Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Poult Sci ; 103(2): 103332, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38128459

ABSTRACT

In late 2020, an outbreak of Tembusu virus (TMUV)-associated disease occurred in a 45-day-old white Roman geese flock in Taiwan. Here, we present the identification and isolation of a novel goose-origin TMUV strain designated as NTU/C225/2020. The virus was successfully isolated using minimal-pathogen-free duck embryos. Phylogenetic analysis of the polyprotein gene showed that NTU/C225/2020 clustered together with the earliest isolates from Malaysia and was most closely related to the first Taiwanese TMUV strain, TP1906. Genomic analysis revealed significant amino acid variations among TMUV isolates in NS1 and NS2A protein regions. In the present study, we characterized the NTU/C225/2020 culture in duck embryos, chicken embryos, primary duck embryonated fibroblasts, and DF-1 cells. All host systems were susceptible to NTU/C225/2020 infection, with observable lesions. In addition, animal experiments showed that the intramuscular inoculation of NTU/C225/2020 resulted in growth retardation and hyperthermia in day-old chicks. Gross lesions in the infected chicks included hepatomegaly, hyperemic thymus, and splenomegaly. Viral loads and histopathological damage were displayed in various tissues of both inoculated and naïve co-housed chicks, confirming the direct chick-to-chick contact transmission of TMUV. This is the first in vivo study of a local TMUV strain in Taiwan. Our findings provide essential information for TMUV propagation and suggest a potential risk of disease outbreak in chicken populations.


Subject(s)
Flavivirus Infections , Flavivirus , Poultry Diseases , Chick Embryo , Animals , Flavivirus Infections/veterinary , Geese , Chickens , Phylogeny , Virulence , Cetuximab , Poultry Diseases/pathology , Ducks
2.
Emerg Microbes Infect ; 12(2): 2220582, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37254830

ABSTRACT

Since the onset of the coronavirus disease 2019 (COVID-19), numerous neutralizing antibodies (NAbs) against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been developed and authorized for emergency use to control the pandemic. Most COVID-19 therapeutic NAbs prevent the S1 subunit of the SARS-CoV-2 spike (S) protein from binding to the human host receptor. However, the emergence of SARS-CoV-2 immune escape variants, which possess frequent mutations on the S1 subunit, may render current NAbs ineffective. In contrast, the relatively conserved S2 subunit of the S protein can elicit NAbs with broader neutralizing potency against various SARS-CoV-2 variants. In this review, the binding specificity and functional features of SARS-CoV-2 NAbs targeting different domains of the S2 subunit are collectively discussed. The knowledge learned from the investigation of the S2-specific NAbs provides insights and potential strategies for developing antibody cocktail therapy and next-generation coronavirus vaccine.


Subject(s)
COVID-19 , Humans , SARS-CoV-2 , COVID-19 Vaccines , Antibodies, Viral , Antibodies, Neutralizing , Spike Glycoprotein, Coronavirus
3.
Appl Microbiol Biotechnol ; 107(9): 2983-2995, 2023 May.
Article in English | MEDLINE | ID: mdl-36988669

ABSTRACT

Vaccination is considered to be the most effective countermeasure to prevent and combat the global health threats of COVID-19. People with obesity are at a greater risk of hospitalization, life-threatening illness, and adverse outcomes after having COVID-19. Therefore, a safe and effective COVID-19 vaccine for obese individuals is urgently needed. In the study, the vaccine composed of the ISA 51 adjuvant and the SARS-CoV-2 spike (S) receptor-binding domain (RBD) in conjugation with the human IgG1 Fc fragment (named as ISA 51-adjuvanted RBD-Fc vaccine) was developed and inoculated in the regular chow diet (RCD) lean mice and the high-fat diet (HFD)-induced obese mice. The S protein-specific IgG titers were largely induced in an increasing manner along with three doses of ISA 51-adjuvanted RBD-Fc vaccine without causing any harmful side effect. In the HFD mice, the S protein-specific IgG titers can be quickly observed 2 weeks post the first inoculation. The antisera elicited by the ISA 51-adjuvanted RBD-Fc vaccine in the RCD and HFD mice exhibited potent SARS-CoV-2 neutralizing activities in the plaque reduction neutralization test (PRNT) assays and showed similar specificity for recognizing the key residues in the RBD which were involved in interacting with angiotensin-converting enzyme 2 (ACE2) receptor. The immune efficacy of the ISA 51-adjuvanted RBD-Fc vaccine in the HFD mice can be sustainably maintained with the PRNT50 values of 1.80-1.91×10-3 for at least 8 weeks post the third inoculation. Collectively, the RBD-Fc-based immunogen and the ISA 51-adjuvanted formulation can be developed as an effective COVID-19 vaccine for obese individuals. KEY POINTS: • The ISA 51-adjuvanted RBD-Fc vaccine can induce potent SARS-CoV-2 neutralizing antibodies in the obese mouse • The antibodies elicited by the ISA 51-adjuvanted RBD-Fc vaccine can bind to the key RBD residues involved in interacting with ACE2 • The immune efficacy of the ISA 51-adjuvanted RBD-Fc vaccine can be sustainably maintained for at least 8 weeks post the third inoculation.


Subject(s)
COVID-19 , Vaccines , Humans , Animals , Mice , Antibodies, Neutralizing , COVID-19 Vaccines , SARS-CoV-2 , Mice, Obese , Angiotensin-Converting Enzyme 2 , COVID-19/prevention & control , Antibodies, Viral , Immunoglobulin G , Spike Glycoprotein, Coronavirus
4.
Appl Microbiol Biotechnol ; 106(24): 8183-8194, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36404356

ABSTRACT

The nucleic acid test is still the standard assessment for the diagnosis of coronavirus disease 2019 (COVID-19), which is caused by human infection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In addition to supporting the confirmation of disease cases, serological assays are used for the analysis of antibody status and epidemiological surveys. In this study, a single Western blot strip (WBS) coated with multiple Escherichia coli (E. coli)-expressed SARS-CoV-2 antigens was developed for comprehensive studies of antibody profiles in COVID-19 patient sera. The levels of specific antibodies directed to SARS-CoV-2 spike (S), S2, and nucleocapsid (N) proteins were gradually increased with the same tendency as the disease progressed after hospitalization. The signal readouts of S, S2, and N revealed by the multi-antigen-coated WBS (mWBS)-based serological assay (mWBS assay) also demonstrated a positive correlation with the SARS-CoV-2 neutralizing potency of the sera measured by the plaque reduction neutralization test (PRNT) assays. Surprisingly, the detection signals against the unstructured receptor-binding domain (RBD) purified from E. coli inclusion bodies were not observed, although the COVID-19 patient sera exhibited strong neutralizing potency in the PRNT assays, suggesting that the RBD-specific antibodies in patient sera mostly recognize the conformational epitopes. Furthermore, the mWBS assay identified a unique and major antigenic epitope at the residues 1148, 1149, 1152, 1155, and 1156 located within the 1127-1167 fragment of the S2 subunit, which was specifically recognized by the COVID-19 patient serum. The mWBS assay can be finished within 14-16 min by using the automatic platform of Western blotting by thin-film direct coating with suction (TDCS WB). Collectively, the mWBS assay can be applied for the analysis of antibody responses, prediction of the protective antibody status, and identification of the specific epitope. KEY POINTS: • A Western blot strip (WBS) coated with multiple SARS-CoV-2 antigens was developed for the serological assay. • The multi-antigen-coated WBS (mWBS) can be utilized for the simultaneous detection of antibody responses to multiple SARS-CoV-2 antigens. • The mWBS-based serological assay (mWBS assay) identified a unique epitope recognized by the COVID-19 patient serum.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Antibody Formation , COVID-19/diagnosis , Escherichia coli/genetics , Blotting, Western
5.
Vaccines (Basel) ; 10(11)2022 Oct 30.
Article in English | MEDLINE | ID: mdl-36366348

ABSTRACT

Recombinant proteins are essential in the development of subunit vaccines. In the design of many recombinant proteins, polyhistidine residues are added to the N- or C-termini of target sequences to facilitate purification. However, whether the addition of tag residues influences the immunogenicity of proteins remains unknown. In this study, the tag-free SARS-CoV-2 RBD and His-tag SARS-CoV-2 RBD proteins were investigated to determine whether there were any differences in their receptor binding affinity and immunogenicity. The results showed that the tag-free RBD protein had a higher affinity for binding with hACE2 receptors than His-tag RBD proteins (EC50: 1.78 µM vs. 7.51 µM). On day 21 after primary immunization with the proteins, the serum ELISA titers of immunized mice were measured and found to be 1:1418 for those immunized with tag-free RBD and only 1:2.4 for His-tag RBD. Two weeks after the booster dose, tag-free-RBD-immunized mice demonstrated a significantly higher neutralizing titer of 1:369 compared with 1:7.9 for His-tag-RBD-immunized mice. Furthermore, neutralizing antibodies induced by tag-free RBD persisted for up to 5 months and demonstrated greater cross-neutralization of the SARS-CoV-2 Delta variant. Evidence from Western blotting showed that the serum of His-tag-RBD-immunized mice recognized irrelevant His-tag proteins. Collectively, we conclude that the addition of a polyhistidine tag on a recombinant protein, when used as a COVID-19 vaccine antigen, may significantly impair protein immunogenicity against SARS-CoV-2. Antibody responses induced were clearly more rapid and robust for the tag-free SARS-CoV-2 RBD than the His-tag SARS-CoV-2 RBD. These findings provide important information for the design of antigens used in the development of COVID-19 subunit vaccines.

6.
Antiviral Res ; 200: 105290, 2022 04.
Article in English | MEDLINE | ID: mdl-35296418

ABSTRACT

Neutralizing antibodies (NAbs) are believed to be promising prophylactic and therapeutic treatment against the coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here, we reported two mouse monoclonal antibodies 7 Eb-4G and 1Ba-3H that specifically recognized the receptor-binding domain (RBD) of SARS-CoV-2 spike (S) protein without exhibiting cross-reactivity with the S proteins of SARS-CoV and MERS-CoV. The binding epitopes of 7 Eb-4G and 1Ba-3H were respectively located in the regions of residues 457-476 and 477-496 in the S protein. Only 1Ba-3H exhibited the neutralizing activity for preventing the pseudotyped lentivirus from binding to the angiotensin-converting enzyme 2 (ACE2)-transfected HEK293T cells. The competitive ELISA further showed that 1Ba-3H interfered with the binding between RBD and ACE2. Epitope mapping experiments demonstrated that a single alanine replacement at residues 480, 482, 484, 485, and 488-491 in the RBD abrogated 1Ba-3H binding. 1Ba-3H exhibited the neutralizing activity against the wild-type, Alpha, Delta, and Epsilon variants of SARS-CoV-2, but lost the neutralizing activity against Gamma variant in the plaque reduction assay. On the contrary, 1Ba-3H enhanced the cellular infection of Gamma variant in a dose-dependent manner. Our findings suggest that the antibody-dependent enhancement of infection mediated by the RBD-specific antibody for different SARS-CoV-2 variants must be considered while developing the NAb.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antibodies, Monoclonal , Epitopes , HEK293 Cells , Humans , Mice , Spike Glycoprotein, Coronavirus
7.
Microbiol Spectr ; 10(2): e0181421, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35293796

ABSTRACT

Most of SARS-CoV-2 neutralizing antibodies (nAbs) targeted the receptor binding domain (RBD) of the SARS-CoV-2 spike (S) protein. However, mutations at RBD sequences found in the emerging SARS-CoV-2 variants greatly reduced the effectiveness of nAbs. Here we showed that four nAbs, S2-4D, S2-5D, S2-8D, and S2-4A, which recognized a conserved epitope in the S2 subunit of the S protein, can inhibit SARS-CoV-2 infection through blocking the S protein-mediated membrane fusion. Notably, these four nAbs exhibited broadly neutralizing activity against SARS-CoV-2 Alpha, Gamma, Delta, and Epsilon variants. Antisera collected from mice immunized with the identified epitope peptides of these four nAbs also exhibited potent virus neutralizing activity. Discovery of the S2-specific nAbs and their unique antigenic epitopes paves a new path for development of COVID-19 therapeutics and vaccines. IMPORTANCE The spike (S) protein on the surface of SARS-CoV-2 mediates receptor binding and virus-host cell membrane fusion during virus entry. Many neutralizing antibodies (nAbs), which targeted the receptor binding domain (RBD) of S protein, lost the neutralizing activity against the newly emerging SARS-CoV-2 variants with sequence mutations at the RBD. In contrast, the nAb against the highly conserved S2 subunit, which plays the key role in virus-host cell membrane fusion, was poorly discovered. We showed that four S2-specific nAbs, S2-4D, S2-5D, S2-8D, and S2-4A, inhibited SARS-CoV-2 infection through blocking the S protein-mediated membrane fusion. These nAbs exhibited broadly neutralizing activity against Alpha, Gamma, Delta, and Epsilon variants. Antisera induced by the identified epitope peptides also possessed potent neutralizing activity. This work not only unveiled the S2-specific nAbs but also discovered an immunodominant epitope in the S2 subunit that can be rationally designed as the broad-spectrum vaccine against the SARS-like coronaviruses.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antibodies, Monoclonal , Antibodies, Neutralizing , Antibodies, Viral , Epitopes , Immune Sera , Membrane Fusion , Mice , Spike Glycoprotein, Coronavirus/genetics
8.
Cell Rep ; 36(9): 109621, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34469741

ABSTRACT

Radiotherapy (RT) resistance is a major cause of treatment failure in cancers that use definitive RT as their primary treatment modality. This study identifies the cancer/testis (CT) antigen G antigen (GAGE) as a mediator of radio resistance in cervical cancers. Elevated GAGE expression positively associates with de novo RT resistance in clinical samples. GAGE, specifically the GAGE12 protein variant, confers RT resistance through synemin-dependent chromatin localization, promoting the association of histone deacetylase 1/2 (HDAC1/2) to its inhibitor actin. This cumulates to elevated histone 3 lysine 56 acetylation (H3K56Ac) levels, increased chromatin accessibility, and improved DNA repair efficiency. Molecular or pharmacological disruption of the GAGE-associated complex restores radiosensitivity. Molecularly, this study demonstrates the role of GAGE in the regulation of chromatin dynamics. Clinically, this study puts forward the utility of GAGE as a pre-screening biomarker to identify poor responders at initial diagnosis and the therapeutic potential of agents that target GAGE and its associated complex in combination with radiotherapy to improve outcomes.


Subject(s)
Antigens, Neoplasm , Chromatin Assembly and Disassembly , Chromatin , Histones , Radiation Tolerance , Uterine Cervical Neoplasms , Animals , Female , Humans , Acetylation , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA Repair , Gene Expression Regulation, Neoplastic , HeLa Cells , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Histones/metabolism , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Lysine , Mice, Inbred BALB C , Mice, Nude , Protein Processing, Post-Translational , Radiation Tolerance/genetics , Signal Transduction , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/radiotherapy , Xenograft Model Antitumor Assays
9.
Appl Microbiol Biotechnol ; 105(11): 4663-4673, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34043078

ABSTRACT

The recent Zika virus (ZIKV) epidemic poses a serious threat to global health due to its association with microcephaly and congenital diseases in newborns and neurological complications and Guillain-Barré syndrome in adults. However, the majority of people infected with ZIKV do not develop symptoms. The platforms aimed to specifically diagnose ZIKV infection are needed for patient care and public health surveillance. In the study, four ZIKV envelope (E) protein-specific monoclonal antibodies (mAbs) (A1, B1, C1, and 9E-1) have been developed by using the conventional mAb technology. The binding epitopes of mAbs A1, B1, C1, and 9E-1 are located at E(238-257), E(410-431), E(258-277), and E(340-356), respectively. mAb 9E-1 performs 1.4- to 47-fold strong affinity to ZIKV E protein compared to another three mAbs. mAbs A1, C1, and 9E-1 do not have cross-reactivity against the recombinant E proteins of dengue virus serotypes 2, 3, and 4. Although these four mAbs do not have ZIKV neutralizing activity, mAbs B1 and 9E-1 have been developed as the lateral flow immunochromatographic assay for specific detection of ZIKV E protein and virions. KEY POINTS: • The mAbs targeting to the regions of E(238-257), E(410-431), E(258-277), and E(340-356) do not have ZIKV neutralizing activity. • The binding epitope of mAb 9E-1 is highly specific to ZIKV E protein. • mAbs B1 and 9E-1 can bind to ZIKV virions and have been developed as the lateral flow immunochromatographic assay.


Subject(s)
Zika Virus Infection , Zika Virus , Adult , Animals , Antibodies, Monoclonal , Antibodies, Neutralizing , Antibodies, Viral , Epitopes , Humans , Infant, Newborn , Mice , Viral Envelope , Viral Envelope Proteins , Zika Virus Infection/diagnosis
10.
Appl Microbiol Biotechnol ; 105(8): 3235-3248, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33770244

ABSTRACT

Many cases of avian influenza A(H7N9) virus infection in humans have been reported since its first emergence in 2013. The disease is of concern because most patients have become severely ill with roughly 30% mortality rate. Because the threat in public health caused by H7N9 virus remains high, advance preparedness is essentially needed. In this study, the recombinant H7N9 hemagglutinin (HA) was expressed in insect cells and purified for generation of two monoclonal antibodies, named F3-2 and 1C6B. F3-2 can only recognize the H7N9 HA without having cross-reactivity with HA proteins of H1N1, H3N2, H5N1, and H7N7. 1C6B has the similar specificity with F3-2, but 1C6B can also bind to H7N7 HA. The binding epitope of F3-2 is mainly located in the region of H7N9 HA(299-307). The binding epitope of 1C6B is located in the region of H7N9 HA(489-506). F3-2 and 1C6B could not effectively inhibit the hemagglutination activity of H7N9 HA. However, F3-2 can prevent H7N9 HA from trypsin cleavage and can bind to H7N9 HA which has undergone pH-induced conformational change. F3-2 also has the ability of binding to H7N9 viral particles and inhibiting H7N9 virus infection to MDCK cells with the IC50 value of 22.18 µg/mL. In addition, F3-2 and 1C6B were utilized for comprising a lateral flow immunochromatographic test strip for specific detection of H7N9 HA. KEY POINTS: • Two mouse monoclonal antibodies, F3-2 and 1C6B, were generated for recognizing the novel binding epitopes in H7N9 HA. • F3-2 can prevent H7N9 HA from trypsin cleavage and inhibit H7N9 virus infection to MDCK cells. • F3-2 and 1C6B were developed as a lateral flow immunochromatographic test for specific detection of H7N9 HA.


Subject(s)
Influenza A Virus, H7N9 Subtype , Influenza, Human , Animals , Antibodies, Monoclonal , Antibodies, Viral , Hemagglutinin Glycoproteins, Influenza Virus , Hemagglutinins , Humans , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Influenza A Virus, H5N1 Subtype , Influenza A Virus, H7N7 Subtype , Influenza, Human/prevention & control , Mice
11.
Appl Microbiol Biotechnol ; 105(1): 235-245, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33245391

ABSTRACT

The highly pathogenic avian influenza (HPAI) H5N8 virus has been detected in wild birds and poultry worldwide. The threat caused by HPAI H5N8 virus still exists with concerns for human infection. The preparedness for epidemic prevention and decreasing the agricultural and economic lost is extremely important. Hemagglutinin (HA), a surface glycoprotein of influenza viruses, is considered as the major target for detection of the influenza virus subtype in the infected samples. In this study, the recombinant H5N8 HA1 and HA2 proteins were expressed in Escherichia coli, and were utilized to generate two monoclonal antibodies, named 7H6C and YC8. 7H6C can bind the HA proteins of H5N1 and H5N8, but cannot bind the HA proteins of H1N1, H3N2, and H7N9, indicating that it has H5-subtype specificity. In contrast, YC8 can bind the HA proteins of H1N1, H5N1, and H5N8, but cannot bind the HA proteins of H3N2 and H7N9, indicating that it has H1-subtype and H5-subtype specificity. The epitope sequences recognized by 7H6C are located in the head domain of H5N8 HA, and are highly conserved in H5 subtypes. The epitope sequences recognized by YC8 are located in the stalk domain of H5N8 HA, and are highly conserved among the H1 and H5 subtypes. 7H6C and YC8 can be applied for specific detection of the HA proteins of H5N8 and H5Nx avian influenza viruses. KEY POINTS: • The mAb 7H6C or YC8 was generated by using the HA1 or HA2 of the HPAI H5N8 virus as the immunogen. • 7H6C recognized the head domain of H5N8 HA, and YC8 recognized the stalk domain of H5N8 HA. • 7H6C and YC8 can detect the HA proteins of H5Nx subtypes specifically.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A Virus, H5N1 Subtype , Influenza A Virus, H7N9 Subtype , Influenza in Birds , Animals , Antibodies, Monoclonal , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinins , Humans , Influenza A Virus, H3N2 Subtype , Influenza A Virus, H5N1 Subtype/genetics , Influenza in Birds/diagnosis
12.
Am J Phys Med Rehabil ; 99(11): 1056-1066, 2020 11.
Article in English | MEDLINE | ID: mdl-32149818

ABSTRACT

Neurogenic bladder disorders are common among patients with spinal cord lesions, which often result in upper and lower urinary tract complications. Urinary tract infection has remained the most frequent type of infection in this population. Our aim is to review systematically the literature on the outcome of different intervention methods to reduce urinary tract infection incidence. A literature search was conducted in the database of Medline, PubMed, Embase, and Scopus. After screening 1559 articles, 42 were included in this review. The intervention methods can be categorized into the four following groups: (1) indwelling catheterization and intermittent catheterization, (2) medications, (3) surgery, and (4) others. Intermittent catheterization is still the most recommended treatment for persons with spinal cord lesions. Hydrophilic catheters are more suitable for adults than children because of complex handling. Bladder management with spontaneous voiding is initially considered for infants and toddlers with spina bifida. Antibiotics treatment should be based on the results of urine cultures. Shortening the course of antibiotics treatment can reduce its adverse effects but may increase urinary tract infection recurrence. Because botulinum toxin injections and bladder surgery can improve urodynamic function, both are conducive toward lowering urinary tract infection incidence.


Subject(s)
Spinal Cord Diseases/complications , Urinary Bladder, Neurogenic/complications , Urinary Catheterization/methods , Urinary Tract Infections/prevention & control , Urologic Surgical Procedures/methods , Urological Agents/therapeutic use , Adolescent , Adult , Anti-Bacterial Agents/administration & dosage , Botulinum Toxins/administration & dosage , Child , Child, Preschool , Female , Humans , Incidence , Infant , Male , Middle Aged , Spinal Cord Injuries/complications , Treatment Outcome , Urinary Bladder, Neurogenic/etiology , Urinary Tract Infections/epidemiology , Urinary Tract Infections/etiology , Young Adult
13.
J Agric Food Chem ; 66(28): 7504-7513, 2018 Jul 18.
Article in English | MEDLINE | ID: mdl-29931974

ABSTRACT

Commercial dietary supplements of calcium pyruvate claim to be beneficial for losing weight, increasing muscle endurance, and regulating metabolism. Most industrial preparations have some impurities, including parapyruvate. Parapyruvate is an inhibitor of the α-ketoglutarate dehydrogenase complex (KGDHC). However, the effect and mechanism of parapyruvate on cell senescence and the content of parapyruvate in the dietary supplements of calcium pyruvate are unknown. In this study, we prepared pure parapyruvate with a purity of 99.8 ± 0.1% and investigated its ability to inhibit KGDHC activity and affect fibroblast senescence. Parapyruvate dose-dependently decreased KGDHC activity, with an IC50 of 4.13 mM and induced Hs68 cell senescence. Calcium ions, a KGDHC activator, antagonized the senescent effects of parapyruvate. The parapyruvate content was 1.4 ± 0.1% to 10.6 ± 0.2% in five brands of calcium pyruvate supplements. In this study, we showed that parapyruvate strongly induces Hs68 cell senescence by inhibiting KGDHC activity. Because of its KGDHC inhibition activity, the parapyruvate content should be an important issue for the food safety of calcium pyruvate supplements.


Subject(s)
Aging/drug effects , Dietary Supplements/analysis , Drug Contamination , Fibroblasts/cytology , Fibroblasts/drug effects , Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors , Pyruvic Acid/pharmacology , Cell Line , Dietary Supplements/adverse effects , Fibroblasts/chemistry , Fibroblasts/enzymology , Humans , Ketoglutarate Dehydrogenase Complex/chemistry , Ketoglutarate Dehydrogenase Complex/metabolism , Pyruvic Acid/chemistry
14.
Sci Rep ; 7(1): 2304, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28536428

ABSTRACT

Ubiquitin (Ub) shares the highest sequence identity with neuronal-precursor-cell-expressed developmentally downregulated protein-8 (NEDD8) in the Ub-like protein family. However, different enzyme systems are precisely employed for targeting Ub and NEDD8 to specific substrates. The molecular determinants for distinguishing between Ub and NEDD8 by Ub-specific peptidases (USPs) remain poorly characterized. By replacing the non-conserved residues of Ub with their NEDD8 equivalents by mutagenesis, and vice versa, we observed that the Ub4K, Ub12E, and Ub14E mutants partially and the Ub4K/12E/14E/72A mutant completely prevented their hydrolysis by USP2. The NEDD84F and NEDD814T mutants were slightly hydrolyzed by USP2; however, the NEDD812T/14T/72R and NEDD84F/12T/14T/72R mutants were accessible for hydrolysis by USP2, suggesting that Ub and NEDD8 residues 4, 12, 14, and 72 serve as the molecular determinants for specific recognition by USP2. We also demonstrated that the level of inhibition caused by Ub mutants with multiple mutation sites was not purely additive when compared with the single mutation results. Furthermore, USP2 was determined to bind to the N-terminus of Ub to form a stable interaction, after which it binds with the C-terminus of Ub to ensure substrate specificity. The same results were also discovered when Ub, Ub4K/12E/14E/72A, NEDD8, and NEDD84F/12T/14T/72R were incubated with USP21.


Subject(s)
Amino Acids/metabolism , Endopeptidases/metabolism , NEDD8 Protein/metabolism , Ubiquitin/metabolism , Amino Acid Sequence , Amino Acids/genetics , Binding Sites/genetics , Binding, Competitive , HeLa Cells , Humans , Hydrolysis , Mutagenesis, Site-Directed , Mutation , NEDD8 Protein/genetics , Protein Binding , Sequence Homology, Amino Acid , Substrate Specificity , Ubiquitin/genetics , Ubiquitin Thiolesterase
15.
PLoS One ; 12(5): e0177115, 2017.
Article in English | MEDLINE | ID: mdl-28472155

ABSTRACT

It has been proposed that malto-oligosaccharides (MOSs) are possibly recycled back into amylopectin biosynthesis via the sequential reactions catalyzed by plastidial α-glucan phosphorylase 1 (Pho1) and disproportionating enzyme 1 (Dpe1). In the present study, the reciprocal co-immunoprecipitation experiments using specific antibodies against Pho1 and Dpe1 demonstrated that these two enzymes can form a complex (the PD complex) in Ipomoea batatas storage roots. The immunohistochemistry analyses also revealed the co-localization of Pho1 and Dpe1 in the amyloplasts, and the protein levels of Pho1 and Dpe1 increased gradually throughout sweet potato storage root development. A high molecular weight PD complex was co-purified from sweet potato storage root lysates by size exclusion chromatography. Enzyme kinetic analyses showed that the PD complex can catalyze maltotriose and maltotetraose to generate glucose-1-phosphate in the presence of inorganic phosphate, and it also performs greater Dpe1 activity toward MOSs than does free form Dpe1. These data suggest that Pho1 and Dpe1 may form a metabolon complex, which provides elevated metabolic fluxes for MOS metabolism via a direct transfer of sugar intermediates, resulting in recycling of glucosyl units back into amylopectin biosynthesis more efficiently.


Subject(s)
Ipomoea/enzymology , Oligosaccharides/metabolism , Phosphorylases/metabolism , Plant Roots/enzymology , Plastids/enzymology , Catalysis , Chromatography, Gel , Immunoprecipitation , Ipomoea/metabolism , Mass Spectrometry , Plant Roots/metabolism
16.
BMC Biotechnol ; 17(1): 2, 2017 01 07.
Article in English | MEDLINE | ID: mdl-28061848

ABSTRACT

BACKGROUND: Human infection with avian influenza A virus (H7N9) was first reported in China in March 2013. Since then, hundreds of cases have been confirmed showing severe symptoms with a high mortality rate. The virus was transmitted from avian species to humans and has spread to many neighboring areas, raising serious concerns over its pandemic potential. Towards containing the disease, the goal of this study is to prepare a virus-like particle (VLP) that consists of hemagglutinin (HA), neuraminidase (NA) and matrix protein 1 (M1) derived from the human isolate A/Taiwan/S02076/2013(H7N9) for potential vaccine development. RESULTS: Full length HA, NA, and M1 protein genes were cloned and expressed using a baculoviral expression system, and the VLPs were generated by co-infecting insect cells with three respective recombinant baculoviruses. Nanoparticle tracking analysis and transmission electron microscopy were applied to verify the VLPs' structure and antigenicity, and the multiplicity of infection of the recombinant baculoviruses was adjusted to achieve the highest hemagglutination activity. In animal experiments, BALB/c mice and specific-pathogen-free chickens receiving the VLP immunization showed elevated hemagglutination inhibition serum titer and antibodies against NA and M1 proteins. In addition, examination of cellular immunity showed the VLP-immunized mice and chickens exhibited an increased splenic antigen-specific cytokines production. CONCLUSIONS: The H7N9 VLPs possess desirable immunogenicity in vivo and may serve as a candidate for vaccine development against avian influenza A (H7N9) infection.


Subject(s)
Antigens, Viral/immunology , Chickens/immunology , Influenza A virus/immunology , Mice/immunology , Vaccines, Virus-Like Particle/immunology , Animals , Antigens, Viral/genetics , Female , Influenza A virus/genetics , Mice, Inbred BALB C , Protein Engineering , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Species Specificity , Vaccines, Virus-Like Particle/genetics
17.
Anal Chem ; 88(12): 6349-56, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27254752

ABSTRACT

Thin-film direct coating (TDC) has been successfully used in Western blotting (WB). In this study, the advanced technique of TDC with suction (TDCS) was developed to reduce the consumption amount of antibody by a factor of up to 10(4) in comparison with the amount consumed by the conventional WB using the capillary tube without any need of special micromachining processes. The operation time for completely finishing a high-quality WB can be reduced from 3 h in conventional WB to about 5 min or even less by TDCS. In addition, the signal-to-noise ratio of the immunoblotting by TDCS can be markedly increased. TDCS WB showed a high linearity within a 6-log2 dynamic range for detecting 90-6000 ng of purified recombinant glutathione-S-transferase (GST) proteins and could particularly detect extrinsic GST proteins added in crude Escherichia coli or 293T cell lysates. Moreover, a protein mixture containing bovine serum albumin, GST, and ubiquitin could be specifically probed in parallel with their corresponding antibodies through multichannel TDCS WB. This simple and innovative TDCS WB offers various potential applications in simultaneously finishing multiple antibody-antigen screenings in a fast and single experiment.

18.
Am J Phys Med Rehabil ; 95(4): 270-6, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26334418

ABSTRACT

BACKGROUND: The aim of this study was to evaluate the efficacy of the Mann Assessment of Swallowing Ability (MASA) to predict the results of videofluoroscopic swallowing studies. METHOD: Children with cerebral palsy with suspicion of aspiration were enrolled. The Functional Dysphagia Scale (FDS) was used to quantify the swallowing dysfunction in videofluoroscopic swallowing studies. Correlation between MASA and FDS scores and differences in these two scores between aspirators and nonaspirators and between silent and overt aspirators were analyzed. RESULTS: Sixteen patients, level IV or V according the Gross Motor Function Classification System, were included. Thirteen patients (81.3%) had aspiration, and 9 (69.2%) were silent aspirators. The MASA scores between aspirators and nonaspirators were not different (median values of total scores, 107.0 and 94.0). The aspirators had higher FDS pharyngeal subtotal scores (P = 0.024) and slightly higher total FDS scores (P = 0.059). The differences in these two scales between silent and overt aspirators were not significant. Correlation coefficients between oral phase subtotal FDS scores and MASA subtotal scores in oral preparation, oral phase, and oral phase total were -0.713 (P < 0.05), -0.428 (P = 0.098), and -0.665 (P < 0.05), respectively. No correlation was found between the pharyngeal subtotal scores in these two scales. CONCLUSION: MASA was not useful in differentiating aspirators and nonaspirators and between silent and overt aspirators in severely disabled cerebral palsy, but it could predict oral dysfunction in videofluoroscopic swallowing studies.


Subject(s)
Cerebral Palsy/complications , Deglutition Disorders/etiology , Fluoroscopy , Respiratory Aspiration/diagnosis , Video Recording , Adolescent , Barium Sulfate , Child , Contrast Media , Deglutition , Deglutition Disorders/complications , Female , Humans , Male , Respiratory Aspiration/etiology , Severity of Illness Index , Young Adult
19.
Neuropsychiatr Dis Treat ; 11: 41-9, 2015.
Article in English | MEDLINE | ID: mdl-25657579

ABSTRACT

The objectives of this study were to determine a safe rotatory vestibular stimulation input for preschool children and to study the effects of grade level and sex of preschool children during active, passive, clockwise, and counterclockwise rotation vestibular input. This study adopted purposive sampling with 120 children from three kindergarten levels (K1, K2, and K3) in Taiwan. The subjects ranged in age from 46 to 79 months of age (mean: 62.1 months; standard deviation =9.60). This study included testing with four types of vestibular rotations. The number, duration, and speed of rotations were recorded. The study found that the mean number of active rotations was 10.28; the mean duration of rotation was 24.17 seconds; and the mean speed was 2.29 seconds per rotation. The mean number of passive rotations was 23.04. The differences in number of rotations in clockwise, counterclockwise, active, and passive rotations were not statistically significant. Sex and grade level were not important related factors in the speed and time of active rotation. Different sexes, rotation methods (active, passive), and grades made significant differences in the number of rotations. The safety and tolerability of rotatory vestibular stimulation input data obtained in this study can provide useful reference data for therapists using sensory integration therapy.

20.
Virus Res ; 197: 101-7, 2015 Feb 02.
Article in English | MEDLINE | ID: mdl-25527464

ABSTRACT

Cleavage of the hemagglutinin (HA) precursor (HA0) by trypsin, which produces the active HA1 and HA2 complex, is a critical step for activating the avian influenza virus (AIV). However, other tryptic cleavage sites on HA might also cause HA degradation and affect the virulence. Otherwise, HA is modified by glycosylation in the host cell. The conjugated glycans on HA may hinder the antigenic epitopes, and thus prevent the virus from being recognized and attacked by the antibodies. In this study, we observed that glycosylation at the Asn-167 (N167) site on the HA1 of the H6N1 AIV strain A/chicken/Taiwan/2838V/00 (2838V) protected Arg-201 (R201) from tryptic cleavage. The 2838V HA protein became sensitive to tryptic cleavage, whereas the glycans at N167 were removed by N-glycosidase F (PNGase-F). Furthermore, the infectivity of 2838V decreased when pretreated with PNGase-F and trypsin. Our observations suggest that the inaccessibility of the R201 residue of HA1 for tryptic cleavage, which is sterically hindered by glycosylation at N167, is a crucial factor for determining the infectivity of the AIV.


Subject(s)
Glycosylation , Influenza A virus/physiology , Trypsin/metabolism , Animals , Arginine/metabolism , Asparagine/metabolism , Cell Line , Chick Embryo , Hemagglutinin Glycoproteins, Influenza Virus , Hydrolysis
SELECTION OF CITATIONS
SEARCH DETAIL