Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Genet ; 19(12): e1011081, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38048317

ABSTRACT

Haploid males of hymenopteran species produce gametes through an abortive meiosis I followed by meiosis II that can either be symmetric or asymmetric in different species. Thus, one spermatocyte could give rise to two spermatids with either equal or unequal amounts of cytoplasm. It is currently unknown what molecular features accompany these postmeiotic sperm cells especially in species with asymmetric meiosis II such as bees. Here we present testis single-cell RNA sequencing datasets from the honeybee (Apis mellifera) drones of 3 and 14 days after emergence (3d and 14d). We show that, while 3d testes exhibit active, ongoing spermatogenesis, 14d testes only have late-stage spermatids. We identify a postmeiotic bifurcation in the transcriptional roadmap during spermatogenesis, with cells progressing toward the annotated spermatids (SPT) and small spermatids (sSPT), respectively. Despite an overall similarity in their transcriptomic profiles, sSPTs express the fewest genes and the least RNA content among all the sperm cell types. Intriguingly, sSPTs exhibit a relatively high expression level for Hymenoptera-restricted genes and a high mutation load, suggesting that the special meiosis II during spermatogenesis in the honeybee is accompanied by phylogenetically young gene activities.


Subject(s)
Semen , Spermatogenesis , Bees/genetics , Male , Animals , Spermatogenesis/genetics , Spermatids/metabolism , Testis , Spermatocytes/metabolism , Meiosis/genetics
2.
Nano Lett ; 24(15): 4423-4432, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38568019

ABSTRACT

The HIV-1 envelope is a heavily glycosylated class 1 trimeric fusion protein responsible for viral entry into CD4+ immune cells. Developing neutralizing antibodies against the specific envelope glycans is an alternative method for antiviral therapies. This work presents the first-ever development and characterization of artificial neutralizing antibodies using molecular imprinting technology to recognize and bind to the envelope protein of HIV-1. The prepared envelope glycan-imprinted nanoparticles (GINPs) can successfully prevent HIV-1 from infecting target cells by shielding the glycans on the envelope protein. In vitro experiments showed that GINPs have strong affinity toward HIV-1 (Kd = 36.7 ± 2.2 nM) and possess high anti-interference and specificity. GINPs demonstrate broad inhibition activity against both tier 1 and tier 2 HIV-1 strains with a pM-level IC50 and exhibit a significant inhibitory effect on long-term viral replication by more than 95%. The strategy provides a promising method for the inhibition and therapy of HIV-1 infection.


Subject(s)
HIV Infections , HIV-1 , Humans , Antibodies, Neutralizing , HIV Antibodies/metabolism , Glycosylation , HIV Infections/drug therapy , Polysaccharides/metabolism
3.
Nano Lett ; 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38922640

ABSTRACT

Enhancing the antitumor immune response and targeting ability of oncolytic viruses will improve the effect of tumor immunotherapy. Through infecting neural stem cells (NSCs) with a capsid dual-modified oncolytic adenovirus (CRAd), we obtained and characterized the "oncolytic extracellular vesicles" (CRAdEV) with improved targeted infection and tumor killing activity compared with CRAd. Both ex vivo and in vivo studies revealed that CRAdEV activated innate immune cells and importantly enhanced the immunomodulatory effect compared to CRAd. We found that CRAdEV effectively increased the number of DCs and activated CD4+ and CD8+ T cells, significantly increased the number and activation of B cells, and produced higher levels of tumor-specific antibodies, thus eliciting enhanced antitumor activity compared with CRAd in a B16 xenograft immunocompetent mice model. This study provides a novel approach to oncolytic adenovirus modification and demonstrates the potential of "oncolytic extracellular vesicles" in antitumor immunotherapy.

4.
J Virol ; 97(1): e0146722, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36475768

ABSTRACT

Assembly of the adenovirus capsid protein hexon depends on the assistance of the molecular chaperone L4-100K. However, the chaperone mechanisms remain unclear. In this study, we found that L4-100K was involved in the hexon translation process and could prevent hexon degradation by the proteasome in cotransfected human cells. Two nonadjacent domains, 84-133 and 656-697, at the N-terminal and C-terminal regions of human adenovirus type 5 L4-100K, respectively, were found to be crucial and cooperatively responsible for hexon trimer expression and assembly. These two chaperone-related domains were conserved in the sequence of L4-100K and in the function of hexon assembly across different adenovirus serotypes. Different degrees of cross-activity of hexon trimerization with different serotypes were detected in subgroups B, C, and D, which were proven to be controlled by the interaction between the C-terminal chaperone-related domain of L4-100K and hypervariable regions (HVR) of hexon. Additionally, HVR-chimeric hexon mutants were successfully assembled with the assistance of the 1-697 mutant. Structural analysis of 656-697 by nuclear magnetic resonance and structural prediction of L4-100K using Robetta showed that the two conserved domains are mainly composed of α-helices and are located on the surface of the highly folded core region. Our research provides a more complete understanding of hexon assembly and guidance for the development of hexon-chimeric adenovirus vectors that will be safer, smarter, and more efficient. IMPORTANCE Adenovirus vectors have been widely used in clinical trials of vaccines and gene therapy, although some deficiencies remain. Chimeric modification of the hexon was expected to improve the potency of preexisting immune evasion and targeting, but in many cases, viral packaging is prevented by the inability of the chimeric hexon to assemble correctly. So far, few studies have examined the mechanisms of hexon trimer assembly. Here, we show how the chaperone protein L4-100K contributes to the assembly of the adenovirus capsid protein hexon, and these data will provide a guide for novel adenovirus vector design and development, as we desired.


Subject(s)
Adenoviruses, Human , Molecular Chaperones , Viral Nonstructural Proteins , Humans , Adenoviruses, Human/genetics , Adenoviruses, Human/metabolism , Capsid/metabolism , Capsid Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
5.
Nano Lett ; 23(23): 11120-11128, 2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38032110

ABSTRACT

An oncolytic virus is a promising strategy for glioblastoma (GBM) therapy. However, there are still some challenges such as the blood-brain barrier (BBB) and preexisting immunity for targeted treatment of GBM with an oncolytic virus. In this study, two kinds of cell membrane-coated oncolytic adenoviruses (NCM-Ad and GCM-Ad) were prepared using neural stem cells (NSCs) and GBM cells as sources of membranes, respectively, and were shown to improve the targeted infectivity on GBM cells and avoid the immune clearance of preexisting neutralizing antibodies in vitro and in vivo. Specifically, NCM-Ad showed a strong ability to cross the BBB and target tumor cells in vivo. To improve the cytotoxicity to GBM, a capsid dual-modified oncolytic adenovirus (A4/k37) was also encapsulated, and NCM-A4/k37 showed outstanding tumor targeting and inhibition capacity in an orthotopic xenograft tumor model of GBM upon intravenous administration. This study provides a promising oncolytic virus-based targeted therapeutic strategy for glioma.


Subject(s)
Brain Neoplasms , Glioblastoma , Oncolytic Virotherapy , Oncolytic Viruses , Humans , Glioblastoma/therapy , Glioblastoma/pathology , Adenoviridae/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Xenograft Model Antitumor Assays , Oncolytic Viruses/genetics , Cell Membrane/metabolism
6.
Int J Mol Sci ; 25(3)2024 Feb 04.
Article in English | MEDLINE | ID: mdl-38339158

ABSTRACT

With the advent of cancer immunotherapy, there is a growing interest in vaccine development as a means to activate the cellular immune system against cancer. Despite the promise of DNA vaccines in this regard, their effectiveness is hindered by poor immunogenicity, leading to modest therapeutic outcomes across various cancers. The role of Type 1 conventional dendritic cells (cDC1), capable of cross-presenting vaccine antigens to activate CD8+T cells, emerges as crucial for the antitumor function of DNA vaccines. To address the limitations of DNA vaccines, a promising approach involves targeting antigens to cDC1 through the fusion of XCL1, a ligand specific to the receptor XCR1 on the surface of cDC1. Here, female C57BL/6 mice were selected for tumor inoculation and immunotherapy. Additionally, recognizing the complexity of cancer, this study explored the use of combination therapies, particularly the combination of cDC1-targeted DNA vaccine with the chemotherapy drug Gemcitabine (Gem) and the anti-PD1 antibody in a mouse lung cancer model. The study's findings indicate that fusion antigens with XCL1 effectively enhance both the immunogenicity and antitumor effects of DNA vaccines. Moreover, the combination of the cDC1-targeted DNA vaccine with Gemcitabine and anti-PD1 antibody in the mouse lung cancer model demonstrates an improved antitumor effect, leading to the prolonged survival of mice. In conclusion, this research provides important support for the clinical investigation of cDC1-targeting DNA vaccines in combination with other therapies.


Subject(s)
Cancer Vaccines , Lung Neoplasms , Vaccines, DNA , Animals , Female , Mice , CD8-Positive T-Lymphocytes , Dendritic Cells , Gemcitabine , Lung Neoplasms/therapy , Mice, Inbred C57BL , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use
7.
J Virol ; 96(17): e0055522, 2022 09 14.
Article in English | MEDLINE | ID: mdl-35950859

ABSTRACT

Apolipoprotein B mRNA-editing catalytic polypeptide-like 3 family members (APOBEC3s) are host restriction factors that inhibit viral replication. Viral infectivity factor (Vif), a human immunodeficiency virus type 1 (HIV-1) accessory protein, mediates the degradation of APOBEC3s by forming the Vif-E3 complex, in which core-binding factor beta (CBFß) is an essential molecular chaperone. Here, we screened nonfunctional Vif mutants with high affinity for CBFß to inhibit HIV-1 in a dominant negative manner. We applied the yeast surface display technology to express Vif random mutant libraries, and mutants showing high CBFß affinity were screened using flow cytometry. Most of the screened Vif mutants containing random mutations of different frequencies were able to rescue APOBEC3G (A3G). In the subsequent screening, three of the mutants restricted HIV-1, recovered G-to-A hypermutation, and rescued APOBEC3s. Among them, Vif-6M showed a cross-protection effect toward APOBEC3C, APOBEC3F, and African green monkey A3G. Stable expression of Vif-6M in T lymphocytes inhibited the viral replication in newly HIV-1-infected cells and the chronically infected cell line H9/HXB2. Furthermore, the expression of Vif-6M provided a survival advantage to T lymphocytes infected with HIV-1. These results suggest that dominant negative Vif mutants acting on the Vif-CBFß target potently restrict HIV-1. IMPORTANCE Antiviral therapy cannot eliminate HIV and exhibits disadvantages such as drug resistance and toxicity. Therefore, novel strategies for inhibiting viral replication in patients with HIV are urgently needed. APOBEC3s in host cells are able to inhibit viral replication but are antagonized by HIV-1 Vif-mediated degradation. Therefore, we screened nonfunctional Vif mutants with high affinity for CBFß to compete with the wild-type Vif (wtVif) as a potential strategy to assist with HIV-1 treatment. Most screened mutants rescued the expression of A3G in the presence of wtVif, especially Vif-6M, which could protect various APOBEC3s and improve the incorporation of A3G into HIV-1 particles. Transduction of Vif-6M into T lymphocytes inhibited the replication of the newly infected virus and the chronically infected virus. These data suggest that Vif mutants targeting the Vif-CBFß interaction may be promising in the development of a new AIDS therapeutic strategy.


Subject(s)
Core Binding Factor beta Subunit , HIV Infections , HIV-1 , vif Gene Products, Human Immunodeficiency Virus , APOBEC Deaminases/genetics , APOBEC Deaminases/metabolism , Animals , Cell Line , Chlorocebus aethiops , Core Binding Factor beta Subunit/genetics , HIV-1/genetics , HIV-1/physiology , Host-Pathogen Interactions , Humans , T-Lymphocytes/virology , Virus Replication , vif Gene Products, Human Immunodeficiency Virus/genetics
8.
J Neuroinflammation ; 19(1): 205, 2022 Aug 15.
Article in English | MEDLINE | ID: mdl-35971179

ABSTRACT

BACKGROUND: Progressive neuronal death is the key pathological feature of Alzheimer's disease (AD). However, the molecular mechanisms underlying the neuronal death in AD patients have not been fully elucidated. Necroptosis reportedly activates and induces neuronal death in patients with Alzheimer's disease (AD); however, the main mediators and mechanisms underlying necroptosis induction in AD remain elusive. METHODS: The function of hyperphosphorylated tau (pTau) in inducing necroptosis in neuronal cell was examined using Western blotting, RT-PCR and flow cytometry. Tau-induced inflammation was identified via RNA sequencing and transwell assay. Pharmacological methods and CRISPR-Cas9 technology were used to verify the role of necrosome proteins in pTau-stimulated neuronal death and inflammation. TauP301S model mice were treated with Nec-1 s to evaluate the role of necroptosis in tau pathology. RESULTS: Hyperphosphorylated tau could induce necroptosis in neuronal cells by promoting the formation of the RIPK1/RIPK3/MLKL necrosome. In addition, pTau significantly stimulated cell-autonomous overexpression of cytokines and chemokines via the intracellular nuclear factor kappa B (NF-κB) signaling pathway. Importantly, the RIPK1/RIPK3/MLKL axis was essential for the pTau-mediated NF-κB activation and cytokine storm. Furthermore, necroptosis stimulation, NF-κB activation, and cytokine induction have been detected in TauP301S mice and blocking necroptosis markedly ameliorated behavioral defects and excessive neuroinflammation in AD mice. CONCLUSIONS: Our study, for the first time, revealed that pTau contributes to neuronal death by inducing necroptosis and inflammation, mediated by activating the RIPK1/RIPK3/MLKL and NF-κB pathways, thereby delineating the hierarchical molecular network of neuronal necroptosis induction in AD.


Subject(s)
Alzheimer Disease , Necroptosis , Alzheimer Disease/genetics , Animals , Apoptosis/genetics , Inflammation/pathology , Mice , NF-kappa B/metabolism , Necrosis/pathology , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
9.
J Biol Chem ; 295(43): 14592-14605, 2020 10 23.
Article in English | MEDLINE | ID: mdl-32817167

ABSTRACT

HIV remains a health challenge worldwide, partly because of the continued development of resistance to drugs. Therefore, it is urgent to find new HIV inhibitors and targets. Apolipoprotein B mRNA-editing catalytic polypeptide-like 3 family members (APOBEC3) are important host restriction factors that inhibit HIV-1 replication by their cytidine deaminase activity. HIV-1 viral infectivity factor (Vif) promotes proteasomal degradation of APOBEC3 proteins by recruiting the E3 ubiquitin ligase complex, in which core-binding factor ß (CBFß) is a necessary molecular chaperone. Interrupting the interaction between Vif and CBFß can release APOBEC3 proteins to inhibit HIV-1 replication and may be useful for developing new drug targets for HIV-1. In this study, we identified a potent small molecule inhibitor CBFß/Vif-3 (CV-3) of HIV-1 replication by employing structure-based virtual screening using the crystal structure of Vif and CBFß (PDB: 4N9F) and validated CV-3's antiviral activity. We found that CV-3 specifically inhibited HIV-1 replication (IC50 = 8.16 µm; 50% cytotoxic concentration >100 µm) in nonpermissive lymphocytes. Furthermore, CV-3 treatment rescued APOBEC3 family members (human APOBEC3G (hA3G), hA3C, and hA3F) in the presence of Vif and enabled hA3G packaging into HIV-1 virions, which resulted in Gly-to-Ala hypermutations in viral genomes. Finally, we used FRET to demonstrate that CV-3 inhibited the interaction between Vif and CBFß by simultaneously forming hydrogen bonds with residues Gln-67, Ile-102, and Arg-131 of CBFß. These findings demonstrate that CV-3 can effectively inhibit HIV-1 by blocking the interaction between Vif and CBFß and that this interaction can serve as a new target for developing HIV-1 inhibitors.


Subject(s)
APOBEC Deaminases/metabolism , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Core Binding Factor beta Subunit/metabolism , HIV-1/drug effects , vif Gene Products, Human Immunodeficiency Virus/metabolism , Cell Line , HIV Infections/drug therapy , HIV Infections/metabolism , HIV-1/physiology , Host-Pathogen Interactions/drug effects , Humans , Molecular Docking Simulation , Protein Interaction Maps/drug effects , Proteolysis/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Virus Replication/drug effects
10.
J Biol Chem ; 295(13): 4252-4264, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32075911

ABSTRACT

SAM and HD domain-containing protein 1 (SAMHD1) is a host factor that restricts reverse transcription of lentiviruses such as HIV in myeloid cells and resting T cells through its dNTP triphosphohydrolase (dNTPase) activity. Lentiviruses counteract this restriction by expressing the accessory protein Vpx or Vpr, which targets SAMHD1 for proteasomal degradation. SAMHD1 is conserved among mammals, and the feline and bovine SAMHD1 proteins (fSAM and bSAM) restrict lentiviruses by reducing cellular dNTP concentrations. However, the functional regions of fSAM and bSAM that are required for their biological functions are not well-characterized. Here, to establish alternative models to investigate SAMHD1 in vivo, we studied the restriction profile of fSAM and bSAM against different primate lentiviruses. We found that both fSAM and bSAM strongly restrict primate lentiviruses and that Vpx induces the proteasomal degradation of both fSAM and bSAM. Further investigation identified one and five amino acid sites in the C-terminal domain (CTD) of fSAM and bSAM, respectively, that are required for Vpx-mediated degradation. We also found that the CTD of bSAM is directly involved in mediating bSAM's antiviral activity by regulating dNTPase activity, whereas the CTD of fSAM is not. Our results suggest that the CTDs of fSAM and bSAM have important roles in their antiviral functions. These findings advance our understanding of the mechanism of fSAM- and bSAM-mediated viral restriction and might inform strategies for improving HIV animal models.


Subject(s)
HIV/genetics , Lentivirus/genetics , Reverse Transcription/genetics , SAM Domain and HD Domain-Containing Protein 1/genetics , Animals , Cats , Cattle , HEK293 Cells , HIV/pathogenicity , Host-Pathogen Interactions/genetics , Humans , Lentivirus/pathogenicity , Myeloid Cells/virology , Protein Domains/genetics , SAM Domain and HD Domain-Containing Protein 1/chemistry , T-Lymphocytes/virology , Virus Replication/genetics
11.
J Neuroinflammation ; 17(1): 72, 2020 Feb 24.
Article in English | MEDLINE | ID: mdl-32093751

ABSTRACT

BACKGROUND: Tau hyper-phosphorylation has been considered a major contributor to neurodegeneration in Alzheimer's disease (AD) and related tauopathies, and has gained prominence in therapeutic development for AD. To elucidate the pathogenic mechanisms underlying AD and evaluate therapeutic approaches targeting tau, numerous transgenic mouse models that recapitulate critical AD-like pathology have been developed. Tau P301S transgenic mice is one of the most widely used mouse models in AD research. Extensive studies have demonstrated that sex significantly influences AD pathology, behavioral status, and therapeutic outcomes, suggesting that studies using mouse models of AD must consider sex- and age-related differences in neuropathology, behavior, and plasma content. METHOD: We systematically investigated differences in tau P301S transgenic mice (PS19 line) and wildtype littermates of different sex behavioral performance, tau neuropathology, and biomarkers in plasma and brain. RESULTS: Male P301S transgenic mice exhibited significant changes in weight loss, survival rate, clasping, kyphosis, composite phenotype assessment, nest building performance, tau phosphorylation at Ser202/Thr205, and astrocyte activation compared to that of wild-type littermates. In contrast, female P301S transgenic mice were only sensitive in the Morris water maze and open field test. In addition, we characterized the absence of macrophage-inflammatory protein (MIP-3α) and the upregulation of interferon (IFN)-γ, interleukin (IL)-5, and IL-6 in the plasma of P301S transgenic mice, which can be served as potential plasma biomarkers in P301S Tg mice. Male P301S transgenic mice expressed more monokine induced by IFN-γ (MIG), tumor necrosis factor-α (TNF-α), IL-10, and IL-13 than those of female P301S mice. CONCLUSION: Our findings highlight sexual dimorphism in the behavior, neuropathology, and plasma proteins in tau P301S transgenic AD mice, indicating that the use of male P301S transgenic mice may be more suitable for assessing anti-phosphorylated tau therapeutic strategies for AD and related tauopathies, and the MIP-3α may be a new potential plasma biomarker.


Subject(s)
Alzheimer Disease , Chemokine CCL20/blood , Disease Models, Animal , Sex Characteristics , tau Proteins/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Biomarkers/blood , Female , Humans , Male , Mice , Mice, Transgenic
12.
Protein Expr Purif ; 175: 105688, 2020 11.
Article in English | MEDLINE | ID: mdl-32681953

ABSTRACT

BST2 is a single-pass type II transmembrane (TM) protein, which has a cytoplasmic domain, a transmembrane domain, and an extracellular domain, each domain is important for biologic function of BST2. BST2 is a host restriction factor that can effectively inhibit retrovirus release. Rhesus monkeys are considered as relevant natural animal models for studying AIDS in humans. In order to recognize rhesus BST2 (RhBST2) protein and detect its function accurately, we prepared a polyclonal antibody (pAb) especially for RhBST2. Meanwhile, we constructed RhBST2 proteins with the addition of an HA-tag at the N-terminus (RhBST2-NHA) or inside of the ectodomain (RhBST2-IHA) to compare the recognition ability of rabbit anti-RhBST2 pAb and anti-HA mAb. The results showed that the anti-HA mAb and rabbit anti-RhBST2 pAb had the same ability to identify RhBST2. RhBST2 demonstrated antiviral activity and the ability to activate NF-κB. Moreover, the N-glycosylation states, cell surface level and intracellular localization of RhBST2 were detected. However, HA tags relatively changed part of the biological function of RhBST2. These results show that the RhBST2 polyclonal antibody is more suitable for analyzing the properties and functions of RhBST2, and the natural domain of RhBST2 is very important for its function.


Subject(s)
Antibodies/immunology , Antigens, CD , Antiviral Agents , Acquired Immunodeficiency Syndrome , Animals , Antigens, CD/biosynthesis , Antigens, CD/chemistry , Antigens, CD/immunology , Antiviral Agents/chemistry , Antiviral Agents/immunology , Antiviral Agents/metabolism , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/immunology , HEK293 Cells , HIV-1/immunology , Humans , Macaca mulatta , Protein Domains , Rabbits
13.
Microbiol Immunol ; 64(1): 52-62, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31544982

ABSTRACT

An integrase-defective SIV (idSIV) vaccine delivered by a DNA prime and viral particle boost approach can suppress viral loads (VLs) during the acute infection stage after intravenous SIVmac239 challenge. This study investigated how idSIV DNA and viral particle immunization alone contributed to the suppression of VLs in Chinese rhesus macaques after SIV challenge. Two macaques were immunized with idSIV DNA five times and two macaques were immunized with idSIV viral particles three times. Cellular and humoral immune responses were measured in the vaccinated macaques after immunization. The VLs and CD4+ T cell counts were monitored for 28 weeks after the intravenous SIVmac239 challenge. The SIV-specific T cell responses were only detected in the DNA-vaccinated macaques. However, binding and neutralizing antibodies against autologous and heterologous viruses were moderately better in macaques immunized with viral particles than in macaques immunized with DNA. After the challenge, the mean peak viremia in the DNA group was 2.3 logs lower than that in the control group, while they were similar between the viral particle immunization and control groups. Similar CD4+ T cell counts were observed among all groups. These results suggest that idSIV DNA immunization alone reduces VLs during acute infection after SIV challenge in macaques and may serve as a key component in combination with other immunogens as prophylactic vaccines.


Subject(s)
Proviruses/immunology , SAIDS Vaccines/immunology , Vaccines, DNA/immunology , Viremia/prevention & control , Animals , Antibodies, Neutralizing , Antibodies, Viral/blood , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes , Disease Models, Animal , Immunity, Humoral , Immunization , Macaca mulatta , Proviruses/genetics , SAIDS Vaccines/genetics , SAIDS Vaccines/therapeutic use , Simian Immunodeficiency Virus/genetics , Vaccination , Vaccines, DNA/therapeutic use , Viral Load
14.
J Cell Mol Med ; 23(5): 3656-3664, 2019 05.
Article in English | MEDLINE | ID: mdl-30905075

ABSTRACT

Castration-resistant progression of prostate cancer is a major cause of prostate cancer mortality, and increased expression and activity of the full-length and the splice variants of androgen receptor (AR) have been indicated to drive castration resistance. Consequently, there is an urgent need to develop agents that can target both the full-length and the splice variants of AR for more effective treatment of prostate cancer. In the present study, we showed that raddeanin A (RA), an oleanane-type triterpenoid saponin, suppresses the transcriptional activities of both the full-length and the splice variants of AR. This is attributable to their decreased expression as a result of RA induction of proteasome-mediated degradation and inhibition of the transcription of the AR gene. We further showed the potential of using RA to enhance the growth inhibitory efficacy of docetaxel, the first-line chemotherapy for prostate cancer. This study identifies RA as a new agent to target both the full-length and the splice variants of AR and provides a rationale for further developing RA for prostate cancer treatment.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Prostatic Neoplasms/genetics , RNA Splicing , Receptors, Androgen/genetics , Saponins/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Docetaxel/therapeutic use , Down-Regulation/drug effects , Drug Synergism , Humans , Male , PC-3 Cells , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Isoforms/genetics
15.
Cancer Sci ; 110(11): 3442-3452, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31432592

ABSTRACT

Functional E-cadherin loss, a hallmark of epithelial-mesenchymal transition (EMT), is important for metastasis. However, the mechanism of Snail2 in hepatocellular carcinoma (HCC) EMT and metastasis remains unclear. Here, we showed that Snail2 was upregulated in primary HCC, and significantly increased during transforming growth factor-ß-induced liver cell EMT. Snail2-overexpressing and knockdown cell lines have been established to determine its function in EMT in HCC. H3K9 methylation was upregulated and H3K4 and H3K56 acetylation were downregulated at the E-cadherin promoter in Snail2-overexpressing cancer cells. Furthermore, Snail2 interacted with G9a and histone deacetylases (HDACs) to form a complex to suppress E-cadherin transcription. Snail2 overexpression enhanced migration and invasion in HCC cells, whereas G9a and HDAC inhibition significantly reversed this effect. Moreover, Snail2 overexpression in cancer cells increased tumor metastasis and shortened survival time in mice, whereas G9a and HDAC inhibitors extended survival. Our study not only reveals a critical mechanism underlying the epigenetic regulation of EMT but also suggests novel treatment strategies for HCC.


Subject(s)
Cadherins/metabolism , Carcinoma, Hepatocellular/metabolism , Epigenesis, Genetic , Epithelial-Mesenchymal Transition/genetics , Histocompatibility Antigens/metabolism , Histone Deacetylases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Liver Neoplasms/metabolism , Snail Family Transcription Factors/metabolism , Acetylation , Animals , Azepines/therapeutic use , Carcinoma, Hepatocellular/secondary , Cell Movement , Disease Progression , Down-Regulation , Female , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydroxamic Acids/therapeutic use , Liver/drug effects , Liver/metabolism , Liver Neoplasms/pathology , Methylation , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Quinazolines/therapeutic use , Transcription, Genetic , Transforming Growth Factor beta/pharmacology
16.
BMC Immunol ; 20(1): 9, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30755174

ABSTRACT

BACKGROUND: Active immunotherapy targeting amyloid-ß (Aß) is a promising treatment for Alzheimer's disease (AD). Numerous preclinical studies and clinical trials demonstrated that a safe and effective AD vaccine should induce high titers of anti-Aß antibodies while avoiding the activation of T cells specific to Aß. RESULTS: An untagged Aß1-6 chimeric protein vaccine against AD based on norovirus (NoV) P particle was expressed in Escherichia coli and obtained by sequential chromatography. Analysis of protein characteristics showed that the untagged Aß1-6 chimeric protein expressed in soluble form exhibited the highest particle homogeneity, with highest purity and minimal host cell protein (HCP) and residual DNA content. Importantly, the untagged Aß1-6 chimeric soluble protein could induce the strongest Aß-specific humoral immune responses without activation of harmful Aß-specific T cells in mice. CONCLUSIONS: The untagged Aß1-6 chimeric protein vaccine is safe and highly immunogenic. Further research will determine the efficacy in cognitive improvement and disease progression delay.


Subject(s)
Alzheimer Vaccines/immunology , Amyloid beta-Peptides/immunology , Antibodies/immunology , Genetic Vectors , Norovirus , Recombinant Fusion Proteins , Alzheimer Disease/therapy , Alzheimer Vaccines/administration & dosage , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/isolation & purification , Animals , Antibody Specificity/immunology , Disease Models, Animal , Immunization , Mice , Norovirus/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
17.
Protein Expr Purif ; 130: 28-34, 2017 02.
Article in English | MEDLINE | ID: mdl-27693623

ABSTRACT

The Norovirus (NoV) P domain, with three surface loops for foreign antigen insertion, has been demonstrated as an excellent platform for antigen presentation and novel vaccine development. The P domain alone can self-assemble into a P dimer, 12-mer small particle or 24-mer P particle, and vaccines based on those particles may elicit different levels of immunogenicity. Currently, P particles are generally produced in soluble expression systems in Escherichia coli, mainly in the 24-mer form. However, the low yield of the soluble protein has hindered further clinical applications of P particle-based protein vaccines. In this study, we inserted the Alzheimer's disease (AD) immunogen Aß1-6 into the three loops of the P particle to generate an AD protein vaccine. To increase the yield of this chimeric protein, we tested the generation of proteins in a soluble expression system and an inclusion body expression system separately in E. coli. The result showed that the inclusion body expression system could greatly enhance the product yield of the chimeric protein compared with the soluble expression system. The refolded protein from the inclusion bodies was mainly in the 12-mer form, while the protein generated from the soluble supernatant was mainly in the 24-mer form. Moreover, the immunogenicity of soluble proteins was significantly stronger than that of the refolded proteins. Thus, comparisons between the two expression methods suggested that the soluble expression system generated chimeric P particles with better immunogenicity, while inclusion body expression system yielded more P particle proteins.


Subject(s)
Antigens, Viral , Gene Expression , Norovirus/genetics , Recombinant Fusion Proteins , Viral Proteins , Viral Vaccines , Animals , Antigens, Viral/biosynthesis , Antigens, Viral/chemistry , Antigens, Viral/genetics , Antigens, Viral/isolation & purification , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Immunogenicity, Vaccine , Mice , Pichia/genetics , Pichia/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Viral Proteins/biosynthesis , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/isolation & purification , Viral Vaccines/biosynthesis , Viral Vaccines/chemistry , Viral Vaccines/genetics , Viral Vaccines/isolation & purification
18.
J Pept Sci ; 23(3): 245-251, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28211253

ABSTRACT

The abnormal deposition of amyloid-ß (Aß) peptides in the brain is the main neuropathological hallmark of Alzheimer's disease (AD). Amyloid deposits are formed by a heterogeneous mixture of Aß peptides, among which the most studied are Aß40 and Aß42. Aß40 is abundantly produced in the human brain, but the level of Aß42 is remarkably increased in the brain of AD patients. Aside from Aß40 and Aß42, recent data have raised the possibility that Aß43 peptides may be instrumental in AD pathogenesis. Besides its length, whether the Aß aggregated form accounts for the neurotoxicity is also particularly controversial. Aß fibrils are generally considered as key pathogenic substances in AD pathogenesis. Nevertheless, recent data implicated soluble Aß oligomers as the main cause of synaptic dysfunction and memory loss in AD. To further address this uncertainty, we analyzed the neurotoxicity of different Aß species and Aß forms at the cellular level. The results showed that Aß42 could form oligomers significantly faster than Aß40 and Aß43 and Aß42 oligomers showed the greatest level of neurotoxicity. Regardless of the length of Aß peptides, Aß oligomers induced significantly higher cytotoxicity compared with the other two Aß forms. Surprisingly, the neurotoxicity of fibrils in PC12 cells was only marginally but not significantly stronger than monomers, contrary to previous reports. Altogether, our findings demonstrate the high pathogenicity of Aß42 among the three Aß species and support the idea that Aß42 oligomers contribute to the pathological events leading to neurodegeneration in AD. Copyright © 2017 European Peptide Society and John Wiley & Sons, Ltd.


Subject(s)
Amyloid beta-Peptides/toxicity , Peptide Fragments/toxicity , Protein Aggregates , Amyloid beta-Peptides/chemical synthesis , Animals , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Kinetics , PC12 Cells , Peptide Fragments/chemical synthesis , Rats
19.
Int J Med Sci ; 14(4): 340-347, 2017.
Article in English | MEDLINE | ID: mdl-28553166

ABSTRACT

Methamphetamine (METH) exerts significant neurotoxicity in experimental animals and humans when taken at high doses or abused chronically. Long-term abusers have decreased dopamine levels, and they are more likely to develop Parkinson's disease (PD). To date, few medications are available to treat the METH-induced damage of neurons. Glial cell line-derived neurotrophic factor (GDNF) has been previously shown to reduce the dopamine-depleting effects of neurotoxic doses of METH. However, the effect of cerebral dopamine neurotrophic factor (CDNF), which has been reported to be more specific and efficient than GDNF in protecting dopaminergic neurons against 6-OHDA toxicity, in attenuating METH neurotoxicity has not been determined. Thus, the present study aimed to evaluate the neuroprotective effect of CDNF against METH-induced damage to the dopaminergic system in vitro and in vivo. In vitro, CDNF protein increased the survival rate and reduced the tyrosine hydroxylase (TH) loss of METH-treated PC12 cells. In vivo, METH was administered to rats following human CDNF overexpression mediated by the recombinant adeno-associated virus. Results demonstrated that CDNF overexpression in the brain could attenuate the METH-induced dopamine and TH loss in the striatum but could not lower METH-induced hyperthermia.


Subject(s)
Gene Transfer Techniques , Methamphetamine/toxicity , Nerve Growth Factors/genetics , Neurotoxicity Syndromes/therapy , Animals , Corpus Striatum/drug effects , Corpus Striatum/pathology , Dopamine/metabolism , Fever/chemically induced , Fever/drug therapy , Fever/genetics , Genetic Therapy , Humans , Nerve Growth Factors/therapeutic use , Neurons/drug effects , Neurons/pathology , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/pathology , PC12 Cells , Rats , Tyrosine 3-Monooxygenase/genetics
20.
Immunopharmacol Immunotoxicol ; 39(1): 37-44, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28004985

ABSTRACT

Cyclophosphamide (CY) is a DNA alkylating agent, which is widely used with other chemotherapy drugs in the treatment of various types of cancer. It can be used not only as a chemotherapeutic but also as an immunomodulatory agent to inhibit IL-10 expression and T regulatory cells (Tregs). Fibroblast activation protein α (FAPα) is expressed in cancer-associated fibroblasts in the tumor microenvironment. Immunotherapy based on FAPα, as a tumor stromal antigen, typically induces specific immune response targeting the tumor microenvironment. This study evaluated the efficacy of a previously unreported CY combination strategy to enhance the limited anti-tumor effect of a DNA vaccine targeting FAPα. The results suggested CY administration could promote the percentage of splenic CD8+ T cells and decrease the proportion of CD4 + CD25 + Foxp3+ Tregs in spleen. In tumor tissues, levels of immunosuppressive cytokines including IL-10 and CXCL-12 were also reduced. Meanwhile, the CY combination did not impair the FAPα-specific immunity induced by the DNA vaccine and further reduced tumor stromal factors. Most importantly, FAP-vaccinated mice also treated with CY chemotherapy showed a marked suppression of tumor growth (inhibition ratio =80%) and a prolongation of survival time. Thus, the combination of FAPα immunotherapy and chemotherapy with CY offers new insights into improving cancer therapies.


Subject(s)
Cancer Vaccines/pharmacology , Cyclophosphamide/pharmacokinetics , Gelatinases/pharmacology , Immunity, Cellular/drug effects , Mammary Neoplasms, Experimental/therapy , Membrane Proteins/pharmacology , Serine Endopeptidases/pharmacology , Vaccines, DNA/pharmacokinetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/immunology , Endopeptidases , Female , Gelatinases/immunology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Serine Endopeptidases/immunology , Vaccines, DNA/immunology
SELECTION OF CITATIONS
SEARCH DETAIL