Breast cancer bone metastasis

  • 文章类型: Journal Article
    乳腺癌骨转移的治疗是一个尚未解决的临床挑战,主要是因为目前的治疗方法不能同时阻断肿瘤生长和修复转移部位的溶骨性骨损伤。在这里,该研究开发了一种治疗乳腺癌骨转移的新型纳米药物。纳米药物基于磷酸盐离子响应性和基于过氧化钙的纳米颗粒,其表面携带骨靶向剂唑来膦酸,并负载有光敏剂吲哚菁绿。在对乳腺癌骨转移的小鼠模型进行静脉内给药后,纳米颗粒有效地积聚在骨转移部位,与游离磷酸盐离子反应,并形成羟基磷灰石纳米聚集体和O2,同时释放光敏剂。羟基磷灰石纳米聚集体引起胶原骨基质的再矿化并引发肿瘤细胞凋亡。用808nm激光源照射荷瘤腿后,O2和游离光敏剂通过纳米颗粒与磷酸根离子反应产生1O2,进一步增强抗肿瘤作用。肿瘤杀伤阻碍了骨转移部位的恶性循环,转化为骨溶解阻滞,进一步促进骨基质的再矿化。这部作品揭示了一部小说的发展,安全,治疗乳腺癌骨转移的有效方法。
    The treatment of breast cancer bone metastasis is an unresolved clinical challenge, mostly because currently therapeutic approaches cannot simultaneously block the tumor growth and repair the osteolytic bone injuries at the metastatic site. Herein, the study develops a novel nanomedicine to treat breast cancer bone metastasis. The nanomedicine is based on phosphate ion-responsive and calcium peroxide-based nanoparticles carrying the bone-targeting agent zoledronic acid on the surface and loaded with the photosensitizer indocyanine green. Following intravenous administration to a mouse model of breast cancer bone metastasis, the nanoparticles efficiently accumulate at the bone metastasis site, react with free phosphate ions, and form hydroxyapatite nanoaggregates and O2, while releasing the photosensitizer. Hydroxyapatite nanoaggregates elicit the remineralization of the collagenous bone matrix and trigger tumor cell apoptosis. Upon irradiating tumor-bearing legs with an 808 nm laser source, the O2 and free photosensitizer produced 1O2 by the reaction of the nanoparticles with phosphate ions, further boosting the anti-tumor effect. Tumor killing hampers the vicious cycle at the site of bone metastasis, translating to osteolysis blockade and further encouraging the remineralization of bone matrix. This work sheds light on the development of a novel, safe, and efficient approach for the treatment of breast cancer bone metastasis.
    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
    乳腺癌骨转移是一种晚期疾病,通常采用放疗和化疗治疗,这导致严重的副作用和有限的有效性。为了改善这一点,声动力疗法可能是未来更安全有效的方法。细菌外膜囊泡(OMV)具有优异的免疫调节特性,包括调节巨噬细胞极化,促进DC细胞成熟,增强抗肿瘤作用。将OMV与声动力疗法结合可以产生协同抗肿瘤作用。因此,我们构建了多功能纳米颗粒用于治疗乳腺癌骨转移。我们将乳腺癌细胞膜和细菌外膜囊泡融合形成杂化膜(HM),然后将负载IR780的PLGA与HM封装在一起以产生纳米颗粒,IR780@PLGA@HM,具有肿瘤靶向性,免疫调节,和声动力学能力。实验表明,IR780@PLGA@HM纳米粒子具有良好的生物相容性,有效靶向4T1肿瘤,促进巨噬细胞I型极化和DC细胞活化,抗肿瘤炎症因子表达增强,并表现出在体外和体内有效杀死肿瘤的能力,对乳腺癌骨转移有很好的治疗效果。因此,我们构建的纳米颗粒为有效治疗乳腺癌骨转移提供了新的策略。
    Breast cancer bone metastasis is a terminal-stage disease and is typically treated with radiotherapy and chemotherapy, which causes severe side effects and limited effectiveness. To improve this, Sonodynamic therapy may be a more safe and effective approach in the future. Bacterial outer membrane vesicles (OMV) have excellent immune-regulating properties, including modulating macrophage polarization, promoting DC cell maturation, and enhancing anti-tumor effects. Combining OMV with Sonodynamic therapy can result in synergetic anti-tumor effects. Therefore, we constructed multifunctional nanoparticles for treating breast cancer bone metastasis. We fused breast cancer cell membranes and bacterial outer membrane vesicles to form a hybrid membrane (HM) and then encapsulated IR780-loaded PLGA with HM to produce the nanoparticles, IR780@PLGA@HM, which had tumor targeting, immune regulating, and Sonodynamic abilities. Experiments showed that the IR780@PLGA@HM nanoparticles had good biocompatibility, effectively targeted to 4T1 tumors, promoted macrophage type I polarization and DC cells activation, strengthened anti-tumor inflammatory factors expression, and presented the ability to effectively kill tumors both in vitro and in vivo, which showed a promising therapeutic effect on breast cancer bone metastasis. Therefore, the nanoparticles we constructed provided a new strategy for effectively treating breast cancer bone metastasis.
    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
    乳腺癌是影响女性健康最常见的恶性肿瘤。骨是最常见的远处转移器官,恶化的生活质量和增加患者的死亡率。早期发现乳腺癌骨转移是阻止疾病进展和改善肿瘤预后的当务之急。最近,具有仿生组织生态位的细胞外基质(ECM)为体外肿瘤模型开辟了新途径。这里,我们开发了一种仿生脱细胞ECM(dECM)系统,以概括不同情况下的骨壁ni,来自成骨细胞的骨模拟物dECM(BM-ECM)和来自骨肉瘤细胞的骨肿瘤模拟物dECM(OS-ECM)。两种dECMs表现出不同的形态,蛋白质组成,和分配。有趣的是,高转移性乳腺癌细胞倾向于在BM-ECM上粘附和迁移,而低转移性乳腺癌细胞更喜欢OS-ECM生态位。上皮-间质转化是在不同的仿生dECMs上启动乳腺癌细胞迁移的潜在机制。在裸鼠模型中,dECM系统早在乳腺垫原位移植后10天捕获转移性乳腺癌细胞,BM-ECM上的信号高于OS-ECM上的信号。总的来说,仿生dECM系统可能是一个很有前途的肿瘤模型,可以区分乳腺癌细胞的体外转移能力,并促进体内转移乳腺癌细胞的早期检测。有助于乳腺癌骨转移的诊断。
    Breast cancer is the most prevalent malignant tumor affecting women\'s health. Bone is the most common distant metastatic organ, worsening the quality of life and increasing the mortality of patients. Early detection of breast cancer bone metastasis is urgent for halting disease progression and improving tumor prognosis. Recently, extracellular matrix (ECM) with biomimetic tissue niches opened a new avenue for tumor models in vitro. Here, we developed a biomimetic decellularized ECM (dECM) system to recapitulate bone niches at different situations, bone mimetic dECM from osteoblasts (BM-ECM) and bone tumor mimetic dECM from osteosarcoma cells (OS-ECM). The two kinds of dECMs exhibited distinct morphology, protein composition, and distribution. Interestingly, highly metastatic breast cancer cells tended to adhere and migrate on BM-ECM, while lowly metastatic breast cancer cells preferred the OS-ECM niche. Epithelial-to-mesenchymal transition was a potential mechanism to initiate the breast cancer cell migration on different biomimetic dECMs. Importantly, in the nude mice model, the dECM system captured metastatic breast cancer cells as early as 10 days after orthotopic transplantation in mammary gland pads, with higher signal on BM-ECM than that on OS-ECM. Collectively, the biomimetic dECM system might be a promising tumor model to distinguish the metastatic ability of breast cancer cells in vitro and to facilitate early detection of metastatic breast cancer cells in vivo, contributing to the diagnosis of breast cancer bone metastasis.
    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
    骨转移(BM)是晚期乳腺癌(BC)的主要表现之一。引起病理性骨折等并发症,严重影响患者的生活质量,甚至导致死亡。在我们的研究中,使用来自BM的单细胞RNA测序数据构建了肿瘤微环境的全球单细胞景观.发现BM中的BC细胞减少,间充质干细胞(MSCs),成纤维细胞和其他细胞在BM中明显更丰富。进一步鉴定了这些细胞的亚群,和路径,讨论了不同亚群的发育轨迹和转录调控。结果表明,随着BM技术的发展,BC细胞易受氧化损伤,表现出高水平的氧化应激,在细胞凋亡中起关键作用。成纤维细胞明显参与骨化和骨重建相关的生物过程(BPs),并在肿瘤细胞接种到骨髓和生长中起重要作用。MSC亚群中大量富含与骨骼生长发育和氧化应激相关的BP,可能是BC细胞归巢和粘附到BM生态位的关键成分。总之,我们的研究结果描述了乳腺癌患者肿瘤微环境细胞亚群的出现,揭示了某些细胞在BM骨重建平衡和BM发育失衡中的重要作用,并为BM提供潜在的治疗靶点。
    Bone metastasis (BM) is one of the main manifestations of advanced breast cancer (BC), causing complications such as pathological fractures, which seriously affects the quality of life of patients and even leads to death. In our study, a global single-cell landscape of the tumor microenvironment was constructed using single cell RNA sequencing data from BM. BC cells were found to be reduced in the BM, while mesenchymal stem cells (MSCs), Fibroblasts and other cells were significantly more abundant in the BM. The subpopulations of these cells were further identified, and the pathways, developmental trajectories and transcriptional regulation of different subpopulations were discussed. The results suggest that with the development of BM, BC cells were vulnerable to oxidative damage, showing a high level of oxidative stress, which played a key role in cell apoptosis. Fibroblasts were obviously involved in the biological processes (BPs) related to ossification and bone remodeling, and play an important role in tumor cell inoculation to bone marrow and growth. MSC subpopulations were significantly enriched in a number of BPs associated with bone growth and development and oxidative stress and may serve as key components of BC cells homing and adhesion to the ecological niche of BM. In conclusion, our research results describe the appearance of tumor microenvironment cell subpopulations in breast cancer patients, reveal the important role of some cells in the balance of BM bone remodeling and the imbalance of BM development, and provide potential therapeutic targets for BM.
    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
    骨是晚期乳腺癌患者易转移的部位之一。破骨细胞和乳腺癌细胞之间的“恶性循环”在乳腺癌的溶骨性骨转移中起着至关重要的作用。为了抑制乳腺癌骨转移,设计并合成了NIR-II光响应性骨靶向纳米系统(CuP@PPy-ZOLNP)。CuP@PPy-ZOLNPs可以触发光热增强的Fenton反应和光动力效应,以增强光热治疗(PTT)效果,从而达到协同抗肿瘤作用。同时,它们表现出光热增强的抑制破骨细胞分化和促进成骨细胞分化的能力,重塑了骨骼微环境。CuP@PPy-ZOLNPs能有效抑制乳腺癌三维骨转移模型中肿瘤细胞的增殖和骨吸收。在乳腺癌骨转移的小鼠模型中,CuP@PPy-ZOLNPs联合PTT与NIR-Ⅱ显著抑制乳腺癌骨转移瘤生长和骨溶解,同时促进骨修复,实现对溶骨性乳腺癌骨转移的逆转。此外,通过条件培养实验和mRNA转录组分析确定了协同治疗的潜在生物学机制。这种纳米系统的设计为治疗溶骨性骨转移提供了有希望的策略。
    Bone is one of the prone metastatic sites of patients with advanced breast cancer. The \"vicious cycle\" between osteoclasts and breast cancer cells plays an essential role in osteolytic bone metastasis from breast cancer. In order to inhibit bone metastasis from breast cancer, NIR-II photoresponsive bone-targeting nanosystems (CuP@PPy-ZOL NPs) are designed and synthesized. CuP@PPy-ZOL NPs can trigger the photothermal-enhanced Fenton response and photodynamic effect to enhance the photothermal treatment (PTT) effect and thus achieve synergistic anti-tumor effect. Meanwhile, they exhibit a photothermal enhanced ability to inhibit osteoclast differentiation and promote osteoblast differentiation, which reshaped the bone microenvironment. CuP@PPy-ZOL NPs effectively inhibited the proliferation of tumor cells and bone resorption in the in vitro 3D bone metastases model of breast cancer. In a mouse model of breast cancer bone metastasis, CuP@PPy-ZOL NPs combined with PTT with NIR-II significantly inhibited the tumor growth of breast cancer bone metastases and osteolysis while promoting bone repair to achieve the reversal of osteolytic breast cancer bone metastases. Furthermore, the potential biological mechanisms of synergistic treatment are identified by conditioned culture experiments and mRNA transcriptome analysis. The design of this nanosystem provides a promising strategy for treating osteolytic bone metastases.
    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
    背景:在发展过程中,胚胎采用不同的策略来去除不需要的细胞。使用类似于一些胚胎细胞的程序,我们从AMPK抑制的外周血单核细胞(PBMC)中的淋巴细胞和单核细胞中产生了一种消除肿瘤的条件培养基(CM).方法:AMPK信号通过应用药物抑制,Dorsomorphin,并使用体外细胞培养物评估其条件培养基(CM)的治疗效果,离体乳腺癌组织,以及乳腺肿瘤和肿瘤诱导的骨溶解的小鼠模型。使用基于质谱的蛋白质组学评估调节机制,西方印迹,免疫沉淀,基因过表达,和RNA干扰。结果:虽然AMPK信号主要起抗肿瘤作用,我们矛盾地抑制了它,以建立诱导的抑瘤细胞及其消除肿瘤的CM。在乳腺癌小鼠模型中,AMPK抑制的淋巴细胞衍生的CM的应用减少了乳腺肿瘤加化疗药物,紫杉醇.它还可以防止荷瘤胫骨的骨丢失。此外,来自患者外周血的CM的应用减少了从同一患者分离的离体乳腺癌组织。值得注意的是,富含CM的蛋白质包括Moesin(MSN),烯醇化酶1(ENO1),和polyA结合蛋白1(PABPC1),在许多类型的癌症中被认为是致瘤性的。MSN和ENO1的肿瘤抑制作用至少部分由Metadherin(Mtdh)介导,这是已知的促进转移播种。结论:我们证明PBMC可用于产生肿瘤抑制性蛋白质组,和细胞外肿瘤抑制蛋白如MSN,ENO1和PABPC1由癌细胞内的肿瘤促进因子转化。结果支持开发自体血液疗法的可能性,其中通过抑制AMPK信号传导在工程PBMC衍生的CM中富集肿瘤抑制蛋白。
    Background: During a developmental process, embryos employ varying tactics to remove unwanted cells. Using a procedure analogous to some of the embryonic cells, we generated a tumor-eliminating conditioned medium (CM) from AMPK-inhibited lymphocytes and monocytes in peripheral blood mononuclear cells (PBMCs). Methods: AMPK signaling was inhibited by the application of a pharmacological agent, Dorsomorphin, and the therapeutic effects of their conditioned medium (CM) were evaluated using in vitro cell cultures, ex vivo breast cancer tissues, and a mouse model of mammary tumors and tumor-induced osteolysis. The regulatory mechanism was evaluated using mass spectrometry-based proteomics, Western blotting, immunoprecipitation, gene overexpression, and RNA interference. Results: While AMPK signaling acted mostly anti-tumorigenic, we paradoxically inhibited it to build induced tumor-suppressing cells and their tumor-eliminating CM. In a mouse model of breast cancer, the application of AMPK-inhibited lymphocyte-derived CM reduced mammary tumors additively to a chemotherapeutic agent, Taxol. It also prevented bone loss in the tumor-bearing tibia. Furthermore, the application of CM from the patient-derived peripheral blood diminished ex vivo breast cancer tissues isolated from the same patients. Notably, proteins enriched in CM included Moesin (MSN), Enolase 1 (ENO1), and polyA-binding protein 1 (PABPC1), which are considered tumorigenic in many types of cancer. The tumor-suppressing actions of MSN and ENO1 were at least in part mediated by Metadherin (Mtdh), which is known to promote metastatic seeding. Conclusion: We demonstrated that PBMCs can be used to generate tumor-suppressive proteomes, and extracellular tumor-suppressing proteins such as MSN, ENO1, and PABPC1 are converted from tumor-promoting factors inside cancer cells. The results support the possibility of developing autologous blood-based therapy, in which tumor-suppressing proteins are enriched in engineered PBMC-derived CM by the inhibition of AMPK signaling.
    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:乳腺癌骨转移可导致严重的骨溶解和剧烈的疼痛。本研究旨在探讨坏死的存在,一种新型的程序性细胞坏死途径,在乳腺癌诱导的成骨细胞死亡中。
    方法:在本研究中,制备乳腺癌细胞条件培养基(CM),以模拟乳腺癌骨转移的微环境,并与成骨细胞共培养。然后通过细胞活力和流式细胞术检测细胞存活和死亡的百分比。采用Westernblot和PCR分别检测RIPK3,MLKL和caspase3的蛋白和mRNA表达水平。
    结果:CM可诱导成骨细胞死亡,在流式细胞术检测中,包括凋亡和坏死抑制素-1和Z-IETD-FMK可以显着降低死亡细胞的百分比。此外,CM可以显着增加裂解的caspase3,RIPK3和p-MLKL,当用Necrostatin-1(Nec-1)处理成骨细胞时,RIPK3和p-MLKL在统计学上降低。此外,三种蛋白的mRNA水平与其相应蛋白水平的变化不一致。
    结论:结论:坏死途径存在于乳腺癌细胞诱导的成骨细胞死亡途径中,并可被Necrostatin-1(Nec-1)抑制。
    BACKGROUND: Bone metastasis of breast cancer could lead to serious osteolysis and severe pain. This study is aimed to investigate the existence of necroptosis, a new type of programmed cell necrosis pathway, in breast cancer-induced osteoblast cell death.
    METHODS: In this study, conditioned medium (CM) of breast cancer cells was prepared to simulate the micro-environment of bone metastasis in breast cancer in vitro and co-cultured with osteoblast. Then the percentage of cell survival and death was detected via cell viability and flow cytometry. Western blot and PCR were taken to measure protein and mRNA expression level of RIPK 3, MLKL and caspase 3 respectively.
    RESULTS: CM could induce osteoblasts death, including apoptosis and necroptosis and necrostatin-1 plus Z-IETD-FMK could decrease the percentage of death cells significantly in the flow cytometry detection. Moreover, CM could increase cleaved caspase 3, RIPK 3 and p-MLKL significantly, while RIPK 3 and p-MLKL was reduced statistically when osteoblasts were treated with Necrostatin-1 (Nec-1). In addition, the mRNA level of three proteins was not consistent with the change of their corresponding protein level.
    CONCLUSIONS: In conclusion, the necroptosis pathway exists in osteoblast cell death pathway induced by breast cancer cells and could be inhibited by Necrostatin-1 (Nec-1).
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    乳腺癌骨转移可能会阻碍正常的骨重建并促进骨降解,在此期间,一些信号通路和小的非编码miRNA可能都起作用。使用基于在线数据的生物信息学分析来分析和选择可能与乳腺癌骨转移相关的miRNA和靶mRNA。使用Targetscan检查与乳腺癌转移相关的关键因子的3'非翻译区的候选miRNA结合位点。验证了预测的结合。研究了单个miRNA的特异性作用和miRNA-mRNA轴对乳腺癌细胞转移的动态影响。根据在线数据集和实验分析,miR-556-5p在乳腺癌样品中下调。在乳腺癌细胞中,miR-556-5p过表达抑制,而miR-556-5p抑制促进癌细胞侵袭和迁移。在与乳腺癌骨转移相关的关键因素中,甲状旁腺激素相关蛋白(PTHrP)3UTR具有miR-556-5p结合位点。通过直接绑定,miR-556-5p负调控PTHrP表达。在乳腺癌细胞系中,miR-556-5p抑制促进,而PTHrP沉默抑制癌细胞迁移,入侵,和上皮-间质转化;PTHrP沉默部分逆转了miR-556-5p的抑制作用。总之,miR-556-5p靶向PTHrP调节乳腺癌细胞迁移和侵袭。
    Breast cancer bone metastases may block normal bone remodeling and promote bone degradation, during which several signaling pathways and small non-coding miRNAs might all play a role. miRNAs and target mRNAs that might be associated with breast cancer bone metastasis were analyzed and selected using bioinformatics analyses based on online data. The 3\' untranslated region of key factors associated with breast cancer metastasis were examined for candidate miRNA binding site using Targetscan. The predicted binding was validated. The specific effects of single miRNA and dynamic effects of the miRNA-mRNA axis on breast cancer cell metastasis were investigated. miR-556-5p was downregulated in breast cancer samples according to online datasets and experimental analyses. In breast cancer cells, miR-556-5p overexpression inhibited, whereas miR-556-5p inhibition promoted cancer cell invasion and migration. Among key factors associated with breast cancer bone metastasis, parathyroid hormone related protein (PTHrP) 3\'UTR possessed miR-556-5p binding site. Through direct binding, miR-556-5p negatively regulated PTHrP expression. In breast cancer cell lines, miR-556-5p inhibition promoted, whereas PTHrP silencing suppressed cancer cell migration, invasion, and epithelial-mesenchymal transition; the effects of miR-556-5p inhibition were partially reversed by PTHrP silencing. In summary, miR-556-5p targets PTHrP to modulate the cell migration and invasion of breast cancer.
    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
    Osteocytes, the most abundant bone cell types embedded in the mineral matrix, express connexin 43 (Cx43) hemichannels that play important roles in bone remodeling and osteocyte survival. Estrogen deficiency decreases osteocytic Cx43 hemichannel activity and causes a loss in osteocytes\' resistance to oxidative stress (OS). In this study, we showed that OS reduced the growth of both human (MDA-MB-231) and murine (Py8119) breast cancer cells. However, co-culturing these cells with osteocytes reduced the inhibitory effect of OS on breast cancer cells, and this effect was ablated by the inhibition of Cx43 hemichannels. Py8119 cells were intratibially implanted in the bone marrow of ovariectomized (OVX) mice to determine the role of osteocytic Cx43 hemichannels in breast cancer bone metastasis in response to OS. Two transgenic mice overexpressing dominant-negative Cx43 mutants, R76W and Δ130-136, were adopted for this study; the former inhibits gap junctions while the latter inhibits gap junctions and hemichannels. Under normal conditions, Δ130-136 mice had significantly more tumor growth in bone than that in WT and R76W mice. OVX increased tumor growth in R76W but had no significant effect on WT mice. In contrast, OVX reduced tumor growth in Δ130-136 mice. To confirm the role of OS, WT and Δ130-136 mice were administered the antioxidant N-acetyl cysteine (NAC). NAC increased tumor burden and growth in Δ130-136 mice but not in WT mice. Together, the data suggest that osteocytes and Cx43 hemichannels play pivotal roles in modulating the oxidative microenvironment and breast cancer growth in the bone.
    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
    Osteolytic bone destruction is found in approximately 60% of advanced breast cancer patients. With the pathogenesis of bone metastasis being unclear, traditional antiresorptive therapeutic strategies might not be ideal for treatment. The Wnt pathway is a highly organized cascade involved in multiple stages of cancer bone metastasis, and Wnt-targeted therapeutic strategies have shown promise in achieving favorable outcomes. In this review, we summarize the current progress of pharmacological Wnt modulators against breast cancer bone metastasis, discuss emerging therapeutic strategies based on Wnt pathway-related targets for bone therapy, and highlight opportunities to better harness the Wnt pathway for bone metastasis therapeutics to further reveal the implications of the Wnt pathway in bone metastasis pathology and provide new ideas for the development of Wnt-based intervention strategies against breast cancer bone metastasis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号