Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
J Biol Chem ; 299(6): 104786, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37146968

RESUMO

The E3 ubiquitin ligase APC/C-Cdh1 maintains the G0/G1 state, and its inactivation is required for cell cycle entry. We reveal a novel role for Fas-associated protein with death domain (FADD) in the cell cycle through its function as an inhibitor of APC/C-Cdh1. Using real-time, single-cell imaging of live cells combined with biochemical analysis, we demonstrate that APC/C-Cdh1 hyperactivity in FADD-deficient cells leads to a G1 arrest despite persistent mitogenic signaling through oncogenic EGFR/KRAS. We further show that FADDWT interacts with Cdh1, while a mutant lacking a consensus KEN-box motif (FADDKEN) fails to interact with Cdh1 and results in a G1 arrest due to its inability to inhibit APC/C-Cdh1. Additionally, enhanced expression of FADDWT but not FADDKEN, in cells arrested in G1 upon CDK4/6 inhibition, leads to APC/C-Cdh1 inactivation and entry into the cell cycle in the absence of retinoblastoma protein phosphorylation. FADD's function in the cell cycle requires its phosphorylation by CK1α at Ser-194 which promotes its nuclear translocation. Overall, FADD provides a CDK4/6-Rb-E2F-independent "bypass" mechanism for cell cycle entry and thus a therapeutic opportunity for CDK4/6 inhibitor resistance.


Assuntos
Proteínas de Ciclo Celular , Ubiquitina-Proteína Ligases , Humanos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Divisão Celular , Expressão Gênica , Células HEK293 , Mutação , Domínios Proteicos , Transporte Proteico/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
2.
Dis Esophagus ; 35(3)2022 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-35022679

RESUMO

Esophageal stricture is the most common delayed sequelae of aerodigestive tract corrosive injuries. Early endoscopic dilatation is an integral part of corrosive injury management. Self-dilatation of the esophagus is effective in preventing stricture recurrence. In this prospective study, we included patients with corrosive aerodigestive tract injury from January 2009 to December 2020. We analyzed the outcome of the endoscopic dilatation and self-dilatation treatments administered to patients with a corrosive esophageal stricture. Among 295 patients, 164 had an esophageal injury, 73 had esophago-gastric injury, 55 had a gastric injury, and 3 had the pharyngeal injury. Of the 295 patients, 194 (81.85%) underwent dilatation, and 13 patients with diffuse esophageal injury underwent upfront surgery. Successful dilatation was performed in 169 (87.11%) patients. Of the 68 patients undergoing self-dilatation, 63 patients achieved nutritional autonomy by 28 days. Early endoscopic dilatation effectively prevents surgery, and self-dilatation appears promising to prevent recurrent esophageal stricture.


Assuntos
Queimaduras Químicas , Cáusticos , Estenose Esofágica , Queimaduras Químicas/complicações , Queimaduras Químicas/terapia , Cáusticos/toxicidade , Dilatação , Estenose Esofágica/induzido quimicamente , Estenose Esofágica/terapia , Esôfago/lesões , Esôfago/cirurgia , Humanos , Estudos Prospectivos , Centros de Atenção Terciária
3.
J Biol Chem ; 295(36): 12661-12673, 2020 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-32669362

RESUMO

The discovery of activating epidermal growth factor receptor (EGFR) mutations spurred the use of EGFR tyrosine kinase inhibitors (TKIs), such as erlotinib, as the first-line treatment of lung cancers. We previously reported that differential degradation of TKI-sensitive (e.g. L858R) and resistant (T790M) EGFR mutants upon erlotinib treatment correlates with drug sensitivity. We also reported that SMAD ubiquitination regulatory factor 2 (SMURF2) ligase activity is important in stabilizing EGFR. However, the molecular mechanisms involved remain unclear. Here, using in vitro and in vivo ubiquitination assays, MS, and superresolution microscopy, we show SMURF2-EGFR functional interaction is important for EGFR stability and response to TKI. We demonstrate that L858R/T790M EGFR is preferentially stabilized by SMURF2-UBCH5 (an E3-E2)-mediated polyubiquitination. We identified four lysine residues as the sites of ubiquitination and showed that replacement of one of them with acetylation-mimicking glutamine increases the sensitivity of mutant EGFR to erlotinib-induced degradation. We show that SMURF2 extends membrane retention of EGF-bound EGFR, whereas SMURF2 knockdown increases receptor sorting to lysosomes. In lung cancer cell lines, SMURF2 overexpression increased EGFR levels, improving TKI tolerance, whereas SMURF2 knockdown decreased EGFR steady-state levels and sensitized lung cancer cells. Overall, we propose that SMURF2-mediated polyubiquitination of L858R/T790M EGFR competes with acetylation-mediated receptor internalization that correlates with enhanced receptor stability; therefore, disruption of the E3-E2 complex may be an attractive target to overcome TKI resistance.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Cloridrato de Erlotinib/farmacologia , Neoplasias Pulmonares/enzimologia , Mutação de Sentido Incorreto , Inibidores de Proteínas Quinases/farmacologia , Ubiquitina-Proteína Ligases/metabolismo , Substituição de Aminoácidos , Animais , Células CHO , Cricetulus , Resistencia a Medicamentos Antineoplásicos/genética , Estabilidade Enzimática/efeitos dos fármacos , Estabilidade Enzimática/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Células HEK293 , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Células MCF-7 , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética
4.
J Biol Chem ; 295(18): 5906-5917, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32165494

RESUMO

We previously reported that overexpression of cytochrome P450 family 24 subfamily A member 1 (CYP24A1) increases lung cancer cell proliferation by activating RAS signaling and that CYP24A1 knockdown inhibits tumor growth. However, the mechanism of CYP24A1-mediated cancer cell proliferation remains unclear. Here, we conducted cell synchronization and biochemical experiments in lung adenocarcinoma cells, revealing a link between CYP24A1 and anaphase-promoting complex (APC), a key cell cycle regulator. We demonstrate that CYP24A1 expression is cell cycle-dependent; it was higher in the G2-M phase and diminished upon G1 entry. CYP24A1 has a functional destruction box (D-box) motif that allows binding with two APC adaptors, CDC20-homologue 1 (CDH1) and cell division cycle 20 (CDC20). Unlike other APC substrates, however, CYP24A1 acted as a pseudo-substrate, inhibiting CDH1 activity and promoting mitotic progression. Conversely, overexpression of a CYP24A1 D-box mutant compromised CDH1 binding, allowing CDH1 hyperactivation, thereby hastening degradation of its substrates cyclin B1 and CDC20, and accumulation of the CDC20 substrate p21, prolonging mitotic exit. These activities also occurred with a CYP24A1 isoform 2 lacking the catalytic cysteine (Cys-462), suggesting that CYP24A1's oncogenic potential is independent of its catalytic activity. CYP24A1 degradation reduced clonogenic survival of mutant KRAS-driven lung cancer cells, and calcitriol treatment increased CYP24A1 levels and tumor burden in Lsl-KRASG12D mice. These results disclose a catalytic activity-independent growth-promoting role of CYP24A1 in mutant KRAS-driven lung cancer. This suggests that CYP24A1 could be therapeutically targeted in lung cancers in which its expression is high.


Assuntos
Adenocarcinoma de Pulmão/patologia , Biocatálise , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética , Vitamina D3 24-Hidroxilase/metabolismo , Adenocarcinoma de Pulmão/genética , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Sobrevivência Celular , Regulação Neoplásica da Expressão Gênica , Humanos , Regulação para Cima , Vitamina D3 24-Hidroxilase/genética
5.
Gastroenterology ; 158(3): 583-597.e1, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31715145

RESUMO

BACKGROUND & AIMS: Barrett's esophagus (BE) can progress to dysplasia and esophageal adenocarcinoma (EAC), accompanied by mutations in TP53 that increase the stability of its product, p53. We analyzed BE tissues for messenger RNAs (mRNAs) that associate with BE progression and identified one that affects the stabilization of p53. METHODS: We obtained 54 BE samples collected from patients with high-grade dysplasia (HGD) or esophageal adenocarcinoma (EAC), from 1992 through 2015, and performed RNA sequence analyses, including isoform-specific analyses. We performed reverse-transcription polymerase chain reaction analyses of 166 samples and immunohistochemical analyses of tissue microarrays that contained BE tissues from 100 patients with HGD or EAC and normal esophageal squamous mucosa (controls). Proteins were expressed from transfected plasmids or knocked down with small interfering RNAs in BE cells and analyzed by immunoblots and in immunoprecipitation and ubiquitin ligase assays. Athymic nude mice bearing EAC xenograft tumors (grown from OE-33 cells) were given intraperitoneal injections of simvastatin; tumor growth was monitored and tumors were collected and analyzed by immunoblotting for levels of RNF128, p53, and acetylated p53. RESULTS: Progression of BE to HGD or EAC associated with changes in expression of mRNAs that encoded mucins and promoted inflammation and activation of ATM and the DNA damage response. As tissues progressed from BE to HGD to EAC, they increased expression of mRNAs encoding isoform 1 of RNF128 (Iso1) and decreased expression of Iso2 of RNF128. RNF128 is an E3 ubiquitin ligase that targets p53 for degradation. Incubation of BE cells with interferon gamma caused them to increase expression of Iso1 and reduce expression of Iso2. Iso1 was heavily glycosylated with limited ubiquitin ligase activity for p53, resulting in p53 stabilization. Knockdown of Iso1 in BE and EAC cells led to degradation of the mutant form of p53 and reduced clonogenic survival. In contrast, Iso2 was a potent ligase that reduced levels of the mutant form of p53 in BE cells. In BE cells, Iso2 was hypoglycosylated and degraded, via ATM and GSK3ß-mediated phosphorylation and activation of the beta-TrCP1-containing SCF ubiquitin ligase complex. Simvastatin, which degrades the mutant form of p53, also degraded RNF128 Iso1 protein in BE cells and slowed growth of EAC xenograft tumors in mice. CONCLUSIONS: We found that isoform 2 of RNF128 is decreased in BE cells, resulting in increased levels of mutant p53, whereas isoform 1 of RNF128 is increased in BE cells, further promoting the stabilization of mutant p53.


Assuntos
Adenocarcinoma/genética , Esôfago de Barrett/genética , Neoplasias Esofágicas/genética , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Esôfago de Barrett/metabolismo , Esôfago de Barrett/patologia , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Esôfago/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Glicosilação , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Interferon gama/farmacologia , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos , Camundongos Nus , Transplante de Neoplasias , RNA Mensageiro/metabolismo , Transdução de Sinais , Sinvastatina/farmacologia , Proteína Supressora de Tumor p53/genética , Ubiquitina-Proteína Ligases/metabolismo
6.
Gastroenterology ; 156(5): 1404-1415, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30578782

RESUMO

BACKGROUND & AIMS: African American and European American individuals have a similar prevalence of gastroesophageal reflux disease (GERD), yet esophageal adenocarcinoma (EAC) disproportionately affects European American individuals. We investigated whether the esophageal squamous mucosa of African American individuals has features that protect against GERD-induced damage, compared with European American individuals. METHODS: We performed transcriptional profile analysis of esophageal squamous mucosa tissues from 20 African American and 20 European American individuals (24 with no disease and 16 with Barrett's esophagus and/or EAC). We confirmed our findings in a cohort of 56 patients and analyzed DNA samples from patients to identify associated variants. Observations were validated using matched genomic sequence and expression data from lymphoblasts from the 1000 Genomes Project. A panel of esophageal samples from African American and European American subjects was used to confirm allele-related differences in protein levels. The esophageal squamous-derived cell line Het-1A and a rat esophagogastroduodenal anastomosis model for reflux-generated esophageal damage were used to investigate the effects of the DNA-damaging agent cumene-hydroperoxide (cum-OOH) and a chemopreventive cranberry proanthocyanidin (C-PAC) extract, respectively, on levels of protein and messenger RNA (mRNA). RESULTS: We found significantly higher levels of glutathione S-transferase theta 2 (GSTT2) mRNA in squamous mucosa from African American compared with European American individuals and associated these with variants within the GSTT2 locus in African American individuals. We confirmed that 2 previously identified genomic variants at the GSTT2 locus, a 37-kb deletion and a 17-bp promoter duplication, reduce expression of GSTT2 in tissues from European American individuals. The nonduplicated 17-bp promoter was more common in tissue samples from populations of African descendant. GSTT2 protected Het-1A esophageal squamous cells from cum-OOH-induced DNA damage. Addition of C-PAC increased GSTT2 expression in Het-1A cells incubated with cum-OOH and in rats with reflux-induced esophageal damage. C-PAC also reduced levels of DNA damage in reflux-exposed rat esophagi, as observed by reduced levels of phospho-H2A histone family member X. CONCLUSIONS: We found GSTT2 to protect esophageal squamous cells against DNA damage from genotoxic stress and that GSTT2 expression can be induced by C-PAC. Increased levels of GSTT2 in esophageal tissues of African American individuals might protect them from GERD-induced damage and contribute to the low incidence of EAC in this population.


Assuntos
Adenocarcinoma/genética , Esôfago de Barrett/genética , Negro ou Afro-Americano/genética , Dano ao DNA , Mucosa Esofágica/enzimologia , Neoplasias Esofágicas/genética , Refluxo Gastroesofágico/genética , Glutationa Transferase/genética , População Branca/genética , Adenocarcinoma/enzimologia , Adenocarcinoma/etnologia , Adenocarcinoma/patologia , Animais , Esôfago de Barrett/enzimologia , Esôfago de Barrett/etnologia , Esôfago de Barrett/patologia , Modelos Animais de Doenças , Mucosa Esofágica/patologia , Neoplasias Esofágicas/enzimologia , Neoplasias Esofágicas/etnologia , Neoplasias Esofágicas/patologia , Feminino , Refluxo Gastroesofágico/enzimologia , Refluxo Gastroesofágico/etnologia , Refluxo Gastroesofágico/patologia , Glutationa Transferase/metabolismo , Células HeLa , Histonas/metabolismo , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Fosfoproteínas/metabolismo , Fosforilação , Fatores de Proteção , Ratos Sprague-Dawley , Fatores de Risco , Estados Unidos/epidemiologia , Regulação para Cima
7.
J Biol Chem ; 288(37): 26879-86, 2013 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-23897823

RESUMO

An eight-amino acid segment is known to be responsible for the marked difference in the rates of degradation of the EGF receptor (ErbB1) and ErbB2 upon treatment of cells with the Hsp90 inhibitor geldanamycin. We have scrambled the first six amino acids of this segment of the EGF receptor (EGFR), which lies in close association with the ATP binding cleft and the dimerization face. Scrambling these six amino acids markedly reduces EGFR stability, EGF-stimulated receptor dimerization, and autophosphorylation activity. Two peptides were synthesized as follows: one containing the wild-type sequence of the eight-amino acid segment, which we call Disruptin; and one with the scrambled sequence. Disruptin inhibits Hsp90 binding to the EGFR and causes slow degradation of the EGFR in two EGFR-dependent cancer cell lines, whereas the scrambled peptide is inactive. This effect is specific for EGFR versus other Hsp90 client proteins. In the presence of EGF, Disruptin, but not the scrambled peptide, inhibits EGFR dimerization and causes rapid degradation of the EGFR. In contrast to the Hsp90 inhibitor geldanamycin, Disruptin inhibits cancer cell growth by a nonapoptotic mechanism. Disruptin provides proof of concept for the development of a new class of anti-tumor drugs that specifically cause EGFR degradation.


Assuntos
Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Fragmentos de Peptídeos/farmacologia , Peptídeos/farmacologia , Animais , Antineoplásicos/farmacologia , Benzoquinonas/farmacologia , Células CHO , Linhagem Celular Tumoral , Cricetulus , Dimerização , Desenho de Fármacos , Receptores ErbB/farmacologia , Humanos , Lactamas Macrocíclicas/farmacologia , Mutagênese Sítio-Dirigida , Mutação , Fosforilação , Ligação Proteica
8.
Cell Death Dis ; 15(3): 194, 2024 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-38453895

RESUMO

The advancement of RNAseq and isoform-specific expression platforms has led to the understanding that isoform changes can alter molecular signaling to promote tumorigenesis. An active area in cancer research is uncovering the roles of ubiquitination on spliceosome assembly contributing to transcript diversity and expression of alternative isoforms. However, the effects of isoform changes on functionality of ubiquitination machineries (E1, E2, E3, E4, and deubiquitinating (DUB) enzymes) influencing onco- and tumor suppressor protein stabilities is currently understudied. Characterizing these changes could be instrumental in improving cancer outcomes via the identification of novel biomarkers and targetable signaling pathways. In this review, we focus on highlighting reported examples of direct, protein-coded isoform variation of ubiquitination enzymes influencing cancer development and progression in gastrointestinal (GI) malignancies. We have used a semi-automated system for identifying relevant literature and applied established systems for isoform categorization and functional classification to help structure literature findings. The results are a comprehensive snapshot of known isoform changes that are significant to GI cancers, and a framework for readers to use to address isoform variation in their own research. One of the key findings is the potential influence that isoforms of the ubiquitination machinery have on oncoprotein stability.


Assuntos
Neoplasias Gastrointestinais , Humanos , Ubiquitinação , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Neoplasias Gastrointestinais/genética , Carcinogênese , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
9.
JCI Insight ; 9(13)2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38781019

RESUMO

Immunosuppression is a common feature of esophageal adenocarcinoma (EAC) and has been linked to poor overall survival (OS). We hypothesized that upstream factors might negatively influence CD3 levels and T cell activity, thus promoting immunosuppression and worse survival. We used clinical data and patient samples of those who progressed from Barrett's to dysplasia to EAC, investigated gene (RNA-Seq) and protein (tissue microarray) expression, and performed cell biology studies to delineate a pathway impacting CD3 protein stability that might influence EAC outcome. We showed that the loss of both CD3-ε expression and CD3+ T cell number correlated with worse OS in EAC. The gene related to anergy in lymphocytes isoform 1 (GRAIL1), which is the prominent isoform in EACs, degraded (ε, γ, δ) CD3s and inactivated T cells. In contrast, isoform 2 (GRAIL2), which is reduced in EACs, stabilized CD3s. Further, GRAIL1-mediated CD3 degradation was facilitated by interferon-stimulated gene 15 (ISG15), a ubiquitin-like protein. Consequently, the overexpression of a ligase-dead GRAIL1, ISG15 knockdown, or the overexpression of a conjugation-defective ISG15-leucine-arginine-glycine-glycine mutant could increase CD3 levels. Together, we identified an ISG15/GRAIL1/mutant p53 amplification loop negatively influencing CD3 levels and T cell activity, thus promoting immunosuppression in EAC.


Assuntos
Adenocarcinoma , Complexo CD3 , Citocinas , Neoplasias Esofágicas , Ubiquitinas , Humanos , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/mortalidade , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/imunologia , Adenocarcinoma/genética , Adenocarcinoma/mortalidade , Adenocarcinoma/patologia , Adenocarcinoma/metabolismo , Adenocarcinoma/imunologia , Complexo CD3/metabolismo , Complexo CD3/genética , Citocinas/metabolismo , Ubiquitinas/metabolismo , Ubiquitinas/genética , Masculino , Linfócitos T/metabolismo , Linfócitos T/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica , Esôfago de Barrett/patologia , Esôfago de Barrett/genética , Esôfago de Barrett/metabolismo , Pessoa de Meia-Idade
10.
Cell Mol Gastroenterol Hepatol ; 13(1): 129-149, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34416429

RESUMO

BACKGROUND & AIMS: TP53 mutations underlie Barrett's esophagus (BE) progression to dysplasia and cancer. During BE progression, the ubiquitin ligase (E3) RNF128/GRAIL switches expression from isoform 2 (Iso2) to Iso1, stabilizing mutant p53. However, the ubiquitin-conjugating enzyme (E2) that partners with Iso1 to stabilize mutant p53 is unknown. METHODS: Single-cell RNA sequencing of paired normal esophagus and BE tissues identified candidate E2s, further investigated in expression data from BE to esophageal adenocarcinoma (EAC) progression samples. Biochemical and cellular studies helped clarify the role of RNF128-E2 on mutant p53 stability. RESULTS: The UBE2D family member 2D3 (UBCH5C) is the most abundant E2 in normal esophagus. However, during BE to EAC progression, loss of UBE2D3 copy number and reduced expression of RNF128 Iso2 were noted, 2 known p53 degraders. In contrast, expression of UBE2D1 (UBCH5A) and RNF128 Iso1 in dysplastic BE and EAC forms an inactive E2-E3 complex, stabilizing mutant p53. To destabilize mutant p53, we targeted RNF128 Iso1 either by mutating asparagine (N48, 59, and 101) residues to block glycosylation to facilitate ß-TrCP1-mediated degradation or by mutating proline (P54 and 105) residues to restore p53 polyubiquitinating ability. In addition, either loss of UBCH5A catalytic activity, or disruption of the Iso1-UBCH5A interaction promoted Iso1 loss. Consequently, overexpression of either catalytically dead or Iso1-binding-deficient UBCH5A mutants destabilized Iso1 to degrade mutant p53, thus compromising the clonogenic survival of mutant p53-dependent BE cells. CONCLUSIONS: Loss of RNF128 Iso2-UBCH5C and persistence of the Iso1-UBCH5A complex favors mutant p53 stability to promote BE cell survival. Therefore, targeting of Iso1-UBCH5A may provide a novel therapeutic strategy to prevent BE progression.


Assuntos
Adenocarcinoma , Esôfago de Barrett , Neoplasias Esofágicas , Proteína Supressora de Tumor p53 , Enzimas de Conjugação de Ubiquitina , Ubiquitina-Proteína Ligases , Adenocarcinoma/patologia , Esôfago de Barrett/genética , Esôfago de Barrett/patologia , Progressão da Doença , Neoplasias Esofágicas/patologia , Humanos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
11.
Transl Oncol ; 21: 101428, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35460942

RESUMO

Grade 2 and higher radiation pneumonitis (RP2) is a potentially fatal toxicity that limits efficacy of radiation therapy (RT). We wished to identify a combined biomarker signature of circulating miRNAs and cytokines which, along with radiobiological and clinical parameters, may better predict a targetable RP2 pathway. In a prospective clinical trial of response-adapted RT for patients (n = 39) with locally advanced non-small cell lung cancer, we analyzed patients' plasma, collected pre- and during RT, for microRNAs (miRNAs) and cytokines using array and multiplex enzyme linked immunosorbent assay (ELISA), respectively. Interactions between candidate biomarkers, radiobiological, and clinical parameters were analyzed using data-driven Bayesian network (DD-BN) analysis. We identified alterations in specific miRNAs (miR-532, -99b and -495, let-7c, -451 and -139-3p) correlating with lung toxicity. High levels of soluble tumor necrosis factor alpha receptor 1 (sTNFR1) were detected in a majority of lung cancer patients. However, among RP patients, within 2 weeks of RT initiation, we noted a trend of temporary decline in sTNFR1 (a physiological scavenger of TNFα) and ADAM17 (a shedding protease that cleaves both membrane-bound TNFα and TNFR1) levels. Cytokine signature identified activation of inflammatory pathway. Using DD-BN we combined miRNA and cytokine data along with generalized equivalent uniform dose (gEUD) to identify pathways with better accuracy of predicting RP2 as compared to either miRNA or cytokines alone. This signature suggests that activation of the TNFα-NFκB inflammatory pathway plays a key role in RP which could be specifically ameliorated by etanercept rather than current therapy of non-specific leukotoxic corticosteroids.

12.
Cell Mol Gastroenterol Hepatol ; 11(2): 449-464, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33130332

RESUMO

The incidence of esophageal adenocarcinoma (EAC) and other gastrointestinal (GI) cancers have risen dramatically, thus defining the oncogenic drivers to develop effective therapies are necessary. Patients with Barrett's Esophagus (BE), have an elevated risk of developing EAC. Around 70%-80% of BE cases that progress to dysplasia and cancer have detectable TP53 mutations. Similarly, in other GI cancers higher rates of TP53 mutation are reported, which provide a significant survival advantage to dysplastic/cancer cells. Targeting molecular chaperones that mediate mutant p53 stability may effectively induce mutant p53 degradation and improve cancer outcomes. Statins can achieve this via disrupting the interaction between mutant p53 and the chaperone DNAJA1, promoting CHIP-mediated degradation of mutant p53, and statins are reported to significantly reduce the risk of BE progression to EAC. However, statins demonstrated sub-optimal efficacy depending on cancer types and TP53 mutation specificity. Besides the well-established role of MDM2 in p53 stability, we reported that individual isoforms of the E3 ubiquitin ligase GRAIL (RNF128) are critical, tissue-specific regulators of mutant p53 stability in BE progression to EAC, and targeting the interaction of mutant p53 with these isoforms may help mitigate EAC development. In this review, we discuss the critical ubiquitin-proteasome and chaperone regulation of mutant p53 stability in EAC and other GI cancers with future insights as to how to affect mutant p53 stability, further noting how the precise p53 mutation may influence the efficacy of treatment strategies and identifying necessary directions for further research in this field.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Gastrointestinais/genética , Chaperonas Moleculares/metabolismo , Proteína Supressora de Tumor p53/genética , Ubiquitina-Proteína Ligases/metabolismo , Antineoplásicos/uso terapêutico , Neoplasias Gastrointestinais/tratamento farmacológico , Neoplasias Gastrointestinais/patologia , Humanos , Chaperonas Moleculares/antagonistas & inibidores , Terapia de Alvo Molecular/métodos , Mutação , Estabilidade Proteica/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases/antagonistas & inibidores
13.
Transl Oncol ; 14(1): 100950, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33395747

RESUMO

INTRODUCTION: Radiation therapy for the management of intrahepatic malignancies can adversely affect liver function. Liver damage has been associated with increased levels of inflammatory cytokines, including tumor necrosis factor alpha (TNFα). We hypothesized that an inflammatory state, characterized by increased soluble TNFα receptor (sTNFR1), mediates sensitivity of the liver to radiation. MATERIALS/METHODS: Plasma samples collected during 3 trials of liver radiation for liver malignancies were assayed for sTNFR1 level via enzyme-linked immunosorbent assay (ELISA). Univariate and multivariate logistic regression and longitudinal models were used to characterize associations between liver toxicity (defined as a ≥2-point increase in Child-Pugh [CP] score within 6 months of radiation treatment) and sTNFR1 levels, ALBI score, biocorrected mean liver dose (MLD), age, and baseline laboratory values. RESULTS: Samples from 78 patients given liver stereotactic body radiation therapy [SBRT] (92%) or hypofractionated radiation were examined. There was a significant association between liver toxicity and sTNFR1 levels, and higher values were associated with increased toxicity over a range of mean liver doses. When ALBI score and biocorrected dose were included in the model with sTNFR1, baseline ALBI score and change in ALBI (ΔALBI) were significantly associated with toxicity, but sTNFR1 was not. Baseline aminotransferase levels also predicted toxicity but not independently of ALBI score. CONCLUSIONS: Elevated plasma sTNFR1 levels are associated with liver injury after liver radiation, suggesting that elevated inflammatory cytokine activity is a predictor of radiation-induced liver dysfunction. Future studies should determine whether administration of agents that decrease inflammation prior to treatment is warranted.

14.
JCI Insight ; 6(1)2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33290281

RESUMO

Esophageal adenocarcinoma (EAC) develops from Barrett's esophagus (BE), a chronic inflammatory state that can progress through a series of transformative dysplastic states before tumor development. While molecular and genetic changes of EAC tumors have been studied, immune microenvironment changes during Barrett's progression to EAC remain poorly understood. In this study, we identify potential immunologic changes that can occur during BE-to-EAC progression. RNA sequencing (RNA-Seq) analysis on tissue samples from EAC patients undergoing surgical resection demonstrated that a subset of chemokines and cytokines, most notably IL6 and CXCL8, increased during BE progression to EAC. xCell deconvolution analysis investigating immune cell population changes demonstrated that the largest changes in expression during BE progression occurred in M2 macrophages, pro-B cells, and eosinophils. Multiplex immunohistochemical staining of tissue microarrays showed increased immune cell populations during Barrett's progression to high-grade dysplasia. In contrast, EAC tumor sections were relatively immune poor, with a rise in PD-L1 expression and loss of CD8+ T cells. These data demonstrate that the EAC microenvironment is characterized by poor cytotoxic effector cell infiltration and increased immune inhibitory signaling. These findings suggest an immunosuppressive microenvironment, highlighting the need for further studies to explore immune modulatory therapy in EAC.


Assuntos
Adenocarcinoma/imunologia , Esôfago de Barrett/imunologia , Neoplasias Esofágicas/imunologia , Adenocarcinoma/genética , Adenocarcinoma/patologia , Esôfago de Barrett/genética , Esôfago de Barrett/patologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Quimiocinas/genética , Quimiocinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Progressão da Doença , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Humanos , Tolerância Imunológica , Imuno-Histoquímica , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Macrófagos/imunologia , Macrófagos/patologia , RNA-Seq , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
15.
Trop Gastroenterol ; 31(4): 291-4, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21568145

RESUMO

BACKGROUND: Radiation induced haemorrhagic proctitis is a well recognized complication of pelvic radiotherapy and its treatment is quite challenging. AIM: To evaluate the efficacy of local instillation of 4% formalin in the management of uncontrolled radiation induced haemorrhagic proctitis. METHODS: This prospective study included twenty-three patients who underwent formalin instillation. We documented the control of the symptoms, assessed the outcome of the treatment, presence of complications & long term results. RESULTS: All patients were followed for a median of 13 months. In 15 patients, the bleeding stopped after a single treatment and in 6 after a second session. Only two patients required formalin instillation for the third time. During the follow-up period all patients were reviewed systemically with reference to the pretreatment complaints. No recurrent rectal bleeding occurred, bowel frequency was decreased and no further blood transfusion was required. CONCLUSION: Local instillation of 4% formalin is an effective treatment for uncontrolled radiation induced haemorrhagic proctitis.


Assuntos
Formaldeído/administração & dosagem , Hemorragia , Proctite , Lesões por Radiação/complicações , Lesões por Radiação/tratamento farmacológico , Adulto , Feminino , Fixadores , Hemorragia/tratamento farmacológico , Hemorragia/etiologia , Hemorragia/patologia , Humanos , Pessoa de Meia-Idade , Proctite/tratamento farmacológico , Proctite/etiologia , Proctite/patologia , Estudos Prospectivos , Resultado do Tratamento , Neoplasias do Colo do Útero/tratamento farmacológico
16.
Neoplasia ; 22(4): 163-178, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32143140

RESUMO

BUB1 (budding uninhibited by benzimidazoles-1) is required for efficient TGF-ß signaling, through its role in stabilizing the TGFBR1 and TGFBR2 complex. Here we demonstrate that TGFBR2 phosphorylates BUB1 at Serine-318, which is conserved in primates. S318 phosphorylation abrogates the interaction of BUB1 with TGFBR1 and SMAD2. Using BUB1 truncation domains (1-241, 241-482 and 482-723), we demonstrate that multiple contact points exist between BUB1 and TGF-ß signaling components and that these interactions are independent of the BUB1 tetratricopeptide repeat (TPR) domain. Moreover, substitutions in the middle domain (241-482) encompassing S318 reveals that efficient interaction with TGFBR2 occurs only in its dephosphorylated state (241-482 S318A). In contrast, the phospho-mimicking mutant (241-482 S318D) exhibits efficient binding with SMAD2 and its over-expression results in a decrease in TGFBR1-TGFBR2 and TGFBR1-SMAD2 interactions. These findings suggest that TGFBR2 mediated BUB1 phosphorylation at S318 may serve as a switch for the dissociation of the SMAD2-TGFBR complex, and therefore represents a regulatory event for TGF-ß signaling. Finally, we provide evidence that the BUB1-TGF-ß signaling axis may mediate aggressive phenotypes in a variety of cancers.


Assuntos
Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Serina/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Sequência de Aminoácidos , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Humanos , Modelos Biológicos , Fosforilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Serina-Treonina Quinases/química , Receptor do Fator de Crescimento Transformador beta Tipo II/química , Fator de Crescimento Transformador beta/química
17.
Cancer Res ; 67(14): 6605-11, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17638870

RESUMO

CDC25A phosphatase activates multiple cyclin-dependent kinases (CDK) during cell cycle progression. Inactivation of CDC25A by ubiquitin-mediated degradation is a major mechanism of DNA damage-induced S-G(2) checkpoint. Although increased CDC25A expression has been reported in various human cancer tissues, it remains unclear whether CDC25A activation is a critical rate-limiting step of carcinogenesis. To assess the role for CDC25A in cell cycle control and carcinogenesis, we used a Cdc25A-null mouse strain we recently generated. Whereas Cdc25A(-/-) mice exhibit early embryonic lethality, Cdc25A(+/-) mice show no appreciable developmental defect. Cdc25A(+/-) mouse embryonic fibroblasts (MEF) exhibit normal kinetics of cell cycle progression at early passages, modestly enhanced G(2) checkpoint response to DNA damage, and shortened proliferative life span, compared with wild-type MEFs. Importantly, Cdc25A(+/-) MEFs are significantly resistant to malignant transformation induced by coexpression of H-ras(V12) and a dominant negative p53 mutant. The rate-limiting role for CDC25A in transformation is further supported by decreased transformation efficiency in MCF-10A human mammary epithelial cells stably expressing CDC25A small interfering RNA. Consistently, Cdc25A(+/-) mice show substantially prolonged latency in mammary tumorigenesis induced by MMTV-H-ras or MMTV-neu transgene, whereas MMTV-myc-induced tumorigenesis is not significantly affected by Cdc25A heterozygosity. Mammary tissues of Cdc25A(+/-);MMTV-neu mice before tumor development display less proliferative response to the oncogene with increased tyrosine phosphorylation of CDK1/2, but show no significant change in apoptosis. These results suggest that Cdc25A plays a rate-limiting role in transformation and tumor initiation mediated by ras activation.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/metabolismo , Fosfatases cdc25/genética , Fosfatases cdc25/fisiologia , Proteínas ras/metabolismo , Animais , Ciclo Celular , Transformação Celular Neoplásica , Células Cultivadas , Fibroblastos/metabolismo , Fase G2 , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fase S , Fatores de Tempo
18.
Cancer Res ; 67(3): 984-91, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17283130

RESUMO

Checkpoint pathways help cells maintain genomic integrity, delaying cell cycle progression in response to various risks of fidelity, such as genotoxic stresses, compromised DNA replication, and impaired spindle control. Cancer cells frequently exhibit genomic instability, and recent studies showed that checkpoint pathways are likely to serve as a tumor-suppressive barrier in vivo. The cell cycle-promoting phosphatase CDC25A is an activator of cyclin-dependent kinases and one of the downstream targets for the CHK1-mediated checkpoint pathway. Whereas CDC25A overexpression is observed in various human cancer tissues, it has not been determined whether deregulated CDC25A expression triggers or promotes tumorigenesis in vivo. Here, we show that transgenic expression of CDC25A cooperates markedly with oncogenic ras or neu in murine mammary tumorigenesis. MMTV-CDC25A transgenic mice exhibit alveolar hyperplasia in the mammary tissue but do not develop spontaneous mammary tumors. The MMTV-CDC25A transgene markedly shortens latency of tumorigenesis in MMTV-ras mice. The MMTV-CDC25A transgene also accelerates tumor growth in MMTV-neu mice with apparent cell cycle miscoordination. CDC25A-overexpressing tumors, which invade more aggressively, exhibit various chromosomal aberrations on fragile regions, including the mouse counterpart of human 1p31-36, according to array-based comparative genomic hybridization and karyotyping. The chromosomal aberrations account for substantial changes in gene expression profile rendered by transgenic expression of CDC25A, including down-regulation of Trp73. These data indicate that deregulated control of cellular CDC25A levels leads to in vivo genomic instability, which cooperates with the neu-ras oncogenic pathway in mammary tumorigenesis.


Assuntos
Instabilidade Genômica , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/genética , Fosfatases cdc25/biossíntese , Animais , Feminino , Genes erbB-2 , Genes ras , Humanos , Hiperplasia , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Fosfatases cdc25/genética
19.
IEEE Trans Radiat Plasma Med Sci ; 3(2): 232-241, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30854500

RESUMO

The purpose of this study is to demonstrate that a Bayesian network (BN) approach can explore hierarchical biophysical relationships that influence tumor response and predict tumor local control (LC) in non-small-cell lung cancer (NSCLC) patients before and during radiotherapy from a large-scale dataset. Our BN building approach has two steps. First, relevant biophysical predictors influencing LC before and during the treatment are selected through an extended Markov blanket (eMB) method. From this eMB process, the most robust BN structure for LC prediction was found via a wrapper-based approach. Sixty-eight patients with complete feature information were used to identify a full BN model for LC prediction before and during the treatment. Fifty more recent patients with some missing information were reserved for independent testing of the developed pre- and during-therapy BNs. A nested cross-validation (N-CV) was developed to evaluate the performance of the two-step BN approach. An ensemble BN model is generated from the N-CV sampling process to assess its similarity with the corresponding full BN model, and thus evaluate the sensitivity of our BN approach. Our results show that the proposed BN development approach is a stable and robust approach to identify hierarchical relationships among biophysical features for LC prediction. Furthermore, BN predictions can be improved by incorporating during treatment information.

20.
Mol Cell Biol ; 25(8): 3338-47, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15798217

RESUMO

Ubiquitin-dependent degradation of Cdc25A is a major mechanism for damage-induced S-phase checkpoint. Two ubiquitin ligases, the Skp1-cullin-beta-TrCP (SCFbeta-TrCP) complex and the anaphase-promoting complex (APCCdh1), are involved in Cdc25A degradation. Here we demonstrate that the transforming growth factor beta (TGF-beta)-Smad3 pathway promotes SCF(beta-TrCP)-mediated Cdc25A ubiquitination. Cells treated with TGF-beta, as well as cells transfected with Smad3 or a constitutively active type I TGF-beta receptor, exhibit increased ubiquitination and markedly shortened half-lives of Cdc25A. Furthermore, Cdc25A is stabilized in cells transfected with Smad3 small interfering RNA (siRNA) and cells from Smad3-null mice. TGF-beta-induced ubiquitination is associated with Cdc25A phosphorylation at the beta-TrCP docking site (DS82G motif) and physical association of Cdc25A with Smad3 and beta-TrCP. Cdc25A mutant proteins deficient in DS82G phosphorylation are resistant to TGF-beta-Smad3-induced degradation, whereas a Cdc25A mutant protein defective in APCCdh1 recognition undergoes efficient degradation. Smad3 siRNA inhibits beta-TrCP-Cdc25A interaction and Cdc25A degradation in response to TGF-beta. beta-TrCP2 siRNA also inhibits Smad3-induced Cdc25A degradation. In contrast, Cdh1 siRNA had no effect on Cdc25A down-regulation by Smad3. These data suggest that Smad3 plays a key role in the regulation of Cdc25A ubiquitination by SCFbeta-TrCP and that Cdc25A stabilization observed in various cancers could be associated with defects in the TGF-beta-Smad3 pathway.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Neoplasias/metabolismo , Transativadores/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Proteínas Contendo Repetições de beta-Transducina/fisiologia , Fosfatases cdc25/metabolismo , Animais , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Meia-Vida , Humanos , Imunoprecipitação , Camundongos , Mutação/genética , Fosforilação , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/fisiologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteína Smad3 , Transativadores/genética , Transativadores/metabolismo , Transfecção , Fator de Crescimento Transformador beta/farmacologia , Ubiquitina/metabolismo , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Fosfatases cdc25/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa