Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 15: 1298721, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38469294

RESUMEN

Subunit vaccines hold substantial promise in controlling infectious diseases, due to their superior safety profile, specific immunogenicity, simplified manufacturing processes, and well-defined chemical compositions. One of the most important end-targets of vaccines is a subset of lymphocytes originating from the thymus, known as T cells, which possess the ability to mount an antigen-specific immune response. Furthermore, vaccines confer long-term immunity through the generation of memory T cell pools. Dendritic cells are essential for the activation of T cells and the induction of adaptive immunity, making them key for the in vitro evaluation of vaccine efficacy. Upon internalization by dendritic cells, vaccine-bearing antigens are processed, and suitable fragments are presented to T cells by major histocompatibility complex (MHC) molecules. In addition, DCs can secrete various cytokines to crosstalk with T cells to coordinate subsequent immune responses. Here, we generated an in vitro model using the immortalized murine dendritic cell line, DC2.4, to recapitulate the process of antigen uptake and DC maturation, measured as the elevation of CD40, MHC-II, CD80 and CD86 on the cell surface. The levels of key DC cytokines, tumor necrosis alpha (TNF-α) and interleukin-10 (IL-10) were measured to better define DC activation. This information served as a cost-effective and rapid proxy for assessing the antigen presentation efficacy of various vaccine formulations, demonstrating a strong correlation with previously published in vivo study outcomes. Hence, our assay enables the selection of the lead vaccine candidates based on DC activation capacity prior to in vivo animal studies.


Asunto(s)
Presentación de Antígeno , Células Dendríticas , Animales , Ratones , Antígenos CD40/metabolismo , Citocinas/metabolismo , Vacunas de Subunidad/metabolismo
2.
Adv Healthc Mater ; 13(7): e2302351, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38198823

RESUMEN

Coxiella burnetti is an intracellular bacterium that causes Q fever, a disease of worldwide importance. Q-VAX® , the approved human Q fever vaccine, is a whole cell vaccine associated with safety concerns. Here a safe particulate subunit vaccine candidate is developed that is ambient-temperature stable and can be cost-effectively manufactured. Endotoxin-free Escherichia coli is bioengineered to efficiently self-assemble biopolymer particles (BPs) that are densely coated with either strings of 18 T-cell epitopes (COX-BP) or two full-length immunodominant antigens (YbgF-BP-Com1) all derived from C. burnetii. BP vaccine candidates are ambient-temperature stable. Safety and immunogenicity are confirmed in mice and guinea pig (GP) models. YbgF-BP-Com1 elicits specific and strong humoral immune responses in GPs with IgG titers that are at least 1 000 times higher than those induced by Q-VAX® . BP vaccine candidates are not reactogenic. After challenge with C. burnetii, YbgF-BP-Com1 vaccine leads to reduced fever responses and pathogen burden in the liver and the induction of proinflammatory cytokines IL-12 and IFN-γ inducible protein (IP-10) when compared to negative control groups. These data suggest that YbgF-BP-Com1 induces functional immune responses reducing infection by C. burnetii. Collectively, these findings illustrate the potential of BPs as effective antigen carrier for Q fever vaccine development.


Asunto(s)
Coxiella burnetii , Fiebre Q , Humanos , Animales , Ratones , Cobayas , Fiebre Q/prevención & control , Coxiella burnetii/metabolismo , Vacunas Bacterianas , Inmunidad , Vacunas de Subunidad/metabolismo
3.
Braz J Microbiol ; 54(4): 3257-3264, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37792270

RESUMEN

Salmonella Typhimurium is the most prevalent non-host specific Salmonella serovars and a major concern for both human and animal health systems worldwide contributing to significant economic loss. Type 3 secretion system (T3SS) of Salmonella plays an important role in bacterial adherence and entry into the host epithelial cells. The product of invH gene of Salmonella is an important component of the needle complex of the type 3 secretion system. Hence, the present study was undertaken to clone and express the 15 kDa InvH surface protein of Salmonella Typhimurium in an E. coli host and to evaluate its immune potency in mice. The purified recombinant InvH (r-InvH) protein provoked a significant (p < 0.01) rise in IgG in the inoculated mice. The immunized mice were completely (100%) protected against the challenge dose of 107.5 LD50, while protection against challenge with the same dose of heterologous serovars was 90%. The bacterin-vaccinated group showed homologous protection of 60% against all three serovars. Findings in this study suggest the potential of the r-InvH protein of S. Typhimurium as an effective vaccine candidate against Salmonella infections.


Asunto(s)
Intoxicación Alimentaria por Salmonella , Salmonelosis Animal , Infecciones por Salmonella , Animales , Ratones , Humanos , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Escherichia coli/genética , Proteínas Bacterianas/metabolismo , Infecciones por Salmonella/prevención & control , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Vacunas de Subunidad/genética , Vacunas de Subunidad/metabolismo , Salmonelosis Animal/microbiología , Vacunas Atenuadas
4.
Circulation ; 147(9): 728-742, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36562301

RESUMEN

BACKGROUND: The metalloprotease ADAMTS-7 (a disintegrin and metalloproteinase with thrombospondin type 1 motif 7) is a novel locus associated with human coronary atherosclerosis. ADAMTS-7 deletion protects against atherosclerosis and vascular restenosis in rodents. METHODS: We designed 3 potential vaccines consisting of distinct B cell epitopic peptides derived from ADAMTS-7 and conjugated with the carrier protein KLH (keyhole limpet hemocyanin) as well as aluminum hydroxide as an adjuvant. Arterial ligation or wire injury was used to induce neointima in mice, whereas ApoE-/- and LDLR-/- (LDLR [low-density lipoprotein receptor]) mice fed a high-fat diet were applied to assess atherosclerosis. In addition, coronary stent implantation was performed on vaccine-immunized Bama miniature pigs, followed by optical coherence tomography to evaluate coronary intimal hyperplasia. RESULTS: A vaccine, ATS7vac, was screened out from 3 candidates to effectively inhibit intimal thickening in murine carotid artery ligation models after vaccination. As well, immunization with ATS7vac alleviated neointima formation in murine wire injury models and mitigated atherosclerotic lesions in both hyperlipidemic ApoE-/- and LDLR-/- mice without lowering lipid levels. Preclinically, ATS7vac markedly impeded intimal hyperplasia in swine stented coronary arteries, but without significant immune-related organ injuries. Mechanistically, ATS7vac vaccination produced specific antibodies against ADAMTS-7, which markedly repressed ADAMTS-7-mediated COMP (cartilage oligomeric matrix protein) and TSP-1 (thrombospondin-1) degradation and subsequently inhibited vascular smooth muscle cell migration but promoted re-endothelialization. CONCLUSIONS: ATS7vac is a novel atherosclerosis vaccine that also alleviates in-stent restenosis. The application of ATS7vac would be a complementary therapeutic avenue to the current lipid-lowering strategy for atherosclerotic disease.


Asunto(s)
Aterosclerosis , Neointima , Animales , Ratones , Proteínas ADAM/metabolismo , Aterosclerosis/patología , Modelos Animales de Enfermedad , Hiperplasia/metabolismo , Lípidos , Miocitos del Músculo Liso/metabolismo , Neointima/metabolismo , Porcinos , Trombospondinas/metabolismo , Vacunas de Subunidad/metabolismo , Proteína ADAMTS7
5.
BMC Bioinformatics ; 23(1): 311, 2022 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-35918631

RESUMEN

BACKGROUND: Cervical cancer is the fourth most common cancer affecting women and is caused by human Papillomavirus (HPV) infections that are sexually transmitted. There are currently commercially available prophylactic vaccines that have been shown to protect vaccinated individuals against HPV infections, however, these vaccines have no therapeutic effects for those who are previously infected with the virus. The current study's aim was to use immunoinformatics to develop a multi-epitope vaccine with therapeutic potential against cervical cancer. RESULTS: In this study, T-cell epitopes from E5 and E7 proteins of HPV16/18 were predicted. These epitopes were evaluated and chosen based on their antigenicity, allergenicity, toxicity, and induction of IFN-γ production (only in helper T lymphocytes). Then, the selected epitopes were sequentially linked by appropriate linkers. In addition, a C-terminal fragment of Mycobacterium tuberculosis heat shock protein 70 (HSP70) was used as an adjuvant for the vaccine construct. The physicochemical parameters of the vaccine construct were acceptable. Furthermore, the vaccine was soluble, highly antigenic, and non-allergenic. The vaccine's 3D model was predicted, and the structural improvement after refinement was confirmed using the Ramachandran plot and ProSA-web. The vaccine's B-cell epitopes were predicted. Molecular docking analysis showed that the vaccine's refined 3D model had a strong interaction with the Toll-like receptor 4. The structural stability of the vaccine construct was confirmed by molecular dynamics simulation. Codon adaptation was performed in order to achieve efficient vaccine expression in Escherichia coli strain K12 (E. coli). Subsequently, in silico cloning of the multi-epitope vaccine was conducted into pET-28a ( +) expression vector. CONCLUSIONS: According to the results of bioinformatics analyses, the multi-epitope vaccine is structurally stable, as well as a non-allergic and non-toxic antigen. However, in vitro and in vivo studies are needed to validate the vaccine's efficacy and safety. If satisfactory results are obtained from in vitro and in vivo studies, the vaccine designed in this study may be effective as a therapeutic vaccine against cervical cancer.


Asunto(s)
Papillomavirus Humano 16 , Neoplasias del Cuello Uterino , Biología Computacional/métodos , Epítopos de Linfocito B , Epítopos de Linfocito T/química , Escherichia coli/metabolismo , Femenino , Papillomavirus Humano 18/genética , Humanos , Simulación del Acoplamiento Molecular , Vacunas de Subunidad/química , Vacunas de Subunidad/metabolismo
6.
BioDrugs ; 36(5): 573-589, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35821564

RESUMEN

The idea of producing vaccines in plants originated in the late 1980s. Initially, it was contemplated that this notion could facilitate the concept of edible vaccines, making them more cost effective and easily accessible. Initial studies on edible vaccines focussed on the use of a variety of different transgenic plant host species for the production of vaccine antigens. However, adequate expression levels of antigens, the difficulties predicted with administration of consistent doses, and regulatory rules required for growth of transgenic plants gave way to the development of vaccine candidates that could be purified and administered parenterally. The field has subsequently advanced with improved expression techniques including a shift from using transgenic to transient expression of antigens, refinement of purification protocols, a deeper understanding of the biological processes and a wealth of evidence of immunogenicity and efficacy of plant-produced vaccine candidates, all contributing to the successful practice of what is now known as biopharming or plant molecular farming. The establishment of this technology has resulted in the development of many different types of vaccine candidates including subunit vaccines and various different types of nanoparticle vaccines targeting a wide variety of bacterial and viral diseases. This has brought further acceptance of plants as a suitable platform for vaccine production and in this review, we discuss the most recent advances in the production of vaccines in plants for human use.


Asunto(s)
Vacunación , Vacunas Comestibles , Antígenos , Humanos , Plantas Modificadas Genéticamente/genética , Plantas Modificadas Genéticamente/metabolismo , Vacunas Comestibles/genética , Vacunas de Subunidad/metabolismo
7.
Mol Pharm ; 19(7): 2638-2650, 2022 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-35621214

RESUMEN

Nanoparticle formulations have long been proposed as subunit vaccine carriers owing to their ability to entrap proteins and codeliver adjuvants. Poly(lactic-co-glycolic acid) (PLGA) remains one of the most studied polymers for controlled release and nanoparticle drug delivery, and numerous studies exist proposing PLGA particles as subunit vaccine carriers. In this work we report using PLGA nanoparticles modified with biotin (bNPs) to deliver proteins via adsorption and stimulate professional antigen-presenting cells (APCs). We present evidence showing bNPs are capable of retaining proteins through the biotin-avidin interaction. Surface accessible biotin bound both biotinylated catalase (bCAT) through avidin and streptavidin horseradish peroxidase (HRP). Analysis of the HRP found that activity on the bNPs was preserved once captured on the surface of bNP. Further, bNPs were found to have self-adjuvant properties, evidenced by bNP induced IL-1ß, IL-18, and IL-12 production in vitro in APCs, thereby licensing the cells to generate Th1-type helper T cell responses. Cytokine production was reduced in avidin precoated bNPs (but not with other proteins), suggesting that the proinflammatory response is due in part to exposed biotin on the surface of bNPs. bNPs injected subcutaneously were localized to draining lymph nodes detectable after 28 days and were internalized by bronchoalveolar lavage dendritic cells and macrophages in mice in a dose-dependent manner when delivered intranasally. Taken together, these data provide evidence that bNPs should be explored further as potential adjuvanting carriers for subunit vaccines.


Asunto(s)
Biotina , Nanopartículas , Adyuvantes Inmunológicos/química , Animales , Avidina , Células Dendríticas , Ratones , Nanopartículas/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Vacunas de Subunidad/metabolismo
8.
Front Immunol ; 12: 732298, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34745102

RESUMEN

Immune modulating therapies and vaccines are in high demand, not least to the recent global spread of SARS-CoV2. To achieve efficient activation of the immune system, professional antigen presenting cells have proven to be key coordinators of such responses. Especially targeted approaches, actively directing antigens to specialized dendritic cells, promise to be more effective and accompanied by reduced payload due to less off-target effects. Although antibody and glycan-based targeting of receptors on dendritic cells have been employed, these are often expensive and time-consuming to manufacture or lack sufficient specificity. Thus, we applied a small-molecule ligand that specifically binds Langerin, a hallmark receptor on Langerhans cells, conjugated to a model protein antigen. Via microneedle injection, this construct was intradermally administered into intact human skin explants, selectively loading Langerhans cells in the epidermis. The ligand-mediated cellular uptake outpaces protein degradation resulting in intact antigen delivery. Due to the pivotal role of Langerhans cells in induction of immune responses, this approach of antigen-targeting of tissue-resident immune cells offers a novel way to deliver highly effective vaccines with minimally invasive administration.


Asunto(s)
Antígenos CD/metabolismo , Antígenos/administración & dosificación , Proteínas Fluorescentes Verdes/administración & dosificación , Células de Langerhans/metabolismo , Lectinas Tipo C/metabolismo , Lectinas de Unión a Manosa/metabolismo , Animales , Antígenos/inmunología , Antígenos/metabolismo , Células COS , Chlorocebus aethiops , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Inyecciones Intradérmicas , Células de Langerhans/inmunología , Ligandos , Miniaturización , Nanomedicina , Agujas , Unión Proteica , Transporte de Proteínas , Proteolisis , Células THP-1 , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/metabolismo
9.
Adv Mater ; 33(51): e2104362, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34651342

RESUMEN

The development of effective vaccines that can be rapidly manufactured and distributed worldwide is necessary to mitigate the devastating health and economic impacts of pandemics like COVID-19. The receptor-binding domain (RBD) of the SARS-CoV-2 spike protein, which mediates host cell entry of the virus, is an appealing antigen for subunit vaccines because it is efficient to manufacture, highly stable, and a target for neutralizing antibodies. Unfortunately, RBD is poorly immunogenic. While most subunit vaccines are commonly formulated with adjuvants to enhance their immunogenicity, clinically-relevant adjuvants Alum, AddaVax, and CpG/Alum are found unable to elicit neutralizing responses following a prime-boost immunization. Here, it has been shown that sustained delivery of an RBD subunit vaccine comprising CpG/Alum adjuvant in an injectable polymer-nanoparticle (PNP) hydrogel elicited potent anti-RBD and anti-spike antibody titers, providing broader protection against SARS-CoV-2 variants of concern compared to bolus administration of the same vaccine and vaccines comprising other clinically-relevant adjuvant systems. Notably, a SARS-CoV-2 spike-pseudotyped lentivirus neutralization assay revealed that hydrogel-based vaccines elicited potent neutralizing responses when bolus vaccines did not. Together, these results suggest that slow delivery of RBD subunit vaccines with PNP hydrogels can significantly enhance the immunogenicity of RBD and induce neutralizing humoral immunity.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Hidrogeles/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunas de Subunidad/inmunología , Adyuvantes Inmunológicos/química , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , COVID-19/prevención & control , COVID-19/virología , Islas de CpG/genética , Femenino , Humanos , Inmunidad Humoral , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química , Polímeros/química , Dominios Proteicos/inmunología , SARS-CoV-2/química , SARS-CoV-2/inmunología , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/aislamiento & purificación , Vacunas de Subunidad/química , Vacunas de Subunidad/metabolismo
10.
Immunogenetics ; 73(6): 459-477, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34542663

RESUMEN

Since 2019, the world was involved with SARS-CoV-2 and consequently, with the announcement by the World Health Organization that COVID-19 was a pandemic, scientific were an effort to obtain the best approach to combat this global dilemma. The best way to prevent the pandemic from spreading further is to use a vaccine against COVID-19. Here, we report the design of a recombinant multi-epitope vaccine against the four proteins spike or crown (S), membrane (M), nucleocapsid (N), and envelope (E) of SARS-CoV-2 using immunoinformatics tools. We evaluated the most antigenic epitopes that bind to HLA class 1 subtypes, along with HLA class 2, as well as B cell epitopes. Beta-defensin 3 and PADRE sequence were used as adjuvants in the structure of the vaccine. KK, GPGPG, and AAY linkers were used to fuse the selected epitopes. The nucleotide sequence was cloned into pET26b(+) vector using restriction enzymes XhoI and NdeI, and HisTag sequence was considered in the C-terminal of the construct. The results showed that the proposed candidate vaccine is a 70.87 kDa protein with high antigenicity and immunogenicity as well as non-allergenic and non-toxic. A total of 95% of the selected epitopes have conservancy with similar sequences. Molecular docking showed a strong binding between the vaccine structure and tool-like receptor (TLR) 7/8. The docking, molecular dynamics, and MM/PBSA analysis showed that the vaccine established a stable interaction with both structures of TLR7 and TLR8. Simulation of immune stimulation by this vaccine showed that it evokes immune responses related to humoral and cellular immunity.


Asunto(s)
Vacunas contra la COVID-19/inmunología , Epítopos de Linfocito B/inmunología , Epítopos de Linfocito T/inmunología , SARS-CoV-2/inmunología , Secuencia de Aminoácidos , Secuencia de Bases , COVID-19/prevención & control , Vacunas contra la COVID-19/genética , Vacunas contra la COVID-19/metabolismo , Biología Computacional , Epítopos de Linfocito B/química , Epítopos de Linfocito B/genética , Epítopos de Linfocito T/química , Epítopos de Linfocito T/genética , Antígenos HLA/inmunología , Humanos , Inmunogenicidad Vacunal , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Peso Molecular , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Receptor Toll-Like 7/química , Receptor Toll-Like 8/química , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/metabolismo , Vacunología , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/inmunología
11.
J Am Chem Soc ; 143(36): 14748-14765, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34490778

RESUMEN

The COVID-19 pandemic highlights the need for platform technologies enabling rapid development of vaccines for emerging viral diseases. The current vaccines target the SARS-CoV-2 spike (S) protein and thus far have shown tremendous efficacy. However, the need for cold-chain distribution, a prime-boost administration schedule, and the emergence of variants of concern (VOCs) call for diligence in novel SARS-CoV-2 vaccine approaches. We studied 13 peptide epitopes from SARS-CoV-2 and identified three neutralizing epitopes that are highly conserved among the VOCs. Monovalent and trivalent COVID-19 vaccine candidates were formulated by chemical conjugation of the peptide epitopes to cowpea mosaic virus (CPMV) nanoparticles and virus-like particles (VLPs) derived from bacteriophage Qß. Efficacy of this approach was validated first using soluble vaccine candidates as solo or trivalent mixtures and subcutaneous prime-boost injection. The high thermal stability of our vaccine candidates allowed for formulation into single-dose injectable slow-release polymer implants, manufactured by melt extrusion, as well as microneedle (MN) patches, obtained through casting into micromolds, for prime-boost self-administration. Immunization of mice yielded high titers of antibodies against the target epitope and S protein, and data confirms that antibodies block receptor binding and neutralize SARS-CoV and SARS-CoV-2 against infection of human cells. We present a nanotechnology vaccine platform that is stable outside the cold-chain and can be formulated into delivery devices enabling single administration or self-administration. CPMV or Qß VLPs could be stockpiled, and epitopes exchanged to target new mutants or emergent diseases as the need arises.


Asunto(s)
Vacunas contra la COVID-19/metabolismo , COVID-19/epidemiología , COVID-19/prevención & control , Preparaciones de Acción Retardada/química , SARS-CoV-2/metabolismo , Vacunas de Subunidad/metabolismo , Animales , Comovirus , Simulación por Computador , Composición de Medicamentos , Epítopos/química , Calor , Humanos , Masculino , Ratones Endogámicos BALB C , Nanopartículas/química , Péptidos/química , Vacunación , Vacunas de Partículas Similares a Virus/química
12.
Biomolecules ; 11(2)2021 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-33671255

RESUMEN

SARS-CoV-2, or COVID-19, has a devastating effect on our society, both in terms of quality of life and death rates; hence, there is an urgent need for developing safe and effective therapeutics against SARS-CoV-2. The most promising strategy to fight against this deadly virus is to develop an effective vaccine. Internalization of SARS-CoV-2 into the human host cell mainly occurs through the binding of the coronavirus spike protein (a trimeric surface glycoprotein) to the human angiotensin-converting enzyme 2 (ACE2) receptor. The spike-ACE2 protein-protein interaction is mediated through the receptor-binding domain (RBD) of the spike protein. Mutations in the spike RBD can significantly alter interactions with the ACE2 host receptor. Due to its important role in virus transmission, the spike RBD is considered to be one of the key molecular targets for vaccine development. In this study, a spike RBD-based subunit vaccine was designed by utilizing a ferritin protein nanocage as a scaffold. Several fusion protein constructs were designed in silico by connecting the spike RBD via a synthetic linker (different sizes) to different ferritin subunits (H-ferritin and L-ferritin). The stability and the dynamics of the engineered nanocage constructs were tested by extensive molecular dynamics simulation (MDS). Based on our MDS analysis, a five amino acid-based short linker (S-Linker) was the most effective for displaying the spike RBD over the surface of ferritin. The behavior of the spike RBD binding regions from the designed chimeric nanocages with the ACE2 receptor was highlighted. These data propose an effective multivalent synthetic nanocage, which might form the basis for new vaccine therapeutics designed against viruses such as SARS-CoV-2.


Asunto(s)
Vacunas contra la COVID-19/química , COVID-19/virología , Ferritinas/química , Nanoestructuras/química , SARS-CoV-2/química , Glicoproteína de la Espiga del Coronavirus/química , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/metabolismo , Vacunas contra la COVID-19/metabolismo , Ferritinas/metabolismo , Humanos , Simulación de Dinámica Molecular , Conformación Proteica , Dominios Proteicos , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Vacunas de Subunidad/química , Vacunas de Subunidad/metabolismo
13.
J Phys Chem Lett ; 11(22): 9920-9930, 2020 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-33174418

RESUMEN

The emergence of severe acute respiratory syndrome from novel Coronavirus (SARS-CoV-2) has put an immense pressure worldwide where vaccination is believed to be an efficient way for developing hard immunity. Herein, we employ immunoinformatic tools to identify B-cell, T-cell epitopes associated with the spike protein of SARS-CoV-2, which is important for genome release. The results showed that the highly immunogenic epitopes located at the stalk part are mostly conserved compared to the receptor binding domain (RDB). Further, two vaccine candidates were computationally modeled from the linear B-cell, T-cell epitopes. Molecular docking reveals the crucial interactions of the vaccines with immune-receptors, and their stability is assessed by MD simulation studies. The chimeric vaccines showed remarkable binding affinity toward the immune cell receptors computed by the MM/PBSA method. van der Waals and electrostatic interactions are found to be the dominant factors for the stability of the complexes. The molecular-level interaction obtained from this study may provide deeper insight into the process of vaccine development against the pandemic of COVID-19.


Asunto(s)
Vacunas contra la COVID-19/inmunología , Epítopos de Linfocito B/inmunología , Epítopos de Linfocito T/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunas de Subunidad/inmunología , Secuencia de Aminoácidos , COVID-19/prevención & control , Vacunas contra la COVID-19/química , Vacunas contra la COVID-19/metabolismo , Epítopos de Linfocito B/química , Epítopos de Linfocito B/metabolismo , Epítopos de Linfocito T/química , Epítopos de Linfocito T/metabolismo , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Dominios Proteicos , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo , Vacunas de Subunidad/química , Vacunas de Subunidad/metabolismo
14.
Biosci Rep ; 40(12)2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33169789

RESUMEN

Immunotherapy is a breakthrough approach for cancer treatment and prevention. By exploiting the fact that cancer cells have overexpression of tumor antigens responsible for its growth and progression, which can be identified and removed by boosting the immune system. In silico techniques have provided efficient ways for developing preventive measures to ward off cancer. Herein, we have designed a potent cytotoxic T-lymphocyte epitope to elicit a desirable immune response against carcinogenic melanoma-associated antigen-A11. Potent epitope was predicted using reliable algorithms and characterized by advanced computational avenue CABS molecular dynamics simulation, for full flexible binding with HLA-A*0201 and androgen receptor to large-scale rearrangements of the complex system. Results showed the potent immunogenic construct (KIIDLVHLL), from top epitopes using five algorithms. Molecular docking analyses showed the strong binding of epitope with HLA-A*0201 and androgen receptor with docking score of -780.6 and -641.06 kcal/mol, respectively. Molecular dynamics simulation analysis revealed strong binding of lead epitope with androgen receptor by involvement of 127 elements through atomic-model study. Full flexibility study showed stable binding of epitope with an average root mean square deviation (RMSD) 2.21 Å and maximum RMSD value of 6.48 Å in optimal cluster density area. The epitope also showed remarkable results with radius of gyration 23.0777 Å, world population coverage of 39.08% by immune epitope database, and transporter associated with antigen processing (TAP) affinity IC50 value of 2039.65 nm. Moreover, in silico cloning approach confirmed the expression and translation capacity of the construct within a suitable expression vector. The present study paves way for a potential immunogenic construct for prevention of cancer.


Asunto(s)
Antígenos de Neoplasias/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Citotoxicidad Inmunológica , Diseño de Fármacos , Epítopos de Linfocito T , Proteínas de Neoplasias/uso terapéutico , Neoplasias/terapia , Linfocitos T Citotóxicos/inmunología , Algoritmos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/metabolismo , Antígeno HLA-A2/inmunología , Antígeno HLA-A2/metabolismo , Humanos , Inmunogenicidad Vacunal , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Unión Proteica , Receptores Androgénicos/inmunología , Receptores Androgénicos/metabolismo , Linfocitos T Citotóxicos/metabolismo , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/metabolismo , Vacunas de Subunidad/uso terapéutico
15.
Infect Genet Evol ; 85: 104517, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32882432

RESUMEN

The present study aimed to predict a novel chimeric vaccine by simultaneously targeting four major structural proteins via the establishment of ancestral relationship among different strains of coronaviruses. Conserved regions from the homologous protein sets of spike glycoprotein, membrane protein, envelope protein and nucleocapsid protein were identified through multiple sequence alignment. The phylogeny analyses of whole genome stated that four proteins reflected the close ancestral relation of SARS-CoV-2 to SARS-COV-1 and bat coronavirus. Numerous immunogenic epitopes (both T cell and B cell) were generated from the common fragments which were further ranked on the basis of antigenicity, transmembrane topology, conservancy level, toxicity and allergenicity pattern and population coverage analysis. Top putative epitopes were combined with appropriate adjuvants and linkers to construct a novel multiepitope subunit vaccine against COVID-19. The designed constructs were characterized based on physicochemical properties, allergenicity, antigenicity and solubility which revealed the superiority of construct V3 in terms safety and efficacy. Essential molecular dynamics and normal mode analysis confirmed minimal deformability of the refined model at molecular level. In addition, disulfide engineering was investigated to accelerate the stability of the protein. Molecular docking study ensured high binding affinity between construct V3 and HLA cells, as well as with different host receptors. Microbial expression and translational efficacy of the constructs were checked using pET28a(+) vector of E. coli strain K12. However, the in vivo and in vitro validation of suggested vaccine molecule might be ensured with wet lab trials using model animals for the implementation of the presented data.


Asunto(s)
Epítopos de Linfocito B/genética , Epítopos de Linfocito T/genética , SARS-CoV-2/clasificación , Vacunas de Subunidad/genética , Proteínas Estructurales Virales/genética , Epítopos de Linfocito B/inmunología , Epítopos de Linfocito T/inmunología , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Evolución Molecular , Genoma Viral , Humanos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Filogenia , Conformación Proteica , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Vacunas de Subunidad/química , Vacunas de Subunidad/metabolismo , Proteínas Estructurales Virales/química , Proteínas Estructurales Virales/inmunología , Vacunas Virales/química , Vacunas Virales/genética , Vacunas Virales/metabolismo
16.
J Biotechnol ; 322: 10-20, 2020 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-32659239

RESUMEN

One of the milestones of vaccinology is the depletion of the global impact of Poliomyelitis. The current vaccines to deal with Polio comprise the Sabin and Salk formulations. The main limitation of the former is the use of attenuated viruses that can revert into pathogenic forms, whereas the latter is more expensive and induces no protection in the intestinal tract; the site of virus replication. Genetically engineered plants cope with such limitations. In addition, they offer a low-cost alternative for production, storage and delivery of vaccines. This technology has been narrowly applied in the development of Polio vaccines. Herein, we explored the ability of tobacco cells to express the immunogenic VP1, VP2, VP3, and VP4 Polio antigens, which are relevant for vaccine development. Evidence on the expression of the plant-made Polio VPs is presented and an immunogenicity assessment proved their capacity to induce local and systemic humoral responses when administered by subcutaneous and oral routes. The plant-made VPs will be useful in the development of low-cost vaccine formulations able to induce effective mucosal immunity without the risks associated to the use of attenuated viruses; therefore there is a potential for this technology to contribute toward Polio eradication.


Asunto(s)
Proteínas de la Cápside , Nicotiana/genética , Vacuna Antipolio Oral , Poliovirus , Vacunas de Subunidad , Animales , Anticuerpos Antivirales/análisis , Anticuerpos Antivirales/sangre , Antígenos Virales/genética , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/metabolismo , Heces/química , Masculino , Ratones , Ratones Endogámicos BALB C , Agricultura Molecular , Plantas Modificadas Genéticamente/genética , Poliomielitis/prevención & control , Poliomielitis/virología , Poliovirus/genética , Poliovirus/inmunología , Vacuna Antipolio Oral/genética , Vacuna Antipolio Oral/inmunología , Vacuna Antipolio Oral/metabolismo , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/metabolismo
17.
Chempluschem ; 85(1): 227-236, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31944609

RESUMEN

Self-adjuvanting vaccines, consisting of recombinant protein antigens and covalently attached Toll-like receptor (TLR) agonists, have the ability to simultaneously and efficiently deliver antigen and TLR adjuvant to antigen presenting cells (APCs). Here, an enzyme-mediated ligation approach was used to overcome difficulties in producing homogeneous, molecularly defined self-adjuvanting vaccine products under native conditions. This process was optimized to allow the incorporation of the lipopeptide TLR2 agonist fibroblast-stimulating lipopeptide (FSL)-1 onto the N- or C-termini of recombinant protein antigens, employing the enzyme Staphylococcus aureus sortase A (SrtAsa) penta mutant. In addition, because SrtAsa-mediated ligations are reversible, a tryptophan zipper derived sequence was introduced into both reactants, which was demonstrated to improve ligation efficiency through the formation of a ß-hairpin structure that hinders the reverse reaction. Finally, it was demonstrated that N- or C-terminal conjugation, and the incorporation of the ß-hairpin structure, did not affect the TLR2-agonist activities of protein antigen TLR agonist conjugates. Overall, this SrtAsa-mediated ligation platform enabled production of antigen TLR2 agonist conjugates with enhanced ligation efficiency, with the conjugates demonstrating potent TLR2 signaling activation (EC50 <1nM).


Asunto(s)
Adyuvantes Inmunológicos/metabolismo , Aminoaciltransferasas/metabolismo , Proteínas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Proteínas Recombinantes/metabolismo , Receptor Toll-Like 2/metabolismo , Vacunas de Subunidad/metabolismo , Adyuvantes Inmunológicos/química , Aminoaciltransferasas/química , Aminoaciltransferasas/genética , Células Presentadoras de Antígenos/inmunología , Antígenos/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/genética , Fibroblastos/metabolismo , Regulación Bacteriana de la Expresión Génica , Humanos , Inmunización , Ligandos , Lipopéptidos/metabolismo , Mutación , Proteínas Recombinantes/química , Staphylococcus aureus/enzimología , Staphylococcus aureus/genética , Receptor Toll-Like 2/química , Triptófano/metabolismo , Vacunas de Subunidad/química , Vacunas de Subunidad/inmunología
18.
Infect Disord Drug Targets ; 20(4): 523-530, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31109281

RESUMEN

BACKGROUND: Chikungunya an arbovirus, is transmitted to humans by the bite of Aedes mosquito. The virus occurrences have been reported in Southeast Asian countries including Pakistan. Its symptoms include typical febrile illness and arthralgic syndrome. The virus has not decisively proved to be life-threatening. METHODS: The attempt was to design T-cell and B-cell epitope-based vaccine for Chikungunya. The proteome of chikungunya was retrieved, antigenic proteins were identified and T-cell epitopes and B-cell epitopes were predicted. Interacting HLA alleles were also identified. The final analysis was done to confirm that predicted T-cell epitopes and B-cell epitopes can be used as a vaccine. RESULTS: About 32 T-cell epitopes and a 10mer B-cell epitope were identified. Both T-cell and Bcell epitopes demonstrated strong interactions with HLA alleles. The predicted T-cell and B-cell epitopes were docked with respective HLA alleles. The docking analysis showed that the predicted respective epitopes best fit into the binding pockets of the alleles. CONCLUSION: On the basis of this computational analysis, it is suggested that these predicted epitopes can be used as a remedy against Alphavirus strain of chikungunya. Further laboratory experiments can be conducted to determine the efficacy and stability of this work.


Asunto(s)
Virus Chikungunya/genética , Epítopos de Linfocito B/genética , Epítopos de Linfocito T/genética , Vacunas de Subunidad/genética , Vacunas Virales/genética , Alelos , Secuencia de Aminoácidos , Fiebre Chikungunya/prevención & control , Fiebre Chikungunya/virología , Biología Computacional/métodos , Simulación por Computador , Epítopos de Linfocito B/química , Epítopos de Linfocito B/metabolismo , Epítopos de Linfocito T/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Vacunas de Subunidad/química , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/metabolismo
19.
Annu Rev Chem Biomol Eng ; 10: 337-359, 2019 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-31173518

RESUMEN

Although vaccines have been the primary defense against widespread infectious disease for decades, there is a critical need for improvement to combat complex and variable diseases. More control and specificity over the immune response can be achieved by using only subunit components in vaccines. However, these often lack sufficient immunogenicity to fully protect, and conjugation or carrier materials are required. A variety of protein and peptide biomaterials have improved effectiveness and delivery of subunit vaccines for infectious, cancer, and autoimmune diseases. They are biodegradable and have control over both material structure and immune function. Many of these materials are built from naturally occurring self-assembling proteins, which have been engineered for incorporation of vaccine components. In contrast, others are de novo designs of structures with immune function. In this review, protein biomaterial design, engineering, and immune functionality as vaccines or immunotherapies are discussed.


Asunto(s)
Materiales Biocompatibles/química , Inmunoterapia , Péptidos/inmunología , Vacunas de Subunidad/inmunología , Antígenos/inmunología , Antígenos/metabolismo , Enfermedades Transmisibles/inmunología , Enfermedades Transmisibles/terapia , Humanos , Nanoestructuras/química , Neoplasias/inmunología , Neoplasias/terapia , Péptidos/química , Péptidos/metabolismo , Vacunas de Subunidad/química , Vacunas de Subunidad/metabolismo , Vacunas de Partículas Similares a Virus/inmunología
20.
Pathog Dis ; 77(3)2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31093663

RESUMEN

Zika virus (ZIKV) is an arbovirus which shares antigenic similarity and the mosquito vector with dengue viruses (DENVs). ZIKV is a neurotropic virus capable of causing congenital neurodevelopmental birth defects. As ZIKV antibodies (Abs) can potentially enhance infection by DENVs, a preventive ZIKV vaccine must be designed to eliminate antibody dependent enhancement of infection. We developed a Zika Subunit Vaccine (ZSV) consisting of two proteins, ZS and S, in a genetically pre-determined ratio of 1:4, using the methylotrophic yeast Pichia pastoris. ZS is an in-frame fusion of ZIKV envelope domain III with the Hepatitis B virus (HBV) surface antigen, and S is the un-fused HBV surface antigen. Using specific monoclonal Abs we showed the presence of ZS and S in the co-purified material which were found to co-assemble into virus-like particles (VLPs), based on dynamic light scattering and electron microscopic analyses. These VLPs were immunogenic in BALB/c mice, eliciting Abs capable of neutralizing ZIKV reporter virus particles. Further, the VLP-induced Abs did not enhance a sub-lethal DENV-2 challenge in AG129 mice. This important safety feature, coupled to the well-documented advantage of P. pastoris expression system, warrants further exploration of ZSV VLP as a possible vaccine candidate.


Asunto(s)
Pichia/metabolismo , Multimerización de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Vacunas de Partículas Similares a Virus/inmunología , Proteínas del Envoltorio Viral/metabolismo , Virosomas/metabolismo , Virus Zika/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Acrecentamiento Dependiente de Anticuerpo , Virus del Dengue/patogenicidad , Femenino , Masculino , Ratones Endogámicos BALB C , Pichia/genética , Proteínas Recombinantes de Fusión/genética , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/aislamiento & purificación , Vacunas de Subunidad/metabolismo , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/aislamiento & purificación , Vacunas Sintéticas/metabolismo , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/aislamiento & purificación , Vacunas de Partículas Similares a Virus/metabolismo , Proteínas del Envoltorio Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...