Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Biomaterials ; 312: 122719, 2024 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-39088912

RESUMO

Acute myeloid leukemia (AML) is a deadly form of leukemia with ineffective traditional treatment and frequent chemoresistance-associated relapse. Personalized drug screening holds promise in identifying optimal regimen, nevertheless, primary AML cells undergo spontaneous apoptosis during cultures, invalidating the drug screening results. Here, we reconstitute a 3D osteogenic niche (3DON) mimicking that in bone marrow to support primary AML cell survival and phenotype maintenance in cultures. Specifically, 3DON derived from osteogenically differentiated mesenchymal stem cells (MSC) from healthy and AML donors are co-cultured with primary AML cells. The AML cells under the AML_3DON niche showed enhanced viability, reduced apoptosis and maintained CD33+ CD34-phenotype, associating with elevated secretion of anti-apoptotic cytokines in the AML_3DON niche. Moreover, AML cells under the AML_3DON niche exhibited low sensitivity to two FDA-approved chemotherapeutic drugs, further suggesting the physiological resemblance of the AML_3DON niche. Most interestingly, AML cells co-cultured with the healthy_3DON niche are highly sensitive to the same sample drugs. This study demonstrates the differential responses of AML cells towards leukemic and healthy bone marrow niches, suggesting the impact of native cancer cell niche in drug screening, and the potential of re-engineering healthy bone marrow niche in AML patients as chemotherapeutic adjuvants overcoming chemoresistance, respectively.

2.
Acta Pharm Sin B ; 12(1): 364-377, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35127392

RESUMO

Up to 70% of patients with late-stage breast cancer have bone metastasis. Current treatment regimens for breast cancer bone metastasis are palliative with no therapeutic cure. Disseminated tumor cells (DTCs) colonize inside the osteogenic niches in the early stage of bone metastasis. Drug delivery into osteogenic niches to inhibit DTC colonization can prevent bone metastasis from entering its late stage and therefore cure bone metastasis. Here, we constructed a 50% DSS6 peptide conjugated nanoparticle to target the osteogenic niche. The osteogenic niche was always located at the endosteum with immature hydroxyapatite. Arsenic-manganese nanocrystals (around 14 nm) were loaded in osteogenic niche-targeted PEG-PLGA nanoparticles with an acidic environment-triggered arsenic release. Arsenic formulations greatly reduced 4T1 cell adhesion to mesenchymal stem cells (MSCs)/preosteoblasts (pre-OBs) and osteogenic differentiation of osteoblastic cells. Arsenic formulations also prevented tumor cell colonization and dormancy via altering the direct interaction between 4T1 cells and MSCs/pre-OBs. The chemotactic migration of 4T1 cells toward osteogenic cells was blocked by arsenic in mimic 3D osteogenic niche. Systemic administration of osteogenic niche-targeted arsenic nanoparticles significantly extended the survival of mice with 4T1 syngeneic bone metastasis. Our findings provide an effective approach for osteogenic niche-specific drug delivery and suggest that bone metastasis can be effectively inhibited by blockage of tumor cell colonization in the bone microenvironment.

3.
Acta Pharmaceutica Sinica B ; (6): 364-377, 2022.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-929300

RESUMO

Up to 70% of patients with late-stage breast cancer have bone metastasis. Current treatment regimens for breast cancer bone metastasis are palliative with no therapeutic cure. Disseminated tumor cells (DTCs) colonize inside the osteogenic niches in the early stage of bone metastasis. Drug delivery into osteogenic niches to inhibit DTC colonization can prevent bone metastasis from entering its late stage and therefore cure bone metastasis. Here, we constructed a 50% DSS6 peptide conjugated nanoparticle to target the osteogenic niche. The osteogenic niche was always located at the endosteum with immature hydroxyapatite. Arsenic-manganese nanocrystals (around 14 nm) were loaded in osteogenic niche-targeted PEG-PLGA nanoparticles with an acidic environment-triggered arsenic release. Arsenic formulations greatly reduced 4T1 cell adhesion to mesenchymal stem cells (MSCs)/preosteoblasts (pre-OBs) and osteogenic differentiation of osteoblastic cells. Arsenic formulations also prevented tumor cell colonization and dormancy via altering the direct interaction between 4T1 cells and MSCs/pre-OBs. The chemotactic migration of 4T1 cells toward osteogenic cells was blocked by arsenic in mimic 3D osteogenic niche. Systemic administration of osteogenic niche-targeted arsenic nanoparticles significantly extended the survival of mice with 4T1 syngeneic bone metastasis. Our findings provide an effective approach for osteogenic niche-specific drug delivery and suggest that bone metastasis can be effectively inhibited by blockage of tumor cell colonization in the bone microenvironment.

4.
Adv Exp Med Biol ; 1329: 35-49, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34664232

RESUMO

Tumor cells frequently disseminate to distant organ sites, where they encounter permissive or restrictive environments that enable them to grow and colonize or enter a dormant state. Tumor dormancy is not strictly defined, but generally describes a tumor cell that is non-proliferative or in a state of balanced equilibrium, in which the proliferation rate of the tumor cell or cells is equal to its rate of cell death. The mechanisms that regulate tumor cell entry into and exit from dormancy are poorly understood, but microenvironmental features as well as tumor cell intrinsic factors play an important role in mediating this transition. Upon homing to distant metastatic sites, tumor cells may disseminate into various niches, most frequently the perivascular, hematopoietic stem cell, or endosteal/osteogenic niche. Tumor cells sense the cytokines, growth factors, and chemo-attractants from each of these niches, and tumor cell expression of cognate ligands and receptors can determine whether a tumor cell enters or exits dormancy. In addition to the secreted factors and cell-cell interactions that regulate dormancy, the cellular milieu also impacts upon disseminated tumor cells to promote or restrain their growth in distant metastatic sites. In this chapter we will discuss the role of the osteogenic and perivascular niche on dormant tumor cells, as well as the impact of hypoxia (low oxygen tensions) and the immune system on the restriction and outgrowth of dormant, disseminated tumor cells.


Assuntos
Neoplasias , Microambiente Tumoral , Morte Celular , Humanos , Neoplasias/genética
5.
Int J Nanomedicine ; 15: 2501-2513, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32368037

RESUMO

PURPOSE: The extracellular matrix (ECM) labyrinthine network secreted by mesenchymal stem cells (MSCs) provides a microenvironment that enhances cell adherence, proliferation, viability, and differentiation. The potential of graphene-based nanomaterials to mimic a tissue-specific ECM has been recognized in designing bone tissue engineering scaffolds. In this study, we investigated the expression of specific ECM proteins when human fat-derived adult MSCs adhered and underwent osteogenic differentiation in the presence of functionalized graphene nanoparticles. METHODS: Graphene nanoparticles with 6-10% oxygen content were prepared and characterized by XPS, FTIR, AFM and Raman spectroscopy. Calcein-am and crystal violet staining were performed to evaluate viability and proliferation of human fat-derived MSCs on graphene nanoparticles. Alizarin red staining and quantitation were used to determine the effect of graphene nanoparticles on osteogenic differentiation. Finally, immunofluorescence assays were used to investigate the expression of ECM proteins during cell adhesion and osteogenic differentiation. RESULTS: Our data show that in the presence of graphene, MSCs express specific integrin heterodimers and exhibit a distinct pattern of the corresponding bone-specific ECM proteins, primarily fibronectin, collagen I and vitronectin. Furthermore, MSCs undergo osteogenic differentiation spontaneously without any chemical induction, suggesting that the physicochemical properties of graphene nanoparticles might trigger the expression of bone-specific ECM. CONCLUSION: Understanding the cell-graphene interactions resulting in an osteogenic niche for MSCs will significantly improve the application of graphene nanoparticles in bone repair and regeneration.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Grafite/farmacologia , Células-Tronco Mesenquimais/metabolismo , Nanopartículas/química , Osteogênese/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Integrinas/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Oxigênio/química , Espectroscopia Fotoeletrônica , Multimerização Proteica
6.
Mater Sci Eng C Mater Biol Appl ; 103: 109836, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31349462

RESUMO

The surface-inspirable biomaterials have captured extensive attention in simulating microenvironments by mimicking nanoscale architectures and signal molecules in the extracellular matrix (ECM). To activate osteogenic differentiation through biomimetic nanostructures, osteoid-like calcium titanate nanorods (CT) were fabricated via anodization and hydrothermal treatment. SEM, EDS, and XPS were employed to confirm the morphology and chemical composition. The functional fibroin and peptide layers on CT (PFCT) were subsequently applied to provide binding sites and bioactive molecules for osteoblast growth and differentiation. The hydrophilicity obviously increased with the protein layers. PFCT presented enhanced cell viability with pronounced pseudopod extension and continuous proliferation. The osteogenic differentiation analysis revealed that the alkaline phosphatase expression improved in the early stage. The calcium nodes and the collagen secretion were also enhanced. Quantitative polymerase chain reaction assessment indicated that osteogenesis-related markers including ALP, Col1, and Runx2, were up-regulated. The in vitro results demonstrated that fibroin/peptide co-functionalized calcium titanate nanorods were efficient to stimulate osteoblasts and improve cell adhesion, proliferation, and differentiation. Therefore, the nanorods can create an osteogenic niche on titanium implants, which can be a promising modification strategy in bone tissue engineering application.


Assuntos
Materiais Biomiméticos/química , Cálcio/química , Diferenciação Celular , Fibroínas/química , Nanotubos/química , Osteogênese , Peptídeos/química , Nicho de Células-Tronco , Titânio/química , Animais , Linhagem Celular , Camundongos
7.
Cancer Cell ; 34(5): 823-839.e7, 2018 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-30423299

RESUMO

The fate of disseminated tumor cells is largely determined by microenvironment (ME) niche. The osteogenic niche promotes cancer cell proliferation and bone metastasis progression. We investigated the underlying mechanisms using pre-clinical models and analyses of clinical data. We discovered that the osteogenic niche serves as a calcium (Ca) reservoir for cancer cells through gap junctions. Cancer cells cannot efficiently absorb Ca from ME, but depend on osteogenic cells to increase intracellular Ca concentration. The Ca signaling, together with previously identified mammalian target of rapamycin signaling, promotes bone metastasis progression. Interestingly, effective inhibition of these pathways can be achieved by danusertib, or a combination of everolimus and arsenic trioxide, which provide possibilities of eliminating bone micrometastases using clinically established drugs.


Assuntos
Antineoplásicos/farmacologia , Trióxido de Arsênio/farmacologia , Benzamidas/farmacologia , Neoplasias Ósseas/patologia , Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Osteogênese/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , Células 3T3 , Animais , Neoplasias Ósseas/prevenção & controle , Neoplasias da Mama/terapia , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Conexina 43/metabolismo , Everolimo/farmacologia , Feminino , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Células RAW 264.7 , Microambiente Tumoral/fisiologia , Células U937
8.
Mol Cell Oncol ; 3(3): e1026526, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27314064

RESUMO

We recently discovered that bone micrometastases of breast cancer predominantly reside in the microenvironment termed the "osteogenic niche". The heterotypic adherens junctions between cancer cells and osteogenic cells promote early-stage bone colonization by activating the mTOR pathway in cancer cells. Here, we discuss a few questions raised by these findings.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...