Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Mol Med (Berl) ; 2024 May 11.
Article in English | MEDLINE | ID: mdl-38733386

ABSTRACT

Atherosclerosis (AS) is a chronic inflammatory arterial disease, in which abnormal lipid metabolism and foam cell formation play key roles. Histamine is a vital biogenic amine catalyzed by histidine decarboxylase (HDC) from L-histidine. Histamine H1 receptor (H1R) antagonist is a commonly encountered anti-allergic agent in the clinic. However, the role and mechanism of H1R in atherosclerosis have not been fully elucidated. Here, we explored the effect of H1R on atherosclerosis using Apolipoprotein E-knockout (ApoE-/-) mice with astemizole (AST, a long-acting H1R antagonist) treatment. The results showed that AST increased atherosclerotic plaque area and hepatic lipid accumulation in mice. The result of microarray study identified a significant change of endothelial lipase (LIPG) in CD11b+ myeloid cells derived from HDC-knockout (HDC-/-) mice compared to WT mice. Blocking H1R promoted the formation of foam cells from bone marrow-derived macrophages (BMDMs) of mice by up-regulating p38 mitogen-activated protein kinase (p38 MAPK) and LIPG signaling pathway. Taken together, these findings demonstrate that blocking H1R signal aggravates atherosclerosis by promoting abnormal lipid metabolism and macrophage-derived foam cell formation via p38 MAPK-LIPG signaling pathway. KEY MESSAGES: Blocking H1R signal with AST aggravated atherosclerosis and increased hepatic lipid accumulation in high-fat diet (HFD)-fed ApoE-/- mice. Blocking H1R signal promoted macrophage-derived foam cell formation via p38 MAPK-LIPG signaling pathway.

2.
World J Surg Oncol ; 22(1): 35, 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38279138

ABSTRACT

BACKGROUND: Lip squamous cell carcinoma (LSCC) was one of the most common cancer types of head and neck tumors. This study aimed to find more predictors of the prognosis in postoperative LSCC patients. METHODS: A total of 147 LSCC patients between June 2012 and June 2018 were collected from two tertiary care institutions. There were 21 clinicopathological factors included and analyzed in our study. The univariate and multivariate Cox regression analyses were performed to find the independent prognostic factors for predicting progression-free survival (PFS) and overall survival (OS) in postoperative LSCC patients. The role of adjuvant radiotherapy in various subgroups was displayed by Kaplan-Meier plots. RESULTS: The 1-, 3-, and 5-year PFS of postoperative LSCC patients were 88.4%, 70.1%, and 57.8%, respectively. Similarly, the 1-, 3-, and 5-year OS of postoperative LSCC patients were 94.6%, 76.9%, and 69.4%, respectively. The results suggested that postoperative LSCC patients with age at diagnosis ≥ 70 years, grade with moderate or poor differentiate, the American Joint Committee on Cancer (AJCC) stage IV, higher systemic immune-inflammation index (SII), surgical margin < 5, and age-adjusted Charlson Comorbidity Index (ACCI) ≥ 5 tend to have a poorer PFS (all P < 0.05). Besides, postoperative LSCC patients with age at diagnosis ≥ 70 years, AJCC stage IV, higher GPS, higher SII, and ACCI ≥ 5 tend to have a worse OS (all P < 0.05). Additionally, postoperative patients with LSCC in the subgroup of ACCI < 5 and AJCC III-IV stage was more likely to benefit from adjuvant radiotherapy, but not for the other subgroups. CONCLUSION: We identified a series of significant immune-inflammation-related and comorbidity-related clinicopathological factors associated with the prognosis of postoperative LSCC patients by local data from two tertiary care institutions in China, which can be helpful for patients and surgeons to pay more attention to nutrition, inflammation, and complications and finally obtained a better prognosis.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Laryngeal Neoplasms , Humans , Aged , Prognosis , Lip , Carcinoma, Squamous Cell/pathology , Squamous Cell Carcinoma of Head and Neck , Inflammation , Laryngeal Neoplasms/pathology , Retrospective Studies
4.
ChemMedChem ; 18(22): e202300439, 2023 11 16.
Article in English | MEDLINE | ID: mdl-37755120

ABSTRACT

Over the past few decades, sophisticated nanomaterials have been used as carries for the targeted delivery of therapeutics to solid tumors. However, the low efficiency of intracellular internalization of nanocarriers in current use restricts their biomedical application. In this work, we demonstrate that novel virus-bionic mesoporous-silica-based nanocarriers can be successfully prepared for programmed precise drug delivery. These unique viral mimic nanovesicles not only present virus bionic counterparts and nanostructures, but also have infectious virus-like properties toward tumor cells and tumor tissues. Encouragingly, their large surface area (322.1 m2 /g) endows them with high loading capacity for therapeutic agents, especially, they have more effective gene transfection properties than the commercially available LipoGeneTM transfection reagent. Thanks to their virus-inspired morphology, they exhibit outstanding cellular uptake efficiency with living tumor cells and the ability to invade cells in large quantities with incubation times as short as 5 min, which is much faster than traditional mesoporous silica nanoparticles (mSN) with smooth appearance. Importantly, after doxorubicin (DOX) loading and surface modification of tumor recognition motifs, RGD (Arg-Gly-Asp, vMN@DOX-RGD), the bionic drug-loaded viral mimics elicit potent tumor cell elimination both in vitro and in vivo, greatly exceeding the mSN-based group. Our work paves the way toward virus bionic nanocarrier design for malignant tumor suppression in the clinic.


Subject(s)
Nanoparticles , Neoplasms , Humans , Silicon Dioxide/chemistry , Bionics , Drug Delivery Systems , Doxorubicin/pharmacology , Doxorubicin/chemistry , Neoplasms/drug therapy , Oligopeptides , Nanoparticles/chemistry , Porosity , Drug Carriers/chemistry
5.
Int Immunopharmacol ; 123: 110774, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37567012

ABSTRACT

Periodontal disease is a chronic inflammatory disease that is highly correlated with cardiovascular disease(CVD). Histamine has been proven to participate in the pathophysiological processes of cardiovascular disease and oral inflammation. However, the role of histamine in the development of cardiac microthrombosis caused by periodontal disease has not been fully elucidated. We established a murine periodontal inflammation model by injecting lipopolysaccharide (LPS) or Porphyromonas gingivalis (P. gingivalis). In order to examine the effect of histamine/H1R signaling on cardiac injury after periodontal disease, we used histidine decarboxylase- knockout (HDC-/-) mice and histamine 1 receptor (H1R) antagonist. Our results demonstrated that LPS-induced periodontal inflammation significantly increased CD11b+Gr-1+ neutrophils in the peripheral blood and myocardial interstitium. Histamine deficiency resulted in further increases in P. gingivalis, neutrophils, inflammatory cytokines, and cardiac microthrombosis in the myocardium of HDC-/- mice compared to wild-type (WT) mice. Mechanistic analysis showed that blocking H1R could synergistically interact with LPS, further increasing the phosphorylation of p65, exacerbating the inflammatory response of neutrophils and endothelial cell damage. Conclusively, the disruption of histamine-H1R signaling exacerbates cardiac microthrombosis after periodontal disease via TLR4/NFκB-p65 pathway. Our findings not only reveal a link between periodontal inflammation and myocardial injury but also provided some thoughts for the use of H1R antagonist in clinical practice.


Subject(s)
Cardiovascular Diseases , Periodontal Diseases , Animals , Mice , Histamine/metabolism , Histamine H1 Antagonists , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Receptors, Histamine H1/metabolism , Toll-Like Receptor 4/metabolism , Receptors, Histamine
6.
Cell Biol Int ; 47(9): 1573-1588, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37303238

ABSTRACT

Intimal thickening caused by the excessive multiplication of vascular smooth muscle cells (VSMCs) is the pathological process central to cardiovascular diseases, including restenosis. In response to vascular injury, VSMCs would undergo phenotypic switching from a fully differentiated, low proliferative rate phenotype to a more pro-proliferative, promigratory, and incompletely-differentiated state. The lack of a full understanding of the molecular pathways coupling the vascular injury stimuli to VSMCs phenotype switching largely limits the development of medical therapies for treating intima hyperplasia-related diseases. The role of signal transducers and activators of transcription 6 (STAT6) in modulating the proliferation and differentiation of various cell types, especially macrophage, has been well investigated, but little is known about its pathophysiological role and target genes in restenosis after vascular injury. In the present work, Stat6-/- mice were observed to exhibit less severe intimal hyperplasia compared with Stat6+/+ mice after carotid injury. The expression of STAT6 was upregulated in VSMCs located in the injured vascular walls. STAT6 deletion leads to decreased proliferation and migration of VSMCs while STAT6 overexpression enhances the proliferation and migration of VSMCs companies with reduced expression of VSMCs marker genes and organized stress fibers. The effect of STAT6 in mouse VSMCs was conserved in human aortic SMCs. RNA-deep-sequencing and experiments verification revealed LncRNA C7orf69/LOC100996318-miR-370-3p/FOXO1-ER stress signaling as the downstream network mediating the pro-dedifferentiation effect of STAT6 in VSMCs. These findings broaden our understanding of vascular pathological molecules and throw a beam of light on the therapy of a variety of proliferative vascular diseases.


Subject(s)
Myocytes, Smooth Muscle , Neointima , STAT6 Transcription Factor , Animals , Mice , STAT6 Transcription Factor/metabolism , Myocytes, Smooth Muscle/cytology , Muscle, Smooth, Vascular/cytology , Neointima/pathology , Vascular System Injuries/metabolism , Vascular System Injuries/pathology , Mice, Knockout , Humans , Mice, Inbred BALB C , Male , Female , Middle Aged , Aged , Cells, Cultured , Aorta/cytology , Cell Dedifferentiation
7.
Zhongguo Zhong Yao Za Zhi ; 48(3): 770-777, 2023 Feb.
Article in Chinese | MEDLINE | ID: mdl-36872241

ABSTRACT

This paper aimed to study the effect of Erjing Pills on the improvement of neuroinflammation of rats with Alzheimer's di-sease(AD) induced by the combination of D-galactose and Aß_(25-35) and its mechanism. SD rats were randomly divided into a sham group, a model control group, a positive drug group(donepezil, 1 mg·kg~(-1)), an Erjing Pills high-dose group(9.0 g·kg~(-1)), and an Erjing Pills low-dose group(4.5 g·kg~(-1)), with 14 rats each group. To establish the rat model of AD, Erjing Pills were intragastrically administrated to rats for 5 weeks after 2 weeks of D-galactose injection. D-galactose was intraperitoneally injected into rats for 3 weeks, and then Aß_(25-35) was injected into the bilateral hippocampus. The new object recognition test was used to evaluate the learning and memory ability of rats after 4 weeks of intragastric administration. Tissues were acquired 24 h after the last administration. The immunofluorescence method was used to detect the activation of microglia in the brain tissue of rats. The positive expressions of Aß_(1-42) and phosphory protein Tau~(404)(p-Tau~(404)) in the CA1 area of the hippocampus were detected by immunohistochemistry. The levels of inflammatory factors interleukin-1ß(IL-1ß), tumor necrosis factor-α(TNF-α), and interleukin-6(IL-6) in the brain tissue were determined by enzyme-linked immunosorbent assay(ELISA). Toll-like receptor 4(TLR4)/nuclear factor kappa B(NF-κB)/nucleotide-binding oligomerization domain-like receptors 3(NLRP3) pathway-associated proteins in the brain tissue were determined by Western blot. The results showed that as compared with the sham group, the new object recognition index of rats in the model control group decreased significantly, the deposition of Aß_(1-42) and p-Tau~(404) positive protein in the hippocampus increased significantly, and the levels of microglia activation increased significantly in the dentate gyrus. The levels of IL-1ß, TNF-α, and IL-6 in the hippocampus of the model control group increased significantly, and the expression levels of TLR4, p-NF-κB p65/NF-κB p65, p-IκBα/IκBα, and NLRP3 proteins in the hippocampus increased significantly. Compared with the model control group, the Erjing Pill groups enhanced the new object recognition index of rats, decreased the deposition of Aß_(1-42) and the expression of p-Tau~(404) positive protein in the hippocampus, inhibited the activation of microglia in the dentate gyrus, reduced the levels of inflammatory factors IL-1ß, TNF-α, and IL-6 in the hippocampus, and down-regulated the expression levels of TLR4, p-NF-κB P65/NF-κB P65, p-IκBα/IκBα, and NLRP3 proteins in the hippocampus. In conclusion, Erjing Pills can improve the learning and memory ability of the rat model of AD presumably by improving the activation of microglia, reducing the expression levels of neuroinflammatory factors IL-1ß, TNF-α, and IL-6, inhibiting the TLR4/NF-κB/NLRP3 neuroinflammation pathway, and decreasing hippocampal deposition of Aß and expression of p-Tau, thereby restoring the hippocampal morphological structure.


Subject(s)
NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Rats , Rats, Sprague-Dawley , NF-KappaB Inhibitor alpha , Galactose , Interleukin-6 , Neuroinflammatory Diseases , Toll-Like Receptor 4 , Tumor Necrosis Factor-alpha
8.
Front Neurosci ; 17: 1124034, 2023.
Article in English | MEDLINE | ID: mdl-36816106

ABSTRACT

Purpose: The stability of fixation is crucial for the development of visual function. In this study, we quantify the deviation of visual target during fixational and saccadic tasks using eye-tracking technology, reflecting the control ability and characteristics of fixational displacement among healthy adults in a convenient method. Methods: One hundred healthy participants aged between 18 and 55 years were recruited in the study. All participants underwent a complete ophthalmic assessment. The eye positions in the fixational and saccadic tasks were documented and analyzed by the Tobii eye-tracking system. Participants were grouped by age and gender. Targeting displacement (TD), defined as the average displacement between visual target and the mean of fixation points corresponding to that stimuli, was used to quantitatively observe fixational displacement in the horizontal and vertical directions. Result: There was a strong reproducibility of TD as an indicator of fixation (ICC 0.812 to 0.891, p < 0.001). The TD in fixational task was significantly smaller than that of the saccadic task (3.884 ± 0.525 vs. 4.484 ± 0.509, p < 0.001) among normal people. Moreover, the difference of TD in the horizontal and vertical meridians was related to the nature of the task: In the fixational task, the TD in horizontal was smaller than that in the vertical (p < 0.001), whereas the TD in horizontal was larger than that in vertical in the saccadic task (p = 0.003). In the different age and gender groups: There was no significant difference between different gender and age groups in fixational task. However, during the saccadic task, males had smaller TD in the vertical direction than females (4.061 ± 0.495 vs. 4.404 ± 0.484, p = 0.002), and the average TD increased with age, mainly in the vertical direction (all p < 0.05). The fixation stability decreased significantly in the group over 50-years-old. Conclusion: By reporting the fixational displacement of different genders and ages in fixational and saccadic tasks, as well as different longitude lines among normal people, our study might provide an objective, quantitative and convenient reference index for the evaluation of fixation stability in visual impairment diseases and aging phenomenon of visual function.

9.
Int Immunopharmacol ; 115: 109630, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36571917

ABSTRACT

Histamine is a versatile biogenic amine, generated by the unique enzyme histidine decarboxylase (Hdc). Accumulating evidence has proven that histamine plays important roles in numerous biological and pathophysiological processes. However, the role and mechanism of Hdc/Histamine signaling in periodontal diseases remain unclear. In our current study, the concentration of histamine increased in the serum, and Hdc gene expression was upregulated in the gingiva of WT mice with LPS-induced periodontal inflammation. With Hdc-GFP mice, we identified that Hdc/GFP in the periodontium was expressed in CD11b+ myeloid cells, rather than in tryptase-positive mast cells. Hdc-expressing CD11b+Gr-1+ neutrophils significantly increased in the peripheral blood of Hdc-GFP mice one day after LPS injection. Lack of histamine in Hdc-/- mice not only promoted the activation and infiltration of more CD11b+ cells into the peripheral blood but also upregulated mRNA expression levels of IL-1ß, IL-6, MCP-1and MMP9 in the gingiva compared to WT mice one day after LPS stimulation. 28 days after LPS treatment, we observed that Hdc-/- mice exhibited more alveolar bone loss and more osteoclasts than WT mice, which was slightly ameliorated by the administration of exogenous histamine. In vivo and in vitro mechanistic studies revealed that the mRNA expression levels of proinflammatory cytokines and protein levels of NLRP3, Caspase-1, and cleaved-Caspase-1 were upregulated after blocking histamine receptor 1 and 2, especially histamine receptor 1. Taken together, CD11b+Gr-1+ neutrophils are the predominant Hdc-expressing sites in periodontal inflammation, and deficiency of endogenous histamine in Hdc-/- mice exacerbates the destruction of the periodontium. Disruption of the histamine/H1R/H2R axis aggravates the inflammatory immune response via NLRP3/Casapse-1 pathway.


Subject(s)
Histamine , Periodontal Diseases , Mice , Animals , Histamine/metabolism , Lipopolysaccharides/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Caspase 1 , Disease Models, Animal , Receptors, Histamine , Inflammation/metabolism , RNA, Messenger
10.
Oxid Med Cell Longev ; 2021: 5577019, 2021.
Article in English | MEDLINE | ID: mdl-34795840

ABSTRACT

Myocardial ischemia-reperfusion injury (MIRI) is a phenomenon that reperfusion leads to irreversible damage to the myocardium and increases mortality in acute myocardial infarction (AMI) patients. There is no effective drug to treat MIRI. Tubeimoside I (TBM) is a triterpenoid saponin purified from Chinese traditional medicine tubeimu. In this study, 4 mg/kg TBM was given to mice intraperitoneally at 15 min after ischemia. And TBM treatment improved postischemic cardiac function, decreased infarct size, diminished lactate dehydrogenase release, ameliorated oxidative stress, and reduced apoptotic index. Notably, ischemia-reperfusion induced a significant decrease in cardiac SIRT3 expression and activity, while TBM treatment upregulated SIRT3's expression and activity. However, the cardioprotective effects of TBM were largely abolished by a SIRT3 inhibitor 3-(1H-1,2,3-triazol-4-yl) pyridine (3-TYP). This suggests that SIRT3 plays an essential role in TBM's cardioprotective effects. In vitro, TBM also protected H9c2 cells against simulated ischemia/reperfusion (SIR) injury by attenuating oxidative stress and apoptosis, and siSIRT3 diminished its protective effects. Taken together, our results demonstrate for the first time that TBM protects against MIRI through SIRT3-dependent regulation of oxidative stress and apoptosis. TBM might be a potential drug candidate for MIRI treatment.


Subject(s)
Apoptosis , Gene Expression Regulation/drug effects , Myocardial Reperfusion Injury/prevention & control , Oxidative Stress , Protective Agents/pharmacology , Saponins/pharmacology , Sirtuin 3/metabolism , Triterpenes/pharmacology , Animals , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Sirtuin 3/genetics
11.
Exp Ther Med ; 22(5): 1252, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34539848

ABSTRACT

Erjing pill, a Traditional Chinese Medicine (TCM) formulation composed of Polygonatum sibiricum and Lycium chinense, has an important role in the treatment of Alzheimer's disease (AD). However, the underlying mechanisms of the action of Erjing pill in AD have remained elusive. In the present study, the key ingredients of Erjing pill were investigated and the active components and their mechanisms of action on AD were analyzed based on networks pharmacology. By using the TCM and TCM Systems Pharmacology and databases, the components of Erjing pill were screened and the data were captured using Discovery Studio. The SwissTarget webserver database was used to predict the potential protein targets of Erjing pill components for pathologies related to AD. The data were further analyzed with the disease targets of AD based on analysis of the Online Mendelian Inheritance in Man, DiGSeE and Therapeutic Target Database. Subsequent analysis of mechanistic pathways of the screened components and protein targets allowed us to construct a network by using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes, which revealed potential molecular mechanisms of Erjing pill against AD. Finally, the protective effect of three active components on neurons was verified using an in vitro injury model of PC12 cells induced by Aß25-35. The results indicated that 65 bioactive components of Erjing pill, including lauric acid and zederone, and 6 targets, including acetylcholinesterase, butylcholinesterase and amyloid protein precursor, were closely associated with the prevention and treatment of AD. The molecular components of Erjing pill were indicated to be involved in various biological signaling processes, mainly in synaptic signal transmission, transsynaptic signal transmission and chemical synaptic transmission. Furthermore, related pathways targeted by Erjing pill in AD included the regulation of neuroactive ligand-receptor interactions, the PI3K-Akt signaling pathway, serotoninergic synapses, calcium signaling pathways and dopaminergic synapses. A cell viability assay indicated that the compounds (polygonatine A, polygonatine C and 4',5-dihydroxyflavone) assessed were able to significantly improve the survival rate and increase the Ca2+ level in a PC12 cell model of AD induced by amyloid-ß25-35. The present study revealed that the mechanisms of action of Erjing pill to prevent and treat AD included a multicompound, multitarget and multipathway regulatory network.

12.
Sci Rep ; 11(1): 11272, 2021 05 28.
Article in English | MEDLINE | ID: mdl-34050207

ABSTRACT

O-GlcNAcylation, an energy-sensitive posttranslational modification, can regulate the activity of endothelial nitric oxide synthase (eNOS). Previous studies found that Thr866 is the key site for low-glucose-mediated regulation of eNOS O-GlcNAc. However, it is not known whether this activity functions through the Thr866 site concomitant with other physical and chemical factors. Therefore, we first explored the effects of physical and chemical factors on eNOS O-GlcNAc and its Thr866 site. In this study, hypertonic stress, hyperthermia and hydrogen peroxide all increased the expression levels of eNOS O-GlcNAc, whereas hypoxia and high levels of alcohol had no effect. on the expression levels of eNOS O-GlcNAc; by contrast, low pH led to a decrease in eNOS O-GlcNAc levels. Notably, eNOS O-GlcNAc protein levels were unchanged after Thr866 site mutation only under hypertonic conditions, suggesting that hypertonic stress may act through the Thr866 site. Upon exploring the mechanism of hypertonic stress on eNOS O-GlcNAc activity and function, we found that hypertonic stress can upregulate the expression of O-linked N-acetylglucosamine (GlcNAc) transferase (OGT), which is dependent on AMPK. When AMPK was knocked out, the upregulation of OGT expression and increased O-GlcNAc modifications induced by hypertonic stress were reversed.


Subject(s)
Acetylglucosamine/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Osmotic Pressure/physiology , Acetylglucosamine/physiology , Animals , Cattle , Cell Line , Gene Expression/genetics , Gene Expression Regulation/genetics , Glycosylation , HEK293 Cells , Humans , N-Acetylglucosaminyltransferases/metabolism , Phosphorylation , Protein Processing, Post-Translational , Threonine/metabolism
13.
Eur J Pharmacol ; 905: 174186, 2021 Aug 15.
Article in English | MEDLINE | ID: mdl-34033817

ABSTRACT

Sepsis-induced cardiac dysfunction (SICD) is one of the key complications in sepsis and it is associated with adverse outcomes and increased mortality. There is no effective drug to treat SICD. Previously, we reported that tubeimoside I (TBM) improved survival of septic mice. The aim of this study is to figure out whether TBM ameliorates SICD. Also, SIRT3 was reported to protects against SICD. Our second aim is to confirm whether SIRT3 plays essential roles in TBM's protective effects against SICD. Our results demonstrated that TBM could alleviate SICD and SICD's key pathological factor, inflammation, oxidative stress, and apoptosis were all reduced by TBM. Notably, SICD induced a significant decrease in cardiac SIRT3 expression, while TBM treatment could reverse SIRT3 expression. To clarify whether TBM provides protection via SIRT3, we injected a specific SIRT3 inhibitor 3-(1H-1,2,3-triazol-4-yl) pyridine (3-TYP) into mice before TBM treatment. Then the cardioprotective effects of TBM were largely abolished by 3-TYP. This suggests that SIRT3 plays an essential role in TBM's cardioprotective effects. In vitro, TBM also protected H9c2 cells against LPS-induced injury, and siSIRT3 diminished these protective effects. Taken together, our results demonstrate that TBM protects against SICD via SIRT3. TBM might be a potential drug candidate for SICD treatment.


Subject(s)
Cardiotonic Agents/pharmacology , Heart Diseases/drug therapy , Heart Diseases/etiology , Saponins/pharmacology , Sepsis/complications , Sirtuin 3/metabolism , Sirtuins/metabolism , Triterpenes/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Apoptosis/drug effects , Cardiotonic Agents/therapeutic use , Heart Diseases/pathology , Inflammation/chemically induced , Inflammation/drug therapy , Lipopolysaccharides/toxicity , Male , Mice, Inbred C57BL , Oxidative Stress/drug effects , Rats , Saponins/therapeutic use , Sirtuin 3/antagonists & inhibitors , Sirtuin 3/genetics , Sirtuins/antagonists & inhibitors , Sirtuins/genetics , Triterpenes/therapeutic use
14.
Inflammation ; 44(5): 1782-1792, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33770326

ABSTRACT

It has been revealed that widespread vascular endothelial dysfunction occurs in septic shock, ultimately resulting in multiple organ failure. The mitochondrial deacetylase sirtuin 3 (SIRT3) is essential in the regulation of metabolism, anti-inflammation, and anti-oxidation. The purpose of this study is to investigate whether SIRT3 is associated with the pathological progression of endothelial dysfunction in sepsis. Septic shock model was induced by cecal ligation and puncture (CLP) surgery on wild-type C57BL/6 mice. We activated and inhibited the function of SIRT3 with honokiol (HKL) and 3-TYP, respectively, and then biochemical, inflammatory, and endothelial function parameters of vascular tissue and survival were determined after CLP. CLP significantly activated NF-κB and NLRP3 pathways and decreased survival rate, endothelium-dependent relaxation function, and expression of Ser1177 phosphorylation of endothelial nitric oxide synthase (p-eNOS). The activation of SIRT3 significantly attenuated the increases of NF-κB and NLRP3 pathways and the declines of p-eNOS, endothelium-dependent relaxation function, and survival rate in septic mice. However, it presented exactly opposite results if SIRT3 was suppressed. We suggested that SIRT3 had a critical protective effect against vascular inflammation and endothelial dysfunction in early sepsis. Our data support a potential therapeutic target in vascular dysfunction and septic shock.


Subject(s)
Endothelium, Vascular/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Punctures/adverse effects , Sepsis/metabolism , Sirtuin 3/metabolism , Acetylcholine/pharmacology , Animals , Aorta/drug effects , Aorta/metabolism , Cecum/pathology , Cecum/surgery , Endothelium, Vascular/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ligation/adverse effects , Male , Mice , Mice, Inbred C57BL , NF-kappa B/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Organ Culture Techniques , Sepsis/etiology , Sepsis/prevention & control , Signal Transduction/drug effects , Signal Transduction/physiology , Vasodilation/drug effects , Vasodilation/physiology
15.
Lab Invest ; 101(7): 897-907, 2021 07.
Article in English | MEDLINE | ID: mdl-33664465

ABSTRACT

Sepsis is life-threatening organ dysfunction caused by a deregulated host response to infection. Endothelial dysfunction is the initial factor leading to organ dysfunction and it is associated with increased mortality. There is no effective drug to treat sepsis-induced endothelial dysfunction. In this study, we detected a favorable effect of tubeimoside I (TBM) in ameliorating sepsis-induced endothelial dysfunction. To unveil the mechanism how TBM protects against sepsis-induced endothelial dysfunction, we examined TBM's effects on oxidative stress and apoptosis both in vivo and in vitro. TBM treatment alleviated oxidative stress by decreasing NOX2 and Ac-SOD2/SOD2 and decreased apoptosis by inhibiting cleaved caspse3 and Bax/Bcl-2. Notably, sepsis induced a significant decrease of SIRT3 expression in vascular endothelium, while TBM treatment reversed SIRT3 expression. To clarify whether TBM provides protection via SIRT3, we knockdown SIRT3 using siRNA before TBM treatment. Then, the cytoprotective effects of TBM were largely abolished by siSIRT3. This suggests that SIRT3 plays an essential role in TBM's endothelial protective effects and TBM might be a potential drug candidate to treat sepsis-induced endothelial dysfunction.


Subject(s)
Endothelium, Vascular/drug effects , Protective Agents/pharmacology , Saponins/pharmacology , Sepsis/metabolism , Sirtuin 3/metabolism , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects
16.
Nat Prod Res ; 35(23): 4945-4952, 2021 Dec.
Article in English | MEDLINE | ID: mdl-32515605

ABSTRACT

A new alkaloid polygonatine C (3-(2'-furaldehyde-5'-methoxymethyl)5,6,7,8 -tetrahydroindolizin-8-one, 1) and a new saponin huangjingsterol B (3-ß-hydroxyspirost-5,25-diene-12-one, 2), together with three known compounds 3-5 were isolated from the ethanolic extract of the Stewed Polygonatum cyrtonema Hua. The structures of these new compounds were identified by extensive spectroscopic analysis including 1D and 2D NMR and MS, as well as chemical methods. The new compounds were tested for their Protective effect against Aß25-35 induced cytotoxicity and oxidative stress damage in PC12 cells. The results indicated that compounds 1-2 can be considered as promising lead compounds for further research on Alzheimer's disease (AD) treatment.


Subject(s)
Alkaloids , Antineoplastic Agents , Polygonatum , Saponins , Alkaloids/pharmacology , Oxidative Stress
17.
J Ethnopharmacol ; 267: 113642, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33264658

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Tubeimoside I (TBM) is a triterpenoid saponin purified from tubeimu (tuber of Bolbostemma paniculatum (Maxim.) Franquet). In traditional Chinese medicine, tubeimu had been used to treat acute mastitis, snake bites, detoxication, inflammatory diseases, and tumors for over 1000 years. AIM OF THE STUDY: This study aimed to investigate whether TBM could promote angiogenesis and how to promote angiogenesis. MATERIALS AND METHODS: In vivo, the pro-angiogenic effects of TBM were examined using the hindlimb ischemia model. After the ischemia operation, 1 mg/kg/day TBM was given via intraperitoneal injection for 28 days and the recovery of blood flow was monitored by Doppler scanner every 7 days. The capillary density in gastrocnemius muscle was detected by immunofluorescence. Expression of related proteins were determined by western blotting. In vitro, the pro-angiogenic effects of TBM on HUVECs were examined by Cell Counting Kit-8, scratch assay, endothelial cell tube formation assay and western blotting. RESULTS: TBM improved recovery from hindlimb ischemia in C57BL/6 mice. TBM promoted endothelial cell viability, migration and tube formation in HUVECs. TBM could activate eNOS-VEGF signaling pathway by enhancing expression of eNOS. And TBM's pro-angiogenesis effects could be abolished by L-NAME (an inhibitor of eNOS). CONCLUSIONS: TBM promoted angiogenesis via the activation of eNOS-VEGF signaling pathway and TBM could be a novel agent for therapeutic angiogenesis in ischemic diseases.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Ischemia/drug therapy , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/metabolism , Saponins/pharmacology , Triterpenes/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Hindlimb , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Male , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/genetics , Regional Blood Flow , Signal Transduction , Vascular Endothelial Growth Factor A/genetics
18.
J Ethnopharmacol ; 259: 112940, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32389853

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Tinospora sinensis (Lour.) Merr. belongs to the family Menispermaceae. It is called LeZhe and is widely used as a kind of folk medicine especially in the Tibetan Plateau of China. T. sinensis has the functions of clearing away heat and detoxification, dispelling wind and dredging collaterals, calming and soothing the nerves. T. sinensis is an effective medicine for the prevention and treatment of aging diseases such as Alzheimer's disease (AD) in the Tibetan Plateau of China, whereas its material basis and underlying mechanisms are not clear. The aim of this study was to investigate the material basis and potential mechanisms of T. sinensis in the treatment of AD by using network pharmacology and molecular docking. MATERIALS AND METHODS: In this study, targets were collected from DrugBank database, Therapeutic Target Database (TTD) and literatures reports for the treatment of AD. Compounds were searched by literatures and systematic separation from T. sinensis. The molecular docking experiment was carried out by using Autodock Vina software to screen the bioactive compounds in T. sinensis and target proteins for AD. Then, the "compound-target network" was constructed by Cytoscape software. The drug-like properties of the active compounds were analyzed by pKCSM performs, and the protein-protein interaction (PPI) network was constructed by Search Tool for the Retrieval of Interacting Genes/Proteins (STRING). The Kyoto Encyclopedia of Genes and Genomes (KEGG) target pathway enrichment analysis was carried out by Database for Annotation, Visualization and Integrated Discovery (DAVID). Furthermore, the protective effect of neurons of two active compounds were verified with the injury cell model of PC12 and primary hippocampus neurons induced by Aß25-35. Finally, the key proteins of related pathways were quantitatively analyzed with Western blot method. RESULTS: In total, 105 compounds and 38 targets have been screened. The main active compounds contained berberine, which belongs to alkaloids, Aurantiamide acetate, N-P-coumaroyltyramine, which belongs to amides, Trans-syringin and 3-demethyl-phillyrin, which belongs to phenylpropanoids. The targets covered inflammation-related proteins, including Protein kinase B (AKT), Phosphoinositide 3-kinase (PI3K), Tyrosine-protein kinase JAK1 (JAK1), mammalian target of rapamycin (mTOR), tumor necrosis factor alpha (TNF-α), Neuronal NOS (NOS1), and cholinergic function-related proteins, including α4-Nicotinic acetylcholine receptor (α4 nAChR), Muscarinic acetylcholine receptor M1 (Muscarnic M1). Inflammation and cholinergic dysfunction were the center of the network and occupy a dominant position. And the results of enrichment analysis shown the pathways mainly contained phosphoinositide-3-kinase/Akt (PI3K/Akt) signal pathway, neurotrophic factors (NTFs) signal pathway, Hypoxia-inducible factor 1 (HIF-1) signal pathway, mechanistic Target of Rapamycin (mTOR) signal pathway, Tumor necrosis factor (TNF) signal pathway, insulin resistance (IR). The results of in vitro assays showed that the tested compounds could significantly improve the survival rate and inhibit the apoptosis of PC12 cells and primary hippocampal neurons injured by Aß25-35. Western blot results showed that T. sinensis had a significant effect on the expression of protein PI3K and Akt. CONCLUSION: Our results revealed that T. sinensis could prevent and treat AD through a multi-compound-multi-target-multi-pathway regulatory network. Our work also expected to provide new ideas and theoretical bases for searching for the active compounds in T. sinensis and potential mechanism in the prevention and treatment of AD by the network pharmacology and molecular docking. The results of in vitro assay and in vivo assay supported the results of molecular docking.


Subject(s)
Alzheimer Disease/drug therapy , Hippocampus/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Systems Biology , Tinospora , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Animals , Female , Gene Expression Regulation , Gene Regulatory Networks , Hippocampus/metabolism , Hippocampus/pathology , Male , Molecular Docking Simulation , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/isolation & purification , PC12 Cells , Peptide Fragments/toxicity , Plant Extracts/isolation & purification , Protein Interaction Maps , Rats , Rats, Sprague-Dawley , Signal Transduction , Tinospora/chemistry
19.
Biomed Pharmacother ; 126: 110083, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32272432

ABSTRACT

Sepsis is a disease with high mortality rate worldwide and inducible nitric oxide (iNOS) induced vascular hyporeactivity plays a key role in it. There is no effective drug to treat vascular hyporeactivity specifically. Tubeimoside I (TBM) is a triterpenoid saponin isolated from Rhizoma Bolbostemmatis. In this study, we found that 4 mg/kg TBM intraperitoneally injected 1 h before cecal ligation and puncture (CLP) partially improved survival, ameliorated mean arterial pressure (MAP) and enhanced vascular responsiveness to norepinephrine (NE) and KCl in wild-type septic mice. CLP activated TLR4-MyD88-NF-κB-iNOS pathway was also inhibited by TBM both in vitro and in vivo. However, iNOS gene knockout counteracted the protection provided by TBM. We conclude that TBM protects mice in sepsis by reducing excessive NO production through inhibiting the TLR4-MyD88-NF-κB-iNOS pathway. Our study suggests a possible therapeutic application of TBM in sepsis.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Gene Expression Regulation/drug effects , Nitric Oxide Synthase/genetics , Saponins/pharmacology , Sepsis/drug therapy , Sepsis/etiology , Triterpenes/pharmacology , Animals , Biomarkers , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gene Expression , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Mice , Mice, Knockout , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Nitric Oxide/metabolism , Norepinephrine/pharmacology , Prognosis , Sepsis/mortality , Signal Transduction/drug effects , Treatment Outcome , Vasoconstrictor Agents/pharmacology
20.
Inflammation ; 43(4): 1561-1571, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32239396

ABSTRACT

Endothelial dysfunction is responsible for multiple organ failure and the high mortality rate of sepsis. Nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome plays an essential role in the progression of sepsis. However, the role of NLRP3 inflammasome in the endothelial dysfunction of sepsis has not been fully elucidated. In this study, septic mice were induced by cecal ligation and puncture (CLP) operation, and human umbilical vein endothelial cells (HUVECs) were treated with lipopolysaccharide (LPS). The 24-h survival rate after CLP was observed. Vasodilation function of the aorta was detected by vascular reactivity experiments. Expression of p-eNOS, eNOS, TLR4, MYD88, p-p65, p65, p-ikbα, ikbα, iNOS, NLRP3, and IL-1ß in the aorta and HUVECs were determined by Western blot. Our results suggest that the p-eNOS expression was downregulated, the endothelium-dependent relaxation function was impaired, and TLR4, MYD88, p-p65, p-ikbα, iNOS, NLRP3, and IL-1ß expression increased after CLP. The onset of death was 12 h after CLP, and the mortality rate was nearly 50% at 24 h after operation. The decline of p-eNOS, endothelium-dependent vasodilation function, and survival rate significantly improved with NLRP3-specific inhibitor MCC950 intervention or NLRP3 knockout in CLP mice. The decrease of p-eNOS in HUVECs induced by LPS was alleviated when pretreated with MCC950 or interleukin-1 receptor antagonist (IL-1Ra). In summary, our results indicate that activation of the NLRP3 inflammasome contributes to the development of endothelial dysfunction of early sepsis in mice, suggesting its potential role as a therapeutic target for the treatment of sepsis.


Subject(s)
Endothelium, Vascular/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sepsis/metabolism , Animals , Endothelium, Vascular/drug effects , Furans , Heterocyclic Compounds, 4 or More Rings/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Indenes , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Sepsis/chemically induced , Sulfonamides , Sulfones/pharmacology , Vasodilation/drug effects , Vasodilation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...