Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cytotherapy ; 23(8): 715-723, 2021 08.
Article in English | MEDLINE | ID: mdl-33863641

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy is a promising therapeutic strategy against lymphoma. However, post-treatment relapses due to antigen loss remain a challenge. Here the authors designed a novel bicistronic CAR construct and tested its functions in vitro and in vivo. The CAR construct consisted of individual anti-CD19 and anti-CD20 single-chain fragment variables equipped with ICOS-CD3ζ and 4-1BB-CD3ζ intracellular domains, respectively. The CD19 and CD20 bicistronic CAR T cells exhibited tumor lytic capacities equivalent to corresponding monospecific CAR T cells. Moreover, when stimulated with CD19 and CD20 simultaneously, the bicistronic CAR T cells showed prolonged persistence and enhanced cytokine generation compared with single stimulations. Interestingly, the authors found that the 4-1BB signal was predominant in the signaling profiles of ICOS and 4-1BB doubly activated CAR T cells. In vivo study using a CD19/CD20 double-positive tumor model revealed that the bicistronic CAR T cells were more efficient than monospecific CD19 CAR T cells in eradicating tumors and prolonging mouse survival. The authors' novel bicistronic CD19/CD20 CAR T cells demonstrate improved anti-tumor efficacy in response to dual antigen stimulations. These data provide optimism that this novel bicistronic CAR construct can improve treatment outcomes in patients with relapsed/refractory B cell malignancy.


Subject(s)
Receptors, Chimeric Antigen , Animals , Antigens, CD19/genetics , Humans , Immunotherapy, Adoptive , Inducible T-Cell Co-Stimulator Protein/genetics , Mice , Neoplasm Recurrence, Local , Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen/genetics , T-Lymphocytes
2.
Sci Rep ; 9(1): 12221, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31434993

ABSTRACT

Recent studies have shown that transforming microglia phenotype from pro-inflammation of M1 phenotype to anti-inflammation and tissue-repairing M2 phenotype may be an effective therapeutic strategy for preventing ischemic stroke brain injury. Isosteviol Sodium (STV-Na) has shown promise as a neuroprotective agent in cerebral ischemia model, although its effect on microglial polarization and chronic recovery after stroke is not clear. Here, we demonstrated that STV-Na treatment significantly reduced cerebral ischemic damage at both acute and chronic time points. STV-Na has a profound regulatory effect on microglia response after stroke. It can promote M2 polarization and inhibit microglia-mediated inflammation (M1) response following stroke in vivo and in vitro. Furthermore, we also found that Growth Arrest-Specific 5 (GAS5) altered OGD/R-induced microglial activation by increasing Notch1 expression via miR-146a-5p, the mRNA level of GAS5 and the protein level of Notch1 in vivo and in vitro, were discovered that both downgraded with STV-Na. Taken together, the present study demonstrated that STV-Na exerted neuroprotective effects by modulating microglia/macrophage polarization in ischemic stroke via the GAS5/miR-146a-5p sponge. These findings provide new evidence that targeting STV-Na could be a treatment for the prevention of stroke-related brain damage.


Subject(s)
Brain Ischemia , Diterpenes, Kaurane/pharmacology , Macrophages , MicroRNAs/metabolism , Microglia , RNA, Long Noncoding/metabolism , Stroke , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/prevention & control , Gene Expression Regulation , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Microglia/metabolism , Microglia/pathology , Receptor, Notch1/biosynthesis , Stroke/metabolism , Stroke/pathology , Stroke/prevention & control
3.
J Stroke Cerebrovasc Dis ; 28(1): 175-184, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30539754

ABSTRACT

BACKGROUND: Stevioside, isolated from the herb Stevia rebaudiana, has been widely used as a food sweetener all over the world. Isosteviol Sodium (STV-Na), an injectable formulation of isosteviol sodium salt, has been proved to possess much greater solubility and bioavailability and exhibit protective effects against cerebral ischemia injury in vivo by inhibiting neuron apoptosis. However, the underlying mechanisms of the neuroprotective effects STV-Na are still not completely known. In the present study, we investigated the effects of STV-Na on neuronal cell death caused by hypoxia in vitro and its underlying mechanisms. METHODS: We used cobalt chloride (CoCl2) to expose mouse neuroblastoma N2a cells to hypoxic conditions in vitro. RESULTS: Our results showed that pretreatment with STV-Na (20 µM) significantly attenuated the decrease of cell viability, lactate dehydrogenase release and cell apoptosis under conditions of CoCl2-induced hypoxia. Meanwhile, STV-Na pretreatment significantly attenuated the upregulation of intracellular Ca2+ concentration and reactive oxygen species production, and inhibited mitochondrial depolarization in N2a cells under conditions of CoCl2-induced hypoxia. Furthermore, STV-Na pretreatment significantly downregulated expressions of nitric oxide synthase, interleukin-1ß, tumor necrosis factor-α, interleukin-6, nuclear factor kappa B (NF-κB), and mitogen-activated protein kinase (MAPK) signalings in N2a cells under conditions of CoCl2-induced hypoxia. CONCLUSIONS: Taken together, STV-Na protects neural cells against hypoxia-induced apoptosis through inhibiting MAPK and NF-κB pathways.


Subject(s)
Apoptosis/drug effects , Cell Hypoxia/drug effects , Diterpenes, Kaurane/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Apoptosis/physiology , Calcium/metabolism , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cobalt/toxicity , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , NF-kappa B/metabolism , Neurons/metabolism , Neurons/pathology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
4.
Brain Res Bull ; 140: 392-401, 2018 06.
Article in English | MEDLINE | ID: mdl-29807144

ABSTRACT

NF-κB signaling pathway plays a critical role in cerebral ischemic stroke. MicroRNA-181b (miR-181b) induces the expression of NF-κB signaling pathways indirectly, and isosteviol sodium (STVNa) protects against ischemic injury via the inhibition of NF-κB-mediated inflammatory and apoptotic responses. However, the function of miR-181b and the actual relationship between STVNa and miR-181b in the ischemia-induced activation of NF-κB signaling pathways remains unclear. In this study, we found that miR-181b expression was significantly decreased in N2A neuroblastoma cells after CoCl2-induced hypoxic injury in vitro. We further found, via western blot analysis and quantitative polymerase chain reaction assay, that altering miR-181b expression could induce changes in one of its target proteins, cylindromatosis (CYLD). Specifically, upregulation and downregulation of miR-181b (through transfection of either pre- or small interfering miR-181b, respectively) could negatively regulate CYLD protein levels as well as N2A cell survival rate and apoptosis following CoCl2-induced injury. Furthermore, STVNa treatment following ischemic injury significantly downregulated the expression of miR-181b to alter apoptotic proteins downstream of the NF-κB signaling pathway through increasing CYLD protein levels in vivo and in vitro. STVNa also had a protective effect on CoCl2-injured N2A cells, increasing cell survival rate, inhibiting apoptosis, reducing the damage of mitochondrial membrane potential (MMP), and the generation of reactive oxygen species (ROS). Together, these results suggest that STVNa may downregulate miRNA-181b to protect mouse brain with ischemia stroke and against hypoxic injury in N2A cells by repressing NF-κB signaling pathways through the activation of CYLD, providing a novel therapy for ischemic stroke.


Subject(s)
Brain Ischemia/drug therapy , Diterpenes, Kaurane/pharmacology , MicroRNAs/metabolism , Neuroprotective Agents/pharmacology , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Brain Ischemia/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Line, Tumor , Cobalt , Down-Regulation/drug effects , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , NF-kappa B/metabolism
5.
Neuroreport ; 29(10): 794-803, 2018 07 04.
Article in English | MEDLINE | ID: mdl-29683870

ABSTRACT

Previous studies have shown that isosteviol sodium (STVNa) protects against permanent cerebral ischemia injury by inhibition of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)-mediated inflammatory responses. Overwhelming evidence shows that toll-like receptors (TLRs) are the upstream regulators of NF-κB. On the basis of the similarity of the pathology caused by traumatic brain injury (TBI) and stroke, we speculated that STVNa may have a therapeutic effect against TBI through regulation of the TLRs/NF-κB signaling-mediated inflammatory response. Thus, we studied the potential therapeutic effects of STVNa and the underlying mechanisms. Male rats, subjected to controlled cortical impact (CCI) injury, were injected intraperitoneally with STVNa (5, 10, 20, 40, and 80 mg/kg, daily for 3 or 7 days) after trauma. Neurobehavioral scores, relative numbers of cortical lesions, and histology were examined. We also measured the mRNA and protein expression levels of TLRs/NF-κB signaling pathway-related genes including TLR2, TLR4, and NF-κB by quantitative real-time-PCR and western blotting, respectively, and concentrations of tumor necrosis factor-α and interleukin-1ß by an enzyme-linked immunosorbent assay. The results indicated that STVNa (20 mg/kg) showed significant neuroprotective effects 3 and 7 days after TBI, including the reduction of cortical lesions, improvement of the neurological severity score, significantly increased number of restored neurons, decreased number of astrocytes, and lower concentrations of tumor necrosis factor-α and interleukin-1ß. Results from quantitative real-time-PCR and western blotting also show that the mRNA and protein expression levels of TLR2, TLR4, and NF-κB were significantly lower in STVNa-treated rats compared with the vehicle-treated rats. The administration of STVNa attenuates the TLR/NF-κB signaling pathway-mediated inflammatory responses in the injured rat brain, and this may be the mechanism by which STVNa improves the outcome following TBI.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Diterpenes, Kaurane/pharmacology , Inflammation/drug therapy , NF-kappa B/drug effects , Toll-Like Receptors/drug effects , Animals , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Male , NF-kappa B/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sodium/metabolism , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/metabolism
6.
Cell Physiol Biochem ; 44(2): 607-617, 2017.
Article in English | MEDLINE | ID: mdl-29161701

ABSTRACT

BACKGROUND/AIMS: Type I interferon (IFN-1) production and IFN-1 signaling play critical roles in the host antiviral innate immune responses. Although transcription factor Yin Yang 1 (YY1) has been reported to have a dual activator/repressor role during the regulation of interferon beta (IFN-ß) promoter activity, the roles of YY1 in the regulation of upstream signaling pathways leading to IFN-1 induction and IFN-1 signaling during viral infection remain to be elucidated. METHODS: The roles of YY1 in IFN-1 production and IFN-1 signaling were investigated using immunoblotting, real-time PCR, small interfering RNA (siRNA)-mediated YY1 knockdown, YY1 overexpression by transient transfection, and co-immunoprecipitation, using mouse cells. RESULTS: YY1 was shown to interact with STAT1 in the absence of viral infection. Following viral infection, YY1 protein expression levels were decreased. YY1 knockdown led to a considerable downregulation of phosphorylated (p) TBK1 and pIRF3 expressions, while YY1 overexpression significantly upregulated pTBK1 and pIRF3 expression levels and promoted virus-induced IFN-ß production. Additionally, YY1 knockdown led to a significant upregulation of pSTAT1, pSTAT2 and antiviral interferon-stimulated genes, and inhibited viral replication. CONCLUSION: We demonstrated here that YY1 interacts with STAT1 and dynamically regulates the induction of IFN-1 production and activation of IFN-1 signaling in different stages during viral infection.


Subject(s)
Immunity, Innate , YY1 Transcription Factor/metabolism , Animals , Cell Line , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Immunoprecipitation , Interferon Regulatory Factor-3/metabolism , Interferon-beta/analysis , Interferon-beta/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction , Simplexvirus/physiology , Transfection , Up-Regulation , Vesiculovirus/physiology , YY1 Transcription Factor/antagonists & inhibitors , YY1 Transcription Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...