Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Biomater Sci ; 12(9): 2292-2301, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38498328

RESUMEN

Colorectal cancer (CRC) ranks among the most prevalent cancers globally, demanding innovative therapeutic strategies. Immunotherapy, a promising avenue, employs cancer vaccines to activate the immune system against tumors. However, conventional approaches fall short of eliciting robust responses within the gastrointestinal (GI) tract, where CRC originates. Harnessing the potential of all-trans retinoic acid (ATRA) and cytosine-phosphorothioate-guanine (CpG), we developed layered nanoparticles using a layer-by-layer assembly method to co-deliver these agents. ATRA, crucial for gut immunity, was efficiently encapsulated alongside CpG within these nanoparticles. Administering these ATRA@CpG-NPs, combined with ovalbumin peptide (OVA), effectively inhibited orthotopic CRC growth in mice. Our approach leveraged the inherent benefits of ATRA and CpG, demonstrating superior efficacy in activating dendritic cells, imprinting T cells with gut-homing receptors, and inhibiting tumor growth. This mucosal adjuvant presents a promising strategy for CRC immunotherapy, showcasing the potential for targeting gut-associated immune responses in combating colorectal malignancies.


Asunto(s)
Neoplasias Colorrectales , Fosfatos de Dinucleósidos , Nanopartículas , Tretinoina , Tretinoina/química , Tretinoina/administración & dosificación , Tretinoina/farmacología , Animales , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/inmunología , Nanopartículas/química , Nanopartículas/administración & dosificación , Ratones , Humanos , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/química , Ratones Endogámicos C57BL , Femenino , Inmunoterapia/métodos , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Ovalbúmina/química , Línea Celular Tumoral , Ratones Endogámicos BALB C , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Nanopartículas Capa por Capa
2.
ACS Nano ; 18(4): 2841-2860, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38251849

RESUMEN

Manganese ions (Mn2+)-coordinated nanoparticles have emerged as a promising class of antitumor nanotherapeutics, capable of simultaneously disrupting the immunosuppressive tumor microenvironment (TME) and triggering the stimulator of interferon genes (STING) pathway-dependent antitumor immunity. However, the activation of STING signaling by Mn2+-based monotherapies is suboptimal for comprehensive stimulation of antigen presenting cells and reversal of immunosuppression in the TME. Here, we report the design of a Mn2+/CpG oligodeoxynucleotides (ODNs) codecorated black phosphorus nanosheet (BPNS@Mn2+/CpG) platform based on the Mn2+ modification of BPNS and subsequent adsorption of synthetic CpG ODNs. The coordination of Mn2+ significantly improved the stability of BPNS and the adsorption of CpG ODNs. The acidic TME and endosomal compartments can disrupt the Mn2+ coordination, triggering pH-responsive release of CpG ODNs and Mn2+ to effectively activate the Toll-like receptor 9 and STING pathways. As a result, M2-type macrophages and immature dendritic cells were strongly stimulated in the TME, thereby increasing T lymphocyte infiltration and reversing the immunosuppression within the TME. Phototherapy and chemodynamic therapy, utilizing the BPNS@Mn2+/CpG platform, have demonstrated efficacy in inducing immunogenic cell death upon 808 nm laser irradiation. Importantly, the treatment of BPNS@Mn2+/CpG with laser irradiation exhibited significant therapeutic efficacy against the irradiated primary tumor and effectively suppressed the growth of nonirradiated distant tumor. Moreover, it induced a robust immune memory, providing long-lasting protection against tumor recurrence. This study demonstrated the enhanced antitumor potency of BPNS@Mn2+/CpG in multimodal therapy, and its proof-of-concept application as a metal ion-modified BPNS material for effective DNA/drug delivery and immunotherapy.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Oligodesoxirribonucleótidos/farmacología , Terapia Combinada , Inmunoterapia , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
3.
ACS Nano ; 17(6): 5340-5353, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36913671

RESUMEN

Cancer immunotherapy, such as the Toll-like receptor (TLR) agonist including CpG oligodeoxynucleotide, has shown potency in clinical settings. However, it is still confronted with multiple challenges, which include the limited efficacy and severe adverse events caused by the rapid clearance and systemic diffusion of CpG. Here we report an improved CpG-based immunotherapy approach composed of a synthetic extracellular matrix (ECM)-anchored DNA/peptide hybrid nanoagonist (EaCpG) via (1) a tailor designed DNA template that encodes tetramer CpG and additional short DNA moieties, (2) generation of elongated multimeric CpG through rolling circle amplification (RCA), (3) self-assembly of densely packaged CpG particles composed of tandem CpG building blocks and magnesium pyrophosphate, and (4) incorporation of multiple copies of ECM binding peptide through hybridization to short DNA moieties. The structurally well-defined EaCpG shows dramatically increased intratumoral retention and marginal systemic dissemination through peritumoral administration, leading to potent antitumor immune response and subsequent tumor elimination, with minimal treatment-related toxicity. Combined with conventional standard-of-care therapies, peritumor administration of EaCpG generates systemic immune responses that lead to a curative abscopal effect on distant untreated tumors in multiple cancer models, which is superior to the unmodified CpG. Taken together, EaCpG provides a facile and generalizable strategy to simultaneously potentiate the potency and safety of CpG for combinational cancer immunotherapies.


Asunto(s)
Neoplasias , Humanos , Animales , Ratones , Neoplasias/tratamiento farmacológico , Oligodesoxirribonucleótidos/farmacología , Adyuvantes Inmunológicos , Inmunoterapia , ADN , Receptores Toll-Like , Receptor Toll-Like 9/agonistas , Ratones Endogámicos C57BL
4.
Pharm Res ; 40(1): 145-156, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36002611

RESUMEN

PURPOSE: Hepatitis B virus (HBV) infection is such a global health problem that hundreds of millions of people are HBV carriers. Current anti-viral agents can inhibit HBV replication, but can hardly eradicate HBV. Cytosine-phosphate-guanosine (CpG) oligodeoxynucleotides (ODNs) are an adjuvant that can activate plasmacytoid dendritic cells (pDCs) and conventional dendritic cells (cDCs) to induce therapeutic immunity for HBV eradication. However, efficient delivery of CpG ODNs into pDCs and cDCs remains a challenge. In this study, we constructed a series of cationic lipid-assisted nanoparticles (CLANs) using different cationic lipids to screen an optimal nanoparticle for delivering CpG ODNs into pDCs and cDCs. METHODS: We constructed different CLANCpG using six cationic lipids and analyzed the cellular uptake of different CLANCpG by pDCs and cDCs in vitro and in vivo, and further analyzed the efficiency of different CLANCpG for activating pDCs and cDCs in both wild type mice and HBV-carrier mice. RESULTS: We found that CLAN fabricated with 1,2-Dioleoyl-3-trimethylammonium propane (DOTAP) showed the highest efficiency for delivering CpG ODNs into pDCs and cDCs, resulting in strong therapeutic immunity in HBV-carrier mice. By using CLANCpG as an immune adjuvant in combination with the injection of recombinant hepatitis B surface antigen (rHBsAg), HBV was successfully eradicated and the chronic liver inflammation in HBV-carrier mice was reduced. CONCLUSION: We screened an optimized CLAN fabricated with DOTAP for efficient delivery of CpG ODNs to pDCs and cDCs, which can act as a therapeutic vaccine adjuvant for treating HBV infection.


Asunto(s)
Hepatitis B , Nanopartículas , Ratones , Animales , Virus de la Hepatitis B , Oligodesoxirribonucleótidos/farmacología , Fosfatos , Citosina , Guanosina , Hepatitis B/tratamiento farmacológico , Ácidos Grasos Monoinsaturados , Adyuvantes Inmunológicos/uso terapéutico , Células Dendríticas
5.
Oncol Res ; 32(2): 309-323, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38186581

RESUMEN

The Myc gene is the essential oncogene in triple-negative breast cancer (TNBC). This study investigates the synergistic effects of combining Myc decoy oligodeoxynucleotides-encapsulated niosomes-selenium hybrid nanocarriers with X-irradiation exposure on the MDA-MB-468 cell line. Decoy and scramble ODNs for Myc transcription factor were designed and synthesized based on promoter sequences of the Bcl2 gene. The nanocarriers were synthesized by loading Myc ODNs and selenium into chitosan (Chi-Se-DEC), which was then encapsulated in niosome-nanocarriers (NISM@Chi-Se-DEC). FT-IR, DLS, FESEM, and hemolysis tests were applied to confirm its characterization and physicochemical properties. Moreover, cellular uptake, cellular toxicity, apoptosis, cell cycle, and scratch repair assays were performed to evaluate its anticancer effects on cancer cells. All anticancer assessments were repeated under X-ray irradiation conditions (fractionated 2Gy). Physicochemical characteristics of niosomes containing SeNPs and ODNs showed that it is synthesized appropriately. It revealed that the anticancer effect of NISM@Chi-Se-DEC can be significantly improved in combination with X-ray irradiation treatment. It can be concluded that NISM@Chi-Se-DEC nanocarriers have the potential as a therapeutic agent for cancer treatment, particularly in combination with radiation therapy and in-vivo experiments are necessary to confirm the efficacy of this nano-drug.


Asunto(s)
Neoplasias de la Mama , Selenio , Humanos , Femenino , Rayos X , Neoplasias de la Mama/genética , Neoplasias de la Mama/radioterapia , Liposomas , Espectroscopía Infrarroja por Transformada de Fourier , Oligodesoxirribonucleótidos/farmacología
6.
Int J Mol Sci ; 22(13)2021 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-34206987

RESUMEN

Hepatitis C virus (HCV) is one of the main triggers of chronic liver disease. Despite tremendous progress in the HCV field, there is still no vaccine against this virus. Potential vaccines can be based on its recombinant proteins. To increase the humoral and, especially, cellular immune response to them, more effective adjuvants are needed. Here, we evaluated a panel of compounds as potential adjuvants using the HCV NS5B protein as an immunogen. These compounds included inhibitors of polyamine biosynthesis and urea cycle, the mTOR pathway, antioxidants, and cellular receptors. A pronounced stimulation of cell proliferation and interferon-γ (IFN-γ) secretion in response to concanavalin A was shown for antioxidant N-acetylcysteine (NAC), polyamine biosynthesis inhibitor 2-difluoromethylornithine (DFMO), and TLR9 agonist CpG ODN 1826 (CpG). Their usage during the immunization of mice with the recombinant NS5B protein significantly increased antibody titers, enhanced lymphocyte proliferation and IFN-γ production. NAC and CpG decreased relative Treg numbers; CpG increased the number of myeloid-derived suppressor cells (MDSCs), whereas neither NAC nor DFMO affected MDSC counts. NAC and DFMO suppressed NO and interleukin 10 (IL-10) production by splenocytes, while DFMO increased the levels of IL-12. This is the first evidence of immunomodulatory activity of NAC and DFMO during prophylactic immunization against infectious diseases.


Asunto(s)
Acetilcisteína/farmacología , Adyuvantes Inmunológicos/farmacología , Eflornitina/farmacología , Hepatitis C/inmunología , Inmunidad Activa/efectos de los fármacos , Proteínas no Estructurales Virales/inmunología , Animales , Proliferación Celular , Células Cultivadas , Femenino , Inmunogenicidad Vacunal/efectos de los fármacos , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Ratones , Ratones Endogámicos DBA , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/inmunología , Óxido Nítrico/metabolismo , Oligodesoxirribonucleótidos/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Vacunas contra Hepatitis Viral/inmunología
7.
Sci Rep ; 11(1): 6267, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33737644

RESUMEN

Antigen-adjuvant conjugation is known to enhance antigen-specific T-cell production in vaccine models, but scalable methods are required to generate site-specific conjugation for clinical translation of this technique. We report the use of the cell-free protein synthesis (CFPS) platform as a rapid method to produce large quantities (> 100 mg/L) of a model antigen, ovalbumin (OVA), with site-specific incorporation of p-azidomethyl-L-phenylalanine (pAMF) at two solvent-exposed sites away from immunodominant epitopes. Using copper-free click chemistry, we conjugated CpG oligodeoxynucleotide toll-like receptor 9 (TLR9) agonists to the pAMF sites on the mutant OVA protein. The OVA-CpG conjugates demonstrate enhanced antigen presentation in vitro and increased antigen-specific CD8+ T-cell production in vivo. Moreover, OVA-CpG conjugation reduced the dose of CpG needed to invoke antigen-specific T-cell production tenfold. These results highlight how site-specific conjugation and CFPS technology can be implemented to produce large quantities of covalently-linked antigen-adjuvant conjugates for use in clinical vaccines.


Asunto(s)
Adyuvantes Inmunológicos/metabolismo , Presentación de Antígeno , Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Proteínas Mutantes/inmunología , Oligodesoxirribonucleótidos/inmunología , Ovalbúmina/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos/genética , Sistema Libre de Células , Química Clic/métodos , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Oligodesoxirribonucleótidos/metabolismo , Oligodesoxirribonucleótidos/farmacología , Ovalbúmina/genética , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Transfección , Vacunación/métodos , Vacunas Conjugadas/administración & dosificación , Vacunas Conjugadas/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología
8.
Front Immunol ; 11: 1725, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193290

RESUMEN

Background and Objectives: The live non-pathogenic Leishmania tarantolae has recently provided a promising approach as an effective vaccine candidate against experimental leishmaniasis (ILL). Here, we evaluated the immunoprotective potential of the live Iranian Lizard Leishmania mixed with CpG adjuvant against L. major infection in BALB/c mice. Methods: Four groups of female BALB/c mice were included in the study. The first and second groups received PBS and CpG, respectively. The immunized groups received 2 × 105 ILL promastigotes and the CpG-mixed ILL (ILL+CpG). Injections were performed subcutaneously in the right footpad. Three weeks later, all mice were challenged with 2 × 105 metacyclic promastigotes of Leishmania majorEGFP ; inoculation was done in the left footpad. The measurement of footpad swelling and in vivo fluorescent imaging were used to evaluate disease progress during infection course. Eight weeks after challenge, all mice were sacrificed and the cytokines levels (IFN-γ, IL-4, and IL-10) and sera antibodies concentrations (IgG2a and IgG1) using ELISA assay, nitric oxide production using Griess assay, and arginase activity in cultured splenocytes, were measured. In addition, direct fluorescent microscopy analysis and qPCR assay were used to quantify the splenic parasite burden. Result: The results showed that mice immunized with ILL+CpG were protected against the development of the dermal lesion. Moreover, they showed a significant reduction in the parasite load, in comparison to the control groups. The observed protection was associated with higher production of IFN-γ, as well as a reduction in IL-4 level. Additionally, the results demonstrated that arginase activity was decreased in ILL+CpG group compared to other groups. Conclusion: Immunization using ILL+CpG induces a protective immunity; indicating that ILL with an appropriate adjuvant would be a suitable choice for vaccination against leishmaniasis.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Leishmania major/inmunología , Vacunas contra la Leishmaniasis/farmacología , Leishmaniasis Cutánea/prevención & control , Lagartos/parasitología , Oligodesoxirribonucleótidos/farmacología , Piel/efectos de los fármacos , Vacunas Vivas no Atenuadas/farmacología , Animales , Anticuerpos Antiprotozoarios/sangre , Arginasa/metabolismo , Células Cultivadas , Citocinas/sangre , Modelos Animales de Enfermedad , Femenino , Inmunización , Inmunogenicidad Vacunal , Leishmaniasis Cutánea/sangre , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/parasitología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/parasitología , Ratones Endogámicos BALB C , Carga de Parásitos , Piel/inmunología , Piel/parasitología , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/metabolismo , Bazo/parasitología , Vacunas Vivas no Atenuadas/inmunología
9.
Int J Nanomedicine ; 15: 5927-5949, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32848397

RESUMEN

PURPOSE: A multi-functional nanoplatform with diagnostic imaging and targeted treatment functions has aroused much interest in the nanomedical research field and has been paid more attention in the field of tumor diagnosis and treatment. However, some existing nano-contrast agents have encountered difficulties in different aspects during clinical promotion, such as complicated preparation process and low specificity. Therefore, it is urgent to find a nanocomplex with good targeting effect, high biocompatibility and significant therapeutic effect for the integration of diagnosis and treatment and clinical transformation. MATERIALS AND METHODS: Nanoparticles (NPs) targeting breast cancer were synthesized by phacoemulsification which had liquid fluorocarbon perfluoropentane(PFP) in the core and were loaded with Iron(II) phthalocyanine (FePc) on the shell. The aptamer (APT) AS1411 was outside the shell used as a molecular probe. Basic characterization and targeting abilities of the NPs were tested, and their cytotoxicity and biological safety in vivo were evaluated through CCK-8 assay and blood bio-chemical analysis. The photoacoustic (PA) and ultrasound (US) imaging system were used to assess the effects of AS1411-PLGA@FePc@PFP (A-FP NPs) as dual modal contrast agent in vitro and in vivo. The effects of photothermal therapy (PTT) in vitro and in vivo were evaluated through MCF-7 cells and tumor-bearing nude mouse models. RESULTS: A-FP NPs, with good stability, great biocompatibility and low toxicity, were of 201.87 ± 1.60 nm in diameter, and have an active targeting effect on breast cancer cells and tissues. With the help of PA/US imaging, it was proved to be an excellent dual modal contrast agent for diagnosis and guidance of targeted therapy. Meanwhile, it can heat up under near-infrared (NIR) laser irradiation and has achieved obvious antitumor effect both in vitro and in vivo experiments. CONCLUSION: As a kind of nanomedicine, A-FP NPs can be used in the integration of diagnosis and treatment. The treatment effects and biocompatibility in vivo may provide new thoughts in the clinical transformation of nanomedicine and early diagnosis and treatment of breast cancer.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/terapia , Indoles/química , Nanopartículas Multifuncionales/química , Oligodesoxirribonucleótidos/farmacología , Animales , Aptámeros de Nucleótidos/administración & dosificación , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/farmacología , Neoplasias de la Mama/patología , Medios de Contraste/química , Femenino , Fluorocarburos/química , Humanos , Hierro/química , Isoindoles , Células MCF-7 , Ratones Endogámicos BALB C , Nanopartículas Multifuncionales/administración & dosificación , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/química , Fototerapia/métodos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Ultrasonografía , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Biosci Biotechnol Biochem ; 84(9): 1846-1855, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32501144

RESUMEN

Here, we prepared the novel combined adjuvants, CTB as intra-molecular adjuvant, CpG and aluminum hydroxide (Alum) to strengthen the immunogenicity of clumping factor A221-550 of Staphylococcus aureus (S. aureus). The protein-immunoactive results showed CTB-ClfA221-550 elicited the strong immune responses to serum from mice immunized with CTB and ClfA221-550, respectively. The mice immunized with CTB-ClfA221-550 plus CpG and Alum adjuvant exhibited significantly stronger CD4+ T cell responses for IFN-γ, IL-2, IL-4, and IL-17 and displayed the higher proliferation response of splenic lymphocytes than the control groups, in addition, these mice generated the strongest humoral immune response against ClfA221-550 among all groups. Our results also showed CTB-ClfA221-550 plus CpG and Alum adjuvant obviously increased the survival percentage of the mice challenged by S. aureus. These data suggested that the novel combined adjuvants, CTB, CpG, and Alum, significantly enhance the immune responses triggered with ClfA221-550, and could provide a new approach against infection of S. aureus. ABBREVIATIONS: CTB: Cholera Toxin B; CpG: Cytosine preceding Guanosine; ODN: Oligodeoxynucleotides; Alum: Aluminum hydroxide; TRAP: Target of RNAIII-activating Protein; TLR9: Toll-like Receptor 9; TMB: 3, 3', 5, 5'-tetramethylbenzidine; mAbs: Monoclonal Antibodies; OD: Optical Densities; S. aureus: Staphylococcus aureus; ClfA: Clumping factor A; FnBPA: Fibronection-binding protein A; IsdB: Iron-regulated surface determinant B; SasA: Staphylococcus aureus Surface Protein A; GapC: Glycer-aldehyde-3-phosphate dehydrogenase-C.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Hidróxido de Aluminio/farmacología , Toxina del Cólera/farmacología , Coagulasa/inmunología , Animales , Proliferación Celular/efectos de los fármacos , Interacciones Farmacológicas , Inmunización , Linfocitos/citología , Linfocitos/efectos de los fármacos , Ratones , Oligodesoxirribonucleótidos/farmacología
11.
Tuberculosis (Edinb) ; 121: 101890, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32279869

RESUMEN

CpG motifs in DNA sequences are recognized by Toll-like receptor 9 and activate immune cells. Bacterial genomic DNA (gDNA) has modified cytosine bases (5-methylcytosine [5 mC]) and modified adenine bases (6-methyladenine [6 mA]). 5 mC inhibits immune activation by CpG DNA; however, it is unclear whether 6 mA inhibits immune activation by CpG DNA. Mycobacterium bovis BCG (BCG) has three adenine methyltransferases (MTases) that act on specific target sequences. In this study, we examined whether the 6 mA at the target sites of adenine MTases affected the immunostimulatory activity of CpG DNA. Our results showed that only 6 mA located at the target sequence of mamA, an adenine MTase from BCG, enhanced interleukin (IL)-12p40 production from murine bone marrow-derived macrophages (BMDMs) stimulated with CpG DNA. Enhancement of IL-12p40 production in BMDMs was also observed when BMDMs were stimulated with CpG DNA ligated to oligodeoxynucleotides (ODNs) harboring 6 mA. Accordingly, we then evaluated whether gDNA from adenine MTase-deficient BCG was less efficient with regard to stimulation of BMDMs. Indeed, gDNA from a mamA-deficient BCG had less ability to activate BMDMs than that from wild-type BCG. We concluded from these results that adenine methylation on ODNs and bacterial gDNA may enhance immune activity induced by CpG DNA.


Asunto(s)
Adenina/análogos & derivados , Adyuvantes Inmunológicos/farmacología , ADN Bacteriano/inmunología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Metiltransferasas/inmunología , Mycobacterium bovis/inmunología , Oligodesoxirribonucleótidos/farmacología , Receptor Toll-Like 9/agonistas , Adenina/inmunología , Animales , Células Cultivadas , ADN Bacteriano/genética , Interacciones Huésped-Patógeno , Subunidad p40 de la Interleucina-12/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Metiltransferasas/deficiencia , Metiltransferasas/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium bovis/enzimología , Mycobacterium bovis/genética , Transducción de Señal , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo
12.
Nanoscale ; 12(6): 3916-3930, 2020 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-32003377

RESUMEN

Palladium nanosheets (Pd NSs) have recently attracted increasing research interest in the biomedical field due to their excellent near-infrared absorption, photothermal conversion capability and biocompatibility. However, the application of Pd NSs in immunotherapy has not been reported. Here, Pd NSs were used as the carriers of immunoadjuvant CpG ODNs for not only efficient delivery of CpG but also for enhancing the immunotherapeutic effects of CpG by the Pd NS-based photothermal therapy (PTT). Pd NSs had no influence on the immune system, and the prepared Pd-CpG nanocomposites, especially Pd(5)-CpG(PS), could significantly increase the uptake of CpG by immune cells and enhance the immunostimulatory activity of CpG in vitro and in vivo. With the combination of Pd(5)-CpG(PS) mediated PTT and immunotherapy, highly efficient tumor inhibition was achieved and the survival rate of the tumor-bearing mice was greatly increased depending on Pd(5)-CpG(PS) with safe near-infrared (NIR) irradiation (808 nm laser, 0.15 W cm-2). Importantly, the combination therapy induced tumor cell death and released tumor-associated antigens, which could be effectively taken up and presented by antigen presenting cells with the assistance of CpG, leading to increased TNF-α and IL-6 production and enhanced cytotoxic T lymphocyte (CTL) activity. This work provides a new paradigm of utilizing photothermal nanomaterials for safe and highly efficient cancer photothermal combined immunotherapy.


Asunto(s)
Antineoplásicos , Inmunoterapia/métodos , Nanocompuestos/química , Oligodesoxirribonucleótidos , Fototerapia/métodos , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacocinética , Adyuvantes Inmunológicos/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Nanomedicina/métodos , Neoplasias Experimentales/terapia , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/farmacocinética , Oligodesoxirribonucleótidos/farmacología , Paladio/química , Células RAW 264.7
13.
Nanoscale ; 11(42): 19912-19922, 2019 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-31599915

RESUMEN

The metastasis and recurrence of tumors are the main reasons for cancer death. In this work, a promising therapy for tumor treatment that can eliminate primary tumors and prevent tumor relapses is introduced by combining chemotherapy, photothermal therapy (PTT) and immunotherapy. Multifunctional FePt/MoS2-FA nanocomposites (FPMF NCs) were obtained via anchoring FePt nanoparticles and folic acid (FA) on MoS2 nanosheets. As an efficient ferroptosis agent, FePt nanoparticles could catalyze the Fenton reaction to produce the reactive oxygen species (ROS). Through the highly effective photothermal conversion of MoS2 nanosheets, the primary tumor cells could be ablated by photothermal therapy (PTT). Moreover, the metastatic tumors were eliminated effectively with the help of oligodeoxynucleotides containing cytosine-guanine (CpG ODNs) combined with systemic checkpoint blockade therapy using an anti-CTLA4 antibody. Even more intriguingly, a strong immunological memory effect was obtained by this synergistic therapy. Taking all these results into consideration, we anticipate that the photo-chemo-immunotherapy strategies show great promise toward the development of a multifunctional platform for anticancer therapeutic applications.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Hipertermia Inducida , Nanopartículas del Metal , Nanocompuestos , Neoplasias Experimentales/terapia , Oligodesoxirribonucleótidos/farmacología , Fototerapia , Microambiente Tumoral/efectos de los fármacos , Animales , Ácido Fólico/química , Ácido Fólico/farmacología , Células HeLa , Humanos , Inmunoterapia , Hierro/química , Hierro/farmacología , Células MCF-7 , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Ratones , Nanocompuestos/química , Nanocompuestos/uso terapéutico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Platino (Metal)/química , Platino (Metal)/farmacología , Microambiente Tumoral/inmunología
14.
Biomater Sci ; 6(12): 3388-3396, 2018 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-30371689

RESUMEN

Cells can communicate with one another through physical connections and chemical signaling, activating various signaling pathways that can affect cellular functions and behaviors. In taste buds, taste cells transmit taste information to neurons via paracrine signaling. However, no previous studies have reported the in vitro co-culture of taste and neuronal cells, which allows us to monitor intercellular communications and better understand the mechanism of taste perception. Here, we introduce the first investigation on the proximate assembly and co-culture of taste cells and neurons to monitor the intercellular transmission of taste signals. Taste cells and neurons are placed closely using a pair of single-stranded oligonucleotides conjugated with polyethylene glycol and a phospholipid. Complementary oligonucleotide conjugates are anchored into the cellular membrane of neonatal taste cells and embryonic hippocampal neuronal cells, respectively, and then the cells are self-assembled into a functional multicellular unit for taste perception. Treatment of the assembled cells with a bitter tastant generates the sequential influx of calcium ions into the cytoplasm in taste cells and then in neuronal cells. Our work demonstrates that the cellular self-assembly is critical for efficient taste signal transduction, which can be used as a promising platform to construct cell-based biosensors for taste sensing.


Asunto(s)
Comunicación Celular , Transducción de Señal , Papilas Gustativas/citología , Animales , Técnicas Biosensibles/métodos , Células Cultivadas , Técnicas de Cocultivo/métodos , Hipocampo/citología , Ratones , Oligodesoxirribonucleótidos/farmacología , Papilas Gustativas/efectos de los fármacos , Papilas Gustativas/fisiología , Percepción del Gusto
15.
Nanoscale ; 10(30): 14516-14524, 2018 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-30024003

RESUMEN

CpG oligodeoxynucleotides (CpG ODNs) possess strong immunostimulatory activity, which hold great promise in cancer immunotherapy. However, their therapeutic efficacy is largely limited due to nuclease degradation and poor cellular internalization. Efficiently delivering CpG ODNs into target cells is crucial to improve their therapeutic efficacy. Boron nitride nanospheres (BNNS) possess advantage as carriers for CpG ODNs. However, their poor aqueous dispersity and low CpG ODN loading capacity became a big obstacle for further applications. Herein, we develop amino group grafted, mesoporous silica (MS)-functionalized BNNS as novel nanovectors for CpG ODN delivery. Modification of BNNS with MS significantly improved the dispersity of BNNS and CpG ODN loading. BNNS@MS-NH2 exhibited no cytotoxicity and enhanced the delivery of CpG ODNs into macrophages. BNNS@MS-NH2/CpG ODN complexes triggered enhanced immunostimulation and induced higher amounts of cytokines. Most importantly, BNNS@MS-NH2/CpG ODN complexes induced bifurcated cytokines, which simultaneously simulated the secretion of IL-6, TNF-α and IFN-α. In contrast, CpG ODN and BNNS/CpG ODN complexes could not. The result of the Transwell plate assay suggested that BNNS@MS-NH2/CpG ODN complexes were more effective in inhibiting cancer cell growth. Taken together, our findings provide a promising strategy for enhancing CpG ODN-mediated cancer immunotherapy.


Asunto(s)
Sistemas de Liberación de Medicamentos , Inmunoterapia , Nanosferas/química , Oligodesoxirribonucleótidos/farmacología , Dióxido de Silicio/química , Adyuvantes Inmunológicos , Animales , Línea Celular Tumoral , Interferón-alfa/sangre , Interleucina-6/sangre , Macrófagos/efectos de los fármacos , Ratones , Células RAW 264.7 , Factor de Necrosis Tumoral alfa/sangre
16.
Theranostics ; 8(8): 2229-2241, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29721075

RESUMEN

Photothermal therapy (PTT) can be an effective antitumor therapy, but it may not completely eliminate tumor cells, leading to the risk of recurrence or metastasis. Here we describe nanocarriers that allow combination therapy involving PTT and immunotherapy. Nanocarriers are prepared by coating Al2O3 nanoparticles with non-toxic, biodegradable polydopamine, which shows high photothermal efficiency. A near-infrared laser irradiation can kill the majority of tumor tissues, resulting in the release of tumor-associated antigens. The Al2O3 within the nanoparticles, together with CpG, acts as an adjuvant to trigger robust cell-mediated immune responses that can help eliminate the residual tumor cells and reduce the risk of tumor recurrence. Methods: The characteristics and photothermal performance of polydopamine-coated Al2O3 nanoparticles were examined after one-step preparation. Then we studied their internalization, photothermal toxicity and immunostimulatory activity in vitro. For in vivo experiments, these nanocarriers were injected directly into B16F10 melanoma allografts in mice to ensure specific localization. After photothermal irradiation on day 0, mice were subcutaneously injected with CpG adjuvant on day 1, 3 and 5. Tumor volumes and number of living mice were recorded every two days. Moreover, various immune responses induced by our combined therapy were tested for mechanism research. Results: 50% of mice after our combined treatment successfully achieved the goal of tumor eradication, and survived for 120 days, which was the end point of the experiment. Mechanism studies demonstrated the combined therapy efficiently led to dendritic cell maturation, resulting in the secretion of antibodies and cytokines as well as the proliferation of splenocytes and lymphocytes for anti-tumor immunotherapy. Conclusion: Taken together, these results demonstrated the promise of our combined photothermal therapy and immunotherapy for tumor shrinkage, which merited further research.


Asunto(s)
Óxido de Aluminio/química , Hipertermia Inducida , Inmunoterapia , Indoles/química , Melanoma Experimental/terapia , Nanopartículas/química , Fototerapia , Polímeros/química , Adyuvantes Inmunológicos/farmacología , Animales , Muerte Celular , Terapia Combinada , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Endocitosis , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Nanopartículas/toxicidad , Nanopartículas/ultraestructura , Oligodesoxirribonucleótidos/farmacología
17.
Pharm Res ; 35(4): 80, 2018 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-29500548

RESUMEN

PURPOSE: Synthetic oligodeoxynucleotides (ODN) containing unmethylated CpG motifs were found to be able to target cells that express Toll-like receptor 9 to modulate innate and adaptive immune reactions. But their in vivo application in immunotherapy against cancer has not been successful. We attempted in this study to examine polyethylene-glycol (PEG) conjugated CpG ODNs and investigated their mechanism of immune modulation in anti-cancer therapy. METHODS: CpG-PEG conjugates with different PEG lengths were synthesized. In vitro activity as well as in vivo pharmacokinetics and pharmacodynamics properties were evaluated. RESULTS: CpG-PEG20Ks were found to be able to persist longer in circulation and activate various downstream effector cells. After intravenous injection, they resulted in higher levels of IL-12p70 in the circulation and lower M-MDSC infiltrates in the tumor microenvironment. Such activities were different from those of CpG ODNs without PEGylation, suggesting different PK-PD profiles systemically and locally. CONCLUSIONS: Our data support the development of CpG-PEGs as a new therapeutic agent that can be systemically administered to modulate immune responses and the microenvironment in tumor tissues.


Asunto(s)
Inmunidad Adaptativa/efectos de los fármacos , Composición de Medicamentos/métodos , Neoplasias/tratamiento farmacológico , Oligodesoxirribonucleótidos/farmacología , Animales , Línea Celular Tumoral/trasplante , Células Cultivadas , Células Dendríticas , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Inyecciones Intravenosas , Interleucina-12/inmunología , Interleucina-12/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias/inmunología , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/uso terapéutico , Ovalbúmina/inmunología , Polietilenglicoles/química , Cultivo Primario de Células , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
18.
Int J Nanomedicine ; 13: 43-62, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29317815

RESUMEN

Conventional class B cytosine-guanine (CpG) (CpG-B) oligodeoxynucleotide (ODNs) consisting of a single-stranded (ss) phosphorothioate (PT) backbone (ss CpG-B-PT) is converted from a proinflammatory cytokine inducer to a type-I interferon (IFN) inducer when complexed with cationic materials. In this study, we designed ss CpG-B and double-stranded (ds) CpG-B ODNs with a phosphodiester (PD) backbone (ss CpG-B-PD and ds CpG-B-PD, respectively) that became type-I IFN inducers upon complexation with Lipofectamine 2000 (Lipo), a cationic liposome. The ds CpG-B-PD complex induced higher IFN-ß expression in mouse macrophage-like RAW264 cells than ss CpG-B-PD and ss CpG-B-PT complexes. The fold induction of IFN-ß increased with the number of CpG motifs in ds CpG-B-PD, and a complex of ds CpG-B-PD consisting of 72 base pairs with nine CpG motifs (ds CpG-B72-PD) and Lipo showed the highest capacity to induce IFN-ß. The materials and method used for complexation influenced the degree of IFN-ß induction: ds CpG-B72-PD entrapped by calcium phosphate (CaP) (ds CpG-B72-PD/CaP) showed a higher induction capacity than ds CpG-B72-PD adsorbed onto the CaP surface. Entrapment of ds CpG-B72-PD by CaP also enhanced the induction of the proinflammatory cytokine interleukin-12. Vaccinating mice with ds CpG-B72-PD/CaP in conjunction with ovalbumin (OVA) increased the ratios of OVA-specific CD8+ T cells to total CD8+ T cells in peripheral blood and of OVA-specific IgG2a associated with helper T (Th)1 cells to OVA-specific IgG1 associated with Th2 cells. These results indicate that ds CpG-B72-PD/CaP is an effective vaccine adjuvant that can activate both cellular and Th1-type humoral immune responses.


Asunto(s)
Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacología , Inmunidad Humoral/efectos de los fármacos , Oligodesoxirribonucleótidos/farmacología , Células TH1/inmunología , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Fosfatos de Calcio/química , Línea Celular , Sistemas de Liberación de Medicamentos/métodos , Inmunoglobulina G/sangre , Interferón beta/genética , Interferón beta/metabolismo , Interleucina-12/sangre , Liposomas/administración & dosificación , Liposomas/química , Ratones , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/inmunología , Ovalbúmina/inmunología , Ovalbúmina/farmacología , Células TH1/efectos de los fármacos , Vacunas/inmunología
19.
Toxicology ; 393: 102-112, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29129814

RESUMEN

Cav3.2 T-type Ca2+ channel activity is suppressed by zinc that binds to the extracellular histidine-191 of Cav3.2, and enhanced by H2S that interacts with zinc. Cav3.2 in nociceptors is upregulated in an activity-dependent manner. The enhanced Cav3.2 activity by H2S formed by the upregulated cystathionine-γ-lyase (CSE) is involved in the cyclophosphamide (CPA)-induced cystitis-related bladder pain in mice. We thus asked if zinc deficiency affects the cystitis-related bladder pain in mice by altering Cav3.2 function and/or expression. Dietary zinc deficiency for 2 weeks greatly decreased zinc concentrations in the plasma but not bladder tissue, and enhanced the bladder pain/referred hyperalgesia (BP/RH) following CPA at 200mg/kg, a subeffective dose, but not 400mg/kg, a maximal dose, an effect abolished by pharmacological blockade or gene silencing of Cav3.2. Acute zinc deficiency caused by systemic N,N,N',N'-tetrakis-(2-pyridylmethyl)-ethylendiamine (TPEN), a zinc chelator, mimicked the dietary zinc deficiency-induced Cav3.2-dependent promotion of BP/RH following CPA at 200mg/kg. CPA at 400mg/kg alone or TPEN plus CPA at 200mg/kg caused Cav3.2 overexpression accompanied by upregulation of Egr-1 and USP5, known to promote transcriptional expression and reduce proteasomal degradation of Cav3.2, respectively, in the dorsal root ganglia (DRG). The CSE inhibitor, ß-cyano-l-alanine, prevented the BP/RH and upregulation of Cav3.2, Egr-1 and USP5 in DRG following TPEN plus CPA at 200mg/kg. Together, zinc deficiency promotes bladder pain accompanying CPA-induced cystitis by enhancing function and expression of Cav3.2 in nociceptors, suggesting a novel therapeutic avenue for treatment of bladder pain, such as zinc supplementation.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Cistitis/metabolismo , Dolor/metabolismo , Zinc/deficiencia , Animales , Quelantes/farmacología , Ciclofosfamida , Cistationina gamma-Liasa/metabolismo , Cistitis/inducido químicamente , Dieta , Modelos Animales de Enfermedad , Etilaminas , Ganglios Espinales/metabolismo , Ratones , Oligodesoxirribonucleótidos/farmacología , Piridinas , Vejiga Urinaria/metabolismo , Zinc/sangre , Zinc/metabolismo
20.
Mol Ther ; 25(7): 1665-1675, 2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-28284981

RESUMEN

Exosomes are small membrane-bound vesicular particles generated by most cells for intercellular communication and regulation. During biogenesis, specific lipids, RNAs, proteins, and carbohydrates are enriched and packaged into the vesicles so that the exosomal contents reflect not only the source but also the physiological conditions of the parental cells. These exosomes transport materials or signals to the target cells for diverse physiological purposes. Our study focused on the exosomes derived from M1-polarized, proinflammatory macrophages for the possibility of using M1 exosomes as an immunopotentiator for a cancer vaccine. The M1 exosomes displayed a tropism toward lymph nodes after subcutaneous injection, primarily taken up by the local macrophages and dendritic cells, and they induced the release of a pool of Th1 cytokines. We found that M1, but not M2, exosomes enhanced activity of lipid calcium phosphate (LCP) nanoparticle-encapsulated Trp2 vaccine, and they induced a stronger antigen-specific cytotoxic T cell response. The M1 exosomes proved to be a more potent immunopotentiator than CpG oligonucleotide when used with LCP nanoparticle vaccine in a melanoma growth inhibition study. Thus, our study indicated that exosomes derived from M1-polarized macrophages could be used as a vaccine adjuvant.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Exosomas/química , Factores Inmunológicos/farmacología , Ganglios Linfáticos/efectos de los fármacos , Melanoma Experimental/terapia , Neoplasias Cutáneas/terapia , Adyuvantes Inmunológicos/farmacología , Animales , Fosfatos de Calcio/química , Portadores de Fármacos , Composición de Medicamentos , Exosomas/inmunología , Femenino , Expresión Génica , Factores Inmunológicos/química , Inflamación/inducido químicamente , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Nanopartículas/administración & dosificación , Nanopartículas/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Oligodesoxirribonucleótidos/farmacología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA