Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Viruses ; 16(4)2024 04 16.
Article in English | MEDLINE | ID: mdl-38675960

ABSTRACT

Reactivation and infection with cytomegalovirus (CMV) are frequently observed in recipients of solid organ transplants, bone marrow transplants, and individuals with HIV infection. This presents an increasing risk of allograft rejection, opportunistic infection, graft failure, and patient mortality. Among immunocompromised hosts, interstitial pneumonia is the most critical clinical manifestation of CMV infection. Recent studies have demonstrated the potential therapeutic benefits of exosomes derived from mesenchymal stem cells (MSC-exos) in preclinical models of acute lung injury, including pneumonia, ARDS, and sepsis. However, the role of MSC-exos in the pathogenesis of infectious viral diseases, such as CMV pneumonia, remains unclear. In a mouse model of murine CMV-induced pneumonia, we observed that intravenous administration of mouse MSC (mMSC)-exos reduced lung damage, decreased the hyperinflammatory response, and shifted macrophage polarization from the M1 to the M2 phenotype. Treatment with mMSC-exos also significantly reduced the infiltration of inflammatory cells and pulmonary fibrosis. Furthermore, in vitro studies revealed that mMSC-exos reversed the hyperinflammatory phenotype of bone marrow-derived macrophages infected with murine CMV. Mechanistically, mMSC-exos treatment decreased activation of the NF-κB/NLRP3 signaling pathway both in vivo and in vitro. In summary, our findings indicate that mMSC-exo treatment is effective in severe CMV pneumonia by reducing lung inflammation and fibrosis through the NF-κB/NLRP3 signaling pathway, thus providing promising therapeutic potential for clinical CMV infection.


Subject(s)
Disease Models, Animal , Exosomes , Mesenchymal Stem Cells , Muromegalovirus , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction , Animals , Exosomes/metabolism , Mesenchymal Stem Cells/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NF-kappa B/metabolism , Muromegalovirus/physiology , Mice, Inbred C57BL , Macrophages/immunology , Cytomegalovirus Infections/therapy , Cytomegalovirus Infections/virology , Lung/virology , Lung/pathology , Pneumonia, Viral/therapy , Pneumonia, Viral/virology , Herpesviridae Infections/therapy , Herpesviridae Infections/virology , Herpesviridae Infections/immunology , Pneumonia/therapy , Pneumonia/virology
3.
PLoS Pathog ; 19(4): e1011316, 2023 04.
Article in English | MEDLINE | ID: mdl-37058447

ABSTRACT

The presence of human cytomegalovirus (HCMV) in glioblastoma (GBM) and improved outcomes of GBM patients receiving therapies targeting the virus have implicated HCMV in GBM progression. However, a unifying mechanism that accounts for the contribution of HCMV to the malignant phenotype of GBM remains incompletely defined. Here we have identified SOX2, a marker of glioma stem cells (GSCs), as a key determinant of HCMV gene expression in gliomas. Our studies demonstrated that SOX2 downregulated promyelocytic leukemia (PML) and Sp100 and consequently facilitated viral gene expression by decreasing the amount of PML nuclear bodies in HCMV-infected glioma cells. Conversely, the expression of PML antagonized the effects of SOX2 on HCMV gene expression. Furthermore, this regulation of SOX2 on HCMV infection was demonstrated in a neurosphere assay of GSCs and in a murine xenograft model utilizing xenografts from patient-derived glioma tissue. In both cases, SOX2 overexpression facilitated the growth of neurospheres and xenografts implanted in immunodeficient mice. Lastly, the expression of SOX2 and HCMV immediate early 1 (IE1) protein could be correlated in tissues from glioma patients, and interestingly, elevated levels of SOX2 and IE1 were predictive of a worse clinical outcome. These studies argue that HCMV gene expression in gliomas is regulated by SOX2 through its regulation of PML expression and that targeting molecules in this SOX2-PML pathway could identify therapies for glioma treatment.


Subject(s)
Glioma , Immediate-Early Proteins , Animals , Humans , Mice , Cytomegalovirus/physiology , Down-Regulation , Gene Expression , Glioma/genetics , Glioma/pathology , Immediate-Early Proteins/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
J Virol ; 97(5): e0031323, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37097169

ABSTRACT

Human cytomegalovirus (HCMV) is a leading cause of congenital birth defects. Though the underlying mechanisms remain poorly characterized, mouse models of congenital CMV infection have demonstrated that the neuronal migration process is damaged. In this study, we evaluated the effects of HCMV infection on connexin 43 (Cx43), a crucial adhesion molecule mediating neuronal migration. We show in multiple cellular models that HCMV infection downregulated Cx43 posttranslationally. Further analysis identified the immediate early protein IE1 as the viral protein responsible for the reduction of Cx43. IE1 was found to bind the Cx43 C terminus and promote Cx43 degradation through the ubiquitin-proteasome pathway. Deletion of the Cx43-binding site in IE1 rendered it incapable of inducing Cx43 degradation. We validated the IE1-induced loss of Cx43 in vivo by introducing IE1 into the fetal mouse brain. Noteworthily, ectopic IE1 expression induced cortical atrophy and neuronal migration defects. Several lines of evidence suggest that these damages result from decreased Cx43, and restoration of Cx43 levels partially rescued IE1-induced interruption of neuronal migration. Taken together, the results of our investigation reveal a novel mechanism of HCMV-induced neural maldevelopment and identify a potential intervention target. IMPORTANCE Congenital CMV (cCMV) infection causes neurological sequelae in newborns. Recent studies of cCMV pathogenesis in animal models reveal ventriculomegaly and cortical atrophy associated with impaired neural progenitor cell (NPC) proliferation and migration. In this study, we investigated the mechanisms underlying these NPC abnormalities. We show that Cx43, a critical adhesion molecule mediating NPC migration, is downregulated by HCMV infection in vitro and HCMV-IE1 in vivo. We provide evidence that IE1 interacts with the C terminus of Cx43 to promote its ubiquitination and consequent degradation through the proteasome. Moreover, we demonstrate that introducing IE1 into mouse fetal brains led to neuronal migration defects, which was associated with Cx43 reduction. Deletion of the Cx43-binding region in IE1 or ectopic expression of Cx43 rescued the IE1-induced migration defects in vivo. Our study provides insight into how cCMV infection impairs neuronal migration and reveals a target for therapeutic interventions.


Subject(s)
Connexin 43 , Cytomegalovirus Infections , Cytomegalovirus , Immediate-Early Proteins , Animals , Humans , Infant, Newborn , Mice , Connexin 43/genetics , Connexin 43/metabolism , Cytomegalovirus/physiology , Cytomegalovirus Infections/metabolism , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism
5.
Esophagus ; 20(3): 502-514, 2023 07.
Article in English | MEDLINE | ID: mdl-36853485

ABSTRACT

BACKGROUND: Cyclin-dependent kinase 5 (CDK5) is a member of the cyclin-dependent kinase family, and unlike the rest of the members of the family, its kinase activity is independent of cyclins. Accumulating evidence has shown that CDK5 plays a significant role in the progress of tumorigenesis except in nervous system. In particular, the expression of CDK5 and its function in esophageal cancer (ESCA) remain unknown. METHODS: With TCGA and GEO databases, CDK5 was analyzed with the expression, predicted value, clinical relationship, functional enrichment, immune cell infiltration and immune molecules in ESCA. In addition, we explored the CDK5 expression with local datasets and the influence of CDK5 on proliferation, migration and invasion behaviors of the esophageal squamous cell carcinoma (ESCC) cells in vitro and in vivo experiments. RESULTS: CDK5 expression was upregulated in ESCA, and this regulation has been verified in cell lines of ESCC. Further analysis has found that the expression of CDK5 was correlated with race, weight, BMI, histological type and tumor central location in ESCA. KEGG analysis revealed that CDK5 was involved in the progress of cancers, innate immune system and PI3K-Akt signaling pathway. CDK5 was closely related to immune cells and immune molecules in ESCA. Functional experiments confirmed CDK5 was an oncogene in ESCC by in vivo and in vitro models. CONCLUSIONS: This study shows that CDK5 is a risk factor to promote tumor progression, and Roscovitine could be one of the effective tools in the therapy of ESCA.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Phosphatidylinositol 3-Kinases , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Biomarkers
6.
PLoS Pathog ; 19(2): e1011166, 2023 02.
Article in English | MEDLINE | ID: mdl-36753521

ABSTRACT

Congenital human cytomegalovirus (HCMV) infection causes severe damage to the fetal brain, and the underlying mechanisms remain elusive. Cytokine signaling is delicately controlled in the fetal central nervous system to ensure proper development. Here we show that suppressor of cytokine signaling 3 (SOCS3), a negative feedback regulator of the IL-6 cytokine family signaling, was upregulated during HCMV infection in primary neural progenitor cells (NPCs) with a biphasic expression pattern. From viral protein screening, pUL97 emerged as the viral factor responsible for prolonged SOCS3 upregulation. Further, by proteomic analysis of the pUL97-interacting host proteins, regulatory factor X 7 (RFX7) was identified as the transcription factor responsible for the regulation. Depletion of either pUL97 or RFX7 prevented the HCMV-induced SOCS3 upregulation in NPCs. With a promoter-luciferase activity assay, we demonstrated that the pUL97 kinase activity and RFX7 were required for SOCS3 upregulation. Moreover, the RFX7 phosphorylation level was increased by either UL97-expressing or HCMV-infection in NPCs, suggesting that pUL97 induces RFX7 phosphorylation to drive SOCS3 transcription. We further revealed that elevated SOCS3 expression impaired NPC proliferation and migration in vitro and caused NPCs migration defects in vivo. Taken together, these findings uncover a novel regulatory mechanism of sustained SOCS3 expression in HCMV-infected NPCs, which perturbs IL-6 cytokine family signaling, leads to NPCs proliferation and migration defects, and consequently affects fetal brain development.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Humans , Cytomegalovirus/physiology , Interleukin-6/metabolism , Proteomics , Transcription Factors/metabolism , Stem Cells , Suppressor of Cytokine Signaling 3 Protein/metabolism
7.
J Med Virol ; 94(11): 5492-5506, 2022 11.
Article in English | MEDLINE | ID: mdl-35879101

ABSTRACT

During the long coevolution of human cytomegalovirus (HCMV) and humans, the host has formed a defense system of multiple layers to eradicate the invader, and the virus has developed various strategies to evade host surveillance programs. The intrinsic immunity primarily orchestrated by promyelocytic leukemia (PML) nuclear bodies (PML-NBs) represents the first line of defense against HCMV infection. Here, we demonstrate that microrchidia family CW-type zinc finger 3 (MORC3), a PML-NBs component, is a restriction factor targeting HCMV infection. We show that depletion of MORC3 through knockdown by RNA interference or knockout by CRISPR-Cas9 augmented immediate-early protein 1 (IE1) gene expression and subsequent viral replication, and overexpressing MORC3 inhibited HCMV replication by suppressing IE1 gene expression. To relief the restriction, HCMV induces transient reduction of MORC3 protein level via the ubiquitin-proteasome pathway during the immediate-early to early stage. However, MORC3 transcription is upregulated, and the protein level recovers in the late stages. Further analyses with temporal-controlled MORC3 expression and the major immediate-early promoter (MIEP)-based reporters show that MORC3 suppresses MIEP activity and consequent IE1 expression with the assistance of PML. Taken together, our data reveal that HCMV enforces temporary loss of MORC3 to evade its repression against the initiation of immediate-early gene expression.


Subject(s)
Cytomegalovirus Infections , Immediate-Early Proteins , Adenosine Triphosphatases/metabolism , Cytomegalovirus/genetics , DNA-Binding Proteins/metabolism , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/metabolism , Virus Replication
8.
Discov Oncol ; 13(1): 58, 2022 Jul 07.
Article in English | MEDLINE | ID: mdl-35798917

ABSTRACT

Acetyl-CoA synthetase 2 (ACSS2), an important member of the acetyl-CoA synthetase (ACSS) family, can catalyze the conversion of acetate to acetyl coenzyme A (acetyl-CoA). Currently, acetyl-CoA is considered an important intermediate metabolite in the metabolism of energy substrates. In addition, nutrients converge through acetyl-CoA into a common metabolic pathway, the tricarboxylic acid cycle and oxidative phosphorylation. Not only does ACSS2 play a crucial role in material energy metabolism, it is also involved in the regulation of various acetylation processes, such as regulation of histone and transcription factor acetylation. ACSS2-mediated regulation of acetylation is related to substance metabolism and tumorigenesis. In mammalian cells, ACSS2 utilizes intracellular acetate to synthesize acetyl-CoA, a step in the process of DNA and histone acetylation. In addition, studies in tumors have shown that cancer cells adapt to the growth conditions in the tumor microenvironment (TME) by activating or increasing the expression level of ACSS2 under metabolic stress. Therefore, this review mainly outlines the role of ACSS2 in substance metabolism and tumors and provides insights useful for investigating ACSS2 as a therapeutic target.

9.
Microbiol Spectr ; 10(3): e0186421, 2022 06 29.
Article in English | MEDLINE | ID: mdl-35467404

ABSTRACT

Autism spectrum disorder (ASD), a highly hereditary and heterogeneous neurodevelopmental disorder, is influenced by genetic and environmental factors. Tuberous sclerosis complex (TSC) is a common syndrome associated with ASD. Cytomegalovirus (CMV) infection is an environmental risk factor for ASD. The similarities in pathological and mechanistic pathways of TSC and CMV intrigued us to investigate whether CMV and TSC interacted in ASD's occurrence. We detected CMV IgG seroprevalence of 308 TSC patients from our prospective cohort (September 2011 to March 2021) and 93 healthy children by magnetic particle indirect chemiluminescence immunoassay. A total of 206 TSC patients enrolled were divided into ASD and non-ASD groups, and the relationship between ASD and CMV seroprevalence was analyzed. Nested PCR and Western blot were used to detect CMV DNAs and proteins in cortical malformations of seven TSC patients with and without ASD. No difference was found in CMV seroprevalence between TSC patients and healthy children (74.0% versus 72.0%, P = 0.704). Univariate analysis showed the seroprevalence in TSC patients with ASD was higher than that in TSC patients without ASD (89.2% versus 75.1%, P = 0.063), and multifactorial analysis showed that CMV seroprevalence was a risk factor for ASD in TSC patients (OR = 3.976, 95% CI = 1.093 to 14.454). Moreover, CMV was more likely to be detected in the cortical malformations in TSC patients with ASD but not in those without ASD. The findings demonstrated that CMV may increase the susceptibility of TSC to ASD. IMPORTANCE CMV is an environmental risk factor for ASD, but its role in syndromic autism with known genetic etiology has been rarely studied. The pathogenesis of ASD is related to the interaction between environmental and genetic factors. This study demonstrated that CMV can contribute to the occurrence of ASD related to TSC, a common genetic syndrome associated with ASD. Our findings provided support for the theory of gene-environment interaction (G × E) in pathogenesis of ASD and a new perspective for the prevention and therapy for TSC related ASD.


Subject(s)
Autism Spectrum Disorder , Cytomegalovirus Infections , Tuberous Sclerosis , Autism Spectrum Disorder/complications , Autism Spectrum Disorder/etiology , Child , Cytomegalovirus/genetics , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/epidemiology , Humans , Prospective Studies , Seroepidemiologic Studies , Tuberous Sclerosis/complications , Tuberous Sclerosis/epidemiology , Tuberous Sclerosis/genetics
10.
Front Oncol ; 12: 847701, 2022.
Article in English | MEDLINE | ID: mdl-35402244

ABSTRACT

Bromodomain-containing protein 4 (BRD4), a member of the bromodomain and extraterminal (BET) family, is considered to be a major driver of cancer cell growth and a new target for cancer therapy. Over 30 targeted inhibitors currently in preclinical and clinical trials have significant inhibitory effects on various tumors, including acute myelogenous leukemia (AML), diffuse large B cell lymphoma, prostate cancer, breast cancer and so on. However, resistance frequently occurs, revealing the limitations of BET inhibitor (BETi) therapy and the complexity of the BRD4 expression mechanism and action pathway. Current studies believe that when the internal and external environmental conditions of cells change, tumor cells can directly modify proteins by posttranslational modifications (PTMs) without changing the original DNA sequence to change their functions, and epigenetic modifications can also be activated to form new heritable phenotypes in response to various environmental stresses. In fact, research is constantly being supplemented with regards to that the regulatory role of BRD4 in tumors is closely related to PTMs. At present, the PTMs of BRD4 mainly include ubiquitination and phosphorylation; the former mainly regulates the stability of the BRD4 protein and mediates BETi resistance, while the latter is related to the biological functions of BRD4, such as transcriptional regulation, cofactor recruitment, chromatin binding and so on. At the same time, other PTMs, such as hydroxylation, acetylation and methylation, also play various roles in BRD4 regulation. The diversity, complexity and reversibility of posttranslational modifications affect the structure, stability and biological function of the BRD4 protein and participate in the occurrence and development of tumors by regulating the expression of tumor-related genes and even become the core and undeniable mechanism. Therefore, targeting BRD4-related modification sites or enzymes may be an effective strategy for cancer prevention and treatment. This review summarizes the role of different BRD4 modification types, elucidates the pathogenesis in the corresponding cancers, provides a theoretical reference for identifying new targets and effective combination therapy strategies, and discusses the opportunities, barriers, and limitations of PTM-based therapies for future cancer treatment.

11.
J Virol ; 96(5): e0182721, 2022 03 09.
Article in English | MEDLINE | ID: mdl-35020472

ABSTRACT

Human cytomegalovirus (HCMV) has a large (∼235 kb) genome with more than 200 predicted open reading frames that exploits numerous cellular factors to facilitate its replication. A key feature of HCMV-infected cells is the emergence of a distinctive membranous cytoplasmic compartment termed the virion assembly compartment (vAC). Here, we report that host protein WD repeat domain 11 (WDR11) plays a key role in vAC formation and virion morphogenesis. We found that WDR11 was upregulated at both mRNA and protein levels during HCMV infection. At the late stage of HCMV replication, WDR11 relocated to the vAC and colocalized with markers of the trans-Golgi network (TGN) and vAC. Depletion of WDR11 hindered HCMV-induced membrane reorganization of the Golgi and TGN, altered vAC formation, and impaired HCMV secondary envelopment and virion morphogenesis. Further, motifs critical for the localization of WDR11 in TGN were identified by alanine-scanning mutagenesis. Mutation of these motifs led to WDR11 mislocation outside the TGN and loss of vAC formation. Taken together, these data indicate that host protein WDR11 is required for efficient viral replication at the stage of virion assembly, possibly by facilitating the remodeling of the endomembrane system for vAC formation and virion morphogenesis. IMPORTANCE During the late phase of human cytomegalovirus (HCMV) infection, the endomembrane system is dramatically reorganized, resulting in the formation of a unique structure termed the virion assembly compartment (vAC), which is critical for the assembly of infectious virions. The mechanism of HCMV-induced vAC formation is still not fully understood. In this report, we identified a host factor, WDR11, that plays an important role in vAC formation. Our findings argue that WDR11 contributes to the relocation of the Golgi and trans-Golgi network to the vAC, a membrane reorganization process that appears to be required for efficient virion maturation. The present work provides new insights into the vAC formation and HCMV virion morphogenesis and a potential novel target for antiviral treatment.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Host Microbial Interactions , WD40 Repeats , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cytomegalovirus Infections/physiopathology , Cytomegalovirus Infections/virology , Humans , Morphogenesis , Virion/metabolism , Virus Assembly/genetics , Virus Replication/genetics , WD40 Repeats/genetics , trans-Golgi Network/metabolism
12.
JCI Insight ; 7(1)2022 01 11.
Article in English | MEDLINE | ID: mdl-35014624

ABSTRACT

Congenital cytomegalovirus (cCMV) infection is the leading infectious cause of neurodevelopmental disorders. However, the neuropathogenesis remains largely elusive due to a lack of informative animal models. In this study, we developed a congenital murine CMV (cMCMV) infection mouse model with high survival rate and long survival period that allowed long-term follow-up study of neurodevelopmental disorders. This model involves in utero intracranial injection and mimics many reported clinical manifestations of cCMV infection in infants, including growth restriction, hearing loss, and impaired cognitive and learning-memory abilities. We observed that abnormalities in MRI/CT neuroimaging were consistent with brain hemorrhage and loss of brain parenchyma, which was confirmed by pathological analysis. Neuropathological findings included ventriculomegaly and cortical atrophy associated with impaired proliferation and migration of neural progenitor cells in the developing brain at both embryonic and postnatal stages. Robust inflammatory responses during infection were shown by elevated inflammatory cytokine levels, leukocyte infiltration, and activation of microglia and astrocytes in the brain. Pathological analyses and CT neuroimaging revealed brain calcifications induced by cMCMV infection and cell death via pyroptosis. Furthermore, antiviral treatment with ganciclovir significantly improved neurological functions and mitigated brain damage as shown by CT neuroimaging. These results demonstrate that this model is suitable for investigation of mechanisms of infection-induced brain damage and long-term studies of neurodevelopmental disorders, including the development of interventions to limit CNS damage associated with cCMV infection.


Subject(s)
Cytomegalovirus Infections , Disease Models, Animal , Neuroimaging , Animals , Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/diagnostic imaging , Cytomegalovirus Infections/physiopathology , Cytomegalovirus Infections/therapy , Female , Follow-Up Studies , Mice , Mice, Inbred ICR , Pregnancy
13.
Ocul Immunol Inflamm ; 30(4): 809-820, 2022 May 19.
Article in English | MEDLINE | ID: mdl-33226275

ABSTRACT

PURPOSES: To understand the pathogenesis in rat corneal endothelial cells (RCECs) induced by murine cytomegalovirus infection in vitro and in vivo. METHODS: In vitro, cultured RCECs were infected with murine cytomegalovirus strain K181-eGFP (MCMV-eGFP). In vivo, experimental rats received intracameral injection of MCMV-eGFP. Replicating viruses and morphology change of RCECs in vivo were evaluated at several time points. RESULTS: In vitro, RCECs became necrosis at 6hpi. MCMV-eGFP began replicating at 12hpi. In vivo, the inflammatory reactions appeared at 12hpi, peaked at 72hpi and gradually subsided. Replicating MCMV-eGFP appeared in RCECs in vivo from 24hpi to 72hpi. RCECs enlarged after 12hpi and capsids in the nuclei were visible at 72hpi. A monocyte was found on a corneal endothelium at 120hpi. CONCLUSIONS: RCECs were sensitive to MCMV in vitro. Replication of MCMV-eGFP in vivo began at 24hpi and ended after 72hpi, later than the inflammatory reactions.


Subject(s)
Cytomegalovirus Infections , Muromegalovirus , Animals , Endothelial Cells , Endothelium, Corneal , Epithelial Cells , Mice , Rats
14.
BMC Cancer ; 21(1): 877, 2021 Jul 31.
Article in English | MEDLINE | ID: mdl-34332557

ABSTRACT

BACKGROUND: Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs), such as gefitinib, have achieved good efficacy in EGFR mutation-positive non-small-cell lung cancer (NSCLC) patients, but eventual drug resistance is inevitable. Thus, new TKI-based combination therapies should be urgently explored to extend the overall survival time of these patients. CD8 + CD56+ natural killer T (NKT) cells are a natural and unique subset of lymphocytes in humans that present characteristics of T and NK cells and exert cytotoxicity on tumour cells in a granzyme B-dependent manner. The aim of this trial was to explore the efficacy and safety of CD8 + CD56+ NKT cell immunotherapy combined with gefitinib in patients with advanced EGFR-mutated NSCLC. METHODS: The study was designed as a prospective, randomized, controlled, open-label, phase I/II trial that includes 30 patients with EGFR mutation-positive stage III/IV NSCLC. All patients will be randomized in blocks at a 1:1 ratio and treated with gefitinib 250 mg/day monotherapy or combination therapy with allogeneic CD8 + CD56+ NKT cell infusions twice per month for 12 cycles or until disease progression occurs. The effectiveness of this treatment will be evaluated based on by progression-free survival (PFS), the time to progression (TTP), overall response rate (ORR), disease control rate (DCR) and overall survival (OS). The safety of the trail is being assessed based on adverse events (AEs). Recruitment and data collection, which started in December 2017, are ongoing. DISCUSSION: Although immunotherapy, including programmed death-1/programmed death-1 ligand (PD-1/PD-L1) immunotherapy, has been used for NSCLC treatment with or without EGFR-TKIs, its clear efficacy still has not been shown. Assessing the safety and therapeutic potential of allogeneic CD8 + CD56+ NKT killer cells in combination with EGFR-TKIs in NSCLC will be of great interest. TRIAL REGISTRATION: This trial (Phase I/II Trails of NKT Cell in Combination With Gefitinib For Non Small Cell Lung Cancer) was registered on 21 November 2017 with www.chictr.org.cn , ChiCTR-IIR-17013471 .


Subject(s)
Adoptive Transfer , Carcinoma, Non-Small-Cell Lung/therapy , Gefitinib/therapeutic use , Lung Neoplasms/therapy , Mutation , Natural Killer T-Cells/immunology , Adoptive Transfer/adverse effects , Adoptive Transfer/methods , B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/etiology , Combined Modality Therapy , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Gefitinib/administration & dosage , Gefitinib/adverse effects , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/etiology , Molecular Targeted Therapy , Natural Killer T-Cells/metabolism , Treatment Outcome
15.
Oncol Lett ; 22(1): 503, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33986864

ABSTRACT

Radiotherapy is an effective therapeutic strategy in esophageal squamous cell carcinoma (ESCC). However, acquired radioresistance of cancer cells leads to radiotherapy failure. The present study aimed to investigate the mechanisms of the effect of high mobility group box 1 (HMGB1) on the radiation sensitivity of ESCC. Small interfering RNA (si) transfection was used to generate three groups of TE-1 cells (TE-1, negative control and TE-1+siHMGB1), and the protein expression levels of HMGB1 in TE-1 cells were detected by western blotting. These groups of TE-1 cells were irradiated with different doses (0, 2, 4, 6 and 8 Gy) of X-rays after transfection. Subsequently, the viability of TE-1 cells was detected using an MTT assay, and the survival fraction of TE-1 cells was observed using a colony formation assay. The apoptotic rate, reactive oxygen species (ROS) content and levels of phosphorylated (p)-histone H2AX at S139 (p-γH2AX) of the cells were detected by flow cytometry. The alterations in mRNA expression levels of nicotinamide adenine nucleotide phosphate oxidase (NOX)1 and NOX5 were detected by reverse transcription-quantitative PCR, while the changes in protein levels of caspase-3, poly(ADP-ribose) polymerase, p-p38, p-ERK1/2 and p-JNK were detected by western blotting. The results revealed that HMGB1 knockdown significantly decreased cell viability, and the apoptosis rate of TE-1 cells transfected with siHMGB1 combined with radiation treatment was increased compared with that in cells with either siHMGB1 transfection or radiation treatment alone. HMGB1 knockdown increased nicotinamide adenine nucleotide phosphate oxidase-mediated ROS production and induced DNA damage via the MAPK signaling pathway, which may promote apoptosis and radiosensitivity after radiation in TE-1 cells. In conclusion, targeting HMGB1 may represent a promising strategy to increase the efficacy of radiation therapy for ESCC.

16.
Viruses ; 13(4)2021 04 06.
Article in English | MEDLINE | ID: mdl-33917368

ABSTRACT

Hearing loss is one of the most prevalent sensory disabilities worldwide with huge social and economic burdens. The leading cause of sensorineural hearing loss (SNHL) in children is congenital cytomegalovirus (CMV) infection. Though the implementation of universal screening and early intervention such as antiviral or anti-inflammatory ameliorate the severity of CMV-associated diseases, direct and targeted therapeutics is still seriously lacking. The major hurdle for it is that the mechanism of CMV induced SNHL has not yet been well understood. In this review, we focus on the impact of CMV infection on the key players in inner ear development including the Wnt and Notch signaling pathways. Investigations on these interactions may gain new insights into viral pathogenesis and reveal novel targets for therapy.


Subject(s)
Cytomegalovirus Infections/complications , Cytomegalovirus/pathogenicity , Gene Expression Regulation , Hearing Loss, Sensorineural/virology , Receptors, Notch/genetics , Signal Transduction , Wnt Proteins/genetics , Animals , Cytomegalovirus/genetics , Humans , Mice , Receptors, Notch/metabolism , Wnt Proteins/metabolism
17.
Oncol Lett ; 20(3): 2855-2869, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32765792

ABSTRACT

Sterol regulatory element-binding protein 1 (SREBP1) is dysregulated in a variety of types of human cancer. However, the functional roles of SREBP1 in esophageal squamous cell carcinoma (ESCC) remain poorly understood. The present study investigated the function of SREBP1 in cell proliferation and motility. Microarray datasets in Oncomine, reverse transcription-quantitative PCR and western blot analysis revealed that SREBP1 was overexpressed in ESCC tumors when compared with normal tissues. In addition, SREBP1 overexpression was significantly associated with tumor differentiation, lymphatic metastasis and Ki67 expression. Results suggested that silencing SREBP1 inhibited the proliferation, migration and invasion of ESCC cells, whereas overexpression of SREBP1 had opposite effects on proliferation and metastasis. In addition, loss of SREBP1 significantly increased E-cadherin and decreased N-cadherin, Vimentin, Snail, matrix metalloproteinase 9 and vascular endothelial growth factor C expression levels, which were restored via SREBP1-overexpression. Mechanistically, loss of SREBP1 suppressed T-cell factor 1/lymphoid enhancer factor 1 (TCF1/LEF1) activity and downregulated TCF1/LEF1 target proteins, including CD44 and cyclin D1. Moreover, knockdown of SREBP1 downregulated the expression levels of stearoyl-CoA desaturase 1 (SCD1), phosphorylated glycogen synthase kinase-3ß and nuclear ß-catenin. Furthermore, the inhibitors of SREBP1 and/or SCD1 and small interfering RNA-SCD1 efficiently inhibited the activation of the Wnt/ß-catenin pathway driven by constitutively active SREBP1. Finally, in vivo results indicated that SREBP1-knockdown suppressed the proliferation and metastasis of ESCC. Taken together, these findings demonstrated that SREBP1 exerts oncogenic effects in ESCC by promoting proliferation and inducing epithelial-mesenchymal transition via the SCD1-induced activation of the Wnt/ß-catenin signaling pathway.

18.
J Cell Mol Med ; 24(13): 7460-7469, 2020 07.
Article in English | MEDLINE | ID: mdl-32436312

ABSTRACT

Peripheral nerve injury and regeneration are complex processes and involve multiple molecular and signalling components. However, the involvement of long non-coding RNA (lncRNA) in this process is not fully clarified. In this study, we evaluated the expression of the lncRNA maternally expressed gene 3 (MEG3) in rats after sciatic nerve transection and explored its potential mechanisms. The expression of lncRNA MEG3 was up-regulated following sciatic nerve injury and observed in Schwann cells (SCs). The down-regulation of lncRNA MEG3 in SCs enhanced the proliferation and migration of SCs via the PTEN/PI3K/AKT pathway. The silencing of lncRNA MEG3 promoted the migration of SCs and axon outgrowth in rats after sciatic nerve transection and facilitated rat nerve regeneration and functional recovery. Our findings indicated that lncRNA MEG3 may be involved in nerve injury and injured nerve regeneration in rats with sciatic nerve defects by regulating the proliferation and migration of SCs. This gene may provide a potential therapeutic target for improving peripheral nerve injury.


Subject(s)
Cell Movement/genetics , Down-Regulation/genetics , Nerve Regeneration/genetics , RNA, Long Noncoding/metabolism , Schwann Cells/pathology , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Animals , Axons/metabolism , Cell Proliferation/genetics , Male , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Transport/genetics , RNA, Long Noncoding/genetics , Rats, Sprague-Dawley , Recovery of Function , Schwann Cells/metabolism , Signal Transduction , Up-Regulation/genetics
19.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 35(10): 926-931, 2019 Oct.
Article in Chinese | MEDLINE | ID: mdl-31814569

ABSTRACT

Objective To investigate the effect of acetyl coenzyme A synthase 2 (ACSS2) on the migration and invasion induced by nutrient stress (NS) in cervical cancer cells. Methods Immunohistochemistry was used to detect the expression and distribution of ACSS2 protein in 20 pairs of cervical tumors and their adjacent normal tissues. Cervical cancer HeLa cells and the normal cervical epithelial HCvEpC cells were cultured, and serum content in culture medium was reduced from 100 to 10 mL/L as NS treatment. Western blot analysis was performed to detect the expression of ACSS2, GSK-3ß, p-GSK-3ß, ß-catenin, ß-actin, E-cadherin, vimentin. Small interfering RNA (siRNA) was used to down-regulate the expression of ACSS2. Cell migration was assessed by wound healing test, and cell invasion was tested by TranswellTM assay. Results The expression level of ACSS2 in 20 cervical tumors was significantly higher as compared with the adjacent normal tissues. The levels of ACSS2 in HeLa cells could be significantly up-regulated by NS, while no marked change was seen in HCvEpC cells. The treatment of NS promoted the epithelial mesenchymal transformation (EMT) of HeLa cells, which could be effectively reversed by siRNA-ACSS2. The scratch results showed that NS increased the healing rate of HeLa cells, which could be blocked by ACSS2 silencing. Coincidently, the number of invasive cells was elevated after NS treatment, which could be partly reversed by siRNA-ACSS2. The expression of ACSS2, p-GSK-3ß and nuclear ß-catenin was up-regulated in HeLa cells treated with NS for 48 hours, while siRNA-ACSS2 down-regulated their expression. Conclusion Silencing ACSS2 expression inhibits migration and invasion of cervical cancer cells induced by NS, which is related to down-regulated Wnt/ß-catenin signaling pathway activity.


Subject(s)
Acetate-CoA Ligase/genetics , Gene Silencing , Uterine Cervical Neoplasms/pathology , Wnt Signaling Pathway , Cell Movement , Cell Proliferation , Culture Media , Epithelial-Mesenchymal Transition , Female , Gene Knockdown Techniques , HeLa Cells , Humans , Neoplasm Invasiveness , Uterine Cervical Neoplasms/genetics
20.
Mol Med Rep ; 20(6): 5286-5296, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31638228

ABSTRACT

Although platinum­based chemotherapy is the first­line choice for locally advanced or metastatic esophageal squamous cell carcinoma (ESCC) patients, accelerated recurrence and chemoresistance remain inevitable. New evidence suggests that metabolism reprogramming under stress involves independent processes that are executed with a variety of proteins. This study investigated the functions of nutrient stress (NS)­mediated acetyl­CoA synthetase short­chain family member 2 (ACSS2) in cell proliferation and cisplatin­resistance and examined its combined effects with proliferating cell nuclear antigen (PCNA), a key regulator of DNA replication and repair. Here, it was demonstrated that under NS, when the AMP­activated protein kinase (AMPK) pathway was activated, ESCC cells maintained proliferation and chemoresistance was distinctly upregulated as determined by CCK­8 assay. As determined using immunoblotting and RT­qPCR, compared with normal esophageal epithelial cells (Het­1A), ESCC cells were less sensitive to NS and showed increased intracellular levels of ACSS2. Moreover, it was shown that ACSS2 inhibition by siRNA not only greatly interfered with proliferation under NS but also participated in DNA repair after cisplatin treatment via PCNA suppression, and the acceleration of cell death was dependent on the activation of the AMPK pathway as revealed by the Annexin V/PI and TUNEL assay results. Our study identified crosstalk between nutrient supply and chemoresistance that could be exploited therapeutically to target AMPK signaling, and the results suggest ACSS2 as a potential biomarker for identifying higher­risk patients.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Acetate-CoA Ligase/metabolism , Drug Resistance, Neoplasm , Nutrients , Proliferating Cell Nuclear Antigen/metabolism , Signal Transduction , Stress, Physiological , Cell Line, Tumor , Cell Proliferation , DNA Damage , DNA Repair , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...