Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 359
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Phytomedicine ; 128: 155404, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38507852

ABSTRACT

BACKGROUND: The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon (IFN) genes (STING) pathway is critical in the innate immune system and can be mobilized by cytosolic DNA. The various inflammatory and autoimmune diseases progression is highly correlated with aberrant cGAS-STING pathway activation. While some cGAS-STING pathway inhibitor were identified, there are no drugs that can be applied to the clinic. Compound Danshen Dripping Pill (CDDP) has been successfully used in clinic around the world, but the most common application is limited to cardiovascular disease. Therefore, the purpose of the present investigation was to examine whether CDDP inhibits the cGAS-STING pathway and could be used as a therapeutic agent for multiple cGAS-STING-triggered diseases. METHODS: BMDMs, THP1 cells or Trex1-/- BMDMs were stimulated with various cGAS-STING-agonists after pretreatment with CDDP to detect the function of CDDP on IFN-ß and ISGs productionn. Next, we detect the influence on IRF3 and P65 nuclear translocation, STING oligomerization and STING-TBK1-IRF3 complex formation of CDDP. Additionally, the DMXAA-mediated activation mice model of cGAS-STING pathway was used to study the effects of CDDP. Trex1-/- mice model and HFD-mediated obesity model were established to clarify the efficacy of CDDP on inflammatory and autoimmune diseases. RESULTS: CDDP efficacy suppressed the IRF3 phosphorylation or the generation of IFN-ß, ISGs, IL-6 and TNF-α. Mechanistically, CDDP did not influence the STING oligomerization and IRF3-TBK1 and STING-IRF3 interaction, but remarkably eliminated the STING-TBK1 interaction, ultimately blocking the downstream responses. In addition, we also clarified that CDDP could suppress cGAS-STING pathway activation triggered by DMXAA, in vivo. Consistently, CDDP could alleviate multi-organ inflammatory responses in Trex1-/- mice model and attenuate the inflammatory disorders, incleding obesity-induced insulin resistance. CONCLUSION: CDDP is a specifically cGAS-STING pathway inhibitor. Furthermore, we provide novel mechanism for CDDP and discovered a clinical agent for the therapy of cGAS-STING-triggered inflammatory and autoimmune diseases.


Subject(s)
Autoimmune Diseases , Camphanes , Drugs, Chinese Herbal , Inflammation , Panax notoginseng , Salvia miltiorrhiza , Mice , Mice, Inbred C57BL , Salvia miltiorrhiza/chemistry , Panax notoginseng/chemistry , Immunity, Innate , Membrane Proteins/agonists , Membrane Proteins/metabolism , Signal Transduction , THP-1 Cells , Exodeoxyribonucleases/genetics , Phosphoproteins/genetics , Macrophages , Interferon-beta/metabolism , Interferon Regulatory Factor-3/metabolism , Transcription Factor RelA/metabolism , Active Transport, Cell Nucleus , Autoimmune Diseases/drug therapy , Inflammation/drug therapy , Obesity/drug therapy , Diet, High-Fat , Protein Serine-Threonine Kinases/metabolism , Humans
2.
Signal Transduct Target Ther ; 8(1): 425, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37945593

ABSTRACT

Proper subcellular localization is crucial for the functioning of biomacromolecules, including proteins and RNAs. Nuclear transport is a fundamental cellular process that regulates the localization of many macromolecules within the nuclear or cytoplasmic compartments. In humans, approximately 60 proteins are involved in nuclear transport, including nucleoporins that form membrane-embedded nuclear pore complexes, karyopherins that transport cargoes through these complexes, and Ran system proteins that ensure directed and rapid transport. Many of these nuclear transport proteins play additional and essential roles in mitosis, biomolecular condensation, and gene transcription. Dysregulation of nuclear transport is linked to major human diseases such as cancer, neurodegenerative diseases, and viral infections. Selinexor (KPT-330), an inhibitor targeting the nuclear export factor XPO1 (also known as CRM1), was approved in 2019 to treat two types of blood cancers, and dozens of clinical trials of are ongoing. This review summarizes approximately three decades of research data in this field but focuses on the structure and function of individual nuclear transport proteins from recent studies, providing a cutting-edge and holistic view on the role of nuclear transport proteins in health and disease. In-depth knowledge of this rapidly evolving field has the potential to bring new insights into fundamental biology, pathogenic mechanisms, and therapeutic approaches.


Subject(s)
Neoplasms , Receptors, Cytoplasmic and Nuclear , Humans , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/therapeutic use , Active Transport, Cell Nucleus/genetics , Karyopherins/genetics , Karyopherins/metabolism , Karyopherins/therapeutic use , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Neoplasms/metabolism , ran GTP-Binding Protein
3.
Gene ; 815: 146178, 2022 Mar 20.
Article in English | MEDLINE | ID: mdl-34995733

ABSTRACT

Frailty develops due to multiple factors, such as sarcopenia, chronic pain, and dementia. Go-sha-jinki-Gan (GJG) is a traditional Japanese herbal medicine used for age-related symptoms. We have reported that GJG improved sarcopenia, chronic pain, and central nervous system function through suppression of tumor necrosis factor-alpha (TNF-α) production. In the present study, GJG was found to reduce the production of TNF-α in the soleus muscle of senescence-accelerated mice at 12 weeks and 36 weeks. GJG did not change the differentiation of C2C12 cells with 2% horse serum. GJG significantly decreased the expression of Muscle atrophy F-box protein (MAFbx) induced by TNF-α in C2C12 cells on real-time PCR. TNF-α significantly decreased the expression of PGC-1α and negated the enhancing effect of GJG for the expression of PGC-1α on digital PCR. Examining 20 chemical compounds derived from GJG, cinnamaldehyde from cinnamon bark and Chikusetsusaponin V (CsV) from Achyrantes Root dose-dependently decreased the production of TNF-⍺ in RAW264.7 cells stimulated by LPS. CsV inhibited the nuclear translocation of nuclear factor-kappa B (NF-κB) p65 in RAW264.7 cells. CsV showed low permeability using Caco-2 cells. However, the plasma concentration of CsV was detected from 30 min to 6 h and peaked at 1 h in the CD1 (ICR) mice after a single dose of GJG. In 8-week-old SAMP8 mice fed 4% (w/w) GJG from one week to four weeks, the plasma CsV concentration ranged from 0.0500 to 10.0 ng/mL. The evidence that CsV plays an important role in various anti-aging effects of GJG via suppression of TNF-⍺ expression is presented.


Subject(s)
Aging/drug effects , Drugs, Chinese Herbal/pharmacology , Saponins/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Active Transport, Cell Nucleus/drug effects , Administration, Oral , Animals , Dose-Response Relationship, Drug , Drug Stability , Drugs, Chinese Herbal/chemistry , Male , Mice , Mice, Inbred ICR , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , RAW 264.7 Cells , SKP Cullin F-Box Protein Ligases/metabolism , Saponins/administration & dosage , Saponins/blood , Solubility , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/pharmacology
4.
Cancer Lett ; 523: 57-71, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34563641

ABSTRACT

High fluence low-level laser (HF-LLL), a mitochondria-targeted tumour phototherapy, results in oxidative damage and apoptosis of tumour cells, as well as damage to normal tissue. To circumvent this, the therapeutic effect of low fluence LLL (LFL), a non-invasive and drug-free therapeutic strategy, was identified for tumours and the underlying molecular mechanisms were investigated. We observed that LFL enhanced antigen-specific immune response of macrophages and dendritic cells by upregulating MHC class II, which was induced by mitochondrial reactive oxygen species (ROS)-activated signalling, suppressing tumour growth in both CD11c-DTR and C57BL/6 mice. Mechanistically, LFL upregulated MHC class II in an MHC class II transactivator (CIITA)-dependent manner. LFL-activated protein kinase C (PKC) promoted the nuclear translocation of CIITA, as inhibition of PKC attenuated the DNA-binding efficiency of CIITA to MHC class II promoter. CIITA mRNA and protein expression also improved after LFL treatment, characterised by direct binding of Src and STAT1, and subsequent activation of STAT1. Notably, scavenging of ROS downregulated LFL-induced Src and PKC activation and antagonised the effects of LFL treatment. Thus, LFL treatment altered the adaptive immune response via the mitochondrial ROS-activated signalling pathway to control the progress of neoplastic disease.


Subject(s)
Histocompatibility Antigens Class II/immunology , Low-Level Light Therapy/methods , Neoplasms, Experimental/therapy , Protein Kinase C/physiology , Reactive Oxygen Species/metabolism , T-Lymphocytes/immunology , src-Family Kinases/physiology , Active Transport, Cell Nucleus , Animals , Antigen Presentation , Dendritic Cells/physiology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Nuclear Proteins/physiology , STAT1 Transcription Factor/physiology , Trans-Activators/physiology
5.
Pharmacol Res ; 170: 105723, 2021 08.
Article in English | MEDLINE | ID: mdl-34116210

ABSTRACT

FAT atypical cadherin 4 (FAT4) has been identified as a tumor suppressor in lung cancers. However, no agent for lung cancer treatment targeting FAT4 has been used in the clinic. Jujuboside A (JUA) is a major active compound in Semen Ziziphi Spinosae. Semen Ziziphi Spinosae is a traditional Chinese herbal medicine used clinically for tumor treatment to improve patients' quality of life. However, the anti-lung cancer activity and the underlying mechanisms of JUA are not yet fully understood. Here, we demonstrated the anti-lung cancer activity of JUA in two lung cancer mice models and three non-small cell lung cancer (NSCLC) cell lines, and further illustrated its underlying mechanisms. JUA suppressed the occurrence and development of lung cancer and extended mice survival in vivo, and suppressed NSCLC cell activities through cell cycle arrest, proliferation suppression, stemness inhibition and senescence promotion. Moreover, JUA directly bound with and activated FAT4, subsequently activating FAT4-HIPPO signaling and inhibiting YAP nuclear translocation. Knockdown of FAT4 diminished JUA's effects on HIPPO signaling, YAP nuclear translocation, cell proliferation and cellular senescence. In conclusion, JUA significantly suppressed NSCLC tumorigenesis by regulating FAT4-HIPPO-YAP signaling. Our findings suggest that JUA is a novel FAT4 activator that can be developed as a promising NSCLC therapeutic agent targeting the FAT4-HIPPO-YAP pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cadherins/agonists , Carcinoma, Non-Small-Cell Lung/drug therapy , Hippo Signaling Pathway/drug effects , Lung Neoplasms/drug therapy , Saponins/pharmacology , Tumor Suppressor Proteins/agonists , YAP-Signaling Proteins/metabolism , Active Transport, Cell Nucleus , Animals , Cadherins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Tumor Suppressor Proteins/metabolism
6.
J Cardiovasc Pharmacol ; 78(3): 411-421, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34132687

ABSTRACT

ABSTRACT: Calcific aortic valve disease is a common heart disease that contributes to increased cardiovascular morbidity and mortality. There is a lack of effective pharmaceutical therapy because its mechanisms are not yet fully known. Ginkgo biloba extract (EGB761) is reported to alleviate vascular calcification. However, whether EGB761 protects against aortic valve calcification, a disease whose pathogenesis shares many similarities with vascular calcification, and potential molecular mechanisms remain unknown. In this study, porcine aortic valve interstitial cell (pAVIC) calcification was induced by warfarin with or without the presence of EGB761. Immunostaining was performed to establish and characterize the pAVIC phenotype. Calcium deposition and calcium content were examined by Alizarin Red S staining and an intracellular calcium content assay. Alkaline phosphatase activity was detected by the p-nitrophenyl phosphate method. The expression levels of bone morphogenetic protein-2 (BMP2), Runt-related transcription factor 2 (Runx2), homeobox protein MSX-2, and phosphorylated (p)-Smad1/5 were detected by reverse transcription-quantitative polymerase chain reaction (PCR) and Western blot analysis. Consistent with these in vitro data, we also confirmed the suppression of in vivo calcification by EGB761 in the warfarin-induced C57/Bl6 mice. The results indicated that both pAVICs and aortic valves tissue of mice stimulated with warfarin showed increased calcium deposition and expression of osteogenic markers (alkaline phosphatase, BMP2, homeobox protein MSX-2, and Runx2) and promoted p-Smad1/5 translocation from the cytoplasm to the nucleus. The addition of EGB761 significantly inhibited p-Smad1/5 translocation from the cytoplasm to the nucleus, thus suppressing calcification. In conclusion, EGB761 could ameliorate warfarin-induced aortic valve calcification through the inhibition of the BMP2-medicated Smad1/5/Runx2 signaling pathway.


Subject(s)
Aortic Valve/drug effects , Bone Morphogenetic Protein 2/metabolism , Calcinosis/prevention & control , Core Binding Factor Alpha 1 Subunit/metabolism , Heart Valve Diseases/prevention & control , Plant Extracts/pharmacology , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Active Transport, Cell Nucleus , Animals , Aortic Valve/metabolism , Aortic Valve/pathology , Calcinosis/chemically induced , Calcinosis/metabolism , Calcinosis/pathology , Calcium/metabolism , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Disease Models, Animal , Ginkgo biloba , Heart Valve Diseases/chemically induced , Heart Valve Diseases/metabolism , Heart Valve Diseases/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice, Inbred C57BL , Osteogenesis/drug effects , Phosphorylation , Signal Transduction , Sus scrofa , Warfarin
7.
Acta Histochem ; 123(5): 151735, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34052674

ABSTRACT

Hyperglycemia-induced oxidative stress has been implicated in diabetes and its complications. Medicinal plants possessing antioxidant activity may decrease oxidative stress by scavenging radicals and reducing power activity and would be a promising strategy for the treatment of inflammatory disorders like diabetes. This study was designed to evaluate the antioxidant effect of Aqueous Extract of S.coccinea leaf (AESL) in HG treated THP-1 cells and streptozotocin (STZ)-induced diabetic Wistar rats. AESL and the standard antidiabetic drug glibenclamide were administered orally by intragastric tube for 14 days and pre-treated HG grown THP-1 cells. AESL treatment reduced HG induced increase in ROS production, NF-κB dependent proinflammatory gene expression by influencing NF-κB nuclear translocation in THP-1 cells. Oral administration of AESL inhibited STZ-induced increase in serum lipid peroxidation, aspartate transaminase, alanine transaminase, and Lactate dehydrogenase of diabetic rats. Significant increase in activity of superoxide dismutase, catalase and glutathione peroxidase, and a reduced level of glutathione, were observed in AESL treatment. The results demonstrate that AESL is useful in controlling blood glucose and also has antioxidant potential to influence the translocation of NF-κB, protect damage caused by hyperglycemia-induced inflammation.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , NF-kappa B/metabolism , Oxidative Stress , Plant Extracts/pharmacology , Plants, Medicinal/metabolism , Active Transport, Cell Nucleus , Administration, Oral , Animals , Antioxidants/metabolism , Blood Glucose/metabolism , Catalase/metabolism , Free Radicals , Glucose Tolerance Test , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glyburide/pharmacology , Humans , Inflammation , Lipid Peroxidation , Pancreas/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species , Salvia , Signal Transduction , THP-1 Cells , Tetrazolium Salts , Thiazoles
8.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article in English | MEDLINE | ID: mdl-33771918

ABSTRACT

Knockout of ERß in the mouse leads to nuclear expression of epidermal growth factor receptor (EGFR) in the prostate. To examine whether ERß plays a similar role in the human prostate, we used four cohorts of men: 1) a Swedish cohort of normal prostates and PCa (prostate cancer) of different Gleason grades; 2) men with benign prostatic hyperplasia (BPH) treated with the 5α-reductase inhibitor, finasteride, and finasteride together with the ERß agonists, soy isoflavones; 3) men with PCa above Gleason grade 4 (GG4), treated with ADT (androgen deprivation therapy) and abiraterone (AA), the blocker of androgen synthesis for different durations; and 4) men with GG4 PCa on ADT or ADT with the AR (androgen receptor) blocker, enzalutamide, for 4 mo to 6 mo. In men with BPH, finasteride treatment induced EGFR nuclear expression, but, when finasteride was combined with isoflavones, EGFR remained on the cell membrane. In GG4 patients, blocking of AR for 4 mo to 6 mo resulted in loss of ERß and PTEN expression and increase in patients with nuclear EGFR from 10 to 40%. In the men with GG4 PCa, blocking of adrenal synthesis of testosterone for 2 mo to 7 mo had the beneficial effect of increasing ERß expression, but, on treatment longer than 8 mo, ERß was lost and EGFR moved to the nucleus. Since nuclear EGFR is a predictor of poor outcome in PCa, addition of ERß agonists together with abiraterone should be considered as a treatment that might sustain expression of ERß and offer some benefit to patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Estrogen Receptor beta/agonists , Prostatic Hyperplasia/drug therapy , Prostatic Neoplasms/drug therapy , Active Transport, Cell Nucleus/drug effects , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , Androstenes/pharmacology , Androstenes/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzamides/pharmacology , Benzamides/therapeutic use , Biopsy , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cohort Studies , ErbB Receptors/metabolism , Estrogen Receptor beta/metabolism , Finasteride/pharmacology , Finasteride/therapeutic use , Humans , Male , Mice , Mice, Knockout , Neoplasm Grading , Nitriles/pharmacology , Nitriles/therapeutic use , PTEN Phosphohydrolase/metabolism , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Phytoestrogens/pharmacology , Phytoestrogens/therapeutic use , Prostate/cytology , Prostate/drug effects , Prostate/pathology , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism
9.
Infect Genet Evol ; 88: 104699, 2021 03.
Article in English | MEDLINE | ID: mdl-33385575

ABSTRACT

Plasmodium falciparum, the prime causative agent of malaria, is responsible for 4, 05,000 deaths per year and fatality rates are higher among the children aged below 5 years. The emerging distribution of the multi-drug resistant P. falciparum becomes a worldwide concern, so the identification of unique targets and novel inhibitors is a prime need now. In the present study, we have employed pharmacoinformatics approaches to analyze 265 lead-like compounds from PubChem databases for virtual screening. Thereafter, 15 lead-like compounds were docked within the active side pocket of importin alpha. Comparative ligand properties and absorption, distribution, metabolism, excretion, and toxicity (ADMET) profile were also assessed. Finally, a novel inhibitor was designed and assessed computationally for its efficacy. From the comparative analysis we have found that our screened compounds possess better results than the existing lead ivermectin; having the highest binding energy of -15.6 kcal/mol, whereas ivermectin has -12.4 kcal/mol. The novel lead compound possessed more fascinating output without deviating any of the rules of Lipinski. It also possessed higher bioavailability and the drug-likeness score of 0.55 and 0.71, respectively compared to ivermectin. Furthermore, the binding study was confirmed by molecular dynamics simulation over 25 ns by evaluating the stability of the complex. Finally, all the screened compounds and the novel compound showed promising ADMET properties likewise. To end, we hope that our proposed screened compounds, as well as the novel compound, might give some advances to treat malaria efficiently in vitro and in vivo.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Antimalarials/pharmacology , Malaria, Falciparum/drug therapy , Plasmodium falciparum/chemistry , Plasmodium falciparum/drug effects , alpha Karyopherins/chemistry , beta Karyopherins/chemistry , Drug Design , Drug Discovery , Drug Evaluation, Preclinical , Drug Resistance , Humans , Ligands , Malaria, Falciparum/parasitology , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , alpha Karyopherins/metabolism , beta Karyopherins/metabolism
10.
Biomolecules ; 11(1)2021 01 13.
Article in English | MEDLINE | ID: mdl-33450991

ABSTRACT

Before replicating, Pospiviroidae viroids must move into the plant nucleus. However, the mechanisms of viroid nuclear import are not entirely understood. To study the nuclear import of viroids, we established a nuclear import assay system using onion cell strips and observed the import of Alexa Fluor-594-labeled citrus exocortis viroid (CEVd). To identify the plant factors involved in the nuclear import of viroids, we cloned the Viroid RNA-binding Protein 1 (VIRP1) gene from a tomato cultivar, Seokwang, and heterologously expressed and purified the VIRP1 protein. The newly prepared VIRP1 protein had alterations of amino acid residues at two points (H52R, A277G) compared with a reference VIRP1 protein (AJ249595). VIRP1 specifically bound to CEVd and promoted its nuclear import. However, it is still uncertain whether VIRP1 is the only factor required for the nuclear import of CEVd because CEVd entered the plant nuclei without VIRP1 in our assay system. The cause of the observed nuclear accumulation of CEVd in the absence of VIRP1 needs to be further clarified.


Subject(s)
Cell Nucleus/metabolism , Citrus/virology , Plant Proteins/metabolism , Viroids/metabolism , Active Transport, Cell Nucleus , Solanum lycopersicum , Onions/cytology , Plant Epidermis/cytology , Plant Proteins/isolation & purification , Protein Binding
11.
PLoS One ; 15(11): e0241739, 2020.
Article in English | MEDLINE | ID: mdl-33206688

ABSTRACT

Due to the challenges for developing vaccines in devastating pandemic situations of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), developing and screening of novel antiviral agents are peremptorily demanded. Herein, we developed EGYVIR as a potent immunomodulatory herbal extract with promising antiviral activity against SARS-CoV-2. It constitutes of a combination of black pepper extract with curcumin extract. The antiviral effect of EGYVIR extract is attributed to the two key phases of the disease in severe cases. First, the inhibition of the nuclear translocation of NF-kß p50, attenuating the SARS-CoV-2 infection-associated cytokine storm. Additionally, the EGYVIR extract has an in vitro virucidal effect for SARS-CoV-2. The in vitro study of EGYVIR extract against SARS-CoV-2 on Huh-7 cell lines, revealed the potential role of NF-kß/TNFα/IL-6 during the infection process. EGYVIR antagonizes the NF-kß pathway in-silico and in-vitro studies. Consequently, it has the potential to hinder the release of IL-6 and TNFα, decreasing the production of essential cytokines storm elements.


Subject(s)
Antiviral Agents/pharmacology , Immunologic Factors/pharmacology , Plant Extracts/pharmacology , SARS-CoV-2/drug effects , Active Transport, Cell Nucleus/drug effects , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chlorocebus aethiops , Curcuma/chemistry , Humans , Interleukin-6/metabolism , Kinetics , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B p50 Subunit/metabolism , Piper nigrum/chemistry , Tumor Necrosis Factor-alpha/metabolism , Vero Cells
12.
J Cell Mol Med ; 24(22): 13208-13222, 2020 11.
Article in English | MEDLINE | ID: mdl-33058500

ABSTRACT

High fructose intake is a risk factor for liver fibrosis. Polydatin is a main constituent of the rhizome of Polygonum cuspidatum, which has been used in traditional Chinese medicine to treat liver fibrosis. However, the underlying mechanisms of fructose-driven liver fibrosis as well as the actions of polydatin are not fully understood. In this study, fructose was found to promote zinc finger E-box binding homeobox 1 (ZEB1) nuclear translocation, decrease microRNA-203 (miR-203) expression, increase survivin, activate transforming growth factor ß1 (TGF-ß1)/Smad signalling, down-regulate E-cadherin, and up-regulate fibroblast specific protein 1 (FSP1), vimentin, N-cadherin and collagen I (COL1A1) in rat livers and BRL-3A cells, in parallel with fructose-induced liver fibrosis. Furthermore, ZEB1 nuclear translocation-mediated miR-203 low-expression was found to target survivin to activate TGF-ß1/Smad signalling, causing the EMT in fructose-exposed BRL-3A cells. Polydatin antagonized ZEB1 nuclear translocation to up-regulate miR-203, subsequently blocked survivin-activated TGF-ß1/Smad signalling, which were consistent with its protection against fructose-induced EMT and liver fibrosis. These results suggest that ZEB1 nuclear translocation may play an essential role in fructose-induced EMT in liver fibrosis by targeting survivin to activate TGF-ß1/Smad signalling. The suppression of ZEB1 nuclear translocation by polydatin may be a novel strategy for attenuating the EMT in liver fibrosis associated with high fructose diet.


Subject(s)
Epithelial-Mesenchymal Transition , Glucosides/pharmacology , Liver Cirrhosis/metabolism , Stilbenes/pharmacology , Zinc Finger E-box-Binding Homeobox 1/metabolism , Active Transport, Cell Nucleus , Animals , Cadherins/metabolism , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Fructose , Liver Cirrhosis/chemically induced , Male , MicroRNAs/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Transforming Growth Factor beta1/metabolism
13.
Med Oncol ; 37(5): 44, 2020 Apr 08.
Article in English | MEDLINE | ID: mdl-32270306

ABSTRACT

Trillin is a constituent of total Trillium Tschonoskii Maxim (TTM), which is extracted from TTM and displayed anti-tumor effect in many tumor cell lines. However, the anti-tumor mechanism of trillin is still unclear. This study demonstrated that trillin could dramatically inhibit hepatoma carcinoma cell proliferation, induce apoptosis and decrease migration and invasion through suppressing phosphorylated STAT3 translocated to nucleus. Trillin could down-regulate Bcl-2 and Survivin, up-regulate cleaved PRAP, leading to dramatically apoptosis; trillin could also down-regulate MMP1, MMP2, MucI and VEGF, which displayed an inhibition effect on hepatocellular tumor cells invasion and development. The results of this study indicated the potential utility of trillin as a STAT3 inhibitor for the treatment of cancers.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , STAT3 Transcription Factor/metabolism , Trillium/chemistry , Active Transport, Cell Nucleus/drug effects , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Plant Extracts/chemistry , Plant Extracts/pharmacology
14.
PLoS Pathog ; 16(4): e1008407, 2020 04.
Article in English | MEDLINE | ID: mdl-32240278

ABSTRACT

Influenza A viruses are human pathogens with limited therapeutic options. Therefore, it is crucial to devise strategies for the identification of new classes of antiviral medications. The influenza A virus genome is constituted of 8 RNA segments. Two of these viral RNAs are transcribed into mRNAs that are alternatively spliced. The M1 mRNA encodes the M1 protein but is also alternatively spliced to yield the M2 mRNA during infection. M1 to M2 mRNA splicing occurs at nuclear speckles, and M1 and M2 mRNAs are exported to the cytoplasm for translation. M1 and M2 proteins are critical for viral trafficking, assembly, and budding. Here we show that gene knockout of the cellular protein NS1-BP, a constituent of the M mRNA speckle-export pathway and a binding partner of the virulence factor NS1 protein, inhibits M mRNA nuclear export without altering bulk cellular mRNA export, providing an avenue to preferentially target influenza virus. We performed a high-content, image-based chemical screen using single-molecule RNA-FISH to label viral M mRNAs followed by multistep quantitative approaches to assess cellular mRNA and cell toxicity. We identified inhibitors of viral mRNA biogenesis and nuclear export that exhibited no significant activity towards bulk cellular mRNA at non-cytotoxic concentrations. Among the hits is a small molecule that preferentially inhibits nuclear export of a subset of viral and cellular mRNAs without altering bulk cellular mRNA export. These findings underscore specific nuclear export requirements for viral mRNAs and phenocopy down-regulation of the mRNA export factor UAP56. This RNA export inhibitor impaired replication of diverse influenza A virus strains at non-toxic concentrations. Thus, this screening strategy yielded compounds that alone or in combination may serve as leads to new ways of treating influenza virus infection and are novel tools for studying viral RNA trafficking in the nucleus.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Antiviral Agents/pharmacology , Cell Nucleus/virology , Influenza A virus/metabolism , Influenza, Human/virology , RNA, Messenger/metabolism , RNA, Viral/metabolism , Drug Evaluation, Preclinical , Humans , Influenza A virus/genetics , RNA, Messenger/genetics , RNA, Viral/genetics , Virus Replication/drug effects
15.
Int J Biol Sci ; 16(9): 1586-1603, 2020.
Article in English | MEDLINE | ID: mdl-32226304

ABSTRACT

Herpes simplex virus (HSV) type 1 (HSV-1) infection exhibited high heterogeneity at individual cells level, including the different gene expression patterns and varying amounts of progeny virus. However, the underlying mechanism of such variability remains obscure. The importance of host long noncoding RNAs (lncRNAs) in virus infection had been recognized, while the contribution of lncRNAs to the heterogeneous infection remains unknown. Herein, a prior single-cell RNA sequencing data using HSV-1 reporter strain expressing ICP4-YFP was re-analyzed to obtain the differentially expressed lncRNA between the successfully initiated viral gene expression (ICP4-YFP+) cells and the aborted infection cells (ICP4-YFP-). The ICP4-YFP+ population show a higher abundance of MAMDC2 antisense 1 (MAMDC2-AS1) lncRNA than ICP4-YFP- population. MAMDC2-AS1 silencing reduces the expression of HSV-1 immediate early (IE) genes and limit HSV-1 infection in human host cells. Consistently, ectopic expression of MAMDC2-AS1 enhances HSV-1 IE genes transcription and facilitates the formation of HSV-1-induced plaques. Mechanically, both RNA-pull down and RNA immunoprecipitation assays show that MAMDC2-AS1 interacts with the RNA binding protein heat shock protein 90α (Hsp90α), a molecular chaperone involving in the nuclear import of HSV-1. The MAMDC2-AS1-Hsp90α interaction facilitates the nuclear transport of viral tegument protein VP16, the core factor initiating the expression of HSV-1 IE genes. The transcription factor YY1 mediates the induction of MAMDC2-AS1 upon HSV-1 infection. Our study elucidates the contribution of lncRNA to HSV-1 infection susceptibility in human cells and the role of Hsp90α RNA binding activity in HSV-1 infection.


Subject(s)
Cell Nucleus/virology , Herpesvirus 1, Human/metabolism , RNA, Long Noncoding/physiology , Active Transport, Cell Nucleus , Cell Line , Genes, Immediate-Early , HSP90 Heat-Shock Proteins/metabolism , Herpes Simplex Virus Protein Vmw65/metabolism , Herpesvirus 1, Human/genetics , Humans , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , RNA-Seq , Single-Cell Analysis , YY1 Transcription Factor/physiology
16.
Biomolecules ; 10(2)2020 02 05.
Article in English | MEDLINE | ID: mdl-32033472

ABSTRACT

Cancer stem cells are responsible for breast cancer initiation, metastasis, and relapse. Targeting breast cancer stem cells (BCSCs) using phytochemicals is a good strategy for the treatment of cancer. A silica gel, a reversed-phase C18 column (ODS), a Sephadex LH-20 gel, thin layer chromatography, and high-performance liquid chromatography (HPLC) were used for compound isolation from Saururus chinensis extracts. The isolated compound was identified as machilin D by mass spectrometry and nuclear magnetic resonance (NMR). Machilin D inhibited the growth and mammosphere formation of breast cancer cells and inhibited tumor growth in a xenograft mouse model. Machilin D reduced the proportions of CD44+/CD24- and aldehyde dehydrogenase 1 (ALDH1)-positive cells. Furthermore, this compound reduced the nuclear localization of the NF-κB protein and decreased the IL-6 and IL-8 secretion in mammospheres. These results suggest that machilin D blocks IL-6 and IL-8 signaling and induces CSC death and thus may be a potential agent targeting BCSCs.


Subject(s)
Breast Neoplasms/drug therapy , Lignans/pharmacology , NF-kappa B/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Saururaceae/chemistry , Signal Transduction/drug effects , Active Transport, Cell Nucleus , Aldehyde Dehydrogenase 1 Family/metabolism , Animals , Breast Neoplasms/metabolism , CD24 Antigen/metabolism , Cell Line, Tumor , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Female , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , MCF-7 Cells , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Mice, Nude , NF-kappa B/metabolism , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Plant Extracts/chemistry , Powders
17.
Int J Mol Sci ; 21(2)2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31952110

ABSTRACT

Maresin-1 (MaR1) is a specialized pro-resolving mediator, derived from omega-3 fatty acids, whose functions are to decrease the pro-inflammatory and oxidative mediators, and also to stimulate cell division. We investigated the hepatoprotective actions of MaR1 in a rat model of liver ischemia-reperfusion (IR) injury. MaR1 (4 ng/gr body weight) was administered prior to ischemia (1 h) and reperfusion (3 h), and controls received isovolumetric vehicle solution. To analyze liver function, transaminases levels and tissue architecture were assayed, and serum cytokines TNF-α, IL-6, and IL-10, mitotic activity index, and differential levels of NF-κB and Nrf-2 transcription factors, were analyzed. Transaminase, TNF-α levels, and cytoarchitecture were normalized with the administration of MaR1 and associated with changes in NF-κB. IL-6, mitotic activity index, and nuclear translocation of Nrf-2 increased in the MaR1-IR group, which would be associated with hepatoprotection and cell proliferation. Taken together, these results suggest that MaR1 alleviated IR liver injury, facilitated by the activation of hepatocyte cell division, increased IL-6 cytokine levels, and the nuclear localization of Nrf-2, with a decrease of NF-κB activity. All of them were related to an improvement of liver injury parameters. These results open the possibility of MaR1 as a potential therapeutic tool in IR and other hepatic pathologies.


Subject(s)
Cell Proliferation/drug effects , Docosahexaenoic Acids/pharmacology , Hepatocytes/drug effects , Liver/drug effects , Reperfusion Injury/prevention & control , Active Transport, Cell Nucleus/drug effects , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytokines/blood , Cytokines/metabolism , Docosahexaenoic Acids/chemistry , Fatty Acids, Omega-3/chemistry , Hepatocytes/cytology , Hepatocytes/metabolism , Liver/blood supply , Liver/physiopathology , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Protective Agents/pharmacology , Rats, Sprague-Dawley , Reperfusion Injury/physiopathology , Transaminases/metabolism
18.
Expert Opin Pharmacother ; 21(4): 399-408, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31957504

ABSTRACT

Introduction: Despite unprecedented advances in the treatment of multiple myeloma (MM), almost all patients develop a disease that is resistant to the five most commonly used and active anti-MM agents. The prognosis for this patient population is particularly poor resulting in an unmet need for additional therapeutic options. Exportin-1 (XPO-1) is a major nuclear export protein of macromolecular cargo frequently overexpressed in MM. Selinexor is a first-in-class, oral Selective-Inhibitor-of-Nuclear-Export (SINE) compound that impedes XPO-1. Based on results of the STORM-trial, selinexor in combination with dexamethasone was granted accelerated FDA approval for patients with penta-refractory MM in July 2019.Areas covered: This article summarizes our up-to-date knowledge on the pathophysiologic role of XPO-1 in MM. Furthermore, it reviews the most recent clinical data on selinexor in combination with dexamethasone and other anti-MM agents; and discusses its safety profile, management strategies; and potential future developments.Expert opinion: Selinexor represents a next-generation-novel agent with an innovative mechanism of action that marks a significant advance in the treatment of heavily pretreated MM patients. Ongoing studies investigate its therapeutic potential also in earlier lines of therapy. Additional data is needed to confirm that selinexor and other SINE compounds are a valuable addition to our current therapeutic armamentarium.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hydrazines/therapeutic use , Karyopherins/antagonists & inhibitors , Multiple Myeloma/drug therapy , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Triazoles/therapeutic use , Active Transport, Cell Nucleus/drug effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Clinical Trials as Topic , Dexamethasone/therapeutic use , Drug Evaluation, Preclinical , Humans , Hydrazines/administration & dosage , Hydrazines/adverse effects , Hydrazines/pharmacokinetics , Karyopherins/genetics , Multiple Myeloma/metabolism , Prognosis , Receptors, Cytoplasmic and Nuclear/genetics , Triazoles/administration & dosage , Triazoles/adverse effects , Triazoles/pharmacokinetics , Exportin 1 Protein
19.
Biochem Biophys Res Commun ; 522(1): 40-46, 2020 01 29.
Article in English | MEDLINE | ID: mdl-31735336

ABSTRACT

Atopic dermatitis (AD) can occur in both children and adults, and the symptoms include itching and eczema, which in turn cause patients to suffer. Ophiopogonin D (OP-D) is a steroidal glycoside from Radix Ophiopogon japonicus, which is well known as an effective anti-inflammatory herbal medicine in many Asian countries. In this study, we aimed to investigate the anti-inflammatory effects of OP-D, using an AD mouse model and inflamed HaCaT cells. Through a histopathological analysis, we were able to confirm the suppressive effects of OP-D on skin thickening and the mast cell activation in AD-like mouse back skin tissues stimulated by DNCB. In addition, we detected significant decreases in cytokine expression levels through multiplex assessment assays of the OP-D-treated mice blood. We observed the anti-inflammatory effect of OP-D in the spleen, causing weight loss in the spleen and in the mRNA expression levels related to diverse cytokines. In human keratinocytes inflamed by TNF-α, OP-D inhibited p38 and ERK protein activation and showed a reduction of NF-κB nuclear translocation. Furthermore, OP-D attenuated pro-inflammatory cytokine mRNA expressions in TNF-α-inflamed HaCaT cells. Accordingly, we came to the conclusion that OP-D is a potential natural drug which can be used in order to treat inflammatory skin diseases, such as AD.


Subject(s)
Dermatitis, Atopic/drug therapy , Keratinocytes/drug effects , Saponins/pharmacology , Spirostans/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Active Transport, Cell Nucleus , Animals , Cell Line , Cytokines/metabolism , Dermatitis, Atopic/chemically induced , Dinitrochlorobenzene/pharmacology , Female , Humans , Inflammation , Mice , Mice, Inbred BALB C , Skin/drug effects , Spleen/drug effects
20.
PLoS One ; 14(11): e0216184, 2019.
Article in English | MEDLINE | ID: mdl-31693670

ABSTRACT

The increased recurrence of Candida albicans infections is associated with greater resistance to antifungal drugs. This involves the establishment of alternative therapeutic protocols, such as probiotic microorganisms whose antifungal potential has already been demonstrated using preclinical models (cell cultures, laboratory animals). Understanding the mechanisms of action of probiotic microorganisms has become a strategic need for the development of new therapeutics for humans. In this study, we investigated the prophylactic anti-C. albicans properties of Lactobacillus rhamnosus Lcr35® using the in vitro Caco-2 cell model and the in vivo Caenorhabditis elegans model. In Caco-2 cells, we showed that the strain Lcr35® significantly inhibited the growth (~2 log CFU.mL-1) and adhesion (150 to 6,300 times less) of the pathogen. Moreover, in addition to having a pro-longevity activity in the nematode (+42.9%, p = 3.56.10-6), Lcr35® protects the animal from the fungal infection (+267% of survival, p < 2.10-16) even if the yeast is still detectable in its intestine. At the mechanistic level, we noticed the repression of genes of the p38 MAPK signalling pathway and genes involved in the antifungal response induced by Lcr35®, suggesting that the pathogen no longer appears to be detected by the worm immune system. However, the DAF-16/FOXO transcription factor, implicated in the longevity and antipathogenic response of C. elegans, is activated by Lcr35®. These results suggest that the probiotic strain acts by stimulating its host via DAF-16 but also by suppressing the virulence of the pathogen.


Subject(s)
Candida albicans , Candidiasis/prevention & control , Lacticaseibacillus rhamnosus , Probiotics/therapeutic use , Active Transport, Cell Nucleus , Animals , Animals, Genetically Modified , Caco-2 Cells , Caenorhabditis elegans/genetics , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Candida albicans/growth & development , Candida albicans/pathogenicity , Disease Models, Animal , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Humans , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL