Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
J Extracell Vesicles ; 12(8): e12354, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37553837

RESUMO

Extracellular vesicles (EVs) can be produced from red blood cells (RBCs) on a large scale and used to deliver therapeutic payloads efficiently. However, not much is known about the native biological properties of RBCEVs. Here, we demonstrate that RBCEVs are primarily taken up by macrophages and monocytes. This uptake is an active process, mediated mainly by endocytosis. Incubation of CD14+ monocytes with RBCEVs induces their differentiation into macrophages with an Mheme-like phenotype, characterized by upregulation of heme oxygenase-1 (HO-1) and the ATP-binding cassette transporter ABCG1. Moreover, macrophages that take up RBCEVs exhibit a reduction in surface CD86 and decreased secretion of TNF-α under inflammatory stimulation. The upregulation of HO-1 is attributed to heme derived from haemoglobin in RBCEVs. Heme is released from internalized RBCEVs in late endosomes and lysosomes via the heme transporter, HRG1. Consequently, RBCEVs exhibit the ability to attenuate foam cell formation from oxidized low-density lipoproteins (oxLDL)-treated macrophages in vitro and reduce atherosclerotic lesions in ApoE knockout mice on a high-fat diet. In summary, our study reveals the uptake mechanism of RBCEVs and their delivery of heme to macrophages, suggesting the potential application of RBCEVs in the treatment of atherosclerosis.


Assuntos
Aterosclerose , Vesículas Extracelulares , Animais , Camundongos , Células Espumosas/metabolismo , Células Espumosas/patologia , Heme/metabolismo , Vesículas Extracelulares/metabolismo , Macrófagos/metabolismo , Eritrócitos/metabolismo , Endocitose
3.
Mol Ther ; 31(5): 1418-1436, 2023 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-37016578

RESUMO

Cancer cachexia is a multifactorial syndrome characterized by a significant loss of skeletal muscle, which negatively affects the quality of life. Inhibition of myostatin (Mstn), a negative regulator of skeletal muscle growth and differentiation, has been proven to preserve muscle mass in muscle atrophy diseases, including cachexia. However, myostatin inhibitors have repeatedly failed clinical trials because of modest therapeutic effects and side effects due to the poor efficiency and toxicity of existing delivery methods. Here, we describe a novel method for delivering Mstn siRNA to skeletal muscles using red blood cell-derived extracellular vesicles (RBCEVs) in a cancer cachectic mouse model. Our data show that RBCEVs are taken up by myofibers via intramuscular administration. Repeated intramuscular administrations with RBCEVs allowed the delivery of siRNAs, thereby inhibiting Mstn, increasing muscle growth, and preventing cachexia in cancer-bearing mice. We observed the same therapeutic effects when delivering siRNAs against malonyl-CoA decarboxylase, an enzyme driving dysfunctional fatty acid metabolism in skeletal muscles during cancer cachexia. We demonstrate that intramuscular siRNA delivery by RBCEVs is safe and non-inflammatory. Hence, this method is useful to reduce the therapeutic dose of siRNAs, to avoid toxicity and off-target effects caused by systemic administration of naked siRNAs at high doses.


Assuntos
Miostatina , Neoplasias , Camundongos , Animais , Miostatina/metabolismo , RNA Interferente Pequeno/metabolismo , Caquexia/etiologia , Caquexia/terapia , Caquexia/metabolismo , Qualidade de Vida , Músculo Esquelético/metabolismo , Neoplasias/complicações , Neoplasias/terapia , Neoplasias/metabolismo , Atrofia Muscular , RNA de Cadeia Dupla
4.
ACS Nano ; 17(6): 5187-5210, 2023 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-36896898

RESUMO

Red blood cells (RBCs) and RBC membrane-derived nanoparticles have been historically developed as bioinspired drug delivery systems to combat the issues of premature clearance, toxicity, and immunogenicity of synthetic nanocarriers. RBC-based delivery systems possess characteristics including biocompatibility, biodegradability, and long circulation time, which make them suited for systemic administration. Therefore, they have been employed in designing optimal drug formulations in various preclinical models and clinical trials to treat a wide range of diseases. In this review, we provide an overview of the biology, synthesis, and characterization of drug delivery systems based on RBCs and their membrane including whole RBCs, RBC membrane-camouflaged nanoparticles, RBC-derived extracellular vesicles, and RBC hitchhiking. We also highlight conventional and latest engineering strategies, along with various therapeutic modalities, for enhanced precision and effectiveness of drug delivery. Additionally, we focus on the current state of RBC-based therapeutic applications and their clinical translation as drug carriers, as well as discussing opportunities and challenges associated with these systems.


Assuntos
Sistemas de Liberação de Medicamentos , Nanopartículas , Portadores de Fármacos , Eritrócitos , Nanopartículas/uso terapêutico , Administração Cutânea
5.
Theranostics ; 12(7): 3288-3315, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35547755

RESUMO

The advent of novel therapeutics in recent years has urged the need for a safe, non-immunogenic drug delivery vector capable of delivering therapeutic payloads specifically to diseased cells, thereby increasing therapeutic efficacy and reducing side effects. Extracellular vesicles (EVs) have garnered attention in recent years as a potentially ideal vector for drug delivery, taking into account their intrinsic ability to transfer bioactive cargo to recipient cells and their biocompatible nature. However, natural EVs are limited in their therapeutic potential and many challenges need to be overcome before engineered EVs satisfy the levels of efficiency, stability, safety and biocompatibility required for therapeutic use. Here, we demonstrate that an enzyme-mediated surface functionalization method in combination with streptavidin-mediated conjugation results in efficient surface functionalization of EVs. Surface functionalization using the above methods permits the stable and biocompatible conjugation of peptides, single domain antibodies and monoclonal antibodies at high copy number on the EV surface. Functionalized EVs demonstrated increased accumulation in target cells expressing common cancer associated markers such as CXCR4, EGFR and EpCAM both in vitro and in vivo. The functionality of this approach was further highlighted by the ability of targeting EVs to specifically deliver therapeutic antisense oligonucleotides to a metastatic breast tumor model, resulting in increased knockdown of a targeted oncogenic microRNA and improved metastasis suppression. The method was also used to equip EVs with a bifunctional peptide that targets EVs to leukemia cells and induces apoptosis, leading to leukemia suppression. Moreover, we conducted extensive testing to verify the biocompatibility, and safety of engineered EVs for therapeutic use, suggesting that surface modified EVs can be used for repeated dose treatment with no detectable adverse effects. This modular, biocompatible method of EV engineering offers a promising avenue for the targeted delivery of a range of therapeutics while addressing some of the safety concerns associated with EV-based drug delivery.


Assuntos
Vesículas Extracelulares , Leucemia , Neoplasias , Sistemas de Liberação de Medicamentos/métodos , Vesículas Extracelulares/química , Humanos , Neoplasias/tratamento farmacológico , Peptídeos
6.
J Extracell Vesicles ; 11(4): e12187, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35430766

RESUMO

The RIG-I pathway can be activated by RNA containing 5' triphosphate, leading to type I interferon release and immune activation. Hence, RIG-I agonists have been used to induce immune responses against cancer as potential immunotherapy. However, delivery of 5' triphosphorylated RNA molecules as RIG-I agonists to tumour cells in vivo is challenging due to the susceptibility of these molecules to degradation. In this study, we demonstrate the use of extracellular vesicles (EVs) from red blood cells (RBCs), which are highly amenable for RNA loading and taken up robustly by cancer cells, for RIG-I agonist delivery. We evaluate the anti-cancer activity of two novel RIG-I agonists, the immunomodulatory RNA (immRNA) with a unique secondary structure for efficient RIG-I activation, and a 5' triphosphorylated antisense oligonucleotide with dual function of RIG-I activation and miR-125b inhibition (3p-125b-ASO). We find that RBCEV-delivered immRNA and 3p-125b-ASO trigger the RIG-I pathway, and induce cell death in both mouse and human breast cancer cells. Furthermore, we observe a significant suppression of tumour growth coupled with increased immune cell infiltration mediated by the activation of RIG-I cascade after multiple intratumoral injections of RBCEVs loaded with immRNA or 3p-125b-ASO. Targeted delivery of immRNA using RBCEVs with EGFR-binding nanobody administrated via intrapulmonary delivery facilitates the accumulation of RBCEVs in metastatic cancer cells, leading to potent tumour-specific CD8+ T cells immune response. This contributes to prominent suppression of breast cancer metastasis in the lung. Hence, this study provides a new strategy for efficient RIG-I agonist delivery using RBCEVs for immunotherapy against cancer and cancer metastasis.


Assuntos
Neoplasias da Mama , Vesículas Extracelulares , Melanoma , Animais , Neoplasias da Mama/tratamento farmacológico , Linfócitos T CD8-Positivos , Vesículas Extracelulares/metabolismo , Feminino , Humanos , Fatores Imunológicos/metabolismo , Imunoterapia , Melanoma/metabolismo , Camundongos , RNA/metabolismo , Neoplasias Cutâneas , Melanoma Maligno Cutâneo
7.
Sci Rep ; 12(1): 6265, 2022 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-35428767

RESUMO

The greenhouse effect of SF6 increasingly limits its application in various gas insulated equipment. C6F12O combines the advantages of insulation resistance, safety and environmental protection. When mixed with buffer gas, C6F12O is considered to have potential application prospects in medium and low voltage equipment. In this paper, about the partial discharge characteristics of the mixed gas, an experimental study was carried out. The partial discharge initiation voltage and discharge extinction voltage of mixed gas under power frequency voltage are measured and compared with the breakdown voltage. The results show that the breakdown voltage is greatly improved after adding C6F12O, with the increase of mixing ratio, the partial discharge initiation voltage and extinction voltage of mixed gas gradually increase, and the effect of gas pressure on high mixing ratio is obvious. The difference between the partial discharge inception voltage and the breakdown voltage is larger than that of pure N2. The research in this paper can provide an important reference for the application, operation and protection of C6F12O mixed gas in medium and low voltage equipment.

8.
Mol Ther ; 29(11): 3258-3273, 2021 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33974998

RESUMO

Dysregulated adenosine-to-inosine (A-to-I) RNA editing is implicated in various cancers. However, no available RNA editing inhibitors have so far been developed to inhibit cancer-associated RNA editing events. Here, we decipher the RNA secondary structure of antizyme inhibitor 1 (AZIN1), one of the best-studied A-to-I editing targets in cancer, by locating its editing site complementary sequence (ECS) at the 3' end of exon 12. Chemically modified antisense oligonucleotides (ASOs) that target the editing region of AZIN1 caused a substantial exon 11 skipping, whereas ECS-targeting ASOs effectively abolished AZIN1 editing without affecting splicing and translation. We demonstrate that complete 2'-O-methyl (2'-O-Me) sugar ring modification in combination with partial phosphorothioate (PS) backbone modification may be an optimal chemistry for editing inhibition. ASO3.2, which targets the ECS, specifically inhibits cancer cell viability in vitro and tumor incidence and growth in xenograft models. Our results demonstrate that this AZIN1-targeting, ASO-based therapeutics may be applicable to a wide range of tumor types.


Assuntos
Proteínas de Transporte/genética , Marcação de Genes , Edição de RNA , Animais , Sequência de Bases , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular/genética , Modelos Animais de Doenças , Éxons , Regulação Neoplásica da Expressão Gênica , Marcação de Genes/métodos , Terapia Genética/métodos , Humanos , Camundongos , Neoplasias/genética , Neoplasias/terapia , Oligonucleotídeos Antissenso/genética , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Extracell Vesicles ; 10(4): e12057, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33643546

RESUMO

Natural extracellular vesicles (EVs) are ideal drug carriers due to their remarkable biocompatibility. Their delivery specificity can be achieved by the conjugation of targeting ligands. However, existing methods to engineer target-specific EVs are tedious or inefficient, having to compromise between harsh chemical treatments and transient interactions. Here, we describe a novel method for the covalent conjugation of EVs with high copy numbers of targeting moieties using protein ligases. Conjugation of EVs with either an epidermal growth factor receptor (EGFR)-targeting peptide or anti-EGFR nanobody facilitates their accumulation in EGFR-positive cancer cells, both in vitro and in vivo. Systemic delivery of paclitaxel by EGFR-targeting EVs at a low dose significantly increases drug efficacy in a xenografted mouse model of EGFR-positive lung cancer. The method is also applicable to the conjugation of EVs with peptides and nanobodies targeting other receptors, such as HER2 and SIRP alpha, and the conjugated EVs can deliver RNA in addition to small molecules, supporting the versatile application of EVs in cancer therapies. This simple, yet efficient and versatile method for the stable surface modification of EVs bypasses the need for genetic and chemical modifications, thus facilitating safe and specific delivery of therapeutic payloads to target cells.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Vesículas Extracelulares , Peptídeos/uso terapêutico , Anticorpos de Domínio Único/uso terapêutico , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Linhagem Celular Tumoral , Portadores de Fármacos/química , Portadores de Fármacos/uso terapêutico , Receptores ErbB/química , Receptores ErbB/uso terapêutico , Eritrócitos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Paclitaxel/uso terapêutico , Peptídeos/química , Anticorpos de Domínio Único/química , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Semin Cancer Biol ; 74: 62-78, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33609665

RESUMO

Cancer is a disease that evolves continuously with unpredictable outcomes. Although conventional chemotherapy can display significant antitumor effects, the lack of specificity and poor bioavailability remain major concerns in cancer therapy. Moreover, with the advent of novel anti-cancer gene therapies, there is an urgent need for drug delivery vectors capable of bypassing cellular barriers and efficiently transferring therapeutic cargo to recipient cells. A number of drug delivery systems have been proposed to overcome these limitations, but their successful clinical translation has been hampered by the onset of unexpected side effects and associated toxicities. The application of extracellular vesicles (EVs), a class of naturally released, cell-derived particles, as drug delivery vectors presents a breakthrough in nanomedicine, taking into account their biocompatibility and natural role in intercellular communication. Combining the advantageous intrinsic properties of EVs with surface functionalization and the encapsulation of drugs allows for a new class of engineered EVs that serve as effective therapeutic carriers. Here, we describe the various successful approaches involving the application of engineered EVs as bio-derived drug delivery vectors in cancer therapy. The latest and most effective strategies of engineering EVs to improve drug loading, stealth properties and tumour targeting capabilities of EVs are debated. Finally, current obstacles and future perspectives of smart engineered EVs are discussed.


Assuntos
Bioengenharia/métodos , Portadores de Fármacos , Sistemas de Liberação de Medicamentos/métodos , Vesículas Extracelulares , Neoplasias/tratamento farmacológico , Animais , Bioengenharia/tendências , Sistemas de Liberação de Medicamentos/tendências , Humanos
11.
Cell Prolif ; 54(2): e12913, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33332677

RESUMO

MicroRNAs (miRNAs) are small and highly conserved non-coding RNAs that silence target mRNAs, and compelling evidence suggests that they play an essential role in the pathogenesis of human diseases, especially cancer. miR-125b, which is the mammalian orthologue of the first discovered miRNA lin-4 in Caenorhabditis elegans, is one of the most important miRNAs that regulate various physiological and pathological processes. The role of miR-125b in many types of cancer has been well established, and so here we review the current knowledge of how miR-125b is deregulated in different types of cancer; its oncogenic and/or tumour-suppressive roles in tumourigenesis and cancer progression; and its regulation with regard to treatment response, all of which are underlined in multiple studies. The emerging information that elucidates the essential functions of miR-125b might help support its potentiality as a diagnostic and prognostic biomarker as well as an effective therapeutic tool against cancer.


Assuntos
MicroRNAs/metabolismo , Neoplasias/patologia , Apoptose/genética , Resistencia a Medicamentos Antineoplásicos/genética , Glicólise/genética , Histonas/metabolismo , Humanos , Metástase Neoplásica , Neoplasias/genética , RNA Longo não Codificante/metabolismo , Transdução de Sinais/genética
12.
Anim Sci J ; 91(1): e13465, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33222358

RESUMO

The present study was conducted to investigate the effect of cold storage time on apoptosis of cumulus cells (CCs) from porcine ovaries, and to compare the sensitivity of four apoptosis-detection methods. Porcine ovaries were stored in physiological saline solution at 4°C for 0, 7, 24 and 48 hr, and then cumulus cells or granulosa cells (GCs) in antral follicles were retrieved to detect cell apoptosis. Cumulus cells isolated from stored ovaries for 24 hr presented obvious apoptosis using terminal deoxynucleotidyl transferase (TdT)-mediated d-UTP nick end-labeling (TUNEL) assay. A typical DNA ladder pattern of apoptosis was observed in GCs 24 hr post storage treatment. The mean Olive Tail Moment of CCs was significantly increased after 24 hr using comet assay; however, the mean tail migration and mean tail DNA increased gradually after 7 hr of storage. In addition, annexin V/PI staining assay showed an obvious increase in apoptotic CCs (Annexin V positive, PI negative) 7 hr after treatment, and the apoptotic rate reached to a peak at 24 hr followed by a decline after 48 hr of storage to the level at 7 hr. In conclusion, cold storage of porcine ovary in physiological saline solution induced a time-dependent increase in apoptosis of cumulus cells, and annexin V/PI staining combined with comet assay provided a sensitive and reliable method to detect early damages in cumulus cells induced by cold storage of ovary.


Assuntos
Apoptose , Temperatura Baixa/efeitos adversos , Células do Cúmulo/patologia , Preservação de Órgãos/efeitos adversos , Preservação de Órgãos/métodos , Folículo Ovariano/citologia , Ovário , Animais , Separação Celular , Células Cultivadas , Células do Cúmulo/fisiologia , DNA , Feminino , Marcação In Situ das Extremidades Cortadas/métodos , Ovário/citologia , Suínos , Fatores de Tempo
13.
Nat Commun ; 10(1): 2883, 2019 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-31253768

RESUMO

A substantial number of mouse genes, about 25%, are embryonically lethal when knocked out. Using current genetic tools, such as the CRISPR-Cas9 system, it is difficult-or even impossible-to produce viable mice with heritable embryonically lethal mutations. Here, we establish a one-step method for microinjection of CRISPR reagents into one blastomere of two-cell embryos to generate viable chimeric founder mice with a heritable embryonically lethal mutation, of either Virma or Dpm1. By examining founder mice, we identify a phenotype and role of Virma in regulating kidney metabolism in adult mice. Additionally, we generate knockout mice with a heritable postnatally lethal mutation, of either Slc17a5 or Ctla-4, and study its function in vivo. This one-step method provides a convenient system that rapidly generates knockout mice possessing lethal phenotypes. This allows relatively easy in vivo study of the associated genes' functions.


Assuntos
Sistemas CRISPR-Cas , Embrião de Mamíferos/fisiologia , Animais , Desenvolvimento Embrionário , Feminino , Edição de Genes/métodos , Engenharia Genética/métodos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Mutação , RNA Guia de Cinetoplastídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
14.
J Extracell Vesicles ; 8(1): 1599680, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31044053

RESUMO

Tumour cells release large quantities of extracellular vesicles (EVs) to mediate their interactions with other cells in the tumour microenvironment. To identify host cells that naturally take up EVs from tumour cells, we created breast cancer cell lines secreting fluorescent EVs. These fluorescent EVs are taken up most robustly by fibroblasts within the tumour microenvironment. RNA sequencing indicated that miR-125b is one of the most abundant microRNAs secreted by mouse triple-negative breast cancer 4T1 and 4TO7 cells. Treatment with 4T1 EVs leads to an increase in fibroblast activation in isogenic 4TO7 tumours, which is reversed by blocking miR-125b in 4T1 EVs; hence, miR-125b delivery by EVs is responsible for fibroblast activation in mouse tumour models. miR-125b is also secreted by human breast cancer cells and the uptake of EVs from these cells significantly increases cellular levels of miR-125b and expression of multiple cancer-associated fibroblast markers in resident fibroblasts. Overexpression of miR-125b in both mouse and human fibroblasts leads to an activated phenotype similar to the knockdown of established miR-125b target mRNAs. These data indicate that miR-125b is transferred through EVs from breast cancer cells to normal fibroblasts within the tumour microenvironment and contributes to their development into cancer-associated fibroblasts.

15.
Chem Commun (Camb) ; 54(77): 10878-10881, 2018 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-30204160

RESUMO

Here we identify hundreds of RNA G-quadruplex (rG4) candidates in microRNAs (miRNAs), characterize the miRNA structure and miRNA-mRNA interactions on several mammalian-conserved miRNAs, and reveal the formation of rG4s in miRNAs. Notably, we study the effect of these rG4s in cells and uncover the role of rG4s in miRNA-mediated post-transcriptional regulation.


Assuntos
Quadruplex G , MicroRNAs/química , Células HEK293 , Humanos , MicroRNAs/metabolismo
16.
Nat Commun ; 9(1): 2359, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29907766

RESUMO

Most of the current methods for programmable RNA drug therapies are unsuitable for the clinic due to low uptake efficiency and high cytotoxicity. Extracellular vesicles (EVs) could solve these problems because they represent a natural mode of intercellular communication. However, current cellular sources for EV production are limited in availability and safety in terms of horizontal gene transfer. One potentially ideal source could be human red blood cells (RBCs). Group O-RBCs can be used as universal donors for large-scale EV production since they are readily available in blood banks and they are devoid of DNA. Here, we describe and validate a new strategy to generate large-scale amounts of RBC-derived EVs for the delivery of RNA drugs, including antisense oligonucleotides, Cas9 mRNA, and guide RNAs. RNA drug delivery with RBCEVs shows highly robust microRNA inhibition and CRISPR-Cas9 genome editing in both human cells and xenograft mouse models, with no observable cytotoxicity.


Assuntos
Sistemas de Liberação de Medicamentos , Eritrócitos/metabolismo , Vesículas Extracelulares , RNA Guia de Cinetoplastídeos , RNA/análise , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Nus , Camundongos SCID , MicroRNAs/genética , Transplante de Neoplasias , Oligonucleotídeos Antissenso/genética
17.
Oncotarget ; 7(36): 58038-58050, 2016 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-27487128

RESUMO

Prolonged treatment of breast cancer with epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) often results in acquired resistance and a narrow therapeutic index. One strategy to improve the therapeutic effects of EGFR TKIs is to combine them with drugs used for other clinical indications. Ethacrynic acid (EA) is an FDA approved drug that may have antitumor effects and may enhance the cytotoxicity of chemotherapeutic agents by binding to glutathione and inhibiting WNT signaling. While the α,ß-unsaturated-keto structure of EA is similar to that of irreversible TKIs, the mechanism of action of EA when combined with irreversible EGFR TKIs in breast cancer remains unknown. We therefore investigated the combination of irreversible EGFR TKIs and EA. We found that irreversible EGFR TKIs and EA synergistically inhibit breast cancer both in vitro and in vivo. The combination of EGFR TKIs and EA induces necrosis and cell cycle arrest and represses WNT/ß-catenin signaling as well as MAPK-ERK1/2 signaling. We conclude that EA synergistically enhances the antitumor effects of irreversible EGFR TKIs in breast cancer.


Assuntos
Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Ácido Etacrínico/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Afatinib , Animais , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Mama/patologia , Neoplasias da Mama/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sinergismo Farmacológico , Ácido Etacrínico/uso terapêutico , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células MCF-7 , Camundongos Endogâmicos BALB C , Inibidores de Proteínas Quinases/uso terapêutico , Quinazolinas/farmacologia , Quinazolinas/uso terapêutico , Quinolinas/farmacologia , Quinolinas/uso terapêutico , Via de Sinalização Wnt/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(7): 876-80, 2016 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-27363264

RESUMO

Objective To investigate the synergistic anti-breast cancer effect of Toll-like receptor 7 agonist T7-ethacrynic acid conjugate (T7-EA) in combination with receptor-tyrosine-kinase-like orphan receptor 1 (ROR1). Methods ROR1 cytotoxic T lymphocyte (CTL) epitope was predicted using Syfpeithi online software. Mouse spleen lymphocytes and bone marrow dendritic cells (DCs) were separately stimulated with 4 µmol/L T7-EA and 4 µmol/L ROR1 alone or in combination. ELISA assay was used to measure the levels of interferon-γ (IFN-γ), interleukin 12 (IL-12) and tumor necrosis factor-α (TNF-α). Xenograft model was established via subcutaneous injection of mouse breast cancer 4T1 cells. The mice were weekly treated through intraperitoneal administration of 3 mg/kg T7-EA, 15 mg/kg ROR1 or the combination of T7-EA and ROR1. After four rounds of treatment, tumor tissues were weighed. Serum level of anti-4T1 tumor protein IgG was measured by ELISA. Specific CTL activity was detected by lactate dehydrogenase (LDH) assay. Results The peptide PYCDETSSV was chosen as an antigen epitope of breast cancer. The T7-EA highly activated in vitro lymphocytes in a dose-dependent manner, which wasn't affected by other relevant peptides. The combination of T7-EA and ROR1 stimulated the secretion of IFN-γ and IL-12 by lymphocytes and TNF-α by bone marrow DCs. The growth of tumor in vivo was significantly inhibited by T7-EA combined with ROR1 compared with T7-EA or ROR1 alone. The specific CTL activity triggered by T7-EA combined with ROR1 was much stronger than that triggered by T7-EA or ROR1 alone. The titer of anti-4T1 tumor protein IgG induced by T7-EA combined with ROR1 was higher than that induced by T7-EA or ROR1. Conclusion The combination of T7-EA and ROR1 has a better killing effect on breast cancer.


Assuntos
Vacinas Anticâncer/imunologia , Ácido Etacrínico/imunologia , Neoplasias Mamárias Experimentais/imunologia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/imunologia , Receptor 7 Toll-Like/imunologia , Sequência de Aminoácidos , Animais , Vacinas Anticâncer/farmacologia , Linhagem Celular Tumoral , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-12/imunologia , Interleucina-12/metabolismo , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Linfócitos/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/prevenção & controle , Camundongos Endogâmicos BALB C , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Fatores de Tempo , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/metabolismo
19.
Oncol Rep ; 35(2): 793-800, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26718332

RESUMO

Immunotherapy is emerging as a powerful and active tumor-specific approach against cancer via triggering the immune system. Toll-like receptors (TLRs) are fundamental elements of the immune system, which facilitate our understanding of the innate and adaptive immune pathways. TLR agonists used as single agents can effectively eradicate tumors due to their potent stimulation of innate and adaptive immunity. We examined the effects of a novel adenine type of TLR7 agonists on both innate and adaptive immune activation in vitro and in vivo. We established the local and distant tumor­bearing mice derived from murine mammary carcinoma cell line (4T1) to model metastatic disease. Our data demonstrated that SZU101 was able to stimulate innate immune cells to release cytokines at the very high level compared with LPS at the same or lower concentration. Locally intratumoral SZU101 injection can elicit a systemic antitumor effect on murine breast tumor model. SZU101 affected the frequency of intratumoral immune cell infiltration, including the percentage of CD4+ and CD8+ increase, and the ratio of Tregs decrease. Our data reveal that the antitumor effect of SZU101 is associated with multiple mechanisms, inducing tumor­specific immune response, activation of innate immune cells and modulation of the tumor microenvironment.


Assuntos
Adenina/análogos & derivados , Antineoplásicos/farmacologia , Neoplasias da Mama/imunologia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Glicoproteínas de Membrana/agonistas , Succinatos/farmacologia , Receptor 7 Toll-Like/agonistas , Microambiente Tumoral/efeitos dos fármacos , Adenina/farmacologia , Animais , Western Blotting , Neoplasias da Mama/patologia , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Microambiente Tumoral/imunologia
20.
Future Oncol ; 12(1): 31-42, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26615672

RESUMO

AIM: To investigate ANXA5 overexpression on in vitro and in vivo malignancies of murine Hca-P cells. MATERIALS & METHODS: Hca-P with low lymph node metastasis (LNM) potential was used as cell model. TEM, CCK-8 and Boyden transwell assays were performed for in vitro Hca-P behaviors. Hca-P-transplanted mouse model was established for in vivo experiment. RESULTS: ANXA5-overexpressing monoclonal Anxa5-Hca-P-1, Anxa5-Hca-P-2 and Anxa5-Hca-P-3 cells were obtained. ANXA5 upregulation alters the proliferation, morphology and rough endoplasmic reticulum of Hca-P cells, enhances in vitro migration and invasions of Hca-P, promotes in vivo malignant degree and LNM rate of Anxa5-Hca-P-3-transplanted mice. CONCLUSION: As a potential indicator for malignancy and lymphatic metastasis, ANXA5 overexpression increases in vitro migration and invasion of Hca-P cell, promotes in vivo malignancy, LNM rate and level of Hca-P-transplanted mice.


Assuntos
Anexina A5/biossíntese , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Metástase Linfática/genética , Animais , Anexina A5/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/patologia , Metástase Linfática/patologia , Camundongos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...