Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Adv Sci (Weinh) ; 11(13): e2305212, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38263873

RESUMO

Platinum (Pt)-based chemotherapy is the main treatment for ovarian cancer (OC); however, most patients develop Pt resistance (Pt-R). This work shows that Pt-R OC cells increase intracellular cholesterol through uptake via the HDL receptor, scavenger receptor type B-1 (SR-B1). SR-B1 blockade using synthetic cholesterol-poor HDL-like nanoparticles (HDL NPs) diminished cholesterol uptake leading to cell death and inhibition of tumor growth. Reduced cholesterol accumulation in cancer cells induces lipid oxidative stress through the reduction of glutathione peroxidase 4 (GPx4) leading to ferroptosis. In turn, GPx4 depletion induces decreased cholesterol uptake through SR-B1 and re-sensitizes OC cells to Pt. Mechanistically, GPx4 knockdown causes lower expression of the histone acetyltransferase EP300, leading to reduced deposition of histone H3 lysine 27 acetylation (H3K27Ac) on the sterol regulatory element binding transcription factor 2 (SREBF2) promoter and suppressing expression of this key transcription factor involved in the regulation of cholesterol metabolism. SREBF2 downregulation leads to decreased SR-B1 expression and diminished cholesterol uptake. Thus, chemoresistance and cancer cell survival under high ROS burden obligates high GPx4 and SR-B1 expression through SREBF2. Targeting SR-B1 to modulate cholesterol uptake inhibits this axis and causes ferroptosis in vitro and in vivo in Pt-R OC.


Assuntos
Nanopartículas , Neoplasias Ovarianas , Humanos , Feminino , Receptores Depuradores Classe B/metabolismo , Colesterol/metabolismo , Fatores de Transcrição/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Oxirredução
2.
J Biol Chem ; 296: 100100, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33208460

RESUMO

Normal human cells can either synthesize cholesterol or take it up from lipoproteins to meet their metabolic requirements. In some malignant cells, de novo cholesterol synthesis genes are transcriptionally silent or mutated, meaning that cholesterol uptake from lipoproteins is required for survival. Recent data suggest that lymphoma cells dependent upon lipoprotein-mediated cholesterol uptake are also subject to ferroptosis, an oxygen- and iron-dependent cell death mechanism triggered by accumulation of oxidized lipids in cell membranes unless the lipid hydroperoxidase, glutathione peroxidase 4 (GPX4), reduces these toxic lipid species. To study mechanisms linking cholesterol uptake with ferroptosis and determine the potential role of the high-density lipoprotein (HDL) receptor as a target for cholesterol depleting therapy, we treated lymphoma cell lines known to be sensitive to the reduction of cholesterol uptake with HDL-like nanoparticles (HDL NPs). HDL NPs are a cholesterol-poor ligand that binds to the receptor for cholesterol-rich HDLs, scavenger receptor type B1 (SCARB1). Our data reveal that HDL NP treatment activates a compensatory metabolic response in treated cells toward increased de novo cholesterol synthesis, which is accompanied by nearly complete reduction in expression of GPX4. As a result, oxidized membrane lipids accumulate, leading to cell death through a mechanism consistent with ferroptosis. We obtained similar results in vivo after systemic administration of HDL NPs in mouse lymphoma xenografts and in primary samples obtained from patients with lymphoma. In summary, targeting SCARB1 with HDL NPs in cholesterol uptake-addicted lymphoma cells abolishes GPX4, resulting in cancer cell death by a mechanism consistent with ferroptosis.


Assuntos
Colesterol/metabolismo , Ferroptose , Linfoma/metabolismo , Animais , Colesterol/genética , Humanos , Células Jurkat , Linfoma/genética , Linfoma/patologia , Camundongos , Camundongos SCID , Proteínas de Neoplasias/metabolismo , Oxirredução , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Receptores Depuradores Classe B/genética , Receptores Depuradores Classe B/metabolismo , Células U937
3.
J Extracell Vesicles ; 10(2): e12042, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33408816

RESUMO

Primary tumours can establish long-range communication with distant organs to transform them into fertile soil for circulating tumour cells to implant and proliferate, a process called pre-metastatic niche (PMN) formation. Tumour-derived extracellular vesicles (EV) are potent mediators of PMN formation due to their diverse complement of pro-malignant molecular cargo and their propensity to target specific cell types (Costa-Silva et al., 2015; Hoshino et al., 2015; Peinado et al., 2012; Peinado et al., 2017). While significant progress has been made to understand the mechanisms by which pro-metastatic EVs create tumour-favouring microenvironments at pre-metastatic organ sites, comparatively little attention has been paid to the factors intrinsic to recipient cells that may modify the extent to which pro-metastatic EV signalling is received and transduced. Here, we investigated the role of recipient cell cholesterol homeostasis in prostate cancer (PCa) EV-mediated signalling and metastasis. Using a bone metastatic model of enzalutamide-resistant PCa, we first characterized an axis of EV-mediated communication between PCa cells and bone marrow that is marked by in vitro and in vivo PCa EV uptake by bone marrow myeloid cells, activation of NF-κB signalling, enhanced osteoclast differentiation, and reduced myeloid thrombospondin-1 expression. We then employed a targeted, biomimetic approach to reduce myeloid cell cholesterol in vitro and in vivo prior to conditioning with PCa EVs. Reducing myeloid cell cholesterol prevented the uptake of PCa EVs by recipient myeloid cells, abolished NF-κB activity and osteoclast differentiation, stabilized thrombospondin-1 expression, and reduced metastatic burden by 77%. These results demonstrate that cholesterol homeostasis in bone marrow myeloid cells regulates pro-metastatic EV signalling and metastasis by acting as a gatekeeper for EV signal transduction.


Assuntos
Biomarcadores Tumorais/metabolismo , Células da Medula Óssea/patologia , Neoplasias Ósseas/secundário , Comunicação Celular , Colesterol/metabolismo , Vesículas Extracelulares/patologia , Neoplasias da Próstata/patologia , Animais , Apoptose , Biomarcadores Tumorais/genética , Células da Medula Óssea/metabolismo , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Proliferação de Células , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Perfilação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
4.
ACS Nano ; 13(9): 10301-10311, 2019 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31487458

RESUMO

B-cell lymphoma cells depend upon cholesterol to maintain pro-proliferation and pro-survival signaling via the B-cell receptor. Targeted cholesterol depletion of lymphoma cells is an attractive therapeutic strategy. We report here high-density lipoprotein mimicking magnetic nanostructures (HDL-MNSs) that can bind to the high-affinity HDL receptor, scavenger receptor type B1 (SR-B1), and interfere with cholesterol flux mechanisms in SR-B1 receptor positive lymphoma cells, causing cellular cholesterol depletion. In addition, the MNS core can be utilized for its ability to generate heat under an external radio frequency field. The thermal activation of MNS can lead to both innate and adaptive antitumor immune responses by inducing the expression of heat shock proteins that lead to activation of antigen presenting cells and finally lymphocyte trafficking. In the present study, we demonstrate SR-B1 receptor mediated binding and cellular uptake of HDL-MNS and prevention of phagolysosome formation by transmission electron microscopy, fluorescence microscopy, and ICP-MS analysis. The combinational therapeutics of cholesterol depletion and thermal activation significantly improves therapeutic efficacy in SR-B1 expressing lymphoma cells. HDL-MNS reduces the T2 relaxation time under magnetic resonance imaging (MRI) more effectively compared with a commercially available contrast agent, and the specificity of HDL-MNS toward the SR-B1 receptor leads to differential contrast between SR-B1 positive and negative cells suggesting its utility in diagnostic imaging. Overall, we have demonstrated that HDL-MNSs have cell specific targeting efficiency, can modulate cholesterol efflux, can induce thermal activation mediated antitumor immune response, and possess high contrast under MRI, making it a promising theranostic platform in lymphoma.


Assuntos
Biomimética , Metabolismo dos Lipídeos , Linfoma de Células B/imunologia , Fenômenos Magnéticos , Nanoestruturas/química , Nanomedicina Teranóstica , Animais , Linhagem Celular Tumoral , Endocitose , Imunomodulação , Ferro/metabolismo , Lipoproteínas HDL/química , Camundongos , Microscopia de Fluorescência , Nanoestruturas/ultraestrutura , Receptores Depuradores Classe B/metabolismo , Temperatura
5.
J Am Chem Soc ; 141(25): 9753-9757, 2019 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-31177775

RESUMO

Synthetic high-density lipoprotein (HDL) mimics have emerged as promising therapeutic agents. However, approaches to date have been unable to reproduce key features of spherical HDLs, which are the most abundant human HDL species. Here, we report the synthesis and characterization of spherical HDL mimics using lipid-conjugated organic core scaffolds. The core design motif constrains and orients phospholipid geometry to facilitate the assembly of soft-core nanoparticles that are approximately 10 nm in diameter and resemble human HDLs in their size, shape, surface chemistry, composition, and protein secondary structure. These particles execute salient HDL functions, including efflux of cholesterol from macrophages, cholesterol delivery to hepatocytes, support lecithin:cholesterol acyltransferase activity, and suppress inflammation. These results represent a significant step toward a genuine functional mimic of human HDLs.


Assuntos
1,2-Dipalmitoilfosfatidilcolina/química , Materiais Biomiméticos/química , Portadores de Fármacos/química , Nanopartículas Metálicas/química , Fosfatidiletanolaminas/química , Materiais Biomiméticos/síntese química , Colesterol/metabolismo , DNA/química , Portadores de Fármacos/síntese química , Ouro/química , Células Hep G2 , Humanos , Inflamação/tratamento farmacológico , Lipoproteínas HDL/química , Lipossomos/química , Monócitos/metabolismo , Subunidade p50 de NF-kappa B/metabolismo
6.
Expert Rev Anticancer Ther ; 19(6): 515-528, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31148521

RESUMO

Introduction: Significant clinical correlations have been observed between serum high-density lipoprotein (HDL) cholesterol and cancer risk, outcomes, and patient response to specific treatments. While the biological processes underlying these correlations remain unclear, evidence suggests that HDLs actively inhibit tumor progression through a variety of mechanisms. As a result, synthetic HDLs have emerged as attractive agents for targeted cancer therapy. Areas covered: We present a focused review of recent developments in the use of synthetic HDLs for cancer therapy, including roles in drug delivery, RNAi, monotherapy, and immunotherapy. In addition to historic references relevant to the field, we searched the following databases for recent articles published from January 1st, 2015 - May 1st, 2019: MEDLINE, Web of Science Core Collection, and Google Scholar. Expert opinion: Synthetic HDLs have already been used in human patients for cardiovascular disease, and have proven to be effective anticancer agents in pre-clinical testing, which should pave the way for future clinical trials in the setting of cancer. Given the growing notoriety of dysregulated cholesterol homeostasis as a key mechanism of cancer progression, and the immense success of synthetic HDLs in animal models, synthetic HDLs are well-poised to make significant strides toward the clinic as cancer therapy.


Assuntos
Lipoproteínas HDL/metabolismo , Nanopartículas , Neoplasias/terapia , Animais , Antineoplásicos/administração & dosagem , Progressão da Doença , Sistemas de Liberação de Medicamentos , Humanos , Imunoterapia/métodos , Neoplasias/patologia , Interferência de RNA
7.
Oncoscience ; 5(5-6): 164-166, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-30035178
8.
EMBO Rep ; 19(3)2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29440125

RESUMO

Trinucleotide repeat (TNR) expansions in the genome cause a number of degenerative diseases. A prominent TNR expansion involves the triplet CAG in the huntingtin (HTT) gene responsible for Huntington's disease (HD). Pathology is caused by protein and RNA generated from the TNR regions including small siRNA-sized repeat fragments. An inverse correlation between the length of the repeats in HTT and cancer incidence has been reported for HD patients. We now show that siRNAs based on the CAG TNR are toxic to cancer cells by targeting genes that contain long reverse complementary TNRs in their open reading frames. Of the 60 siRNAs based on the different TNRs, the six members in the CAG/CUG family of related TNRs are the most toxic to both human and mouse cancer cells. siCAG/CUG TNR-based siRNAs induce cell death in vitro in all tested cancer cell lines and slow down tumor growth in a preclinical mouse model of ovarian cancer with no signs of toxicity to the mice. We propose to explore TNR-based siRNAs as a novel form of anticancer reagents.


Assuntos
Proteína Huntingtina/genética , Neoplasias/genética , RNA Interferente Pequeno/farmacologia , Repetições de Trinucleotídeos/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Modelos Animais de Doenças , Humanos , Proteína Huntingtina/antagonistas & inibidores , Doença de Huntington/genética , Doença de Huntington/patologia , Camundongos , Neoplasias/patologia , Neoplasias/terapia , Fases de Leitura Aberta , RNA Interferente Pequeno/genética , Expansão das Repetições de Trinucleotídeos/genética , Repetições de Trinucleotídeos/efeitos dos fármacos
9.
Sci Rep ; 8(1): 1211, 2018 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-29352211

RESUMO

Medulloblastoma is the most common paediatric malignant brain cancer and there is a need for new targeted therapeutic approaches to more effectively treat these malignant tumours, which can be divided into four molecular subtypes. Here, we focus on targeting sonic hedgehog (SHH) subtype medulloblastoma, which accounts for approximately 25% of all cases. The SHH subtype relies upon cholesterol signalling for tumour growth and maintenance of tumour-initiating cancer stem cells (CSCs). To target cholesterol signalling, we employed biomimetic high-density lipoprotein nanoparticles (HDL NPs) which bind to the HDL receptor, scavenger receptor type B-1 (SCARB1), depriving cells of natural HDL and their cholesterol cargo. We demonstrate uptake of HDL NPs in SCARB1 expressing medulloblastoma cells and depletion of cholesterol levels in cancer cells. HDL NPs potently blocked proliferation of medulloblastoma cells, as well as hedgehog-driven Ewing sarcoma cells. Furthermore, HDL NPs disrupted colony formation in medulloblastoma and depleted CSC populations in medulloblastoma and Ewing sarcoma. Altogether, our findings provide proof of principle for the development of a novel targeted approach for the treatment of medulloblastoma using HDL NPs. These findings present HDL-mimetic nanoparticles as a promising therapy for sonic hedgehog (SHH) subtype medulloblastoma and possibly other hedgehog-driven cancers.


Assuntos
Neoplasias Cerebelares/metabolismo , Proteínas Hedgehog/metabolismo , Lipoproteínas HDL/metabolismo , Meduloblastoma/metabolismo , Nanopartículas , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Neoplasias Cerebelares/genética , Colesterol/metabolismo , Perfilação da Expressão Gênica , Humanos , Lipoproteínas HDL/química , Meduloblastoma/genética , Nanopartículas/metabolismo , Ligação Proteica , Receptores de Lipoproteínas/metabolismo , Receptores Depuradores Classe B/metabolismo , Transdução de Sinais
10.
Mol Cancer Ther ; 17(3): 686-697, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29282300

RESUMO

Myeloid-derived suppressor cells (MDSC) are innate immune cells that potently inhibit T cells. In cancer, novel therapies aimed to activate T cells can be rendered ineffective due to the activity of MDSCs. Thus, targeted inhibition of MDSCs may greatly enhance T-cell-mediated antitumor immunity, but mechanisms remain obscure. Here we show, for the first time, that scavenger receptor type B-1 (SCARB1), a high-affinity receptor for spherical high-density lipoprotein (HDL), is expressed by MDSCs. Furthermore, we demonstrate that SCARB1 is specifically targeted by synthetic high-density lipoprotein-like nanoparticles (HDL NP), which reduce MDSC activity. Using in vitro T-cell proliferation assays, data show that HDL NPs specifically bind SCARB1 to inhibit MDSC activity. In murine cancer models, HDL NP treatment significantly reduces tumor growth, metastatic tumor burden, and increases survival due to enhanced adaptive immunity. Flow cytometry and IHC demonstrate that HDL NP-mediated suppression of MDSCs increased CD8+ T cells and reduced Treg cells in the metastatic tumor microenvironment. Using transgenic mice lacking SCARB1, in vivo data clearly show that the HDL NPs specifically target this receptor for suppressing MDSCs. Ultimately, our data provide a new mechanism and targeted therapy, HDL NPs, to modulate a critical innate immune cell checkpoint to enhance the immune response to cancer. Mol Cancer Ther; 17(3); 686-97. ©2017 AACR.


Assuntos
Lipoproteínas HDL/toxicidade , Células Supressoras Mieloides/efeitos dos fármacos , Nanopartículas/toxicidade , Neoplasias Experimentais/prevenção & controle , Receptores Depuradores Classe B/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células Supressoras Mieloides/metabolismo , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Receptores Depuradores Classe B/genética , Análise de Sobrevida , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/genética
11.
Nat Commun ; 8(1): 1319, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-29105655

RESUMO

Metastatic cancers produce exosomes that condition pre-metastatic niches in remote microenvironments to favor metastasis. In contrast, here we show that exosomes from poorly metastatic melanoma cells can potently inhibit metastasis to the lung. These "non-metastatic" exosomes stimulate an innate immune response through the expansion of Ly6Clow patrolling monocytes (PMo) in the bone marrow, which then cause cancer cell clearance at the pre-metastatic niche, via the recruitment of NK cells and TRAIL-dependent killing of melanoma cells by macrophages. These events require the induction of the Nr4a1 transcription factor and are dependent on pigment epithelium-derived factor (PEDF) on the outer surface of exosomes. Importantly, exosomes isolated from patients with non-metastatic primary melanomas have a similar ability to suppress lung metastasis. This study thus demonstrates that pre-metastatic tumors produce exosomes, which elicit a broad range of PMo-reliant innate immune responses via trigger(s) of immune surveillance, causing cancer cell clearance at the pre-metastatic niche.


Assuntos
Exossomos/imunologia , Melanoma Experimental/imunologia , Melanoma Experimental/secundário , Monócitos/imunologia , Animais , Diferenciação Celular/imunologia , Proteínas do Olho/imunologia , Feminino , Humanos , Imunidade Inata , Vigilância Imunológica , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Macrófagos/imunologia , Macrófagos/patologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Monócitos/patologia , Fatores de Crescimento Neural/imunologia , Fagocitose/imunologia , Serpinas/imunologia , Microambiente Tumoral/imunologia
12.
Oncotarget ; 8(49): 84643-84658, 2017 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-29156673

RESUMO

The death receptor CD95/Fas can be activated by immune cells to kill cancer cells. shRNAs and siRNAs derived from CD95 or CD95 ligand (CD95L) are highly toxic to most cancer cells. We recently found that these sh/siRNAs kill cancer cells in the absence of the target by targeting the 3'UTRs of critical survival genes through canonical RNAi. We have named this unique form of off-target effect DISE (for death induced by survival gene elimination). DISE preferentially kills transformed cells and cancer stem cells. We demonstrate that DISE induction occurs in cancer cells in vivo after introducing a lentiviral CD95L derived shRNA (shL3) into HeyA8 ovarian cancer cells grown as i.p. xenografts in mice, when compared to a scrambled shRNA. To demonstrate the possibility of therapeutically inducing DISE, we coupled siRNAs to templated lipoprotein nanoparticles (TLP). In vitro, TLPs loaded with a CD95L derived siRNA (siL3) selectively silenced a biosensor comprised of Venus and CD95L ORF and killed ovarian cancer cells. In vivo, two siRNA-TLPs (siL2-TLP and siL3-TLP) reduced tumor growth similarly as observed for cells expressing the shL3 vector. These data suggest that it is possible to kill ovarian cancer cells in vivo via DISE induction using siRNA-TLPs.

13.
Mol Pharm ; 14(11): 4042-4051, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-28933554

RESUMO

Cancer cells have altered metabolism and, in some cases, an increased demand for cholesterol. It is important to identify novel, rational treatments based on biology, and cellular cholesterol metabolism as a potential target for cancer is an innovative approach. Toward this end, we focused on diffuse large B-cell lymphoma (DLBCL) as a model because there is differential cholesterol biosynthesis driven by B-cell receptor (BCR) signaling in germinal center (GC) versus activated B-cell (ABC) DLBCL. To specifically target cellular cholesterol homeostasis, we employed high-density lipoprotein-like nanoparticles (HDL NP) that can generally reduce cellular cholesterol by targeting and blocking cholesterol uptake through the high-affinity HDL receptor, scavenger receptor type B-1 (SCARB1). As we previously reported, GC DLBCL are exquisitely sensitive to HDL NP as monotherapy, while ABC DLBCL are less sensitive. Herein, we report that enhanced BCR signaling and resultant de novo cholesterol synthesis in ABC DLBCL drastically reduces the ability of HDL NPs to reduce cellular cholesterol and induce cell death. Therefore, we combined HDL NP with the BCR signaling inhibitor ibrutinib and the SYK inhibitor R406. By targeting both cellular cholesterol uptake and BCR-associated de novo cholesterol synthesis, we achieved cellular cholesterol reduction and induced apoptosis in otherwise resistant ABC DLBCL cell lines. These results in lymphoma demonstrate that reduction of cellular cholesterol is a powerful mechanism to induce apoptosis. Cells rich in cholesterol require HDL NP therapy to reduce uptake and molecularly targeted agents that inhibit upstream pathways that stimulate de novo cholesterol synthesis, thus, providing a new paradigm for rationally targeting cholesterol metabolism as therapy for cancer.


Assuntos
Linfoma Difuso de Grandes Células B/metabolismo , Nanopartículas/química , Receptores de Antígenos de Linfócitos B/metabolismo , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Colesterol/metabolismo , Humanos , Lipoproteínas HDL/metabolismo , Receptores de Lipoproteínas/metabolismo , Receptores Depuradores Classe B/metabolismo , Transdução de Sinais/fisiologia
14.
Oncotarget ; 8(7): 11219-11227, 2017 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-28061439

RESUMO

Chronic lymphocytic leukemia (CLL) remains incurable despite the introduction of new drugs. Therapies targeting receptors and pathways active specifically in malignant B cells might provide better treatment options. For instance, in B cell lymphoma, our group has previously shown that scavenger receptor type B-1 (SR-B1), the high-affinity receptor for cholesterol-rich high-density lipoproteins (HDL), is a therapeutic target. As evidence suggests that targeting cholesterol metabolism in CLL cells may have therapeutic benefit, we examined SR-B1 expression in primary CLL cells from patients. Unlike normal B cells that do not express SR-B1, CLL cells express the receptor. As a result, we evaluated cholesterol-poor synthetic HDL nanoparticles (HDL NP), known for targeting SR-B1, as a therapy for CLL. HDL NPs potently and selectively induce apoptotic cell death in primary CLL cells. HDL NPs had no effect on normal peripheral blood mononuclear cells from healthy individuals or patients with CLL. These data implicate SR-B1 as a target in CLL and HDL NPs as targeted monotherapy for CLL.


Assuntos
Apoptose/efeitos dos fármacos , Antígenos CD36/metabolismo , Leucemia Linfocítica Crônica de Células B/metabolismo , Lipoproteínas HDL/metabolismo , Ligação Competitiva , Western Blotting , Antígenos CD36/antagonistas & inibidores , Células Cultivadas , Feminino , Citometria de Fluxo , Humanos , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Leucemia Linfocítica Crônica de Células B/patologia , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Lipoproteínas HDL/síntese química , Lipoproteínas HDL/farmacologia , Masculino , Nanopartículas , Ligação Proteica
15.
Proc Natl Acad Sci U S A ; 113(38): 10655-60, 2016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27601638

RESUMO

We report the identification of a molecular signature using the Scano-miR profiling platform based on the differential expression of circulating microRNAs (miRNA, miR) in serum samples specific to patients with very high-risk (VHR) prostate cancer (PCa). Five miRNA PCa biomarkers (miR-200c, miR-605, miR-135a*, miR-433, and miR-106a) were identified as useful for differentiating indolent and aggressive forms of PCa. All patients with VHR PCa in the study had elevated serum levels of miR-200c. Circulating miR-433, which was differentially expressed in patients with VHR versus low-risk (LR) forms of PCa, was not detectable by quantitative real-time PCR in samples from healthy volunteers. In blind studies, the five miRNA PCa biomarkers were able to differentiate patients with VHR PCas from those with LR forms as well as healthy individuals with at least 89% accuracy. Biological pathway analysis showed the predictive capability of these miRNA biomarkers for the diagnosis and prognosis of VHR aggressive PCa.


Assuntos
Biomarcadores Tumorais/sangue , MicroRNA Circulante/sangue , Neoplasias da Próstata/sangue , Idoso , MicroRNA Circulante/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Fatores de Risco , Transcriptoma
16.
Sci Rep ; 6: 22915, 2016 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-26964503

RESUMO

Exosomes are produced by cells to mediate intercellular communication, and have been shown to perpetuate diseases, including cancer. New tools are needed to understand exosome biology, detect exosomes from specific cell types in complex biological media, and to modify exosomes. Our data demonstrate a cellular pathway whereby membrane-bound scavenger receptor type B-1 (SR-B1) in parent cells becomes incorporated into exosomes. We tailored synthetic HDL-like nanoparticles (HDL NP), high-affinity ligands for SR-B1, to carry a fluorescently labeled phospholipid. Data show SR-B1-dependent transfer of the fluorescent phospholipid from HDL NPs to exosomes. Modified exosomes are stable in serum and can be directly detected using flow cytometry. As proof-of-concept, human serum exosomes were found to express SR-B1, and HDL NPs can be used to label and isolate them. Ultimately, we discovered a natural cellular pathway and nanoparticle-receptor pair that enables exosome modulation, detection, and isolation.


Assuntos
Técnicas Biossensoriais , Comunicação Celular/genética , Exossomos/metabolismo , Receptores Depuradores Classe B/isolamento & purificação , Exossomos/química , Humanos , Ligantes , Metabolismo dos Lipídeos/genética , Lipoproteínas HDL/química , Nanopartículas/química , Fosfolipídeos/química , Fosfolipídeos/metabolismo , Ligação Proteica , Receptores Depuradores Classe B/sangue , Receptores Depuradores Classe B/química , Receptores Depuradores Classe B/genética
17.
Adv Funct Mater ; 26(43): 7824-7835, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28717350

RESUMO

Efficient systemic administration of therapeutic short interfering RNA (siRNA) is challenging. High-density lipoproteins (HDL) are natural in vivo RNA delivery vehicles. Specifically, native HDLs: 1) Load single-stranded RNA; 2) Are anionic, which requires charge reconciliation between the RNA and HDL, and 3) Actively target scavenger receptor type B-1 (SR-B1) to deliver RNA. Emphasizing these particular parameters, we employed templated lipoprotein particles (TLP), mimics of spherical HDLs, and self-assembled them with single-stranded complements of, presumably, any highly unmodified siRNA duplex pair after formulation with a cationic lipid. Resulting siRNA templated lipoprotein particles (siRNA-TLP) are anionic and tunable with regard to RNA assembly and function. Data demonstrate that the siRNA-TLPs actively target SR-B1 to potently reduce androgen receptor (AR) and enhancer of zeste homolog 2 (EZH2) proteins in multiple cancer cell lines. Systemic administration of siRNA-TLPs demonstrated no off-target toxicity and significantly reduced the growth of prostate cancer xenografts. Thus, native HDLs inspired the synthesis of a hybrid siRNA delivery vehicle that can modularly load single-stranded RNA complements after charge reconciliation with a cationic lipid, and that function due to active targeting of SR-B1.

18.
Sci Rep ; 5: 15724, 2015 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-26511855

RESUMO

Exosomes are nanoscale vesicles that mediate intercellular communication. Cellular exosome uptake mechanisms are not well defined partly due to the lack of specific inhibitors of this complex cellular process. Exosome uptake depends on cholesterol-rich membrane microdomains called lipid rafts, and can be blocked by non-specific depletion of plasma membrane cholesterol. Scavenger receptor type B-1 (SR-B1), found in lipid rafts, is a receptor for cholesterol-rich high-density lipoproteins (HDL). We hypothesized that a synthetic nanoparticle mimic of HDL (HDL NP) that binds SR-B1 and removes cholesterol through this receptor would inhibit cellular exosome uptake. In cell models, our data show that HDL NPs bind SR-B1, activate cholesterol efflux, and attenuate the influx of esterified cholesterol. As a result, HDL NP treatment results in decreased dynamics and clustering of SR-B1 contained in lipid rafts and potently inhibits cellular exosome uptake. Thus, SR-B1 and targeted HDL NPs provide a fundamental advance in studying cholesterol-dependent cellular uptake mechanisms.


Assuntos
Materiais Biomiméticos , Colesterol/metabolismo , Exossomos/metabolismo , Lipoproteínas HDL , Nanopartículas/química , Receptores Depuradores Classe B/metabolismo , Animais , Transporte Biológico Ativo , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Linhagem Celular Tumoral , Humanos , Lipoproteínas HDL/química , Lipoproteínas HDL/farmacologia , Camundongos
19.
Cancer Treat Res ; 166: 129-50, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25895867

RESUMO

High-density lipoproteins (HDL) are diverse natural nanoparticles that carry cholesterol and are best known for the role that they play in cardiovascular disease. However, due to their unique targeting capabilities, diverse molecular cargo, and natural functions beyond cholesterol transport, it is becoming increasingly appreciated that HDLs are critical to cancer development and progression. Accordingly, this chapter highlights ongoing research focused on the connections between HDL and cancer in order to design new drugs and targeted drug delivery vehicles. Research is focused on synthesizing biomimetic HDL-like nanoparticles (NP) that can be loaded with diverse therapeutic cargo (e.g., chemotherapies, nucleic acids, proteins) and specifically targeted to cancer cells. Beyond drug delivery, new data is emerging that HDL-like NPs may be therapeutically active in certain tumor types, for example, B cell lymphoma. Overall, HDL-like NPs are becoming increasingly appreciated as targeted, biocompatible, and efficient therapies for cancer, and may soon become indispensable agents in the cancer therapeutic armamentarium.


Assuntos
Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Lipoproteínas HDL/uso terapêutico , Nanoconjugados/uso terapêutico , Nanomedicina/métodos , Neoplasias/tratamento farmacológico , Animais , Humanos , Lipoproteínas HDL/química
20.
J Lipid Res ; 56(5): 972-85, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25652088

RESUMO

The ability of HDL to support macrophage cholesterol efflux is an integral part of its atheroprotective action. Augmenting this ability, especially when HDL cholesterol efflux capacity from macrophages is poor, represents a promising therapeutic strategy. One approach to enhancing macrophage cholesterol efflux is infusing blood with HDL mimics. Previously, we reported the synthesis of a functional mimic of HDL (fmHDL) that consists of a gold nanoparticle template, a phospholipid bilayer, and apo A-I. In this work, we characterize the ability of fmHDL to support the well-established pathways of cellular cholesterol efflux from model cell lines and primary macrophages. fmHDL received cell cholesterol by unmediated (aqueous) and ABCG1- and scavenger receptor class B type I (SR-BI)-mediated diffusion. Furthermore, the fmHDL holoparticle accepted cholesterol and phospholipid by the ABCA1 pathway. These results demonstrate that fmHDL supports all the cholesterol efflux pathways available to native HDL and thus, represents a promising infusible therapeutic for enhancing macrophage cholesterol efflux. fmHDL accepts cholesterol from cells by all known pathways of cholesterol efflux: unmediated, ABCG1- and SR-BI-mediated diffusion, and through ABCA1.


Assuntos
Apolipoproteína A-I/farmacologia , Cardiotônicos/farmacologia , Colesterol/metabolismo , Nanopartículas/metabolismo , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Apolipoproteína A-I/metabolismo , Transporte Biológico , Linhagem Celular , Doença da Artéria Coronariana/tratamento farmacológico , Cricetinae , Avaliação Pré-Clínica de Medicamentos , Estabilidade de Medicamentos , Ouro/metabolismo , Lipoproteínas/metabolismo , Macrófagos/metabolismo , Mimetismo Molecular , Fosfolipídeos/farmacologia , Receptores Depuradores Classe B/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA