Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Acta Pharmacol Sin ; 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38914677

ABSTRACT

Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have obvious advantages over MSC therapy. But the strong procoagulant properties of MSC-EVs pose a potential risk of thromboembolism, an issue that remains insufficiently explored. In this study, we systematically investigated the procoagulant activity of large EVs derived from human umbilical cord MSCs (UC-EVs) both in vitro and in vivo. UC-EVs were isolated from cell culture supernatants. Mice were injected with UC-EVs (0.125, 0.25, 0.5, 1, 2, 4 µg/g body weight) in 100 µL PBS via the tail vein. Behavior and mortality were monitored for 30 min after injection. We showed that these UC-EVs activated coagulation in a dose- and tissue factor-dependent manner. UC-EVs-induced coagulation in vitro could be inhibited by addition of tissue factor pathway inhibitor. Notably, intravenous administration of high doses of the UC-EVs (1 µg/g body weight or higher) led to rapid mortality due to multiple thrombus formations in lung tissue, platelets, and fibrinogen depletion, and prolonged prothrombin and activated partial thromboplastin times. Importantly, we demonstrated that pulmonary thromboembolism induced by the UC-EVs could be prevented by either reducing the infusion rate or by pre-injection of heparin, a known anticoagulant. In conclusion, this study elucidates the procoagulant characteristics and mechanisms of large UC-EVs, details the associated coagulation risk during intravenous delivery, sets a safe upper limit for intravenous dose, and offers effective strategies to prevent such mortal risks when high doses of large UC-EVs are needed for optimal therapeutic effects, with implications for the development and application of large UC-EV-based as well as other MSC-EV-based therapies.

2.
ACS Nano ; 18(9): 7046-7063, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38381372

ABSTRACT

Type 2 alveolar epithelial cell (AEC2) senescence is crucial to the pathogenesis of pulmonary fibrosis (PF). The nicotinamide adenine dinucleotide (NAD+)-consuming enzyme cluster of differentiation 38 (CD38) is a marker of senescent cells and is highly expressed in AEC2s of patients with PF, thus rendering it a potential treatment target. Umbilical cord mesenchymal stem cell (MSC)-derived extracellular vesicles (MSC-EVs) have emerged as a cell-free treatment with clinical application prospects in antiaging and antifibrosis treatments. Herein, we constructed CD38 antigen receptor membrane-modified MSC-EVs (CD38-ARM-MSC-EVs) by transfecting MSCs with a lentivirus loaded with a CD38 antigen receptor-CD8 transmembrane fragment fusion plasmid to target AEC2s and alleviate PF. Compared with MSC-EVs, the CD38-ARM-MSC-EVs engineered in this study showed a higher expression of the CD38 antigen receptor and antifibrotic miRNAs and targeted senescent AEC2s cells highly expressing CD38 in vitro and in naturally aged mouse models after intraperitoneal administration. CD38-ARM-MSC-EVs effectively restored the NAD+ levels, reversed the epithelial-mesenchymal transition phenotype, and rejuvenated senescent A549 cells in vitro, thereby mitigating multiple age-associated phenotypes and alleviating PF in aged mice. Thus, this study provides a technology to engineer MSC-EVs and support our CD38-ARM-MSC-EVs to be developed as promising agents with high clinical potential against PF.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Pulmonary Fibrosis , Humans , Mice , Animals , Pulmonary Fibrosis/therapy , Pulmonary Fibrosis/metabolism , Alveolar Epithelial Cells , NAD/metabolism , Extracellular Vesicles/metabolism , Receptors, Antigen/metabolism
3.
Acta Pharmacol Sin ; 45(1): 180-192, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37644132

ABSTRACT

Adhesion molecules play essential roles in the homeostatic regulation and malignant transformation of hematopoietic cells. The dysregulated expression of adhesion molecules in leukemic cells accelerates disease progression and the development of drug resistance. Thus, targeting adhesion molecules represents an attractive anti-leukemic therapeutic strategy. In this study, we investigated the prognostic role and functional significance of cytohesin-1 (CYTH1) in acute myeloid leukemia (AML). Analysis of AML patient data from the GEPIA and BloodSpot databases revealed that CYTH1 was significantly overexpressed in AML and independently correlated with prognosis. Functional assays using AML cell lines and an AML xenograft mouse model confirmed that CYTH1 depletion significantly inhibited the adhesion, migration, homing, and engraftment of leukemic cells, delaying disease progression and prolonging animal survival. The CYTH1 inhibitor SecinH3 exerted in vitro and in vivo anti-leukemic effects by disrupting leukemic adhesion and survival programs. In line with the CYTH1 knockdown results, targeting CYTH1 by SecinH3 suppressed integrin-associated adhesion signaling by reducing ITGB2 expression. SecinH3 treatment efficiently induced the apoptosis and inhibited the growth of a panel of AML cell lines (MOLM-13, MV4-11 and THP-1) with mixed-lineage leukemia gene rearrangement, partly by reducing the expression of the anti-apoptotic protein MCL1. Moreover, we showed that SecinH3 synergized with the BCL2-selective inhibitor ABT-199 (venetoclax) to inhibit the proliferation and promote the apoptosis of ABT-199-resistant leukemic cells. Taken together, our results not only shed light on the role of CYTH1 in cell-adhesion-mediated leukemogenesis but also propose a novel combination treatment strategy for AML.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Humans , Mice , Animals , Leukemia, Myeloid, Acute/drug therapy , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Adhesion Molecules , Disease Progression , Cell Line, Tumor
4.
Acta Pharmacol Sin ; 43(11): 2885-2894, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35365781

ABSTRACT

Few therapies can reverse the proangiogenic activity of senescent mesenchymal stromal/stem cells (MSCs). In this study, we investigated the effects of rapamycin on the proangiogenic ability of senescent human umbilical cord MSCs (UCMSCs). An in vitro replicative senescent cell model was established in cultured UCMSCs. We found that late passage (P25 or later) UCMSCs (LP-UCMSCs) exhibited impaired proangiogenic abilities. Treatment of P25 UCMSCs with rapamycin (900 nM) reversed the senescent phenotype and notably enhanced the proangiogenic activity of senescent UCMSCs. In a nude mouse model of hindlimb ischemia, intramuscular injection of rapamycin-treated P25 UCMSCs into the ischemic limb significantly promoted neovascularization and ischemic limb salvage. We further analyzed the changes in the expression of angiogenesis-associated genes in rapamycin-primed MSCs and found higher expression of several genes related to angiogenesis, such as VEGFR2 and CTGF/CCN2, in primed cells than in unprimed MSCs. Taken together, our data demonstrate that rapamycin is a potential drug to restore the proangiogenic activity of senescent MSCs, which is of importance in treating ischemic diseases and tissue engineering.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Mice , Animals , Limb Salvage , Hindlimb , Neovascularization, Physiologic , Sirolimus/pharmacology , Sirolimus/therapeutic use , Ischemia/therapy , Ischemia/metabolism , Mesenchymal Stem Cells/metabolism , Neovascularization, Pathologic/metabolism , Mice, Nude , Cells, Cultured
5.
Vaccines (Basel) ; 10(2)2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35214784

ABSTRACT

(1) Background: Although there are extensive data on admission co-variates and outcomes of persons with coronavirus infectious disease-2019 (COVID-19) at diverse geographic sites, there are few, if any, subject-level comparisons between sites in regions and countries. We investigated differences in hospital admission co-variates and outcomes of hospitalized people with COVID-19 between Wuhan City, China and the New York City region, USA. (2) Methods: We retrospectively analyzed clinical data on 1859 hospitalized subjects with COVID-19 in Wuhan City, China, from 20 January to 4 April 2020. Data on 5700 hospitalized subjects with COVID-19 in the New York City region, USA, from 1 March to 4 April 2020 were extracted from an article by Richardson et al. Hospital admission co-variates (epidemiological, demographic, and laboratory co-variates) and outcomes (rate of intensive care unit [ICU] admission, invasive mechanical ventilation [IMV], major organ failure and death, and length of hospital stay) were compared between the cohorts. (3) Results: Wuhan subjects were younger, more likely female, less likely to have co-morbidities and fever, more likely to have a blood lymphocyte concentration > 1 × 109/L, and less likely to have abnormal liver and cardiac function tests compared with New York subjects. There were outcomes data on all Wuhan subjects and 2634 New York subjects. Wuhan subjects had higher blood nadir median lymphocyte concentrations and longer hospitalizations, and were less likely to receive IMV, ICU hospitalization, and interventions for kidney failure. Amongst subjects not receiving IMV, those in Wuhan were less likely to die compared with New York subjects. In contrast, risk of death was similar in subjects receiving IMV at both sites. (4) Conclusions: We found different hospital admission co-variates and outcomes between hospitalized persons with COVID-19 between Wuhan City and the New York region, which should be useful developing a comprehensive global understanding of the SARS-CoV-2 pandemic and COVID-19.

6.
Sci Transl Med ; 13(578)2021 01 27.
Article in English | MEDLINE | ID: mdl-33504653

ABSTRACT

Stem cell senescence increases alongside the progressive functional declines that characterize aging. The effects of extracellular vesicles (EVs) are now attracting intense interest in the context of aging and age-related diseases. Here, we demonstrate that neonatal umbilical cord (UC) is a source of EVs derived from mesenchymal stem cells (MSC-EVs). These UC-produced MSC-EVs (UC-EVs) contain abundant anti-aging signals and rejuvenate senescing adult bone marrow-derived MSCs (AB-MSCs). UC-EV-rejuvenated AB-MSCs exhibited alleviated aging phenotypes and increased self-renewal capacity and telomere length. Mechanistically, UC-EVs rejuvenate AB-MSCs at least partially by transferring proliferating cell nuclear antigen (PCNA) into recipient AB-MSCs. When tested in therapeutic context, UC-EV-triggered rejuvenation enhanced the regenerative capacities of AB-MSCs in bone formation, wound healing, and angiogenesis. Intravenously injected UC-EVs conferred anti-aging phenotypes including decreased bone and kidney degeneration in aged mice. Our findings reveal that UC-EVs are of high translational value in anti-aging intervention.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Animals , Bone Marrow , Cellular Senescence , Mice , Proliferating Cell Nuclear Antigen
7.
Leukemia ; 34(9): 2384-2391, 2020 09.
Article in English | MEDLINE | ID: mdl-32690880

ABSTRACT

The impact of cancer on outcome of persons with coronavirus disease 2019 (COVID-19) after infection with acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is controversial. We studied 1859 subjects with COVID-19 from seven centers in Wuhan, China, 65 of whom had cancer. We found having cancer was an independent risk factor for in-hospital death from COVID-19 in persons <65 years (hazard ratio [HR] = 2.45, 95% confidence interval [CI], 1.04, 5.76; P = 0.041) but not in those ≥65 years (HR = 1.12 [0.56, 2.24]; P = 0.740). It was also more common in those not in complete remission. Risks of in-hospital death were similar in subjects with solid cancers and those with hematological cancers. These data may help predict outcomes of persons with cancer and COVID-19.


Subject(s)
Coronavirus Infections/mortality , Neoplasms/complications , Pneumonia, Viral/mortality , Adult , Age Factors , Aged , Betacoronavirus , COVID-19 , China , Coronavirus Infections/complications , Female , Hospital Mortality , Humans , Male , Middle Aged , Neoplasms/mortality , Pandemics , Pneumonia, Viral/complications , Remission Induction , Risk Factors , SARS-CoV-2
8.
Leukemia ; 34(8): 2163-2172, 2020 08.
Article in English | MEDLINE | ID: mdl-32528042

ABSTRACT

We studied admission and dynamic demographic, hematological and biochemical co-variates in 1449 hospitalized subjects with coronavirus infectious disease-2019 (COVID-19) in five hospitals in Wuhan, Hubei province, China. We identified two admission co-variates: age (Odds Ratio [OR] = 1.18, 95% Confidence Interval [CI] [1.02, 1.36]; P = 0.026) and baseline D-dimer (OR = 3.18 [1.48, 6.82]; P = 0.003) correlated with an increased risk of death in persons with COVID-19. We also found dynamic changes in four co-variates, Δ fibrinogen (OR = 6.45 [1.31, 31.69]; P = 0.022), Δ platelets (OR = 0.95 [0.90-0.99]; P = 0.029), Δ C-reactive protein (CRP) (OR = 1.09 [1.01, 1.18]; P = 0.037), and Δ lactate dehydrogenase (LDH) (OR = 1.03 [1.01, 1.06]; P = 0.007) correlated with an increased risk of death. The potential risk factors of old age, high baseline D-dimer, and dynamic co-variates of fibrinogen, platelets, CRP, and LDH could help clinicians to identify and treat subjects with poor prognosis.


Subject(s)
Betacoronavirus/isolation & purification , Biomarkers/blood , Coronavirus Infections/mortality , Hematologic Diseases/blood , Mortality/trends , Pneumonia, Viral/mortality , Adult , Aged , Aged, 80 and over , C-Reactive Protein/analysis , COVID-19 , Coronavirus Infections/blood , Coronavirus Infections/complications , Coronavirus Infections/diagnosis , Female , Fibrin Fibrinogen Degradation Products/analysis , Follow-Up Studies , Hematologic Diseases/diagnosis , Hematologic Diseases/virology , Humans , L-Lactate Dehydrogenase/blood , Lymphocyte Count , Lymphocytes/metabolism , Male , Middle Aged , Pandemics , Pneumonia, Viral/blood , Pneumonia, Viral/complications , Pneumonia, Viral/diagnosis , Prognosis , SARS-CoV-2 , Severity of Illness Index , Survival Rate
9.
Leukemia ; 34(8): 2173-2183, 2020 08.
Article in English | MEDLINE | ID: mdl-32546725

ABSTRACT

We studied 1859 subjects with confirmed COVID-19 from seven centers in Wuhan 1651 of whom recovered and 208 died. We interrogated diverse covariates for correlations with risk of death from COVID-19. In multi-variable Cox regression analyses increased hazards of in-hospital death were associated with several admission covariates: (1) older age (HR = 1.04; 95% Confidence Interval [CI], 1.03, 1.06 per year increase; P < 0.001); (2) smoking (HR = 1.84 [1.17, 2.92]; P = 0.009); (3) admission temperature per °C increase (HR = 1.32 [1.07, 1.64]; P = 0.009); (4) Log10 neutrophil-to-lymphocyte ratio (NLR; HR = 3.30 [2.10, 5.19]; P < 0.001); (5) platelets per 10 E + 9/L decrease (HR = 0.996 [0.994, 0.998]; P = 0.001); (6) activated partial thromboplastin (aPTT) per second increase (HR = 1.04 [1.02, 1.05]; P < 0.001); (7) Log10 D-dimer per mg/l increase (HR = 3.00 [2.17, 4.16]; P < 0.001); and (8) Log10 serum creatinine per µmol/L increase (HR = 4.55 [2.72, 7.62]; P < 0.001). In piecewise linear regression analyses Log10NLR the interval from ≥0.4 to ≤1.0 was significantly associated with an increased risk of death. Our data identify covariates associated with risk of in hospital death in persons with COVID-19.


Subject(s)
Betacoronavirus/isolation & purification , Biomarkers/blood , Coronavirus Infections/mortality , Lymphocytes/pathology , Mortality/trends , Neutrophils/pathology , Pneumonia, Viral/mortality , Severity of Illness Index , Adult , Aged , Aged, 80 and over , COVID-19 , Coronavirus Infections/blood , Coronavirus Infections/pathology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Pandemics , Pneumonia, Viral/blood , Pneumonia, Viral/pathology , Prognosis , ROC Curve , Risk Factors , SARS-CoV-2 , Survival Rate
10.
Curr Med Sci ; 38(1): 35-42, 2018 Feb.
Article in English | MEDLINE | ID: mdl-30074149

ABSTRACT

Low-dose cytarabine combined with differentiating or DNA hypomethylating agents, such as vitamin D compounds, is a potential regimen to treat acute myeloid leukemia (AML) patients who are unfit for high-intensity chemotherapy. The present study aimed to determine which subset of AML would be most responsive to low-dose cytarabine with the differentiating agent 1,25-dihydroxyvitamin D3 (1,25-D3). Here, firstly, cBioPortal database was used and we found out that vitamin D receptor (VDR) was highly expressed in acute monocytic leukemia (M5) and high VDR expression was associated with a poor survival of AML patients. Then, we confirmed that 1,25-D3 at clinical available concentration could induce more significant differentiation in acute monocytic leukemia cell lines (U937, MOLM-13, THP-1) and blasts from M5 patients than in non-monocytic cell lines (KGla and K562) and blasts from M2 patient. Finally, it was shown that the combination of 1,25-D3 and low-dose cytarabine further increased the differentiating rate, growth inhibition and G0/G1 arrest, while mild changes were found in the apoptosis in acute monocytic leukemia cell lines. Our study demonstrates that the enhanced response of acute monocytic leukemia cells to low-dose cytarabine by 1,25-D3 might indicate a novel therapeutic direction for patients with acute monocytic leukemia, especially for elderly and frail ones.


Subject(s)
24,25-Dihydroxyvitamin D 3/pharmacology , Antineoplastic Agents/pharmacology , Cytarabine/pharmacology , Leukemia, Monocytic, Acute/drug therapy , Vitamins/pharmacology , 24,25-Dihydroxyvitamin D 3/administration & dosage , 24,25-Dihydroxyvitamin D 3/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytarabine/administration & dosage , Cytarabine/therapeutic use , Drug Synergism , Humans , Vitamins/administration & dosage , Vitamins/therapeutic use
11.
Theranostics ; 7(10): 2673-2689, 2017.
Article in English | MEDLINE | ID: mdl-28819455

ABSTRACT

Senescence in human mesenchymal stem cells (MSCs) not only contributes to organism aging and the development of a variety of diseases but also severely impairs their therapeutic properties as a promising cell therapy. Studies searching for efficient biomarkers that represent cellular senescence have attracted much attention; however, no single marker currently provides an accurate cell-free representation of cellular senescence. Here, we studied characteristics of MSC-derived microvesicles (MSC-MVs) that may reflect the senescence in their parental MSCs. We found that senescent late passage (LP) MSCs secreted higher levels of MSC-MVs with smaller size than did early passage (EP) MSCs, and the level of CD105+ MSC-MVs decreased with senescence in the parental MSCs. Also, a substantially weaker ability to promote osteogenesis in MSCs was observed in LP than EP MSC-MVs. Comparative analysis of RNA sequencing showed the same trend of decreasing number of highly-expressed miRNAs with increasing number of passages in both MSCs and MSC-MVs. Most of the highly-expressed genes in LP MSCs and the corresponding MSC-MVs were involved in the regulation of senescence-related diseases, such as Alzheimer's disease. Furthermore, based on the miRNA profiling, transcription factors (TF) and genes regulatory networks of MSC senescence, and the datasets from GEO database, we confirmed that expression of miR-146a-5p in MSC-MVs resembled the senescent state of their parental MSCs. Our findings provide evidence that MSC-MVs are a key factor in the senescence-associated secretory phenotype of MSCs and demonstrate that their integrated characteristics can dynamically reflect the senescence state of MSCs representing a potential biomarker for monitoring MSC senescence.


Subject(s)
Aging/pathology , Biomarkers/analysis , Extracellular Vesicles/chemistry , Mesenchymal Stem Cells/physiology , MicroRNAs/analysis , Cells, Cultured , Humans
12.
J Huazhong Univ Sci Technolog Med Sci ; 37(2): 179-184, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28397044

ABSTRACT

Increasing studies have demonstrated that interferon gamma (IFN-γ), which serves as a critical inflammatory cytokine, is essential to induce the immunosuppressive effects of mesenchymal stem cells (MSCs). However, the mechanisms underlying the enhanced immunosuppressive effects of IFN-γ-stimulated MSCs (γMSCs) are not fully understood. MSC-derived microvesicles (MSC-MVs) have been viewed as potential pivotal mediators of the immunosuppressive effects of MSCs. Moreover, microRNAs (miRNAs) are important regulators of immunological processes and can be shuttled from cell to cell by MVs. The aim of our study was to analyze the the miRNA expression signature of MVs derived from γMSCs (γMSC-MVs), which may provide better understanding of the immunosuppressive property of their parent cells. Through miRNA microarray and bioinformatics analysis, we found 62 significantly differentially expressed miRNAs (DEMs) in γMSC-MVs compared with MSC-MVs. And the potential target genes and signaling pathways regulated by DEMs were predicted and analyzed. Interestingly, many DEMs and predicted signaling pathways had been demonstrated to be involved in immunoregulation. Furthermore, the network between immunoregulation-related pathways and relevant DEMs was constructed. Collectively, our research on the miRNA repertoires of γMSC-MVs not only provides new perspectives into the mechanisms underlying the enhanced immunosuppressive property of γMSCs, but also paves the way to clinical application of these potent organelles in the future.


Subject(s)
Cell-Derived Microparticles/genetics , Interferon-gamma/pharmacology , Mesenchymal Stem Cells/immunology , MicroRNAs/genetics , Cell-Derived Microparticles/drug effects , Cells, Cultured , Computational Biology/methods , Gene Expression Regulation/drug effects , Gene Regulatory Networks , Humans , Mesenchymal Stem Cells/drug effects , Oligonucleotide Array Sequence Analysis , Signal Transduction
13.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-238378

ABSTRACT

Increasing studies have demonstrated that interferon gamma (IFN-γ),which serves as a critical inflammatory cytokine,is essential to induce the immunosuppressive effects of mesenchymal stem cells (MSCs).However,the mechanisms underlying the enhanced immunosuppressive effects of IFN-γ-stimulated MSCs (γMSCs) are not fully understood.MSC-derived rnicrovesicles (MSC-MVs) have been viewed as potential pivotal mediators of the immunosuppressive effects of MSCs.Moreover,microRNAs (miRNAs) are important regulators of immunological processes and can be shuttled from cell to cell by MVs.The aim of our study was to analyze the the miRNA expression signature of MVs derived from γMSCs (γMSC-MVs),which may provide better understanding of the immunosuppressive property of their parent cells.Through miRNA microarray and bioinformatics analysis,we found 62 significantly differentially expressed miRNAs (DEMs) in γMSC-MVs compared with MSC-MVs.And the potential target genes and signaling pathways regulated by DEMs were predicted and analyzed.Interestingly,many DEMs and predicted signaling pathways had been.demonstrated to be involved in immunoregulation.Furthermore,the network between immunoregulation-related pathways and relevant DEMs was constructed.Collectively,our research on the miRNA repertoires of γMSC-MVs not only provides new perspectives into the mechanisms underlying the enhanced immunosuppressive property of γMSCs,but also paves the way to clinical application of these potent organelles in the future.

14.
J Huazhong Univ Sci Technolog Med Sci ; 35(3): 343-349, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26072071

ABSTRACT

This study examined the mechanism of the inhibitory effect of parthenolide (PTL) on the activity of NF-κB in multiple myeloma (MM). Human multiple myeloma cell line RPMI 8226 cells were treated with or without different concentrations of PTL for various time periods, and then MTT assay was used to detect cell proliferation. Cell cycle and apoptosis were flow cytometrically detected. The level of protein ubiquitination was determined by using immunoprecipitation. Western blotting was employed to measure the level of total protein ubiquitination, the expression of IκB-α in cell plasma and the content of p65 in nucleus. The content of p65 in nucleus before and after PTL treatment was also examined with immunofluorescence. Exposure of RPMI 8226 cells to PTL attenuated the level of ubiquitinated Nemo, increased the expression of IκB-α and reduced the level of p65 in nucleus, finally leading to the decrease of the activity of NF-κB. PTL inhibited cell proliferation, induced apoptosis and blocked cell cycle. Furthermore, the levels of ubiquitinated tumor necrosis factor receptor-associated factor 6 (TRAF6) and total proteins were decreased after PTL treatment. By using Autodock software package, we predicted that PTL could bind to TRAF6 directly and tightly. Taken together, our findings suggest that PTL inhibits the activation of NF-κB signaling pathway via directly binding with TRAF6, thereby suppressing MM cell proliferation and inducing apoptosis.


Subject(s)
Multiple Myeloma/metabolism , NF-kappa B/antagonists & inhibitors , Sesquiterpenes/pharmacology , TNF Receptor-Associated Factor 6/metabolism , Apoptosis , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Multiple Myeloma/drug therapy , NF-kappa B/blood , Transcription Factor RelA/metabolism , Ubiquitination/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...