Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Pharm ; 16(6): 2376-2384, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-30951315

RESUMEN

Skeletal muscle is ideally suited and highly desirable as a target for therapeutic gene delivery because of its abundance, high vascularization, and high levels of protein expression. However, efficient gene delivery to skeletal muscle remains a current challenge. Besides the major obstacle of cell-specific targeting, efficient intracellular trafficking, or the cytosolic transport of DNA to the nucleus, must be demonstrated. To overcome the challenge of cell-specific targeting, herein we develop a generation 5-polyamidoamine dendrimer (G5-PAMAM) functionalized with a skeletal muscle-targeted peptide, ASSLNIA (G5-SMTP). Specifically, to demonstrate the feasibility of our approach, we prepared a complex of our G5-SMTP dendrimer with a plasmid encoding firefly luciferase and investigated its delivery to skeletal muscle cells. Luciferase assays indicated a threefold increase in transfection efficiency of C2C12 murine skeletal muscle cells using G5-SMTP when compared with nontargeting nanocarriers using unmodified G5. To further improve the transfection yield, we employed a cationic dynein light chain 8 protein (DLC8)-binding peptide (DBP) containing an internal sequence known to bind to the DLC8 of the dynein motor protein complex. Complexation of DBP with our targeting nanocarrier, that is, G5-SMTP, and our luciferase plasmid cargo resulted in a functional nanocarrier that showed an additional sixfold increase in transfection efficiency compared with G5-SMTP transfection alone. To our knowledge, this is the first successful use of two different functional nanocarrier components that enable targeted skeletal muscle cell recognition and increased efficiency of intracellular trafficking to synergistically enhance gene delivery to skeletal muscle cells. This strategy of targeting and trafficking can also be universally applied to any cell/tissue type for which a recognition domain exists.


Asunto(s)
Dendrímeros/química , Dineínas/química , Músculo Esquelético/metabolismo , Plásmidos/administración & dosificación , Animales , Línea Celular , Citoplasma/metabolismo , Dineínas Citoplasmáticas/metabolismo , Ratones , Plásmidos/genética
2.
J Immunol ; 198(10): 4166-4177, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28396317

RESUMEN

Myeloid cells play a key role in tumor progression and metastasis by providing nourishment and immune protection, as well as facilitating cancer invasion and seeding to distal sites. Although advances have been made in understanding the biology of these tumor-educated myeloid cells (TEMCs), their intrinsic plasticity challenges our further understanding of their biology. Indeed, in vitro experiments only mimic the in vivo setting, and current gene-knockout technologies do not allow the simultaneous, temporally controlled, and cell-specific silencing of multiple genes or pathways. In this article, we describe the 4PD nanoplatform, which allows the in vivo preferential transfection and in vivo tracking of TEMCs with the desired RNAs. This platform is based on the conjugation of CD124/IL-4Rα-targeting peptide with G5 PAMAM dendrimers as the loading surface and can convey therapeutic or experimental RNAs of interest. When injected i.v. in mice bearing CT26 colon carcinoma or B16 melanoma, the 4PD nanoparticles predominantly accumulate at the tumor site, transfecting intratumoral myeloid cells. The use of 4PD to deliver a combination of STAT3- and C/EBPß-specific short hairpin RNA or miR-142-3p confirmed the importance of these genes and microRNAs in TEMC biology and indicates that silencing of both genes is necessary to increase the efficacy of immune interventions. Thus, the 4PD nanoparticle can rapidly and cost effectively modulate and assess the in vivo function of microRNAs and mRNAs in TEMCs.


Asunto(s)
Dendrímeros/metabolismo , Silenciador del Gen , Células Mieloides/metabolismo , Nanotecnología/métodos , Animales , Línea Celular Tumoral , Neoplasias del Colon , Dendrímeros/administración & dosificación , Subunidad alfa del Receptor de Interleucina-4/inmunología , Subunidad alfa del Receptor de Interleucina-4/metabolismo , Melanoma Experimental , Ratones , MicroARNs , Células Mieloides/inmunología , Nanopartículas/administración & dosificación , Nanopartículas/metabolismo , Nanotecnología/normas , Receptores de Interleucina-4/inmunología , Receptores de Interleucina-4/metabolismo
3.
Anal Biochem ; 498: 1-7, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26772160

RESUMEN

Here we describe the design and construction of an imaging construct with high bioluminescent resonance energy transfer (BRET) efficiency that is composed of multiple quantum dots (QDs; λem = 655 nm) self-assembled onto a bioluminescent protein, Renilla luciferase (Rluc). This is facilitated by the streptavidin-biotin interaction, allowing the facile formation of a hybrid-imaging construct (HIC) comprising up to six QDs (acceptor) grafted onto a light-emitting Rluc (donor) core. The resulting assembly of multiple acceptors surrounding a donor permits this construct to exhibit high resonance energy transfer efficiency (∼64.8%). The HIC was characterized using fluorescence excitation anisotropy measurements and high-resolution transmission electron microscopy. To demonstrate the application of our construct, a generation-5 (G5) polyamidoamine dendrimer (PAMAM) nanocarrier was loaded with our HIC for in vitro and in vivo imaging. We envision that this design of multiple acceptors and bioluminescent donor will lead to the development of new BRET-based systems useful in sensing, imaging, and other bioanalytical applications.


Asunto(s)
Dendrímeros/química , Diseño de Fármacos , Transferencia de Energía , Luciferasas de Renilla/química , Mediciones Luminiscentes , Nanoestructuras/química , Puntos Cuánticos , Dendrímeros/síntesis química , Luciferasas de Renilla/metabolismo
4.
Clin Nephrol ; 83(2): 104-10, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24691016

RESUMEN

Anti-glomerular basement membrane (GBM) disease is a severe inflammatory renal disorder due to pathogenic autoantibodies directed mainly against the α3 chain of type IV collagen. In ~1/4 of patients with anti-GBM disease, antineutrophil cytoplasmic antibodies (ANCA) predominantly with myeloperoxidase (MPO) specificity can be detected. Although the inciting stimuli leading to the development of an immune response against the type IV collagen and neutrophils are unknown, evidence indicates that both genetic and environmental factors play a role. Of note, molecular mimicry between self-antigens and nonself-antigens such as antigenic determinants of microorganisms has been implicated in the pathogenesis of anti-GBM disease and ANCA-associated vasculitis. A mosquito-borne viral illness highly prevalent in the tropics and subtropics, dengue can be complicated by acute renal failure, proteinuria, hematuria and glomerulonephritis. We present a 66-year-old woman who was diagnosed with dengue infection and rapidly progressive glomerulonephritis during an outbreak of dengue in Honduras in the summer of 2013. Renal biopsy revealed severe crescentic glomerulonephritis. Immunofluorescence examination demonstrated strong linear IgG deposition along glomerular capillary walls. Serologic tests demonstrated antibodies against GBM, MPO and platelet glycoproteins. The patient was diagnosed with anti-GBM disease associated with p-ANCA with MPO specificity. Despite heavy immunosuppression and plasmapheresis, IgG titers against dengue virus continued to rise confirming the diagnosis of acute dengue infection. We present the first reported case of anti-GBM disease associated with p-ANCA with MPO specificity during dengue infection. This report calls for a heightened awareness of autoimmunity leading to crescentic glomerulonephritis in patients with dengue infection.


Asunto(s)
Enfermedad por Anticuerpos Antimembrana Basal Glomerular/virología , Anticuerpos Anticitoplasma de Neutrófilos/inmunología , Dengue/inmunología , Dengue/patología , Anciano , Enfermedad por Anticuerpos Antimembrana Basal Glomerular/inmunología , Anticuerpos Anticitoplasma de Neutrófilos/sangre , Femenino , Humanos
5.
J Infect Dis ; 208(11): 1914-22, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23901083

RESUMEN

BACKGROUND: Amphotericin B (AmB), the most effective drug against leishmaniasis, has serious toxicity. As Leishmania species are obligate intracellular parasites of antigen presenting cells (APC), an immunopotentiating APC-specific AmB nanocarrier would be ideally suited to reduce the drug dosage and regimen requirements in leishmaniasis treatment. Here, we report a nanocarrier that results in effective treatment shortening of cutaneous leishmaniasis in a mouse model, while also enhancing L. major specific T-cell immune responses in the infected host. METHODS: We used a Pan-DR-binding epitope (PADRE)-derivatized-dendrimer (PDD), complexed with liposomal amphotericin B (LAmB) in an L. major mouse model and analyzed the therapeutic efficacy of low-dose PDD/LAmB vs full dose LAmB. RESULTS: PDD was shown to escort LAmB to APCs in vivo, enhanced the drug efficacy by 83% and drug APC targeting by 10-fold and significantly reduced parasite burden and toxicity. Fortuitously, the PDD immunopotentiating effect significantly enhanced parasite-specific T-cell responses in immunocompetent infected mice. CONCLUSIONS: PDD reduced the effective dose and toxicity of LAmB and resulted in elicitation of strong parasite specific T-cell responses. A reduced effective therapeutic dose was achieved by selective LAmB delivery to APC, bypassing bystander cells, reducing toxicity and inducing antiparasite immunity.


Asunto(s)
Anfotericina B/administración & dosificación , Antiprotozoarios/administración & dosificación , Dendrímeros/administración & dosificación , Leishmania major/efectos de los fármacos , Leishmaniasis Cutánea/tratamiento farmacológico , Vacunas contra la Malaria/administración & dosificación , Inmunidad Adaptativa , Anfotericina B/toxicidad , Animales , Células Presentadoras de Antígenos/inmunología , Antiprotozoarios/toxicidad , Modelos Animales de Enfermedad , Portadores de Fármacos , Epítopos , Femenino , Inyecciones Intraperitoneales , Leishmania major/inmunología , Vacunas contra la Leishmaniasis , Leishmaniasis Cutánea/inmunología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Nanopartículas
6.
Vaccine ; 39(15): 2110-2116, 2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33744048

RESUMEN

The success of SARS-CoV-2 (CoV-2) vaccines is measured by their ability to mount immune memory responses that are long-lasting. To achieve this goal, it is important to identify surrogates of immune protection, namely, CoV-2 MHC Class I and II immunodominant pieces/epitopes and methodologies to measure them. Here, we present results of flow cytometry-based MHC Class I and II QuickSwitchTM platforms for assessing SARS-CoV-2 peptide binding affinities to various human alleles as well as the H-2 Kb mouse allele. Multiple SARS-CoV-2 potential MHC binders were screened and validated by QuickSwitch testing. The screen included 31 MHC Class I and 19 MHC Class II peptides predicted to be good binders by the IEDB web resource provided by NIAID. While several predicted peptides with acceptable theoretical Kd showed poor MHC occupancies, fourteen MHC class II and three MHC class I peptides showed promiscuity in that they bind to multiple MHC molecule types. In addition to providing important data towards the study of the SARS-CoV-2 virus and its presented antigenic epitopes, the peptides identified in this study can be used in the QuickSwitch platform to generate MHC tetramers. With those tetramers, scientists can assess CD4 + and CD8 + immune responses to these different MHC/peptide complexes.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , COVID-19/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Animales , Epítopos de Linfocito T/inmunología , Humanos , Epítopos Inmunodominantes/inmunología , Ratones , SARS-CoV-2/inmunología
7.
Sci Rep ; 11(1): 9561, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33953256

RESUMEN

Extracellular adenosine suppresses T cell immunity in the tumor microenvironment and in vitro treatment of memory T cells with adenosine can suppress antigen-mediated memory T cell expansion. We describe utilizing the recall antigen assay platform to screen small molecule drug off-target effects on memory T cell expansion/function using a dosing regimen based on adenosine treatment. As a proof of principle, we show low dose GS-5734, a monophosphoramidate prodrug of an adenosine analog, does not alter memory T cell recall at lower doses whereas toxicity observed at high dose favors antigen-specific memory T cell survival/proliferation over non-specific CD8+ T cells. Conversely, parent nucleoside GS-441524 at high dosage does not result in cellular toxicity and reduces antigen-specific T cell recall in most donors. Despite similar chemical structure, these drugs displayed opposing effects on memory T cell expansion and viability highlighting the sensitivity of this assay setup in screening compounds for off-target effects.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Adenosina/análogos & derivados , Adenosina/farmacología , Alanina/análogos & derivados , Memoria Inmunológica/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Adenosina Monofosfato/farmacología , Alanina/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Proliferación Celular/efectos de los fármacos , Humanos , Memoria Inmunológica/inmunología , Linfocitos T/inmunología
8.
Vaccine ; 38(50): 7989-7997, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33158592

RESUMEN

Pharmacological therapies for the treatment of cocaine addiction have had disappointing efficacy, and the lack of recent developments in the clinical care of cocaine-addicted patients indicates a need for novel treatment strategies. Recent studies have shown that vaccination against cocaine to elicit production of antibodies that reduce concentrations of free drug in the blood is a promising method to protect against the effects of cocaine and reduce rates of relapse. However, the poorly immunogenic nature of cocaine remains a major hurdle to active immunization. Therefore, we hypothesized that strategies to increase targeted exposure of cocaine to the immune system may produce a more effective vaccine. To specifically direct an immune response against cocaine, in the present study we have conjugated a cocaine analog to a dendrimer-based nanoparticle carrier with MHC II-binding moieties that previously has been shown to activate antigen-presenting cells necessary for antibody production. This strategy produced a rapid, prolonged, and high affinity anti-cocaine antibody response without the need for an adjuvant. Surprisingly, additional evaluation using multiple adjuvant formulations in two strains of inbred mice found adjuvants were either functionally redundant or deleterious in the vaccination against cocaine using this platform. The use of conditioned place preference in rats after administration of this vaccine provided proof of concept for the ability of this vaccine to diminish cocaine reward. Together these data demonstrate the intrinsic efficacy of an immune-targeting dendrimer-based cocaine vaccine, with a vast potential for design of future vaccines against other poorly immunogenic antigens by substitution of the conjugated cargo.


Asunto(s)
Cocaína , Dendrímeros , Nanopartículas , Vacunas , Adyuvantes Inmunológicos , Animales , Humanos , Ratones , Ratas , Vacunación
9.
Immunotherapy ; 10(16): 1349-1360, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30474481

RESUMEN

AIM: Lung cancer gene therapies require reagents to selectively transfect lung tumors after systemic administration. MATERIALS & METHODS: We created a reagent called NSCLC-NP by attaching a peptide with binding affinity for lung cancer to polyamidoamine dendrimers. The positively charged dendrimers electrostatically bind negatively charged nucleic acids, inhibit endogenous nucleases and transfect cells targeted by the attached peptide. RESULTS: In vitro, NSCLC-NP complexed to DNA plasmids bound and transfected three human lung cancer cell lines producing protein expression of the plasmid's gene. In vivo, systemically administered NSCLC-NP selectively transfected lung cancer cells growing in RAG1KO mice. CONCLUSION: The capability of NSCLC-NP to selectively transfect lung cancer allows its future use as a vehicle to implement human lung cancer gene therapy strategies.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Dendrímeros/farmacología , Neoplasias Pulmonares , Plásmidos/farmacología , Transfección/métodos , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Dendrímeros/química , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Ratones , Ratones Noqueados , Plásmidos/química
10.
J Transl Med ; 5: 26, 2007 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-17555571

RESUMEN

The incidence of cancer increases significantly in later life, yet few pre-clinical studies of cancer immunotherapy use mice of advanced age. A novel vaccine delivery platform (VacciMax,VM) is described that encapsulates antigens and adjuvants in multilamellar liposomes in a water-in-oil emulsion. The therapeutic potential of VM-based vaccines administered as a single dose was tested in HLA-A2 transgenic mice of advanced age (48-58 weeks old) bearing large palpable TC1/A2 tumors. The VM-based vaccines contained one or more peptides having human CTL epitopes derived from HPV 16 E6 and E7. VM formulations contained a single peptide, a mixture of four peptides or the same four peptides linked together in a single long peptide. All VM formulations contained PADRE and CpG as adjuvants and ISA51 as the hydrophobic component of the water-in-oil emulsion. VM-formulated vaccines containing the four peptides as a mixture or linked together in one long peptide eradicated 19-day old established tumors within 21 days of immunization. Peptide-specific cytotoxic cellular responses were confirmed by ELISPOT and intracellular staining for IFN-gamma producing CD8+ T cells. Mice rendered tumor-free by vaccination were re-challenged in the opposite flank with 10 million HLF-16 tumor cells, another HLA-A2/E6/E7 expressing tumor cell line. None of these mice developed tumors following the re-challenge. In summary, this report describes a VM-formulated therapeutic vaccine with the following unprecedented outcome: a) eradication of large tumors (> 700 mm3) b) in mice of advanced age c) in less than three weeks post-immunization d) following a single vaccination.


Asunto(s)
Papillomavirus Humano 16/inmunología , Inmunización/instrumentación , Neoplasias/inmunología , Neoplasias/patología , Péptidos/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Envejecimiento/inmunología , Envejecimiento/patología , Animales , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Citocinas/metabolismo , Humanos , Interferón gamma/metabolismo , Espacio Intracelular/metabolismo , Ratones , Bazo/citología , Coloración y Etiquetado
11.
J Transl Med ; 5: 20, 2007 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-17451606

RESUMEN

BACKGROUND: Melanoma tumors are known to express antigens that usually induce weak immune responses of short duration. Expression of both tumor-associated antigens p53 and TRP2 by melanoma cells raises the possibility of simultaneously targeting more than one antigen in a therapeutic vaccine. In this report, we show that VacciMax (VM), a novel liposome-based vaccine delivery platform, can increase the immunogenicity of melanoma associated antigens, resulting in tumor elimination. METHODS: C57BL/6 mice bearing B16-F10 melanoma tumors were vaccinated subcutaneously 6 days post tumor implantation with a mixture of synthetic peptides (modified p53: 232-240, TRP-2: 181-188 and PADRE) and CpG. Tumor growth was monitored and antigen-specific splenocyte responses were assayed by ELISPOT. RESULTS: Vaccine formulated in VM increased the number of both TRP2- and p53-specific IFN-gamma producing splenocytes following a single vaccination. Vaccine formulated without VM resulted only in enhanced IFN-gamma producing splenocytes to one CTL epitopes (TRP2:180-188), suggesting that VM overcomes antigen dominance and enhances immunogenicity of multiple epitopes. Vaccination of mice bearing 6-day old B16-F10 tumors with both TRP2 and p53-peptides formulated in VM successfully eradicated tumors in all mice. A control vaccine which contained all ingredients except liposomes resulted in eradication of tumors in no more than 20% of mice. CONCLUSION: A single administration of VM is capable of inducing an effective CTL response to multiple tumor-associated antigens. The responses generated were able to reject 6-day old B16-F10 tumors.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Péptidos/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Vacunación/instrumentación , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Femenino , Interferón gamma/biosíntesis , Recuento de Linfocitos , Melanoma Experimental/prevención & control , Ratones , Ratones Endogámicos C57BL , Bazo/citología , Bazo/inmunología
12.
Cell Death Dis ; 8(5): e2826, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28542141

RESUMEN

The process of controlled cellular death known as apoptosis has an important central role not only in normal homeostatic maintenance of tissues, but also in numerous diseases such as cancer, neurodegenerative, autoimmune, and cardiovascular diseases. As a result, new technologies with the capability to selectively detect apoptotic cells represent a central focus of research for the study of these conditions. We have developed a new biosensor for the detection of apoptotic cells, incorporating the targeted selectivity for apoptotic cells from Annexin V with the sensitivity of bioluminescence signal generation from a serum-stable mutant of Renilla luciferase (RLuc8). Our data presents a complete characterization of the structural and biochemical properties of this new Annexin-Renilla fusion protein (ArFP) construct, as well as a validation of its ability to detect apoptosis in vitro. Moreover, this work represents the first report of a bioluminescent Annexin V apoptosis sensor utilized in vivo. With this new construct, we examine apoptosis within disease-relevant animal models of surgery-induced ischemia/reperfusion, corneal injury, and retinal cell death as a model of age-related macular degeneration. In each of these experiments, we demonstrate successful application of the ArFP construct for detection and bioluminescence imaging of apoptosis within each disease or treatment model. ArFP represents an important new tool in the continuously growing kit of technologies for apoptosis detection, and our results from both in vitro and in vivo experiments suggest a diverse range of potential clinically relevant applications including cancer therapeutic screening and efficacy analysis, atherosclerosis and cardiovascular disease detection, and the monitoring of any number of other conditions in which apoptosis has a central role.


Asunto(s)
Anexina A5/metabolismo , Apoptosis , Luminiscencia , Sondas Moleculares/metabolismo , Animales , Calorimetría , Modelos Animales de Enfermedad , Femenino , Humanos , Células Jurkat , Luciferasas de Renilla/metabolismo , Ratones Endogámicos BALB C , Modelos Biológicos , Ratas , Proteínas Recombinantes de Fusión/metabolismo
13.
Cancer Res ; 64(15): 5407-14, 2004 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15289349

RESUMEN

The p53 gene product is overexpressed by almost 50% of cancers, making it an ideal target for cancer immunotherapy. We previously demonstrated rejection of established p53-overexpressing tumors without stimulating autoimmunity by immunization with modified vaccinia Ankara-expressing murine p53 (MVAp53). Tumor rejection was enhanced through antibody-mediated CTL-associated antigen 4 (CTLA-4) blockade. We examined the role of synthetic oligodeoxynucleotides (ODN) containing unmethylated cytosine-phosphate-guanine (CpG) motifs (CpG ODN) in enhancing MVAp53-mediated tumor rejection. CpG ODN with MVAp53 resulted in tumor rejection in BALB/c mice bearing poorly immunogenic 11A-1 murine mammary carcinomas or Meth A sarcomas and C57Bl/6 mice bearing MC-38 colon carcinomas. The effect was similar to that seen in tumor-bearing mice treated with MVAp53 along with CTLA-4 blockade. Monoclonal antibody depletion experiments demonstrated that the adjuvant effects of CpG ODN and CTLA-4 blockades were CD8 dependent. CpG ODN were partially natural killer cell dependent and ineffective in Toll-like Receptor 9(-/-) and interleukin 6(-/-) mice, whereas CTLA-4 blockade was partially CD4 dependent and functional in Toll-like Receptor 9(-/-) and interleukin 6(-/-) mice. In addition, when administered with MVAp53, both adjuvants enhanced p53-specific cytotoxicity and demonstrated an additive effect when combined. The combination of CpG ODN and CTLA-4 blockade worked synergistically to reject palpable 11A-1 and MC-38 tumors. These experiments demonstrate the potential for augmenting MVAp53-mediated antitumor immunity using CpG ODN and CTLA-4 blockade. This cell-free immunotherapy approach is a candidate for evaluation in cancer patients.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Neoplasias del Colon/terapia , Neoplasias Mamarias Animales/terapia , Sarcoma Experimental/terapia , Transducción de Señal , Proteína p53 Supresora de Tumor/farmacología , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/uso terapéutico , Animales , Anticuerpos Monoclonales/metabolismo , Antígenos CD , Antígenos de Diferenciación/química , Antígeno CTLA-4 , Células Cultivadas , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Terapia Combinada , Cricetinae , Pruebas Inmunológicas de Citotoxicidad , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Sinergismo Farmacológico , Femenino , Homocigoto , Humanos , Inmunización , Interleucina-6/genética , Interleucina-6/fisiología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Oligodesoxirribonucleótidos/farmacología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Sarcoma Experimental/genética , Sarcoma Experimental/inmunología , Receptor Toll-Like 9 , Proteína p53 Supresora de Tumor/genética
14.
PLoS One ; 11(4): e0154053, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27104647

RESUMEN

Stem cell therapy has emerged as a promising approach for treatment of a number of diseases, including delayed and non-healing wounds. However, targeted systemic delivery of therapeutic cells to the dysfunctional tissues remains one formidable challenge. Herein, we present a targeted nanocarrier-mediated cell delivery method by coating the surface of the cell to be delivered with dendrimer nanocarriers modified with adhesion molecules. Infused nanocarrier-coated cells reach to destination via recognition and association with the counterpart adhesion molecules highly or selectively expressed on the activated endothelium in diseased tissues. Once anchored on the activated endothelium, nanocarriers-coated transporting cells undergo transendothelial migration, extravasation and homing to the targeted tissues to execute their therapeutic role. We now demonstrate feasibility, efficacy and safety of our targeted nanocarrier for delivery of bone marrow cells (BMC) to cutaneous wound tissues and grafted corneas and its advantages over conventional BMC transplantation in mouse models for wound healing and neovascularization. This versatile platform is suited for targeted systemic delivery of virtually any type of therapeutic cell.


Asunto(s)
Selectina E/metabolismo , Neovascularización Fisiológica , Células Madre/citología , Cicatrización de Heridas , Animales , Membrana Celular/metabolismo , Dendrímeros , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión
15.
EBioMedicine ; 9: 161-169, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27333040

RESUMEN

Bacteria in humans play an important role in health and disease. Considerable emphasis has been placed in understanding the role of bacteria in host-microbiome interkingdom communication. Here we show that serotonin, responsible for mood in the brain and motility in the gut, can also act as a bacterial signaling molecule for pathogenic bacteria. Specifically, we found that serotonin acts as an interkingdom signaling molecule via quorum sensing and that it stimulates the production of bacterial virulence factors and increases biofilm formation in vitro and in vivo in a novel mouse infection model. This discovery points out at roles of serotonin both in bacteria and humans, and at phenotypic implications not only manifested in mood behavior but also in infection processes in the host. Thus, regulating serotonin concentrations in the gut may provide with paradigm shifting therapeutic approaches.


Asunto(s)
Pseudomonas aeruginosa/fisiología , Percepción de Quorum/efectos de los fármacos , Serotonina/farmacología , Virulencia/efectos de los fármacos , Animales , Proteínas Bacterianas/metabolismo , Catalasa/metabolismo , Citocinas/análisis , Femenino , Inyecciones Intraperitoneales , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Intestinos/patología , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Rastreo , Péptido Hidrolasas/metabolismo , Peroxidasa/metabolismo , Fenotipo , Embarazo , Pseudomonas aeruginosa/enzimología , Pseudomonas aeruginosa/metabolismo
16.
PLoS One ; 11(7): e0158579, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27367859

RESUMEN

The increased importance of in vivo diagnostics has posed new demands for imaging technologies. In that regard, there is a need for imaging molecules capable of expanding the applications of current state-of-the-art imaging in vivo diagnostics. To that end, there is a desire for new reporter molecules capable of providing strong signals, are non-toxic, and can be tailored to diagnose or monitor the progression of a number of diseases. Aequorin is a non-toxic photoprotein that can be used as a sensitive marker for bioluminescence in vivo imaging. The sensitivity of aequorin is due to the fact that bioluminescence is a rare phenomenon in nature and, therefore, it does not suffer from autofluorescence, which contributes to background emission. Emission of bioluminescence in the blue-region of the spectrum by aequorin only occurs when calcium, and its luciferin coelenterazine, are bound to the protein and trigger a biochemical reaction that results in light generation. It is this reaction that endows aequorin with unique characteristics, making it ideally suited for a number of applications in bioanalysis and imaging. Herein we report the site-specific incorporation of non-canonical or non-natural amino acids and several coelenterazine analogues, resulting in a catalog of 72 cysteine-free, aequorin variants which expand the potential applications of these photoproteins by providing several red-shifted mutants better suited to use in vivo. In vivo studies in mouse models using the transparent tissue of the eye confirmed the activity of the aequorin variants incorporating L-4-iodophehylalanine and L-4-methoxyphenylalanine after injection into the eye and topical addition of coelenterazine. The signal also remained localized within the eye. This is the first time that aequorin variants incorporating non-canonical amino acids have shown to be active in vivo and useful as reporters in bioluminescence imaging.


Asunto(s)
Aequorina/genética , Aequorina/metabolismo , Sustitución de Aminoácidos , Imagen Molecular , Aequorina/química , Animales , Color , Femenino , Luminiscencia , Ratones , Modelos Moleculares , Conformación Proteica
18.
Cancer Res ; 71(24): 7452-62, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21987727

RESUMEN

DNA-based vaccines hold promise to outperform conventional antigen-based vaccines by virtue of many unique features. However, DNA vaccines have thus far fallen short of expectations, due in part to poor targeting of professional antigen-presenting cells (APC) and low immunogenicity. In this study, we describe a new platform for effective and selective delivery of DNA to APCs in vivo that offers intrinsic immune-enhancing characteristics. This platform is based on conjugation of fifth generation polyamidoamine (G5-PAMAM) dendrimers, a DNA-loading surface, with MHC class II-targeting peptides that can selectively deliver these dendrimers to APCs under conditions that enhance their immune stimulatory potency. DNA conjugated with this platform efficiently transfected murine and human APCs in vitro. Subcutaneous administration of DNA-peptide-dendrimer complexes in vivo preferentially transfected dendritic cells (DC) in the draining lymph nodes, promoted generation of high affinity T cells, and elicited rejection of established tumors. Taken together, our findings show how PAMAM dendrimer complexes can be used for high transfection efficiency and effective targeting of APCs in vivo, conferring properties essential to generate effective DNA vaccines.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Dendrímeros/química , Péptidos/inmunología , Vacunas de ADN/inmunología , Secuencia de Aminoácidos , Animales , Células Presentadoras de Antígenos/metabolismo , Línea Celular Tumoral , Células Cultivadas , ADN/genética , ADN/inmunología , ADN/metabolismo , Citometría de Flujo , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Péptidos/química , Péptidos/metabolismo , Unión Proteica/inmunología , Electricidad Estática , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Vacunación/métodos , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética
19.
Hybridoma (Larchmt) ; 30(5): 409-18, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22008067

RESUMEN

In vivo electroporation has become a gold standard method for DNA immunization. The method assists the DNA entry into cells, results in expression and the display of the native form of antigens to professional cells of the immune system, uses both arms of immune system, has a built-in adjuvant system, is relatively safe, and is cost-effective. However, there are challenges for achieving an optimized reproducible process for eliciting strong humoral responses and for the screening of specific immune responses, in particular, when the aim is to mount humoral responses or to generate monoclonal antibodies via hybridoma technology. Production of monoclonal antibodies demands generation of high numbers of primed B and CD4 T helper cells in lymphoid organs needed for the fusion that traditionally is achieved by a final intravenous antigen injection. The purified antigen is also needed for screening of hundreds of clones obtained upon fusion of splenocytes. Such challenges make DNA vaccination dependent on purified proteins. Here, we have optimized methods for in vivo electroporation, production, and use of cells expressing the antigen and an in-cell Western screening method. These methods resulted in (1) reproducibly mounting robust humoral responses against antigens with different cell localizations, and (2) the ability to screen for antigen eliminating a need for protein/antigen purification. This process includes optimized parameters for in vivo electroporation, the use of transfected cells for final boost, and mild fixation/permeabilization of cells for screening. Using this process, upon two vaccinations via in vivo electroporation (and final boost), monoclonal antibodies against nucleus and cytoplasmic and transmembrane proteins were achieved.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Vacunas de ADN , Animales , Western Blotting/métodos , Proteínas Adaptadoras de Señalización CARD , Células COS , Chlorocebus aethiops , Proteínas del Citoesqueleto/biosíntesis , Proteínas del Citoesqueleto/inmunología , Electroporación/métodos , Femenino , Células HEK293 , Humanos , Sueros Inmunes , Complejo de Antígeno L1 de Leucocito/biosíntesis , Complejo de Antígeno L1 de Leucocito/inmunología , Ratones , Ratones Endogámicos BALB C , Proteínas de Microfilamentos/biosíntesis , Proteínas de Microfilamentos/inmunología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/inmunología , Ovalbúmina/biosíntesis , Ovalbúmina/inmunología , Conformación Proteica , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/inmunología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Receptores del Activador de Plasminógeno Tipo Uroquinasa/inmunología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología
20.
Nat Med ; 17(8): 952-60, 2011 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-21804539

RESUMEN

Focal segmental glomerulosclerosis (FSGS) is a cause of proteinuric kidney disease, compromising both native and transplanted kidneys. Treatment is limited because of a complex pathogenesis, including unknown serum factors. Here we report that serum soluble urokinase receptor (suPAR) is elevated in two-thirds of subjects with primary FSGS, but not in people with other glomerular diseases. We further find that a higher concentration of suPAR before transplantation underlies an increased risk for recurrence of FSGS after transplantation. Using three mouse models, we explore the effects of suPAR on kidney function and morphology. We show that circulating suPAR activates podocyte ß(3) integrin in both native and grafted kidneys, causing foot process effacement, proteinuria and FSGS-like glomerulopathy. Our findings suggest that the renal disease only develops when suPAR sufficiently activates podocyte ß(3) integrin. Thus, the disease can be abrogated by lowering serum suPAR concentrations through plasmapheresis, or by interfering with the suPAR-ß(3) integrin interaction through antibodies and small molecules targeting either uPAR or ß(3) integrin. Our study identifies serum suPAR as a circulating factor that may cause FSGS.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/etiología , Integrina beta3/metabolismo , Podocitos/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/sangre , Adolescente , Adulto , Animales , Western Blotting , Femenino , Citometría de Flujo , Glomeruloesclerosis Focal y Segmentaria/sangre , Humanos , Inmunohistoquímica , Inmunoprecipitación , Trasplante de Riñón/fisiología , Masculino , Ratones , Microscopía Electrónica , Microscopía Fluorescente , Modelos Biológicos , Plasmaféresis , Podocitos/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA