Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.037
Filter
1.
FASEB J ; 38(16): e70002, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39162680

ABSTRACT

Breast cancer is one of the threatening malignant tumors with the highest mortality and incidence rate over the world. There are a lot of breast cancer patients dying every year due to the lack of effective and safe therapeutic drugs. Therefore, it is highly necessary to develop more effective drugs to overcome breast cancer. As a glycoside derivative of apigenin, cosmosiin is characterized by low toxicity, high water solubility, and wide distribution in nature. Additionally, cosmosiin has been shown to perform anti-tumor effects in cervical cancer, hepatocellular carcinoma and melanoma. However, its pharmacological effects on breast cancer and its mechanisms are still unknown. In our study, the anti-breast cancer effect and mechanism of cosmosiin were investigated by using breast cancer models in vivo and in vitro. The results showed that cosmosiin inhibited the proliferation, migration, and adhesion of breast cancer cells in vitro and suppressed the growth of tumor in vivo through binding with AhR and inhibiting it, thus regulating the downstream CYP1A1/AMPK/mTOR and PPARγ/Wnt/ß-catenin signaling pathways. Collectively, our findings have made contribution to the development of novel drugs against breast cancer by targeting AhR and provided a new direction for the research in the field of anti-breast cancer therapy.


Subject(s)
Breast Neoplasms , Cell Proliferation , Cytochrome P-450 CYP1A1 , PPAR gamma , Receptors, Aryl Hydrocarbon , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , PPAR gamma/metabolism , Animals , Receptors, Aryl Hydrocarbon/metabolism , Mice , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A1/genetics , Cell Proliferation/drug effects , Mice, Nude , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Mice, Inbred BALB C , Cell Movement/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Xenograft Model Antitumor Assays , Wnt Signaling Pathway/drug effects
2.
Signal Transduct Target Ther ; 9(1): 195, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39098923

ABSTRACT

Accumulated evidence suggested that gut microbial dysbiosis interplayed with progressive chronic kidney disease (CKD). However, no available therapy is effective in suppressing progressive CKD. Here, using microbiomics in 480 participants including healthy controls and patients with stage 1-5 CKD, we identified an elongation taxonomic chain Bacilli-Lactobacillales-Lactobacillaceae-Lactobacillus-Lactobacillus johnsonii correlated with patients with CKD progression, whose abundance strongly correlated with clinical kidney markers. L. johnsonii abundance reduced with progressive CKD in rats with adenine-induced CKD. L. johnsonii supplementation ameliorated kidney lesion. Serum indole-3-aldehyde (IAld), whose level strongly negatively correlated with creatinine level in CKD rats, decreased in serum of rats induced using unilateral ureteral obstruction (UUO) and 5/6 nephrectomy (NX) as well as late CKD patients. Treatment with IAld dampened kidney lesion through suppressing aryl hydrocarbon receptor (AHR) signal in rats with CKD or UUO, and in cultured 1-hydroxypyrene-induced HK-2 cells. Renoprotective effect of IAld was partially diminished in AHR deficiency mice and HK-2 cells. Our further data showed that treatment with L. johnsonii attenuated kidney lesion by suppressing AHR signal via increasing serum IAld level. Taken together, targeting L. johnsonii might reverse patients with CKD. This study provides a deeper understanding of how microbial-produced tryptophan metabolism affects host disease and discovers potential pathways for prophylactic and therapeutic treatments for CKD patients.


Subject(s)
Lactobacillus johnsonii , Renal Insufficiency, Chronic , Renal Insufficiency, Chronic/microbiology , Renal Insufficiency, Chronic/therapy , Renal Insufficiency, Chronic/pathology , Animals , Rats , Humans , Mice , Male , Lactobacillus johnsonii/genetics , Indoles , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Gastrointestinal Microbiome , Female
3.
Theranostics ; 14(11): 4218-4239, 2024.
Article in English | MEDLINE | ID: mdl-39113799

ABSTRACT

Rationale: The aryl hydrocarbon receptor (AhR) functions in the regulation of intestinal inflammation, but knowledge of the underlying mechanisms in innate immune cells is limited. Here, we investigated the role of AhR in modulating the functions of macrophages in inflammatory bowel disease pathogenesis. Methods: The cellular composition of intestinal lamina propria CD45+ leukocytes in a dextran sulfate sodium (DSS)-induced mouse colitis model was determined by single-cell RNA sequencing. Macrophage pyroptosis was quantified by analysis of lactate dehydrogenase release, propidium iodide staining, enzyme-linked immunosorbent assay, western blot, and flow cytometry. Differentially expressed genes were confirmed by RNA-seq, RT-qPCR, luciferase assay, chromatin immunoprecipitation, and immunofluorescence staining. Results: AhR deficiency mediated dynamic remodeling of the cellular composition of intestinal lamina propria (LP) CD45+ immune cells in a colitis model, with a significant increase in monocyte-macrophage lineage. Mice with AhR deficiency in myeloid cells developed more severe dextran sulfate sodium induced colitis, with concomitant increased macrophage pyroptosis. Dietary supplementation with an AhR pre-ligand, indole-3-carbinol, conferred protection against colitis while protection failed in mice lacking AhR in myeloid cells. Mechanistically, AhR signaling inhibited macrophage pyroptosis by promoting ornithine decarboxylase 1 (Odc1) transcription, to enhance polyamine biosynthesis. The increased polyamine, particularly spermine, inhibited NLRP3 inflammasome assembly and subsequent pyroptosis by suppressing K+ efflux. AHR expression was positively correlated with ODC1 in intestinal mucosal biopsies from patients with ulcerative colitis. Conclusions: These findings suggest a functional role for the AhR/ODC1/polyamine axis in maintaining intestinal homeostasis, providing potential targets for treatment of inflammatory bowel disease.


Subject(s)
Colitis , Dextran Sulfate , Macrophages , Polyamines , Pyroptosis , Receptors, Aryl Hydrocarbon , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Animals , Mice , Macrophages/metabolism , Macrophages/immunology , Colitis/metabolism , Colitis/chemically induced , Colitis/pathology , Humans , Polyamines/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice, Knockout , Inflammation/metabolism , Male , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Basic Helix-Loop-Helix Transcription Factors
4.
Environ Health Perspect ; 132(8): 87005, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39140734

ABSTRACT

BACKGROUND: Exposure to persistent organic pollutants (POPs) and disruptions in the gastrointestinal microbiota have been positively correlated with a predisposition to factors such as obesity, metabolic syndrome, and type 2 diabetes; however, it is unclear how the microbiome contributes to this relationship. OBJECTIVE: This study aimed to explore the association between early life exposure to a potent aryl hydrocarbon receptor (AHR) agonist and persistent disruptions in the microbiota, leading to impaired metabolic homeostasis later in life. METHODS: This study used metagenomics, nuclear magnetic resonance (NMR)- and mass spectrometry (MS)-based metabolomics, and biochemical assays to analyze the gut microbiome composition and function, as well as the physiological and metabolic effects of early life exposure to 2,3,7,8-tetrachlorodibenzofuran (TCDF) in conventional, germ-free (GF), and Ahr-null mice. The impact of TCDF on Akkermansia muciniphila (A. muciniphila) in vitro was assessed using optical density (OD 600), flow cytometry, transcriptomics, and MS-based metabolomics. RESULTS: TCDF-exposed mice exhibited lower abundances of A. muciniphila, lower levels of cecal short-chain fatty acids (SCFAs) and indole-3-lactic acid (ILA), as well as lower levels of the gut hormones glucagon-like peptide 1 (GLP-1) and peptide YY (PYY), findings suggestive of disruption in the gut microbiome community structure and function. Importantly, microbial and metabolic phenotypes associated with early life POP exposure were transferable to GF recipients in the absence of POP carry-over. In addition, AHR-independent interactions between POPs and the microbiota were observed, and they were significantly associated with growth, physiology, gene expression, and metabolic activity outcomes of A. muciniphila, supporting suppressed activity along the ILA pathway. CONCLUSIONS: These data obtained in a mouse model point to the complex effects of POPs on the host and microbiota, providing strong evidence that early life, short-term, and self-limiting POP exposure can adversely impact the microbiome, with effects persisting into later life with associated health implications. https://doi.org/10.1289/EHP13356.


Subject(s)
Benzofurans , Gastrointestinal Microbiome , Homeostasis , Mice, Inbred C57BL , Receptors, Aryl Hydrocarbon , Animals , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Receptors, Aryl Hydrocarbon/metabolism , Mice , Homeostasis/drug effects , Persistent Organic Pollutants , Male , Ligands
5.
Environ Health Perspect ; 132(8): 87003, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39133094

ABSTRACT

BACKGROUND: Currently, many emerging polycyclic aromatic hydrocarbons (PAHs) have been found to be widely present in the environment. However, little has been reported about their toxicity, particularly in relation to CYP1A1. OBJECTIVES: This study aimed to explore the toxicity of naphtho[2,1-a]pyrene (N21aP) and elucidate the mechanism underlying N21aP-induced expression of CYP1A1. METHODS: The concentration and sources of N21aP were detected and analyzed by gas chromatography-triple quadrupole mass spectrometry (GC-MS/MS) and diagnostic ratio analysis. Then the effects of CYP1A1 on the toxicity of N21aP were conducted in male wild-type (WT) and Cyp1a1 knockout mice exposed to N21aP (0.02, 0.2, and 2mg/kg) through intratracheal instillation. Further, the aryl hydrocarbon receptor (AhR) pathway was examined through luciferase and chromatin immunoprecipitation (ChIP) assays. N6-methyladenosine (m6A) modification levels were measured on global RNA and specifically on CYP1A1 mRNA using dot blotting and methylated RNA immunoprecipitation-quantitative real-time polymerase chain reaction (MeRIP qRT-PCR), with validation by m6A inhibitors, DAA and SAH. m6A sites on CYP1A1 were identified by bioinformatics and luciferase assays, and CYP1A1 mRNA's interaction with IGF2BP3 was confirmed by RNA pull-down, luciferase, and RNA binding protein immunoprecipitation (RIP) assays. RESULTS: N21aP was of the same environmental origin as benzo[a]pyrene (BaP) but was more stably present in the environment. N21aP could be metabolically activated by CYP1A1 to produce epoxides, causing DNA damage and further leading to lung inflammation. Importantly, in addition to the classical AhR pathway (i.e., BaP), N21aP also induced CYP1A1 expression with a posttranscriptional modification of m6A in CYP1A1 mRNA via the METTL14-IGF2BP3-CYP1A1 axis. Specifically, in the two recognition sites of METTL14 on the CYP1A1 mRNA transcript (position at 2700 and 5218), a methylation site (position at 5218) in the 3'-untranslated region (UTR) was recognized by IGF2BP3, enhanced the stability of CYP1A1 mRNA, and finally resulted in an increase in CYP1A1 expression. DISCUSSION: This study systematically demonstrated that in addition to AhR-mediated transcriptional regulation, N21aP, had a new additional mechanism of m6A-mediated posttranscriptional modification, jointly contributing to CYP1A1 expression. Given that PAHs are the metabolic substrates of CYP1A1, this study not only helps to understand the significance of environment-genetic interactions for the toxicity of PAHs but also helps to better understand the health risks of the emerging PAHs at environmental exposure levels. https://doi.org/10.1289/EHP14055.


Subject(s)
Cytochrome P-450 CYP1A1 , Receptors, Aryl Hydrocarbon , Animals , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A1/genetics , Mice , Male , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Mice, Knockout , Adenosine/analogs & derivatives , Adenosine/metabolism , Environmental Pollutants/toxicity , RNA Processing, Post-Transcriptional/drug effects
6.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39125717

ABSTRACT

Acute myeloid leukemia (AML) is the most prevalent type of hematopoietic malignancy. Despite recent therapeutic advancements, the high relapse rate associated with extramedullary involvement remains a challenging issue. Moreover, therapeutic targets that regulate the extramedullary infiltration of AML cells are still not fully elucidated. The Aryl Hydrocarbon Receptor (AHR) is known to influence the progression and migration of solid tumors; however, its role in AML is largely unknown. This study explored the roles of AHR in the invasion and migration of AML cells. We found that suppressed expression of AHR target genes correlated with an elevated relapse rate in AML. Treatment with an AHR agonist on patient-derived AML cells significantly decreased genes associated with leukocyte trans-endothelial migration, cell adhesion, and regulation of the actin cytoskeleton. These results were further confirmed in THP-1 and U937 AML cell lines using AHR agonists (TCDD and FICZ) and inhibitors (SR1 and CH-223191). Treatment with AHR agonists significantly reduced Matrigel invasion, while inhibitors enhanced it, regardless of the Matrigel's stiffness. AHR agonists significantly reduced the migration rate and chemokinesis of both cell lines, but AHR inhibitors enhanced them. Finally, we found that the activity of AHR and the expression of NMIIA are negatively correlated. These findings suggest that AHR activity regulates the invasiveness and motility of AML cells, making AHR a potential therapeutic target for preventing extramedullary infiltration in AML.


Subject(s)
Cell Movement , Leukemia, Myeloid, Acute , Myosin Heavy Chains , Neoplasm Invasiveness , Receptors, Aryl Hydrocarbon , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/agonists , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/genetics , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Nonmuscle Myosin Type IIA/metabolism , Nonmuscle Myosin Type IIA/genetics , Cell Line, Tumor , Female , Male , Gene Expression Regulation, Leukemic , Middle Aged , Aged , THP-1 Cells , U937 Cells , Adult , Basic Helix-Loop-Helix Transcription Factors
7.
Science ; 385(6711): eabm6131, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39172838

ABSTRACT

Impaired cerebral glucose metabolism is a pathologic feature of Alzheimer's disease (AD), with recent proteomic studies highlighting disrupted glial metabolism in AD. We report that inhibition of indoleamine-2,3-dioxygenase 1 (IDO1), which metabolizes tryptophan to kynurenine (KYN), rescues hippocampal memory function in mouse preclinical models of AD by restoring astrocyte metabolism. Activation of astrocytic IDO1 by amyloid ß and tau oligomers increases KYN and suppresses glycolysis in an aryl hydrocarbon receptor-dependent manner. In amyloid and tau models, IDO1 inhibition improves hippocampal glucose metabolism and rescues hippocampal long-term potentiation in a monocarboxylate transporter-dependent manner. In astrocytic and neuronal cocultures from AD subjects, IDO1 inhibition improved astrocytic production of lactate and uptake by neurons. Thus, IDO1 inhibitors presently developed for cancer might be repurposed for treatment of AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Astrocytes , Glucose , Glycolysis , Hippocampus , Indoleamine-Pyrrole 2,3,-Dioxygenase , Kynurenine , Long-Term Potentiation , Neurons , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Animals , Hippocampus/metabolism , Glucose/metabolism , Mice , Humans , Astrocytes/metabolism , Amyloid beta-Peptides/metabolism , Kynurenine/metabolism , Neurons/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Cognition/drug effects , Disease Models, Animal , tau Proteins/metabolism , Monocarboxylic Acid Transporters/metabolism , Male , Receptors, Aryl Hydrocarbon/metabolism , Lactic Acid/metabolism , Tryptophan/metabolism , Memory/drug effects
8.
Proc Natl Acad Sci U S A ; 121(35): e2400385121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39167602

ABSTRACT

Type 2 diabetes (T2D) is potentially linked to disordered tryptophan metabolism that attributes to the intricate interplay among diet, gut microbiota, and host physiology. However, underlying mechanisms are substantially unknown. Comparing the gut microbiome and metabolome differences in mice fed a normal diet (ND) and high-fat diet (HFD), we uncover that the gut microbiota-dependent tryptophan metabolite 5-hydroxyindole-3-acetic acid (5-HIAA) is present at lower concentrations in mice with versus without insulin resistance. We further demonstrate that the microbial transformation of tryptophan into 5-HIAA is mediated by Burkholderia spp. Additionally, we show that the administration of 5-HIAA improves glucose intolerance and obesity in HFD-fed mice, while preserving hepatic insulin sensitivity. Mechanistically, 5-HIAA promotes hepatic insulin signaling by directly activating AhR, which stimulates TSC2 transcription and thus inhibits mTORC1 signaling. Moreover, T2D patients exhibit decreased fecal levels of 5-HIAA. Our findings identify a noncanonical pathway of microbially producing 5-HIAA from tryptophan and indicate that 5-HIAA might alleviate the pathogenesis of T2D.


Subject(s)
Diet, High-Fat , Gastrointestinal Microbiome , Insulin Resistance , Liver , Mechanistic Target of Rapamycin Complex 1 , Receptors, Aryl Hydrocarbon , Signal Transduction , Tryptophan , Tuberous Sclerosis Complex 2 Protein , Animals , Diet, High-Fat/adverse effects , Mechanistic Target of Rapamycin Complex 1/metabolism , Tryptophan/metabolism , Gastrointestinal Microbiome/drug effects , Mice , Receptors, Aryl Hydrocarbon/metabolism , Liver/metabolism , Humans , Tuberous Sclerosis Complex 2 Protein/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/microbiology , Male , Mice, Inbred C57BL , Obesity/metabolism , Obesity/microbiology , Basic Helix-Loop-Helix Transcription Factors
9.
Neurobiol Dis ; 199: 106603, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39002811

ABSTRACT

Caffeine consumption outcomes on Amyotrophic Lateral Sclerosis (ALS) including progression, survival and cognition remain poorly defined and may depend on its metabolization influenced by genetic variants. 378 ALS patients with a precise evaluation of their regular caffeine consumption were monitored as part of a prospective multicenter study. Demographic, clinical characteristics, functional disability as measured with revised ALS Functional Rating Scale (ALSFRS-R), cognitive deficits measured using Edinburgh Cognitive and Behavioural ALS Screen (ECAS), survival and riluzole treatment were recorded. 282 patients were genotyped for six single nucleotide polymorphisms tagging different genes involved in caffeine intake and/or metabolism: CYP1A1 (rs2472297), CYP1A2 (rs762551), AHR (rs4410790), POR (rs17685), XDH (rs206860) and ADORA2A (rs5751876) genes. Association between caffeine consumption and ALSFRS-R, ALSFRS-R rate, ECAS and survival were statistically analyzed to determine the outcome of regular caffeine consumption on ALS disease progression and cognition. No association was observed between caffeine consumption and survival (p = 0.25), functional disability (ALSFRS-R; p = 0.27) or progression of ALS (p = 0.076). However, a significant association was found with higher caffeine consumption and better cognitive performance on ECAS scores in patients carrying the C/T and T/T genotypes at rs2472297 (p-het = 0.004). Our results support the safety of regular caffeine consumption on ALS disease progression and survival and also show its beneficial impact on cognitive performance in patients carrying the minor allele T of rs2472297, considered as fast metabolizers, that would set the ground for a new pharmacogenetic therapeutic strategy.


Subject(s)
Amyotrophic Lateral Sclerosis , Caffeine , Cytochrome P-450 CYP1A2 , Disease Progression , Polymorphism, Single Nucleotide , Receptor, Adenosine A2A , Humans , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/drug therapy , Female , Male , Middle Aged , Aged , Receptor, Adenosine A2A/genetics , Cytochrome P-450 CYP1A2/genetics , Cognition/physiology , Cognition/drug effects , Prospective Studies , Cytochrome P-450 CYP1A1/genetics , Receptors, Aryl Hydrocarbon/genetics , Adult , Cognitive Dysfunction/genetics , Riluzole/therapeutic use , Central Nervous System Stimulants/therapeutic use , Basic Helix-Loop-Helix Transcription Factors
10.
Sci Total Environ ; 947: 174721, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39002591

ABSTRACT

The 7-ethoxyresorufin-O-deethylase (EROD) activity was first time characterized in the neotropical fish Cnesterodon decemmaculatus as a biomarker for assessing environmental health in aquatic ecosystems of the Rio de la Plata Basin impacted by organic pollutants agonist of the aryl-hydrocarbon receptor (AhR). Both laboratory and field studies were conducted. Laboratory experiments were run using ß-naphthoflavone (BNF) as an AhR agonist model. A clear concentration-response relationship was found between 1 and 100 µg/L, with a NOEC and LOEC of 1 and 10 µg/L. A fast time-dependent response was observed with a significant induction after 24 h and a plateau from 24 to 48 h up to 264 h of exposure. Differences in basal activity were found between juveniles, females, and males, but induction levels were similar. Both basal activities and induction levels were distinct in the whole body, liver, gill, muscle, brain, and embryos. Fold-change inductions in the respective tissues were: 20, 114, 3, 5, 1, and 14. Maternal transfer and early cyp1a activation were unveiled by embryonic induction. Clear differences in EROD activity were found among juveniles collected in hydrocarbon-polluted streams, beside the La Plata Petrochemical hub, and a reference stream. Similar EROD activities were observed in laboratory and feral fish, usually with values below or above 1,000 pmol/min x mg protein for unexposed or exposed organisms. The study contributes with original information about EROD activity in C. decemmaculatus that encourages the use of both the response as a robust biomarker of exposure and the species as a good sentinel organism to be included in surveillant programs for assessing aquatic pollution by AhR agonist chemicals within the Rio de la Plata Basin within the One Health paradigm.


Subject(s)
Biomarkers , Cytochrome P-450 CYP1A1 , Environmental Monitoring , Receptors, Aryl Hydrocarbon , Water Pollutants, Chemical , Animals , Water Pollutants, Chemical/toxicity , Water Pollutants, Chemical/analysis , Cytochrome P-450 CYP1A1/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Environmental Monitoring/methods , Biomarkers/metabolism , Brazil
11.
Elife ; 122024 Jul 09.
Article in English | MEDLINE | ID: mdl-38980302

ABSTRACT

Trained immunity is the long-term functional reprogramming of innate immune cells, which results in altered responses toward a secondary challenge. Despite indoxyl sulfate (IS) being a potent stimulus associated with chronic kidney disease (CKD)-related inflammation, its impact on trained immunity has not been explored. Here, we demonstrate that IS induces trained immunity in monocytes via epigenetic and metabolic reprogramming, resulting in augmented cytokine production. Mechanistically, the aryl hydrocarbon receptor (AhR) contributes to IS-trained immunity by enhancing the expression of arachidonic acid (AA) metabolism-related genes such as arachidonate 5-lipoxygenase (ALOX5) and ALOX5 activating protein (ALOX5AP). Inhibition of AhR during IS training suppresses the induction of IS-trained immunity. Monocytes from end-stage renal disease (ESRD) patients have increased ALOX5 expression and after 6 days training, they exhibit enhanced TNF-α and IL-6 production to lipopolysaccharide (LPS). Furthermore, healthy control-derived monocytes trained with uremic sera from ESRD patients exhibit increased production of TNF-α and IL-6. Consistently, IS-trained mice and their splenic myeloid cells had increased production of TNF-α after in vivo and ex vivo LPS stimulation compared to that of control mice. These results provide insight into the role of IS in the induction of trained immunity, which is critical during inflammatory immune responses in CKD patients.


Subject(s)
Indican , Kidney Failure, Chronic , Receptors, Aryl Hydrocarbon , Animals , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Kidney Failure, Chronic/immunology , Kidney Failure, Chronic/metabolism , Humans , Mice , Monocytes/immunology , Monocytes/metabolism , Monocytes/drug effects , Arachidonic Acid/metabolism , Male , Immunity, Innate/drug effects , Mice, Inbred C57BL , Arachidonate 5-Lipoxygenase/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Trained Immunity
12.
Nature ; 631(8022): 857-866, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38987586

ABSTRACT

Systemic lupus erythematosus (SLE) is prototypical autoimmune disease driven by pathological T cell-B cell interactions1,2. Expansion of T follicular helper (TFH) and T peripheral helper (TPH) cells, two T cell populations that provide help to B cells, is a prominent feature of SLE3,4. Human TFH and TPH cells characteristically produce high levels of the B cell chemoattractant CXCL13 (refs. 5,6), yet regulation of T cell CXCL13 production and the relationship between CXCL13+ T cells and other T cell states remains unclear. Here, we identify an imbalance in CD4+ T cell phenotypes in patients with SLE, with expansion of PD-1+/ICOS+ CXCL13+ T cells and reduction of CD96hi IL-22+ T cells. Using CRISPR screens, we identify the aryl hydrocarbon receptor (AHR) as a potent negative regulator of CXCL13 production by human CD4+ T cells. Transcriptomic, epigenetic and functional studies demonstrate that AHR coordinates with AP-1 family member JUN to prevent CXCL13+ TPH/TFH cell differentiation and promote an IL-22+ phenotype. Type I interferon, a pathogenic driver of SLE7, opposes AHR and JUN to promote T cell production of CXCL13. These results place CXCL13+ TPH/TFH cells on a polarization axis opposite from T helper 22 (TH22) cells and reveal AHR, JUN and interferon as key regulators of these divergent T cell states.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , CD4-Positive T-Lymphocytes , Chemokine CXCL13 , Interferon Type I , Lupus Erythematosus, Systemic , Proto-Oncogene Proteins c-jun , Receptors, Aryl Hydrocarbon , Female , Humans , Male , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Chemokine CXCL13/metabolism , Epigenomics , Gene Expression Profiling , Interferon Type I/immunology , Interferon Type I/metabolism , Interleukin-22/immunology , Interleukin-22/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/genetics , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Aryl Hydrocarbon/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
13.
Sci Rep ; 14(1): 15706, 2024 07 08.
Article in English | MEDLINE | ID: mdl-38977770

ABSTRACT

Maintaining the mucus layer is crucial for the innate immune system. Urolithin A (Uro A) is a gut microbiota-derived metabolite; however, its effect on mucin production as a physical barrier remains unclear. This study aimed to elucidate the protective effects of Uro A on mucin production in the colon. In vivo experiments employing wild-type mice, NF-E2-related factor 2 (Nrf2)-deficient mice, and wild-type mice treated with an aryl hydrocarbon receptor (AhR) antagonist were conducted to investigate the physiological role of Uro A. Additionally, in vitro assays using mucin-producing cells (LS174T) were conducted to assess mucus production following Uro A treatment. We found that Uro A thickened murine colonic mucus via enhanced mucin 2 expression facilitated by Nrf2 and AhR signaling without altering tight junctions. Uro A reduced mucosal permeability in fluorescein isothiocyanate-dextran experiments and alleviated dextran sulfate sodium-induced colitis. Uro A treatment increased short-chain fatty acid-producing bacteria and propionic acid concentration. LS174T cell studies confirmed that Uro A promotes mucus production through the AhR and Nrf2 pathways. In conclusion, the enhanced intestinal mucus secretion induced by Uro A is mediated through the actions of Nrf-2 and AhR, which help maintain intestinal barrier function.


Subject(s)
Colitis , Coumarins , Intestinal Mucosa , NF-E2-Related Factor 2 , Receptors, Aryl Hydrocarbon , Animals , NF-E2-Related Factor 2/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Mice , Intestinal Mucosa/metabolism , Coumarins/pharmacology , Colitis/metabolism , Colitis/chemically induced , Mucin-2/metabolism , Mucin-2/genetics , Humans , Colon/metabolism , Mice, Inbred C57BL , Signal Transduction/drug effects , Male , Gastrointestinal Microbiome , Mice, Knockout , Dextran Sulfate , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Intestinal Barrier Function
14.
Sci Rep ; 14(1): 14975, 2024 06 28.
Article in English | MEDLINE | ID: mdl-38951170

ABSTRACT

Glioblastoma (GBM) continues to exhibit a discouraging survival rate despite extensive research into new treatments. One factor contributing to its poor prognosis is the tumor's immunosuppressive microenvironment, in which the kynurenine pathway (KP) plays a significant role. This study aimed to explore how KP impacts the survival of newly diagnosed GBM patients. We examined tissue samples from 108 GBM patients to assess the expression levels of key KP markers-tryptophan 2,3-dioxygenase (TDO2), indoleamine 2,3-dioxygenase (IDO1/2), and the aryl hydrocarbon receptor (AhR). Using immunohistochemistry and QuPath software, three tumor cores were analyzed per patient to evaluate KP marker expression. Kaplan-Meier survival analysis and stepwise multivariate Cox regression were used to determine the effect of these markers on patient survival. Results showed that patients with high expression of TDO2, IDO1/2, and AhR had significantly shorter survival times. This finding held true even when controlling for other known prognostic variables, with a hazard ratio of 3.393 for IDO1, 2.775 for IDO2, 1.891 for TDO2, and 1.902 for AhR. We suggest that KP markers could serve as useful tools for patient stratification, potentially guiding future immunomodulating trials and personalized treatment approaches for GBM patients.


Subject(s)
Biomarkers, Tumor , Glioblastoma , Indoleamine-Pyrrole 2,3,-Dioxygenase , Kynurenine , Receptors, Aryl Hydrocarbon , Tryptophan Oxygenase , Humans , Kynurenine/metabolism , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Female , Male , Prognosis , Middle Aged , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Biomarkers, Tumor/metabolism , Tryptophan Oxygenase/metabolism , Aged , Adult , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Kaplan-Meier Estimate , Tumor Microenvironment , Aged, 80 and over , Basic Helix-Loop-Helix Transcription Factors
15.
Cells ; 13(13)2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38994936

ABSTRACT

Although our skin is not the primary visual organ in humans, it acts as a light sensor, playing a significant role in maintaining our health and overall well-being. Thanks to the presence of a complex and sophisticated optotransduction system, the skin interacts with the visible part of the electromagnetic spectrum and with ultraviolet (UV) radiation. Following a brief overview describing the main photosensitive molecules that detect specific electromagnetic radiation and their associated cell pathways, we analyze their impact on physiological functions such as melanogenesis, immune response, circadian rhythms, and mood regulation. In this paper, we focus on 6-formylindolo[3,2-b]carbazole (FICZ), a photo oxidation derivative of the essential amino acid tryptophan (Trp). This molecule is the best endogenous agonist of the Aryl hydrocarbon Receptor (AhR), an evolutionarily conserved transcription factor, traditionally recognized as a signal transducer of both exogenous and endogenous chemical signals. Increasing evidence indicates that AhR is also involved in light sensing within the skin, primarily due to its ligand FICZ, which acts as both a chromophore and a photosensitizer. The biochemical reactions triggered by their interaction impact diverse functions and convey crucial data to our body, thus adding a piece to the complex puzzle of pathways that allow us to decode and elaborate environmental stimuli.


Subject(s)
Carbazoles , Receptors, Aryl Hydrocarbon , Skin , Humans , Receptors, Aryl Hydrocarbon/metabolism , Skin/metabolism , Carbazoles/pharmacology , Light , Animals , Vision, Ocular/physiology , Signal Transduction
16.
Chemosphere ; 363: 142885, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39025314

ABSTRACT

Particulate matter of aerodynamic diameter ≤2.5 µm (PM2.5) exposure induces oxidative stress in lung tissues. Ferroptosis is a form of regulated cell death based on oxidative damage and lipid peroxidation. Whether PM2.5 exposure-induced oxidative stress can promote ferroptosis to aggravate asthma is not known. To investigate if PM2.5 exposure induces oxidative stress to promote ferroptosis and influence asthma development, a cockroach extract-induced asthma model in mice was used for in vivo studies. Airway epithelial cell (AEC) ferroptosis was detected by assays (CCK8, malonaldehyde, and 4-hydroxynonenal). Molecular mechanisms were investigated by real-time reverse transcription-quantitative polymerase chain reaction, western blotting, flow cytometry, liquid chromatography-tandem mass spectrometry, and chromatin immunoprecipitation. We found that exposure to PM2.5 and Indeno[1,2,3-cd] pyrene (IP; one of the prominent absorbed polycyclic aromatic hydrocarbons in PM2.5) enhanced the sensitivity of AECs to ferroptosis to aggravate asthma, whereas ferroptosis inhibitors and cytosolic phospholipase A2 (cPLA2) inhibitors reversed this augmented inflammatory response in mice suffering from asthma. IP treatment enhanced cPLA2 expression/activation through aryl hydrocarbon receptor (AhR) genomic and non-genomic pathways, resulting in arachidonic-acid release to promote the sensitivity of AECs to ferroptosis. IP exposure enhanced the release of leukotriene-B4 from lung macrophages, resulting in enhanced expression of acyl-coA synthetase long chain family member4 (ACSL4) and the sensitivity of AECs to ferroptosis. This finding suggests that exposure to PM2.5 and IP promote ferroptosis sensitivity in AECs to aggravate asthma, which may provide new targets for the prevention and treatment of asthma.


Subject(s)
Asthma , Epithelial Cells , Ferroptosis , Particulate Matter , Ferroptosis/drug effects , Animals , Asthma/chemically induced , Asthma/metabolism , Mice , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Particulate Matter/toxicity , Oxidative Stress/drug effects , Air Pollutants/toxicity , Lung/drug effects , Lung/pathology , Lung/metabolism , Mice, Inbred BALB C , Receptors, Aryl Hydrocarbon/metabolism , Lipid Peroxidation/drug effects
18.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39000041

ABSTRACT

Persistent systemic chronic inflammatory conditions are linked with many pathologies, including cardiovascular diseases (CVDs), a leading cause of death across the globe. Among various risk factors, one of the new possible contributors to CVDs is the metabolism of essential amino acid tryptophan. Proinflammatory signals promote tryptophan metabolism via the kynurenine (KYN) pathway (KP), thereby resulting in the biosynthesis of several immunomodulatory metabolites whose biological effects are associated with the development of symptoms and progression of various inflammatory diseases. Some participants in the KP are agonists of aryl hydrocarbon receptor (AhR), a central player in a signaling pathway that, along with a regulatory influence on the metabolism of environmental xenobiotics, performs a key immunomodulatory function by triggering various cellular mechanisms with the participation of endogenous ligands to alleviate inflammation. An AhR ligand with moderate affinity is the central metabolite of the KP: KYN; one of the subsequent metabolites of KYN-kynurenic acid (KYNA)-is a more potent ligand of AhR. Understanding the role of AhR pathway-related metabolites of the KP that regulate inflammatory factors in cells of the cardiovascular system is interesting and important for achieving effective treatment of CVDs. The purpose of this review was to summarize the results of studies about the participation of the KP metabolite-KYNA-and of the AhR signaling pathway in the regulation of inflammation in pathological conditions of the heart and blood vessels and about the possible interaction of KYNA with AhR signaling in some CVDs.


Subject(s)
Cardiovascular Diseases , Inflammation , Kynurenic Acid , Receptors, Aryl Hydrocarbon , Signal Transduction , Humans , Receptors, Aryl Hydrocarbon/metabolism , Cardiovascular Diseases/metabolism , Kynurenic Acid/metabolism , Inflammation/metabolism , Animals , Kynurenine/metabolism , Tryptophan/metabolism , Basic Helix-Loop-Helix Transcription Factors
19.
J Agric Food Chem ; 72(32): 17912-17923, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39078661

ABSTRACT

Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a complex gastrointestinal condition influenced by genetic, microbial, and environmental factors, among which the gut microbiota plays a crucial role and has emerged as a potential therapeutic target. Ganoderic acid A (GAA), which is a lanostane triterpenoid compound derived from edible mushroom Ganoderma lucidum, has demonstrated the ability to modulate gut dysbiosis. Thus, we investigated the impact of GAA on IBD using a dextran sodium sulfate (DSS)-induced colitis mouse model. GAA effectively prevented colitis, preserved epithelial and mucus layer integrity, and modulated the gut microbiota. In addition, GAA promoted tryptophan metabolism, especially 3-IAld generation, activated the aryl hydrocarbon receptor (AhR), and induced IL-22 production. Fecal microbiota transplantation validated the mediating role of the gut microbiota in the IBD protection conferred by GAA. Our study suggests that GAA holds potential as a nutritional intervention for ameliorating IBD by influencing the gut microbiota, thereby regulating tryptophan metabolism, enhancing AhR activity, and ultimately improving gut barrier function.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Lanosterol , Receptors, Aryl Hydrocarbon , Tryptophan , Animals , Humans , Male , Mice , Bacteria/classification , Bacteria/metabolism , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/drug effects , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Heptanoic Acids , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/microbiology , Interleukin-22 , Lanosterol/analogs & derivatives , Lanosterol/pharmacology , Mice, Inbred C57BL , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Tryptophan/metabolism
20.
Cell Host Microbe ; 32(8): 1347-1364.e10, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39013472

ABSTRACT

Mitochondrial dysfunction is associated with inflammatory bowel diseases (IBDs). To understand how microbial-metabolic circuits contribute to intestinal injury, we disrupt mitochondrial function in the epithelium by deleting the mitochondrial chaperone, heat shock protein 60 (Hsp60Δ/ΔIEC). This metabolic perturbation causes self-resolving tissue injury. Regeneration is disrupted in the absence of the aryl hydrocarbon receptor (Hsp60Δ/ΔIEC;AhR-/-) involved in intestinal homeostasis or inflammatory regulator interleukin (IL)-10 (Hsp60Δ/ΔIEC;Il10-/-), causing IBD-like pathology. Injury is absent in the distal colon of germ-free (GF) Hsp60Δ/ΔIEC mice, highlighting bacterial control of metabolic injury. Colonizing GF Hsp60Δ/ΔIEC mice with the synthetic community OMM12 reveals expansion of metabolically flexible Bacteroides, and B. caecimuris mono-colonization recapitulates the injury. Transcriptional profiling of the metabolically impaired epithelium reveals gene signatures involved in oxidative stress (Ido1, Nos2, Duox2). These signatures are observed in samples from Crohn's disease patients, distinguishing active from inactive inflammation. Thus, mitochondrial perturbation of the epithelium causes microbiota-dependent injury with discriminative inflammatory gene profiles relevant for IBD.


Subject(s)
Chaperonin 60 , Gastrointestinal Microbiome , Mitochondria , Animals , Mice , Mitochondria/metabolism , Humans , Chaperonin 60/genetics , Chaperonin 60/metabolism , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Oxidative Stress , Bacteroides/genetics , Mice, Inbred C57BL , Mice, Knockout , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Gene Expression Profiling , Intestines/microbiology , Intestines/pathology , Disease Models, Animal , Crohn Disease/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL