Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Front Public Health ; 10: 904550, 2022.
Article in English | MEDLINE | ID: covidwho-2154831

ABSTRACT

Objective: After the unprecedented coronavirus disease 2019 (COVID-19) outbreak, the health status of the general population has suffered a huge threat, and the mental health of front-line healthcare providers has also encountered great challenges. Therefore, this study aims to: (1) investigate the prevalence and influencing factors of post-traumatic stress disorder (PTSD) among healthcare providers, and (2) verify the moderating role of self-efficacy in the influence of PTSD on mental health. Methods: A cross-sectional study was conducted using an online survey of 1993 participants. The presence of depression, anxiety, self-efficacy, and PTSD was evaluated using screening tests from March 1. Sociodemographic and COVID-19-related data were also collected. A data analysis was performed using descriptive statistics, Pearson's correlation coefficient, and multiple linear regression. Results: The prevalence of PTSD among healthcare providers was 9.3%. PTSD was negatively correlated with self-efficacy (r = -0.265, P < 0.01), anxiety (r = -0.453, P < 0.01), and depression (r = 0.708, P < 0.01). Profession, daily working hours, maximum continuous working days, and daily sleep time were influencing factors of PTSD. A binary logistic regression analysis showed that physicians (OR = 2.254, 95% CI = 1.298, 3.914) and nurses (OR = 2.176, 95% CI = 1.337, 3.541) were more likely to experience PTSD than other healthcare providers. Conclusion: Self-efficacy has a moderating effect on the influence of PTSD on anxiety and depression. This suggests that health managers need to respond to the current psychological crisis of healthcare providers, implement appropriate psychological interventions, and minimize the psychological harm caused by COVID-19.


Subject(s)
COVID-19 , Stress Disorders, Post-Traumatic , COVID-19/epidemiology , Cross-Sectional Studies , Depression/epidemiology , Health Personnel/psychology , Humans , Mental Health , Prevalence , Stress Disorders, Post-Traumatic/epidemiology
2.
Frontiers in public health ; 10, 2022.
Article in English | EuropePMC | ID: covidwho-1971023

ABSTRACT

Objective After the unprecedented coronavirus disease 2019 (COVID-19) outbreak, the health status of the general population has suffered a huge threat, and the mental health of front-line healthcare providers has also encountered great challenges. Therefore, this study aims to: (1) investigate the prevalence and influencing factors of post-traumatic stress disorder (PTSD) among healthcare providers, and (2) verify the moderating role of self-efficacy in the influence of PTSD on mental health. Methods A cross-sectional study was conducted using an online survey of 1993 participants. The presence of depression, anxiety, self-efficacy, and PTSD was evaluated using screening tests from March 1. Sociodemographic and COVID-19-related data were also collected. A data analysis was performed using descriptive statistics, Pearson's correlation coefficient, and multiple linear regression. Results The prevalence of PTSD among healthcare providers was 9.3%. PTSD was negatively correlated with self-efficacy (r = −0.265, P < 0.01), anxiety (r = −0.453, P < 0.01), and depression (r = 0.708, P < 0.01). Profession, daily working hours, maximum continuous working days, and daily sleep time were influencing factors of PTSD. A binary logistic regression analysis showed that physicians (OR = 2.254, 95% CI = 1.298, 3.914) and nurses (OR = 2.176, 95% CI = 1.337, 3.541) were more likely to experience PTSD than other healthcare providers. Conclusion Self-efficacy has a moderating effect on the influence of PTSD on anxiety and depression. This suggests that health managers need to respond to the current psychological crisis of healthcare providers, implement appropriate psychological interventions, and minimize the psychological harm caused by COVID-19.

3.
Front Immunol ; 13: 848961, 2022.
Article in English | MEDLINE | ID: covidwho-1963440

ABSTRACT

CoronaVac (Sinovac), an inactivated vaccine for SARS-CoV-2, has been widely used for immunization. However, analysis of the underlying molecular mechanisms driving CoronaVac-induced immunity is still limited. Here, we applied a systems biology approach to understand the mechanisms behind the adaptive immune response to CoronaVac in a cohort of 50 volunteers immunized with 2 doses of CoronaVac. Vaccination with CoronaVac led to an integrated immune response that included several effector arms of the adaptive immune system including specific IgM/IgG, humoral response and other immune response, as well as the innate immune system as shown by complement activation. Metabolites associated with immunity were also identified implicating the role of metabolites in the humoral response, complement activation and other immune response. Networks associated with the TCA cycle and amino acids metabolic pathways, such as phenylalanine metabolism, phenylalanine, tyrosine and tryptophan biosynthesis, and glycine, serine and threonine metabolism were tightly coupled with immunity. Critically, we constructed a multifactorial response network (MRN) to analyze the underlying interactions and compared the signatures affected by CoronaVac immunization and SARS-CoV-2 infection to further identify immune signatures and related metabolic pathways altered by CoronaVac immunization. These results help us to understand the host response to vaccination of CoronaVac and highlight the utility of a systems biology approach in defining molecular correlates of protection to vaccination.


Subject(s)
COVID-19 , Viral Vaccines , Adaptive Immunity , COVID-19/prevention & control , COVID-19 Vaccines , Humans , Phenylalanine , Proteomics , SARS-CoV-2 , Vaccines, Inactivated
4.
J Med Virol ; 94(11): 5304-5324, 2022 Nov.
Article in English | MEDLINE | ID: covidwho-1935705

ABSTRACT

To control the ongoing coronavirus disease-2019 (COVID-19) pandemic, CoronaVac (Sinovac), an inactivated vaccine, has been granted emergency use authorization by many countries. However, the underlying mechanisms of the inactivated COVID-19 vaccine-induced immune response remain unclear, and little is known about its features compared to (Severe acute respiratory syndrome coronavirus 2) SARS-CoV-2 infection. Here, we implemented single-cell RNA sequencing (scRNA-seq) to profile longitudinally collected PBMCs (peripheral blood mononuclear cells) in six individuals immunized with CoronaVac and compared these to the profiles of COVID-19 infected patients from a Single Cell Consortium. Both inactivated vaccines and SARS-CoV-2 infection altered the proportion of different immune cell types, caused B cell activation and differentiation, and induced the expression of genes associated with antibody production in the plasma. The inactivated vaccine and SARS-COV-2 infection also caused alterations in peripheral immune activity such as interferon response, inflammatory cytokine expression, innate immune cell apoptosis and migration, effector T cell exhaustion and cytotoxicity, however, the magnitude of change was greater in COVID-19 patients, especially those with severe disease, than in immunized individuals. Further analyses revealed a distinct peripheral immune cell phenotype associated with CoronaVac immunization (HLA class II upregulation and IL21R upregulation in naïve B cells) versus SARS-CoV-2 infection (HLA class II downregulation and IL21R downregulation in naïve B cells from severe disease individuals). There were also differences in the expression of important genes associated with proinflammatory cytokines and thrombosis. In conclusion, this study provides a single-cell atlas of the systemic immune response to CoronaVac immunization and revealed distinct immune responses between inactivated vaccines and SARS-CoV-2 infection.


Subject(s)
COVID-19 , Viral Vaccines , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines , Cytokines , Humans , Leukocytes, Mononuclear , Receptors, Interleukin-21 , SARS-CoV-2 , Transcriptome , Vaccines, Inactivated
5.
Frontiers in immunology ; 13, 2022.
Article in English | EuropePMC | ID: covidwho-1888296

ABSTRACT

CoronaVac (Sinovac), an inactivated vaccine for SARS-CoV-2, has been widely used for immunization. However, analysis of the underlying molecular mechanisms driving CoronaVac-induced immunity is still limited. Here, we applied a systems biology approach to understand the mechanisms behind the adaptive immune response to CoronaVac in a cohort of 50 volunteers immunized with 2 doses of CoronaVac. Vaccination with CoronaVac led to an integrated immune response that included several effector arms of the adaptive immune system including specific IgM/IgG, humoral response and other immune response, as well as the innate immune system as shown by complement activation. Metabolites associated with immunity were also identified implicating the role of metabolites in the humoral response, complement activation and other immune response. Networks associated with the TCA cycle and amino acids metabolic pathways, such as phenylalanine metabolism, phenylalanine, tyrosine and tryptophan biosynthesis, and glycine, serine and threonine metabolism were tightly coupled with immunity. Critically, we constructed a multifactorial response network (MRN) to analyze the underlying interactions and compared the signatures affected by CoronaVac immunization and SARS-CoV-2 infection to further identify immune signatures and related metabolic pathways altered by CoronaVac immunization. These results help us to understand the host response to vaccination of CoronaVac and highlight the utility of a systems biology approach in defining molecular correlates of protection to vaccination.

6.
Front Bioeng Biotechnol ; 9: 748746, 2021.
Article in English | MEDLINE | ID: covidwho-1507012

ABSTRACT

The ongoing Corona virus disease (COVID-19) outbreak has become a huge global health concern. Here, we reported a novel detection platform based on the loop-mediated isothermal amplification (LAMP), termed real-time reverse transcription LAMP (rRT-LAMP) and applied it for the diagnosis of COVID-19 (COVID-19 rRT-LAMP). rRT-LAMP integrates reverse transcription, LAMP amplification, restriction endonuclease cleavage and real-time fluorescence detection into one-pot reaction, and facilitates the diagnosis of COVID-19 at 64°C for only 35 min. The ORF1ab (opening reading frame 1a/b) and NP (nucleoprotein) genes of SARS-CoV-2 were detected for diagnosing COVID-19. The limit of detection (LoD) of COVID-19 rRT-LAMP assay was 14 copies (for each marker) per vessel, and no positive results were obtained from non-SARS-CoV-2 templates. To demonstrate its feasibility, a total of 33 oropharynx swab samples collected from COVID-19 patients also were diagnosed as SARS-CoV-2 infection using COVID-19 rRT-LAMP protocol. No cross-reactivity was yielded from 41 oropharynx swab samples collected from non-COVID-19 patients. These data suggesting that the COVID-19 rRT-LAMP assay is a potential detection tool for the diagnosis of SARS-CoV-2 infection in clinical, field and disease control laboratories, and will be valuable for controlling the COVID-19 epidemic.

7.
Biosens Bioelectron ; 166: 112437, 2020 Oct 15.
Article in English | MEDLINE | ID: covidwho-645435

ABSTRACT

The ongoing global pandemic (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a huge public health issue. Hence, we devised a multiplex reverse transcription loop-mediated isothermal amplification (mRT-LAMP) coupled with a nanoparticle-based lateral flow biosensor (LFB) assay (mRT-LAMP-LFB) for diagnosing COVID-19. Using two LAMP primer sets, the ORF1ab (opening reading frame 1a/b) and N (nucleoprotein) genes of SARS-CoV-2 were simultaneously amplified in a single-tube reaction, and detected with the diagnosis results easily interpreted by LFB. In presence of FITC (fluorescein)-/digoxin- and biotin-labeled primers, mRT-LAMP produced numerous FITC-/digoxin- and biotin-attached duplex amplicons, which were determined by LFB through immunoreactions (FITC/digoxin on the duplex and anti-FITC/digoxin on the test line of LFB) and biotin/treptavidin interaction (biotin on the duplex and strptavidin on the polymerase nanoparticle). The accumulation of nanoparticles leaded a characteristic crimson band, enabling multiplex analysis of ORF1ab and N gene without instrumentation. The limit of detection (LoD) of COVID-19 mRT-LAMP-LFB was 12 copies (for each detection target) per reaction, and no cross-reactivity was generated from non-SARS-CoV-2 templates. The analytical sensitivity of SARS-CoV-2 was 100% (33/33 oropharynx swab samples collected from COVID-19 patients), and the assay's specificity was also 100% (96/96 oropharynx swab samples collected from non-COVID-19 patients). The total diagnostic test can be completed within 1 h from sample collection to result interpretation. In sum, the COVID-19 mRT-LAMP-LFB assay is a promising tool for diagnosing SARS-CoV-2 infections in frontline public health field and clinical laboratories, especially from resource-poor regions.


Subject(s)
Betacoronavirus/genetics , Betacoronavirus/isolation & purification , Biosensing Techniques , Clinical Laboratory Techniques , Coronavirus Infections/diagnosis , Coronavirus Infections/virology , Pandemics , Pneumonia, Viral/diagnosis , Pneumonia, Viral/virology , Biosensing Techniques/instrumentation , Biosensing Techniques/methods , Biosensing Techniques/statistics & numerical data , COVID-19 , COVID-19 Testing , China/epidemiology , Clinical Laboratory Techniques/instrumentation , Clinical Laboratory Techniques/methods , Clinical Laboratory Techniques/statistics & numerical data , Coronavirus Infections/epidemiology , Equipment Design , Feasibility Studies , Humans , Limit of Detection , Molecular Diagnostic Techniques , Multiplex Polymerase Chain Reaction/methods , Multiplex Polymerase Chain Reaction/statistics & numerical data , Nanoparticles , Nanotechnology , Nucleic Acid Amplification Techniques , Pneumonia, Viral/epidemiology , RNA, Viral/analysis , RNA, Viral/genetics , SARS-CoV-2 , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL