Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Phytomedicine ; 103: 154229, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35691076

ABSTRACT

BACKGROUND: Modified Jianpi Yangzheng decoction (mJPYZ), as an empirical decoction of Traditional Chinese medicine has been shown significantly to prolong the survival of patients with advanced stage gastric cancer. Pyruvate kinase M2 (PKM2), has attracted attention for its important role on cellular aerobic glycolysis, however, few studies focus on PKM2 non-metabolic roles in tumor progression. PURPOSE: Our study aimed to investigate the potential role of gastric cancer exosomes containing PKM2 in regulating tumor-associated macrophages (TAM) and the mechanism of mJPYZ against gastric cancer. METHODS: Colony Formation Assay, flow cytometry and TUNEL staining were employed to estimate the effect of mJPYZ on gastric cancer in tumor-bearing mice and cells. Western blot analyzed apoptosis-related protein expression changes. Network pharmacology and bioinformatics predicted potential exosomes modulation of mJPYZ in gastric cancer. Exosomes were isolated and co-cultured with TAM. Diff-Quik Staining observed the TAM morphological changes when incubating with gastric cancer cells exosomes. Flow cytometry and immunofluorescence were performed to demonstrate whether exosomes PKM2 involved in TAM polarization. RESULTS: mJPYZ induced apoptosis of gastric cancer cells by targeting PKM2 and downregulating PI3K/Akt/mTOR axis in vivo and in vitro. Network pharmacology showed potential exosomes modulation of mJPYZ in gastric cancer. We extracted exosomes and found mJPYZ decreased the abundance of serum exosomes PKM2 in patients with advanced gastric cancer and xenograft tumor model. Additionally, we firstly detected and confirmed that PKM2 is a package protein of exosomes extracted from gastric cancer cells, and mJPYZ could diminish the content of exosomal PKM2 in gastric cancer cells. Importantly, mJPYZ reduced the delivery of exosomal PKM2 from tumor cells to macrophages, and alleviated exosomal PKM2-induced differentiation of M2-TAM in tumor microenvironment, eventually inhibited gastric cancer progression. CONCLUSION: Gastric cancer exosomes containing PKM2 could lead to M2 macrophages differentiation, thereby promoting gastric cancer progression. Our findings provide a rationale for potential application of mJPYZ in the treatment of gastric cancer via PKM2.


Subject(s)
Drugs, Chinese Herbal , Exosomes , Pyruvate Kinase , Stomach Neoplasms , Animals , Carrier Proteins/metabolism , Cell Line, Tumor , Drugs, Chinese Herbal/pharmacology , Exosomes/drug effects , Exosomes/metabolism , Exosomes/pathology , Humans , Membrane Proteins/metabolism , Mice , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pyruvate Kinase/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Thyroid Hormones/metabolism , Tumor Microenvironment , Thyroid Hormone-Binding Proteins
2.
Food Funct ; 12(23): 11790-11807, 2021 Nov 29.
Article in English | MEDLINE | ID: mdl-34761788

ABSTRACT

Exosomes are extracellular membranous nanovesicles that carry functional molecules to mediate cell-to-cell communication. To date, whether probiotics improve the immune function of broilers by plasmal exosome cargo is unclear. In this study, 300 broilers were allocated to three treatments: control diet (CON group), control diet + dexamethasone injection (DEX group), and control diet containing 1 × 108 cfu g-1 P8 + DEX injection (P8 + DEX group). The growth performance, meat quality and immune function of plasma and jejunal mucosa were detected. Exosomes were isolated from the plasma and characterized. Then, the exosome protein profile was determined by proteomic analysis. Correlation analyses between the exosomal proteins and growth performance, meat quality, immune function were performed. Lastly, the related protein levels were verified by multiple reaction monitoring (MRM). Results showed that P8 treatment increased the growth performance, meat quality and immune function of DEX-induced broilers with immunological stress. Moreover, the average diameters, cup-shaped morphology and expressed exosomal proteins confirmed that the isolated extracellular vesicles were exosomes. A total of 784 proteins were identified in the exosomes; among which, 126 differentially expressed proteins (DEPs) were found between the DEX and CON groups and 102 DEPs were found between the P8 + DEX and DEX groups. Gene ontology analysis indicated that DEPs between the DEX and CON groups are mainly involved in the metabolic process, cellular anatomical entity, cytoplasm, etc. DEPs between the P8 + DEX and DEX groups are mainly involved in the multicellular organismal process, response to stimulus, cytoplasm, etc. Pathway analysis revealed that most of the DEPs between the DEX and CON groups participated in the ECM-receptor interaction, focal adhesion, regulation of actin cytoskeleton, etc. Most of the DEPs between the P8 + DEX and DEX groups participated in the ErbB and PPAR signaling pathways. Moreover, many DEPs were correlated with the altered parameters of growth performance, meat quality and immunity in P8-treated broilers. MRM further revealed that the upregulated FABP6 and EPCAM in the DEX group were decreased by P8 + DEX treatment, and the downregulated C1QTNF3 in the DEX group was increased by P8 + DEX treatment. In conclusion, our findings demonstrated that P8 may promote the immune function, growth performance and meat quality of broilers with immunological stress by regulating the plasma exosomal proteins, especially the proteins of FABP6, EPCAM and C1QTNF3 and the pathway of PPAR (ILK/FABP6).


Subject(s)
Chickens , Exosomes , Lactobacillus plantarum , Meat , Probiotics , Animal Feed , Animals , Dietary Supplements , Exosomes/chemistry , Exosomes/drug effects , Exosomes/immunology , Male , Meat/analysis , Meat/standards , Probiotics/administration & dosage , Probiotics/pharmacology , Proteomics , Stress, Physiological/immunology
3.
Molecules ; 26(8)2021 Apr 12.
Article in English | MEDLINE | ID: mdl-33921245

ABSTRACT

Endometrial cancer (EC) cells metastasize to various regions, including the ovaries, fallopian tubes, cervix, blood, liver, bone, and brain. Various carcinogens are known to cause EC. Exosomes are released from several types of cells and contain various cellular components. In this study, flow cytometry and quantitative PCR were used to evaluate marker levels, cell migration, cell invasion, and mitochondrial membrane potential, and cellular senescence tests were used to estimate cancer activity. The microRNAs were profiled using next-generation sequencing. Although tocopherol-α and rutin content in Aurea helianthus is high, A. helianthus extract was more useful in modulating tumor activity compared to the two aforementioned substances. Notably, we established that the extract induced bioactive exosomes in EC cells, and profiling of miRNAs in the extract-inducing exosomes (EIE) indicated their potency to be developed as a biological drug. The extract and EIE contributed to the following five biological process categories for EC cells: (1) cell migration and invasion suppression, (2) cellular senescence activation by attenuating mitochondrial membrane potential and enhancing autophagy, (3) reproductive cancer activity attenuation, (4) drug susceptibility activation, and (5) EIE containing miRNAs associated with decreasing inflammation.


Subject(s)
Endometrial Neoplasms/metabolism , Exosomes/metabolism , Malvaceae/chemistry , Plant Extracts/pharmacology , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Endometrial Neoplasms/genetics , Exosomes/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , MicroRNAs/genetics , MicroRNAs/metabolism
4.
Daru ; 28(2): 647-659, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32974883

ABSTRACT

BACKGROUND: Despite the advances in the treatment of multiple myeloma (MM), complete remission is usually challenging. The interactions between tumor and host cells, in which exosomes (EXs) play critical roles, have been shown to be among the major deteriorative tumor-promoting factors herein. Therefore, any endeavor to beneficially target these EX-mediated interactions could be of high importance. OBJECTIVES: a) To investigate the effects of myeloma EXs on natural killer (NK) cell functions. b) To check whether treatment of myeloma cells with eicosapentaenoic acid (EPA) or docosahexaenoic acid (DHA), two polyunsaturated omega-3 fatty acids with known anti-cancer effects, can modify myeloma EXs in terms of their effects on natural killer functions. METHODS: L363 cells were treated with either EPA or DHA or left untreated and the released EXs (designated as E-EX, D-EX and C-EX, respectively) were used to treat NK cells for functional studies. RESULTS: Myeloma EXs (C-EXs) significantly reduced NK cytotoxicity against K562 cells (P ≤ 0.05), while the cytotoxicity suppression was significantly lower (P ≤ 0.05) in the (E-EX)- and (D-EX)-treated NK cells compared to the (C-EX)-treated cells. The expression of the activating NK receptor NKG2D and NK degranulation, after treatment with the EXs, were both altered following the same pattern. However, C-EXs could increase IFN-γ production in NK cells (P < 0.01), which was not significantly affected by EPA/DHA treatment. This indicates a dual effect of myeloma EXs on NK cells functions. CONCLUSION: Our observations showed that myeloma EXs have both suppressive and stimulatory effects on different NK functions. Treatment of myeloma cells with EPA/DHA can reduce the suppressive effects of myeloma EXs while maintaining their stimulatory effects. These findings, together with the previous findings on the anti-cancer effects of EPA/DHA, provide stronger evidence for the repositioning of the currently existing EPA/DHA supplements to be used in the treatment of MM as an adjuvant treatment. EXs released from L363 (myeloma) cells in their steady state increase IFN-γ production of NK cells, while reduce their cytotoxicity against the K562 cell line (right blue trace). EXs from L363 cells pre-treated with either EPA or DHA are weaker stimulators of IFN-γ production. These EXs also increase NK cytotoxicity and NKG2D expression (left brown trace) compared to the EXs obtained from untreated L363 cells. Based on these findings, myeloma EXs have both suppressive and stimulatory effects on different NK functions depending on the properties of their cells of origin, which can be exploited in the treatment of myeloma.


Subject(s)
Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Exosomes/physiology , Killer Cells, Natural/cytology , Multiple Myeloma/metabolism , Cell Line, Tumor , Chemotherapy, Adjuvant , Exosomes/drug effects , Humans , Interferon-gamma/metabolism , K562 Cells , Killer Cells, Natural/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism
5.
J Neurosci ; 40(41): 7965-7979, 2020 10 07.
Article in English | MEDLINE | ID: mdl-32887744

ABSTRACT

Microglia, a type of CNS immune cell, have been shown to contribute to ethanol-activated neuronal death of the stress regulatory proopiomelanocortin (POMC) neuron-producing ß-endorphin peptides in the hypothalamus in a postnatal rat model of fetal alcohol spectrum disorders. We determined whether the microglial extracellular vesicle exosome is involved in the ethanol-induced neuronal death of the ß-endorphin neuron. Extracellular vesicles were prepared from hypothalamic tissues collected from postnatal rats (both males and females) fed daily with 2.5 mg/kg ethanol or control milk formula for 5 d or from hypothalamic microglia cells obtained from postnatal rats, grown in cultures for several days, and then challenged with ethanol or vehicle for 24 h. Nanoparticle tracking analysis and transmission electron microscopy indicated that these vesicles had the size range and shape of exosomes. Ethanol treatments increased the number and the ß-endorphin neuronal killing activity of microglial exosomes both in vivo and in vitro Proteomics analyses of exosomes of cultured microglial cells identified a large number of proteins, including various complements, which were elevated following ethanol treatment. Proteomics data involving complements were reconfirmed using quantitative protein assays. Ethanol treatments also increased deposition of the complement protein C1q in ß-endorphin neuronal cells in both in vitro and in vivo systems. Recombinant C1q protein increased while C1q blockers reduced ethanol-induced C3a/b, C4, and membrane attack complex/C5b9 formations; ROS production; and ultimately cellular death of ß-endorphin neurons. These data suggest that the complement system involving C1q-C3-C4-membrane attack complex and ROS regulates exosome-mediated, ethanol-induced ß-endorphin neuronal death.SIGNIFICANCE STATEMENT Neurotoxic action of alcohol during the developmental period is recognized for its involvement in fetal alcohol spectrum disorders, but the lack of clear understanding of the mechanism of alcohol action has delayed the progress in therapeutic intervention of this disease. Proopiomelanocortin neurons known to regulate stress, energy homeostasis, and immune functions are reported to be killed by developmental alcohol exposure because of activation of microglial immune cells in the brain. While microglia are known to use extracellular vesicles to communicate with neurons for maintaining homeostasis, we show here that ethanol exposure during the developmental period hijacks this system to spread apoptotic factors, including complement protein C1q, to induce the membrane attack complex and reactive super-oxygen species for proopiomelanocortin neuronal killing.


Subject(s)
Central Nervous System Depressants/pharmacology , Complement C1q/pharmacology , Ethanol/pharmacology , Exosomes/drug effects , Fetal Alcohol Spectrum Disorders/pathology , Microglia/drug effects , Pro-Opiomelanocortin/genetics , Animals , Animals, Newborn , Cell Death/drug effects , Cells, Cultured , Female , Fetal Alcohol Spectrum Disorders/metabolism , Hypothalamus/metabolism , Hypothalamus/pathology , Male , Neurons/drug effects , Neurons/metabolism , Pregnancy , Proteomics , Rats , Rats, Sprague-Dawley , beta-Endorphin/metabolism
6.
Toxicol Appl Pharmacol ; 401: 115109, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32544403

ABSTRACT

Bladder cancer (BCa) is the fourth leading cause of cancer deaths worldwide due to its aggressiveness and resistance against therapies. Intricate interactions between cancer cells and the tumor microenvironment (TME) are essential for both disease progression and regression. Thus, interrupting molecular communications within the TME could potentially provide improved therapeutic efficacies. M2-polarized tumor-associated macrophages (M2 TAMs) were shown to contribute to BCa progression and drug resistance. We attempted to provide evidence for ovatodiolide (OV) as a potential therapeutic agent that targets both TME and BCa cells. First, tumor-suppressing functions of OV were determined by cell viability, colony, and tumor-sphere formation assays using a coculture system composed of M2 TAMs/BCa cells. Subsequently, we demonstrated that extracellular vesicles (EVs) isolated from M2 TAMs containing oncomiR-21 and mRNAs, including Akt, STAT3, mTOR, and ß-catenin, promoted cisplatin (CDDP) resistance, migration, and tumor-sphere generation in BCa cells, through increasing CDK6, mTOR, STAT3, and ß-catenin expression. OV treatment also prevented M2 polarization and reduced EV cargos from M2 TAMs. Finally, in vivo data demonstrated that OV treatment overcame CDDP resistance. OV only and the OV + CDDP combination both resulted in significant reductions in mTOR, ß-catenin, CDK6, and miR-21 expression in tumor samples and EVs isolated from serum. Collectively, we demonstrated that M2 TAMs induced malignant properties in BCa cells, in part via oncogenic EVs. OV treatment prevented M2 TAM polarization, reduced EV cargos derived from M2 TAMs, and suppressed ß-catenin/mTOR/CDK6 signaling. These findings provide preclinical evidence for OV as a single or adjuvant agent for treating drug-resistant BCa.


Subject(s)
Cyclin-Dependent Kinase 6/metabolism , Diterpenes/therapeutic use , MicroRNAs/metabolism , TOR Serine-Threonine Kinases/metabolism , Urinary Bladder Neoplasms/metabolism , beta Catenin/metabolism , Animals , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Cell Line, Tumor , Coculture Techniques , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Diterpenes/isolation & purification , Diterpenes/pharmacology , Dose-Response Relationship, Drug , Exosomes/drug effects , Exosomes/metabolism , Exosomes/pathology , Female , Humans , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/antagonists & inhibitors , Plants, Medicinal , TOR Serine-Threonine Kinases/antagonists & inhibitors , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , beta Catenin/antagonists & inhibitors
7.
J Ethnopharmacol ; 256: 112802, 2020 Jun 28.
Article in English | MEDLINE | ID: mdl-32240782

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Jinfukang has long been used for the clinical treatment of lung cancer. Previous studies have shown that Jinfukang can induce the apoptosis of circulating tumor cells by intervening ROS-mediated DNA damage pathway. However, whether Jinfukang can inhibit the metastasis of circulating tumor cells and its mechanism are still unclear. AIM OF THE STUDY: To further investigate the mechanism of Jinfukang in anti-metastasis of lung cancer from the perspective of intervention of tumor exosomes. MATERIALS AND METHODS: The invadopodia formation was determined with immunofluorescence. Invasion and migration were detected using the Transwell assay. Ultracentrifugation was used to isolate exosomes. Exosomes were characterized by electron microscopy, nanoparticle tracking analysis and immunoblotting, and the protein profile was evaluated by proteomic analysis. The molecular functions, biological processes and signaling pathway enrichment analysis were performed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Key differentially expressed proteins were verified by Western blot. RESULTS: Jinfukang can inhibit the expression of MMP14, cortactin, Tks5 and the formation of invadopodia of CTC-TJH-01 cells. Furthermore, Jinfukang can significantly inhibit the invasion and migration of CTC-TJH-01 cells. The diameter of exosomes extracted from the CTC-TJH-01 cells treated by Jinfukang was 30-100 nm, and the exosomal markers CD63, CD81 and TSG101 were expressed. We identified 680 deferentially expressed proteins. Gene oncology analysis indicated that exosomes were mostly derived from plasma membrane and mainly involved in protein localization and intracellular signaling. The ingenuity pathway analysis showed that the EGF pathway was significantly inhibited, whereas the GP6 signaling pathway was significantly activated. We also confirmed that Jinfukang inhibited the expression of EGF pathway-related proteins in CTC-TJH-01 cells. Besides, when EGF was used to activate EGF signaling pathway, the inhibition of Jinfukang on CTC cell metastasis was reversed. CONCLUSION: Jinfukang inhibits the metastasis of CTC-TJH-01 cells through the EGF pathway.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Exosomes/drug effects , Exosomes/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , MicroRNAs/metabolism , Proteomics/methods , Signal Transduction/drug effects
8.
J Biochem Mol Toxicol ; 34(5): e22462, 2020 May.
Article in English | MEDLINE | ID: mdl-32045083

ABSTRACT

Heart failure (HF) is a medical condition inability of the heart to pump sufficient blood to meet the metabolic demand of the body to take place. The number of hospitalized patients with cardiovascular diseases is estimated to be more than 1 million each year, of which 80% to 90% of patients ultimately progress to decompensated HF. Digitalis glycosides exert modest inotropic actions when administered to patients with decompensated HF. Although its efficacy in patients with HF and atrial fibrillation is clear, its value in patients with HF and sinus rhythm has often been questioned. A series of recent studies have cast serious doubt on the benefit of digoxin when added to contemporary HF treatment. We are hypothesizing the role and mechanism of exosome and its biological constituents responsible for worsening the disease state and mortality in decompensated HF patients on digitalis.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/drug therapy , Cardiotonic Agents/therapeutic use , Digitalis/chemistry , Digoxin/therapeutic use , Exosomes/drug effects , Heart Failure/drug therapy , Plant Extracts/therapeutic use , Anti-Arrhythmia Agents/pharmacology , Cardiotonic Agents/pharmacology , Digoxin/pharmacology , Humans , Plant Extracts/pharmacology , Wnt Signaling Pathway/drug effects
9.
Fitoterapia ; 141: 104481, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31954179

ABSTRACT

In this study, seven previously undescribed steroidal glycoalkaloids, compounds 1-7, were isolated from Solanum lyratum, along with two known ones (8 and 9). Comprehensive spectroscopy techniques were used to determine their structures. Although 1-8 only showed a weak inhibitory effect on the proliferation of the tumor-derived vascular endothelial cells, however, in a former study we found both total steroidal glycoalkaloids from Solanum lyratum (TSGS) and 9 significantly inhibited tumor angiogenesis and its mechanism was linked to its ability to interfere with cell membrane lipid rafts. Lipid rafts are closely related to the functions of tumor-derived exosomes, a vital factor in cancer progression. Thus, we investigated the impacts of TSGS and 9 on the functions of A549-derived exosomes. Our results indicated that A549-derived exosomes can significantly enhance the angiogenesis abilities of human umbilical vein endothelial cells, whereas the intervention of TSGS or 9 significantly inhibited this activity of A549-derived exosomes. These findings suggest that TSGS and 9 exert anti-tumor angiogenesis by inhibiting the pro-angiogenic activity of A549-derived exosomes.


Subject(s)
Alkaloids/chemistry , Alkaloids/pharmacology , Exosomes/drug effects , Neovascularization, Physiologic/drug effects , A549 Cells , Alkaloids/classification , Humans , Molecular Structure
10.
Aging (Albany NY) ; 11(24): 12278-12294, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31848325

ABSTRACT

Spinal cord injury (SCI) remains the most common cause of paralysis, and there are no effective therapies for SCI patients. Neural stem cell (NSC)-derived exosomes can attenuate apoptosis and neuroinflammation after traumatic spinal cord injury, but the mechanisms underlying these effects remain unclear. Here, we examined the efficacy of miRNAs isolated from exosomes as treatments for SCI and characterized their mechanisms of action. Furthermore, we evaluated the effects of exosomes formed in the presence of insulin growth factor-1 (IFG-1, IGF-Exo), which promotes neural proliferation and regeneration, as well as normal exosomes (Nor-Exo) and compared control and H2O2-treated groups both invitro and invivo. Using microRNA sequencing and qRT-PCR, we identified miR-219a-2-3p, levels of which were higher in the IGF-Exo than Nor-Exo group and played crucial anti-inflammatory and anti-apoptosis roles. Additional experiments revealed that IGF-Exo inhibits YY1 expression through up-regulation of miR-219a-2-3p. This in turn inhibits the NF-κB pathway, partly inhibiting neuroinflammation and promoting the neuroprotective effects after SCI.


Subject(s)
Exosomes/metabolism , Insulin-Like Growth Factor I/pharmacology , MicroRNAs/metabolism , Neural Stem Cells/drug effects , Stem Cell Transplantation , YY1 Transcription Factor/metabolism , Animals , Apoptosis , Cell Survival , Embryonic Stem Cells , Exosomes/drug effects , Female , Hindlimb , MicroRNAs/genetics , Motor Activity , Neural Stem Cells/physiology , PC12 Cells , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries , YY1 Transcription Factor/genetics
11.
J Exp Biol ; 222(Pt 20)2019 10 16.
Article in English | MEDLINE | ID: mdl-31537653

ABSTRACT

Microvesicles are key players in cellular communication. As glandular secretions present a rich source of active exosomes, we hypothesized that exosome-like vesicles are present in Apis mellifera hypopharyngeal gland secretomal products (honey, royal jelly and bee pollen), and participate in their known antibacterial and pro-regenerative effects. We developed an isolation protocol based on serial centrifugation and ultracentrifugation steps and demonstrated the presence of protein-containing exosome-like vesicles in all three bee-derived products. Assessing their antibacterial properties, we found that exosome-like vesicles had bacteriostatic, bactericidal and biofilm-inhibiting effects on Staphylococcus aureus Furthermore, we demonstrated that mesenchymal stem cells (MSCs) internalize bee-derived exosome-like vesicles and that these vesicles influence the migration potential of the MSCs. In an in vitro wound-healing assay, honey and royal jelly exosome-like vesicles increased migration of human MSCs, demonstrating their inter-kingdom activity. In summary, we have discovered exosome-like vesicles as a new, active compound in bee pollen, honey and royal jelly.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bees/metabolism , Exosomes/metabolism , Fatty Acids/chemistry , Honey , Pollen/chemistry , Regeneration/drug effects , Animals , Cell Movement/drug effects , Endocytosis/drug effects , Exosomes/drug effects , Humans , Pollen/ultrastructure
12.
Biochem Pharmacol ; 169: 113619, 2019 11.
Article in English | MEDLINE | ID: mdl-31465776

ABSTRACT

Immunoglobulin A nephropathy (IgAN) is an autoimmune kidney disease with complex pathogenesis leading to end-stage renal damage. The prime pathological characteristics of IgAN are IgA immune complexes deposition accompany with mesangial cell proliferation and urine protein elevation. Artemisinin (ART) is extracted from traditional Chinese medicine Artemisia annua L. Hydroxychloroquine (HCQ) is a classical antimalarial drug applied in the treatment of autoimmune diseases. Both of them possess anti-inflammatory and immunomodulatory properties. The purpose of this research was to investigate the pharmacological effects of ART combined with HCQ (AH) and discuss thoroughly the potential molecular mechanisms in IgAN. In vivo, our results demonstrated that AH could efficiently ameliorate kidney damage by improving kidney dysfunction and reducing the levels of 24 h urine protein, IgA and IgG immune complexes deposition in glomerulus of IgAN rats. Interestingly, AH obviously promoted the secretion of exosomes in renal tissues, inhibited the expressions of nuclear factor-κB (NF-κB) signaling and NLRP3 inflammasome-related proteins, including IκB-α, p-p65, NLRP3, ASC, IL-1ß and caspase-1 in IgAN rats. In vitro, further mechanistic study illustrated that exosomes derived from human renal tubular epithelial cells (HK-2) were significantly enhanced by AH, which could be utterly taken up in human mesangial cells (HMCs) and inhibited the activation of NF-κB pathway and NLRP3 inflammasome after AH intervention. However, GW4869 interdicted the promotive effect of AH on exosomes from HK-2 cells and the suppression of exosomes on NF-κB/NLRP3 activation in HMCs. Taken together, this study demonstrated that there was an inhibitory effect of AH therapy on NF-κB/NLRP3 signaling via mediating exosomes release in IgAN rats, which provided an alternative approach for IgAN treatment.


Subject(s)
Artemisinins/administration & dosage , Artemisinins/pharmacology , Exosomes/drug effects , Glomerulonephritis, IGA/drug therapy , Hydroxychloroquine/administration & dosage , Inflammasomes/antagonists & inhibitors , Kidney/drug effects , NF-kappa B/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Aniline Compounds/pharmacology , Animals , Benzylidene Compounds/pharmacology , Cells, Cultured , Drug Therapy, Combination , Exosomes/physiology , Humans , Inflammasomes/physiology , Male , NF-kappa B/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
13.
J Pineal Res ; 66(4): e12561, 2019 May.
Article in English | MEDLINE | ID: mdl-30659651

ABSTRACT

Adipocyte-derived exosomes (Exos) serve as bioinformation-containing messengers in cell-to-cell communications, and numerous reports demonstrate that resistin, an adipokine, is strongly associated with hepatic steatosis and other fatty liver diseases, suggesting that adipose dysfunction-generated altered pattern of exosomal cytokines may contribute to shaping the physiological activities in liver. Admittedly, melatonin-mediated positive effects on various tissues/organs have been respectively reported, but regulatory mechanisms of melatonin on the crosstalk between adipose tissue and liver have been investigated rarely. Overall, we hypothesize that the crosstalk originating from adipose tissue may be another worthy regulatory pathway for melatonin ameliorating of hepatic steatosis. Here, we first found the amount of adipocyte-derived exosomal resistin to be significantly decreased by melatonin supplementation. Compared to mice with ExosHFD or Exosresistin treatment, ExosMT remarkably ameliorated hepatic steatosis. Further test demonstrated that resistin was a pivotal cytokine which repressed phosphorylation of 5' adenosine monophosphate-activated protein kinase α (pAMPKα Thr172 ) to trigger endoplasmic reticulum (ER) stress, resulting in hepatic steatosis, whereas ExosMT reversed these risks in hepatocytes. In adipocytes, we identified melatonin to reduce the production of resistin through the brain and muscle arnt-like protein 1 (Bmal1) transcriptional inhibition. Notably, we also confirmed that melatonin enhanced N6 -Methyladenosine (m6 A) RNA demethylation to degrade resistin mRNA in adipocytes. Overall, melatonin decreases traffic volume of adipocyte-generated exosomal resistin from adipocytes to hepatocytes, which further alleviates ER stress-induced hepatic steatosis. Our findings illustrate a novel melatonin-mediated regulatory pathway from adipocytes to hepatocytes, indicating that adipocyte-derived exosome is a new potential target for treating obesity and related hepatorenal syndrome.


Subject(s)
Adipocytes/metabolism , Exosomes/drug effects , Exosomes/metabolism , Fatty Liver/drug therapy , Fatty Liver/metabolism , Melatonin/therapeutic use , Resistin/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Random Allocation
14.
Microcirculation ; 26(1): e12515, 2019 01.
Article in English | MEDLINE | ID: mdl-30431204

ABSTRACT

OBJECTIVE: To investigate the effects of TNF-α-induced exosomes release on the biological behavior, metabolism, and bioenergetics of HUVECs. METHODS: Exosomes were isolated from conditioned media of HUVECs by ultracentrifugation after treatment with or without TNF-α. HUVECs were treated with or without TNF-α, or different concentrations of exosomes isolated from conditioned media with or without TNF-α induction (TExo and CExo , respectively). RESULTS: The results showed that TNF-α significantly inhibited migration, tube formation, and increased apoptosis rate of HUVECs compared with controls. Furthermore, TNF-α-induced exosomes (TExo ) rather than CExo , indicated similar effects to inhibit migration, tube formation and promote endothelial apoptosis. Although TNF-α treatment did not show a statistical difference, TExo significantly inhibited extracellular OCR compared with controls. TExo could significantly inhibited intracellular OCR in a hypoxia condition. TNF-α significantly increased L-ECA compared with control cells, and TExo showed similar stimulative effect on L-ECA. CONCLUSIONS: TNF-α-induced exosomes could significantly (a) change migration, tube formation, and apoptosis; (b) inhibit endothelial extracellular OCR and intracellular OCR (hypoxia); (c) increase glycolysis rate of the endothelial cells. These data provide new evidence for exploring endothelial behavior regulation using exosomes and their effects on endothelial metabolism and bioenergetics.


Subject(s)
Exosomes/physiology , Human Umbilical Vein Endothelial Cells/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis , Cell Movement , Cells, Cultured , Energy Metabolism , Exosomes/drug effects , Glycolysis , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/physiology , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Hypoxia , Lactic Acid/metabolism , Oxygen Consumption
15.
Int J Mol Sci ; 19(12)2018 Dec 19.
Article in English | MEDLINE | ID: mdl-30572617

ABSTRACT

In healthy or pathological brains, the neuroinflammatory state is supported by a strong communication involving microglia and neurons. Recent studies indicate that extracellular vesicles (EVs), including exosomes and microvesicles, play a key role in the physiological interactions between cells allowing central nervous system (CNS) development and/or integrity. The present report used medicinal leech CNS to investigate microglia/neuron crosstalk from ex vivo approaches as well as primary cultures. The results demonstrated a large production of exosomes from microglia. Their incubation to primary neuronal cultures showed a strong interaction with neurites. In addition, neurite outgrowth assays demonstrated microglia exosomes to exhibit significant neurotrophic activities using at least a Transforming Growth Factor beta (TGF-ß) family member, called nGDF (nervous Growth/Differentiation Factor). Of interest, the results also showed an EV-mediated dialog between leech microglia and rat cells highlighting this communication to be more a matter of molecules than of species. Taken together, the present report brings a new insight into the microglia/neuron crosstalk in CNS and would help deciphering the molecular evolution of such a cell communication in brain.


Subject(s)
Central Nervous System/metabolism , Exosomes/metabolism , Hirudo medicinalis/physiology , Microglia/metabolism , Neurons/metabolism , Amino Acid Sequence , Animals , Central Nervous System/drug effects , Coculture Techniques , Exosomes/drug effects , Exosomes/ultrastructure , Microglia/drug effects , Nerve Growth Factors/pharmacology , Neurites/drug effects , Neurites/metabolism , Neurons/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
Biosci Rep ; 38(5)2018 10 31.
Article in English | MEDLINE | ID: mdl-30061173

ABSTRACT

d Rhamnose ß-hederin (DRß-H), an active component extracted from the traditional Chinese medicinal plant Clematis ganpiniana, has been reported to be effective against breast cancer. Recent studies have also indicated that the isolated exosomes (D/exo) from docetaxel-resistant breast cancer cells MCF-7 (MCF-7/Doc) were associated with resistance transmission by delivering genetic cargo. However, the relevance of D/exo during DRß-H exposure remains largely unclear. In the present work, exosomes were characterized by morphology and size distribution. We reinforced the significant role of D/exo in spreading chemoresistance from MCF-7/Doc to recipient sensitive cells after absorption and internalization. DRß-H could reduce the formation and release of D/exo. Next, we demonstrated that DRß-H was able to reverse docetaxel resistance and that D/exo was responsible for DRß-H-mediated resistance reversal. We also found that DRß-H could decrease the expressions of several most abundant miRNAs (miR-16, miR-23a, miR-24, miR-26a, and miR-27a) transported by D/exo. Target gene prediction and pathway analysis showed the involvement of these selected miRNAs in pathways related to treatment failure. Our results suggested that DRß-H could reduce D/exo secretion from MCF-7/Doc cells and induce the reduction in resistance transmission via D/exo.


Subject(s)
Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Exosomes/drug effects , Oleanolic Acid/analogs & derivatives , Saponins/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Docetaxel/pharmacology , Drug Resistance, Neoplasm/physiology , Exosomes/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , MicroRNAs/genetics , Oleanolic Acid/pharmacology
17.
Hepatology ; 65(5): 1543-1556, 2017 05.
Article in English | MEDLINE | ID: mdl-28027586

ABSTRACT

Diseases of the liver related to metabolic syndrome have emerged as the most common and undertreated hepatic ailments. The cause of nonalcoholic fatty liver disease is the aberrant accumulation of lipid in hepatocytes, though the mechanisms whereby this leads to hepatocyte dysfunction, death, and hepatic fibrosis are still unclear. Insulin-sensitizing thiazolidinediones have shown efficacy in treating nonalcoholic steatohepatitis (NASH), but their widespread use is constrained by dose-limiting side effects thought to be due to activation of the peroxisome proliferator-activated receptor γ. We sought to determine whether a next-generation thiazolidinedione with markedly diminished ability to activate peroxisome proliferator-activated receptor γ (MSDC-0602) would retain its efficacy for treating NASH in a rodent model. We also determined whether some or all of these beneficial effects would be mediated through an inhibitory interaction with the mitochondrial pyruvate carrier 2 (MPC2), which was recently identified as a mitochondrial binding site for thiazolidinediones, including MSDC-0602. We found that MSDC-0602 prevented and reversed liver fibrosis and suppressed expression of markers of stellate cell activation in livers of mice fed a diet rich in trans-fatty acids, fructose, and cholesterol. Moreover, mice with liver-specific deletion of MPC2 were protected from development of NASH on this diet. Finally, MSDC-0602 directly reduced hepatic stellate cell activation in vitro, and MSDC-0602 treatment or hepatocyte MPC2 deletion also limited stellate cell activation indirectly by affecting secretion of exosomes from hepatocytes. CONCLUSION: Collectively, these data demonstrate the effectiveness of MSDC-0602 for attenuating NASH in a rodent model and suggest that targeting hepatic MPC2 may be an effective strategy for pharmacologic development. (Hepatology 2017;65:1543-1556).


Subject(s)
Acetophenones/therapeutic use , Anion Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Non-alcoholic Fatty Liver Disease/drug therapy , Thiazolidinediones/therapeutic use , Acetophenones/pharmacology , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Exosomes/drug effects , Hepatic Stellate Cells/drug effects , Male , Mice, Inbred C57BL , Molecular Targeted Therapy , Random Allocation , Thiazolidinediones/pharmacology
18.
Molecules ; 21(6)2016 Jun 16.
Article in English | MEDLINE | ID: mdl-27322220

ABSTRACT

Shikonin is a naphthoquinone isolated from the traditional Chinese medicine Lithospermum. It has been used in the treatment of various tumors. However, the effects of shikonin on such diseases have not been fully elucidated. In the present study, we detected the exosome release of a breast cancer cell line (MCF-7) with shikonin treatment and found a positive relationship between the level of secreted exosomes and cell proliferation. We next analyzed miRNA profiles in MCF-7 cells and exosomes and found that some miRNAs are specifically sorted and abundant in exosomes. Knockdown of the most abundant miRNAs in exosomes and the MCF-7 proliferation assay showed that miR-128 in exosomes negatively regulates the level of Bax in MCF-7 recipient cells and inhibits cell proliferation. These results show that shikonin inhibits the proliferation of MCF-7 cells through reducing tumor-derived exosomal miR-128. The current study suggests that shikonin suppresses MCF-7 growth by the inhibition of exosome release.


Subject(s)
Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Naphthoquinones/administration & dosage , Apoptosis/drug effects , Breast Neoplasms/genetics , Exosomes/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , MicroRNAs/biosynthesis
19.
Biochim Biophys Acta ; 1851(9): 1123-33, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25900100

ABSTRACT

Little is known about the effects of fatty acids on adiponectin oligomer assembly and trafficking. The aim of this study was to examine the effects of different fatty acids on adiponectin transport and secretion in differentiated 3T3-L1 adipocytes. Subcellular fractionation and immunofluorescence microscopy revealed that the majority of cellular adiponectin was located in the endoplasmic reticulum (ER). Adiponectin secretion was increased by treatment with fatty acids, including eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), and several fatty acids changed the cellular localization of adiponectin. Adiponectin secretion has been shown to be altered by ER stress and interactions with ER chaperone proteins. However these mechanisms were not influenced by fatty acids, suggesting that alternative mechanisms must be responsible for the increased secretion of adiponectin observed with fatty acid treatment. Secretion of adiponectin was blocked by Brefeldin A, but we identified a minor pool of adiponectin that could be secreted from beyond the Brefeldin A block. Exosomes appeared to contribute to a minor amount of adiponectin secreted from the cell, and exosome release was increased by treatment with DHA. These data suggest that the ER is an important site of adiponectin accumulation and that treatment with long chain omega-3 fatty acids increases adiponectin release. Furthermore, the secretory pathway of adiponectin is complex, involving both the classical ER-Golgi pathway as well as unconventional secretory mechanisms such as an exosome-mediated pathway.


Subject(s)
Adipocytes/drug effects , Adiponectin/metabolism , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Endoplasmic Reticulum/drug effects , Exosomes/drug effects , 3T3-L1 Cells , Adipocytes/metabolism , Adiponectin/agonists , Adiponectin/genetics , Animals , Biological Transport/drug effects , Brefeldin A/pharmacology , Cell Differentiation , Endoplasmic Reticulum/metabolism , Exosomes/metabolism , Gene Expression , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Mice , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Signal Transduction
20.
PLoS One ; 10(4): e0124436, 2015.
Article in English | MEDLINE | ID: mdl-25919665

ABSTRACT

HIV-1 Nef is an important pathogenic factor for HIV/AIDS pathogenesis. Several recent studies including ours have demonstrated that Nef can be transferred to neighboring cells and alters the function of these cells. However, how the intercellular Nef transfer occurs is in dispute. In the current study, we attempted to address this important issue using several complementary strategies, a panel of exosomal markers, and human CD4+ T lymphocyte cell line Jurkat and a commonly used cell line 293T. First, we showed that Nef was transferred from Nef-expressing or HIV-infected Jurkat to naïve Jurkat and other non-Jurkat cells and that the transfer required the membrane targeting function of Nef and was cell density-dependent. Then, we showed that Nef transfer was cell-cell contact-dependent, as exposure to culture supernatants or exosomes from HIV-infected Jurkat or Nef-expressing Jurkat and 293T led to little Nef detection in the target cells Jurkat. Thirdly, we demonstrated that Nef was only detected to be associated with HIV virions but not with acetylcholinesterase (AChE+) exosomes from HIV-infected Jurkat and not in the exosomes from Nef-expressing Jurkat. In comparison, when it was over-expressed in 293T, Nef was detected in detergent-insoluble AChE+/CD81 low/TSG101 low exosomes, but not in detergent-soluble AChE-/CD81 high/TSG101 high exosomes. Lastly, microscopic imaging showed no significant Nef detection in exosomal vesicle-like structures in and out 293T. Taken together, these results show that exosomes are unlikely involved in intercellular Nef transfer. In addition, this study reveals existence of two types of exosomes: AChE+/CD81 low/TSG101 low exosomes and AChE-/CD81 high/TSG101 high exosomes.


Subject(s)
Exosomes/metabolism , Intracellular Space/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , Acetylcholinesterase/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Bystander Effect/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Detergents/pharmacology , Exosomes/drug effects , Flow Cytometry , Green Fluorescent Proteins/metabolism , HEK293 Cells , HIV Infections/metabolism , HIV Infections/virology , HIV-1/physiology , Humans , Jurkat Cells , Protein Transport/drug effects , Tetraspanin 28/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL