Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Immunol ; 209(11): 2114-2132, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36261171

RESUMEN

MPYS/STING (stimulator of IFN genes) senses cyclic dinucleotides (CDNs), generates type I IFNs, and plays a critical role in infection, inflammation, and cancer. In this study, analyzing genotype and haplotype data from the 1000 Genomes Project, we found that the R71H-G230A-R293Q (HAQ) MPYS allele frequency increased 57-fold in East Asians compared with sub-Saharan Africans. Meanwhile, the G230A-R293Q (AQ) allele frequency decreased by 98% in East Asians compared with sub-Saharan Africans. We propose that the HAQ and AQ alleles underwent a natural selection during the out-of-Africa migration. We used mouse models of HAQ and AQ to investigate the underlying mechanism. We found that the mice carrying the AQ allele, which disappeared in East Asians, had normal CDN-type I IFN responses. Adult AQ mice, however, had less fat mass than did HAQ or wild-type mice on a chow diet. AQ epididymal adipose tissue had increased regulatory T cells and M2 macrophages with protein expression associated with enhanced fatty acid oxidation. Conditional knockout mice and adoptive cell transfer indicate a macrophage and regulatory T cell-intrinsic role of MPYS in fatty acid metabolism. Mechanistically, AQ/IFNAR1-/- mice had a similar lean phenotype as for the AQ mice. MPYS intrinsic tryptophan fluorescence revealed that the R71H change increased MPYS hydrophilicity. Lastly, we found that the second transmembrane (TM) and the TM2-TM3 linker region of MPYS interact with activated fatty acid, fatty acyl-CoA. In summary, studying the evolution of the human MPYS gene revealed an MPYS function in modulating fatty acid metabolism that may be critical during the out-of-Africa migration.


Asunto(s)
Ácidos Grasos , Tolerancia Inmunológica , Proteínas de la Membrana , Adulto , Animales , Humanos , Ratones , Ácidos Grasos/metabolismo , Homeostasis , Proteínas de la Membrana/metabolismo , Ratones Noqueados , Interferón Tipo I
2.
J Immunol ; 206(9): 2233-2245, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33879579

RESUMEN

Induction of lung mucosal immune responses is highly desirable for vaccines against respiratory infections. We recently showed that monocyte-derived dendritic cells (moDCs) are responsible for lung IgA induction. However, the dendritic cell subset inducing lung memory TH cells is unknown. In this study, using conditional knockout mice and adoptive cell transfer, we found that moDCs are essential for lung mucosal responses but are dispensable for systemic vaccine responses. Next, we showed that mucosal adjuvant cyclic di-GMP differentiated lung moDCs into Bcl6+ mature moDCs promoting lung memory TH cells, but they are dispensable for lung IgA production. Mechanistically, soluble TNF mediates the induction of lung Bcl6+ moDCs. Our study reveals the functional heterogeneity of lung moDCs during vaccination and paves the way for an moDC-targeting vaccine strategy to enhance immune responses on lung mucosa.


Asunto(s)
GMP Cíclico/análogos & derivados , Pulmón/inmunología , Monocitos/inmunología , Proteínas Proto-Oncogénicas c-bcl-6/inmunología , Vacunas/inmunología , Adyuvantes Inmunológicos , Animales , Diferenciación Celular/inmunología , GMP Cíclico/inmunología , Células Dendríticas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células TH1/inmunología
3.
ACS Appl Bio Mater ; 7(2): 727-751, 2024 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-38166376

RESUMEN

The immune system usually provides a defense against invading pathogenic microorganisms and any other particulate contaminants. Nonetheless, it has been recently reported that nanomaterials can evade the immune system and modulate immunological responses due to their unique physicochemical characteristics. Consequently, nanomaterial-based activation of immune components, i.e., neutrophils, macrophages, and other effector cells, may induce inflammation and alter the immune response. Here, it is essential to distinguish the acute and chronic modulations triggered by nanomaterials to determine the possible risks to human health. Nanomaterials size, shape, composition, surface charge, and deformability are factors controlling their uptake by immune cells and the resulting immune responses. The exterior corona of molecules adsorbed over nanomaterials surfaces also influences their immunological effects. Here, we review current nanoengineering trends for targeted immunomodulation with an emphasis on the design, safety, and potential toxicity of nanomaterials. First, we describe the characteristics of engineered nanomaterials that trigger immune responses. Then, the biocompatibility and immunotoxicity of nanoengineered particles are debated, because these factors influence applications. Finally, future nanomaterial developments in terms of surface modifications, synergistic approaches, and biomimetics are discussed.


Asunto(s)
Nanoestructuras , Humanos , Nanoestructuras/toxicidad , Nanoestructuras/química , Macrófagos , Inflamación , Inmunidad , Inmunomodulación
4.
Curr Protein Pept Sci ; 25(4): 307-325, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38265408

RESUMEN

The global pandemic caused by COVID-19 posed a significant challenge to public health, necessitating rapid scientific interventions to tackle the spread of infection. The review discusses the key areas of research on COVID-19 including viral genomics, epidemiology, pathogenesis, diagnostics, and therapeutics. The genome sequencing of the virus facilitated the tracking of its evolution, transmission dynamics, and identification of variants. Epidemiological studies have provided insights into disease spread, risk factors, and the impact of public health infrastructure and social distancing measures. Investigations of the viral pathogenesis have elucidated the mechanisms underlying immune responses and severe manifestations including the long-term effects of COVID-19. Overall, the article provides an updated overview of the diagnostic methods developed for SARS-CoV-2 and discusses their strengths, limitations, and appropriate utilization in different clinical and public health settings. Furthermore, therapeutic approaches including antiviral drugs, immunomodulatory therapies, and repurposed medications have been investigated to alleviate disease severity and improve patient outcomes. Through a comprehensive analysis of these scientific efforts, the review provides an overview of the advancements made in understanding and tackling SARS-CoV-2, while underscoring the need for continued research to address the evolving challenges posed by this global health crisis.


Asunto(s)
Antivirales , COVID-19 , SARS-CoV-2 , Humanos , COVID-19/virología , COVID-19/epidemiología , COVID-19/transmisión , SARS-CoV-2/genética , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/aislamiento & purificación , Antivirales/uso terapéutico , Pandemias/prevención & control , Genoma Viral , Salud Global , Tratamiento Farmacológico de COVID-19
5.
Front Immunol ; 14: 1277677, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38090593

RESUMEN

Advances in understanding the genetic basis of cancer have driven alternative treatment approaches. Recent findings have demonstrated the potential of bacteria and it's components to serve as robust theranostic agents for cancer eradication. Compared to traditional cancer therapies like surgery, chemotherapy, radiotherapy, bacteria mediated tumor therapy has exhibited superior cancer suppressing property which is attributed a lot to it's tumor proliferating and accumulating characteristics. Genetically modified bacteria has reduced inherent toxicity and enhanced specificity towards tumor microenvironment. This anti- tumor activity of bacteria is attributed to its toxins and other active components from the cell membrane, cell wall and spores. Furthermore, bacterial genes can be regulated to express and deliver cytokines, antibodies and cancer therapeutics. Although there is less clinical data available, the pre- clinical research clearly indicates the feasibility and potential of bacteria- mediated cancer therapy.


Asunto(s)
Neoplasias , Humanos , Neoplasias/genética , Bacterias/genética , Bacterias/metabolismo , Inmunoterapia , Terapia Combinada , Microambiente Tumoral
6.
Front Immunol ; 12: 699702, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34512626

RESUMEN

The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway senses DNA and induces type I interferon (IFN) production. Whether and how the STING pathway crosstalk to other innate immune pathways during pathogen infection, however, remains unclear. Here, we showed that STING was needed for Streptococcus pneumoniae-induced late, not early, stage of lung IFNγ production. Using knockout mice, IFNγ reporter mice, intracellular cytokine staining, and adoptive cell transfer, we showed that cGAS-STING-dependent lung IFNγ production was independent of type I IFNs. Furthermore, STING expression in monocyte/monocyte-derived cells governed IFNγ production in the lung via the production of IL-12p70. Surprisingly, DNA stimulation alone could not induce IL-12p70 or IFNγ in Ly6Chi monocyte. The production of IFNγ required the activation by both DNA and heat-killed S. pneumococcus. Accordingly, MyD88-/- monocyte did not generate IL-12p70 or IFNγ. In summary, the cGAS-STING pathway synergizes with the MyD88 pathway in monocyte to promote late-stage lung IFNγ production during pulmonary pneumococcal infection.


Asunto(s)
Interferón gamma/biosíntesis , Proteínas de la Membrana/inmunología , Monocitos/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Nucleotidiltransferasas/inmunología , Infecciones Neumocócicas/inmunología , Animales , Femenino , Pulmón/inmunología , Pulmón/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Nucleotidiltransferasas/metabolismo , Infecciones Neumocócicas/metabolismo , Transducción de Señal/inmunología , Streptococcus pneumoniae
7.
Pulm Circ ; 11(1): 2045894021996574, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33738095

RESUMEN

For as long as nucleic acids have been utilized to vertically and horizontally transfer genetic material, living organisms have had to develop methods of recognizing cytosolic DNA as either pathogenic (microbial invasion) or physiologic (mitosis and cellular proliferation). Derangement in key signaling molecules involved in these pathways of DNA sensing result in a family of diseases labeled interferonopathies. An interferonopathy, characterized by constitutive expression of type I interferons, ultimately manifests as severe autoimmune disease at a young age. Afflicted patients present with a constellation of immune-mediated conditions, including primary lung manifestations such as pulmonary fibrosis and pulmonary hypertension. The latter condition is especially interesting in light of the known role that DNA damage plays in a variety of types of inherited and induced pulmonary hypertension, with free DNA detection elevated in the circulation of affected individuals. While little is known regarding the role of cytosolic DNA sensing in development of pulmonary vascular disease, exciting new research in the related fields of immunology and oncology potentially sheds light on future areas of fruitful exploration. As such, the goal of this review is to summarize the state of the field of nucleic acid sensing, extrapolating common shared pathways that parallel our knowledge of pulmonary hypertension, in a molecular and cell-specific manner. Principles of DNA sensing related to known pulmonary injury inducing stimuli are also evaluated, in addition to potential therapeutic targets. Finally, future directions in pulmonary hypertension research and treatments will be briefly discussed.

8.
Sci Immunol ; 6(61)2021 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-34244314

RESUMEN

Asthma is a common inflammatory lung disease with no known cure. Previously, we uncovered a lung TNFR2+ conventional DC2 subset (cDC2s) that induces regulatory T cells (Tregs) maintaining lung tolerance at steady state but promotes TH2 response during house dust mite (HDM)-induced asthma. Lung IFNß is essential for TNFR2+ cDC2s-mediated lung tolerance. Here, we showed that exogenous IFNß reprogrammed TH2-promoting pathogenic TNFR2+ cDC2s back to tolerogenic DCs, alleviating eosinophilic asthma and preventing asthma exacerbation. Mechanistically, inhaled IFNß, not IFNα, activated ERK2 signaling in pathogenic lung TNFR2+ cDC2s, leading to enhanced fatty acid oxidation (FAO) and lung Treg induction. Last, human IFNß reprogrammed pathogenic human lung TNFR2+ cDC2s from patients with emphysema ex vivo. Thus, we identified an IFNß-specific ERK2-FAO pathway that might be harnessed for DC therapy.


Asunto(s)
Asma/inmunología , Células Dendríticas/trasplante , Interferón beta/uso terapéutico , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Células Th2/inmunología , Traslado Adoptivo , Animales , Asma/patología , Células Cultivadas , Células Dendríticas/inmunología , Dermatophagoides pteronyssinus/inmunología , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Humanos , Tolerancia Inmunológica/inmunología , Factores Inmunológicos/uso terapéutico , Interferón-alfa/farmacología , Pulmón/citología , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Oxidación-Reducción , Receptor de Interferón alfa y beta/genética , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología
9.
Vaccines (Basel) ; 8(3)2020 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-32823563

RESUMEN

As prophylactic vaccine adjuvants for infectious diseases, cyclic dinucleotides (CDNs) induce safe, potent, long-lasting humoral and cellular memory responses in the systemic and mucosal compartments. As therapeutic cancer vaccine adjuvants, CDNs induce potent anti-tumor immunity, including cytotoxic T cells and NK cells activation that achieve durable regression in multiple mouse models of tumors. Clinical trials are ongoing to fulfill the promise of CDNs (ClinicalTrials.gov: NCT02675439, NCT03010176, NCT03172936, and NCT03937141). However, in October 2018, the first clinical data with Merck's CDN MK-1454 showed zero activity as a monotherapy in patients with solid tumors or lymphomas (NCT03010176). Lately, the clinical trial from Aduro's CDN ADU-S100 monotherapy was also disappointing (NCT03172936). The emerging hurdle in CDN vaccine development calls for a timely re-evaluation of our understanding on CDN vaccine adjuvants. Here, we review the status of CDN vaccine adjuvant research, including their superior adjuvant activities, in vivo mode of action, and confounding factors that affect their efficacy in humans. Lastly, we discuss the strategies to overcome the hurdle and advance promising CDN adjuvants in humans.

10.
Vaccines (Basel) ; 8(4)2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-33019545

RESUMEN

Aluminium salts have been the adjuvant of choice in more than 100 licensed vaccines. Here, we have studied the synergistic effect of aluminium hydroxide nanoparticles (AH np) and non-ionic surfactant-based vesicles (NISV) in modulating the immune response against protective antigen domain 4 (D4) of Bacillus anthracis. NISV was prepared from Span 60 and cholesterol, while AH np was prepared from aluminium chloride and sodium hydroxide. AH np was co-administered with NISV encapsulating D4 (NISV-D4) to formulate AHnp/NISV-D4. The antigen-specific immune response of AHnp/NISV-D4 was compared with that of commercial alhydrogel (alhy) co-administered with NISV-D4 (alhydrogel/NISV-D4), NISV-D4, AHnp/D4, and alhydrogel/D4. Co-administration of NISV-D4 with AH np greatly improved the D4-specific antibody titer as compared to the control groups. Based on IgG isotyping and ex vivo cytokine analysis, AHnp/NISV-D4 generated a balanced Th1/Th2 response. Furthermore, AH np/NISV-D4 showed superior protection against anthrax spore challenge in comparison to other groups. Thus, we demonstrate the possibility of developing a novel combinatorial nanoformulation capable of augmenting both humoral and cellular response, paving the way for adjuvant research.

11.
Front Immunol ; 11: 1674, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32849581

RESUMEN

Cyclic dinucleotides (CDNs) are promising vaccine adjuvants inducing balanced, potent humoral, and cellular immune responses. How aging influences CDN efficacy is unclear. We examined the vaccine efficacy of 3',5'-cyclic diguanylic acid (cyclic di-GMP, CDG), the founding member of CDNs, in 1-year-old (middle-aged) and 2-year-old (aged) C57BL/6J mice. We found that 1- and 2-year-old C57BL/6J mice are defective in CDG-induced memory T helper (Th)1 and Th17 responses and high-affinity serum immunoglobulin (Ig)G, mucosal IgA production. Next, we generated two novel tumor necrosis factor (TNF) fusion proteins that target soluble TNF (solTNF) and transmembrane TNF (tmTNF) to monocyte-derived dendritic cells (moDCs) to enhance CDG vaccine efficacy in 1- and 2-year-old mice. The moDC-targeting TNF fusion proteins restored CDG-induced memory Th1, Th17, and high-affinity IgG, IgA responses in the 1- and 2-year-old mice. Together, the data suggested that aging negatively impacts CDG vaccine adjuvanticity. MoDC-targeting TNF fusion proteins enhanced CDG adjuvanticity in the aging mice.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , GMP Cíclico/análogos & derivados , Células Dendríticas/efectos de los fármacos , Fragmentos Fc de Inmunoglobulinas/farmacología , Pulmón/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Adyuvantes Inmunológicos/administración & dosificación , Administración Intranasal , Factores de Edad , Envejecimiento/inmunología , Envejecimiento/metabolismo , Animales , Células Cultivadas , GMP Cíclico/administración & dosificación , GMP Cíclico/farmacología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Inmunidad Mucosa/efectos de los fármacos , Inmunización , Inmunogenicidad Vacunal , Inmunoglobulina A/inmunología , Inmunoglobulina A/metabolismo , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Pulmón/inmunología , Pulmón/metabolismo , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión/administración & dosificación , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/metabolismo , Factor de Necrosis Tumoral alfa/administración & dosificación
12.
Int J Nanomedicine ; 15: 239-252, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32021177

RESUMEN

INTRODUCTION: Aluminum salts, although they have been used as adjuvants in many vaccine formulations since 1926, exclusively induce a Th2-biased immune response, thereby limiting their use against intracellular pathogens like Mycobacterium tuberculosis. METHODS AND RESULTS: Herein, we synthesized amorphous and crystalline forms of aluminum hydroxide nanoparticles (AH nps) of 150-200 nm size range. Using Bacillus anthracis protective antigen domain 4 (D4) as a model antigen, we demonstrated that both amorphous and crystalline forms of AH nps displayed enhanced antigen D4 uptake by THP1 cells as compared to commercial adjuvant aluminum hydroxide gel (AH gel). In a mouse model, both amorphous and crystalline AH nps triggered an enhanced D4-specific Th2- and Th1-type immune response and conferred superior protection against anthrax spore challenge as compared to AH gel. Physicochemical characterization of crystalline and amorphous AH nps revealed stronger antigen D4 binding and release than AH gel. CONCLUSION: These results demonstrate that size and crystallinity of AH nps play important roles in mediating enhanced antigen presenting cells (APCs) activation and potentiating a strong antigen-specific immune response, and are critical parameters for the rational design of alum-based Th1-type adjuvant to induce a more balanced antigen-specific immune response.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Hidróxido de Aluminio/química , Carbunco/prevención & control , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/inmunología , Nanopartículas del Metal/química , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacocinética , Hidróxido de Aluminio/inmunología , Hidróxido de Aluminio/farmacología , Animales , Carbunco/inmunología , Vacunas contra el Carbunco/química , Vacunas contra el Carbunco/inmunología , Vacunas contra el Carbunco/farmacología , Línea Celular , Modelos Animales de Enfermedad , Dispersión Dinámica de Luz , Femenino , Humanos , Ratones , Células RAW 264.7 , Espectroscopía Infrarroja por Transformada de Fourier , Células TH1/inmunología
13.
Mucosal Immunol ; 13(4): 595-608, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31959883

RESUMEN

The lung is a naturally tolerogenic organ. Lung regulatory T cells (T-regs) control lung mucosal tolerance. Here, we identified a lung IFNAR1hiTNFR2+ conventional DC2 (iR2D2) population that induces T-regs in the lung at steady state. Using conditional knockout mice, adoptive cell transfer, receptor blocking antibodies, and TNFR2 agonist, we showed that iR2D2 is a lung microenvironment-adapted dendritic cell population whose residence depends on the constitutive TNFR2 signaling. IFNß-IFNAR1 signaling in iR2D2 is necessary and sufficient for T-regs induction in the lung. The Epcam+CD45- epithelial cells are the sole lung IFNß producer at the steady state. Surprisingly, iR2D2 is plastic. In a house dust mite model of asthma, iR2D2 generates lung TH2 responses. Last, healthy human lungs have a phenotypically similar tolerogenic iR2D2 population, which became pathogenic in lung disease patients. Our findings elucidate lung epithelial cells IFNß-iR2D2-T-regs axis in controlling lung mucosal tolerance and provide new strategies for therapeutic interventions.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Tolerancia Inmunológica , Receptor de Interferón alfa y beta/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Biomarcadores , Femenino , Humanos , Interferón beta/metabolismo , Masculino , Ratones , Factor de Crecimiento Transformador beta1/metabolismo
14.
Int J Nanomedicine ; 13: 7427-7440, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30532531

RESUMEN

INTRODUCTION: In this study, we have investigated the immunogenicity and protective efficacy of a niosomal formulation encapsulating protective antigen (PA) and PA domain 4 (D4) of Bacillus anthracis. METHODS: Nonionic surfactant-based vesicles (NISV) + PA and NISV + D4 were prepared from span-60 and cholesterol by reverse-phase evaporation method and were evaluated for in vitro characteristics and immunological studies. RESULTS: Particle characterization using transmission electron microscopy and atomic force microscopy analysis showed that the niosomal formulation was spherical in shape. The entrapment efficiency values were calculated to be 58.5% and 44.75% for PA and D4, respectively. Confocal microscopy and flow cytometry studies showed an enhanced uptake of antigen in THP1 macrophages by niosome as compared to antigen only. An in vitro release assay showed a burst release of antigen from niosome within 24 hours followed by a gradual release for 144 hours. Immunological studies showed that both PA- and D4-encapsulated niosome elicited a robust IgG titer. Antibody isotyping and cytokine profile showed that NISV + PA and NISV + D4 enhanced both Th1 and Th2 responses in mice, suggesting a mixed Th1/Th2 response. Both NISV + PA and NISV + D4 elicited high levels of anti-inflammatory cytokine interleukin-10 with low levels of pro-inflammatory cytokine tumor necrosis factor-α, suggesting the anti-inflammatory property of niosome. Both the niosomal formulations were also able to confer protection against BA infection as compared to only PA and D4. CONCLUSION: PA and D4 encapsulated NISV formulation could modulate both the Th1 and Th2 adaptive immune system and was found to be a better prophylactic against anthrax.


Asunto(s)
Carbunco/inmunología , Carbunco/prevención & control , Bacillus anthracis/inmunología , Inmunidad , Esporas Bacterianas/inmunología , Células TH1/inmunología , Células Th2/inmunología , Animales , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/inmunología , Línea Celular , Citocinas/metabolismo , Dispersión Dinámica de Luz , Humanos , Inmunización , Liposomas , Ratones , Bazo/metabolismo , Electricidad Estática , Tensoactivos/química
15.
Int J Nanomedicine ; 13: 7959-7970, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30538470

RESUMEN

The application of natural carbohydrate polysaccharides for antigen delivery and its adjuvanation potential has garnered interest in the scientific community in the recent years. These biomaterials are considered favorable candidates for adjuvant development due to their desirable properties like enormous bioavailability, non-toxicity, biodegradability, stability, affordability, and immunostimulating ability. Chitosan is the one such extensively studied natural polymer which has been appreciated for its excellent applications in pharmaceuticals. Trimethyl chitosan (TMC), a derivative of chitosan, possesses these properties. In addition it has the properties of high aqueous solubility, high charge density, mucoadhesive, permeation enhancing (ability to cross tight junction), and stability over a range of ionic conditions which makes the spectrum of its applicability much broader. It has also been seen to perform analogously to alum, complete Freund's adjuvant, incomplete Freund's adjuvant, and cyclic guanosine monophosphate adjuvanation, which justifies its role as a potent adjuvant. Although many review articles detailing the applications of chitosan in vaccine delivery are available, a comprehensive review of the applications of TMC as an adjuvant is not available to date. This article provides a comprehensive overview of structural and chemical properties of TMC which affect its adjuvant characteristics; the efficacy of various delivery routes for TMC antigen combination; and the recent advances in the elucidation of its mechanism of action.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Quitosano/química , Sistemas de Liberación de Medicamentos , Vacunas/administración & dosificación , Animales , Quitosano/administración & dosificación , Quitosano/síntesis química , Vías de Administración de Medicamentos , Portadores de Fármacos/química , Humanos
16.
Front Immunol ; 9: 562, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29616046

RESUMEN

Anthrax is an era old deadly disease against which there are only two currently available licensed vaccines named anthrax vaccine adsorbed and precipitated (AVP). Though they can provide a protective immunity, their multiple side-effects owing to their ill-defined composition and presence of toxic proteins (LF and EF) of Bacillus anthracis, the causative organism of anthrax, in the vaccine formulation makes their widespread use objectionable. Hence, an anthrax vaccine that contains well-defined and controlled components would be highly desirable. In this context, we have evaluated the potential of various vaccine formulations comprising of protective antigen (PA) encapsulated trimethyl-chitosan nanoparticles (TMC-PA) in conjunction with either CpG-C ODN 2395 (CpG) or Poly I:C. Each formulation was administered via three different routes, viz., subcutaneous (SC), intramuscular (IM), and intraperitoneal in female BALB/c mice. Irrespective of the route of immunization, CpG or Poly I:C adjuvanted TMC-PA nanoparticles induced a significantly higher humoral response (total serum IgG and its isotypes viz., IgG1, IgG2a, and IgG2b), compared to their CpG or Poly I:C PA counterparts. This clearly demonstrates the synergistic behavior of CpG and Poly I:C with TMC nanoparticles. The adjuvant potential of TMC nanoparticles could be observed in all the three routes as the TMC-PA nanoparticles by themselves induced IgG titers (1-1.5 × 105) significantly higher than both CpG PA and Poly I:C PA groups (2-8 × 104). The effect of formulations on T-helper (Th) cell development was assessed by quantifying the Th1-dependant (TNF-α, IFN-γ, and IL-2), Th2-dependant (IL-4, IL-6, and IL-10), and Th17-type (IL-17A) cytokines. Adjuvanation with CpG and Poly I:C, the TMC-PA nanoparticles triggered a Th1 skewed immune response, as suggested by an increase in the levels of total IgG2a along with IFN-γ cytokine production. Interestingly, the TMC-PA group showed a Th2-biased immune response. Upon challenge with the B. anthracis Ames strain, CpG and Poly I:C adjuvanted TMC-PA nanoparticles immunized via the SC and IM routes showed the highest protective efficacy of ~83%. Altogether, the results suggest that CpG or Poly I:C adjuvanted, PA-loaded TMC nanoparticles could be used as an effective, non-toxic, second generation subunit-vaccine candidate against anthrax.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Carbunco/inmunología , Antígenos Bacterianos/inmunología , Bacillus anthracis/inmunología , Quitosano/inmunología , Animales , Carbunco/microbiología , Carbunco/prevención & control , Vacunas contra el Carbunco/administración & dosificación , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/química , Bacillus anthracis/fisiología , Quitosano/administración & dosificación , Quitosano/química , Femenino , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Metilación , Ratones Endogámicos BALB C , Nanopartículas/administración & dosificación , Nanopartículas/química , Nanopartículas/ultraestructura , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/inmunología , Análisis de Supervivencia , Vacunación
17.
Pathog Dis ; 75(4)2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28472295

RESUMEN

The pleiotropism of the GTP-sensing transcriptional regulator CodY is evident by the gamut of processes that it regulates in almost all low G+C Gram-positive bacteria, including general metabolism, biosynthesis of some amino acids and transport systems, nitrogen uptake, sporulation, biofilm formation, motility and virulence. The role of CodY in virulence has been established in Bacillus anthracis, the top rated bioterrorism agent. In this study, we investigated the biochemical attributes of this global regulator. Homology modeling and sequence/structure analysis revealed putative GTP-binding residues in CodY of B. anthracis. CodY exhibited an interaction with the GTP as tested by ultraviolet cross-linking experiments. It could autophosphorylate itself at a conserved Ser215 residue. This was further corroborated by the impairment of autophosphorylation activity in the CodYS215A mutant. Autophosphorylation may be speculated as an additional mechanism regulating CodY activity in the cell. The protein could also hydrolyze GTP, albeit weakly, as indicated by thin- layer chromatography and spectrophotometric quantification of its kinetic parameters. Altogether, these observations provide us an insight into the mechanism of action of this global regulator and a better understanding of its functional regulation.


Asunto(s)
Bacillus anthracis/fisiología , Guanosina Trifosfato/metabolismo , Factores de Transcripción/metabolismo , Sitios de Unión , Hidrólisis , Cinética , Modelos Moleculares , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Factores de Transcripción/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA