Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Proc Natl Acad Sci U S A ; 121(20): e2306776121, 2024 May 14.
Article En | MEDLINE | ID: mdl-38709933

A high-fat diet (HFD) is a high-risk factor for the malignant progression of cancers through the disruption of the intestinal microbiota. However, the role of the HFD-related gut microbiota in cancer development remains unclear. This study found that obesity and obesity-related gut microbiota were associated with poor prognosis and advanced clinicopathological status in female patients with breast cancer. To investigate the impact of HFD-associated gut microbiota on cancer progression, we established various models, including HFD feeding, fecal microbiota transplantation, antibiotic feeding, and bacterial gavage, in tumor-bearing mice. HFD-related microbiota promotes cancer progression by generating polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). Mechanistically, the HFD microbiota released abundant leucine, which activated the mTORC1 signaling pathway in myeloid progenitors for PMN-MDSC differentiation. Clinically, the elevated leucine level in the peripheral blood induced by the HFD microbiota was correlated with abundant tumoral PMN-MDSC infiltration and poor clinical outcomes in female patients with breast cancer. These findings revealed that the "gut-bone marrow-tumor" axis is involved in HFD-mediated cancer progression and opens a broad avenue for anticancer therapeutic strategies by targeting the aberrant metabolism of the gut microbiota.


Breast Neoplasms , Cell Differentiation , Diet, High-Fat , Disease Progression , Gastrointestinal Microbiome , Leucine , Myeloid-Derived Suppressor Cells , Animals , Diet, High-Fat/adverse effects , Leucine/metabolism , Female , Humans , Mice , Myeloid-Derived Suppressor Cells/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/microbiology , Breast Neoplasms/metabolism , Obesity/microbiology , Obesity/metabolism , Obesity/pathology , Cell Line, Tumor
2.
EBioMedicine ; 100: 104959, 2024 Feb.
Article En | MEDLINE | ID: mdl-38215690

BACKGROUND: Exclusive enteral nutrition (EEN) is an important alternative strategy for patients with Crohn's disease (CD), and during this process, microbiota alterations have been observed. However, the underlying mechanisms by which EEN reduces intestinal inflammation are currently unclear. METHODS: The therapeutic potential of enteral nutrition (EN) was assessed using various mouse models. Fecal full-length 16S rDNA sequencing analysis and several CD metagenome datasets were used to identify the candidate therapeutic bacteria Faecalibaculum rodentium (F. rodentium). Whole genome sequencing of F. rodentium and widely-targeted metabolome analysis of the supernatant showed that EN-induced F. rodentium accumulation protected against colitis via histidine biosynthesis. FINDINGS: The therapeutic potential of EN therapy was observed in both dextran sulfate sodium (DSS)-induced colitis and Il10-/- spontaneous colitis mouse models. Accumulation of F. rodentium after EN therapy was determined using full-length 16S rDNA sequencing and verified with several metagenome datasets from patients with CD. Colonization of an isolated F. rodentium could reduce colitis in Il10-/- mice. Significant histidine enrichment was observed in the F. rodentium culture supernatant, and a series of histidine biosynthesis genes were observed in the F. rodentium genome. Engineered Escherichia coli Nissle 1917 (EcN), encoding the heterologous hisG of F. rodentium (EcN-hisG), which was a key driver of histidine biosynthesis in F. rodentium, was found to protect against colitis. INTERPRETATION: This study suggests that EN-induced F. rodentium accumulation protects against colitis in mice via gut bacteria-mediated histidine biosynthesis. FUNDING: A full list of funding bodies can be found in the Acknowledgements section.


Colitis , Crohn Disease , Firmicutes , Humans , Animals , Mice , Enteral Nutrition , Interleukin-10/genetics , Histidine , Colitis/etiology , Colitis/therapy , Crohn Disease/microbiology , Bacteria/genetics , Disease Models, Animal , DNA, Ribosomal
3.
Gut Microbes ; 15(1): 2221978, 2023.
Article En | MEDLINE | ID: mdl-37312410

Dietary iron intake is closely related to the incidence of colorectal cancer. However, the interactions among dietary iron, gut microbiota, and epithelial cells in promoting tumorigenesis have rarely been discussed. Here, we report that gut microbiota plays a crucial role in promoting colorectal tumorigenesis in multiple mice models under excessive dietary iron intake. Gut microbiota modulated by excessive dietary iron are pathogenic, irritating the permeability of the gut barrier and causing leakage of lumen bacteria. Mechanistically, epithelial cells released more secretory leukocyte protease inhibitor (SLPI) to combat the leaked bacteria and limit inflammation. The upregulated SLPI acted as a pro-tumorigenic factor and promoted colorectal tumorigenesis by activating the MAPK signaling pathway. Moreover, excessive dietary iron significantly depleted Akkermansiaceae in the gut microbiota; while supplementation with Akkermansia muciniphila could successfully attenuate the tumorigenic effect from excessive dietary iron. Overall, excessive dietary iron perturbs diet - microbiome-epithelium interactions, which contributes to intestinal tumor initiation.


Colorectal Neoplasms , Gastrointestinal Microbiome , Animals , Mice , Iron, Dietary , Secretory Leukocyte Peptidase Inhibitor , Carcinogenesis , Iron
4.
Biomater Sci ; 11(12): 4385-4396, 2023 Jun 13.
Article En | MEDLINE | ID: mdl-37158209

Design of multifunctional nanoplatforms combined with ultrasound-targeted microbubble destruction (UTMD) technology for enhanced tumor accumulation is feasible to solve the bottleneck of theranostics. Herein, we present the development of zwitterion-modified gadolinium (Gd)-chelated core-shell tecto dendrimers (CSTDs) as a nanomedicine platform (PCSTD-Gd) for enhanced magnetic resonance (MR) imaging-guided chemo-gene therapy of orthotopic breast cancer with the assistance of UTMD. In our design, CSTDs synthesized via supramolecular recognition of ß-cyclodextrin and adamantane were covalently linked with tetraazacyclododecane tetraacetic acid-Gd(III) chelators, modified with 1,3-propane sultone to achieve good protein-resistance property, and used for co-delivery of an microRNA 21 inhibitor (miR 21i) and an anticancer drug doxorubicin (DOX). The overall design is quite advantageous and cooperative. The CSTDs with a greater size than single-generation core dendrimers have amplified the enhanced permeability and retention effect for better passive tumor targeting, with a larger r1 relaxivity for sensitive MR imaging and serum-enhanced gene delivery efficiency due to the better compaction ability as well as the protein resistance ability, and with larger interior space for improved drug loading. Through the unique design and the assistance of UTMD, the obtained PCSTD-Gd/DOX/miR 21i polyplexes enable enhanced MR imaging-guided combined chemo-gene therapy of an orthotopic breast cancer model in vivo.


Breast Neoplasms , Dendrimers , MicroRNAs , Humans , Female , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Precision Medicine , Nanomedicine , Doxorubicin/pharmacology , Magnetic Resonance Imaging/methods , Theranostic Nanomedicine/methods
5.
Aging Cell ; 22(6): e13848, 2023 06.
Article En | MEDLINE | ID: mdl-37132117

Gut microbiota associated with longevity plays an important role in the adaptation to damaging stimuli accumulated during the aging process. The mechanism by which the longevity-associated microbiota protects the senescent host remains unclear, while the metabolites of the gut bacteria are of particular interest. Here, an integrated analysis of untargeted metabolomics and 16S rRNA gene sequencing was used to characterize the metabolite and microbiota profiles of long-lived individuals (aged ≥90 years) in comparison to old-elderly (aged 75-89 years), young-elderly (aged 60-74 years), and young to middle-aged (aged ≤59 years) individuals. This novel study constructed both metabolite and microbiota trajectories across aging in populations from Jiaoling county (the seventh longevity town of the world) in China. We found that the long-lived group exhibited remarkably differential metabolomic signatures, highlighting the existence of metabolic heterogeneity with aging. Importantly, we also discovered that long-lived individuals from the familial longevity cohort harbored a microbiome distinguished from that of the general population. Specifically, we identified that the levels of a candidate metabolite, pinane thromboxane A2 (PTA2), which is positively associated with aging, were consistently higher in individuals with familial longevity and their younger descendants than in those of the general population. Furtherly, functional analysis revealed that PTA2 potentiated the efficiency of microglial phagocytosis of ß-amyloid 40 and enhanced an anti-inflammatory phenotype, indicating a protective role of PTA2 toward host health. Collectively, our results improve the understanding of the role of the gut microbiome in longevity and may facilitate the development of strategies for healthy aging.


Healthy Aging , Microbiota , Humans , Healthy Aging/genetics , RNA, Ribosomal, 16S/genetics , Longevity/genetics , Aging/genetics , Metabolomics
7.
Cell Discov ; 9(1): 26, 2023 Mar 07.
Article En | MEDLINE | ID: mdl-36878899

Metabolic reprogramming is a hallmark of cancer. However, it is not well known how metabolism affects cancer progression. We identified that metabolic enzyme acyl-CoA oxidase 1 (ACOX1) suppresses colorectal cancer (CRC) progression by regulating palmitic acid (PA) reprogramming. ACOX1 is highly downregulated in CRC, which predicts poor clinical outcome in CRC patients. Functionally, ACOX1 depletion promotes CRC cell proliferation in vitro and colorectal tumorigenesis in mouse models, whereas ACOX1 overexpression inhibits patient-derived xenograft growth. Mechanistically, DUSP14 dephosphorylates ACOX1 at serine 26, promoting its polyubiquitination and proteasomal degradation, thereby leading to an increase of the ACOX1 substrate PA. Accumulated PA promotes ß-catenin cysteine 466 palmitoylation, which inhibits CK1- and GSK3-directed phosphorylation of ß-catenin and subsequent ß-Trcp-mediated proteasomal degradation. In return, stabilized ß-catenin directly represses ACOX1 transcription and indirectly activates DUSP14 transcription by upregulating c-Myc, a typical target of ß-catenin. Finally, we confirmed that the DUSP14-ACOX1-PA-ß-catenin axis is dysregulated in clinical CRC samples. Together, these results identify ACOX1 as a tumor suppressor, the downregulation of which increases PA-mediated ß-catenin palmitoylation and stabilization and hyperactivates ß-catenin signaling thus promoting CRC progression. Particularly, targeting ß-catenin palmitoylation by 2-bromopalmitate (2-BP) can efficiently inhibit ß-catenin-dependent tumor growth in vivo, and pharmacological inhibition of DUSP14-ACOX1-ß-catenin axis by Nu-7441 reduced the viability of CRC cells. Our results reveal an unexpected role of PA reprogramming induced by dephosphorylation of ACOX1 in activating ß-catenin signaling and promoting cancer progression, and propose the inhibition of the dephosphorylation of ACOX1 by DUSP14 or ß-catenin palmitoylation as a viable option for CRC treatment.

8.
Adv Sci (Weinh) ; 10(12): e2205272, 2023 04.
Article En | MEDLINE | ID: mdl-36802200

Mesenteric adipose tissue (MAT) in Crohn's disease (CD) is associated with transmural inflammation. Extended mesenteric excision can reduce surgical recurrence and improve long-term outcomes, indicating that MAT plays an important role in the pathogenesis of CD. Bacterial translocation has been reported to occur in the MAT of patients with CD (CD-MAT), but the mechanisms by which translocated bacteria lead to intestinal colitis remain unclear. Here it is shown that members of Enterobacteriaceae are highly enriched in CD-MAT compared with non-CD controls. Viable Klebsiella variicola in Enterobacteriaceae is isolated exclusively in CD-MAT and can induce a pro-inflammatory response in vitro and exacerbates colitis both in dextran sulfate sodium (DSS)-induced colitis mice model and IL-10-/- spontaneous colitis mice model. Mechanistically, active type VI secretion system (T6SS) is identified in the genome of K. variicola, which can impair the intestinal barrier by inhibiting the zonula occludens (ZO-1) expression. Dysfunction of T6SS by CRISPR interference system alleviates the inhibitory effect of K. variicola on ZO-1 expression and attenuated colitis in mice. Overall, these findings demonstrate that a novel colitis-promoting bacteria exist in the mesenteric adipose tissue of CD, opening a new therapeutic avenue for colitis management.


Colitis , Type VI Secretion Systems , Animals , Mice , Type VI Secretion Systems/metabolism , Type VI Secretion Systems/therapeutic use , Colitis/chemically induced , Intestines , Adipose Tissue/metabolism
9.
Mol Ther ; 31(2): 585-598, 2023 Feb 01.
Article En | MEDLINE | ID: mdl-38556635

Inflammatory bowel disease (IBD) is a predisposing factor for colitis-associated cancer (CAC). The association between bile acids and the gut microbiota has been demonstrated in colon neoplasia; however, the effect of ursodeoxycholic acid (UDCA) on gut microbiota alteration in development of colitis and CAC is unknown. Our analysis of publicly available datasets demonstrated the association of UDCA treatment and accumulation of Akkermansia. UDCA-mediated alleviation of DSS-induced colitis was microbially dependent. UDCA treatment significantly upregulated Akkermansia colonization in a mouse model. Colonization of Akkermansia was associated with enhancement of the mucus layer upon UDCA treatment as well as activation of bile acid receptors in macrophages. UDCA played a role in CAC prevention and treatment in the AOM-DSS and ApcMin/+-DSS models through downregulation of inflammation and accumulation of Akkermansia. This study suggests that UDCA intervention could reshape intestinal gut homeostasis, facilitating colonization of Akkermansia and preventing and treating colitis and CAC.


Colitis-Associated Neoplasms , Colitis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Mice , Animals , Ursodeoxycholic Acid/adverse effects , Colitis-Associated Neoplasms/complications , Colitis/chemically induced , Colitis/complications , Colitis/drug therapy , Dextran Sulfate/adverse effects , Disease Models, Animal , Mice, Inbred C57BL , Colon
10.
Adv Mater ; 34(47): e2206861, 2022 Nov.
Article En | MEDLINE | ID: mdl-36125843

Construction of multifunctional nanoplatforms to elevate chemotherapeutic efficacy and induce long-term antitumor immunity still remains to be an extreme challenge. Herein, the design of an advanced redox-responsive nanomedicine formulation based on phosphorus dendrimer-copper(II) complexes (1G3 -Cu)- and toyocamycin (Toy)-loaded polymeric nanoparticles (GCT NPs) coated with cancer cell membranes (CM) are reported. The designed GCT@CM NPs with a size of 210 nm are stable under physiological conditions but are rapidly dissociated in the reductive tumor microenvironment to deplete glutathione and release drugs. The co-loading of 1G3 -Cu and Toy within the NPs causes significant tumor cell apoptosis and immunogenic cell death through 1G3 -Cu-induced mitochondrial dysfunction and Toy-mediated amplification of endoplasmic reticulum stress, respectively, thus effectively suppressing tumor growth, promoting dendritic cell maturation, and increasing tumor-infiltrating cytotoxic T lymphocytes. Likewise, the coated CM and the loaded 1G3 -Cu render the GCT@CM NPs with homotypic targeting and T1 -weighted magnetic resonance imaging of tumors, respectively. With the assistance of programmed cell death ligand 1 antibody, the GCT@CM NP-mediated chemotherapy can significantly potentiate tumor immunotherapy for effective inhibition of tumor recurrence and metastasis. The developed GCT@CM NPs hold a great potential for chemotherapy-potentiated immunotherapy of different tumor types through different mechanisms or synergies.


Nanoparticles , Neoplasms , Humans , Endoplasmic Reticulum Stress , Biomimetics , Polymers , Immunotherapy , Neoplasms/drug therapy , Mitochondria , Cell Line, Tumor , Tumor Microenvironment
11.
Microbiome ; 9(1): 228, 2021 11 23.
Article En | MEDLINE | ID: mdl-34814945

BACKGROUND: Mesenteric adipose tissue (mAT) hyperplasia, known as creeping fat is a pathologic characteristic of Crohn's disease (CD). The reserve of creeping fat in surgery is associated with poor prognosis of CD patients, but the mechanism remains unknown. METHODS: Mesenteric microbiome, metabolome, and host transcriptome were characterized using a cohort of 48 patients with CD and 16 non-CD controls. Multidimensional data including 16S ribosomal RNA gene sequencing (16S rRNA), host RNA sequencing, and metabolome were integrated to reveal network interaction. Mesenteric resident bacteria were isolated from mAT and functionally investigated both in the dextran sulfate sodium (DSS) model and in the Il10 gene-deficient (Il10-/-) mouse colitis model to validate their pro-inflammatory roles. RESULTS: Mesenteric microbiota contributed to aberrant metabolites production and transcripts in mATs from patients with CD. The presence of mAT resident microbiota was associated with the development of CD. Achromobacter pulmonis (A. pulmonis) isolated from CD mAT could translocate to mAT and exacerbate both DSS-induced and Il10 gene-deficient (Il10-/-) spontaneous colitis in mice. The levels of A. pulmonis in both mAT and mucous layer from CD patients were higher compared to those from the non-CD group. CONCLUSIONS: This study suggests that the mesenteric microbiota from patients with CD sculpt a detrimental microenvironment and promote intestinal inflammation. Video abstract.


Colitis , Crohn Disease , Microbiota , Adipose Tissue/metabolism , Animals , Colitis/chemically induced , Colitis/metabolism , Crohn Disease/microbiology , Dextran Sulfate , Humans , Mice , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism
12.
Front Immunol ; 11: 2101, 2020.
Article En | MEDLINE | ID: mdl-32983166

Background and Purpose: The mechanism underlying the pathology of neuromyelitis optica spectrum disorders (NMOSD) remains unclear even though antibodies to the water channel protein aquaporin-4 (AQP4) on astrocytes play important roles. Our previous study showed that dysbiosis occurred in the fecal microbiota of NMOSD patients. In this study, we further investigated whether the intestinal barrier and mucosal flora balance are also interrupted in NMOSD patients. Methods: Sigmoid mucosal biopsies were collected by endoscopy from six patients with NMOSD and compared with samples from five healthy control (HC) individuals. These samples were processed for electron microscopy and immunohistochemistry to investigate changes in ultrastructure and in the number and size of intestinal inflammatory cells. Changes in mucosal flora were also analyzed by high-throughput 16S ribosomal RNA gene amplicon sequencing. Results: The results from bacterial rRNA gene sequencing showed that bacterial diversity was decreased, but Streptococcus and Granulicatella were abundant in the colonic mucosa specimens of NMOSD patients compared to the HC individuals. The intercellular space between epithelia of the colonic mucosa was wider in NMOSD patients compared to the HC subjects (p < 0.01), and the expression of tight junction proteins [occludin, claudin-1 and zonula occludens-1 (ZO-1)] in NMOSD patients significantly decreased compared to that in the HC subjects. We also found numerous activated macrophages with many inclusions within the cytoplasm, mast cells with many particles in their cytoplasm, and enlarged plasma cells with rich developed rough endoplasmic reticulum in the lamina propria of the mucosa of the patients with NMOSD. Quantitative analysis showed that the percentages of small CD38+ and CD138+ cells (plasma cells) were lower, but the percentage of larger plasma cells was higher in NMOSD patients. Conclusion: The present study demonstrated that the intestinal barrier was disrupted in the patients with NMOSD, accompanied by dysbiosis and inflammatory activation of the gut. The mucosal microbiota imbalance and inflammatory responses might allow pathogens to cross the damaged intestinal barrier and participate in pathological process in NMOSD. However, further study on the pathological mechanism of NMOSD underlying gut dysbiosis is warranted in the future.


Dysbiosis/microbiology , Gastrointestinal Microbiome , Intestinal Mucosa/metabolism , Neuromyelitis Optica/microbiology , Adult , Bacteria/isolation & purification , Colon, Sigmoid/microbiology , Colon, Sigmoid/pathology , Dysbiosis/immunology , Feces/microbiology , Female , Humans , Inflammation , Intercellular Junctions/ultrastructure , Intestinal Mucosa/microbiology , Intestinal Mucosa/ultrastructure , Male , Microscopy, Electron, Transmission , Middle Aged , Neuromyelitis Optica/immunology , Plasma Cells/pathology , Ribotyping , Young Adult
13.
Mult Scler ; 25(9): 1316-1325, 2019 08.
Article En | MEDLINE | ID: mdl-30113252

BACKGROUND: Intestinal microbiota is an important environmental factor in the initiation and progression of autoimmune diseases. However, investigations on the gut microbiome in neuromyelitis optica spectrum disorders (NMOSD) are relatively insufficient, especially for that of the Asia population. OBJECTIVES: To evaluate whether or not the intestinal microbiota of NMOSD patients had specific microbial signatures. METHODS: Next-generation sequencing and gas chromatography were employed to compare the fecal microbial composition and short-chain fatty acids (SCFAs) spectrum between patients with NMOSD (n = 84) and healthy controls (n = 54). RESULTS: The gut microbial composition of NMOSD distinguished from healthy individuals. Streptococcus, significantly increased in NMOSD, is positively correlated with disease severities (p < 0.05). The use of immunosuppressants results in a decrease of Streptococcus, suggesting that Streptococcus might play a significant role in the pathogenesis of NMOSD. A striking depletion of fecal SCFAs was observed in NMOSD patients (p < 0.0001), with acetate and butyrate showing significantly negative correlation with disease severities (p < 0.05). CONCLUSION: The fecal organismal structures and SCFAs level of patients with NMOSD were distinctive from healthy individuals. These findings not only could be critical events driving the aberrant immune response responsible for the pathogenesis of these disorders but could also provide suggestions for disease therapy.


Dysbiosis/microbiology , Fatty Acids, Volatile , Feces , Gastrointestinal Microbiome , Neuromyelitis Optica/microbiology , Streptococcus , Adult , China , Feces/microbiology , Female , Humans , Male , Middle Aged , Severity of Illness Index
14.
Exp Ther Med ; 14(3): 2633-2636, 2017 Sep.
Article En | MEDLINE | ID: mdl-28962205

Acute renal infarction (ARI) is an uncommon disease and delayed diagnosis or misdiagnosis often leads to rapid deterioration of the patient. As patients with atrial fibrillation and left atrial myxomas are at risk of developing ARI due to cardiogenic thromboembolism, it is pivotal for clinicians to make a correct diagnosis and start a timely treatment. The present study reported on the case of a 68-year-old man who suffered from ARI secondary to thromboembolism associated with atrial fibrillation and left atrial myxomas. His renal function deteriorated rapidly after the onset of the attack. He was treated with anti-coagulant agents with excellent results. Classical anti-coagulation therapy remains to be the effective and safe treatment of ARI.

15.
Can J Physiol Pharmacol ; 94(9): 996-1006, 2016 Sep.
Article En | MEDLINE | ID: mdl-27376621

Edaravone, a radical scavenger, has been recognized as a potential protective agent for cardiovascular diseases. However, little is known about the effect of edaravone in cardiac complications associated with diabetes. Here, we have demonstrated that edaravone prevents cardiac dysfunction and apoptosis in the streptozotocin-induced type 1 diabetic rat heart. Mechanistic studies revealed that edaravone treatment improved cardiac function and restored superoxide dismutase levels. In addition, treatment of diabetic animals by edaravone increased protein expressions of sirtuin-1 (SIRT-1), peroxisome proliferator activated receptor γ coactivator α (PGC-1α), nuclear factor like-2 (NRF-2), and B cell lymphoma 2 (Bcl-2), and reduced protein expressions of Bax and Caspase-3 compared to the control group. High glucose incubation resulted in the production of reactive oxygen species (ROS) and cell death. Treatment of high-glucose-incubated H9c2 cells by edaravone reduced ROS production and cell death. In addition, the treatment of high-glucose-incubated H9c2 cells by edaravone increased the activity of antioxidative stress by increasing SIRT-1, PGC-1α, and NRF-2, and this treatment also reduced apoptosis by increasing Bcl-2 expression and reducing Bax and Caspase-3 expressions. Knockdown SIRT-1 with small interferer RNA abolished the effects of edaravone. Overall, our data demonstrated that edaravone may be an effective agent against the development of diabetic cardiomyopathy.


Antioxidants/pharmacology , Antipyrine/analogs & derivatives , Diabetes Mellitus, Experimental/metabolism , Glucose/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Animals , Antioxidants/metabolism , Antipyrine/antagonists & inhibitors , Antipyrine/pharmacology , Antipyrine/therapeutic use , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Blood Glucose/metabolism , Caspase 3/metabolism , Cell Culture Techniques , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/drug therapy , Edaravone , Free Radical Scavengers/pharmacokinetics , Free Radical Scavengers/therapeutic use , Gene Knockdown Techniques , Hypertrophy/drug therapy , Insulin , Male , NF-E2-Related Factor 2/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/pharmacology , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sirtuin 1/metabolism , Superoxide Dismutase/metabolism , bcl-2-Associated X Protein/metabolism
16.
Minerva Cardioangiol ; 64(6): 586-94, 2016 Dec.
Article En | MEDLINE | ID: mdl-26334114

BACKGROUND: In this study we aimed to investigate molecular mechanisms for rhamnetin in oxidative damaged myocardial cells and provide some new treatment targets for myocardial oxidative stress injury by bioinformatic analysis. METHODS: Two RNA libraries (control and rhamnetin exposure) from H9c2 cells were constructed and sequenced. After differentially expressed genes (DEGs) identified by NOISeq package, pathways and function analyses were conducted by KEGG and GO BP through DAVID online tool, respectively. Additionally, STRING database was used to construct protein-protein interaction (PPI) network and MCODE was performed for module analysis. RESULTS: A total of 703 up- and 749 downregulated genes were screened between rhamnetin and control groups. The up-regulated genes were mainly enriched in Adherens junction, Notch signaling pathway and Focal adhesion pathways as well as biological and cellular regulating processes, while the down-regulated genes were mainly enriched in pathways and processes related with oxidative phosphorylation, diseases cell cycle and metabolism. PPI network was constructed with 609 nodes and the top ten nodes were selected with high degrees. Additionally, two modules were obtained with score >6. CONCLUSIONS: Our study supplied the theoretical basis for uncovering the molecular mechanism of rhamnetin and potential therapeutic targets for myocardial diseases.


Mutagens/toxicity , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Quercetin/analogs & derivatives , Animals , Cell Line , Computational Biology , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Microarray Analysis , Myocytes, Cardiac/pathology , Quercetin/toxicity , Rats
17.
Asian Pac J Cancer Prev ; 12(8): 1973-7, 2011.
Article En | MEDLINE | ID: mdl-22292636

BACKGROUND: Human arrest defective 1 protein (ARD1), as a N-terminal acetyltransferase, has been reported to play a crucial role in tumorigenesis, but the results are somewhat controversial. To explore the clinical and pathological significance of ARD1 in breast tumorigenesis, we analyzed ARD1 status in multiple types of breast disease. METHODS: The expression of ARD1 protein was assessed by immunohistochemistry in 356 cases including 82 invasive ductal carcinomas (IDC), 159 fibroadenomas, 66 hyperplasia of mammary glands, 19 inflammatory breast disease, 30 breast cysts, and in 29 postoperative treatment patients. We assessed the relationship of ARD1 protein with clinical and pathological characteristics using χ2 test. RESULTS: ARD1 protein was observed at 61.0% (50/82), 54.7% (87/159), 37.9% (25/66), 36.8% (7/19) in IDC, fibroadenoma, hyperplasia, and inflammation, respectively, and less than 30.0% for breast cyst. Thus, high ARD1 expression correlated with breast cancer (relative risk = 1.32, P < 0.005). Moreover, the level of ARD1 protein in carcinoma patients was distinctly related to lymph node metastasis and ER status, with 94.0% (47/50) as copmpared to 6.0% (3/50) in metastatic and non-metastatic (P < 0.001), and 84.0% (42/50) and 16.0% (8/50) for ER + and ER - (P < 0.01), respectively. In addition, the level of ARD1 appeared to have potential for evaluation of prognosis in breast cancer patients after postoperative therapy. CONCLUSIONS: These results suggest that ARD1 expression may be as a potential target for exploring the mechanism of breast cancer metastasic to lymph nodes and hormone-responsive regulation.


Acetyltransferases/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Acetyltransferases/biosynthesis , Acetyltransferases/metabolism , Adult , Breast Cyst/genetics , Breast Cyst/metabolism , Breast Cyst/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/secondary , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Female , Fibroadenoma/genetics , Fibroadenoma/metabolism , Fibroadenoma/pathology , Humans , Hyperplasia/genetics , Hyperplasia/metabolism , Hyperplasia/pathology , Immunohistochemistry , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Lymphatic Metastasis , Middle Aged , N-Terminal Acetyltransferase A , N-Terminal Acetyltransferase E , Phenotype , Postoperative Period , Prognosis , Up-Regulation
18.
Sheng Wu Gong Cheng Xue Bao ; 26(1): 57-62, 2010 Jan.
Article Zh | MEDLINE | ID: mdl-20353093

Human arrest defective 1(hARD1) is an acetyltransferase catalyzing the N-terminal acetylation of proteins after translation. The high expression of hARD1 could be an indicator of the breast cancer. In current study, we produced an anti-hARD lp monoclonal antibody that could specifically recognize ARD1 in breast cancer tissues by using the immunohistochemical assay. The full-length His-tag hARD1 protein (1-235 aa) was over-expressed in Escherichia coli, and purified recombinant protein was injected into Balb/c mice to perform immunization procedure. Eight stable positive monoclonal cell lines were isolated. ELISA results demonstrated that all light chains of antibodies were kappa, and the heavy chains displayed three subtypes IgG1, IgG2a and IgG2b, respectively. A monoclonal antibody, which could specifically recognize hARD1 protein in breast cancer tissues, was identified by screening different cancer tissues using antibody-specificity method. Further, the specificity of the antibody was confirmed by Western blotting analysis. Our study would facilitate breast cancer diagnosis by using this ARD1 monoclonal antibody in clinic. Also, this antibody could be used as an important tool for further investigating the role of ARD1 in tumorigenesis.


Acetyltransferases/immunology , Antibodies, Monoclonal/biosynthesis , Biomarkers, Tumor , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Acetyltransferases/genetics , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/immunology , Breast Neoplasms/immunology , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Humans , Immunization , Mice , Mice, Inbred BALB C , N-Terminal Acetyltransferase A , N-Terminal Acetyltransferase E , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology
19.
Oncol Rep ; 21(4): 909-15, 2009 Apr.
Article En | MEDLINE | ID: mdl-19287988

The arrest-defective-1 (ARD1) gene has been reported to be important in yeast cell cycle regulation, and recent studies have shown that human arrest-defective-1 (hARD1) is related to cancer cell proliferation. To investigate the expression pattern of hARD1 protein in cancer tissues, immunohistochemical analysis was performed to analyze the hARD1 expression pattern in 400 cases of 19 types of common cancer and 133 non-cancer samples from 11 tissue types. hARD1 protein was expressed extensively in cancer tissues including glandular carcinoma and squamous cancer, and the positive rate was 71.5% (15/20) in urinary bladder cancer, 62.5% (30/48) in breast cancer and 57.1% (8/14) in cervical carcinoma. The average hARD1-positive rate was 52.3% in cancers and 31.5% in non-cancers, for which the difference was significant (p<0.005). Comparing the staining intensity of different fields in the same section, the hARD1 protein was highly accumulated in cancer cells when compared to the cells adjacent to cancer. The positive rate of breast and intestinal cancer was obviously higher than corresponding non-cancers (p<0.05 and 0.01). These findings suggest that the accumulation of hARD1 protein may be related to carcinogenesis of various types of cancer.


Acetyltransferases/analysis , Neoplasms/chemistry , Acetyltransferases/physiology , Adult , Aged , Aged, 80 and over , Female , Humans , Immunohistochemistry , Male , Middle Aged , N-Terminal Acetyltransferase A , N-Terminal Acetyltransferase E
...