Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Small ; 20(20): e2308680, 2024 May.
Article in English | MEDLINE | ID: mdl-38225709

ABSTRACT

Gut microbiota function has numerous effects on humans and the diet humans consume has emerged as a pivotal determinant of gut microbiota function. Here, a new concept that gut microbiota can be trained by diet-derived exosome-like nanoparticles (ELNs) to release healthy outer membrane vesicles (OMVs) is introduced. Specifically, OMVs released from garlic ELN (GaELNs) trained human gut Akkermansia muciniphila (A. muciniphila) can reverse high-fat diet-induced type 2 diabetes (T2DM) in mice. Oral administration of OMVs released from GaELNs trained A. muciniphila can traffick to the brain where they are taken up by microglial cells, resulting in inhibition of high-fat diet-induced brain inflammation. GaELNs treatment increases the levels of OMV Amuc-1100, P9, and phosphatidylcholines. Increasing the levels of Amuc-1100 and P9 leads to increasing the GLP-1 plasma level. Increasing the levels of phosphatidylcholines is required for inhibition of cGas and STING-mediated inflammation and GLP-1R crosstalk with the insulin pathway that leads to increasing expression of Insulin Receptor Substrate (IRS1 and IRS2) on OMV targeted cells. These findings reveal a molecular mechanism whereby OMVs from plant nanoparticle-trained gut bacteria regulate genes expressed in the brain, and have implications for the treatment of brain dysfunction caused by a metabolic syndrome.


Subject(s)
Brain-Gut Axis , Diabetes Mellitus, Type 2 , Exosomes , Garlic , Gastrointestinal Microbiome , Nanoparticles , Diabetes Mellitus, Type 2/metabolism , Garlic/chemistry , Animals , Nanoparticles/chemistry , Exosomes/metabolism , Mice , Akkermansia , Humans , Male , Diet, High-Fat , Mice, Inbred C57BL , Brain/metabolism , Brain/pathology
2.
J Extracell Vesicles ; 12(2): e12307, 2023 02.
Article in English | MEDLINE | ID: mdl-36754903

ABSTRACT

Extracellular vesicles (EVs) contain more than 100 proteins. Whether there are EVs proteins that act as an 'organiser' of protein networks to generate a new or different biological effect from that identified in EV-producing cells has never been demonstrated. Here, as a proof-of-concept, we demonstrate that EV-G12D-mutant KRAS serves as a leader that forms a protein complex and promotes lung inflammation and tumour growth via the Fn1/IL-17A/FGF21 axis. Mechanistically, in contrast to cytosol derived G12D-mutant KRAS complex from EVs-producing cells, EV-G12D-mutant KRAS interacts with a group of extracellular vesicular factors via fibronectin-1 (Fn1), which drives the activation of the IL-17A/FGF21 inflammation pathway in EV recipient cells. We show that: (i), depletion of EV-Fn1 leads to a reduction of a number of inflammatory cytokines including IL-17A; (ii) induction of IL-17A promotes lung inflammation, which in turn leads to IL-17A mediated induction of FGF21 in the lung; and (iii) EV-G12D-mutant KRAS complex mediated lung inflammation is abrogated in IL-17 receptor KO mice. These findings establish a new concept in EV function with potential implications for novel therapeutic interventions in EV-mediated disease processes.


Subject(s)
Extracellular Vesicles , Lung Neoplasms , Pneumonia , Mice , Animals , Interleukin-17/metabolism , Interleukin-17/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Mutant Proteins/metabolism , Mutant Proteins/therapeutic use , Extracellular Vesicles/metabolism , Lung Neoplasms/drug therapy , Pneumonia/genetics
3.
Cell Host Microbe ; 30(7): 944-960.e8, 2022 07 13.
Article in English | MEDLINE | ID: mdl-35654045

ABSTRACT

The intestinal microbiome releases a plethora of small molecules. Here, we show that the Ruminococcaceae metabolite isoamylamine (IAA) is enriched in aged mice and elderly people, whereas Ruminococcaceae phages, belonging to the Myoviridae family, are reduced. Young mice orally administered IAA show cognitive decline, whereas Myoviridae phage administration reduces IAA levels. Mechanistically, IAA promotes apoptosis of microglial cells by recruiting the transcriptional regulator p53 to the S100A8 promoter region. Specifically, IAA recognizes and binds the S100A8 promoter region to facilitate the unwinding of its self-complementary hairpin structure, thereby subsequently enabling p53 to access the S100A8 promoter and enhance S100A8 expression. Thus, our findings provide evidence that small molecules released from the gut microbiome can directly bind genomic DNA and act as transcriptional coregulators by recruiting transcription factors. These findings further unveil a molecular mechanism that connects gut metabolism to gene expression in the brain with implications for disease development.


Subject(s)
Bacteriophages , Cognitive Dysfunction , Gastrointestinal Microbiome , Amines , Animals , Bacteria , Bacteriophages/genetics , Humans , Mice , Microglia , Tumor Suppressor Protein p53
4.
Theranostics ; 12(3): 1220-1246, 2022.
Article in English | MEDLINE | ID: mdl-35154484

ABSTRACT

Background: Obesity is becoming a global epidemic and reversing the pathological processes underlying obesity and metabolic co-morbidities is challenging. Obesity induced chronic inflammation including brain inflammation is a hallmark of obesity via the gut-brain axis. The objective of this study was to develop garlic exosome-like nanoparticles (GaELNs) that inhibit systemic as well as brain inflammatory activity and reverse a HFD induced obesity in mice. Methods: GELNs were isolated and administrated orally into HFD fed mice. GaELNs were fluorescent labeled for monitoring their in vivo trafficking route after oral administration and quantified the number particles in several tissues. The brain inflammation was determined by measuring inflammatory cytokines by ELISA and real-time PCR. Mitochondrial membrane permeability of microglial cells was determined using JC-10 fluorescence dye. The in vivo apoptotic cell death was quantified by TUNEL assay. The brain metabolites were identified and quantified by LC-MS analysis. Memory function of the mice was determined by several memory functional analysis. The effect of GaELNs on glucose and insulin response of the mice was determined by glucose and insulin tolerance tests. c-Myc localization and interaction with BASP1 and calmodulin was determined by confocal microscopy. Results: Our results show that GaELNs is preferentially taken up microglial cells and inhibits the brain inflammation in HFD mice. GaELN phosphatidic acid (PA) (36:4) is required for the uptake of GaELNs via interaction with microglial BASP1. Formation of the GaELNs/BASP1 complex is required for inhibition of c-Myc mediated expression of STING. GaELN PA binds to BASP1, leading to inhibition of c-Myc expression and activity through competitively binding to CaM with c-Myc transcription factor. Inhibition of STING activity leads to reducing the expression of an array of inflammatory cytokines including IFN-γ and TNF-α. IFN-γ induces the expression of IDO1, which in turn the metabolites generated as IDO1 dependent manner activate the AHR pathway that contributes to developing obesity. The metabolites derived from the GaELNs treated microglial cells promote neuronal differentiation and inhibit mitochondrial mediated neuronal cell death. GaELNs treated HFD mice showed improved memory function and increased glucose tolerance and insulin sensitivity in these mice. Conclusion: Collectively, these results demonstrate how nanoparticles from a healthy diet can inhibit unhealthy high-fat diet induced brain inflammation and reveal a link between brain microglia/diet to brain inflammatory disease outcomes via diet-derived exosome-like nanoparticles.


Subject(s)
Encephalitis , Garlic , Nanoparticles , Animals , Antioxidants , Brain/metabolism , Cytokines/metabolism , Diet, High-Fat/adverse effects , Garlic/metabolism , Glucose , Inflammation/metabolism , Insulin , Mice , Mice, Inbred C57BL , Obesity/metabolism
5.
Theranostics ; 12(3): 1388-1403, 2022.
Article in English | MEDLINE | ID: mdl-35154496

ABSTRACT

Rationale: The obesity epidemic has expanded globally, due in large part to the increased consumption of high-fat diets (HFD), and has increased the risk of major chronic diseases, including type 2 diabetes. Diet manipulation is the foundation of prevention and treatment of obesity and diabetes. The molecular mechanisms that mediate the diet-based prevention of insulin resistance, however, remain to be identified. Here, we report that treatment with orally administered ginger-derived nanoparticles (GDNP) prevents insulin resistance by restoring homeostasis in gut epithelial Foxa2 mediated signaling in mice fed a high-fat diet (HFD). Methods: Ginger-derived nanoparticles (GDNP) were added into drinking water to treat high-fat diet fed mice for at least one year or throughout their life span. A micro array profile of intestinal, liver and fat tissue of GDNP treated mice was used to analyze their gene expression profile. Genes associated with metabolism or insulin signaling were further quantified using the real time polymerase chain reaction (RT-PCR). Surface plasmon resonance (SPR) was used for determining the interaction between Foxa2 protein and phosphatic acid lipid nanoparticles. Results: HFD-feeding inhibited the expression of Foxa2; the GDNPs increased the expression of Foxa2 and protected Foxa2 against Akt-1 mediated phosphorylation and subsequent inactivation of Foxa2. Increasing expression of Foxa2 leads to altering the composition of intestinal epithelial cell (IEC) exosomes of mice fed a HFD and prevents IEC exosome mediated insulin resistance. Collectively, oral administration of GDNP prevents insulin resistance in HFD mice. Interestingly, oral administration of GDNP also extended the life span of the mice and inhibited skin inflammation. Conclusion: Our findings showed that GDNP treatment can prevent HFD-induced obesity and insulin resistance via protecting the Foxa2 from Akt-1 mediated phosphorylation. GDNP treatment provides an alternative approach based on diet manipulation for the development of therapeutic interventions for obesity.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Nanoparticles , Zingiber officinale , Animals , Diet, High-Fat/adverse effects , Hepatocyte Nuclear Factor 3-beta/genetics , Insulin Resistance/physiology , Liposomes , Mice , Mice, Inbred C57BL , Obesity/metabolism , Proto-Oncogene Proteins c-akt
6.
EMBO Rep ; 23(3): e53365, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34994476

ABSTRACT

Bark protects the tree against environmental insults. Here, we analyzed whether this defensive strategy could be utilized to broadly enhance protection against colitis. As a proof of concept, we show that exosome-like nanoparticles (MBELNs) derived from edible mulberry bark confer protection against colitis in a mouse model by promoting heat shock protein family A (Hsp70) member 8 (HSPA8)-mediated activation of the AhR signaling pathway. Activation of this pathway in intestinal epithelial cells leads to the induction of COP9 Constitutive Photomorphogenic Homolog Subunit 8 (COPS8). Utilizing a gut epithelium-specific knockout of COPS8, we demonstrate that COPS8 acts downstream of the AhR pathway and is required for the protective effect of MBELNs by inducing an array of anti-microbial peptides. Our results indicate that MBELNs represent an undescribed mode of inter-kingdom communication in the mammalian intestine through an AhR-COPS8-mediated anti-inflammatory pathway. These data suggest that inflammatory pathways in a microbiota-enriched intestinal environment are regulated by COPS8 and that edible plant-derived ELNs may hold the potential as new agents for the prevention and treatment of gut-related inflammatory disease.


Subject(s)
Colitis , Exosomes , Morus , Nanoparticles , Animals , Colitis/chemically induced , Colitis/metabolism , Colitis/prevention & control , Disease Models, Animal , Exosomes/metabolism , Mice , Mice, Inbred C57BL , Plant Bark/metabolism
7.
Chemosphere ; 292: 133462, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34973255

ABSTRACT

Potentially toxic elements (PTEs) are harmful to plant growth and reduce crop productivity. In this work, we studied three rice genotypes (T-35, RZ-1, and RZ-2) to quantify the diverse PTE effects and tolerances by examining morphology, physiology, and DNA methylation patterns. Morphological results showed that T-35 exhibits the highest tolerance to all studied PTE stressors (Cu, Cd, Cr). Physiological responses under PTE stresses confirmed earlier findings, where T-35 showed a higher potassium (K+) content and more peroxidase (POD) accumulation in the roots than the other two rice genotypes. The differences in PTE tolerance levels observed among the three rice genotypes were also associated with variations in the heavy metal transportation (HMT) gene expression level. Moreover, methylation-sensitive blotting analysis of the selected genes showed that the DNA methylation changes occurring due to PTE treatments are mainly CHG hypomethylation in T-35 but hypermethylation in RZ-1 and RZ-2. Our results demonstrate a tight relationship among physiological response, expression levels of the HMT genes, and DNA methylation pattern under PTEs stresses. It is also indicated that plants use generic mechanisms to tolerate stresses; however, different genotypes employ different combinations of such tactics to confer tolerance, which results in diverse PTE stress tolerances. These findings shed light on the PTE stresses tolerance mechanism and help direct future breeding activities in rice.


Subject(s)
Metals, Heavy , Oryza , DNA Methylation , Gene Expression Regulation, Plant , Genotype , Oryza/genetics , Plant Roots , Stress, Physiological/genetics
8.
Small ; 18(6): e2105385, 2022 02.
Article in English | MEDLINE | ID: mdl-34897972

ABSTRACT

Microglia modulate pro-inflammatory and neurotoxic activities. Edible plant-derived factors improve brain function. Current knowledge of the molecular interactions between edible plant-derived factors and the microglial cell is limited. Here an alcohol-induced chronic brain inflammation model is used to identify that the microglial cell is the novel target of oat nanoparticles (oatN). Oral administration of oatN inhibits brain inflammation and improves brain memory function of mice that are fed alcohol. Mechanistically, ethanol activates dectin-1 mediated inflammatory pathway. OatN is taken up by microglial cells via ß-glucan mediated binding to microglial hippocalcin (HPCA) whereas oatN digalactosyldiacylglycerol (DGDG) prevents assess of oatN ß-glucan to dectin-1. Subsequently endocytosed ß-glucan/HPCA is recruited in an endosomal recycling compartment (ERC) via interaction with Rab11a. This complex then sequesters the dectin-1 in the ERC in an oatN ß-glucan dependent manner and alters the location of dectin-1 from Golgi to early endosomes and lysosomes and increases exportation of dectin-1 into exosomes in an Rab11a dependent manner. Collectively, these cascading actions lead to preventing the activation of the alcoholic induced brain inflammation signing pathway(s). This coordinated assembling of the HPCA/Rab11a/dectin-1 complex by oral administration of oatN may contribute to the prevention of brain inflammation.


Subject(s)
Exosomes , Lectins, C-Type , Memory , Microglia , Nanoparticles , Animals , Avena , Brain , Ethanol/administration & dosage , Lectins, C-Type/metabolism , Memory/physiology , Mice , Microglia/metabolism
9.
iScience ; 24(6): 102511, 2021 Jun 25.
Article in English | MEDLINE | ID: mdl-34142028

ABSTRACT

Diet and bile play critical roles in shaping gut microbiota, but the molecular mechanism underlying interplay with intestinal microbiota is unclear. Here, we showed that lemon-derived exosome-like nanoparticles (LELNs) enhance lactobacilli toleration to bile. To decipher the mechanism, we used Lactobacillus rhamnosus GG (LGG) as proof of concept to show that LELNs enhance LGG bile resistance via limiting production of Msp1 and Msp3, resulting in decrease of bile accessibility to cell membrane. Furthermore, we found that decline of Msps protein levels was regulated through specific tRNAser UCC and tRNAser UCG decay. We identified RNase P, an essential housekeeping endonuclease, being responsible for LELNs-induced tRNAser UCC and tRNAser UCG decay. We further identified galacturonic acid-enriched pectin-type polysaccharide as the active factor in LELNs to increase bile resistance and downregulate tRNAser UCC and tRNAser UCG level in the LGG. Our study demonstrates a tRNA-based gene expression regulation mechanism among lactobacilli to increase bile resistance.

10.
Mol Ther ; 29(8): 2424-2440, 2021 08 04.
Article in English | MEDLINE | ID: mdl-33984520

ABSTRACT

Lung inflammation is a hallmark of coronavirus disease 2019 (COVID-19). In this study, we show that mice develop inflamed lung tissue after being administered exosomes released from the lung epithelial cells exposed to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Nsp12 and Nsp13 (exosomesNsp12Nsp13). Mechanistically, we show that exosomesNsp12Nsp13 are taken up by lung macrophages, leading to activation of nuclear factor κB (NF-κB) and the subsequent induction of an array of inflammatory cytokines. Induction of tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1ß from exosomesNsp12Nsp13-activated lung macrophages contributes to inducing apoptosis in lung epithelial cells. Induction of exosomesNsp12Nsp13-mediated lung inflammation was abolished with ginger exosome-like nanoparticle (GELN) microRNA (miRNA aly-miR396a-5p. The role of GELNs in inhibition of the SARS-CoV-2-induced cytopathic effect (CPE) was further demonstrated via GELN aly-miR396a-5p- and rlcv-miR-rL1-28-3p-mediated inhibition of expression of Nsp12 and spike genes, respectively. Taken together, our results reveal exosomesNsp12Nsp13 as potentially important contributors to the development of lung inflammation, and GELNs are a potential therapeutic agent to treat COVID-19.


Subject(s)
COVID-19/metabolism , Exosomes/metabolism , MicroRNAs/metabolism , Plants/metabolism , Pneumonia/metabolism , A549 Cells , Animals , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Cytokines/metabolism , Epithelial Cells/metabolism , Humans , Interleukin-6/metabolism , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , SARS-CoV-2/pathogenicity , Tumor Necrosis Factor-alpha/metabolism , U937 Cells , Vero Cells
11.
Theranostics ; 11(9): 4061-4077, 2021.
Article in English | MEDLINE | ID: mdl-33754048

ABSTRACT

Background: Diet manipulation is the basis for prevention of obesity and diabetes. The molecular mechanisms that mediate the diet-based prevention of insulin resistance are not well understood. Here, as proof-of-concept, ginger-derived nanoparticles (GDNP) were used for studying molecular mechanisms underlying GDNP mediated prevention of high-fat diet induced insulin resistance. Methods: Ginger-derived nanoparticles (GDNP) were isolated from ginger roots and administered orally to C57BL/6 high-fat diet mice. Fecal exosomes released from intestinal epithelial cells (IECs) of PBS or GDNP treated high-fat diet (HFD) fed mice were isolated by differential centrifugation. A micro-RNA (miRNA) polymerase chain reaction (PCR) array was used to profile the exosomal miRs and miRs of interest were further analyzed by quantitative real time (RT) PCR. miR-375 or antisense-miR375 was packed into nanoparticles made from the lipids extracted from GDNP. Nanoparticles was fluorescent labeled for monitoring their in vivo trafficking route after oral administration. The effect of these nanoparticles on glucose and insulin response of mice was determined by glucose and insulin tolerance tests. Results: We report that HFD feeding increased the expression of AhR and inhibited the expression of miR-375 and VAMP7. Treatment with orally administered ginger-derived nanoparticles (GDNP) resulted in reversing HFD mediated inhibition of the expression of miR-375 and VAMP7. miR-375 knockout mice exhibited impaired glucose homeostasis and insulin resistance. Induction of intracellular miR-375 led to inhibition of the expression of AhR and VAMP7 mediated exporting of miR-375 into intestinal epithelial exosomes where they were taken up by gut bacteria and inhibited the production of the AhR ligand indole. Intestinal exosomes can also traffic to the liver and be taken up by hepatocytes, leading to miR-375 mediated inhibition of hepatic AhR over-expression and inducing the expression of genes associated with the hepatic insulin response. Altogether, GDNP prevents high-fat diet-induced insulin resistance by miR-375 mediated inhibition of the aryl hydrocarbon receptor mediated pathways over activated by HFD feeding. Conclusion: Collectively our findings reveal that oral administration of GDNP to HFD mice improves host glucose tolerance and insulin response via regulating AhR expression by GDNP induced miR-375 and VAMP7.


Subject(s)
Bacteria/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Diet, High-Fat/adverse effects , Insulin Resistance/genetics , Insulin/genetics , MicroRNAs/genetics , Receptors, Aryl Hydrocarbon/genetics , Tryptophanase/genetics , Adult , Animals , Cells, Cultured , Zingiber officinale/chemistry , Hepatocytes/drug effects , Humans , Lipids/genetics , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Nanoparticles/administration & dosage , Obesity/genetics , R-SNARE Proteins/genetics
12.
Nat Commun ; 12(1): 213, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431899

ABSTRACT

High-fat diet (HFD) decreases insulin sensitivity. How high-fat diet causes insulin resistance is largely unknown. Here, we show that lean mice become insulin resistant after being administered exosomes isolated from the feces of obese mice fed a HFD or from patients with type II diabetes. HFD altered the lipid composition of exosomes from predominantly phosphatidylethanolamine (PE) in exosomes from lean animals (L-Exo) to phosphatidylcholine (PC) in exosomes from obese animals (H-Exo). Mechanistically, we show that intestinal H-Exo is taken up by macrophages and hepatocytes, leading to inhibition of the insulin signaling pathway. Moreover, exosome-derived PC binds to and activates AhR, leading to inhibition of the expression of genes essential for activation of the insulin signaling pathway, including IRS-2, and its downstream genes PI3K and Akt. Together, our results reveal HFD-induced exosomes as potential contributors to the development of insulin resistance. Intestinal exosomes thus have potential as broad therapeutic targets.


Subject(s)
Diet, High-Fat , Exosomes/metabolism , Insulin Resistance/genetics , Phosphatidylcholines/metabolism , Up-Regulation/genetics , Adipose Tissue/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Dyslipidemias/complications , Dyslipidemias/genetics , Dyslipidemias/pathology , Epithelial Cells/metabolism , Fatty Liver/complications , Fatty Liver/genetics , Fatty Liver/pathology , Feces , Gene Expression Regulation , Glucose Intolerance , Green Fluorescent Proteins/metabolism , Humans , Insulin/metabolism , Interleukin-6/blood , Intestines/cytology , Lipids/chemistry , Liver/metabolism , Liver/pathology , Macrophage Activation , Mice, Inbred C57BL , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Tetraspanin 30/metabolism , Tumor Necrosis Factor-alpha/blood
13.
iScience ; 23(10): 101571, 2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33083738

ABSTRACT

Clostridioides difficile (C. diff) is the leading cause of antibiotic-associated colitis. Here, we report that lemon exosome-like nanoparticles (LELNs) manipulated probiotics to inhibit C. diff infection (CDI). LELN-manipulated Lactobacillus rhamnosus GG (LGG) and Streptococcus thermophilus ST-21 (STH) (LELN-LS) decrease CDI mortality via an LELN-mediated increase in bile resistance and gut survivability. LELN-LS treatment increases the AhR ligands indole-3-lactic acid (I3LA) and indole-3-carboxaldehyde (I3Ald), leading to induction of IL-22, and increases lactic acid leading to a decrease of C. diff fecal shedding by inhibiting C. diff growth and indole biosynthesis. A synergistic effect between STH and LGG was identified. The STH metabolites inhibit gluconeogenesis of LGG and allow fructose-1,6-bisphosphate (FBP) to accumulate in LGG; accumulated FBP then activates lactate dehydrogenase of LGG (LGG-LDH) and enhances production of lactic acid and the AhR ligand. Our findings provide a new strategy for CDI prevention and treatment with a new type of prebiotics.

15.
iScience ; 21: 308-327, 2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31678913

ABSTRACT

Plant exosomes protect plants against infection; however, whether edible plant exosomes can protect mammalian hosts against infection is not known. In this study, we show that ginger exosome-like nanoparticles (GELNs) are selectively taken up by the periodontal pathogen Porphyromonas gingivalis in a GELN phosphatidic acid (PA) dependent manner via interactions with hemin-binding protein 35 (HBP35) on the surface of P. gingivalis. Compared with PA (34:2), PA (34:1) did not interact with HBP35, indicating that the degree of unsaturation of PA plays a critical role in GELN-mediated interaction with HBP35. On binding to HBP35, pathogenic mechanisms of P. gingivalis were significantly reduced following interaction with GELN cargo molecules, including PA and miRs. These cargo molecules interacted with multiple pathogenic factors in the recipient bacteria simultaneously. Using edible plant exosome-like nanoparticles as a potential therapeutic agent to prevent/treat chronic periodontitis was further demonstrated in a mouse model.

16.
Cell Host Microbe ; 24(5): 637-652.e8, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30449315

ABSTRACT

The gut microbiota can be altered by dietary interventions to prevent and treat various diseases. However, the mechanisms by which food products modulate commensals remain largely unknown. We demonstrate that plant-derived exosome-like nanoparticles (ELNs) are taken up by the gut microbiota and contain RNAs that alter microbiome composition and host physiology. Ginger ELNs (GELNs) are preferentially taken up by Lactobacillaceae in a GELN lipid-dependent manner and contain microRNAs that target various genes in Lactobacillus rhamnosus (LGG). Among these, GELN mdo-miR7267-3p-mediated targeting of the LGG monooxygenase ycnE yields increased indole-3-carboxaldehyde (I3A). GELN-RNAs or I3A, a ligand for aryl hydrocarbon receptor, are sufficient to induce production of IL-22, which is linked to barrier function improvement. These functions of GELN-RNAs can ameliorate mouse colitis via IL-22-dependent mechanisms. These findings reveal how plant products and their effects on the microbiome may be used to target specific host processes to alleviate disease.


Subject(s)
Exosome Multienzyme Ribonuclease Complex/pharmacology , Gastrointestinal Microbiome/drug effects , Intestines/microbiology , Intestines/physiology , MicroRNAs/pharmacology , Plants/chemistry , Animals , Bacterial Proteins , Colitis/therapy , Disease Models, Animal , Disease Susceptibility , Female , Food , Gastrointestinal Microbiome/genetics , Germ-Free Life , Host-Pathogen Interactions , Immunity, Mucosal , Indoles/metabolism , Interleukins/metabolism , Lacticaseibacillus rhamnosus/drug effects , Lacticaseibacillus rhamnosus/genetics , Male , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S/genetics , Receptors, Aryl Hydrocarbon/metabolism , Serine Endopeptidases , Tryptophan/metabolism , Interleukin-22
17.
Theranostics ; 8(18): 4912-4924, 2018.
Article in English | MEDLINE | ID: mdl-30429877

ABSTRACT

Tumor-specific delivery of therapeutics is challenging. One of the major hurdles for successfully delivering targeted agents by nanovectors is the filtering role of the liver in rapidly sequestering nanovectors from the circulation. Exosomes, a type of endogenous nanoparticle, circulate continuously in the peripheral blood and play a role in intercellular communication. The aim of this study was to determine whether the level of endogenous exosomes has an effect on nanovector delivery efficiency of targeted agents. Methods: Exosomes were isolated from peripheral blood and intravenously (I.V.) injected into tumor-bearing mice. Subsequently, 1,1-dioctadecyl-3,3,3'3'-tetramethylindotricarbocyanine-iodide (DiR) fluorescent dye-labeled nanoparticles, including grapefruit nanovectors (GNV) and standard liposomes, were I.V. injected in the mice. The efficiency of redirecting GNVs from liver to other organs of injected mice was further analyzed with in vivo imaging. The concentration of chemo drugs delivered by GNV was measured by HPLC and the anti-lung metastasis therapeutic effects of chemo drugs delivered by GNVs in mouse breast cancer and melanoma cancer models were evaluated. Results: We show that tail vein-injected exosomes isolated from mouse peripheral blood were predominately taken up by liver Kupffer cells. Injection of peripheral blood-derived exosomes before I.V. injection of grapefruit-derived nanovector (GNV) decreased the deposition of GNV in the liver and redirected the GNV to the lung and to the tumor in breast and melanoma tumor-bearing mouse models. Enhanced therapeutic efficiency of doxorubicin (Dox) or paclitaxel (PTX) carried by GNVs for lung metastases was demonstrated when there was an I.V. injection of exosomes before therapeutic treatment. Furthermore, we found that CD36 and IGFR1 receptor-mediated pathways played a critical role in the exosome-mediated inhibitory effect of GNV entry into liver macrophages. Conclusions: Collectively, our findings provide a foundation for using autologous exosomes to enhance therapeutic vector targeted delivery to the lung.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , CD36 Antigens/metabolism , Exosomes , Lung Neoplasms/secondary , Receptors, IgG/metabolism , Tissue Distribution , Administration, Intravenous , Animals , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Disease Models, Animal , Drug Carriers/administration & dosage , Liposomes/administration & dosage , Liposomes/pharmacokinetics , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/drug therapy , Melanoma/diagnostic imaging , Melanoma/drug therapy , Mice , Nanostructures/administration & dosage , Optical Imaging , Treatment Outcome
18.
Mol Ther ; 25(7): 1641-1654, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28274798

ABSTRACT

The intestinal immune system is continuously exposed to massive amounts of nanoparticles derived from food. Whether nanoparticles from plants we eat daily have a role in maintaining intestinal immune homeostasis is poorly defined. Here, we present evidence supporting our hypothesis that edible nanoparticles regulate intestinal immune homeostasis by targeting dendritic cells (DCs). Using three mouse colitis models, our data show that orally given nanoparticles isolated from broccoli extracts protect mice against colitis. Broccoli-derived nanoparticle (BDN)-mediated activation of adenosine monophosphate-activated protein kinase (AMPK) in DCs plays a role in not only prevention of DC activation but also induction of tolerant DCs. Adoptively transferring DCs pre-pulsed with total BDN lipids, but not sulforaphane (SFN)-depleted BDN lipids, prevented DSS-induced colitis in C57BL/6 (B6) mice, supporting the role of BDN SFN in the induction of DC tolerance. Adoptively transferring AMPK+/+, but not AMPK-/-, DCs pre-pulsed with SFN prevented DSS-induced colitis in B6 mice, further supporting the DC AMPK role in SFN-mediated prevention of DSS-induced colitis. This finding could open new preventive or therapeutic avenues to address intestinal-related inflammatory diseases via activating AMPK.


Subject(s)
AMP-Activated Protein Kinases/genetics , Anti-Inflammatory Agents/pharmacology , Brassica/chemistry , Colitis, Ulcerative/prevention & control , Dendritic Cells/drug effects , Nanoparticles/chemistry , AMP-Activated Protein Kinases/metabolism , Administration, Oral , Adoptive Transfer , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/transplantation , Disease Models, Animal , Enzyme Activation/drug effects , Gene Expression , Humans , Immune Tolerance , Isothiocyanates/chemistry , Lipids/isolation & purification , Lipids/pharmacology , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Plant Extracts/chemistry , Sodium Dodecyl Sulfate , Sulfoxides
19.
Nat Commun ; 8: 14448, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28211508

ABSTRACT

Exosomes are emerging mediators of intercellular communication; whether the release of exosomes has an effect on the exosome donor cells in addition to the recipient cells has not been investigated to any extent. Here, we examine different exosomal miRNA expression profiles in primary mouse colon tumour, liver metastasis of colon cancer and naive colon tissues. In more advanced disease, higher levels of tumour suppressor miRNAs are encapsulated in the exosomes. miR-193a interacts with major vault protein (MVP). Knockout of MVP leads to miR-193a accumulation in the exosomal donor cells instead of exosomes, inhibiting tumour progression. Furthermore, miR-193a causes cell cycle G1 arrest and cell proliferation repression through targeting of Caprin1, which upregulates Ccnd2 and c-Myc. Human colon cancer patients with more advanced disease show higher levels of circulating exosomal miR-193a. In summary, our data demonstrate that MVP-mediated selective sorting of tumour suppressor miRNA into exosomes promotes tumour progression.


Subject(s)
Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Progression , Exosomes/metabolism , MicroRNAs/metabolism , Vault Ribonucleoprotein Particles/metabolism , Animals , Base Sequence , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Colonic Neoplasms/blood , Colonic Neoplasms/genetics , Female , Humans , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Mice, Inbred BALB C , MicroRNAs/blood , MicroRNAs/genetics , Models, Biological , Neoplasm Invasiveness , RNA Transport , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...