Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Nanobiotechnology ; 22(1): 328, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38858780

RESUMO

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.


Assuntos
Membrana Externa Bacteriana , Neoplasias Ósseas , Neoplasias da Mama , Camundongos Endogâmicos BALB C , Feminino , Animais , Neoplasias da Mama/terapia , Neoplasias da Mama/patologia , Camundongos , Neoplasias Ósseas/secundário , Neoplasias Ósseas/terapia , Linhagem Celular Tumoral , Terapia por Ultrassom/métodos , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Humanos , Nanopartículas/química , Nanopartículas/uso terapêutico , Células RAW 264.7 , Membrana Celular , Nanopartículas Multifuncionais/química
2.
Small ; 19(38): e2301003, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37211708

RESUMO

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.


Assuntos
Neoplasias Ósseas , Osteólise , Animais , Camundongos , Terapia Fototérmica , Microambiente Tumoral , Osso e Ossos/patologia , Neoplasias Ósseas/terapia , Neoplasias Ósseas/patologia , Osteoclastos , Osteólise/terapia , Osteólise/patologia , Linhagem Celular Tumoral
3.
J Cell Mol Med ; 23(2): 1224-1234, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30461194

RESUMO

Bone is the most common metastatic site for breast cancer. The excessive osteoclast activity in the metastatic bone lesions often produces osteolysis. The cyclic-AMP (cAMP)-response element binding protein (CREB) serves a variety of biological functions including the transformation and immortalization of breast cancer cells. In addition, evidence has shown that CREB plays a key role in osteoclastgenesis and bone resorption. Small organic molecules with good pharmacokinetic properties and specificity, targeting CREB-CBP (CREB-binding protein) interaction to inhibit CREB-mediated gene transcription have attracted more considerations as cancer therapeutics. We recently identified naphthol AS-E (nAS-E) as a cell-permeable inhibitor of CREB-mediated gene transcription through inhibiting CREB-CBP interaction. In this study, we tested the effect of nAS-E on breast cancer cell proliferation, survival, migration as well as osteoclast formation and bone resorption in vitro for the first time. Our results demonstrated that nAS-E inhibited breast cancer cell proliferation, migration, survival and suppressed osteoclast differentiation as well as bone resorption through inhibiting CREB-CBP interaction. In addition, the in vivo effect of nAS-E in protecting against breast cancer-induced osteolysis was evaluated. Our results indicated that nAS-E could reverse bone loss induced by MDA-MB-231 tumour. These results suggest that small molecules targeting CREB-CBP interaction to inhibit CREB-mediated gene transcription might be a potential approach for the treatment of breast cancer bone metastasis.


Assuntos
Neoplasias Ósseas/prevenção & controle , Reabsorção Óssea/tratamento farmacológico , Neoplasias da Mama/prevenção & controle , Proteína de Ligação a CREB/antagonistas & inibidores , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/antagonistas & inibidores , Naftóis/farmacologia , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Animais , Apoptose , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ciclo Celular , Movimento Celular , Proliferação de Células , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Fosforilação , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Mol Pharm ; 16(7): 2872-2883, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31150251

RESUMO

Although the prognosis of patients with breast cancer continues to improve, breast cancer metastasis to bones remains high in incidence and challenging to manage. Here, we report the development of bone-homing alendronate (ALN)-anchored biodegradable polymeric micelles for the targeted treatment of metastatic cancer to bone. These micelles exhibited bone protective capacity including the recruitment, differentiation, and resorption activity of the osteoclasts. Encapsulation of docetaxel (DTX), the first-line chemotherapeutic for treatment of metastatic breast cancer, in ALN-modified micelles results in a sustained release, enhanced cytotoxicity, and improved pharmacokinetics. In the syngeneic animal model of late-stage disseminated breast cancer bone metastasis, the treatment with targeted DTX-loaded micelles attenuated the tumorigenesis and significantly improved animal lifespan compared to the conventional surfactant-based formulation (free DTX). These findings indicate potential applications of the osteotropic nanomedicines for bone metastasis treatment.


Assuntos
Alendronato/uso terapêutico , Antineoplásicos/uso terapêutico , Conservadores da Densidade Óssea/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Neoplasias da Mama/tratamento farmacológico , Docetaxel/uso terapêutico , Micelas , Polímeros , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Liberação Controlada de Fármacos , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Células RAW 264.7 , Transplante Homólogo , Resultado do Tratamento
5.
Bioeng Transl Med ; 9(1): e10597, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38193110

RESUMO

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.

6.
Theranostics ; 13(4): 1247-1263, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36923539

RESUMO

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.


Assuntos
Neoplasias Ósseas , Neoplasias Mamárias Animais , Camundongos , Animais , Proteínas Quinases Ativadas por AMP/metabolismo , Leucócitos Mononucleares/metabolismo , Proteoma , Meios de Cultivo Condicionados/farmacologia , Transdução de Sinais , Neoplasias Ósseas/tratamento farmacológico , Linhagem Celular Tumoral
7.
Aging (Albany NY) ; 15(14): 6950-6968, 2023 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-37470685

RESUMO

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.


Assuntos
Neoplasias Ósseas , Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/patologia , Microambiente Tumoral/genética , Qualidade de Vida , Neoplasias Ósseas/patologia , Células da Medula Óssea/metabolismo , Estresse Oxidativo
8.
J Mol Histol ; 53(2): 297-308, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35000027

RESUMO

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.


Assuntos
Neoplasias da Mama , MicroRNAs , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Invasividade Neoplásica/genética , Proteína Relacionada ao Hormônio Paratireóideo/genética , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo
9.
J Mol Med (Berl) ; 100(3): 373-384, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34821953

RESUMO

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.


Assuntos
Neoplasias Ósseas , Neoplasias da Mama , Osteólise , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/terapia , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Feminino , Humanos , Via de Sinalização Wnt
10.
Theranostics ; 11(11): 5143-5159, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33859739

RESUMO

Background: Advanced breast cancer metastasizes to many organs including bone, but few effective treatments are available. Here we report that induced tumor-suppressing (iTS) MSCs protected bone from metastases while un-induced MSCs did not. Methods: iTS MSCs were generated by overexpressing Lrp5, ß-catenin, Snail, or Akt. Their tumor-suppressing capability was tested using a mouse model of mammary tumors and bone metastasis, human breast cancer tissues and cancer cell lines. Results: In a mouse model, the induced MSC-derived conditioned medium (MSC CM) reduced mammary tumors and suppressed tumor-induced osteolysis. Tumor-promoting genes such as CXCL2 and LIF, as well as PDL1, a blocker of T-cell-based immune responses were downregulated. Proteomics analysis revealed that heat shock protein 90 (Hsp90ab1), calreticulin (Calr) and peptidylprolyl isomerase B (Ppib), which are highly expressed intracellular proteins in many cancers, were enriched in MSC CM as atypical tumor suppressors. Thus, overexpressing selected genes that were otherwise tumorigenic rendered MSCs the tumor-suppressing capability through the atypical suppressors, as well as p53 and Trail. Notably, the inhibitory effect of Lrp5- and Akt-overexpressing MSC CMs, Hsp90ab1 and Calr presented selective inhibition to tumor cells than non-tumor cells. The development of bone-resorbing osteoclasts was also suppressed by MSC CMs. Conclusion: Collectively, the results showed an anti-tumor effect of iTS MSCs and suggested novel therapeutic approaches to suppress the progression of tumors into the bone.


Assuntos
Neoplasias Ósseas/patologia , Osso e Ossos/patologia , Células-Tronco Mesenquimais/citologia , Animais , Neoplasias Ósseas/metabolismo , Osso e Ossos/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinogênese/metabolismo , Carcinogênese/patologia , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Meios de Cultivo Condicionados/metabolismo , Progressão da Doença , Feminino , Humanos , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células PC-3 , Células RAW 264.7 , Transdução de Sinais/fisiologia
11.
Cancers (Basel) ; 13(24)2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34944962

RESUMO

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.

12.
Theranostics ; 10(9): 4042-4055, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32226538

RESUMO

Rationale: Bone is the most frequent site for breast cancer metastasis, which accounts for the leading cause of death in advanced breast cancer patients. Serious skeletal-related events (SREs) caused by bone metastasis have a decisive impact on the life expectancy of breast cancer patients, making breast cancer almost incurable. Metastatic breast cancer cell induced pathological osteoclastogenesis is a key driver of bone metastasis and osteolytic bone lesions. We previously reported that gold clusters can prevent inflammation induced osteoclastogenesis and osteolysis in vivo. In this study, we investigated the effects of a BSA-coated gold cluster on metastatic breast cancer-induced osteoclastogenesis in vitro and tumor-induced osteolysis in vivo, and elucidated its possible mechanism. Methods: Breast cancer cell line MDA-MB-231 was used to evaluate the regulatory effects of gold clusters on breast cancer metastasis and tumor induced osteoclastogenesis in vitro. Cell counting kit-8, transwell, wound-healing and colony formation assays were performed to evaluate the effect of gold clusters on proliferation and metastasis of MDA-MB-231 cells. Tartrate-resistant acid phosphatase (TRAP) staining and filamentous-actin rings analysis were used to detect the regulatory effects of gold clusters on MDA-MB-231 cell-conditioned medium (MDA-MB-231 CM) triggered and receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclastogenesis in mouse bone marrow-derived mononuclear cells (BMMs). A mouse model of breast cancer bone metastasis was used to evaluate the in vivo activity of the gold cluster on the tumor induced osteolysis. Results: The gold clusters suppressed the migration, invasion and colony formation of MDA-MB-231 cells in a dose-dependent manner in vitro. The gold clusters strongly inhibited both MDA-MB-231 CM triggered and RANKL-induced osteoclast formation from BMMs in vitro. Cell studies indicated that the gold clusters suppressed the expression of osteolysis-related factors in MDA-MB-231 cells and inhibited the subsequent activation of NF-κB pathway in BMMs. Treatment with the clusters at a dose of 10 mg Au/kg.bw significantly reduces the breast cancer cell induced osteolysis in vivo. Conclusion: Therefore, the gold clusters may offer new therapeutic agents for preventing breast cancer bone metastasis and secondary osteolysis to improve patient outcomes.


Assuntos
Neoplasias Ósseas , Neoplasias da Mama , Ouro , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Animais , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Feminino , Ouro/administração & dosagem , Ouro/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , NF-kappa B/metabolismo
13.
Colloids Surf B Biointerfaces ; 195: 111224, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32634713

RESUMO

Metastatic breast cancer cells on arriving at bone site interact with the bone cells to influence their growth, proliferation, and chemoresistance. There are currently no effective therapeutics available in the clinic for bone metastases. Many existing anti-cancer therapeutics are ineffective at the metastatic bone site due to a lack of accurate models of breast cancer bone metastasis for drug screening. Here, we report the development of an effective in vitro model using osteogenically differentiated human mesenchymal stem cells (MSCs) and human breast cancer cells on 3D nanoclay scaffolds as a testbed for screening drugs. Our results demonstrate that breast cancer cells grown in 3D bone-mimetic scaffolds exhibited altered physiological and biochemical properties, including tumoroids formation, elevated levels of cytokine such as IL-6, and its downstream effector-mediated inhibition of apoptosis and upregulation of multidrug transporters proteins, leading to drug resistance against paclitaxel. Most importantly, Signal Transducer and Activator of Transcription 3 (STAT3), a potential biomarker for chemoresistance in many cancers, was activated in the 3D breast cancer bone metastasis model. Thus, our data suggest that 3D bone-mimetic nanoclay scaffolds-based in vitro tumor model is a promising testbed for screening new therapeutics for breast cancer bone metastasis where bone interface governs drug resistance in breast cancer cells.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Neoplasias da Mama , Resistencia a Medicamentos Antineoplásicos , Neoplasias Ósseas/secundário , Osso e Ossos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Resistência a Medicamentos , Feminino , Humanos
14.
Transl Cancer Res ; 9(2): 500-507, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35117394

RESUMO

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).

15.
ACS Biomater Sci Eng ; 6(5): 2600-2611, 2020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33463270

RESUMO

Breast cancer shows a high affinity toward bone, causing bone-related complications, leading to a poor clinical prognosis. The Wnt/ß-catenin signaling pathway has been well-documented for the bone regenerative process; however, the regulation of the Wnt/ß-catenin pathway in breast cancer bone metastasis is poorly explored. Here, we report that the Wnt/ß-catenin signaling pathway has a significant effect on osteogenesis during breast cancer bone metastasis. In this study, we have created a 3D in vitro breast cancer bone metastatic microenvironment using nanoclay-based scaffolds along with osteogenically differentiated human mesenchymal stem cells (MSCs) and human breast cancer cells (MCF-7 and MDA-MB-231). The results showed upregulation in expressions of Wnt-related factors (Wnt-5a, ß-catenin, AXIN2, and LRP5) in sequential cultures of MSCs with MCF-7 as compared to sequential cultures of MSCs with MDA-MB-231. Sequential cultures of MSCs with MCF-7 also showed higher ß-catenin expression on the protein levels than sequential cultures of MSCs with MDA-MB-231. Stimulation of Wnt/ß-catenin signaling in sequential cultures of MSCs with MCF-7 by ET-1 resulted in increased bone formation, whereas inactivation of Wnt/ß-catenin signaling by DKK-1 displayed a significant decrease in bone formation, mimicking bone lesions in breast cancer patients. These data collectively demonstrate that Wnt/ß-catenin signaling governs osteogenesis within the tumor-harboring bone microenvironment, leading to bone metastasis. The nanoclay scaffold provides a unique testbed approach for analysis of the pathways of cancer metastasis.


Assuntos
Neoplasias da Mama , Células-Tronco Mesenquimais , Diferenciação Celular , Humanos , Osteogênese , Microambiente Tumoral , Via de Sinalização Wnt
16.
Int J Biol Sci ; 15(11): 2448-2460, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31595162

RESUMO

The mTOR complex 2 (mTORC2) is recognized as a promising target for breast cancer treatment. As mTORC2-specific inhibitors do not yet exist, studies into the role of mTORC2 in cancer are performed by deleting Rictor or by RNAi-mediated Rictor silencing. The purpose of this study was to explore the effects of Rictor ablation in bone mesenchymal stromal cells (BMSCs) on bone metastasis of breast cancer. First, female mice with the genotype of Prx1-Cre;Rictorf/f (hereafter RiCKO) or Rictorf/f (as control) were injected intratibially with cells of the breast cancer cell line (TM40D) at 4 months of age. Three weeks later, osteolytic bone destruction was detected in metastatic legs by X-ray and micro-CT. We found that Rictor ablation in BMSCs inhibited TM40D-induced osteolytic bone destruction and resulted in greater bone volume maintenance in vivo. Lower CTX-I serum level, a decreased number of TRAP+ osteoclasts and lower Cathepsin-K expression observed at the tumor-bone interface indicated that osteoclastogenesis was inhibited in RiCKO mice. Additionally, co-culture experiments confirmed that Rictor deletion in BMSCs diminished osteoclast differentiation partly via down regulation of RANKL expression. Furthermore, Rictor deficiency was found to reduce the transition of BMSCs to CAFs coupled with decreased secretion of cytokines (IL-6, RANKL, TGFß), which resulted in lower chemotaxis and less proliferation in TM40D cells. These results suggest that Rictor ablation in BMSCs plays dual roles in breast cancer bone metastasis: (1) repression of osteolytic bone destruction; (2) inhibition of tumor growth.


Assuntos
Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/prevenção & controle , Células-Tronco Mesenquimais/metabolismo , Proteína Companheira de mTOR Insensível à Rapamicina/metabolismo , Animais , Neoplasias Ósseas/secundário , Neoplasias da Mama/complicações , Linhagem Celular Tumoral , Citocinas/metabolismo , Feminino , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Camundongos , Osteoclastos/metabolismo , Osteogênese/genética , Osteogênese/fisiologia , Proteína Companheira de mTOR Insensível à Rapamicina/deficiência
17.
Int J Nanomedicine ; 14: 7353-7362, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31571856

RESUMO

BACKGROUNDS: Profiting from the development of nanomaterials, photothermal therapy (PTT) has been discovered as efficient tumor ablation strategy for breast cancer. MATERIALS AND METHODS: Novel oxygen vacancy-rich tungsten bronze nanoparticles (NaxWO3) were synthesized through a simple pyrogenic decomposition process. TEM, XRD, UV-vis-NIR, photothermal conversion ability, and photothermal stability were performed. The viabilities of 293T and 4T1 cells after treating with 200 µg/mL NaxWO3 nanoparticles for 24 or 48 hrs were both above 80%, which proved the good biosafety and cytotoxicity of NaxWO3 in vitro. Two in vivo breast cancer models, namely percutaneous and intratibial 4T1 models were established and NaxWO3 (20 mg/kg) with power intensity of 1.5 W/cm2 980 nm laser photothermal treatment was used in vivo. RESULTS: We successfully synthesized ~150 nm NaxWO3 nanoparticles with desirable PTT effects, as evidenced by the temperature increase from 25.8°C to 41.8°C in 5 mins under the irradiation of 980 nm laser (1 mg/mL). Also, cellular compatibility of NaxWO3 nanoparticles was found upon physiologic 293T cells, in contrast with significant cytotoxicity against breast cancer 4T1 cell in vitro dose-dependently. Besides, two in vivo breast cancer models showed the decent tumor ablation ability of NaxWO3 nanoparticles, demonstrating percutaneous 4T1 tumor elimination without recurrence during 2 weeks observation as well as intratibial breast cancer inhibition with decreased bone destruction and tumor volume after NaxWO3+PTT in vivo. CONCLUSION: For the first time, we developed a novel oxygen vacancy-rich tungsten bronze nanoparticles (NaxWO3) through a simple pyrogenic decomposition process for PTT. Both in vitro and in vivo experiments showed the good PTT ability and tumor ablation effects of synthesized NaxWO3 nanoparticles against breast cancer osteolytic bone metastasis. Additionally, our oxygen-deficient NaxWO3 nanoparticles will expand the research horizons of PTT nanomaterials.


Assuntos
Neoplasias Ósseas/secundário , Neoplasias Ósseas/terapia , Neoplasias da Mama/patologia , Hipertermia Induzida , Nanopartículas/química , Fototerapia , Tungstênio/química , Animais , Neoplasias Ósseas/diagnóstico por imagem , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/ultraestrutura , Proteínas de Neoplasias/metabolismo , Recidiva Local de Neoplasia , Distribuição Tecidual , Microtomografia por Raio-X
18.
ACS Nano ; 13(7): 7556-7567, 2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31259530

RESUMO

Bone metastasis, a clinical complication of patients with advanced breast cancer, seriously reduces the quality of life. To avoid destruction of the bone matrix, current treatments focus on inhibiting the cancer cell growth and the osteoclast activity through combination therapy. Therefore, it could be beneficial to develop a bone-targeted drug delivery system to treat bone metastasis. Here, a bone-targeted nanoplatform was developed using gold nanorods enclosed inside mesoporous silica nanoparticles (Au@MSNs) which were then conjugated with zoledronic acid (ZOL). The nanoparticles (Au@MSNs-ZOL) not only showed bone-targeting ability in vivo but also inhibited the formation of osteoclast-like cells and promoted osteoblast differentiation in vitro. The combination of Au@MSNs-ZOL and photothermal therapy (PTT), triggered by near-infrared irradiation, inhibited tumor growth both in vitro and in vivo and relieved pain and bone resorption in vivo by inducing apoptosis in cancer cells and improving the bone microenvironment. This single nanoplatform combines ZOL and PTT to provide an exciting strategy for treating breast cancer bone metastasis.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Ósseas/secundário , Neoplasias Ósseas/terapia , Neoplasias da Mama/terapia , Nanopartículas/química , Fototerapia , Ácido Zoledrônico/farmacologia , Células 3T3-L1 , Animais , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Neoplasias Ósseas/patologia , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Camundongos , Camundongos Endogâmicos ICR , Camundongos Nus , Neoplasias Experimentais/patologia , Neoplasias Experimentais/secundário , Neoplasias Experimentais/terapia , Imagem Óptica , Tamanho da Partícula , Porosidade , Propriedades de Superfície , Células Tumorais Cultivadas , Microambiente Tumoral/efeitos dos fármacos , Ácido Zoledrônico/administração & dosagem
19.
ACS Biomater Sci Eng ; 4(2): 324-336, 2018 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-29756030

RESUMO

Biological models are necessary tools for gaining insight into underlying mechanisms governing complex pathologies such as cancer in the bone. Models range from in vitro tissue culture systems to in vivo models and can be used with corresponding epidemiological and clinical data to understand disease etiology, progression, driver mutations, and signaling pathways. In bone cancer, as with many other cancers, in vivo models are often too complex to study specific cell-cell interactions or protein roles, and 2D models are often too simple to accurately represent disease processes. Consequently, researchers have increasingly turned to 3D in vitro tissue engineered models as a useful compromise. In this review, tissue engineered 3D models of bone and cancer are described in depth and compared to 2D models. Biomaterials and cell types used are described, and future directions in the field of tissue engineered bone cancer models are proposed.

20.
Oncotarget ; 7(48): 79688-79705, 2016 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-27806311

RESUMO

Bone is one of the most common organs of breast cancer metastasis. Cancer cells that mimic osteoblasts by expressing bone matrix proteins and factors have a higher likelihood of metastasizing to bone. However, the molecular mechanisms of osteomimicry formation of cancer cells remain undefined. Herein, we identified a set of bone-related genes (BRGs) that are ectopically co-expressed in primary breast cancer tissues and determined that osteomimetic feature is obtained due to the osteoblast-like transformation of epithelial breast cancer cells that have undergone epithelial-mesenchymal transition (EMT) followed by bone morphogenetic protein-2 (BMP2) stimulation. Furthermore, we demonstrated that breast cancer cells that transformed into osteoblast-like cells with high expression of BRGs showed enhanced chemotaxis, adhesion, proliferation and multidrug resistance in an osteoblast-mimic bone microenvironment in vitro. During these processes, runt-related transcription factor 2 (RUNX2) functioned as a master mediator by suppressing or activating the transcription of BRGs that underlie the dynamic antagonism between the TGF-ß/SMAD and BMP/SMAD signaling pathways in breast cancer cells. Our findings suggest a novel mechanism of osteomimicry formation that arises in primary breast tumors, which may explain the propensity of breast cancer to metastasize to the skeleton and contribute to potential strategies for predicting and targeting breast cancer bone metastasis and multidrug resistance.


Assuntos
Mimetismo Biológico , Proteína Morfogenética Óssea 2/farmacologia , Neoplasias Ósseas/metabolismo , Neoplasias da Mama/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Antineoplásicos/farmacologia , Neoplasias Ósseas/genética , Neoplasias Ósseas/secundário , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Osteoblastos/metabolismo , Osteoblastos/patologia , Fenótipo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transcrição Gênica , Microambiente Tumoral , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa