assuntos
Vesículas Extracelulares , Gamopatia Monoclonal de Significância Indeterminada , Mieloma Múltiplo , Proteoma , Proteômica , Humanos , Vesículas Extracelulares/metabolismo , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/sangue , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Proteômica/métodos , Masculino , Gamopatia Monoclonal de Significância Indeterminada/diagnóstico , Gamopatia Monoclonal de Significância Indeterminada/sangue , Gamopatia Monoclonal de Significância Indeterminada/metabolismo , Gamopatia Monoclonal de Significância Indeterminada/patologia , Feminino , Pessoa de Meia-Idade , Idoso , Biomarcadores Tumorais/sangue , Lesões Pré-Cancerosas/diagnóstico , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/sangue , Lesões Pré-Cancerosas/patologia , Biomarcadores , Prognóstico
2.
Cancer Lett ; 565: 216209, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37169162

RESUMO

The development of androgen receptor signaling inhibitor (ARSI) drug resistance in prostate cancer (PC) remains therapeutically challenging. Our group has described the role of sex determining region Y-box 2 (SOX2) overexpression in ARSI-resistant PC. Continuing this work, we report that NR3C1, the gene encoding glucocorticoid receptor (GR), is a novel SOX2 target in PC, positively regulating its expression. Similar to ARSI treatment, SOX2-positive PC cells are insensitive to GR signaling inhibition using a GR modulating therapy. To understand SOX2-mediated nuclear hormone receptor signaling inhibitor (NHRSI) insensitivity, we performed RNA-seq in SOX2-positive and -negative PC cells following NHRSI treatment. RNA-seq prioritized differentially regulated genes mediating the cell cycle, including G2 checkpoint WEE1 Kinase (WEE1) and cyclin-dependent kinase 1 (CDK1). Additionally, WEE1 and CDK1 were differentially expressed in PC patient tumors dichotomized by high vs low SOX2 gene expression. Importantly, pharmacological targeting of WEE1 (WEE1i) in combination with an ARSI or GR modulator re-sensitizes SOX2-positive PC cells to nuclear hormone receptor signaling inhibition in vitro, and WEE1i combined with ARSI significantly slowed tumor growth in vivo. Collectively, our data suggest SOX2 predicts NHRSI resistance, and simultaneously indicates the addition of WEE1i to improve therapeutic efficacy of NHRSIs in SOX2-positive PC.


Assuntos
Antineoplásicos , Neoplasias de Próstata Resistentes à Castração , Masculino , Humanos , Proteína Quinase CDC2/genética , Proteína Quinase CDC2/metabolismo , Transdução de Sinais , Antineoplásicos/farmacologia , Proteínas de Ciclo Celular/metabolismo , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/genética , Antagonistas de Receptores de Andrógenos/farmacologia , Receptores Citoplasmáticos e Nucleares , Linhagem Celular Tumoral , Proteínas Tirosina Quinases/metabolismo , Fatores de Transcrição SOXB1/genética
3.
Cell Death Dis ; 12(11): 1061, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34750354

RESUMO

Aberrant microRNA (miR) expression plays an important role in pathogenesis of different types of cancers, including B-cell lymphoid malignancies and in the development of chemo-sensitivity or -resistance in chronic lymphocytic leukemia (CLL) as well as diffuse large B-cell lymphoma (DLBCL). Ibrutinib is a first-in class, oral, covalent Bruton's tyrosine kinase (BTK) inhibitor (BTKi) that has shown impressive clinical activity, yet many ibrutinib-treated patients relapse or develop resistance over time. We have reported that acquired resistance to ibrutinib is associated with downregulation of tumor suppressor protein PTEN and activation of the PI3K/AKT pathway. Yet how PTEN mediates chemoresistance in B-cell malignancies is not clear. We now show that the BTKi ibrutinib and a second-generation compound, acalabrutinib downregulate miRNAs located in the 14q32 miRNA cluster region, including miR-494, miR-495, and miR-543. BTKi-resistant CLL and DLBCL cells had striking overexpression of miR-494, miR-495, miR-543, and reduced PTEN expression, indicating further regulation of the PI3K/AKT/mTOR pathway in acquired BTKi resistance. Additionally, unlike ibrutinib-sensitive CLL patient samples, those with resistance to ibrutinib treatment, demonstrated upregulation of 14q32 cluster miRNAs, including miR-494, miR-495, and miR-543 and decreased pten mRNA expression. Luciferase reporter gene assay showed that miR-494 directly targeted and suppressed PTEN expression by recognizing two conserved binding sites in the PTEN 3'-UTR, and subsequently activated AKTSer473. Importantly, overexpression of a miR-494 mimic abrogated both PTEN mRNA and protein levels, further indicating regulation of apoptosis by PTEN/AKT/mTOR. Conversely, overexpression of a miR-494 inhibitor in BTKi-resistant cells restored PTEN mRNA and protein levels, thereby sensitizing cells to BTKi-induced apoptosis. Inhibition of miR-494 and miR-495 sensitized cells by cooperative targeting of pten, with additional miRNAs in the 14q32 cluster that target pten able to contribute to its regulation. Therefore, targeting 14q32 cluster miRNAs may have therapeutic value in acquired BTK-resistant patients via regulation of the PTEN/AKT/mTOR signaling axis.


Assuntos
Tirosina Quinase da Agamaglobulinemia/antagonistas & inibidores , Leucemia Linfocítica Crônica de Células B/genética , MicroRNAs/genética , PTEN Fosfo-Hidrolase/metabolismo , Humanos , Leucemia Linfocítica Crônica de Células B/patologia , Transdução de Sinais , Transfecção
4.
Clin Ophthalmol ; 15: 617-622, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33628007

RESUMO

AIM: To assess the demography, magnitude, and type of corneal astigmatism in patients undergoing cataract surgery in North India. METHODS: It is a clinic-based, cross-sectional, observational study. Keratometric values and demographic data were collected for eligible patients who had undergone phacoemulsification at a tertiary eye care center between January 2010 and December 2017, using a non contact, optical low coherence reflectometry (OLCR). RESULTS: A total of 3597 eyes were recruited for the study. There were 1810 (50.3%) females and 1787 (49.7%) males. The mean age was 59.121±15.19 (range 5-100 years). A total of 3559 eyes were qualified for astigmatism analysis. The mean corneal astigmatism among all patients was 1.17±1.15 D (range 0-12.5 D). There was no astigmatism in 99 eyes (2.78%), with-the-rule (WTR) in 1062 eyes (29.83%), against-the-rule (ATR) in 1843 eyes (51.72%) and oblique astigmatism (OA) in 555 eyes (15.59%). The tendency of a gradual change from with the rule (WTR) to against the rule (ATR) astigmatism was noted as the age advanced. CONCLUSION: In the present study around 56.69% of eyes had corneal astigmatism of <1.0 D that can be managed by simple cost-effective keratorefractive procedures especially in developing countries. However, our 40.49% patients had >1.0 D of corneal astigmatism, which may benefit by toric intraocular lenses.

5.
Cell Death Dis ; 11(11): 941, 2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-33139702

RESUMO

Defects in apoptosis can promote tumorigenesis and impair responses of malignant B cells to chemotherapeutics. Members of the B-cell leukemia/lymphoma-2 (BCL-2) family of proteins are key regulators of the intrinsic, mitochondrial apoptotic pathway. Overexpression of antiapoptotic BCL-2 family proteins is associated with treatment resistance and poor prognosis. Thus, inhibition of BCL-2 family proteins is a rational therapeutic option for malignancies that are dependent on antiapoptotic BCL-2 family proteins. Venetoclax (ABT-199, GDC-0199) is a highly selective BCL-2 inhibitor that represents the first approved agent of this class and is currently widely used in the treatment of chronic lymphocytic leukemia (CLL) as well as acute myeloid leukemia (AML). Despite impressive clinical activity, venetoclax monotherapy for a prolonged duration can lead to drug resistance or loss of dependence on the targeted protein. In this review, we provide an overview of the mechanism of action of BCL-2 inhibition and the role of this approach in the current treatment paradigm of B-cell malignancies. We summarize the drivers of de novo and acquired resistance to venetoclax that are closely associated with complex clonal shifts, interplay of expression and interactions of BCL-2 family members, transcriptional regulators, and metabolic modulators. We also examine how tumors initially resistant to venetoclax become responsive to it following prior therapies. Here, we summarize preclinical data providing a rationale for efficacious combination strategies of venetoclax to overcome therapeutic resistance by a targeted approach directed against alternative antiapoptotic BCL-2 family proteins (MCL-1, BCL-xL), compensatory prosurvival pathways, epigenetic modifiers, and dysregulated cellular metabolism/energetics for durable clinical remissions.


Assuntos
Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Leucemia de Células B/tratamento farmacológico , Linfoma de Células B/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Animais , Humanos , Leucemia de Células B/metabolismo , Leucemia de Células B/patologia , Linfoma de Células B/metabolismo , Linfoma de Células B/patologia , Terapia de Alvo Molecular
6.
Arterioscler Thromb Vasc Biol ; 40(10): 2440-2453, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32787518

RESUMO

OBJECTIVE: Calcium channel blockers, such as dihydropyridines, are commonly used to inhibit enhanced activity of vascular CaV1.2 channels in hypertension. However, patients who are insensitive to such treatments develop calcium channel blocker-resistant hypertension. The function of CaV1.2 channel is diversified by alternative splicing, and the splicing factor PTBP (polypyrimidine tract-binding protein) 1 influences the utilization of mutually exclusive exon 8/8a of the CaV1.2 channel during neuronal development. Nevertheless, whether and how PTBP1 makes a role in the calcium channel blocker sensitivity of vascular CaV1.2 channels, and calcium channel blocker-induced vasodilation remains unknown. Approach and Results: We detected high expression of PTBP1 and, inversely, low expression of exon 8a in CaV1.2 channels (CaV1.2E8a) in rat arteries. In contrast, the opposite expression patterns were observed in brain and heart tissues. In comparison to normotensive rats, the expressions of PTBP1 and CaV1.2E8a channels were dysregulated in mesenteric arteries of hypertensive rats. Notably, PTBP1 expression was significantly downregulated, and CaV1.2E8a channels were aberrantly increased in dihydropyridine-resistant arteries compared with dihydropyridine-sensitive arteries of rats and human. In rat vascular smooth muscle cells, PTBP1 knockdown resulted in shifting of CaV1.2 exon 8 to 8a. Using patch-clamp recordings, we demonstrated a concomitant reduction of sensitivity of CaV1.2 channels to nifedipine, due to the higher expression of CaV1.2E8a isoform. In vascular myography experiments, small interfering RNA-mediated knockdown of PTBP1 attenuated nifedipine-induced vasodilation of rat mesenteric arteries. CONCLUSIONS: PTBP1 finely modulates the sensitivities of CaV1.2 channels to dihydropyridine by shifting the utilization of exon 8/8a and resulting in changes of responses in dihydropyridine-induced vasodilation.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Resistência a Medicamentos , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Hipertensão/tratamento farmacológico , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Nifedipino/farmacologia , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Processamento Alternativo , Animais , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Éxons , Células HEK293 , Ribonucleoproteínas Nucleares Heterogêneas/genética , Humanos , Hipertensão/genética , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Masculino , Potenciais da Membrana , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/metabolismo , Artérias Mesentéricas/fisiopatologia , Camundongos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Ratos Sprague-Dawley , Transdução de Sinais
7.
Nucleic Acids Res ; 48(2): 736-747, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31740976

RESUMO

Ionizing radiation (IR)-induced DNA double-strand breaks (DSBs) are predominantly repaired by non-homologous end joining (NHEJ). IR-induced DNA damage activates autophagy, an intracellular degradation process that delivers cytoplasmic components to the lysosome. We identified the deubiquitinase USP14 as a novel autophagy substrate and a regulator of IR-induced DNA damage response (DDR) signaling. Inhibition of autophagy increased levels and DSB recruitment of USP14. USP14 antagonized RNF168-dependent ubiquitin signaling and downstream 53BP1 chromatin recruitment. Here we show that autophagy-deficient cells are defective in NHEJ, as indicated by decreased IR-induced foci (IRIF) formation by pS2056-, pT2609-DNA-PKcs, pS1778-53BP1, RIF1 and a reporter assay activation. Moreover, chromatin recruitment of key NHEJ proteins, including, Ku70, Ku80, DNA-PKcs and XLF was diminished in autophagy-deficient cells. USP14 inhibition rescued the activity of NHEJ-DDR proteins in autophagy-deficient cells. Mass spectrometric analysis identified USP14 interaction with core NHEJ proteins, including Ku70, which was validated by co-immunoprecipitation. An in vitro assay revealed that USP14 targeted Ku70 for deubiquitination. AKT, which mediates Ser432-USP14 phosphorylation, was required for IRIF formation by USP14. Similar to USP14 block, AKT inhibition rescued the activity of NHEJ-DDR proteins in autophagy- and PTEN-deficient cells. These findings reveal a novel negative PTEN/Akt-dependent regulation of NHEJ by USP14.


Assuntos
Reparo do DNA por Junção de Extremidades/efeitos da radiação , PTEN Fosfo-Hidrolase/genética , Proteínas Proto-Oncogênicas c-akt/genética , Ubiquitina Tiolesterase/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Autofagia/efeitos da radiação , Cromatina/genética , Cromatina/efeitos da radiação , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Dano ao DNA/efeitos da radiação , Reparo do DNA/efeitos da radiação , Células HEK293 , Humanos , Autoantígeno Ku/genética , PTEN Fosfo-Hidrolase/deficiência , Radiação Ionizante , Transdução de Sinais/genética , Transdução de Sinais/efeitos da radiação , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética
8.
Cell Death Dis ; 10(12): 924, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31801949

RESUMO

Chronic activation of the Bruton's tyrosine kinase (BTK)-mediated B-cell receptor (BCR) signaling is a hallmark of many B-cell lymphoid malignancies, including chronic lymphocytic leukemia (CLL) and diffuse large B-cell lymphoma (DLBCL). Ibrutinib, an FDA approved, orally administered BTK inhibitor, has demonstrated high response rates, however, complete responses are infrequent and acquired resistance to BTK inhibition can emerge. In this study, we generated ibrutinib-resistant (IB-R) cell lines by chronic exposure of CLL and activated B-cell (ABC)-DLBCL cells to ibrutinib in order to investigate the mechanism of acquired resistance to ibrutinib. IB-R cell lines demonstrated downregulation of FOXO3a and PTEN levels and activation of AKT, with their levels being low in the nuclei of resistant cells in comparison to the sensitive counterparts. Inhibition of PI3K and AKT using idelalisib and MK2206, respectively increased ibrutinib-induced apoptosis in IB-R cells by downregulation of pAKT473 and restoring FOXO3a levels, demonstrating the importance of these cell survival factors for ibrutinib-resistance. Notably, the exportin 1 inhibitor, selinexor synergized with ibrutinib in IB-R cells and restored nuclear abundance of FOXO3a and PTEN, suggesting that nuclear accumulation of FOXO3a and PTEN facilitates increase in ibrutinib-induced apoptosis in IB-R cells. These data demonstrate that reactivation of FOXO3a nuclear function enhances the efficacy of ibrutinib and overcomes acquired resistance to ibrutinib. Together, these findings reveal a novel mechanism that confers ibrutinib resistance via aberrant nuclear/cytoplasmic subcellular localization of FOXO3a and could be exploited by rational therapeutic combination regimens for effectively treating lymphoid malignancies.


Assuntos
Tirosina Quinase da Agamaglobulinemia/genética , Proteína Forkhead Box O3/genética , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Linfoma Difuso de Grandes Células B/tratamento farmacológico , PTEN Fosfo-Hidrolase/genética , Tirosina Quinase da Agamaglobulinemia/antagonistas & inibidores , Idoso , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/patologia , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/patologia , Masculino , Pessoa de Meia-Idade , Fosfatidilinositol 3-Quinases/genética , Inibidores de Proteínas Quinases/administração & dosagem , Proteínas Proto-Oncogênicas c-akt/genética , Purinas/farmacologia , Quinazolinonas/farmacologia , Transdução de Sinais/efeitos dos fármacos
9.
Biochim Biophys Acta Mol Cell Res ; 1864(10): 1545-1553, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28578910

RESUMO

Granulocyte colony-stimulating factor receptor (G-CSFR) plays a crucial role in regulating myeloid cell survival, proliferation, and neutrophilic granulocyte precursor cells maturation. Previously, we demonstrated that Fbw7α negatively regulates G-CSFR and its downstream signaling through ubiquitin-proteasome mediated degradation. However, whether additional ubiquitin ligases for G-CSFR exist is not known. Identifying multiple E3 ubiquitin ligases for G-CSFR shall improve our understanding of activation and subsequent attenuation of G-CSFR signaling required for differentiation and proliferation. Here, for the first time we demonstrate that E6 associated protein (E6AP), an E3 ubiquitin ligase physically associates with G-CSFR and targets it for ubiquitin-mediated proteasome degradation and thereby attenuates its functions. We further show that E6AP promoted G-CSFR degradation leads to reduced phosphorylation of signal transducer and activator of transcription 3 (STAT3) which is required for G-CSF dependent granulocytic differentiation. More importantly, our finding shows that E6AP also targets mutant form of G-SCFR (G-CSFR-T718), frequently observed in severe congenital neutropenia (SCN) patients that very often culminate to AML, however, at a quite slower rate than wild type G-CSFR. In addition, our data showed that knockdown of E6AP restores G-CSFR and its signaling thereby promoting granulocytic differentiation. Collectively, our data demonstrates that E6AP facilitates ubiquitination and subsequent degradation of G-CSFR leading to attenuation of its downstream signaling and inhibition of granulocytic differentiation.


Assuntos
Proteína 7 com Repetições F-Box-WD/genética , Receptores de Fator Estimulador de Colônias de Granulócitos/genética , Ubiquitina-Proteína Ligases/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Proteína 7 com Repetições F-Box-WD/metabolismo , Técnicas de Silenciamento de Genes , Granulócitos/metabolismo , Granulócitos/patologia , Humanos , Células Mieloides/metabolismo , Células Mieloides/patologia , Complexo de Endopeptidases do Proteassoma/genética , Proteólise , Receptores de Fator Estimulador de Colônias de Granulócitos/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/metabolismo
10.
Mol Cancer Res ; 14(11): 1097-1109, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27470268

RESUMO

Drosophila caudal-related homeobox transcription factor 2 (CDX2) drives differentiation of the intestinal epithelium. Loss of CDX2 expression has been reported in several colorectal cancers and cancer cell lines with a potential inverse correlation between CDX2 levels and tumor stage. Ubiquitination of CDX2 leading to its downregulation has been implicated in several studies; however, the E3 ubiquitin ligases involved in CDX2 ubiquitination have largely remained unknown. Here, it is mechanistically determined that the E3 ubiquitin ligase Fbw7 promotes CDX2 ubiquitination and degradation through two phosphodegron motifs present within CDX2 in a GSK3ß-dependent manner leading to its reduced expression and function in colon cancer cells. Fbw7, through its WD domain, interacted with CDX2 both in a heterologous HEK293T cell system and in colon cancer cells. GSK3ß was also present in the same complex as determined by coimmunoprecipitation. Furthermore, overexpression of both Fbw7 or GSK3ß down regulated endogenous CDX2 expression and function; however, both failed to inhibit endogenous CDX2 when either of them were depleted in colon cancer cells. Fbw7-mediated inhibition of CDX2 expression also led to reduced CDX2 transactivation and growth arrest of colon cancer cells. Both GSK3ß and Fbw7 degraded mutant-CDX2 having either of the Cdc4-phosphodegron (CPD) motifs disrupted (CDX2-S60A or CDX-S281A), but were unable to degrade mutant-CDX2 having both CPDs disrupted (CDX2-S60,64,281A). IMPLICATIONS: Taken together, these findings demonstrate that Fbw7 negatively regulates CDX2 expression in a GSK3ß-dependent manner through two CPDs present in CDX2. Mol Cancer Res; 14(11); 1097-109. ©2016 AACR.


Assuntos
Fator de Transcrição CDX2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Neoplasias do Colo/metabolismo , Proteínas F-Box/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Motivos de Aminoácidos , Fator de Transcrição CDX2/química , Células CACO-2 , Proteínas de Ciclo Celular/química , Linhagem Celular Tumoral , Proteínas F-Box/química , Proteína 7 com Repetições F-Box-WD , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Fosforilação , Domínios Proteicos , Transdução de Sinais , Ubiquitina-Proteína Ligases/química , Ubiquitinação
12.
Biochim Biophys Acta ; 1863(4): 510-9, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26778333

RESUMO

Osteogenic transcription factor Runx2 is essential for osteoblast differentiation. The activity of Runx2 is tightly regulated at transcriptional as well as post-translational level. However, regulation of Runx2 stability by ubiquitin mediated proteasomal degradation by E3 ubiquitin ligases is little-known. Here, for the first time we demonstrate that Skp2, an SCF family E3 ubiquitin ligase negatively targets Runx2 by promoting its polyubiquitination and proteasome dependent degradation. Co-immunoprecipitation studies revealed that Skp2 physically interacts with Runx2 both in a heterologous as well as physiologically relevant system. Functional consequences of Runx2-Skp2 physical interaction were then assessed by promoter reporter assay. We show that Skp2-mediated downregulation of Runx2 led to reduced Runx2 transactivation and osteoblast differentiation. On the contrary, inhibition of Skp2 restored Runx2 levels and promoted osteoblast differentiation. We further show that Skp2 and Runx2 proteins are co-expressed and show inverse relation in vivo such as in lactating, ovariectomized and estrogen-treated ovariectomized animals. Together, these data demonstrate that Skp2 targets Runx2 for ubiquitin mediated degradation and hence negatively regulate osteogenesis. Therefore, the present study provides a plausible therapeutic target for osteoporosis or cleidocranial dysplasia caused by the heterozygous mutation of Runx2 gene.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Osteogênese/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Proteínas Quinases Associadas a Fase S/fisiologia , Animais , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Osteogênese/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Proteínas Quinases Associadas a Fase S/antagonistas & inibidores , Proteínas Quinases Associadas a Fase S/genética , Ubiquitina/metabolismo
13.
Oncotarget ; 7(7): 7640-56, 2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26506232

RESUMO

Perturbed stability of regulatory proteins is a major cause of transformations leading to cancer, including several leukemia subtypes. Here, for the first time we demonstrate that E6-associated protein (E6AP), an E3 ubiquitin ligase negatively targets MAX binding protein MNT for ubiquitin-mediated proteasome degradation and impedes ATRA mediated myeloid cell differentiation. MNT is a member of the Myc/Max/Mad network of transcription factor that regulates cell proliferation, differentiation, cellular transformation and tumorigenesis. Wild-type E6AP promoted proteasome dependent degradation of MNT, while catalytically inactive E6AP having cysteine replaced with alanine at amino-acid 843 position (E6APC843A) rather stabilized it. Further, these proteins physically associated with each other both in non-myeloid (HEK293T) and myeloid cells. MNT overexpression induced G0-G1 growth arrest and promoted myeloid differentiation while its knockdown mitigated even ATRA induced differentiation suggesting MNT to be crucial for myeloid differentiation. We further showed that ATRA inhibited E6AP and stabilized MNT expression by protecting it from E6AP mediated ubiquitin-proteasome degradation. Notably, E6AP knockdown in HL60 cells restored MNT expression and promoted myeloid differentiation. Taken together, our data demonstrated that E6AP negatively regulates granulocytic differentiation by targeting MNT for degradation which is required for growth arrest and subsequent myeloid differentiation by various differentiation inducing agents.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Biomarcadores/metabolismo , Diferenciação Celular , Leucemia Promielocítica Aguda/metabolismo , Proteômica/métodos , Proteínas Repressoras/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Western Blotting , Células Cultivadas , Citometria de Fluxo , Imunofluorescência , Células HEK293 , Humanos , Imunoprecipitação , Leucemia Promielocítica Aguda/patologia , Microscopia de Fluorescência , Ligação Proteica , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
14.
J Biol Chem ; 290(52): 30975-87, 2015 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-26542806

RESUMO

Runx2, a master regulator of osteoblast differentiation, is tightly regulated at both transcriptional and post-translational levels. Post-translational modifications such as phosphorylation and ubiquitination have differential effects on Runx2 functions. Here, we show that the reduced expression and functions of Runx2 upon its phosphorylation by GSK3ß are mediated by its ubiquitin-mediated degradation through E3 ubiquitin ligase Fbw7α. Fbw7α through its WD domain interacts with Runx2 both in a heterologous (HEK293T cells) system as well as in osteoblasts. GSK3ß was also present in the same complex as determined by co-immunoprecipitation. Furthermore, overexpression of either Fbw7α or GSK3ß was sufficient to down-regulate endogenous Runx2 expression and function; however, both failed to inhibit endogenous Runx2 when either of them was depleted in osteoblasts. Fbw7α-mediated inhibition of Runx2 expression also led to reduced Runx2 transactivation and osteoblast differentiation. In contrast, inhibition of Fbw7α restored Runx2 levels and promoted osteoblast differentiation. We also observed reciprocal expression levels of Runx2 and Fbw7α in models of bone loss such as lactating (physiological bone loss condition) and ovariectomized (induction of surgical menopause) animals that show reduced Runx2 and enhanced Fbw7α, whereas this was reversed in the estrogen-treated ovariectomized animals. In addition, methylprednisolone (a synthetic glucocorticoid) treatment to neonatal rats showed a temporal decrease in Runx2 with a reciprocal increase in Fbw7 in their calvarium. Taken together, these data demonstrate that Fbw7α negatively regulates osteogenesis by targeting Runx2 for ubiquitin-mediated degradation in a GSK3ß-dependent manner and thus provides a plausible explanation for GSK3ß-mediated bone loss as described before.


Assuntos
Diferenciação Celular , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Proteínas F-Box/metabolismo , Osteoblastos/metabolismo , Proteólise , Ubiquitina-Proteína Ligases/metabolismo , Animais , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Proteínas F-Box/genética , Proteína 7 com Repetições F-Box-WD , Feminino , Quinase 3 da Glicogênio Sintase/biossíntese , Glicogênio Sintase Quinase 3 beta , Células HEK293 , Humanos , Camundongos , Osteogênese/genética , Ratos , Ratos Sprague-Dawley , Ativação Transcricional , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética
15.
Malays J Pathol ; 36(2): 115-24, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25194534

RESUMO

There are 106 bones in hands and feet but their lesions are not commonly reported. This was a retrospective study of all osteolytic lesions involving bones of the hands or feet presenting to the only tertiary referral centre of the north Indian state of Uttarakhand during the 7-year period from January 2006 to December 2012. A compilation of the various demographic, clinical, radiological and histopathological findings was made. Of the 52 lesions encountered in the 7-year record, 75% were asymptomatic. 20 (38.4%) were benign tumours, 20 (38.4%) tumour-like lesions, 9 (17.3%) inflammatory and post traumatic lesions and only 3 (5.7%) were malignant lesions. Giant cell tumour was the most common benign tumour, aneurysmal bone cyst the most common tumour-like lesion and non-specific osteomyelitis was the most common inflammatory and post-traumatic pathology. All phalangeal lesions were non-malignant and 62% were either giant cell tumours or giant cell reactions. Giant cell reaction was confined to upper limb bones; metatarsals were afflicted exclusively with giant cell tumours (n=3) while malignant lesions affected the metacarpals in two and carpal bones in one instance. Aneurysmal bone cysts were seen exclusively in the tarsal (n=4) and carpal bones (n=2), a very rare finding. More cases need to be studied to define patterns of lesions of hands and feet. The definitive diagnosis is essential as many patients with osteolytic lesions may not require surgical intervention.


Assuntos
Cistos Ósseos Aneurismáticos/diagnóstico por imagem , Neoplasias Ósseas/diagnóstico por imagem , Ossos do Pé/diagnóstico por imagem , Tumor de Células Gigantes do Osso/diagnóstico por imagem , Ossos da Mão/diagnóstico por imagem , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Feminino , Humanos , Índia , Masculino , Pessoa de Meia-Idade , Radiografia , Estudos Retrospectivos , Centros de Atenção Terciária , Adulto Jovem
16.
Biochim Biophys Acta ; 1833(12): 2639-2652, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23820376

RESUMO

Tight control between activation and attenuation of granulocyte colony stimulating factor receptor (G-CSFR) signaling is essential to regulate survival, proliferation and differentiation of myeloid progenitor cells. Previous studies demonstrated negative regulation of G-CSFR through endosomal-lysosomal routing and ubiquitin-proteasome mediated degradation. However, very few E3 ubiquitin ligases are known to target G-CSFR for ubiquitin-proteasome pathway. Here we identified F-box and WD repeat domain-containing 7 (Fbw7), a substrate recognizing component of Skp-Cullin-F box (SCF) E3 ubiquitin Ligase physically associates with G-CSFR and promotes its ubiquitin-mediated proteasomal degradation. Our data shows that Fbw7 also interacts with and degrades G-CSFR-T718 (a truncated mutant of G-CSFR found in severe congenital neutropenia/acute myeloid leukemia (SCN/AML patients)) though at a quite slower rate compared to G-CSFR. We further show that glycogen synthase kinase 3 beta (GSK3ß), like Fbw7 also targets G-CSFR and G-CSFR-T718 for degradation; however, Fbw7 and GSK3ß are interdependent in targeting G-CSFR/G-CSFR-T718 for degradation because they are unable to degrade G-CSFR individually when either of them is knocked down. We further show that Fbw7 mediated downregulation of G-CSFR inhibits signal transducer and activator of transcription 3 (STAT3) phosphorylation which is required for G-CSF dependent granulocytic differentiation. In addition, our data also shows that inhibition of Fbw7 restores G-CSFR signaling leading to enhanced STAT3 activity resulting in massive granulocytic differentiation. These data indicate that Fbw7 together with GSK3ß negatively regulates G-CSFR expression and its downstream signaling.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Proteínas F-Box/metabolismo , Granulócitos/citologia , Granulócitos/metabolismo , Proteólise , Receptores de Fator Estimulador de Colônias de Granulócitos/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Linhagem Celular , Proteína 7 com Repetições F-Box-WD , Técnicas de Silenciamento de Genes , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Cinética , Camundongos , Proteínas Mutantes/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Fator de Transcrição STAT3/metabolismo , Ubiquitina/metabolismo
17.
PLoS One ; 8(6): e65330, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23762344

RESUMO

CCAAT/Enhancer Binding Protein Alpha (C/EBPα) is a key transcription factor involved in the adipocyte differentiation. Here for the first time we demonstrate that E6AP, an E3 ubiquitin ligase inhibits adipocyte differentiation in 3T3-L1 cells as revealed by reduced lipid staining with oil red. Knock down of E6AP in mouse 3T3L1 preadipocytes is sufficient to convert them to adipocytes independent of external hormonal induction. C/EBPα protein level is drastically increased in E6AP deficient 3T3L1 preadipocytes while inverse is observed when wild type E6AP is over expressed. We show that transient transfection of wild type E6AP downregulates C/EBPα protein expression in a dose dependent manner while catalytically inactive E6AP-C843A rather stabilizes it. In addition, wild type E6AP inhibits expression of proadipogenic genes while E6AP-C843A enhances them. More importantly, overexpression of E6AP-C843A in mesenchymal progenitor cells promotes accumulation of lipid droplets while there is drastically reduced lipid droplet formation when E6AP is over expressed. Taken together, our finding suggests that E6AP may negatively control adipogenesis by inhibiting C/EBPα expression by targeting it to ubiquitin-proteasome pathway for degradation.


Assuntos
Adipogenia , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Regulação para Baixo , Ubiquitina-Proteína Ligases/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Biocatálise , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Diferenciação Celular , Separação Celular , Técnicas de Silenciamento de Genes , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Proteínas Mutantes/metabolismo , RNA Interferente Pequeno/metabolismo , Ativação Transcricional/genética , Regulação para Cima
18.
Proteomics ; 13(14): 2100-12, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23576398

RESUMO

Nuclear receptor coregulators play an important role in the transcriptional regulation of nuclear receptors. In the present study, we aimed to identify estrogen receptor α (ERα) interacting proteins in Tamoxifen treated MCF7 cells. Using in vitro GST-pull down assay with ERα ligand-binding domain (ERα-LBD) and MS-based proteomics approach we identified Profilin1 as a novel ERα interacting protein. Profilin1 contains I/LXX/L/H/I amino acid signature motif required for corepressor interaction with ERα. We show that these two proteins physically interact with each other both in vitro as well as in vivo by GST-pull down and coimmunoprecipitation, respectively. We further show that these two proteins also colocalize together in the nucleus. Previous studies have reported reduced expression of Profilin1 in breast cancer; and here we found that Tamoxifen increases Profilin1 expression in MCF7 cells. Our data demonstrate that over expression of Profilin1 inhibits ERα-mediated transcriptional activation as well as its downstream target genes in ERα positive breast cancer cells MCF7. In addition, Profilin1 overexpression in MCF7 cells leads to inhibition of cell proliferation that apparently is due to enhanced apoptosis. In nutshell, these data indicate that MS-based proteomics approach identifies a novel ERα interacting protein Profilin1 that serves as a putative corepressor of ERα functions.


Assuntos
Neoplasias da Mama/metabolismo , Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/metabolismo , Profilinas/química , Profilinas/metabolismo , Proteoma/análise , Motivos de Aminoácidos , Sequência de Aminoácidos , Neoplasias da Mama/tratamento farmacológico , Feminino , Humanos , Células MCF-7 , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Proteômica/métodos , Tamoxifeno/farmacologia
19.
Expert Opin Drug Discov ; 7(12): 1165-75, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22971110

RESUMO

INTRODUCTION: Global protein expression profiling between healthy vs diseased states helps identifying differential expression and post-translational modifications of proteins, thereby providing better insights into the molecular changes of disease diagnosis and prognosis. In addition, analytical separation and identification of proteins from complex mixtures can provide insight into targeted drug therapy and prediction of response to different therapeutics. AREAS COVERED: In the present review the authors summarize the readily available quantitative proteomics tools for the analytical separation and identification of target proteins in myeloid leukemia, AML in particular, and its future perspectives in its diagnostics and therapeutics. Within, the authors highlight some of the proteomics approaches such as gel-based quantitation strategies (2D, 2D-DIGE); MS-based quantitative proteomics tools (metabolic labeling (SILAC), chemical labeling (ITRAQ, ICAT)); MS techniques (MALDI-MS/MS). In addition, some of the target proteins identified using proteomics approaches in myeloid leukemia are also discussed that may encourage cancer biology investigators to undertake proteomics as a vital tool in their study. EXPERT OPINION: With suitable, selective application of diverse set of quantitative proteomics strategies integrated with bioinformatics software and precise statistical analysis in myeloid leukemia holds tremendous promise in deciphering cancer proteome, understanding tumor pathophysiology and development of personalized molecular medicine and therapy.


Assuntos
Técnicas de Química Analítica/métodos , Descoberta de Drogas/métodos , Leucemia Mieloide/tratamento farmacológico , Proteínas de Neoplasias/metabolismo , Proteoma/metabolismo , Proteômica/métodos , Antineoplásicos/uso terapêutico , Cromatografia Líquida/métodos , Eletroforese em Gel Bidimensional/métodos , Humanos , Leucemia Mieloide/metabolismo , Espectrometria de Massas/métodos
20.
Proteomics ; 12(9): 1363-77, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22589186

RESUMO

Tamoxifen (Tam) is most widely used selective estrogen receptor modulator (SERM) for treatment of hormone-responsive breast cancer. Despite being regularly used in clinical therapy for breast cancer since 1971, the mechanism of Tam action remains largely unclear. In order to gain insights into Tam-mediated antibreast cancer actions, we applied 2DE and MS based proteomics approach to identify target proteins of Tam. We identified E6-associated protein, i.e. E6AP (UBE3A) among others to be regulated by Tam that otherwise is upregulated in breast tumors. We confirmed our 2DE finding by immunoblotting and further show that Tam leads to inhibition of E6AP expression presumably by promoting its autoubiquitination, which is coupled with nuclear export and subsequent proteasome-mediated degradation. Furthermore, we show that Tam- and siE6AP-mediated inhibition of E6AP leads to enhanced G0-G1 growth arrest and apoptosis, which is also evident from significant upregulation of cytochrome-c, Bax, p21, and PARP cleavage. Taken together, our data suggest that, Tam-targeted E6AP inhibition is in fact required for Tam-mediated antibreast cancer actions. Thus, E6AP may be a therapeutic target in breast cancer.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Tamoxifeno/farmacologia , Ubiquitina-Proteína Ligases/metabolismo , Antineoplásicos Hormonais/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citoplasma/metabolismo , Eletroforese em Gel Bidimensional , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Espectrometria de Massas , Terapia de Alvo Molecular/métodos , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas/química , Proteínas/classificação , Proteínas/metabolismo , Proteoma/análise , Proteoma/efeitos dos fármacos , Proteoma/metabolismo , Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA