Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Cell ; 152(4): 714-26, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-23415222

RESUMO

Clonal evolution is a key feature of cancer progression and relapse. We studied intratumoral heterogeneity in 149 chronic lymphocytic leukemia (CLL) cases by integrating whole-exome sequence and copy number to measure the fraction of cancer cells harboring each somatic mutation. We identified driver mutations as predominantly clonal (e.g., MYD88, trisomy 12, and del(13q)) or subclonal (e.g., SF3B1 and TP53), corresponding to earlier and later events in CLL evolution. We sampled leukemia cells from 18 patients at two time points. Ten of twelve CLL cases treated with chemotherapy (but only one of six without treatment) underwent clonal evolution, predominantly involving subclones with driver mutations (e.g., SF3B1 and TP53) that expanded over time. Furthermore, presence of a subclonal driver mutation was an independent risk factor for rapid disease progression. Our study thus uncovers patterns of clonal evolution in CLL, providing insights into its stepwise transformation, and links the presence of subclones with adverse clinical outcomes.


Assuntos
Leucemia Linfocítica Crônica de Células B/genética , Mutação , Algoritmos , Animais , Linfócitos B/metabolismo , Variações do Número de Cópias de DNA , Estudo de Associação Genômica Ampla , Humanos , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Ploidias
2.
Arch Biochem Biophys ; 734: 109497, 2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-36574914

RESUMO

NFIC is a potent transcriptional factor involved in many physiological and pathological processes, including tumorigenesis. However, the role of NFIC1, the longest isoform of NFIC, in the progression of triple negative breast cancer (TNBC) remains elusive. Our study demonstrates that overexpression of NFIC1 inhibits the migration and invasion of TNBC MDA-MB-231 cells. NFIC1 regulates the expression of S100A2, and knockdown of S100A2 reverses the inhibitive effects of NFIC1 on the migration and invasion of MDA-MB-231 cells. Furthermore, knockdown of S100A2 activates the MEK/ERK signaling transduction pathway that is inhibited by NFIC1 overexperssion. Treatment with MEK/ERK pathway inhibitor, U0126, abolishes the effects of S100A2 knockdown. In addition, overexpression of NFIC1 in MDA-MB-231 cells increases the expression of epithelial markers and decreases the expression of mesenchymal markers, and these effects could also be reversed by knockdown of S100A2. Collectively, these results demonstrate that NFIC1 inhibits the Epithelial-mesenchymal transition (EMT) of MDA-MB-231 cells by regulating S100A2 expression, which suppress the activation of MEK/ERK pathway. Therefore, our study confirms the role of NFIC1 as a tumor repressor in TNBC, and reveals the molecular mechanism through which NFIC1 inhibits the migration and invasion of MDA-MB-231 cells.


Assuntos
Sistema de Sinalização das MAP Quinases , Neoplasias de Mama Triplo Negativas , Humanos , Células MDA-MB-231 , Proliferação de Células , Movimento Celular , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/farmacologia , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Fatores Quimiotáticos/metabolismo , Fatores Quimiotáticos/farmacologia , Proteínas S100/metabolismo , Proteínas S100/farmacologia
3.
Cancer Sci ; 113(2): 489-499, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34839571

RESUMO

Lysine-specific demethylase 1 (LSD1) is an important histone demethylase that mediates epithelial to mesenchymal transition (EMT). The E239K mutation of LSD1 was identified in a luminal breast cancer patient from the COSMIC Breast Cancer dataset. To investigate the functional effects of the E239K mutation of LSD1, a stable LSD1 knockdown MCF7 cell line was generated. Rescue with WT LSD1, but not E239K mutated LSD1, suppressed the invasion and migration of the LSD1 knockdown cells, indicating that the E239K mutation abolished the suppressive effects of LSD1 on the invasion and migration of MCF7 cells. Further analysis showed that the E239K mutation abolished LSD1-mediated invasion and migration of MCF7 cells through downregulation of estrogen receptor α (ERα). Most importantly, the E239K mutation disrupted the interaction between LSD1 and GATA3, which reduced the enrichment of LSD1 at the promoter region of the ERα gene; the reduced enrichment of LSD1 at the promoter region of the ERα gene caused enhanced histone H3K9 methylation, which subsequently suppressed the transcription of the ERα gene. In summary, the E239K mutation abolishes the suppressive function of LSD1 on migration and invasion of breast cancer cells by disrupting the interaction between LSD1 and GATA3.


Assuntos
Movimento Celular , Histona Desmetilases/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Adesão Celular , Transição Epitelial-Mesenquimal , Receptor alfa de Estrogênio/genética , Feminino , Fator de Transcrição GATA3/metabolismo , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases/genética , Humanos , Células MCF-7 , Camundongos , Mutação
4.
Arch Biochem Biophys ; 727: 109346, 2022 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-35798053

RESUMO

NFIC1, the longest isoform of NFIC, is essential for the regulation on spatiotemporal expression of drug-metabolizing genes in liver. However, the role of NFIC1 in breast cancer is not clear. Here we showed that increased expression of NFIC1 suppressed the migration and invasion of MCF-7 cells. NFIC1 overexpression increased the expression of IFNB1, IFNL1, IFNL2 and IFNL3, and the activation of interferon-mediated Jak-STAT pathway was enhanced by NFIC1 overexpression. Treatment with Jak-STAT pathway inhibitors, Filgotinib or Ruxolitinib, reversed the suppressive effects of NFIC1 overexpression on migration and invasion of MCF-7 cells. In addition, we found that MX1 and MX2, two target genes of Jak-STAT pathway, mediated the migration and invasion of MCF-7 cells. These results demonstrated that NFIC1 inhibited the migration and invasion in MCF-7 cells through interferon-mediated activation of Jak-STAT pathway, indicating that Jak-STAT pathway might be a potential therapeutic target for preventing breast cancer metastasis.


Assuntos
Neoplasias da Mama , Janus Quinases , Fatores de Transcrição NFI/metabolismo , Neoplasias da Mama/patologia , Movimento Celular , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Interferons/genética , Interferons/metabolismo , Interferons/farmacologia , Janus Quinases/metabolismo , Melanoma , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Neoplasias Cutâneas , Melanoma Maligno Cutâneo
5.
Biochem Biophys Res Commun ; 546: 21-28, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33561744

RESUMO

SF3B1, an essential component of the U2 snRNP, is frequently mutated in cancers. Cancer-associated SF3B1 mutation causes aberrant RNA splicing, mostly at 3' splice sites (3'ss). RNA splicing of DVL2, a regulator of Notch signaling, is affected by SF3B1 mutation. Here, we report that the mutated SF3B1 use an alternative branchpoint sequence (BPS) for the aberrant splicing of DVL2, which has a higher affinity to U2 snRNA than the BPS for the canonical splicing of DVL2. Swapping the position of the alternative BPS with the position of the canonical BPS decreased the aberrant splicing of DVL2, suggesting that the mutated SF3B1 prefers to use BPS with high affinity to U2 snRNA for splicing. Additionally, swapping the positions of two BPSs associated with the canonical splicing of DVL2 demonstrated that both the affinity to the U2 snRNA and the distance to the 3'ss are important to the selection of BPS. Importantly, the aberrant splicing of DVL2 does not require the canonical 3'ss and the canonical polypyrimidine tract, which reveals a novel type of aberrant splicing induced by SF3B1 mutation. These findings provide a more comprehensive understanding of the mechanisms underlying aberrant splicing induced by SF3B1 mutation in cancer.


Assuntos
Processamento Alternativo , Proteínas Desgrenhadas/genética , Mutação , Neoplasias/genética , Fosfoproteínas/genética , Fatores de Processamento de RNA/genética , Sequência de Bases , Proteínas Desgrenhadas/química , Humanos , Fosfoproteínas/química , Sítios de Splice de RNA/genética , Fatores de Processamento de RNA/química , RNA Nuclear Pequeno/genética
6.
Int J Mol Sci ; 20(5)2019 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-30857183

RESUMO

The aminotransferase from Bacillus circulans (BtrR), which is involved in the biosynthesis of butirosin, catalyzes the pyridoxal phosphate (PLP)-dependent transamination reaction to convert valienone to ß-valienamine (a new ß-glycosidase inhibitor for the treatment of lysosomal storage diseases) with an optical purity enantiomeric excess value. To explore the stereoselective mechanism of valienamine generated by BtrR, multiple molecular dynamics (MD) simulations were performed for the BtrR/PLP/valienamine and BtrR/PLP/ß-valienamine complexes. The theoretical results showed that ß-valienamine could make BtrR more stable and dense than valienamine. ß-valienamine could increase the hydrogen bond probability and decrease the binding free energy between coenzyme PLP and BtrR by regulating the protein structure of BtrR, which was conducive to the catalytic reaction. ß-valienamine maintained the formation of cation-p interactions between basic and aromatic amino acids in BtrR, thus enhancing its stability and catalytic activity. In addition, CAVER 3.0 analysis revealed that ß-valienamine could make the tunnel of BtrR wider and straight, which was propitious to the removal of products from BtrR. Steered MD simulation results showed that valienamine interacted with more residues in the tunnel during dissociation compared with ß-valienamine, resulting in the need for a stronger force to be acquired from BtrR. Taken together, BtrR was more inclined to catalyze the substrates to form ß-valienamine, either from the point of view of the catalytic reaction or product removal.


Assuntos
Bacillus/metabolismo , Cicloexenos/metabolismo , Hexosaminas/metabolismo , Simulação de Dinâmica Molecular , Transaminases/metabolismo , Bacillus/química , Bacillus/enzimologia , Ligação de Hidrogênio , Simulação de Acoplamento Molecular , Fosfato de Piridoxal/metabolismo , Estereoisomerismo , Especificidade por Substrato , Transaminases/química
7.
Small ; 14(21): e1800239, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29682859

RESUMO

Nanocavities composed of lipids and block polymers have demonstrated great potential in biomedical applications such as sensors, nanoreactors, and delivery vectors. However, it remains a great challenge to produce nanocavities from fluorescent semiconducting polymers owing to their hydrophobic rigid polymer backbones. Here, we describe a facile, yet general strategy that combines photocrosslinking with nanophase separation to fabricate multicolor, water-dispersible semiconducting polymer nanocavities (PNCs). A photocrosslinkable semiconducting polymer is blended with a porogen such as degradable macromolecule to form compact polymer dots (Pdots). After crosslinking the polymer and removing the porogen, this approach yields semiconducting polymer nanospheres with open cavities that are tunable in diameter. Both small molecules and macromolecules can be loaded in the nanocavities, where molecular size can be differentiated by the efficiency of the energy transfer from host polymer to guest molecules. An anticancer drug doxorubicin (Dox) is loaded into the nanocavities and the intracellular release is monitored in real time by the fluorescence signal. Finally, the efficient delivery of small interfering RNA (siRNA) to silence gene expression without affecting cell viability is demonstrated. The combined features of bright fluorescence, tunable cavity, and efficient drug/siRNA delivery makes these nanostructures promising for biomedical imaging and drug delivery.


Assuntos
Sistemas de Liberação de Medicamentos , Nanoestruturas/química , Polímeros/química , RNA Interferente Pequeno/administração & dosagem , Semicondutores , Cátions , Sobrevivência Celular , Reagentes de Ligações Cruzadas/química , Células HeLa , Humanos , Lipídeos/química , Células MCF-7 , Peso Molecular , Nanoestruturas/ultraestrutura , Polímeros/síntese química , Porosidade
8.
Int J Mol Sci ; 19(9)2018 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-30231501

RESUMO

Zearalenone hydrolase (ZHD) is the only reported α/ß-hydrolase that can detoxify zearalenone (ZEN). ZHD has demonstrated its potential as a treatment for ZEN contamination that will not result in damage to cereal crops. Recent researches have shown that the V153H mutant ZHD increased the specific activity against α-ZOL, but decreased its specific activity to ß-ZOL. To understand whyV153H mutation showed catalytic specificity for α-ZOL, four molecular dynamics simulations combining with protein network analysis for wild type ZHD α-ZOL, ZHD ß-ZOL, V153H α-ZOL, and V153H ß-ZOL complexes were performed using Gromacs software. Our theoretical results indicated that the V153H mutant could cause a conformational switch at the cap domain (residues Gly161⁻Thr190) to affect the relative position catalytic residue (H242). Protein network analysis illustrated that the V153H mutation enhanced the communication with the whole protein and residues with high betweenness in the four complexes, which were primarily assembled in the cap domain and residues Met241 to Tyr245 regions. In addition, the existence of α-ZOL binding to V153H mutation enlarged the distance from the OAE atom in α-ZOL to the NE2 atom in His242, which prompted the side chain of H242 to the position with catalytic activity, thereby increasing the activity of V153H on the α-ZOL. Furthermore, α-ZOL could easily form a right attack angle and attack distance in the ZHD and α-ZOL complex to guarantee catalytic reaction. The alanine scanning results indicated that modifications of the residues in the cap domain produced significant changes in the binding affinity for α-ZOL and ß-ZOL. Our results may provide useful theoretical evidence for the mechanism underlying the catalytic specificity of ZHD.


Assuntos
Hidrolases/metabolismo , Hypocreales/enzimologia , Zearalenona/metabolismo , Zeranol/análogos & derivados , Substituição de Aminoácidos , Hidrolases/química , Hidrolases/genética , Hypocreales/química , Hypocreales/genética , Hypocreales/metabolismo , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Mutação Puntual , Ligação Proteica , Conformação Proteica , Estabilidade Proteica , Zearalenona/química , Zeranol/química , Zeranol/metabolismo
9.
EMBO J ; 32(4): 583-96, 2013 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-23376919

RESUMO

Toll-like receptors transduce their signals through the adaptor molecule MyD88 and members of the IL-1R-associated kinase family (IRAK-1, 2, M and 4). IRAK-1 and IRAK-2, known to form Myddosomes with MyD88-IRAK-4, mediate TLR7-induced TAK1-dependent NFκB activation. IRAK-M was previously known to function as a negative regulator that prevents the dissociation of IRAKs from MyD88, thereby inhibiting downstream signalling. However, we now found that IRAK-M was also able to interact with MyD88-IRAK-4 to form IRAK-M Myddosome to mediate TLR7-induced MEKK3-dependent second wave NFκB activation, which is uncoupled from post-transcriptional regulation. As a result, the IRAK-M-dependent pathway only induced expression of genes that are not regulated at the post-transcriptional levels (including inhibitory molecules SOCS1, SHIP1, A20 and IκBα), exerting an overall inhibitory effect on inflammatory response. On the other hand, through interaction with IRAK-2, IRAK-M inhibited TLR7-mediated production of cytokines and chemokines at translational levels. Taken together, IRAK-M mediates TLR7-induced MEKK3-dependent second wave NFκB activation to produce inhibitory molecules as a negative feedback for the pathway, while exerting inhibitory effect on translational control of cytokines and chemokines.


Assuntos
Citocinas/biossíntese , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Glicoproteínas de Membrana/metabolismo , NF-kappa B/metabolismo , Receptores de Interleucina-1/metabolismo , Transdução de Sinais/fisiologia , Receptor 7 Toll-Like/metabolismo , Animais , Linhagem Celular , Citocinas/genética , Humanos , Quinases Associadas a Receptores de Interleucina-1/genética , MAP Quinase Quinase Quinase 3/genética , MAP Quinase Quinase Quinase 3/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , NF-kappa B/genética , Receptores de Interleucina-1/genética , Receptor 7 Toll-Like/genética
10.
Blood ; 121(23): 4627-34, 2013 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-23568491

RESUMO

SF3B1 is a critical component of the splicing machinery, which catalyzes the removal of introns from precursor messenger RNA (mRNA). Next-generation sequencing studies have identified mutations in SF3B1 in chronic lymphocytic leukemia (CLL) at high frequency. In CLL, SF3B1 mutation is associated with more aggressive disease and shorter survival, and recent studies suggest that it can be incorporated into prognostic schema to improve the prediction of disease progression. Mutations in SF3B1 are predominantly subclonal genetic events in CLL, and hence are likely later events in the progression of CLL. Evidence of altered pre-mRNA splicing has been detected in CLL cases with SF3B1 mutations. Although the causative link between SF3B1 mutation and CLL pathogenesis remains unclear, several lines of evidence suggest SF3B1 mutation might be linked to genomic stability and epigenetic modification.


Assuntos
Leucemia Linfocítica Crônica de Células B/genética , Mutação/genética , Fosfoproteínas/genética , Ribonucleoproteína Nuclear Pequena U2/genética , Humanos , Leucemia Linfocítica Crônica de Células B/patologia , Fatores de Processamento de RNA
11.
N Engl J Med ; 365(26): 2497-506, 2011 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-22150006

RESUMO

BACKGROUND: The somatic genetic basis of chronic lymphocytic leukemia, a common and clinically heterogeneous leukemia occurring in adults, remains poorly understood. METHODS: We obtained DNA samples from leukemia cells in 91 patients with chronic lymphocytic leukemia and performed massively parallel sequencing of 88 whole exomes and whole genomes, together with sequencing of matched germline DNA, to characterize the spectrum of somatic mutations in this disease. RESULTS: Nine genes that are mutated at significant frequencies were identified, including four with established roles in chronic lymphocytic leukemia (TP53 in 15% of patients, ATM in 9%, MYD88 in 10%, and NOTCH1 in 4%) and five with unestablished roles (SF3B1, ZMYM3, MAPK1, FBXW7, and DDX3X). SF3B1, which functions at the catalytic core of the spliceosome, was the second most frequently mutated gene (with mutations occurring in 15% of patients). SF3B1 mutations occurred primarily in tumors with deletions in chromosome 11q, which are associated with a poor prognosis in patients with chronic lymphocytic leukemia. We further discovered that tumor samples with mutations in SF3B1 had alterations in pre-messenger RNA (mRNA) splicing. CONCLUSIONS: Our study defines the landscape of somatic mutations in chronic lymphocytic leukemia and highlights pre-mRNA splicing as a critical cellular process contributing to chronic lymphocytic leukemia.


Assuntos
DNA de Neoplasias/análise , Leucemia Linfocítica Crônica de Células B/genética , Mutação , Spliceossomos/genética , Adulto , Deleção Cromossômica , Cromossomos Humanos Par 11/genética , Exoma/genética , Biblioteca Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Mutação de Sentido Incorreto , Splicing de RNA
12.
J Biochem ; 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39259498

RESUMO

Mutations in SF3B1 are common in many types of cancer, which promotes cancer progression through aberrant RNA splicing. Recently, mRNA nuclear export has been reported to be defective in cells with SF3B1 K700E mutation. However, the mechanism remains unclear. Our study reveals that the K700E mutation in SF3B1 attenuates its interaction with THOC5, an essential component of mRNA nuclear export complex THO. Furthermore, SF3B1 mutation caused reduced binding of THOC5 with some mRNA and inhibited the nuclear export of these mRNA. Interestingly, THOC5 overexpression restores the nuclear export of these mRNA in cells with SF3B1 K700E mutation. Importantly, other types of cancer-associated SF3B1 mutations also inhibited mRNA nuclear export similarly, suggesting that it is common for cancer-associated SF3B1 mutation to inhibit mRNA nuclear export. Our research highlights the critical role of the THOC5-SF3B1 interaction in the regulation of mRNA nuclear export and provides valuable insights into the impact of SF3B1 mutations on mRNA nuclear export.

13.
Cell Death Discov ; 10(1): 115, 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38448424

RESUMO

Bone metastasis is a key contributor to morbidity and mortality of breast cancer patients. We have previously shown that exosomal miRNAs derived from LSD1 knockdown (KD) breast cancer cells inhibit osteoblast differentiation and promote osteoclast differentiation. However, how LSD1 regulates exosomal miRNAs and whether miRNAs promote bone metastasis through the formation of pre-metastatic niches remains unclear. In vivo experiments demonstrates that exosomes derived from LSD1 KD breast cancer cells significantly promoted bone metastasis. To explore the mechanism underlying the effect of LSD1 on exosomes in breast cancer cells, exosomal and cellular miRNAs from control, LSD1 KD, and rescue cells were sequenced. Interestingly, approximately 80% of LSD1-associated miRNAs were downregulated in exosomes from LSD1 KD cells. The consensus sequence UAGGGC, was identified in many miRNAs downregulated in LSD1 KD exosomes. We found that hnRNPA2B1 regulated the exosomal sorting of miR-6881-3p and some other miRNAs. LSD1 deficiency reduced hnRNPA2B1 expression in breast cancer cells by decreasing the level of H3K9me2 demethylation in the promoter region of the hnRNPA2B1 gene. Our study revealed that LSD1 plays a crucial role in the regulation of exosomal sorting of miRNA.

14.
J Immunol ; 186(5): 3006-14, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21270393

RESUMO

Bone marrow-derived plasmacytoid dendritic cells (pDCs) from IL-1R-associated kinase (IRAK)2-deficient mice produced more IFNs than did wild-type pDCs upon stimulation with the TLR9 ligand CpG. Furthermore, in CpG-stimulated IRAK2-deficient pDCs there was increased nuclear translocation of IFN regulatory factor 7, the key transcription factor for IFN gene transcription in these cells. In IRAK2-deficient macrophages, enhanced NF-κB activation and increased expression of CpG-induced genes were detected within 2 h after treatment. However, at later times, NF-κB activation was decreased and, in contrast to the results with IFN, there was less secretion of other proinflammatory cytokines (such as TNF-α) and chemokines in CpG-stimulated IRAK2-deficient pDCs and macrophages. Therefore, although IRAK2 is a negative regulator of TLR9-mediated IFN production through its modulation of the transcriptional activity of IFN regulatory factor 7, it is also a positive regulator of TLR9-mediated proinflammatory cytokine and chemokine production at some level subsequent to transcription.


Assuntos
Citocinas/biossíntese , Mediadores da Inflamação/metabolismo , Interferon Tipo I/biossíntese , Interferon beta/biossíntese , Quinases Associadas a Receptores de Interleucina-1/fisiologia , Receptor Toll-Like 9/fisiologia , Animais , Células Cultivadas , Ilhas de CpG/imunologia , Citocinas/antagonistas & inibidores , Citocinas/fisiologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Regulação para Baixo/imunologia , Infecções por Herpesviridae/enzimologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/patologia , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/fisiologia , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/fisiologia , Interferon Tipo I/antagonistas & inibidores , Interferon-alfa , Interferon beta/antagonistas & inibidores , Quinases Associadas a Receptores de Interleucina-1/deficiência , Ligantes , Camundongos , Camundongos Knockout , Proteínas Recombinantes , Rhadinovirus/imunologia , Receptor Toll-Like 9/metabolismo , Infecções Tumorais por Vírus/enzimologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/patologia
15.
Front Nutr ; 10: 1144589, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36960204

RESUMO

Background: There are many causes of acute liver injury (ALI), such as alcohol, drugs, infection, and toxic materials, which have caused major health problems around the world. Among these causes, alcohol consumption induced liver injury is a common alcoholic liver disease, which can further lead to liver failure even liver cancer. A number of traditional Chinese medicine (TCM) and TCM derived compounds have been used in treating the liver-associated diseases and combination use of probiotics with TCM phytochemicals has attracted interests for enhanced biological effects. Methods: This study investigated the hepatoprotective effect of TCM-probiotics complex (TCMPC) and its underlying mechanism for the treatment of ALI in mice. The TCMPC is composed of TCM phytochemicals puerarin, curcumin, ginsenosides, and 5 lactobacteria strains. We first established a mouse model of alcohol-induced ALI, then the therapeutic effects of TCMPC on alcohol-induced ALI were monitored. A series of measurements have been performed on antioxidation, anti-inflammation, and lipid metabolism regulation. Results: The results showed that TCMPC can reduce the level of liver injury biomarkers and regulate oxidative stress. Histopathological results indicated that TCMPC could ameliorate ALI in mice. In addition, it can also significantly reduce the production of inflammatory cytokines caused by ALI. Conclusion: Our research has proved the therapeutic effect of TCMPC on alcohol-induced ALI. The potential mechanism of hepatoprotective effects of TCMPC may be related to its antioxidative and anti-inflammatory effects. Our research might provide a new way for liver disease treatment.

16.
Int J Biol Macromol ; 235: 123792, 2023 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-36828097

RESUMO

Bone metastasis is a common and incurable complication of breast cancer. Lysine-specific demethylase 1 (LSD1), a histone demethylase, plays an important role in the metastasis of breast cancer. However, the role of LSD1 in bone metastasis of breast cancer is unclear. We hypothesized that exosomes from LSD1 knockdown breast cancer cells promote bone metastasis by remodeling bone microenvironment. To verify this hypothesis, exosomes from LSD1 knockdown Estrogen receptor-positive cancer cell lines, MCF7 and T47D, were isolated, and the effects of these exosomes on osteoblast and osteoclast differentiation were investigated. Interestingly, exosomes from LSD1 knockdown breast cancer cells inhibited osteoblast differentiation and promoted osteoclast differentiation. Mechanistically, miR-6881-3p was decreased in the exosomes from LSD1 knockdown cells, and miR-6881-3p suppressed the expression of pre-B-cell leukemia homeobox 1 (PBX1) and additional sex combs like-2 (ASXL2), two genes with essential functions in osteoblast and osteoclast differentiations respectively. Transfection of miR-6881-3p into LSD1 knockdown cells reversed the effects of the exosomes on osteoblast and osteoclast differentiations. Our study reveals important roles of LSD1 on the regulation of exosomal miRNAs and the formation of favorable bone microenvironment for metastasis.


Assuntos
Neoplasias da Mama , Exossomos , MicroRNAs , Humanos , Feminino , Osteogênese/genética , Exossomos/genética , Exossomos/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , MicroRNAs/genética , MicroRNAs/metabolismo , Histona Desmetilases/genética , Microambiente Tumoral
17.
J Biochem ; 172(2): 117-126, 2022 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-35652295

RESUMO

While cancer-associated SF3B1 mutations causes alternative RNA splicing, the molecular mechanism underlying the alternative RNA splicing is not fully elucidated. Here, we analysed the proteins that interacted with the wild-type and K700E-mutated SF3B1 and found that the interactions of two RNA helicases, DDX42 and DDX46, with the mutated SF3B1 were reduced. Overexpression of DDX42 restored the decreased interaction between DDX42 and the K700E-mutated SF3B1, and suppressed some alternative RNA splicing associated with the SF3B1 mutation. Mutation that decreased the ATP hydrolysis activities of DDX42 abolished the suppressive effects of DDX42 on the alternative RNA splicing, suggesting that the ATP hydrolysis activity of DDX42 is involved in the mechanism of the altered RNA splicing associated with the SF3B1 mutation. Our study demonstrates an important function of the interaction between DDX42 and SF3B1 on regulating RNA splicing and revealed a potential role of DDX42 in the altered RNA splicing associated with the SF3B1 mutation.


Assuntos
RNA Helicases DEAD-box , Neoplasias , Fosfoproteínas , Fatores de Processamento de RNA , Ribonucleoproteína Nuclear Pequena U2 , Trifosfato de Adenosina , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Humanos , Mutação , Neoplasias/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Splicing de RNA , Fatores de Processamento de RNA/genética , Fatores de Processamento de RNA/metabolismo , Ribonucleoproteína Nuclear Pequena U2/genética , Ribonucleoproteína Nuclear Pequena U2/metabolismo
18.
Food Funct ; 13(24): 12602-12618, 2022 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-36373867

RESUMO

Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by recurrent gastrointestinal inflammation caused by abnormal immune response, and patients usually have intestinal flora imbalance. At present, the pathogenesis of UC is not well understood, and it appears that there is chronic activation of the immune and inflammatory cascade in genetically susceptible individuals. Some food supplements such as specific peptides and probiotics have been investigated and shown the potential for the treatment of UC. The purpose of this study is to investigate the therapeutic effect and potential mechanism of tetrapeptide from maize (TPM) and probiotic treatment on dextran sulfate sodium (DSS)-induced UC in C57BL/6J mice. Our results indicated that the therapeutic effects of TPM and probiotics are positively associated with a reduction in pro-inflammatory cytokine levels and restoration of the gut microbiota. Treatment with TPM or probiotics effectively alleviated the adverse effects of UC, including weight loss, shortened colon length, and colon and kidney tissue damage in mice. Additionally, both TPM and probiotics significantly reduced pro-inflammatory cytokine levels and oxidative stress in UC mice, and the effect was more pronounced when both were used together. Moreover, co-treatment with TPM and probiotics increased the diversity of gut microbes in UC mice, reduced the ratio of Firmicutes to Bacteroidetes (F/B) and increased the abundance of bacterial species, including Muribaculaceae, Alistipes, Ligilactobacillus and Lactobacillus, and has been shown to be beneficial for a variety of pathological conditions.


Assuntos
Colite Ulcerativa , Colite , Microbioma Gastrointestinal , Probióticos , Camundongos , Animais , Sulfato de Dextrana/efeitos adversos , Zea mays , Camundongos Endogâmicos C57BL , Colite/induzido quimicamente , Colite/tratamento farmacológico , Probióticos/farmacologia , Colo/microbiologia , Colite Ulcerativa/tratamento farmacológico , Citocinas , Anti-Inflamatórios/uso terapêutico , Modelos Animais de Doenças
19.
J Biochem ; 170(1): 69-77, 2021 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-33751071

RESUMO

SF3B1, an essential RNA splicing factor, is frequently mutated in various types of cancers, and the cancer-associated SF3B1 mutation causes aberrant RNA splicing. The aberrant splicing of several transcripts, including MAP3K7, promotes tumorigenesis. Here, we identify a premature termination codon in the aberrantly spliced transcript of MAP3K7. Treatment of HEK293T cells transfected with the K700E-mutated SF3B1 with cycloheximide leads to increased accumulation of the aberrant spliced transcript of MAP3K7, demonstrating that the aberrantly spliced transcript of MAP3K7 is targeted by nonsense-mediated decay. The aberrantly spliced MAP3K7 transcript uses an aberrant 3' splice sites and an alternative branchpoint sequence. In addition, the aberrant splicing of MAP3K7 requires not only the polypyrimidine tract associated with normal splicing but also an alternative polypyrimidine tract upstream of the aberrant 3' splice site. Other cancer-associated SF3B1 mutations also cause the aberrant splicing of MAP3K7, which depends on the same sequence features. Our data provide a further understanding of the mechanisms underlying aberrant splicing induced by cancer-associated SF3B1 mutation, and reveal an important role of alternative polypyrimidine tract in diseases.


Assuntos
MAP Quinase Quinase Quinases/genética , Neoplasias/genética , Fosfoproteínas/genética , Fatores de Processamento de RNA/genética , Splicing de RNA/genética , Células Cultivadas , Células HEK293 , Humanos , MAP Quinase Quinase Quinases/metabolismo , Mutação , Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Fatores de Processamento de RNA/metabolismo
20.
Int J Biol Macromol ; 142: 782-789, 2020 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-31622717

RESUMO

TRIM21 mediates the ubiquitination and proteasomal degradation of Snail, a master regulator of the epithelial to mesenchymal transition, and suppresses the migration and invasion of breast cancer cells. R64Q, a breast cancer-associated TRIM21 mutation, abolishes the interaction between TRIM21 and Snail, and the TRIM21-mediated ubiquitination and degradation of Snail. More importantly, comparing to the xenograft tumors derived from MDA-MB-231 cells with the wild-type TRIM21, xenograft tumors derived from MDA-MB-231 cells with the R64Q mutated TRIM21 showed greatly increased infiltration into neighboring muscle fibers. Furthermore, the R64Q mutation eliminates the effects of TRIM21 on the expression of genes that regulate the epithelial to mesenchymal transition. Collectively, our study demonstrates that the R64Q mutation abolishes the suppressive effects of TRIM21 on the invasion of breast cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Transição Epitelial-Mesenquimal/genética , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos Endogâmicos BALB C , Mutação , Transdução de Sinais , Ubiquitinação , Cicatrização/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA