Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Fish Shellfish Immunol ; 154: 109893, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39260531

RESUMEN

Piscirickettsia salmonis, the primary bacterial disease in Chilean salmon farming, necessitates a constant refinement of control strategies. This study hypothesized that the current vaccination strategy for SRS control in the Chilean Atlantic salmon aquaculture industry, which has been in place since 2017 (ALPHA JECT® 5.1 plus LiVac®), solely relies on vaccines formulated with the EM-90 genogroup of P. salmonis (PS-EM-90), triggering a partial cross-immunity response in fish infected with the LF-89 genogroup (PS-LF-89). Relative Percent Survival (RPS) and cell-mediated immune (CMI) response were evaluated in Atlantic salmon post-smolts vaccinated with the standard vaccination strategy but challenged with both PS-EM-90 and PS-LF-89, in addition to other vaccination strategies considering primo vaccination and booster with other commercial vaccines and the possible enhancing effects of the combination with a natural immunomodulator (PAQ-Xtract®) administered orally. The intraperitoneal (I.P.) challenge was performed after 2395°-days (DD) after the start of the immunostimulant delivery, 1905 DD after the primo vaccination, and 1455 DD after the booster vaccination. Unvaccinated fish showed 73.6 and 41.7 % mortality when challenged with PS-EM-90 and PS-LF-89, respectively. Fish infected with PS-LF-89 died significantly faster (21 days post-infection, dpi) than fish challenged with PS-EM-90 (28 dpi) (p = 0.0043) and had a higher probability of death (0.4626) than fish challenged with PS-EM-90. RPS had a significant positive correlation with the PS-EM-90 load of the P. salmonis genogroup (r = 0.540, p < 0.01) but not with the PS-LF-89 load (r = 0.155, p > 0.05). This demonstrated that the immunization strategies were more effective in lowering PS-EM-90 loads, resulting in higher survival rates in fish challenged with PS-EM-90. The current industry vaccination strategy recorded a 100 % RPS when fish were challenged with PS-EM-90, but the RPS dropped significantly to 77 % when fish were challenged with PS-LF-89, meaning that the strategy did not show complete cross-protection. But after adding PAQ-Xtract®, the RPS improved from 77 % to 92 % in fish that were vaccinated with the standard method but then challenged with PS-LF-89. The most effective vaccination strategy was based on LiVac® as primo vaccination and ALPHA JECT® 5.1 plus LiVac® as booster vaccination, with or without PAQ-Xtract®, in both PS-EM-90 (100 %) and PS-LF-89 (96 %) challenged fish. The serum concentration of anti-P. salmonis IgM did not show a correlation with the protection of immunization strategies expressed in survival. Low serum IL-12 and high serum IFNγ concentrations showed a correlation with higher bacterial loads and lower survival. Aggregate analysis showed a significant correlation between higher numbers of CD8+ cells in the head-kidney, higher fish survival, and a lower bacterial load. The immunization strategies were safe for fish and induced only mild microscopic lesions in the gut. Taken together, our results help to better understand the biological interaction between P. salmonis and post-smolt vaccinated Atlantic salmon to deepen the knowledge on vaccine-induced protection, CMI immune response, and cross-immunity applied to improve the current immunization strategy for SRS control in the Chilean salmon industry.

2.
Epidemics ; 48: 100776, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38944025

RESUMEN

Influenza A has two hemagglutinin groups, with stronger cross-immunity to reinfection within than between groups. Here, we explore the implications of this heterogeneity for proposed cross-protective influenza vaccines that may offer broad, but not universal, protection. While the development goal for the breadth of human influenza A vaccine is to provide cross-group protection, vaccines in current development stages may provide better protection against target groups than non-target groups. To evaluate vaccine formulation and strategies, we propose a novel perspective: a vaccine population-level target product profile (PTPP). Under this perspective, we use dynamical models to quantify the epidemiological impacts of future influenza A vaccines as a function of their properties. Our results show that the interplay of natural and vaccine-induced immunity could strongly affect seasonal subtype dynamics. A broadly protective bivalent vaccine could lower the incidence of both groups and achieve elimination with sufficient vaccination coverage. However, a univalent vaccine at low vaccination rates could permit a resurgence of the non-target group when the vaccine provides weaker immunity than natural infection. Moreover, as a proxy for pandemic simulation, we analyze the invasion of a variant that evades natural immunity. We find that a future vaccine providing sufficiently broad and long-lived cross-group protection at a sufficiently high vaccination rate, could prevent pandemic emergence and lower the pandemic burden. This study highlights that as well as effectiveness, breadth and duration should be considered in epidemiologically informed TPPs for future human influenza A vaccines.


Asunto(s)
Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Humanos , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Gripe Humana/epidemiología , Gripe Humana/inmunología , Virus de la Influenza A/inmunología , Protección Cruzada/inmunología
3.
J Gen Virol ; 105(6)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38861287

RESUMEN

Increased human-to-human transmission of monkeypox virus (MPXV) is cause for concern, and antibodies directed against vaccinia virus (VACV) are known to confer cross-protection against Mpox. We used 430 serum samples derived from the Scottish patient population to investigate antibody-mediated cross-neutralization against MPXV. By combining electrochemiluminescence immunoassays with live-virus neutralization assays, we show that people born when smallpox vaccination was routinely offered in the United Kingdom have increased levels of antibodies that cross-neutralize MPXV. Our results suggest that age is a risk factor of Mpox infection, and people born after 1971 are at higher risk of infection upon exposure.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Monkeypox virus , Mpox , Vacuna contra Viruela , Humanos , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Vacuna contra Viruela/inmunología , Vacuna contra Viruela/administración & dosificación , Adulto , Persona de Mediana Edad , Monkeypox virus/inmunología , Adulto Joven , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Mpox/inmunología , Mpox/prevención & control , Femenino , Adolescente , Anciano , Masculino , Protección Cruzada/inmunología , Escocia , Factores de Edad , Pruebas de Neutralización , Niño , Vacunación , Viruela/prevención & control , Viruela/inmunología , Preescolar , Reacciones Cruzadas , Anciano de 80 o más Años
4.
Front Immunol ; 15: 1401728, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38827749

RESUMEN

Background: Immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is now widespread; however, the degree of cross-immunity between SARS-CoV-2 and endemic, seasonal human coronaviruses (HCoVs) remains unclear. Methods: SARS-CoV-2 and HCoV cross-immunity was evaluated in adult participants enrolled in a US sub-study in the phase III, randomized controlled trial (NCT04516746) of AZD1222 (ChAdOx1 nCoV-19) primary-series vaccination for one-year. Anti-HCoV spike-binding antibodies against HCoV-229E, HCoV-HKU1, HCoV-OC43, and HCoV-NL63 were evaluated in participants following study dosing and, in the AZD1222 group, after a non-study third-dose booster. Timing of SARS-CoV-2 seroconversion (assessed via anti-nucleocapsid antibody levels) and incidence of COVID-19 were evaluated in those who received AZD1222 primary-series by baseline anti-HCoV titers. Results: We evaluated 2,020/21,634 participants in the AZD1222 group and 1,007/10,816 in the placebo group. At the one-year data cutoff (March 11, 2022) mean duration of follow up was 230.9 (SD: 106.36, range: 1-325) and 94.3 (74.12, 1-321) days for participants in the AZD1222 (n = 1,940) and placebo (n = 962) groups, respectively. We observed little elevation in anti-HCoV humoral titers post study-dosing or post-boosting, nor evidence of waning over time. The occurrence and timing of SARS-CoV-2 seroconversion and incidence of COVID-19 were not largely impacted by baseline anti-HCoV titers. Conclusion: We found limited evidence for cross-immunity between SARS-CoV-2 and HCoVs following AZD1222 primary series and booster vaccination. Susceptibility to future emergence of novel coronaviruses will likely persist despite a high prevalence of SARS-CoV-2 immunity in global populations.


Asunto(s)
Anticuerpos Antivirales , Vacunas contra la COVID-19 , COVID-19 , ChAdOx1 nCoV-19 , Inmunidad Humoral , SARS-CoV-2 , Humanos , COVID-19/inmunología , COVID-19/prevención & control , ChAdOx1 nCoV-19/inmunología , Adulto , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , SARS-CoV-2/inmunología , Masculino , Femenino , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/administración & dosificación , Persona de Mediana Edad , Inmunidad Humoral/efectos de los fármacos , Reacciones Cruzadas/inmunología , Estaciones del Año , Adulto Joven , Vacunación , Método Doble Ciego
5.
mBio ; 15(2): e0272223, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38270455

RESUMEN

Numerous clinical parameters link to severe coronavirus disease 2019, but factors that prevent symptomatic disease remain unknown. We investigated the impact of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) and endemic human coronavirus (HCoV) antibody responses on symptoms in a longitudinal children cohort (n = 2,917) and a cross-sectional cohort including children and adults (n = 882), all first exposed to SARS-CoV-2 (March 2020 to March 2021) in Switzerland. Saliva (n = 4,993) and plasma (n = 7,486) antibody reactivity to the four HCoVs (subunit S1 [S1]) and SARS-CoV-2 (S1, receptor binding domain, subunit S2 [S2], nucleocapsid protein) was determined along with neutralizing activity against SARS-CoV-2 Wuhan, Alpha, Delta, and Omicron (BA.2) in a subset of individuals. Inferred recent SARS-CoV-2 infection was associated with a strong correlation between mucosal and systemic SARS-CoV-2 anti-spike responses. Individuals with pre-existing HCoV-S1 reactivity exhibited significantly higher antibody responses to SARS-CoV-2 in both plasma (IgG regression coefficients = 0.20, 95% CI = [0.09, 0.32], P < 0.001) and saliva (IgG regression coefficient = 0.60, 95% CI = [0.088, 1.11], P = 0.025). Saliva neutralization activity was modest but surprisingly broad, retaining activity against Wuhan (median NT50 = 32.0, 1Q-3Q = [16.4, 50.2]), Alpha (median NT50 = 34.9, 1Q-3Q = [26.0, 46.6]), and Delta (median NT50 = 28.0, 1Q-3Q = [19.9, 41.7]). In line with a rapid mucosal defense triggered by cross-reactive HCoV immunity, asymptomatic individuals presented with higher pre-existing HCoV-S1 activity in plasma (IgG HKU1, odds ratio [OR] = 0.53, 95% CI = [0.29,0.97], P = 0.038) and saliva (total HCoV, OR = 0.55, 95% CI = [0.33, 0.91], P = 0.019) and higher SARS-CoV-2 reactivity in saliva (IgG S2 fold change = 1.26, 95% CI = [1.03, 1.54], P = 0.030). By investigating the systemic and mucosal immune responses to SARS-CoV-2 and HCoVs in a population without prior exposure to SARS-CoV-2 or vaccination, we identified specific antibody reactivities associated with lack of symptom development.IMPORTANCEKnowledge of the interplay between human coronavirus (HCoV) immunity and severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection is critical to understanding the coexistence of current endemic coronaviruses and to building knowledge potential future zoonotic coronavirus transmissions. This study, which retrospectively analyzed a large cohort of individuals first exposed to SARS-CoV-2 in Switzerland in 2020-2021, revealed several key findings. Pre-existing HCoV immunity, particularly mucosal antibody responses, played a significant role in improving SARS-CoV-2 immune response upon infection and reducing symptoms development. Mucosal neutralizing activity against SARS-CoV-2, although low in magnitude, retained activity against SARS-CoV-2 variants underlining the importance of maintaining local mucosal immunity to SARS-CoV-2. While the cross-protective effect of HCoV immunity was not sufficient to block infection by SARS-CoV-2, the present study revealed a remarkable impact on limiting symptomatic disease. These findings support the feasibility of generating pan-protective coronavirus vaccines by inducing potent mucosal immune responses.


Asunto(s)
COVID-19 , Adulto , Niño , Humanos , SARS-CoV-2 , Estudios Transversales , Estudios Retrospectivos , Inmunoglobulina G , Anticuerpos Antivirales , Glicoproteína de la Espiga del Coronavirus
6.
BMC Infect Dis ; 23(1): 757, 2023 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-37924004

RESUMEN

BACKGROUND: The general human immune responses similarity against different coronaviruses may reflect some degree of cross-immunity, whereby exposure to one coronavirus may confer partial immunity to another. The aim was to determine whether previous MERS-CoV infection was associated with a lower risk of subsequent COVID-19 disease and its related outcomes. METHODS: We conducted a retrospective cohort study among all patients screened for MERS-CoV at a tertiary care hospital in Saudi Arabia between 2012 and early 2020. Both MERS-CoV positive and negative patients were followed up from early 2020 to September 2021 for developing COVID-19 infection confirmed by RT-PCR testing. RESULTS: A total of 397 participants followed for an average 15 months during COVID-19 pandemic (4.9 years from MERS-CoV infection). Of the 397 participants, 93 (23.4%) were positive for MERS-CoV at baseline; 61 (65.6%) of the positive cases were symptomatic. Out of 397, 48 (12.1%) participants developed COVID-19 by the end of the follow-up period. Cox regression analysis adjusted for age, gender, and major comorbidity showed a marginally significant lower risk of COVID-19 disease (hazard ratio = 0.533, p = 0.085) and hospital admission (hazard ratio = 0.411, p = 0.061) in patients with positive MERS-CoV. Additionally, the risk of COVID-19 disease was further reduced and became significant in patients with symptomatic MERS-CoV infection (hazard ratio = 0.324, p = 0.034) and hospital admission (hazard ratio = 0.317, p = 0.042). CONCLUSIONS: The current findings may indicate a partial cross-immunity, where patients with symptomatic MERS-CoV have a lower risk of future COVID-19 infection and related hospitalization. The present results may need further examination nationally using immunity markers.


Asunto(s)
COVID-19 , Coronavirus del Síndrome Respiratorio de Oriente Medio , Humanos , COVID-19/epidemiología , Estudios Retrospectivos , Pandemias , Riesgo , Arabia Saudita/epidemiología
7.
Math Biosci Eng ; 20(9): 16083-16113, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37920004

RESUMEN

We introduce a two-strain model with asymmetric temporary immunity periods and partial cross-immunity. We derive explicit conditions for competitive exclusion and coexistence of the strains depending on the strain-specific basic reproduction numbers, temporary immunity periods, and degree of cross-immunity. The results of our bifurcation analysis suggest that, even when two strains share similar basic reproduction numbers and other epidemiological parameters, a disparity in temporary immunity periods and partial or complete cross-immunity can provide a significant competitive advantage. To analyze the dynamics, we introduce a quasi-steady state reduced model which assumes the original strain remains at its endemic steady state. We completely analyze the resulting reduced planar hybrid switching system using linear stability analysis, planar phase-plane analysis, and the Bendixson-Dulac criterion. We validate both the full and reduced models with COVID-19 incidence data, focusing on the Delta (B.1.617.2), Omicron (B.1.1.529), and Kraken (XBB.1.5) variants. These numerical studies suggest that, while early novel strains of COVID-19 had a tendency toward dramatic takeovers and extinction of ancestral strains, more recent strains have the capacity for co-existence.


Asunto(s)
COVID-19 , Enfermedades Transmisibles , Humanos , Enfermedades Transmisibles/epidemiología , Número Básico de Reproducción , COVID-19/epidemiología
8.
Math Biosci ; 365: 109086, 2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37821025

RESUMEN

The bacteria Vibrio cholerae relies heavily upon an aquatic reservoir as a transmission route with two distinct serotypes observed in many recent outbreaks. In this paper, we extend previously studied ordinary differential equation epidemiological models to create a two-strain SIRP (susceptible-infectious-recovered-pathogen) system which incorporates both partial cross-immunity between disease strains and environmental pathogen transmission. Of particular interest are undamped anti-phase periodic solutions, as these display a type of coexistence where strains routinely switch dominance, and understanding what drives this switch can optimize the efficiency of the host population's control measures against the disease. We derive the basic reproduction number R0 and use stability analysis to examine the disease free and single-strain equilibria. We formulate a unique coexistence equilibrium and prove uniform persistence of both strains when R0>1. In addition, we simulate solutions to this system, along with seasonally forced versions of the model with and without host coinfection. Cross-immunity and transmission pathways influence damped or sustained oscillatory dynamics, where the presence of seasonality can modify, amplify or synchronize the period and phase of serotypes, driving epidemic waves. Cycling of serotypes over large time intervals, similar to observed data, is found for a range of cross-immunity levels, and the inclusion of coinfection in the model contributes to sustained anti-phase periodic solutions.

9.
Vaccine ; 41(43): 6470-6482, 2023 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-37718187

RESUMEN

Coxsackievirus A2 (CVA2) is one of the causative agents of hand-foot-and-mouth disease (HFMD), which poses a great challenge for global public health. However, presently, there are no available commercial vaccines or antivirals to prevent CVA2 infection. Here, we present an inactivated Vero cell-based whole CVA2 vaccine candidate and evaluate its safety and efficacy in this study. Neonatal BALB/c mice were vaccinated at 5 and 7 days old, respectively, and then challenged with either homologous or heterologous strain of CVA2 at a lethal dose at 10 days old. The inactivated whole CVA2 vaccine candidate showed a high protective efficacy. Additionally, our inactivated vaccine stimulated the production of CVA2-specific IgG1 and IgG2a antibodies in vivo and high titers of neutralization antibodies (NtAbs) in the serum of immunized mice. Maternal immunization with the inactivated CVA2 vaccine provided full protection to pups against lethal infection. Compared with mice inoculated with only alum, the viral loads were decreased, and pathological changes were relieved in tissue samples of immunized mice. Moreover, the transcription levels of some genes related to cytokines (IFN-γ and TNF-α, MCP-1, IL-6, CXCL-10 etc.) were significantly reduced. The number of immune cells and levels of cytokines in peripheral blood of mice inoculated with only alum were higher than that of immunized mice. It is noteworthy that this vaccine showed a good cross-immunity efficacy against Enterovirus A71 (EVA71) challenge. In conclusion, our findings suggest that this experimental inactivated CVA2 vaccine is a promising component of polyvalent vaccines related to HFMD in the near future.

10.
J Math Biol ; 87(3): 42, 2023 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-37573266

RESUMEN

Multi-strain diseases lead to the development of some degree of cross-immunity among people. In the present paper, we propose a multi-delayed SIRC epidemic model with incubation and immunity time delays. Here we aim to examine and investigate the effects of incubation delay [Formula: see text] and the impact of vaccine which provides partial/cross-immunity with immunity delay parameter ([Formula: see text]) on the disease dynamics. Also, we study the impact of the strength of cross-immunity [Formula: see text] on the disease prevalence. The positivity and boundedness of the solutions of the epidemic model have been established. Two different types of equilibrium points (disease-free and endemic) have been deduced. Expression for basic reproduction number has been derived. The stability conditions and Hopf-bifurcation about both the equilibrium points in the absence and presence of both delays have been discussed. The Lyapunov stability conditions about the endemic equilibrium point have been established. Numerical simulations have been performed to support our analytical results. We quantitatively demonstrate how oscillations and Hopf-bifurcation allow time delays to alter the dynamics of the system. The combined impacts of both the delays on disease prevalence has been studied. Through parameter sensitivity analysis, we observe that the infected population decreases with an increase in vaccination rate and the system starts to stabilize early with the increase in cross-immunity rate. Global sensitivity analysis for the basic reproduction number has been performed using Latin hypercube sampling and partial rank correlation coefficients techniques. The combined effect of vaccination rate with transmission rate and vaccination rate with re-infection probability (i.e. strength of cross-immunity) on [Formula: see text] have been discussed. Our research underlines the need to take cross-immunity and time delays into account in the epidemic model in order to better understand disease dynamics.


Asunto(s)
Epidemias , Humanos , Simulación por Computador , Factores de Tiempo , Epidemias/prevención & control , Número Básico de Reproducción , Vacunación , Modelos Biológicos
11.
Evolution ; 77(11): 2392-2408, 2023 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-37592809

RESUMEN

The time-honored paradigm in the theory of virulence evolution assumes a positive relation between infectivity and harmfulness. However, the etiology of respiratory diseases yields a negative relation, with diseases of the lower respiratory tract being less infective and more harmful. We explore the evolutionary consequences in a simple model incorporating cross-immunity between disease strains that diminishes with their distance in the respiratory tract, assuming that docking rate follows the match between the local mix of cell surface types and the pathogen's surface and cross-immunity the similarity of the pathogens' surfaces. The assumed relation between fitness components causes virulent strains infecting the lower airways to evolve to milder more transmissible variants. Limited cross-immunity, generally, causes a readiness to diversify that increases with host population density. In respiratory diseases that diversity will be highest in the upper respiratory tract. More tentatively, emerging respiratory diseases are likely to start low and virulent, to evolve up, and become milder. Our results extend to a panoply of realistic generalizations of the disease's ecology to including additional epitope axes. These extensions allow us to apply our results quantitatively to elucidate the differences in diversification between rhino- and coronavirus caused common colds.


Asunto(s)
Evolución Biológica , Enfermedades Respiratorias , Humanos , Virulencia
12.
Front Cell Infect Microbiol ; 13: 1226088, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37492527

RESUMEN

Malaria remains one of the most significant health issues worldwide, accounting for 2.6% of the total global disease burden, and efforts to eliminate this threat continue. The key focus is to develop an efficient and long-term immunity to this disease via vaccination or therapeutic approach, and innovative strategies would enable us to achieve this target. Previously, using a mouse co-infection disease model, cross-protection was illustrated between Babesia microti and Plasmodium chabaudi. Hence, this study was planned to elucidate the impact of acute B. microti Peabody mjr and Plasmodium berghei ANKA co-infection on the consequence of complicated malaria in the C57BL/6J mouse model of malaria. Furthermore, immune response and pathological features were analyzed, and the course of the disease was compared among experimental groups. Our study established that acute B. microti infection activated immunity which was otherwise suppressed by P. berghei. The immunosuppressive tissue microenvironment was counteracted as evidenced by the enhanced immune cell population in co-infected mice, in contrast to P. berghei-infected control mice. Parasite sequestration in the brain, liver, lung, and spleen of co-infected mice was significantly decreased and tissue injury was ameliorated. Meanwhile, the serum levels of IFN-γ, TNF-α, and IL-12p70 were reduced while the secretion of IL-10 was promoted in co-infected mice. Eventually, co-infected mice showed an extended rate of survival. Hereby, the principal cytokines associated with the severity of malaria by P. berghei infection were TNF-α, IFN-γ, and IL-12p70. Moreover, it was evident from our flow cytometry results that innate immunity is crucial and macrophages are at the frontline of immunity against P. berghei infection. Our study recommended further investigations to shed light on the effects of babesiosis in suppressing malaria with the goal of developing Babesia-based therapy against malaria.


Asunto(s)
Babesia microti , Coinfección , Malaria , Animales , Ratones , Plasmodium berghei , Factor de Necrosis Tumoral alfa , Ratones Endogámicos C57BL , Malaria/complicaciones , Malaria/tratamiento farmacológico
13.
Bull Math Biol ; 85(9): 81, 2023 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-37507538

RESUMEN

A high mutation rate of the RNA virus results in the emergence of novel mutants that may escape the immunity activated by the original (wild-type) strain. However, many of them go extinct because of the stochasticity due to the small initial number of infected cells. In a previous paper, we studied the probability of escaping stochastic extinction when the novel mutant has a faster rate of infection and when it is resistant to a drug that suppresses the wild-type virus. In this study, we examine the effect of escaping the immune reaction of the host. Based on a continuous-time branching process with time-dependent rates, we conclude the chance for a mutant strain to be established [Formula: see text] decreases with time [Formula: see text] since the wild-type infection when the mutant is produced. The number of novel mutants that can escape extinction risk has a peak soon after the wild-type infection. The number of novel escape mutations produced per patient in the early phase of host infection is small both for very strong and very weak immune responses, and it attains its maximum value when immune activity is of an intermediate strength.


Asunto(s)
Modelos Biológicos , Virus , Humanos , Conceptos Matemáticos , Virus/genética , Probabilidad , Tasa de Mutación , Mutación
14.
Virol J ; 20(1): 167, 2023 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-37507719

RESUMEN

Since SARS-CoV-2 was first reported in late 2019, multiple variations of the original virus have emerged. Each variant harbors accumulations of mutations, particularly within the spike glycoprotein, that are associated with increased viral transmissibility and escape immunity. The different mutations in the spike protein of different variants shape the subsequent antibody and T cell responses, such that exposure to different spike proteins can result in reduced or enhanced responses to heterologous variants further down the line. Globally, people have been exposed and re-exposed to multiple variations of the Ancestral strain, including the five variants of concerns. Studies have shown that the protective immune response of an individual is influenced by which strain or combination of strains they are exposed to. The initial exposure to a specific strain may also shape their subsequent immune patterns and response to later infections with a heterologous virus. Most immunological observations were carried out early during the pandemic when the Ancestral strain was circulating. However, SARS-CoV-2 variants exhibit varying patterns of disease severity, waning immunity, immune evasion and sensitivity to therapeutics. Here we investigated the cross-protection in hamsters previously infected with a variant of concern (VOC) and subsequently re-infected with a heterologous variant. We also determined if cross-protection and immunity were dependent on the specific virus to which the hamster was first exposed. We further profiled the host cytokine response induced by each SARS-CoV-2 variants as well as subsequent to re-infection. A comparative analysis of the three VOCs revealed that Alpha variant was the most pathogenic VOC to emerge. We showed that naturally acquired immunity protected hamsters from subsequent re-infection with heterologous SARS-CoV-2 variant, regardless which variant the animal was first exposed to. Our study supports observations that heterologous infection of different SARS-CoV-2 variants do not exacerbate disease in subsequent re-infections. The continual emergence of new SARS-CoV-2 variants mandates a better understanding of cross-protection and immune imprinting in infected individuals. Such information is essential to guide vaccine strategy and public policy to emerging SARS-CoV-2 VOCs and future novel pandemic coronaviruses.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Cricetinae , Humanos , SARS-CoV-2/genética , Protección Cruzada , Reinfección , Inmunidad Adaptativa , Glicoproteína de la Espiga del Coronavirus/genética
15.
Vopr Virusol ; 68(3): 252-264, 2023 07 06.
Artículo en Ruso | MEDLINE | ID: mdl-37436416

RESUMEN

INTRODUCTION: The WHO regularly updates influenza vaccine recommendations to maximize their match with circulating strains. Nevertheless, the effectiveness of the influenza A vaccine, specifically its H3N2 component, has been low for several seasons. The aim of the study is to develop a mathematical model of cross-immunity based on the array of published WHO hemagglutination inhibition assay (HAI) data. MATERIALS AND METHODS: In this study, a mathematical model was proposed, based on finding, using regression analysis, the dependence of HAI titers on substitutions in antigenic sites of sequences. The computer program we developed can process data (GISAID, NCBI, etc.) and create real-time databases according to the set tasks. RESULTS: Based on our research, an additional antigenic site F was identified. The difference in 1.6 times the adjusted R2, on subsets of viruses grown in cell culture and grown in chicken embryos, demonstrates the validity of our decision to divide the original data array by passage histories. We have introduced the concept of a degree of homology between two arbitrary strains, which takes the value of a function depending on the Hamming distance, and it has been shown that the regression results significantly depend on the choice of function. The provided analysis showed that the most significant antigenic sites are A, B, and E. The obtained results on predicted HAI titers showed a good enough result, comparable to similar work by our colleagues. CONCLUSION: The proposed method could serve as a useful tool for future forecasts, with further study to confirm its sustainability.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Embrión de Pollo , Animales , Humanos , Subtipo H3N2 del Virus de la Influenza A/genética , Vacunas contra la Influenza/genética , Antígenos Virales/genética , Epítopos , Modelos Teóricos , Gripe Humana/epidemiología , Gripe Humana/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza , Estaciones del Año
16.
Vet Microbiol ; 283: 109796, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37285792

RESUMEN

There are three main genotypes of porcine circovirus type 2 (PCV2), namely PCV2a, PCV2b and PCV2d, of which PCV2b and PCV2d are currently the most common. There are antigenic differences between these different genotypes. To explore the effect of PCV2 antigen differences on the immune protection provided by vaccines, a cross-immune protection test was carried out in pigs. Three genotype strains, PCV2a-CL, PCV2b-MDJ and PCV2d-LNHC, were inactivated and emulsified to prepare inactivated vaccines to immunize pigs, who were then challenged with the circulating strains PCV2b-BY and PCV2d-LNHC. Immunoperoxidase monolayer assays (IPMAs) and micro-neutralization assays were used to detect antibodies against the three different genotypes of PCV2. The results showed that the three genotype vaccines induced pigs to produce antibodies against the same and different genotypes of PCV2, but the levels of IPMA and neutralizing antibodies against the same genotype were higher than those against different genotypes. Quantitative Polymerase Chain Reaction (qPCR), virus titration and immunohistochemistry were used to detect PCV2 genomic DNA, live virus and antigen, respectively, in inguinal lymph nodes of experimental pigs. Following challenge with the PCV2b-BY strain, the viral DNA load in the inguinal lymph nodes of pigs immunized with the three genotype vaccines was reduced by more than 99 % compared to the unimmunized group. Following challenge with the PCV2d-LNHC strain, the viral DNA loads in the inguinal lymph nodes of pigs immunized with PCV2a, PCV2b and PCV2d genotype vaccines were reduced by 93.8 %, 99.8 % and 98.3 %, respectively, compared to unimmunized controls. In addition, neither live PCV2 virus nor antigen were detected in the inguinal lymph nodes of pigs immunized with any of the genotype vaccines (0/18), but both were detected in the lymph nodes of experimental pigs in the unimmunized control group (6/6). These findings suggest that, although the antigenic differences of the three genotype strains induce significant differences in antibody levels, they seem to have little effect on cross-protection between different genotypes.


Asunto(s)
Infecciones por Circoviridae , Circovirus , Enfermedades de los Porcinos , Vacunas Virales , Animales , Porcinos , Anticuerpos Antivirales , Circovirus/genética , ADN Viral/genética , Genotipo , Infecciones por Circoviridae/prevención & control , Infecciones por Circoviridae/veterinaria
17.
Qual Theory Dyn Syst ; 22(3): 87, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37124841

RESUMEN

In this paper, we analyze a stochastic SIRC model with Ornstein-Uhlenbeck process. Firstly, we give the existence and uniqueness of global solution of stochastic SIRC model and prove it. In addition, the existence of ergodic stationary distributions for stochastic SIRC system is proved by constructing a suitable series of Lyapunov functions. A quasi-endemic equilibrium related to endemic equilibrium of deterministic systems is defined by considering randomness. And we obtain the probability density function of the linearized system near the equilibrium point. After the proof of probability density function, the sufficient condition of disease extinction is given and proved. We prove the theoretical results in the paper by numerical simulation at the end of the paper.

18.
Microorganisms ; 11(4)2023 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-37110282

RESUMEN

We studied the effect of transmissibility and vaccination on the time required for an emerging strain of an existing virus to dominate in the infected population using a simulation-based experiment. The emergent strain is assumed to be completely resistant to the available vaccine. A stochastic version of a modified SIR model for emerging viral strains was developed to simulate surveillance data for infections. The proportion of emergent viral strain infections among the infected was modeled using a logistic curve and the time to dominance (TTD) was recorded for each simulation. A factorial experiment was implemented to compare the TTD values for different transmissibility coefficients, vaccination rates, and initial vaccination coverage. We discovered a non-linear relationship between TTD and the relative transmissibility of the emergent strain for populations with low vaccination coverage. Furthermore, higher vaccination coverage and high vaccination rates in the population yielded significantly lower TTD values. Vaccinating susceptible individuals against the current strain increases the susceptible pool of the emergent virus, which leads to the emergent strain spreading faster and requiring less time to dominate the infected population.

19.
Math Biosci ; 360: 109010, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37088125

RESUMEN

Within-host models of infection can provide important insights into the processes that affect parasite spread and persistence in host populations. However, modeling can be limited by the availability of empirical data, a problem commonly encountered in natural systems. Here, we used six years of immune-infection observations of two gastrointestinal helminths (Trichostrongylus retortaeformis and Graphidium strigosum) from a population of European rabbits (Oryctolagus cuniculus) to develop an age-dependent, mathematical model that explicitly included species-specific and cross-reacting antibody (IgA and IgG) responses to each helminth in hosts with single or dual infections. Different models of single infection were formally compared to test alternative mechanisms of parasite regulation. The two models that best described single infections of each helminth species were then coupled through antibody cross-immunity to examine how the presence of one species could alter the host immune response to, and the within-host dynamics of, the other species. For both single infections, model selection suggested that either IgA or IgG responses could equally explain the observed parasite intensities by host age. However, the antibody attack rate and affinity level changed between the two helminths, it was stronger against T. retortaeformis than against G. strigosum and caused contrasting age-intensity profiles. When the two helminths coinfect the same host, we found variation of the species-specific antibody response to both species together with an asymmetric cross-immune response driven by IgG. Lower attack rate and affinity of antibodies in dual than single infections contributed to the significant increase of both helminth intensities. By combining mathematical modeling with immuno-infection data, our work provides a tractable model framework for disentangling some of the complexities generated by host-parasite and parasite-parasite interactions in natural systems.


Asunto(s)
Helmintos , Animales , Conejos , Incidencia , Helmintos/fisiología , Inmunoglobulina G , Inmunoglobulina A , Interacciones Huésped-Parásitos
20.
Evol Med Public Health ; 11(1): 80-89, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37007165

RESUMEN

Non-pharmaceutical interventions (NPIs), such as social distancing and contact tracing, are important public health measures that can reduce pathogen transmission. In addition to playing a crucial role in suppressing transmission, NPIs influence pathogen evolution by mediating mutation supply, restricting the availability of susceptible hosts, and altering the strength of selection for novel variants. Yet it is unclear how NPIs might affect the emergence of novel variants that are able to escape pre-existing immunity (partially or fully), are more transmissible or cause greater mortality. We analyse a stochastic two-strain epidemiological model to determine how the strength and timing of NPIs affect the emergence of variants with similar or contrasting life-history characteristics to the wild type. We show that, while stronger and timelier NPIs generally reduce the likelihood of variant emergence, it is possible for more transmissible variants with high cross-immunity to have a greater probability of emerging at intermediate levels of NPIs. This is because intermediate levels of NPIs allow an epidemic of the wild type that is neither too small (facilitating high mutation supply), nor too large (leaving a large pool of susceptible hosts), to prevent a novel variant from becoming established in the host population. However, since one cannot predict the characteristics of a variant, the best strategy to prevent emergence is likely to be an implementation of strong, timely NPIs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA