Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 785
Filter
Add more filters

Complementary Medicines
Publication year range
1.
BMC Complement Med Ther ; 24(1): 28, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38195460

ABSTRACT

BACKGROUND: Indigofera suffruticosa Mill. is used as a folk medicine for treating patients with leukemia, however very little is known regarding the molecular mechanism of its anti-leukemic activity and the chemical profile of the active extract. The present study aimed to reveal the molecular effect of I. suffruticosa aerial parts extract (ISAE) on leukemia cells and its chemical constituents. METHODS: Cytotoxicity of ISAE were determined by resazurin viability assay, multitox - Glo multiplex cytotoxicity assay, and Annexin V staining assay. Cell cycle profiles were revealed by propidium iodide staining assay. The effects of ISAE on G2/M arrest signaling and DNA damage were evaluated by Western blot assay and phospho-H2A.X staining assay. The chemical profile of ISAE were determined by tandem mass spectroscopy and molecular networking approach. RESULTS: We showed that the acute lymphoblastic leukemia cell line Jurkat cell was more responsive to ISAE treatment than other leukemia cell lines. In contrast, ISAE did not induce cytotoxic effects in normal fibroblast cells. Cell cycle analysis revealed that ISAE triggered G2/M arrest in Jurkat cells in dose- and time-dependent manners. Elevation of annexin V-stained cells and caspase 3/7 activity suggested ISAE-induced apoptosis. Furthermore, ISAE alone could increase the phosphorylation of CDK1 at Y15 and activate the ATR/CHK1/Wee1/CDC25C signaling pathway. However, the addition of caffeine, a widely used ATR inhibitor to ISAE, reduced the phosphorylation of ATR, CHK1, and CDK1, as well as G2/M arrest in Jurkat cells. Moreover, increased phospho-H2A.X stained cells indicated the involvement of DNA damage in the anti-leukemic effect of ISAE. Finally, qualitative analysis using UPLC-tandem mass spectroscopy and molecular networking revealed that tryptanthrin was the most abundant organoheterocyclic metabolite in ISAE. At equivalent concentrations to ISAE, tryptanthrin induced G2/M arrest of Jurkat cells, which can be prevented by caffeine. CONCLUSIONS: ISAE causes G2/M arrest via activating ATR/CHK1/CDK1 pathway and tryptanthrin is one of the active components of ISAE. Our findings provide subtle support to the traditional use of I. suffruitcosa in leukemia management in folk medicine.


Subject(s)
Indigofera , Leukemia , Humans , Jurkat Cells , Annexin A5 , Apoptosis , Caffeine , Cell Line, Tumor , G2 Phase Cell Cycle Checkpoints , Plant Components, Aerial , Plant Extracts/pharmacology , Ataxia Telangiectasia Mutated Proteins
2.
Bioorg Med Chem ; 91: 117408, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37453188

ABSTRACT

Infection with the retrovirus human T-cell leukemia virus type 1 (HTLV-1) sometimes causes diseases that are difficult to cure. To find anti-HTLV-1 natural compounds, we opted to screen using the HTLV-1-infected T-cell line, MT-2. Based on our results, an extract of the pulp/seeds of Akebia quinata Decaisne fruit killed MT-2 cells but did not affect the Jurkat cell line that was not infected with virus. To determine the active ingredients, seven saponins with one-six sugar moieties were isolated from A. quinata seeds, and their activities against the two cell lines were examined. Both cell lines were killed in a similar manner by Akebia saponins A and B. Further, Akebia saponins D, E, PK and G did not exhibit cytotoxicity. Akebia saponin C had a similar activity to the extract found in the screening. This compound was found to enhance Gag aggregation, induce the abnormal cleavage of Gag, suppress virion release, and preferentially kill HTLV-1 infected cells; however, their relationship remains elusive. Our findings may lead to the development of new therapies for infectious diseases based on the removal of whole-virus-infected cells.


Subject(s)
Human T-lymphotropic virus 1 , Saponins , Humans , Cell Line , Saponins/pharmacology , Jurkat Cells , Plant Extracts
3.
J Biol Chem ; 299(8): 105009, 2023 08.
Article in English | MEDLINE | ID: mdl-37406814

ABSTRACT

Selenoprotein P (SeP, encoded by the SELENOP gene) is a plasma protein that contains selenium in the form of selenocysteine residues (Sec, a cysteine analog containing selenium instead of sulfur). SeP functions for the transport of selenium to specific tissues in a receptor-dependent manner. Apolipoprotein E receptor 2 (ApoER2) has been identified as a SeP receptor. However, diverse variants of ApoER2 have been reported, and the details of its tissue specificity and the molecular mechanism of its efficiency remain unclear. In the present study, we found that human T lymphoma Jurkat cells have a high ability to utilize selenium via SeP, while this ability was low in human rhabdomyosarcoma cells. We identified an ApoER2 variant with a high affinity for SeP in Jurkat cells. This variant had a dissociation constant value of 0.67 nM and a highly glycosylated O-linked sugar domain. Moreover, the acidification of intracellular vesicles was necessary for selenium transport via SeP in both cell types. In rhabdomyosarcoma cells, SeP underwent proteolytic degradation in lysosomes and transported selenium in a Sec lyase-dependent manner. However, in Jurkat cells, SeP transported selenium in Sec lyase-independent manner. These findings indicate a preferential selenium transport pathway involving SeP and high-affinity ApoER2 in a Sec lyase-independent manner. Herein, we provide a novel dynamic transport pathway for selenium via SeP.


Subject(s)
Lyases , Selenium , Humans , Lyases/metabolism , Selenium/metabolism , Selenocysteine/genetics , Selenocysteine/metabolism , Selenoprotein P/genetics , Selenoprotein P/metabolism , Selenoproteins , Jurkat Cells
4.
Pharm Biol ; 61(1): 259-270, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36656546

ABSTRACT

CONTEXT: Due to the poor prognosis of T-cell acute lymphoblastic leukaemia (T-ALL), there is an urgent need to identify safer and more cost-effective drugs. OBJECTIVE: This study evaluated the antitumour activity of Shuanghuanglian (SHL) on T-ALL cells and elucidated the mechanism. MATERIALS AND METHODS: Jurkat and Molt4 cells were treated with SHL (0.1, 0.2 and 0.4 mg/mL) for 24 and 48 h. The controls were treated with RPMI 1640 containing 10% foetal bovine serum. Cell viability was evaluated through Cell Counting Kit-8 assay. Patterns of death and signalling pathway alterations caused by SHL were identified by network pharmacology combined with GO enrichment analysis and then were verified by Hoechst 33342 staining, Annexin V-FITC/PI staining and Western blotting. Interactions of the active ingredients with targets were analysed by molecular docking. RESULTS: The IC50 values of SHL in Jurkat and Molt4 cells were 0.30 ± 0.10 and 0.48 ± 0.07 mg/mL, respectively, at 24 h and 0.27 ± 0.05 and 0.30 ± 0.03 mg/mL at 48 h. In T-ALL, 117 target genes of SHL were mainly enriched in the apoptosis and NOTCH signalling pathways. SHL induced apoptosis was confirmed by Hoechst 33342 staining and flow cytometry. The protein levels of cleaved caspase-7 and cleaved PARP were significantly increased but those of cleaved NOTCH1 and MYC were reduced. The active ingredients of SHL can interact with γ-secretase.Discussion and conclusions: SHL induces apoptosis in T-ALL cells via the NOTCH1-MYC pathway and may be a potential drug for the treatment of T-ALL.


Subject(s)
Drugs, Chinese Herbal , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Apoptosis , Molecular Docking Simulation , Network Pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Drugs, Chinese Herbal/pharmacology , Jurkat Cells
5.
J Ethnopharmacol ; 305: 116120, 2023 Apr 06.
Article in English | MEDLINE | ID: mdl-36610674

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The genus Eryngium is known for producing a wide range of bioactive compounds with proved medicinal properties. In the last years, research has focused on E. maritimum, with previous studies reporting anticancer, antimicrobial, antioxidant, and anti-inflammatory activities. Ethnobotanical literature suggests that it has been traditionally used to treat a wide range of illnesses, having antitussive, diuretic and aphrodisiac properties. Being rhizome one of the most bioactive organs, much of the available references from traditional uses suggest that it has been specifically used to treat renal diseases. In this sense, inflammation and oxidative processes play a major role in kidney dysfunctions, which could be associated to the mechanism of action of the plant extracts. AIM OF THE STUDY: The main aim of the study was to investigate the effects of E. maritimum rhizome extract on the antioxidant and inflammatory response in human immune cells. MATERIAL AND METHODS: Rhizome extracts were obtained from plants growing in Mallorca (Balearic Islands), and its composition was determined using HPLC-DAD, highlighting simple phenolic compounds such as trans-ferulic acid, catechin, chlorogenic acid, epicatechin and rosmarinic acid as the major constituents. Total antioxidant capacity was determined using the FRAP assay. Jurkat cells were cultured to analyse cytotoxicity by cell viability assay. In parallel, cells were stimulated with phytohemagglutinin and treated with different extract concentrations. Gene and protein expression, as well as nitrite and cytokine levels were evaluated as indicators of metabolic responses. RESULTS: The plant extract showed a high diversity of pharmacologically bioactive compounds with potential therapeutic uses. The extract presented null cytotoxicity and exerted antioxidant and anti-inflammatory effects on Jurkat cells by inducing an antioxidant response and reducing cytokine and nitric oxide release and the expression of pro-inflammatory genes. CONCLUSION: The present findings suggest that E. maritimum is a promising phytotherapeutic species because of its strong antioxidant and anti-inflammatory potential, which could explain some of its traditional uses.


Subject(s)
Antioxidants , Eryngium , Humans , Antioxidants/pharmacology , Rhizome , Jurkat Cells , Anti-Inflammatory Agents/pharmacology , Plant Extracts/pharmacology
6.
Chem Biol Interact ; 367: 110178, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36113632

ABSTRACT

Selenium is an essential trace element. High dosage of selenite exhibits a great potential in treating leukemia. Previous study discovered selenite could promote leukemia cells apoptosis through inducing DNA damage and cell cycle arrest, while the switch mechanisms of these events and autophagy were still unclear. Current study discovered selenite promoted autophagy and apoptosis of leukemia Jurkat cells. In this process, DNA damage related ATM/IKK alpha axis was activated. This axis could stabilize pro-apoptotic P73, and promote autophagy through regulating NF-kappaB signaling pathway. Moreover, survivin-2B was also confirmed to be necessary for the ATM-induced nuclear location of IKK alpha, and therefore stood at the node position of apoptosis and autophagy cascades inside Jurkat cells. Finally, our in vivo experiments proved that selenite exhibited some anti-tumor effects on Jurkat cells-bearing mice. Moreover, alterations of ATM and IKK alpha expression observed in vivo were similar to that identified in vitro. Therefore, our findings had fully confirmed survivin-2B dependent activation of ATM/IKK alpha axis might be another crosstalk between autophagy and apoptosis of selenite-treated leukemia cells.


Subject(s)
Leukemia , Selenium , Trace Elements , Animals , Apoptosis , Ataxia Telangiectasia Mutated Proteins/metabolism , Autophagy , Humans , I-kappa B Kinase/metabolism , Jurkat Cells , Leukemia/pathology , Mice , NF-kappa B/metabolism , Selenious Acid/metabolism , Selenious Acid/pharmacology , Selenium/pharmacology , Survivin/metabolism , Trace Elements/metabolism
7.
Int J Mol Sci ; 23(3)2022 Jan 26.
Article in English | MEDLINE | ID: mdl-35163318

ABSTRACT

The infection of CD4 T-lymphocytes with human immunodeficiency virus (HIV), the etiological agent of acquired immunodeficiency syndrome (AIDS), disrupts cellular homeostasis, increases oxidative stress and interferes with micronutrient metabolism. Viral replication simultaneously increases the demand for micronutrients and causes their loss, as for selenium (Se). In HIV-infected patients, selenium deficiency was associated with a lower CD4 T-cell count and a shorter life expectancy. Selenium has an important role in antioxidant defense, redox signaling and redox homeostasis, and most of these biological activities are mediated by its incorporation in an essential family of redox enzymes, namely the selenoproteins. Here, we have investigated how selenium and selenoproteins interplay with HIV infection in different cellular models of human CD4 T lymphocytes derived from established cell lines (Jurkat and SupT1) and isolated primary CD4 T cells. First, we characterized the expression of the selenoproteome in various human T-cell models and found it tightly regulated by the selenium level of the culture media, which was in agreement with reports from non-immune cells. Then, we showed that selenium had no significant effect on HIV-1 protein production nor on infectivity, but slightly reduced the percentage of infected cells in a Jurkat cell line and isolated primary CD4 T cells. Finally, in response to HIV-1 infection, the selenoproteome was slightly altered.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , HIV Infections/metabolism , HIV Infections/virology , HIV-1/metabolism , Selenium/metabolism , Selenoproteins/metabolism , Virus Replication/physiology , Acquired Immunodeficiency Syndrome/metabolism , Antioxidants/metabolism , Cell Line, Tumor , Glutathione Peroxidase/metabolism , HEK293 Cells , Humans , Jurkat Cells , Oxidative Stress/physiology
8.
Biochem Pharmacol ; 197: 114940, 2022 03.
Article in English | MEDLINE | ID: mdl-35120895

ABSTRACT

Programmed death ligand-1 (PD-L1) and indoleamine 2, 3-dioxygenase 1 (IDO1) are immune checkpoints induced by interferon-γ (IFN-γ) in the tumor microenvironment, leading to immune escape of tumors. Myricetin (MY) is a flavonoid distributed in many edible and medicinal plants. In this study, MY was identified to inhibit IFN-γ-induced PD-L1 expression in human lung cancer cells. It also reduced the expression of IDO1 and the production of kynurenine which is the product catalyzed by IDO1, while didn't show obvious effect on the expression of major histocompatibility complex-I (MHC-I), a crucial molecule for antigen presentation. In addition, the function of T cells was evaluated using a co-culture system consist of lung cancer cells and the Jurkat-PD-1 T cell line overexpressing PD-1. MY restored the survival, proliferation, CD69 expression and interleukin-2 (IL-2) secretion of Jurkat-PD-1 T cells suppressed by IFN-γ-treated lung cancer cells. Mechanistically, IFN-γ up-regulated PD-L1 and IDO1 at the transcriptional level through the JAK-STAT-IRF1 axis, which was targeted and inhibited by MY. Together, our research revealed a new mechanism of MY mediated anti-tumor activity and highlighted the potential implications of MY in tumor immunotherapy.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Flavonoids/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Interferon-gamma/pharmacology , Lung Neoplasms/metabolism , A549 Cells , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/genetics , Coculture Techniques , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/physiology , HCT116 Cells , HEK293 Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Jurkat Cells , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology
9.
Biomed Pharmacother ; 147: 112577, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35078092

ABSTRACT

Cowanin, a xanthone derivative extracted from the Garcinia fusca plant, has been recognized for various biological activities including, antimicrobial, anti-inflammatory, and anticancer activities. However, the mechanism to induce cancer cell death in cancer cells remains to be fully elucidated. Our previous report showed that other xanthones from these plants could act as histone deacetylase inhibitors (HDACi), so we deeply analyzed the role of cowanin, a major compound of G.fusca, and investigated through the mode of cell death both apoptosis and autophagy that have never been reported. As a result, it was demonstrated that cowanin indicated the role of HDACi as other xanthones. The molecular docking analysis showed that cowanin could interact within the catalytic pocket region of HDAC class I (HDAC2, 8) and II (HDAC4, 7) proteins and inhibit their activity. Also, the level of protein expression of HDAC2, 4, 7, and 8 was distinctly decreased, and the level of histone H3 and H4 acetylation increased in cowanin treated cells. For the mode of cell death, cowanin demonstrated both apoptosis and autophagy activation in Jurkat cells. Besides, cowanin significantly suppressed phosphorylation of PI3K, Akt, and mTOR signaling. Therefore, these findings revealed that cowanin represents a new promising candidate for development as an anticancer agent by inducing apoptosis and autophagy via PI3K/AKT/mTOR pathway and effectively inhibiting HDAC activity.


Subject(s)
Garcinia , Histone Deacetylase Inhibitors , Plant Extracts , Humans , Apoptosis/drug effects , Autophagy/drug effects , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Jurkat Cells/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phytotherapy , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
10.
Anticancer Res ; 42(1): 547-554, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34969764

ABSTRACT

BACKGROUND/AIM: This study analysed the effect of α-tocopheryl succinate (α-TS) on the redox-state of leukemia and normal lymphocytes, as well as their sensitization to fifteen anticancer drugs. MATERIALS AND METHODS: Cell viability was analyzed by trypan blue staining and automated counting of live and dead cells. Apoptosis was analyzed by FITC-Annexin V test. Oxidative stress was evaluated by the intracellular levels of reactive oxygen species (ROS) and protein-carbonyl products. RESULTS: Most combinations (α-TS plus anticancer drug) exerted additive or antagonistic effects on the proliferation and viability of leukemia lymphocytes. α-TS combined with barasertib, bortezomib or lonafarnib showed a strong synergistic cytotoxic effect, which was best expressed in the case of barasestib. It was accompanied by impressive induction of apoptosis and increased production of ROS, but insignificant changes in protein-carbonyl levels. α-TS plus barasertib did not alter the viability and did not induce oxidative stress and apoptosis in normal lymphocytes. CONCLUSION: α-TS could be a promising adjuvant in second-line anticancer therapy, particularly in acute lymphoblastic leukemia, to reduce the therapeutic doses of barasertib, bortezomib, and lonafarnib, increasing their effectiveness and minimizing their side effects.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Leukemia/drug therapy , alpha-Tocopherol/pharmacology , Apoptosis/drug effects , Cell Survival/drug effects , Humans , Jurkat Cells/drug effects , Leukemia/genetics , Leukemia/pathology , Lymphocytes/drug effects , Lymphocytes/pathology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species , Succinates/pharmacology
11.
Nat Commun ; 12(1): 7344, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34937876

ABSTRACT

Manipulating lymphocyte functions with gene silencing approaches is promising for treating autoimmunity, inflammation, and cancer. Although oligonucleotide therapy has been proven to be successful in treating several conditions, efficient in vivo delivery of oligonucleotide to lymphocyte populations remains a challenge. Here, we demonstrate that intravenous injection of a heteroduplex oligonucleotide (HDO), comprised of an antisense oligonucleotide (ASO) and its complementary RNA conjugated to α-tocopherol, silences lymphocyte endogenous gene expression with higher potency, efficacy, and longer retention time than ASOs. Importantly, reduction of Itga4 by HDO ameliorates symptoms in both adoptive transfer and active experimental autoimmune encephalomyelitis models. Our findings reveal the advantages of HDO with enhanced gene knockdown effect and different delivery mechanisms compared with ASO. Thus, regulation of lymphocyte functions by HDO is a potential therapeutic option for immune-mediated diseases.


Subject(s)
Lymphocytes/metabolism , Nucleic Acid Heteroduplexes/metabolism , Oligonucleotides/metabolism , RNA/metabolism , Administration, Intravenous , Adoptive Transfer , Animals , Demyelinating Diseases/genetics , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Endocytosis/drug effects , Female , Gene Expression Regulation , Gene Silencing , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Humans , Integrin alpha4/genetics , Integrin alpha4/metabolism , Jurkat Cells , Male , Mice, Inbred C57BL , Nucleic Acid Heteroduplexes/administration & dosage , Nucleic Acid Heteroduplexes/pharmacokinetics , Nucleic Acid Heteroduplexes/pharmacology , Oligonucleotides/administration & dosage , Oligonucleotides/pharmacokinetics , Oligonucleotides/pharmacology , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spinal Cord/pathology , Tissue Distribution/drug effects
12.
Front Immunol ; 12: 737076, 2021.
Article in English | MEDLINE | ID: mdl-34659228

ABSTRACT

Immune checkpoints such as programmed death-1 (PD-1) have been proven as antitumor targets by enhancing cytotoxic T cell activity. All immune checkpoint blockades are antibody therapeutics that have large size and high affinity, as well as known immune-related side effects and low responses. To overcome the limitation of antibody therapeutics, we have explored PD-1/PD-L1 (programmed death-ligand 1) blockades in traditional oriental medicine, which has a long history but has not yet studied PD-1/PD-L1 blockades. Sanguisorbae Radix extract (SRE) blocked PD-1 and PD-L1 binding in competitive ELISA. SRE effectively inhibited the PD-1/PD-L1 interaction, thereby improving T cell receptor (TCR) signaling and the NFAT-mediated luciferase activity of T cells. SRE treatment reduced tumor growth in the humanized PD-L1 MC38 cell allograft humanized PD-1 mouse model. Additionally, the combination of SRE and pembrolizumab (anti-PD-1 antibody) suppressed tumor growth and increased infiltrated cytotoxic T cells to a greater extent did either agent alone. This study showed that SRE alone has anticancer effects via PD-1/PD-L1 blockade and that the combination therapy of SRE and pembrolizumab has enhanced immuno-oncologic effects.


Subject(s)
Adenocarcinoma/drug therapy , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/drug effects , Colorectal Neoplasms/drug therapy , Immune Checkpoint Inhibitors/pharmacology , Lymphocytes, Tumor-Infiltrating/drug effects , Plant Extracts/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Sanguisorba , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CHO Cells , Coculture Techniques , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cricetulus , Humans , Jurkat Cells , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Plant Extracts/isolation & purification , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Sanguisorba/chemistry , Signal Transduction , Tumor Burden
13.
Fitoterapia ; 155: 105053, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34610355

ABSTRACT

It is commonly known that radiotherapy is still a key modality for treatment of cancer. Though this effect is desirable during radiotherapy, it leads to radiotoxicity on normal healthy cells. In the present research, we designed, synthesized and analyzed a series of nitronyl nitroxide radical (NITR) spin-labeled resveratrol (RES) derivatives. The cytotoxicity of the newly synthesized substances was tested on Jurkat T cells. The derivatives were studied as reactive oxygen species (ROS) scavenger to protect ionizing radiation of Jurkat T cells upon 6 Gy X-irradiation. The experimental results revealed that compound 2 and 3 could significantly alleviate the damage of Jurkat T cells, as evidenced by decreasing ROS production and restoring the cell apoptosis. Further mechanism investigations indicated that the radioprotective effects of the novel derivatives were largely associated with modulating the expression of apoptotic proteins including cIAP-1, cIAP-2, cytochrome c, caspase-3 and caspase-9. Based on the experimental result, we disclosed that the novel NITR spin-labeled RES derivatives exhibit the potential to be used as the novel radioprotective candidates to ameliorate the injury induced by ionizing radiation.


Subject(s)
Apoptosis/drug effects , Nitrogen Oxides/pharmacology , Radiation-Protective Agents/pharmacology , Resveratrol/pharmacology , Antioxidants/pharmacology , Humans , Jurkat Cells , Molecular Structure , Radiation, Ionizing , Reactive Oxygen Species/metabolism , Resveratrol/analogs & derivatives , Spin Labels
14.
Phytomedicine ; 93: 153796, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34689117

ABSTRACT

BACKGROUND: Bavachin, a flavonoid compound isolated from the seeds and fruits of Psoralea corylifolia l. (family Fabaceae), is used as a traditional medicine in Asia. Indeed, it is reported to have various medicinal functions such as estrogenic and antiinflammatory activities among others. However, to date, the effects of bavachin on T cell activation have yet to be reported. PURPOSE AND STUDY DESIGN: We aimed to determine the effects of bavachin on the activation of a human T cell line in vitro and on antigen-specific immune responses in mice in vivo. METHODS: In a nuclear factor of activated T cells (NFAT) activity assay, the Jurkat T cell line expressing a luciferase reporter driven by an NFAT-response element was stimulated with antihuman CD3/CD28 antibody and bavachin. Furthermore, the level of cytokine production was measured in the Jurkat T cell line stimulated with phorbol 12-myristate 13-acetate/ionomycin and bavachin using an IL-2 ELISA and a cytometric bead array assay. For in vivo analyses, mice were subcutaneously immunized with an antigen (ovalbumin protein) and bavachin, and the immune responses of mice were analyzed by FACS analysis, a T cell proliferation assay, a cytokine ELISA, and an antiovalbumin-specific antibody ELISA. RESULTS: We found that bavachin activated NFAT-mediated transcription in the human T cell line in vitro. In mice, when bavachin was administered with the antigen, an increase in T cell responses and antibody production specific to the antigen was observed. CONCLUSION: Our results suggest that bavachin has immunoadjuvant and immunomodulation effects, which arise through activation of the NFAT signaling pathway.


Subject(s)
Adjuvants, Immunologic , NFATC Transcription Factors , Animals , Flavonoids , Humans , Interleukin-2 , Jurkat Cells , Mice , Signal Transduction
15.
Commun Biol ; 4(1): 1123, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34556786

ABSTRACT

The highly cytotoxic marine natural product callyspongiolide holds great promise as a warhead of antibody-drug conjugate in cancer therapeutics; however, the mechanism underlying its cytotoxicity remains unclear. To elucidate how callyspongiolide kills cells, we employed label-free target identification with thermal stability-shift-based fluorescence difference in two-dimensional (2-D) gel electrophoresis (TS-FITGE), which allowed observation of a unique phenomenon of protein-spot separation on 2-D gels upon treatment with callyspongiolide at increasing temperatures. During our exploration of what proteins were associated with this phenomenon as well as why it happens, we found that callyspongiolide induces mitochondrial/lysosomal dysfunction and autophagy inhibition. Moreover, molecular biology studies revealed that callyspongiolide causes lysosomal dysfunction, which induces cellular iron depletion and leads to mitochondrial dysfunction and subsequent cytotoxicity. Notably, these effects were rescued through iron supplementation. Although our approach was unable to reveal the direct protein targets of callyspongiolide, unique phenomena observed only by TS-FITGE provided critical insight into the mechanism of action of callyspongiolide and specifically its cytotoxic activity via induction of mitochondrial dysfunction through cellular iron depletion caused by lysosomal deacidification, which occurred independent of known programmed cell death pathways.


Subject(s)
Antineoplastic Agents/pharmacology , Iron/metabolism , Macrolides/pharmacology , Mitochondria/metabolism , A549 Cells , HCT116 Cells , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Jurkat Cells , MCF-7 Cells , PC-3 Cells
16.
Mol Biol Rep ; 48(9): 6249-6258, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34478011

ABSTRACT

BACKGROUND: Britannin, a Sesquiterpene Lactone isolated from Inula aucheriana, has recently gained attraction in the therapeutic fields due to its anti-tumor properties. This study was designed to evaluate the effect of this agent on Acute Lymphoblastic Leukemia (ALL) cell lines, either as a monotherapy or in combination with Vincristine (VCR). METHODS AND RESULTS: To determine the anti-leukemic effects of Britannin on ALL-derived cell lines and suggest a mechanism of action for the agent, we used MTT assay, Annexin-V/PI staining, ROS assay, and real-time PCR analysis. Moreover, by using a combination index (CI), we evaluated the synergistic effect of Britannin on Vincristine. We found that unlike normal Peripheral Blood Mononuclear Cells (PBMCs) and L929 cells, Britannin reduced the viability of NALM-6, REH, and JURKAT cells. Among tested cells, NALM-6 cells had the highest sensitivity to Britannin, and this agent was able to induce p21/p27-mediated G1 cell cycle arrest and Reactive Oxygen Specious (ROS)-mediated apoptotic cell death in this cell line. When NALM-6 cells were treated with Nacetyl-L-Cysteine (NAC), a scavenger of ROS, Britannin could induce neither apoptosis nor reduce the survival of the cells suggesting that the cytotoxic effect of Britannin is induced through ROS-dependent manner. Moreover, we found that a low dose of Britannin enhanced the effect of Vincristine in NALM-6 cells by inducing apoptotic cell death via altering the expression of apoptotic-related genes. CONCLUSIONS: Overall, our results proposed a mechanism for the cytotoxic effect of Britannin, either as a single agent or in combination with Vincristine, in NALM-6 cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Catharanthus/chemistry , Inula/chemistry , Lactones/pharmacology , Phytochemicals/pharmacology , Plant Extracts/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Reactive Oxygen Species/metabolism , Sesquiterpenes/pharmacology , Vincristine/pharmacology , Acetylcysteine/pharmacology , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Survival/drug effects , Drug Synergism , Free Radical Scavengers/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Jurkat Cells , Lactones/isolation & purification , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Phytochemicals/isolation & purification , Plant Extracts/isolation & purification , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Sesquiterpenes/isolation & purification , Signal Transduction/drug effects
17.
Sci Rep ; 11(1): 18086, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34508147

ABSTRACT

Leukotoxin (LtxA) (Trade name, Leukothera) is a protein that is secreted from the oral bacterium Aggregatibacter actinomycetemcomitans, which targets and kills activated white blood cells (WBCs) by binding to lymphocyte function associated antigen-1 (LFA-1). Interaction between LtxA and Jurkat T-cells results in cell death and is characterized by increased intracellular Ca2+, activation of caspases, clustering of LtxA and LFA-1 within lipid rafts, and involvement of the Fas death receptor. Here, we show that LtxA can kill malignant lymphocytes via apoptotic and necrotic forms of cell death. We show that LtxA causes activation of caspases and PARP, cleavage of pannexin-1 (Panx1) channels, and expulsion of ATP, ultimately leading to cell death via apoptosis and necrosis. CRISPR-Cas9 mediated knockout (K/O) of Panx1 in Jurkat cells prevented ATP expulsion and resulted in resistance to LtxA for both apoptotic and necrotic forms of death. Resistance to necrosis could only be overcome when supplementing LtxA with endogenous ATP (bzATP). The combination of LtxA and bzATP promoted only necrosis, as no Panx1 K/O cells stained positive for phosphatidylserine (PS) exposure following the combined treatment. Inhibition of LtxA/bzATP-induced necrosis was possible when pretreating Jurkat cells with oATP, a P2X7R antagonist. Similarly, blockage of P2X7Rs with oATP prevented the intracellular mobilization of Ca2+, an important early step in LtxA induced cell death. We show that LtxA is able to kill malignant lymphocytes through an apoptotic death pathway which is potentially linked to a Panx1/P2X7R mediated necrotic form of death. Thus, inhibition of ATP release appears to significantly delay the onset of LtxA induced apoptosis while completely disabling the necrotic death pathway in T-lymphocytes, demonstrating the crucial role of ATP release in LtxA-mediated cell death.


Subject(s)
Connexins/metabolism , Exotoxins/metabolism , Lymphocytes/metabolism , Nerve Tissue Proteins/metabolism , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Calcium/metabolism , Cell Death , Cell Membrane/drug effects , Cell Membrane/metabolism , Connexins/deficiency , Exotoxins/pharmacology , Gene Knockdown Techniques , Humans , Jurkat Cells , Leukemia, Lymphoid/etiology , Leukemia, Lymphoid/metabolism , Leukemia, Lymphoid/pathology , Lymphocytes/pathology , Lymphoma/etiology , Lymphoma/metabolism , Lymphoma/pathology , Nerve Tissue Proteins/deficiency , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/metabolism , Signal Transduction/drug effects
18.
Exp Hematol ; 101-102: 58-67, 2021 09.
Article in English | MEDLINE | ID: mdl-34450221

ABSTRACT

Huaier, a traditional Chinese medicine, is currently used to treat certain types of cancer in the clinic and is also regarded as an immune-modulating and immune-enhancing agent that regulates immune cells. Emerging evidence indicates that an imbalance of immune cells, such as CD4+ T helper (Th) lymphocytes, contributes to the progression of immune thrombocytopenia (ITP), but the effects of Huaier on the regulation of CD4+ T cells are not yet fully elucidated. In the present study, Jurkat cells and peripheral blood mononuclear cells (PBMCs) from patients with ITP and healthy volunteers were treated with Huaier aqueous extract (HR). The CCK-8 assay revealed that HR suppressed the proliferation of Jurkat cells in a dose-dependent manner, whereas 3 mg/mL could decrease cell viability by 50%. At the latter concentration, the activation of CD4+ T cells from patients with ITP was partially attenuated. In addition, HR could correct the unbalanced Th1/Th2 polarization and inhibit the secretion of pro-inflammatory factors interleukin (IL)-2, tumor necrosis factor-α, and interferon-γ. It also suppressed Treg and facilitated Th17 differentiation, but did not change the levels of IL-10 and transforming growth factor-ß. Thus, this study provides more information on how Huaier regulates cellular immunity and improves our understanding of the use of Huaier in ITP.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Immunomodulating Agents/pharmacology , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Complex Mixtures/chemistry , Humans , Immunomodulating Agents/chemistry , Jurkat Cells , Male , Medicine, Chinese Traditional , Middle Aged , Purpura, Thrombocytopenic, Idiopathic/immunology , Trametes/chemistry , Young Adult
19.
Chin J Nat Med ; 19(8): 608-620, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34419260

ABSTRACT

Brucea javanica oil emulsion (BJOE) has been used to treat tumor in China for more than 40 years. However, its components and effectiveness in the treatment of acute lymphocytic leukemia (ALL) and its mechanism of anti-cancer activity remain unknown. In the current study, high-performance liquid chromatography-evaporative light scattering detector (HPLC-ELSD) was used to analyze the components of BJOE. Then, the anti-leukemia effects of BJOE were examined both in vitro and in vivo using ALL Jurkat cells and the p388 mouse leukemia transplant model, respectively. The primary ALL leukemia cells were also used to confirm the anti-leukemia effects of BJOE. The apoptotic-related results indicated that BJOE induced apoptosis in Jurkat cells and were suggestive of intrinsic apoptotic induction. Moreover, BJOE inhibited Akt (protein kinase B) activation and upregulated its downstream targets p53 and FoxO1 (forkhead box gene, group O-1) to initiate apoptosis. The activation of GSK3ß was also involved. Our findings demonstrate that BJOE has anti-leukemia effects on ALL cells and can induce apoptosis in Jurkat cells through the phosphoinositide3-kinase (PI3K) /Akt signaling pathway.


Subject(s)
Apoptosis , Brucea , Plant Oils/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Animals , Brucea/chemistry , Glycogen Synthase Kinase 3 , Humans , Jurkat Cells , Mice , Phosphatidylinositol 3-Kinases/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Proto-Oncogene Proteins c-akt/genetics , Seeds/chemistry , Signal Transduction
20.
Int J Mol Sci ; 22(16)2021 Aug 18.
Article in English | MEDLINE | ID: mdl-34445589

ABSTRACT

Crohn's Disease (CD) and Rheumatoid Arthritis (RA) share some single nucleotide polymorphisms (SNPs) in protein tyrosine phosphatase non-receptor types 2 and 22 (PTPN2/22). Recently, we reported that clinical samples from CD and RA patients associated with PTPN2:rs478582 or PTPN22:rs2476601 genotypes were linked to overactive immune response and exacerbation of inflammation. Here, we investigated in vitro the effects of these SNPs in Jurkat T-cells using CRISPR-Cas9. All cells were evaluated for PTPN22/22 loss of function and effects on cell response. We measured gene expression via RT-qPCR and cytokines by ELISA. We also measured cell proliferation using a BrdU labeling proliferation ELISA, and T-cell activation using CD-25 fluorescent immunostaining. In PTPN2 SNP-edited cells, PTPN2 expression decreased by 3.2-fold, and proliferation increased by 10.2-fold compared to control. Likewise, expression of PTPN22 decreased by 2.4-fold and proliferation increased by 8.4-fold in PTPN22 SNP-edited cells. IFN-γ and TNF-α secretions increased in both edited cell lines. CD25 expression (cell activation) was 80.32% in PTPN2 SNP-edited cells and 85.82% in PTPN22 SNP-edited cells compared to 70.48% in unedited Jurkat T-cells. Treatment of PTPN2 and PTPN22-edited cells with a maximum 20 µM spermidine restored PTPN2/22 expression and cell response including cell proliferation, activation, and cytokines secretion. Most importantly, the effect of spermidine on edited cells restored normal expression and secretion of IFN-γ and TNF-α. The data clearly demonstrated that edited SNPs in PTPN2 or PTPN22 were associated with reduced gene expression, which resulted in an increase in cell proliferation and activation and overactive immune response. The data validated our earlier observations in CD and RA clinical samples. Surprisingly, spermidine restored PTPN2/22 expression in edited Jurkat T-cells and the consequent beneficial effect on cell response and inflammation. The study supports the use of polyamines dietary supplements for management of CD and in RA patients.


Subject(s)
CRISPR-Cas Systems , Gene Expression Regulation, Leukemic/drug effects , Leukemia, T-Cell/pathology , Polymorphism, Single Nucleotide , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Spermidine/pharmacology , Arthritis, Rheumatoid/genetics , Crohn Disease/genetics , Genetic Predisposition to Disease , Humans , Jurkat Cells , Leukemia, T-Cell/drug therapy , Leukemia, T-Cell/genetics , Lymphocyte Activation , Protein Tyrosine Phosphatase, Non-Receptor Type 2/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 22/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL