1.Novel and potent inhibitors targeting DHODH are broad-spectrum antivirals against RNA viruses including newly-emerged coronavirus SARS-CoV-2.
Rui XIONG ; Leike ZHANG ; Shiliang LI ; Yuan SUN ; Minyi DING ; Yong WANG ; Yongliang ZHAO ; Yan WU ; Weijuan SHANG ; Xiaming JIANG ; Jiwei SHAN ; Zihao SHEN ; Yi TONG ; Liuxin XU ; Yu CHEN ; Yingle LIU ; Gang ZOU ; Dimitri LAVILLETE ; Zhenjiang ZHAO ; Rui WANG ; Lili ZHU ; Gengfu XIAO ; Ke LAN ; Honglin LI ; Ke XU
Protein & Cell 2020;11(10):723-739
Emerging and re-emerging RNA viruses occasionally cause epidemics and pandemics worldwide, such as the on-going outbreak of the novel coronavirus SARS-CoV-2. Herein, we identified two potent inhibitors of human DHODH, S312 and S416, with favorable drug-likeness and pharmacokinetic profiles, which all showed broad-spectrum antiviral effects against various RNA viruses, including influenza A virus, Zika virus, Ebola virus, and particularly against SARS-CoV-2. Notably, S416 is reported to be the most potent inhibitor so far with an EC of 17 nmol/L and an SI value of 10,505.88 in infected cells. Our results are the first to validate that DHODH is an attractive host target through high antiviral efficacy in vivo and low virus replication in DHODH knock-out cells. This work demonstrates that both S312/S416 and old drugs (Leflunomide/Teriflunomide) with dual actions of antiviral and immuno-regulation may have clinical potentials to cure SARS-CoV-2 or other RNA viruses circulating worldwide, no matter such viruses are mutated or not.
Animals
;
Antiviral Agents
;
pharmacology
;
therapeutic use
;
Betacoronavirus
;
drug effects
;
physiology
;
Binding Sites
;
drug effects
;
Cell Line
;
Coronavirus Infections
;
drug therapy
;
virology
;
Crotonates
;
pharmacology
;
Cytokine Release Syndrome
;
drug therapy
;
Drug Evaluation, Preclinical
;
Gene Knockout Techniques
;
Humans
;
Influenza A virus
;
drug effects
;
Leflunomide
;
pharmacology
;
Mice
;
Mice, Inbred BALB C
;
Orthomyxoviridae Infections
;
drug therapy
;
Oseltamivir
;
therapeutic use
;
Oxidoreductases
;
antagonists & inhibitors
;
metabolism
;
Pandemics
;
Pneumonia, Viral
;
drug therapy
;
virology
;
Protein Binding
;
drug effects
;
Pyrimidines
;
biosynthesis
;
RNA Viruses
;
drug effects
;
physiology
;
Structure-Activity Relationship
;
Toluidines
;
pharmacology
;
Ubiquinone
;
metabolism
;
Virus Replication
;
drug effects
2.High dose vitamin C inhibits proliferation of breast cancer cells through reducing glycolysis and protein synthesis.
Qingmei WANG ; Qianzi XU ; Anyi WEI ; Shishuo CHEN ; Chong ZHANG ; Linghui ZENG
Journal of Zhejiang University. Medical sciences 2019;48(3):296-302
OBJECTIVE:
To investigate the effects of high dose vitamin C (VC) on proliferation of breast cancer cells and to explore its mechanisms.
METHODS:
Human breast cancer cells Bcap37 and MDA-MB-453 were treated with VC at low dose (0.01 mmol/L), medium dose (0.10 mmol/L) and high dose (2.00 mmol/L). Cell proliferation was determined with CCK-8 assay, protein expression was evaluated by Western blot, and the secretion of lactic acid in tumor cells was detected by colorimetric method. Bcap37 cells were inoculated in nude mice, and tumor baring nude mice were intraperitoneally injected with high VC(4 g/kg, VC group, =5)or normal saline (control group, =5) for 24 d. Tumor weight and body weight were calculated.
RESULTS:
experiments demonstrated that high dose VC significantly inhibited cell proliferation in Bcap37 and MDA-MB-453 cells (all <0.01); the expressions of Glut1 and mTOR signaling pathway-related proteins were decreased (all <0.05); and the secretion of lactic acid was also markedly reduced (all <0.05). experiment showed that the tumor weight was decreased in mice treated with high-dose VC as compared with control group (<0.05), but no difference in body weights between two groups was observed.
CONCLUSIONS
High dose VC may inhibit proliferation of breast cancer cells both and through reducing glycolysis and protein synthesis.
Animals
;
Ascorbic Acid
;
pharmacology
;
Breast Neoplasms
;
drug therapy
;
Cell Line, Tumor
;
Cell Proliferation
;
drug effects
;
Glycolysis
;
drug effects
;
Humans
;
Mice
;
Mice, Nude
;
Protein Biosynthesis
;
drug effects
3.Effects of estrogen receptor GPR30 agonist G1 on neuronal apoptosis and microglia polarization in traumatic brain injury rats.
Meng-Xian PAN ; Jun-Chun TANG ; Rui LIU ; Yu-Gong FENG ; Qi WAN
Chinese Journal of Traumatology 2018;21(4):224-228
PURPOSETo investigate the effects of estrogen G protein-coupled receptor 30 (GPR30) agonist G1 on hippocampal neuronal apoptosis and microglial polarization in rat traumatic brain injury (TBI).
METHODSMale SD rats were randomly divided into sham group, TBI + vehicle group, TBI + G1 group. Experimental moderate TBI was induced using Feeney's weigh-drop method. G1 (100μg/kg) or vehicle was intravenously injected from femoral vein at 30 min post-injury. Rats were sacrificed at 24 h after injury for detection of neuronal apoptosis and microglia polarization. Neuronal apoptosis was assayed by immunofluorescent staining of active caspase-3. M1 type microglia markers (iNOS and IL-1β) and M2 type markers (Arg1 and IL-4) were examined by immunoblotting or ELISA. Total protein level of Akt and phosphorylated Akt were assayed by immunoblotting.
RESULTSG1 significantly reduced active caspase-3 positive neurons in hippocampus. Meanwhile G1 increased the ratio of Arg1/iNOS. IL-1β production was decreased but IL-4 was increased after G1 treatment. G1 treatment also increased the active form of Akt.
CONCLUSIONSGPR30 agonist G1 inhibited neuronal apoptosis and favored microglia polarization to M2 type.
Animals ; Apoptosis ; drug effects ; Brain Injuries, Traumatic ; drug therapy ; pathology ; Cell Polarity ; Hippocampus ; drug effects ; Interleukin-1beta ; biosynthesis ; Male ; Microglia ; drug effects ; Neurons ; drug effects ; Proto-Oncogene Proteins c-akt ; metabolism ; Rats ; Rats, Sprague-Dawley ; Receptors, G-Protein-Coupled ; agonists
4.Osthole decreases collagen I/III contents and their ratio in TGF-β1-overexpressed mouse cardiac fibroblasts through regulating the TGF-β/Smad signaling pathway.
Jin-Cheng LIU ; Lei ZHOU ; Feng WANG ; Zong-Qi CHENG ; Chen RONG
Chinese Journal of Natural Medicines (English Ed.) 2018;16(5):321-329
The present study was designed to elucidate whether the mechanism by which osthole decreases collagenI/III contents and their ratio is regulating the TGF-β/Smad signaling pathway in TGF-β1-overexpressed mouse cardiac fibroblasts (CFs). These CFs were cultured and treated with different concentrations of osthole. Our results showed that the TGF-β1 expression in the CFs transfected with that the recombinant expression plasmids pcDNA3.1(+)-TGF-β1 was significantly enhanced. After the CFs were treated with 1.25-5 μg·mL of osthole for 24 h, the mRNA and protein expression levels of collagensIand III were reduced. The collagen I/III ratio was also reduced. The mRNA and protein expression levels of TGF-β1, TβRI, Smad2/3, P-Smad2/3, Smad4, and α-SMA were decreased, whereas the expression level of Smad7 was increased. These effects suggested that osthole could inhibit collagen I and III expression and reduce their ratio via the TGF-β/Smad signaling pathway in TGF-β1 overexpressed CFs. These effects of osthole may play beneficial roles in the prevention and treatment of myocardial fibrosis.
Actins
;
genetics
;
Animals
;
Cells, Cultured
;
Collagen
;
biosynthesis
;
genetics
;
Coumarins
;
pharmacology
;
Fibroblasts
;
drug effects
;
metabolism
;
Gene Expression Regulation
;
drug effects
;
Mice
;
Myocardium
;
cytology
;
Protein-Serine-Threonine Kinases
;
genetics
;
RNA, Messenger
;
genetics
;
Real-Time Polymerase Chain Reaction
;
Receptor, Transforming Growth Factor-beta Type I
;
Receptors, Transforming Growth Factor beta
;
genetics
;
Signal Transduction
;
drug effects
;
Smad Proteins
;
genetics
;
Transforming Growth Factor beta1
;
genetics
5.Differential Inhibition of Nav1.7 and Neuropathic Pain by Hybridoma-Produced and Recombinant Monoclonal Antibodies that Target Nav1.7 : Differential activities of Nav1.7-targeting monoclonal antibodies.
Sangsu BANG ; Jiho YOO ; Xingrui GONG ; Di LIU ; Qingjian HAN ; Xin LUO ; Wonseok CHANG ; Gang CHEN ; Sang-Taek IM ; Yong Ho KIM ; Judith A STRONG ; Ma-Zhong ZHANG ; Jun-Ming ZHANG ; Seok-Yong LEE ; Ru-Rong JI
Neuroscience Bulletin 2018;34(1):22-41
The voltage-gated Na channel subtype Nav1.7 is important for pain and itch in rodents and humans. We previously showed that a Nav1.7-targeting monoclonal antibody (SVmab) reduces Na currents and pain and itch responses in mice. Here, we investigated whether recombinant SVmab (rSVmab) binds to and blocks Nav1.7 similar to SVmab. ELISA tests revealed that SVmab was capable of binding to Nav1.7-expressing HEK293 cells, mouse DRG neurons, human nerve tissue, and the voltage-sensor domain II of Nav1.7. In contrast, rSVmab showed no or weak binding to Nav1.7 in these tests. Patch-clamp recordings showed that SVmab, but not rSVmab, markedly inhibited Na currents in Nav1.7-expressing HEK293 cells. Notably, electrical field stimulation increased the blocking activity of SVmab and rSVmab in Nav1.7-expressing HEK293 cells. SVmab was more effective than rSVmab in inhibiting paclitaxel-induced mechanical allodynia. SVmab also bound to human DRG neurons and inhibited their Na currents. Finally, potential reasons for the differential efficacy of SVmab and rSVmab and future directions are discussed.
Animals
;
Antibodies, Monoclonal
;
therapeutic use
;
Biotin
;
metabolism
;
Cells, Cultured
;
Disease Models, Animal
;
Female
;
Ganglia, Spinal
;
cytology
;
HEK293 Cells
;
Humans
;
Hybridomas
;
chemistry
;
Hyperalgesia
;
drug therapy
;
Male
;
Mice
;
Mice, Inbred C57BL
;
NAV1.5 Voltage-Gated Sodium Channel
;
metabolism
;
NAV1.7 Voltage-Gated Sodium Channel
;
chemistry
;
immunology
;
metabolism
;
Neuralgia
;
drug therapy
;
metabolism
;
Protein Binding
;
drug effects
;
Recombinant Proteins
;
biosynthesis
;
therapeutic use
;
Sensory Receptor Cells
;
drug effects
;
physiology
6.Aldosterone induces inflammatory cytokines in penile corpus cavernosum by activating the NF-κB pathway.
Fei WU ; Zu-Quan XIONG ; Shan-Hua MAO ; Ji-Meng HU ; Jian-Qing WANG ; Hao-Wen JIANG ; Qiang DING
Asian Journal of Andrology 2018;20(1):24-29
Emerging evidence indicates that aldosterone and mineralocorticoid receptors (MRs) are associated with the pathogenesis of erectile dysfunction. However, the molecular mechanisms remain largely unknown. In this study, freshly isolated penile corpus cavernosum tissue from rats was treated with aldosterone, with or without MRs inhibitors. Nuclear factor (NF)-kappa B (NF-κB) activity was evaluated by real-time quantitative PCR, luciferase assay, and immunoblot. The results demonstrated that mRNA levels of the NF-κB target genes, including inhibitor of NF-κB alpha (IκB-α), NF-κB1, tumor necrosis factor-alpha (TNF-α), and interleukin 6 (IL-6), were higher after aldosterone treatment. Accordingly, phosphorylation of p65/RelA, IκB-α, and inhibitor of NF-κB kinase-β was markedly increased by aldosterone. Furthermore, knockdown of MRs prevented activation of the NF-κB canonical pathway by aldosterone. Consistent with this finding, ectopic overexpression of MRs enhanced the transcriptional activation of NF-κB by aldosterone. More importantly, the MRs antagonist, spironolactone blocked aldosterone-mediated activation of the canonical NF-κB pathway. In conclusion, aldosterone has an inflammatory effect in the corpus cavernosum penis, inducing NF-κB activation via an MRs-dependent pathway, which may be prevented by selective MRs antagonists. These data reveal the possible role of aldosterone in erectile dysfunction as well as its potential as a novel pharmacologic target for treatment.
Aldosterone/pharmacology*
;
Animals
;
Cytokines/biosynthesis*
;
Gene Knockdown Techniques
;
I-kappa B Kinase/antagonists & inhibitors*
;
Interleukin-6/genetics*
;
Male
;
Mineralocorticoid Receptor Antagonists/pharmacology*
;
NF-kappa B/genetics*
;
Penis/metabolism*
;
Protein Serine-Threonine Kinases/antagonists & inhibitors*
;
RNA, Messenger/biosynthesis*
;
Rats
;
Rats, Inbred WKY
;
Receptors, Mineralocorticoid/genetics*
;
Signal Transduction/drug effects*
;
Spironolactone/pharmacology*
;
Transcriptional Activation
;
Tumor Necrosis Factor-alpha/biosynthesis*
;
NF-kappaB-Inducing Kinase
7.Hyperbaric Oxygen Pretreatment Improves Cognition and Reduces Hippocampal Damage Via p38 Mitogen-Activated Protein Kinase in a Rat Model.
Baisong ZHAO ; Yongying PAN ; Zixin WANG ; Haiping XU ; Xingrong SONG
Yonsei Medical Journal 2017;58(1):131-138
PURPOSE: To investigate the effects of hyperbaric oxygen (HBO) pretreatment on cognitive decline and neuronal damage in an Alzheimer’s disease (AD) rat model. MATERIALS AND METHODS: Rats were divided into three groups: normal saline (NS), AD, and HBO+AD. In the AD group, amyloid β peptide (Aβ)₁₋₄₀ was injected into the hippocampal CA1 region of the brain. NS rats received NS injection. In the HBO+AD group, rats received 5 days of daily HBO therapy following Aβ₁₋₄₀ injection. Learning and memory capabilities were examined using the Morris water maze task. Neuronal damage and astrocyte activation were evaluated by hematoxylin-eosin staining and immunohistochemistry, respectively. Dendritic spine density was determined by Golgi-Cox staining. Tumor necrosis factor-α, interleukin-1β, and interleukin-10 production was assessed by enzyme-linked immunosorbent assay. Neuron apoptosis was evaluated by terminal deoxynucleotidyl transferase dUTP nick end labeling. Protein expression was examined by western blotting. RESULTS: Learning and memory dysfunction was ameliorated in the HBO+AD group, as shown by significantly lower swimming distances and escape latency, compared to the AD group. Lower rates of neuronal damage, astrocyte activation, dendritic spine loss, and hippocampal neuron apoptosis were seen in the HBO+AD than in the AD group. A lower rate of hippocampal p38 mitogen-activated protein kinase (MAPK) phosphorylation was observed in the HBO+AD than in the AD group. CONCLUSION: HBO pretreatment improves cognition and reduces hippocampal damage via p38 MAPK in AD rats.
Alzheimer Disease/*therapy
;
Amyloid beta-Peptides/*administration & dosage
;
Animals
;
Apoptosis
;
*Cognition/drug effects
;
Disease Models, Animal
;
Enzyme-Linked Immunosorbent Assay
;
Hippocampus/*enzymology
;
*Hyperbaric Oxygenation
;
In Situ Nick-End Labeling
;
Interleukin-10/biosynthesis
;
Interleukin-1beta/biosynthesis
;
Learning/drug effects
;
Male
;
Memory/drug effects
;
Neurons
;
Peptide Fragments/*administration & dosage
;
Rats
;
Rats, Sprague-Dawley
;
Sodium Chloride/administration & dosage
;
Tumor Necrosis Factor-alpha/biosynthesis
;
p38 Mitogen-Activated Protein Kinases/*metabolism
8.An inducible CRISPR-ON system for controllable gene activation in human pluripotent stem cells.
Jianying GUO ; Dacheng MA ; Rujin HUANG ; Jia MING ; Min YE ; Kehkooi KEE ; Zhen XIE ; Jie NA
Protein & Cell 2017;8(5):379-393
Human pluripotent stem cells (hPSCs) are an important system to study early human development, model human diseases, and develop cell replacement therapies. However, genetic manipulation of hPSCs is challenging and a method to simultaneously activate multiple genomic sites in a controllable manner is sorely needed. Here, we constructed a CRISPR-ON system to efficiently upregulate endogenous genes in hPSCs. A doxycycline (Dox) inducible dCas9-VP64-p65-Rta (dCas9-VPR) transcription activator and a reverse Tet transactivator (rtTA) expression cassette were knocked into the two alleles of the AAVS1 locus to generate an iVPR hESC line. We showed that the dCas9-VPR level could be precisely and reversibly controlled by the addition and withdrawal of Dox. Upon transfection of multiplexed gRNA plasmid targeting the NANOG promoter and Dox induction, we were able to control NANOG gene expression from its endogenous locus. Interestingly, an elevated NANOG level promoted naïve pluripotent gene expression, enhanced cell survival and clonogenicity, and enabled hESCs to integrate with the inner cell mass (ICM) of mouse blastocysts in vitro. Thus, iVPR cells provide a convenient platform for gene function studies as well as high-throughput screens in hPSCs.
Animals
;
Cell Line
;
Clustered Regularly Interspaced Short Palindromic Repeats
;
Doxycycline
;
pharmacology
;
Gene Expression Regulation
;
drug effects
;
Human Embryonic Stem Cells
;
metabolism
;
Humans
;
Mice
;
Nanog Homeobox Protein
;
biosynthesis
;
genetics
;
Pluripotent Stem Cells
;
metabolism
9.Telmisartan reduced cerebral edema by inhibiting NLRP3 inflammasome in mice with cold brain injury.
Xin WEI ; Chen-Chen HU ; Ya-Li ZHANG ; Shang-Long YAO ; Wei-Ke MAO
Journal of Huazhong University of Science and Technology (Medical Sciences) 2016;36(4):576-583
The aim of this study was to investigate the possible beneficial role of telmisartan in cerebral edema after traumatic brain injury (TBI) and the potential mechanisms related to the nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) pyrin domain-containing 3 (NLRP3) inflammasome activation. TBI model was established by cold-induced brain injury. Male C57BL/6 mice were randomly assigned into 3, 6, 12, 24, 48 and 72 h survival groups to investigate cerebral edema development with time and received 0, 5, 10, 20 and 40 mg/kg telmisartan by oral gavage, 1 h prior to TBI to determine the efficient anti-edemic dose. The therapeutic window was identified by post-treating 30 min, 1 h, 2 h and 4 h after TBI. Blood-brain barrier (BBB) integrity, the neurological function and histological injury were assessed, at the same time, the mRNA and protein expression levels of NLRP3 inflammasome, IL-1β and IL-18 concentrations in peri-contused brain tissue were measured 24 h post TBI. The results showed that the traumatic cerebral edema occurred from 6 h, reached the peak at 24 h and recovered to the baseline 72 h after TBI. A single oral dose of 5, 10 and 20 mg/kg telmisartan could reduce cerebral edema. Post-treatment up to 2 h effectively limited the edema development. Furthermore, prophylactic administration of telmisartan markedly inhibited BBB impairment, NLRP3, apoptotic speck-containing protein (ASC) and Caspase-1 activation, as well as IL-1β and IL-18 maturation, subsequently improved the neurological outcomes. In conclusion, telmisartan can reduce traumatic cerebral edema by inhibiting the NLRP3 inflammasome-regulated IL-1β and IL-18 accumulation.
Animals
;
Benzimidazoles
;
administration & dosage
;
Benzoates
;
administration & dosage
;
Blood-Brain Barrier
;
drug effects
;
Brain Edema
;
drug therapy
;
genetics
;
pathology
;
Brain Injuries, Traumatic
;
drug therapy
;
genetics
;
pathology
;
Caspase 1
;
biosynthesis
;
Gene Expression Regulation
;
drug effects
;
Humans
;
Inflammasomes
;
adverse effects
;
genetics
;
Interleukin-18
;
biosynthesis
;
Interleukin-1beta
;
biosynthesis
;
Male
;
Mice
;
NLR Family, Pyrin Domain-Containing 3 Protein
;
biosynthesis
;
genetics
;
Signal Transduction
;
drug effects
10.Gel-based chemical cross-linking analysis of 20S proteasome subunit-subunit interactions in breast cancer.
Hai SONG ; Hua XIONG ; Jing CHE ; Qing-Song XI ; Liu HUANG ; Hui-Hua XIONG ; Peng ZHANG
Journal of Huazhong University of Science and Technology (Medical Sciences) 2016;36(4):564-570
The ubiquitin-proteasome system plays a pivotal role in breast tumorigenesis by controlling transcription factors, thus promoting cell cycle growth, and degradation of tumor suppressor proteins. However, breast cancer patients have failed to benefit from proteasome inhibitor treatment partially due to proteasome heterogeneity, which is poorly understood in malignant breast neoplasm. Chemical crosslinking is an increasingly important tool for mapping protein three-dimensional structures and proteinprotein interactions. In the present study, two cross-linkers, bis (sulfosuccinimidyl) suberate (BS(3)) and its water-insoluble analog disuccinimidyl suberate (DSS), were used to map the subunit-subunit interactions in 20S proteasome core particle (CP) from MDA-MB-231 cells. Different types of gel electrophoresis technologies were used. In combination with chemical cross-linking and mass spectrometry, we applied these gel electrophoresis technologies to the study of the noncovalent interactions among 20S proteasome subunits. Firstly, the CP subunit isoforms were profiled. Subsequently, using native/SDSPAGE, it was observed that 0.5 mmol/L BS(3) was a relatively optimal cross-linking concentration for CP subunit-subunit interaction study. 2-DE analysis of the cross-linked CP revealed that α1 might preinteract with α2, and α3 might pre-interact with α4. Moreover, there were different subtypes of α1α2 and α3α4 due to proteasome heterogeneity. There was no significant difference in cross-linking pattern for CP subunits between BS(3) and DSS. Taken together, the gel-based characterization in combination with chemical cross-linking could serve as a tool for the study of subunit interactions within a multi-subunit protein complex. The heterogeneity of 20S proteasome subunit observed in breast cancer cells may provide some key information for proteasome inhibition strategy.
Amino Acid Sequence
;
Breast Neoplasms
;
drug therapy
;
genetics
;
pathology
;
Cell Line, Tumor
;
Cross-Linking Reagents
;
administration & dosage
;
Electrophoresis, Gel, Two-Dimensional
;
Female
;
Humans
;
Mass Spectrometry
;
Proteasome Endopeptidase Complex
;
drug effects
;
Protein Binding
;
drug effects
;
Protein Isoforms
;
genetics
;
Protein Subunits
;
biosynthesis
;
genetics
;
Proteomics
;
Succinimides
;
administration & dosage

Result Analysis
Print
Save
E-mail