HDACi

HDACi
  • 文章类型: Journal Article
    背景:肝细胞癌(HCC)的患病率增加仍然是全球健康挑战。肝癌化疗耐药是其治疗的临床障碍。异常的miRNA表达是癌症进展和耐药性的标志。然而,目前尚不清楚哪些miRNA与肝癌化疗耐药有关。
    方法:MicroRNA微阵列分析揭示了肝细胞癌HA22T细胞系和HDACi-R细胞系之间microRNA的差异表达谱,通过定量实时PCR(qRT-PCR)验证。探讨miR-342-5p的生物学功能及microRNA-342-5p/CFL1轴在肝癌HDACi耐药中的作用机制,在体外进行了功能丧失和功能获得研究。
    结果:在这里,我们证明了肝癌中组蛋白去乙酰化酶抑制剂(HDACi)耐药的分子机制。差异miRNA表达分析显示miR-342-5p在HDACi-R细胞中的显著下调比在亲本HA22T细胞中的显著下调。miR-342-5p的模拟物通过上调Bax增强细胞凋亡,cyto-C,HDACi-R细胞中caspase-3的表达伴随抗凋亡蛋白(Bcl-2)的下降。虽然HDACi没有增加HDACi-R的细胞活力,miR-342-5p的过表达降低了cofilin-1的表达,上调活性氧(ROS)介导的细胞凋亡,并以剂量依赖性方式使HDACi-R对HDACi敏感。
    结论:我们的研究结果表明miR-342-5p在HCC的HDACi耐药中的关键作用,并且这种机制可能归因于miR-342-5p/cofilin-1调节。
    BACKGROUND: Increased prevalence of hepatocellular carcinoma (HCC) remains a global health challenge. HCC chemoresistance is a clinical obstacle for its management. Aberrant miRNA expression is a hallmark for both cancer progression and drug resistance. However, it is unclear which miRNAs are involved in HCC chemoresistance.
    METHODS: MicroRNA microarray analysis revealed a differential expression profile of microRNAs between the hepatocellular carcinoma HA22T cell line and the HDACi-R cell line, which was validated by quantitative real-time PCR (qRT-PCR). To determine the biological function of miR-342-5p and the mechanism of the microRNA-342-5p/CFL1 axis in hepatocellular carcinoma HDACi resistance, loss- and gain-of-function studies were conducted in vitro.
    RESULTS: Here we demonstrated the molecular mechanism of histone deacetylase inhibitor (HDACi) resistance in HCC. Differential miRNA expression analysis showed significant down regulation of miR-342-5p in HDACi-R cells than in parental HA22T cells. Mimics of miR-342-5p enhanced apoptosis through upregulation of Bax, cyto-C, cleaved-caspase-3 expressions with concomitant decline in anti-apoptotic protein (Bcl-2) in HDACi-R cells. Although HDACi did not increase cell viability of HDACi-R, overexpression of miR-342-5p decreased cofilin-1 expression, upregulated reactive oxygen species (ROS) mediated apoptosis, and sensitized HDACi-R to HDACi in a dose-dependent manner.
    CONCLUSIONS: Our findings demonstrated the critical role of miR-342-5p in HDACi resistance of HCC and that this mechanism might be attributed to miR-342-5p/cofilin-1 regulation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    上皮-间充质转化(EMT)是指在特定的生理或病理条件下,极性上皮细胞向活动的间充质细胞转化,从而促进癌细胞的转移。上皮钙粘蛋白(E-cadherin)是一种在肿瘤细胞运动性的获得中起重要作用的蛋白质,并且作为关键的EMT上皮标志物。在本研究中,AW01178,一种具有潜在治疗功效的小分子化合物,通过以E-cadherin为靶标的细胞内Western高通量筛选技术进行鉴定。该化合物在mRNA和蛋白质水平上诱导E-cadherin的上调,并在体外和体内抑制乳腺癌细胞的EMT。机械上,AW01178是一种新型的苯并乙酰胺组蛋白脱乙酰酶抑制剂(HDACi),主要针对I类组蛋白脱乙酰酶。AW01178通过增强E-cadherin启动子区组蛋白H3的乙酰化水平促进E-cadherin的转录和表达,从而抑制乳腺癌细胞的转移。集体发现支持了本研究中确定的新型HDACi化合物的潜在用途,AW01178,作为乳腺癌的治疗药物,突出了其未来发展HDACi结构作为抗癌药物的价值。
    Epithelial-mesenchymal transition (EMT) refers to the transformation of polar epithelial cells into motile mesenchymal cells under specific physiological or pathological conditions, thus promoting the metastasis of cancer cells. Epithelial cadherin (E-cadherin) is a protein that plays an important role in the acquisition of tumor cell motility and serves as a key EMT epithelial marker. In the present study, AW01178, a small-molecule compound with potential therapeutic efficacy, was identified via in-cell Western high-throughput screening technology using E-cadherin as the target. The compound induced the upregulation of E-cadherin at both mRNA and protein levels and inhibited the EMT of breast cancer cells in vitro as well as metastasis in vivo. Mechanistically, AW01178 is a novel benzacetamide histone deacetylase inhibitor (HDACi) mainly targeting class I histone deacetylases. AW01178 promoted the transcription and expression of E-cadherin through enhancing the acetylation level of histone H3 in the E-cadherin promoter region, thereby inhibiting the metastasis of breast cancer cells. The collective findings support the potential utility of the novel HDACi compound identified in this study, AW01178, as a therapeutic drug for breast cancer and highlight its value for the future development of HDACi structures as anticancer drugs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:软组织肉瘤(STS)是罕见的疾病,通常由儿童和成人的结缔组织引起。然而,参与STS治疗的化疗可能会引起毒副作用和多药耐药,使治疗更具挑战性。组蛋白脱乙酰酶抑制剂(HDACi)是表观遗传剂,已显示出作为单一药物以及与其他药物联合使用的抗肿瘤作用。我们的项目旨在证明STS中的相同效果。
    方法:根据我们以前的研究,选择帕比司他(LBH589)加阿霉素进行研究。在上皮样肉瘤模型中尝试肿瘤异种移植物以验证体内良好的协同作用,并且使用平滑肌肉瘤模型作为阴性比较组。随后研究了基因谱变化。对HDACi可能引起的通路变化进行了探索和验证。
    结果:在STS细胞系和上皮样肉瘤小鼠模型中成功验证了LBH589加多柔比星的协同作用。我们试图将阿霉素的剂量降低到较低水平,发现该药物组合仍然可以抑制小鼠的肿瘤大小。此外,通过RNA测序分析研究了由LBH589引起的基因谱变化。结果表明,LBH589可以对一组靶基因发挥作用,这些靶基因可以调节潜在的生物学功能,尤其是在细胞周期通路中。
    BACKGROUND: Soft tissue sarcomas (STS) are rare diseases typically arising from connective tissues in children and adults. However, chemotherapies involved in the treatment of STS may cause toxic side effects and multi-drug chemoresistance, making the treatment even more challenging. Histone deacetylase inhibitors (HDACi) are epigenetic agents which have shown anti-tumor effects as single agent as well as combination use with other drugs. Our project intends to prove the same effects in STS.
    METHODS: Panobinostat (LBH589) plus doxorubicin was selected for investigations based on our previous research. Tumor xenografts were tried in an epithelioid sarcoma model to validate good synergy effects in vivo and a leiomyosarcoma model was used as a negative comparison group. Gene profile changes were studied afterwards. The possible pathway changes caused by HDACi were explored and validated by several assays.
    RESULTS: Synergy effect of LBH589 plus doxorubicin was successfully validated in STS cell lines and an epithelioid sarcoma mice model. We tried to reduce the dose of doxorubicin to a lower level and found the drug combination can still inhibit tumor size in mice. Furthermore, gene profile changes caused by LBH589 was studied by RNA-Sequencing analysis. Results showed LBH589 can exert effects on a group of target genes which can regulate potential biological functions especially in the cell cycle pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    焦亡,一种炎症形式的细胞死亡,促进免疫原性物质的释放并刺激免疫细胞募集,一个过程,可以把冷肿瘤变成热肿瘤。因此,在三阴性乳腺癌(TNBC)中激发焦亡是恢复抗肿瘤免疫力的可行方法。我们使用细胞计数试剂盒-8和集落形成测定分析了组蛋白脱乙酰酶抑制剂(HDACi)对TNBC细胞的作用。利用细胞凋亡和乳酸脱氢酶(LDH)释放测定来确定细胞死亡的形式。通过定量实时聚合酶链反应和westernblot验证了热解执行器。分析转录组以研究焦凋亡诱导机制。在BALB/c小鼠中产生皮下移植肿瘤模型以评估免疫细胞的浸润。HDACi显著减少细胞增殖,和变性的“气球”样细胞变得明显。HDACi导致细胞内和细胞外物质交换,以LDH的释放和碘化丙啶的摄取为标志。在Gasdermin家族中,TNBC细胞表达最大量的GSDME,和GSDMA的表达,GSDMB,HDACi治疗后GSDME增强。通过caspase3-GSDME途径的激活引起细胞凋亡,其潜在机制是细胞周期停滞和由于谷胱甘肽代谢异常而改变的细胞内REDOX平衡。体内实验表明,HDACi可以激活焦亡,限制肿瘤生长,肿瘤中CD8+淋巴细胞和CD11b+细胞浸润以及颗粒酶B的存在增加。HDACi可以在TNBC中引起焦亡,促进免疫细胞的浸润,从而增强抗癌免疫的功效。
    Pyroptosis, an inflammatory form of cell death, promotes the release of immunogenic substances and stimulates immune cell recruitment, a process, which could turn cold tumors into hot ones. Thus, instigating pyroptosis in triple-negative breast cancer (TNBC) serves as a viable method for restoring antitumor immunity. We analyzed the effects of Histone Deacetylase Inhibitors (HDACi) on TNBC cells using the Cell Counting Kit-8 and colony formation assay. Apoptosis and lactate dehydrogenase (LDH) release assays were utilized to determine the form of cell death. The pyroptotic executor was validated by quantitative real-time polymerase chain reaction and western blot. Transcriptome was analyzed to investigate pyroptosis-inducing mechanisms. A subcutaneously transplanted tumor model was generated in BALB/c mice to evaluate infiltration of immune cells. HDACi significantly diminished cell proliferation, and pyroptotic \"balloon\"-like cells became apparent. HDACi led to an intra and extracellular material exchange, signified by the release of LDH and the uptake of propidium iodide. Among the gasdermin family, TNBC cells expressed maximum quantities of GSDME, and expression of GSDMA, GSDMB, and GSDME were augmented post HDACi treatment. Pyroptosis was instigated via the activation of the caspase 3-GSDME pathway with the potential mechanisms being cell cycle arrest and altered intracellular REDOX balance due to aberrant glutathione metabolism. In vivo experiments demonstrated that HDACi can activate pyroptosis, limit tumor growth, and escalate CD8+ lymphocyte and CD11b+ cell infiltration along with an increased presence of granzyme B in tumors. HDACi can instigate pyroptosis in TNBC, promoting infiltration of immune cells and consequently intensifying the efficacy of anticancer immunity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    胚胎干细胞(ESC)可以分化成胚胎胚层的所有细胞类型。ESC还可以产生全能2C样细胞和滋养外胚层细胞。然而,由于表观遗传障碍,这些后一种转变以低频率发生,其性质尚未完全理解。这里,我们表明,用丁酸钠(NaB)处理小鼠ESCs可增加2C样细胞的数量,并可将ESCs直接重编程为滋养干细胞(TSCs),而不会转变为2C样状态.机械上,NaB抑制LSD1-HDAC1/2共阻遏复合物中的组蛋白脱乙酰酶活性。这增加了2C-和TSC-特异性基因调节区的乙酰化水平,促进他们的表达。此外,NaB处理的细胞获得产生胚泡样结构的能力,该结构可以在体外发育超过植入阶段并在体内形成蜕膜。这些结果确定了表观遗传学如何限制小鼠ESC中的全能性和滋养外胚层命运。
    Embryonic stem cells (ESCs) can differentiate into all cell types of the embryonic germ layers. ESCs can also generate totipotent 2C-like cells and trophectodermal cells. However, these latter transitions occur at low frequency due to epigenetic barriers, the nature of which is not fully understood. Here, we show that treating mouse ESCs with sodium butyrate (NaB) increases the population of 2C-like cells and enables direct reprogramming of ESCs into trophoblast stem cells (TSCs) without a transition through a 2C-like state. Mechanistically, NaB inhibits histone deacetylase activities in the LSD1-HDAC1/2 corepressor complex. This increases acetylation levels in the regulatory regions of both 2C- and TSC-specific genes, promoting their expression. In addition, NaB-treated cells acquire the capacity to generate blastocyst-like structures that can develop beyond the implantation stage in vitro and form deciduae in vivo. These results identify how epigenetics restrict the totipotent and trophectoderm fate in mouse ESCs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    化疗是最完善和有效的癌症治疗方法之一。然而,非肿瘤相关损伤限制了治疗的有效性和安全性。近几十年来,我们对癌症表观遗传学的日益理解导致了新的治疗选择和更好的患者预后的潜力。在癌症中,表观遗传变化很普遍,特别是组蛋白脱乙酰酶(HDAC)的表达和活性增加。药物是修饰DNA和染色质结构的化学试剂,其促进转录和转录后变化的破坏。第一代表观药物包括HDAC抑制剂(HDACi)(批准用于治疗血液恶性肿瘤)具有各种不良反应,要求发现和开发可能有益于癌症治疗的潜在天然HDACi,尤其是血液恶性肿瘤。姜黄素(二铁酰基甲烷),一种多酚,姜黄的成分,是一种众所周知的消炎药,抗氧化,和抗血脂药,最近被证明是一种泛HDACi。然而,姜黄中其他类姜黄素作为panHDACi的潜力仍未被开发。(i)针对人类组蛋白去乙酰化酶(HDAC)I类,虚拟筛选姜黄素和类姜黄素(去甲氧基姜黄素[DMC]和双去甲氧基姜黄素[BDMC]),II和IV酶与FDA批准的人类HDAC相比,它们的panHDAC抑制活性可在市场上购买,并且还可以(ii)预测姜黄素的药物相似性和ADME/毒性,类姜黄素和通过计算方法批准的HDACis。对人类HDACI类进行了同源建模,然后进行对接,II和IV酶与姜黄素,去甲氧基姜黄素,双去甲氧基姜黄素和5个参考HDACi化合物伏立诺他(SAHA),曲古他汀A(TSA),Chidamide,Romidepsin,和Panobinostat来了解蛋白质-配体相互作用和结合效率。Further,预测了具有低结合能的研究配体的药代动力学特性和Lipinski\的5法则。我们的研究表明,与标准HDACi(姜黄素,DMC,BDMC,Belinostat,Chidamide,Romidepsin,Panobinostat,曲古他汀A和伏立诺他)。同样,所有选择的配体分子,除了Romidepsin(折射率>130m3mol-1),遵守Lipinski的规则,没有天然化合物(姜黄素,DMC,BDMC)也报告了任何毒性和诱变性质,与化学药物相比,所有天然化合物的致死剂量(LD50)更高。由于人Zn+HDAC之间的高结合亲和力,BDMC可能是比姜黄素和DMC潜在的panHDACi。我们本研究的结果可用于设计和开发针对几种类型的癌症具有更好的HDAC抑制活性的新型化合物。此外,这些发现可以通过体外研究和临床试验得到验证,以评估天然HDACi联合标准化疗方案治疗癌症患者的生存结局.
    在线版本包含补充材料,可在10.1007/s40203-024-00221-4获得。
    Chemotherapy is one of the most well-established and effective cancer treatments available. However, non-tumor-associated damage restrict the treatment\'s effectiveness and safety. Our growing understanding of cancer epigenetics has resulted in new therapeutic options and the potential of better patient outcomes in recent decades. In cancer, epigenetic changes are widespread, particularly increased expression and activity of histone deacetylases (HDACs). Epi-drugs are chemical agents that modify the structure of DNA and chromatin facilitating disruption of transcriptional and post-transcriptional changes. First generation epi-drugs include HDAC inhibitors (HDACi) (approved to treat hematological malignancies) harbor various adverse effects demanding the discovery and development of potential natural HDACi that might benefit cancer treatment especially in hematological malignancies. Curcumin (diferuloylmethane), a polyphenolic, component of Curcuma longa, is a well-known anti-inflammatory, anti-oxidative, and anti-lipidemic agent and has recently been shown to be a pan HDACi. Yet the potential of other curcuminoids in Curcuma longa as pan HDACi remains unexplored. (i) To virtually screen curcumin and curcuminoids (Desmethoxycurcumin [DMC] & Bisdemethoxycurcumin [BDMC]) against human Histone deacetylase (HDAC) class I, II and IV enzymes in comparison to their pan HDAC inhibition activity with FDA approved human HDACis available in market and also (ii) to predict the drug likeness property and ADME/ toxicity of curcumin, curcuminoids and approved HDACis via computational approach. Homology modelling followed by docking was performed for human HDAC class I, II and IV enzymes with curcumin, Desmethoxycurcumin, Bisdemethoxycurcumin and with 5 reference HDACi compounds Vorinostat (SAHA), Trichostatin A (TSA), Chidamide, Romidepsin, and Panobinostat to understand the protein -ligand interactions and binding efficiencies. Further, the study ligands with low binding energy were predicted for pharmacokinetic properties and Lipinski\'s rule of 5. Our study revealed that BDMC followed by DMC and curcumin had high inhibitory effect by interacting at the active site of Zn+ HDACs similar to that of the standard HDACi (curcumin, DMC, BDMC, Belinostat, Chidamide, Romidepsin, Panobinostat, Trichostatin A and Vorinostat). Likewise, all of the chosen ligand molecules, with the exception of Romidepsin (refractive index > 130 m3mol-1), adhered to Lipinski\'s rule of five and none of the natural compounds (curcumin, DMC, BDMC) did report any toxicity and mutagenic property also, the lethal doses (LD50) of all the natural compounds were higher when compared to chemical drugs. BDMC could be a potential pan HDACi than curcumin and DMC owing to high binding affinity among human Zn+ HDACs. The results of our present study can be useful for the design and development of novel compounds having better HDAC inhibitory activity against several types of cancers. Moreover, these findings could be validated with invitro investigations and by clinical trials to evaluate the survival outcomes in cancer patients when treated with the natural HDACi along with standard chemo regimen.
    UNASSIGNED: The online version contains supplementary material available at 10.1007/s40203-024-00221-4.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:肝内胆管癌(ICCA)是一类异质性恶性肿瘤,其特点是复发率高,预后差。异染色质蛋白1α(HP1α)是通过异染色质形成和结构维持参与转录沉默的最重要的非组蛋白染色体蛋白之一。HP1α对ICCA进展的影响尚不清楚。
    方法:通过在两种细胞系和两种ICCA小鼠模型中的实验来检测对ICCA增殖的影响。使用电喷雾电离质谱(ESI-MS)确定HP1α与组蛋白脱乙酰酶1(HDAC1)之间的相互作用,并使用免疫沉淀测定(co-IP)研究结合机制。通过RNA测序(RNA-seq)筛选出目标基因。通过生物信息学方法预测DNA结合蛋白和组蛋白修饰的占用,并通过靶标和标签下的裂解(CUT&Tag)和染色质免疫沉淀(ChIP)进行评估。
    结果:HP1α在肝内胆管癌(ICCA)组织中上调,并通过抑制干扰素途径,以信号转导和转录激活因子1(STAT1)依赖性方式调节ICCA细胞的增殖。机械上,STAT1由HP1α-HDAC1复合物直接和表观遗传地通过启动子结合和不同组蛋白修饰的变化进行转录调节,通过高通量测序验证。广谱HDAC抑制剂(HDACi)通过下调HP1α和靶向异源二聚体激活干扰素途径并抑制ICCA细胞的增殖。发现广谱HDACi加干扰素制备方案可改善体内和体外的抗增殖作用并延迟ICCA的发展,它利用了基础激活以及干扰素途径的直接激活。HP1α参与介导对两种试剂的细胞抗性。
    结论:HP1α-HDAC1复合物通过在转录水平上直接和表观调节STAT1影响干扰素途径的激活。广谱HDACi加干扰素制剂方案抑制ICCA发展,为ICCA治疗提供可行的策略。靶向HP1α-HDAC1-STAT1轴是治疗ICCA的可能策略,尤其是HP1α阳性病例。
    BACKGROUND: Intrahepatic cholangiocarcinoma (ICCA) is a heterogeneous group of malignant tumors characterized by high recurrence rate and poor prognosis. Heterochromatin Protein 1α (HP1α) is one of the most important nonhistone chromosomal proteins involved in transcriptional silencing via heterochromatin formation and structural maintenance. The effect of HP1α on the progression of ICCA remained unclear.
    METHODS: The effect on the proliferation of ICCA was detected by experiments in two cell lines and two ICCA mouse models. The interaction between HP1α and Histone Deacetylase 1 (HDAC1) was determined using Electrospray Ionization Mass Spectrometry (ESI-MS) and the binding mechanism was studied using immunoprecipitation assays (co-IP). The target gene was screened out by RNA sequencing (RNA-seq). The occupation of DNA binding proteins and histone modifications were predicted by bioinformatic methods and evaluated by Cleavage Under Targets and Tagmentation (CUT & Tag) and Chromatin immunoprecipitation (ChIP).
    RESULTS: HP1α was upregulated in intrahepatic cholangiocarcinoma (ICCA) tissues and regulated the proliferation of ICCA cells by inhibiting the interferon pathway in a Signal Transducer and Activator of Transcription 1 (STAT1)-dependent manner. Mechanistically, STAT1 is transcriptionally regulated by the HP1α-HDAC1 complex directly and epigenetically via promoter binding and changes in different histone modifications, as validated by high-throughput sequencing. Broad-spectrum HDAC inhibitor (HDACi) activates the interferon pathway and inhibits the proliferation of ICCA cells by downregulating HP1α and targeting the heterodimer. Broad-spectrum HDACi plus interferon preparation regimen was found to improve the antiproliferative effects and delay ICCA development in vivo and in vitro, which took advantage of basal activation as well as direct activation of the interferon pathway. HP1α participates in mediating the cellular resistance to both agents.
    CONCLUSIONS: HP1α-HDAC1 complex influences interferon pathway activation by directly and epigenetically regulating STAT1 in transcriptional level. The broad-spectrum HDACi plus interferon preparation regimen inhibits ICCA development, providing feasible strategies for ICCA treatment. Targeting the HP1α-HDAC1-STAT1 axis is a possible strategy for treating ICCA, especially HP1α-positive cases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    尿路上皮癌(UC)迫切需要新的治疗选择。组蛋白脱乙酰酶(HDAC)在UC中经常失调,并构成开发替代疗法选择的有趣靶标。因此,我们研究了第二代HDAC抑制剂(HDACi)quisinostat在五种UC细胞系(UCC)和两种正常对照细胞系中的作用,一种特征良好的HDACi,以前显示可诱导细胞死亡和细胞周期停滞。在UCC中,quisinostat导致细胞周期改变,细胞死亡诱导和DNA损伤,但正常细胞耐受性良好。喹诺司他与顺铂或PARP抑制剂talazoparib的组合导致细胞活力降低,并且在五个UCC和铂抗性亚系中具有显着的协同作用,从而可以减少剂量。在低剂量比的UM-UC-3和J82中的进一步分析显示,其机制包括细胞周期紊乱,细胞凋亡诱导和DNA损伤。这些组合在正常细胞中表现出良好的耐受性。总之,我们的结果表明新的有希望的联合方案治疗UC,在顺铂耐药的情况下也是如此。
    Urothelial carcinoma (UC) urgently requires new therapeutic options. Histone deacetylases (HDAC) are frequently dysregulated in UC and constitute interesting targets for the development of alternative therapy options. Thus, we investigated the effect of the second generation HDAC inhibitor (HDACi) quisinostat in five UC cell lines (UCC) and two normal control cell lines in comparison to romidepsin, a well characterized HDACi which was previously shown to induce cell death and cell cycle arrest. In UCC, quisinostat led to cell cycle alterations, cell death induction and DNA damage, but was well tolerated by normal cells. Combinations of quisinostat with cisplatin or the PARP inhibitor talazoparib led to decrease in cell viability and significant synergistic effect in five UCCs and platinum-resistant sublines allowing dose reduction. Further analyses in UM-UC-3 and J82 at low dose ratio revealed that the mechanisms included cell cycle disturbance, apoptosis induction and DNA damage. These combinations appeared to be well tolerated in normal cells. In conclusion, our results suggest new promising combination regimes for treatment of UC, also in the cisplatin-resistant setting.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    癌症是全世界死亡的主要原因之一。癌症的病因尚未完全阐明,和进一步增强是必要的,以优化治疗效果。丁酸,短链脂肪酸,是通过肠道微生物发酵产生的膳食纤维。研究揭示了丁酸盐在恶性肿瘤中的相关性,全面了解其在癌症中的作用对于充分发挥其在肿瘤治疗中的潜力至关重要。它通过激活G蛋白偶联受体和抑制组蛋白脱乙酰酶的完全抗肿瘤作用也得到了证实。然而,潜在的机械细节仍不清楚。本研究旨在综述丁酸盐在癌变过程中的作用及其分子机制,特别强调其与肿瘤免疫疗法疗效的关系,以及讨论丁酸酯作为肿瘤疾病治疗靶点的相关临床研究,为肿瘤治疗提供新的见解。
    Cancer is one of the leading causes of mortality worldwide. The etiology of cancer has not been fully elucidated yet, and further enhancements are necessary to optimize therapeutic efficacy. Butyrate, a short‑chain fatty acid, is generated through gut microbial fermentation of dietary fiber. Studies have unveiled the relevance of butyrate in malignant neoplasms, and a comprehensive understanding of its role in cancer is imperative for realizing its full potential in oncological treatment. Its full antineoplastic effects via the activation of G protein‑coupled receptors and the inhibition of histone deacetylases have been also confirmed. However, the underlying mechanistic details remain unclear. The present study aimed to review the involvement of butyrate in carcinogenesis and its molecular mechanisms, with a particular emphasis on its association with the efficacy of tumor immunotherapy, as well as discussing relevant clinical studies on butyrate as a therapeutic target for neoplastic diseases to provide new insights into cancer treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    我们以前已经表明,异源三聚体G蛋白亚基α2(Gαi2)对于前列腺中的细胞迁移和侵袭至关重要,卵巢癌和乳腺癌细胞,和靶向Gαi2的新型小分子抑制剂阻断其对迁移和侵袭行为的影响。在这项研究中,我们已经确定了有效的,代谢稳定,第二代Gαi2抑制剂抑制前列腺癌细胞的细胞迁移。最近的研究表明,化疗可以诱导癌细胞迁移到远处部位形成转移。在本研究中,我们确定了紫杉烷(多西他赛)的作用,抗雄激素(恩扎鲁他胺和比卡鲁胺)和组蛋白去乙酰化酶(HDAC)抑制剂(SAHA和SBI-I-19)对前列腺癌细胞迁移的影响。所有治疗都诱导细胞迁移,同时用新的Gαi2抑制剂治疗阻断了它们对细胞迁移的影响。我们得出的结论是,Gαi2抑制剂和化学疗法的联合治疗可能会削弱癌细胞迁移和形成转移的能力。
    We have previously shown that heterotrimeric G-protein subunit alphai2 (Gαi2) is essential for cell migration and invasion in prostate, ovarian and breast cancer cells, and novel small molecule inhibitors targeting Gαi2 block its effects on migratory and invasive behavior. In this study, we have identified potent, metabolically stable, second generation Gαi2 inhibitors which inhibit cell migration in prostate cancer cells. Recent studies have shown that chemotherapy can induce the cancer cells to migrate to distant sites to form metastases. In the present study, we determined the effects of taxanes (docetaxel), anti-androgens (enzalutamide and bicalutamide) and histone deacetylase (HDAC) inhibitors (SAHA and SBI-I-19) on cell migration in prostate cancer cells. All treatments induced cell migration, and simultaneous treatments with new Gαi2 inhibitors blocked their effects on cell migration. We concluded that a combination treatment of Gαi2 inhibitors and chemotherapy could blunt the capability of cancer cells to migrate and form metastases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号