Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters











Publication year range
1.
Invest Ophthalmol Vis Sci ; 65(8): 50, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39083310

ABSTRACT

Purpose: Retinal microvascular changes are associated with ischemic stroke, and optical coherence tomography angiography (OCTA) is a potential tool to reveal the retinal microvasculature. We investigated the feasibility of using the OCTA image to automatically identify ischemic stroke and its subtypes (i.e. lacunar and non-lacunar stroke), and exploited the association of retinal biomarkers with the subtypes of ischemic stroke. Methods: Two cohorts were included in this study and a total of 1730 eyes from 865 participants were studied. A deep learning model was developed to discriminate the subjects with ischemic stroke from healthy controls and to distinguish the subtypes of ischemic stroke. We also extracted geometric parameters of the retinal microvasculature at different retinal layers to investigate the correlations. Results: Superficial vascular plexus (SVP) yielded the highest areas under the receiver operating characteristic curve (AUCs) of 0.922 and 0.871 for the ischemic stroke detection and stroke subtypes classification, respectively. For external data validation, our model achieved an AUC of 0.822 and 0.766 for the ischemic stroke detection and stroke subtypes classification, respectively. When parameterizing the OCTA images, we showed individuals with ischemic strokes had increased SVP tortuosity (B = 0.085, 95% confidence interval [CI] = 0.005-0.166, P = 0.038) and reduced FAZ circularity (B = -0.212, 95% CI = -0.42 to -0.005, P = 0.045); non-lacunar stroke had reduced SVP FAZ circularity (P = 0.027) compared to lacunar stroke. Conclusions: Our study demonstrates the applicability of artificial intelligence (AI)-enhanced OCTA image analysis for ischemic stroke detection and its subtypes classification. Biomarkers from retinal OCTA images can provide useful information for clinical decision-making and diagnosis of ischemic stroke and its subtypes.


Subject(s)
Biomarkers , Ischemic Stroke , ROC Curve , Retinal Vessels , Tomography, Optical Coherence , Humans , Male , Female , Tomography, Optical Coherence/methods , Retinal Vessels/diagnostic imaging , Retinal Vessels/pathology , Ischemic Stroke/classification , Ischemic Stroke/diagnosis , Ischemic Stroke/diagnostic imaging , Middle Aged , Biomarkers/metabolism , Aged , Deep Learning , Fluorescein Angiography/methods , Fundus Oculi
2.
Front Med (Lausanne) ; 11: 1400137, 2024.
Article in English | MEDLINE | ID: mdl-38808141

ABSTRACT

Background: Ultra-wide-field (UWF) fundus photography represents an emerging retinal imaging technique offering a broader field of view, thus enhancing its utility in screening and diagnosing various eye diseases, notably diabetic retinopathy (DR). However, the application of computer-aided diagnosis for DR using UWF images confronts two major challenges. The first challenge arises from the limited availability of labeled UWF data, making it daunting to train diagnostic models due to the high cost associated with manual annotation of medical images. Secondly, existing models' performance requires enhancement due to the absence of prior knowledge to guide the learning process. Purpose: By leveraging extensively annotated datasets within the field, which encompass large-scale, high-quality color fundus image datasets annotated at either image-level or pixel-level, our objective is to transfer knowledge from these datasets to our target domain through unsupervised domain adaptation. Methods: Our approach presents a robust model for assessing the severity of diabetic retinopathy (DR) by leveraging unsupervised lesion-aware domain adaptation in ultra-wide-field (UWF) images. Furthermore, to harness the wealth of detailed annotations in publicly available color fundus image datasets, we integrate an adversarial lesion map generator. This generator supplements the grading model by incorporating auxiliary lesion information, drawing inspiration from the clinical methodology of evaluating DR severity by identifying and quantifying associated lesions. Results: We conducted both quantitative and qualitative evaluations of our proposed method. In particular, among the six representative DR grading methods, our approach achieved an accuracy (ACC) of 68.18% and a precision (pre) of 67.43%. Additionally, we conducted extensive experiments in ablation studies to validate the effectiveness of each component of our proposed method. Conclusion: In conclusion, our method not only improves the accuracy of DR grading, but also enhances the interpretability of the results, providing clinicians with a reliable DR grading scheme.

3.
Redox Biol ; 67: 102877, 2023 11.
Article in English | MEDLINE | ID: mdl-37690164

ABSTRACT

The senescence of adipose stem cells (ASCs) impairs healthy adipose tissue remodeling, causing metabolic maladaptation to energy surplus. The intrinsic molecular pathways and potential therapy targets for ASC senescence are largely unclear. Here, we showed that visceral ASCs were prone to senescence that was caused by reactive oxygen species (ROS) overload, especially mitochondrial ROS. These senescent ASCs failed to sustain efficient glucose influx, pentose phosphate pathway (PPP) and redox homeostasis. We showed that CD90 silence restricted the glucose uptake by ASCs and thus disrupted their PPP and anti-oxidant system, resulting in ASC senescence. Notably, fibroblast growth factor 21 (FGF21) treatment significantly reduced the senescent phenotypes of ASCs by augmenting CD90 protein via glycosylation, which promoted glucose influx via the AKT-GLUT4 axis and therefore mitigated ROS overload. For diet-induced obese mice, chronic administration of low-dose FGF21 relieved their visceral white adipose tissue (VAT) dysfunction and systemic metabolic disorders. In particular, VAT homeostasis was restored in FGF21-treated obese mice, where ASC repertoire was markedly recovered, accompanied by CD90 elevation and anti-senescent phenotypes in these ASCs. Collectively, we reveal a molecular mechanism of ASC senescence by which CD90 downregulation interferes glucose influx into PPP and redox homeostasis. And we propose a FGF21-based strategy for healthy VAT remodeling, which targets CD90 glycosylation to correct ASC senescence and therefore combat obesity-related metabolic dysfunction.


Subject(s)
Adipose Tissue, White , Glucose , Animals , Mice , Adipose Tissue/metabolism , Adipose Tissue, White/metabolism , Cellular Senescence , Glucose/metabolism , Glycosylation , Mice, Obese , Obesity/metabolism , Reactive Oxygen Species/metabolism , Thy-1 Antigens/metabolism
4.
Metabolism ; 148: 155690, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37717724

ABSTRACT

BACKGROUND: The liver regulates metabolic balance during fasting-feeding cycle. Hepatic adaptation to fasting is precisely modulated on multiple levels. Tumor necrosis factor-α-induced protein 8-like 2 (TIPE2) is a negative regulator of immunity that reduces several liver pathologies, but its physiological roles in hepatic metabolism are largely unknown. METHODS: TIPE2 expression was examined in mouse liver during fasting-feeding cycle. TIPE2-knockout mice, liver-specific TIPE2-knockout mice, liver-specific TIPE2-overexpressed mice were examined for fasting blood glucose and pyruvate tolerance test. Primary hepatocytes or liver tissues from these mice were evaluated for glucose production, lipid accumulation, gene expression and regulatory pathways. TIPE2 interaction with Raf-1 and TIPE2 transcription regulated by PPAR-α were examined using gene overexpression or knockdown, co-immunoprecipitation, western blot, luciferase reporter assay and DNA-protein binding assay. RESULTS: TIPE2 expression was upregulated in fasted mouse liver and starved hepatocytes, which was positively correlated with gluconeogenic genes. Liver-specific TIPE2 deficiency impaired blood glucose homeostasis and gluconeogenic capacity in mice upon fasting, while liver-specific TIPE2 overexpression elevated fasting blood glucose and hepatic gluconeogenesis in mice. In primary hepatocytes upon starvation, TIPE2 interacted with Raf-1 to accelerate its ubiquitination and degradation, resulting in ERK deactivation and FOXO1 maintenance to sustain gluconeogenesis. During prolonged fasting, hepatic TIPE2 deficiency caused aberrant activation of ERK-mTORC1 axis that increased hepatic lipid accumulation via lipogenesis. In hepatocytes upon starvation, PPAR-α bound with TIPE2 promoter and triggered its transcriptional expression. CONCLUSIONS: Hepatocyte TIPE2 is a PPAR-α-induced Raf-1 inactivator that sustains hepatic gluconeogenesis and prevents excessive hepatic lipid accumulation, playing beneficial roles in hepatocyte adaptation to fasting.

5.
Comput Methods Programs Biomed ; 237: 107583, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37167882

ABSTRACT

BACKGROUND AND OBJECTIVES: Hematologic malignancies, including the associated multiple subtypes, are critically threatening to human health. The timely detection of malignancies is crucial for their effective treatment. In this regard, the examination of bone marrow smears constitutes a crucial step. Nonetheless, the conventional approach to cell identification and enumeration is laborious and time-intensive. Therefore, the present study aimed to develop a method for the efficient diagnosis of these malignancies directly from bone marrow microscopic images. METHODS: A deep learning-based framework was developed to facilitate the diagnosis of common hematologic malignancies. First, a total of 2033 microscopic images of bone marrow analysis, including the images for 6 disease types and 1 healthy control, were collected from two Chinese medical websites. Next, the collected images were classified into the training, validation, and test datasets in the ratio of 7:1:2. Subsequently, a method of stain normalization to multi-domains (stain domain augmentation) based on the MultiPathGAN model was developed to equalize the stain styles and expand the image datasets. Afterward, a lightweight hybrid model named MobileViTv2, which integrates the strengths of both CNNs and ViTs, was developed for disease classification. The resulting model was trained and utilized to diagnose patients based on multiple microscopic images of their bone marrow smears, obtained from a cohort of 61 individuals. RESULTS: MobileViTv2 exhibited an average accuracy of 94.28% when applied to the test set, with multiple myeloma, acute lymphocytic leukemia, and lymphoma revealed as the three diseases diagnosed with the highest accuracy values of 98%, 96%, and 96%, respectively. Regarding patient-level prediction, the average accuracy of MobileViTv2 was 96.72%. This model outperformed both CNN and ViT models in terms of accuracy, despite utilizing only 9.8 million parameters. When applied to two public datasets, MobileViTv2 exhibited accuracy values of 99.75% and 99.72%, respectively, and outperformed previous methods. CONCLUSIONS: The proposed framework could be applied directly to bone marrow microscopic images with different stain styles to efficiently establish the diagnosis of common hematologic malignancies.


Subject(s)
Hematologic Neoplasms , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Bone Marrow/diagnostic imaging , Bone Marrow/pathology , Hematologic Neoplasms/diagnostic imaging
6.
Cell Rep ; 42(5): 112424, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37086405

ABSTRACT

Adipose-derived stem cells (ASCs) drive healthy visceral adipose tissue (VAT) expansion via adipocyte hyperplasia. Obesity induces ASC senescence that causes VAT dysfunction and metabolic disorders. It is challenging to restrain this process by biological intervention, as mechanisms of controlling VAT ASC senescence remain unclear. We demonstrate that a population of CX3CR1hi macrophages is maintained in mouse VAT during short-term energy surplus, which sustains ASCs by restraining their senescence, driving adaptive VAT expansion and metabolic health. Long-term overnutrition induces diminishment of CX3CR1hi macrophages in mouse VAT accompanied by ASC senescence and exhaustion, while transferring CX3CR1hi macrophages restores ASC reservoir and triggers VAT beiging to alleviate the metabolic maladaptation. Mechanistically, visceral ASCs attract macrophages via MCP-1 and shape their CX3CR1hi phenotype via exosomes; these macrophages relieve ASC senescence by promoting the arginase1-eIF5A hypusination axis. These findings identify VAT CX3CR1hi macrophages as ASC supporters and unravel their therapeutic potential for metabolic maladaptation to obesity.


Subject(s)
Adipocytes , Intra-Abdominal Fat , Animals , Mice , Intra-Abdominal Fat/metabolism , Adipocytes/metabolism , Macrophages/metabolism , Obesity/metabolism , Cellular Senescence , Adipose Tissue/metabolism , CX3C Chemokine Receptor 1/metabolism
7.
Heliyon ; 9(3): e14104, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36890807

ABSTRACT

Background: Olfactory impairment is a major symptom of COVID-19. Is it necessary for COVID-19 patients to perform the detection of olfactory function, even how to select the olfactory psychophysical assessment tool. Methods: Patients infected with SARS-CoV-2 Delta variant were firstly taken into three categories (mild, moderate, and severe) according to the clinical classification. The Odor Stick Identification Test for the Japanese (OSIT-J) and the Simple Olfactory Test were used to assess olfactory function. Moreover, these patients were divided into three groups based on the results of the olfactory degree (euosmia, hyposmia, and dysosmia), too. The statistical analysis of the correlations between olfaction and clinical characteristics of patients were performed. Results: Our study demonstrated that the elderly men of Han were more susceptible to infected SARS-CoV-2, the clinical symptoms of the COVID-19 patients showed a clear correspondence with the disease type and the degree of olfactory disturbance. Whether or not to vaccinate and whether to complete the whole course of vaccination was closely related to the patient's condition. OSIT-J Test and Simple Test were consistent in our work, indicating that olfactory grading would worsen with the aggravation of symptoms. Furthermore, the OSIT-J method maybe better than Simple Olfactory Test. Conclusion: The vaccination has an important protective effect on the general population, and vaccination should be vigorously promoted. Moreover, it is necessary for COVID-19 patients to perform the detection of olfactory function, and the easier, faster and less expensive method for determination of olfactory function should be utilized to COVID-19 patients as the vital physical examination.

8.
Biol Reprod ; 108(3): 408-422, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36617174

ABSTRACT

Recurrent pregnancy loss (RPL) is a common pathological problem during pregnancy, and its clinical etiology is complex and unclear. Dysfunction of trophoblasts may cause a series of pregnancy complications, including preeclampsia, fetal growth restriction, and RPL. Recently, lncRNAs have been found to be closely related to the occurrence and regulation of pregnancy-related diseases, but few studies have focused on their role in RPL. In this study, we identified a novel lncRNA BBOX1-AS1 that was significantly upregulated in villous tissues and serum of RPL patients. Functionally, BBOX1-AS1 inhibited proliferation, migration, invasion, tube formation and promoted apoptosis of trophoblast cells. Mechanistically, overexpression of BBOX1-AS1 activated the p38 and JNK MAPK signaling pathways by upregulating GADD45A expression. Further studies indicated that BBOX1-AS1 could increase the stability of GADD45A mRNA by binding hnRNPK and ultimately cause abnormal trophoblast function. Collectively, our study highlights that the BBOX1-AS1/hnRNPK/GADD45A axis plays an important role in trophoblast-induced RPL and that BBOX1-AS1 may serve as a potential target for the diagnosis of RPL.


Subject(s)
MicroRNAs , Pre-Eclampsia , RNA, Long Noncoding , Female , Pregnancy , Humans , Trophoblasts/metabolism , Cell Proliferation/genetics , MAP Kinase Signaling System , Pre-Eclampsia/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Cell Movement/genetics , MicroRNAs/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism
9.
FEBS J ; 289(12): 3489-3504, 2022 06.
Article in English | MEDLINE | ID: mdl-35037402

ABSTRACT

As an indispensable process for breast cancer metastasis, tumour angiogenesis requires a tight interaction between cancer cells and endothelial cells in tumour microenvironment. Here, we explored the participation of small extracellular vesicles (sEVs) derived from breast cancer cells in modulating angiogenesis and investigated the effect of IL-35 in facilitating this process. Firstly, we characterized breast cancer cells-derived sEVs untreated or treated with IL-35 and visualized the internalization of these sEVs by human umbilical vein endothelial cells (HUVECs). Breast cancer cells-derived sEVs promoted endothelial cell proliferation through facilitating cell cycle progression and enhanced capillary-like structures formation and microvessel formation. Subsequent results proved that IL-35 further reinforced the angiogenic effect induced by breast cancer cells-derived sEVs. Moreover, sEVs from breast cancer cells significantly enhanced tumour growth and microvessel density in breast tumour-bearing mice model. Microarray analysis showed that IL-35 might alter the mRNA profiles of sEVs and activate the Ras/Raf/MEK/ERK signalling pathway. These findings demonstrated that IL-35 indirectly promoted angiogenesis in breast cancer through regulating the content of breast cancer cells-derived sEVs, which could be internalized by HUVECs.


Subject(s)
Breast Neoplasms , Extracellular Vesicles , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukins/genetics , Interleukins/metabolism , Interleukins/pharmacology , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Tumor Microenvironment
10.
Mol Nutr Food Res ; 65(19): e2100315, 2021 10.
Article in English | MEDLINE | ID: mdl-34363644

ABSTRACT

INTRODUCTION: Obesity causes many life-threatening diseases. It is important to develop effective approaches for obesity treatment. Oral supplementation with spermidine retards age-related processes, but its influences on obesity and various metabolic tissues remain largely unknow. This study aims to investigate the effects of oral spermidine on brown adipose tissue (BAT) and skeletal muscle as well as its roles in counteracting obesity and metabolic disorders. METHODS AND RESULTS: Spermidine is orally administrated into high-fat diet (HFD)-fed mice. The weight gain, insulin resistance, and hepatic steatosis are attenuated by oral spermidine in HFD-fed mice, accompanied by an alleviation of white adipose tissue inflammation. Oral spermidine promotes BAT activation and metabolic adaptation of skeletal muscle in HFD-fed mice, evidenced by UCP-1 induction and CREB activation in both tissues. Notably, oral spermidine upregulates tyrosine hydroxylase in hypothalamus of HFD-fed mice; spermidine treatment increases tyrosine hydroxylase expression and norepinephrine production in neurocytes, which leads to CREB activation and UCP-1 induction in brown adipocytes and myotubes. Spermidine also directly promotes UCP-1 and PGC-1α expression in brown adipocytes and myotubes. CONCLUSION: Spermidine serves as an oral supplement to attenuate obesity and metabolic disorders through hypothalamus-dependent or -independent BAT activation and skeletal muscle adaptation.


Subject(s)
Adipose Tissue, Brown/drug effects , Muscle, Skeletal/drug effects , Obesity/drug therapy , Spermidine/administration & dosage , Spermidine/pharmacology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Administration, Oral , Animals , Diet, High-Fat/adverse effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Insulin Resistance , Male , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity/etiology , Panniculitis/drug therapy , Panniculitis/etiology , Tyrosine 3-Monooxygenase/metabolism
11.
Mol Hum Reprod ; 27(8)2021 08 07.
Article in English | MEDLINE | ID: mdl-34264302

ABSTRACT

Long noncoding RNAs (lncRNAs) have been reported to be involved in various cellular processes and to participate in a variety of human diseases. Recently, increasing studies have reported that lncRNAs are related to many reproductive diseases, such as pathogenesis of recurrent pregnancy loss (RPL), preeclampsia (PE) and gestational diabetes mellitus (GDM). In this study, we aimed to investigate the effect of LINC01088 in trophoblast cells and its potential role in pathogenesis of RPL. LINC01088 was found to be upregulated in first-trimester chorionic villi tissues from RPL patients. Increased LINC01088 repressed proliferation, migration and invasion of trophoblast cells, and promoted apoptosis of trophoblast cells. Further exploration indicated that LINC01088 decreased the production of nitric oxide (NO) by binding and increasing Arginase-1 and decreasing eNOS protein levels. Importantly, JNK and p38 MAPK-signaling pathways were active after overexpression of LINC01088. In conclusion, our studies demonstrated that LINC01088 plays an important role in the pathogenesis of RPL, and is a potential therapeutic target for the treatment of RPL.


Subject(s)
Abortion, Habitual/genetics , MAP Kinase Signaling System/physiology , RNA, Long Noncoding/genetics , Trophoblasts/metabolism , Abortion, Habitual/physiopathology , Adult , Apoptosis , Arginase/metabolism , CRISPR-Cas Systems , Cell Cycle , Cell Division , Cell Line , Cell Movement , Chorionic Villi/metabolism , Chorionic Villi/pathology , Female , HEK293 Cells , Humans , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Pregnancy , Pregnancy Trimester, First , RNA, Guide, Kinetoplastida/genetics , RNA, Long Noncoding/biosynthesis , Trophoblasts/pathology , Up-Regulation , Young Adult
12.
Cell Death Dis ; 12(7): 670, 2021 07 03.
Article in English | MEDLINE | ID: mdl-34218256

ABSTRACT

Breast cancer is the major cause of cancer death worldwide in women. Patients with metastasis have poor prognosis and the mechanisms of breast cancer metastasis are not completely understood. Long non-coding RNAs (lncRNAs) have been shown to have crucial roles in breast cancer development and progression. However, the underlying mechanisms by which lncRNA-driven breast cancer metastasis are unknown. The main objective of this paper is to explore a functional lncRNA and its mechanisms in breast cancer. Here we identified a novel lncRNA AC073352.1 that was significantly upregulated in breast cancer tissues and was associated with advanced TNM stages and poor prognosis in breast cancer patients. In addition, AC073352.1 was found to promote the migration and invasion of breast cancer cells in vitro and enhance breast cancer metastasis in vivo. Mechanistically, we elucidated that AC073352.1 interacted with YBX1 and stabilized its protein expression. Knock down of YBX1 reduced breast cancer cell migration and invasion and could partially reverse the stimulative effects of AC073352.1 overexpressed on breast cancer metastasis. Moreover, AC073352.1 might be packaged into exosomes by binding to YBX1 in breast cancer cells resulting in angiogenesis. Collectively, our results demonstrated that AC073352.1 promoted breast cancer metastasis and angiogenesis via binding YBX1, and it could serve as a promising, novel biomarker for prognosis and a therapeutic target in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , Neovascularization, Pathologic , RNA, Long Noncoding/metabolism , Y-Box-Binding Protein 1/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , MCF-7 Cells , Neoplasm Invasiveness , Neoplasm Metastasis , RNA, Long Noncoding/genetics , Signal Transduction , Y-Box-Binding Protein 1/genetics
13.
Front Oncol ; 10: 515421, 2020.
Article in English | MEDLINE | ID: mdl-33194577

ABSTRACT

Deregulations of long non-coding RNAs (lncRNAs) have been implicated in the progression of breast cancer (BC). However, the prognostic values of those lncRNAs in BC remain elusive. This study aimed at constructing a lncRNA-based prognostic model to improve the clinical management of BC. Systematic investigation of lncRNA expression profiles and clinical data from The Cancer Genome Atlas (TCGA) database were utilized to establish a 10-lncRNA signature. The prognostic signature efficiently discriminated patients with significantly different prognosis regardless of intrinsic molecular subtypes and tumor-node-metastasis (TNM) stage. A combined model was constructed by multivariate Cox proportional hazards regression (CPHR) analysis, which combined the lncRNA-based signature with certain clinical risk factors (TNM stage, age, and human epidermal growth factor receptor 2 status). This model predicted a survival probability that closely corresponds to the actual survival probability. With respect to the entire set, the time-dependent receiver-operating characteristic curves revealed that the area under the curve of this model was the highest than any of the clinical risk factors. Moreover, functional enrichment analysis indicated that the molecular signature was mainly involved in DNA replication, which was firmly related to BC tumorigenesis. Consistent with the discovery, the knockdown of LHX1-DT, one of the 10 prognostic lncRNAs, attenuated the proliferation of BC cells in vitro and in vivo. Taken together, our study constructed a novel 10-lncRNA signature for prediction prognosis, and the signature-based model could provide new insight into accurate management of BC patients.

14.
J Cell Mol Med ; 24(19): 11318-11329, 2020 10.
Article in English | MEDLINE | ID: mdl-32889785

ABSTRACT

Long non-coding RNAs (lncRNAs) have come out as critical molecular regulators of human tumorigenesis. In this study, we sought to identify and functionally characterize lncRNAs as potential mediators of colorectal cancer progression. We screened and identified a novel lncRNA, ADAMTS9-AS1, which was significantly decreased in colorectal cancer tissues and was correlated with clinical outcome of patients according to The Cancer Genome Atlas (TCGA) database. In addition, ADAMTS9-AS1 regulated cell proliferation and migration both in vitro and in vivo. Bioinformatics analysis revealed that overexpression of lncRNA-ADAMTS9-AS1 preferentially affected genes that were linked to proliferation and migration. Mechanistically, we found that ADAMTS9-AS1 obviously suppressed ß-catenin, suggesting that Wnt signalling pathway participates in ADAMTS9-AS1-mediated gene transcriptional regulation in the suppression of colorectal tumorigenesis. Finally, we found that exosomal ADAMTS9-AS1 could serve as a diagnostic biomarker for colorectal cancer with AUC = 0.835 and 95% confidence interval = 0.777-0.911. Our data demonstrated that ADAMTS9-AS1 might play important roles in colorectal cancer by suppressing oncogenesis. Targeting ADAMTS9-AS1 may have potential clinical applications in colorectal cancer prognosis and treatment as an ideal therapeutic target. Finally, exosomal lncRNA-ADAMTS9-AS1 is a promising, novel diagnostic biomarker for colorectal cancer.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , RNA, Long Noncoding/metabolism , Wnt Signaling Pathway/genetics , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/genetics , Down-Regulation/genetics , Epithelial-Mesenchymal Transition/genetics , Exosomes/metabolism , Exosomes/ultrastructure , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , RNA, Long Noncoding/genetics
15.
Aging (Albany NY) ; 12(3): 2857-2879, 2020 02 12.
Article in English | MEDLINE | ID: mdl-32047140

ABSTRACT

Bladder cancer (BCa) is a heterogeneous disease with various tumorigenic mechanisms and clinical behaviors. The current tumor-node-metastasis (TNM) staging system is inadequate to predict overall survival (OS) in BCa patients. We developed a BCa-specific, long-non-coding-RNA (lncRNA)-based nomogram to improve survival prediction in BCa. We obtained the large-scale gene expression profiles of samples from 414 BCa patients in The Cancer Genome Atlas database. Using an lncRNA-mining computational framework, we identified three OS-related lncRNAs among 826 lncRNAs that were differentially expressed between BCa and normal samples. We then constructed a three-lncRNA signature, which efficiently distinguished high-risk from low-risk patients and was even viable in the TNM stage-II, TNM stage-III and ≥65-year-old subgroups (all P<0.05). Using clinical risk factors, we developed a signature-based nomogram, which performed better than the molecular signature or clinical factors alone for prognostic prediction. A bioinformatical analysis revealed that the three OS-related lncRNAs were co-expressed with genes involved in extracellular matrix organization. Functional assays demonstrated that RNF144A-AS1, one of the three OS-related lncRNAs, promoted BCa cell migration and invasion in vitro. Our three-lncRNA signature-based nomogram effectively predicts the prognosis of BCa patients, and could potentially be used for individualized management of such patients.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Nomograms , RNA, Long Noncoding/metabolism , Urinary Bladder Neoplasms/metabolism , Aged , Cell Movement , Female , Humans , Male , Middle Aged , Prognosis , RNA, Long Noncoding/genetics , Risk Factors
16.
Nat Commun ; 10(1): 4601, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31601798

ABSTRACT

During pregnancy, trophoblast cells sustain the maternal-fetal tolerance via expressing and secreting various chemokines and cytokines. Our previous study revealed the expression of interleukin-35 (IL-35) in human first-trimester trophoblasts. Here we show that IL-35 is expressed in both human first-trimester primary trophoblast cells and a trophoblast cell line. Trophoblast cells inhibit the proliferation of human naive conventional T cells (Tconv cells) and convert suppressed Tconv cells into iTR35 in an IL-35-dependent manner. Mechanistically, trophoblast cell derived IL-35 mediates its function through phosphorylation of STAT1 and STAT3. In vivo studies confirm that mice with immunologically spontaneous abortion have lower levels of IL-35 and iTR35 cells at the maternal-fetal interface, and neutralizing anti-IL-35 mAb enhances abortion rates. Meanwhile, exogenous IL-35 induces iTR35 and prevents immunological abortion. Our findings thus suggest that trophoblast cells have a critical function in preserving maternal-fetal tolerance via secreting IL-35 during pregnancy.


Subject(s)
Interleukins/metabolism , Maternal-Fetal Exchange/physiology , T-Lymphocytes, Regulatory/immunology , Trophoblasts/cytology , Animals , Cell Proliferation , Female , Humans , Immune Tolerance , Interleukins/blood , Interleukins/immunology , Interleukins/pharmacology , Male , Maternal-Fetal Exchange/immunology , Mice, Inbred BALB C , Mice, Inbred DBA , Placenta/cytology , Placenta/immunology , Pregnancy , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Trophoblasts/immunology , Trophoblasts/metabolism
17.
Exp Cell Res ; 383(2): 111513, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31362000

ABSTRACT

Regulatory B cells (Breg cells) play critical roles in modulating immune responses during autoimmune diseases and infection. Here we explored the participation of two main Breg subsets, including IL-10+ Breg (B10) and IL-35+ Breg cells, in maintaining successful pregnancy. We first observed an elevated percentage of B10 cells in peripheral blood from first-trimester pregnant women compared with non-pregnant controls. Serum from pregnancy induced the augmentation of B10  in peripheral blood mononuclear cells from non-pregnant women. In animal models, we demonstrated that there were significant augmentation of B10 cells and obvious increase of IL-10 level in splenic B cells from normal pregnant mice compared to that in abortion-prone pregnant mice and virgin mice. Further analysis showed that both hCG and IL-35 suppressed the proliferation of mouse splenic B cells. Moreover, IL-35 induced the expansion of both mouse splenic B10 and IL-35+ Breg cells while hCG only mediated the generation of B10 cells. Subsequent study in mice demonstrated that the activation of STAT1 and STAT3 in B cells caused by IL-35 and the activation of STAT3 caused by hCG were the predominant mechanism of IL-35+ Breg and B10 cells augmentation. These findings suggested that hCG and IL-35 induced the amplification of B10 and IL-35+ Breg cells which played a vital peripheral regulatory role during pregnancy.


Subject(s)
B-Lymphocytes, Regulatory/physiology , Chorionic Gonadotropin/physiology , Immune Tolerance , Interleukin-10/metabolism , Interleukins/physiology , Pregnancy Maintenance/immunology , Abortion, Spontaneous/blood , Abortion, Spontaneous/immunology , Abortion, Spontaneous/pathology , Adult , Animals , B-Lymphocytes, Regulatory/drug effects , B-Lymphocytes, Regulatory/metabolism , Case-Control Studies , Cell Proliferation/drug effects , Cells, Cultured , Chorionic Gonadotropin/blood , Chorionic Gonadotropin/pharmacology , Disease Models, Animal , Female , Humans , Immune Tolerance/drug effects , Interleukins/blood , Interleukins/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Mice, Inbred DBA , Pregnancy , Pregnancy Maintenance/drug effects , Pregnancy Trimester, First/blood , Pregnancy Trimester, First/immunology , Young Adult
18.
Carcinogenesis ; 39(12): 1488-1496, 2018 12 31.
Article in English | MEDLINE | ID: mdl-30321288

ABSTRACT

Interleukin 35 (IL-35) is a potent immunosuppressive cytokine, consisting of an Epstein-Barr virus-induced gene 3 (EBI3) subunit and a p35 subunit. IL-35 is mainly produced by regulatory T and regulatory B cells, and plays a crucial role in the development and prevention of infectious and autoimmune diseases. However, the effect of IL-35 in malignant disease is not well understood. In this study, we demonstrated that breast cancer cells (BCCs) also expressed and secreted IL-35 and higher level of IL-35 in BCCs was closely associated with poor prognosis of patients and was an independent unfavorable prognostic factor for breast cancer. Subsequent study revealed that BCC-derived IL-35 inhibited conventional T (Tconv) cell proliferation and further induced suppressed Tconv cells into IL-35-producing induced regulatory T (iTr35) cells. Furthermore, BCC-derived IL-35 promoted the secretion of inhibitory cytokine IL-10 and obviously decreased the secretion of Th1-type cytokine IFN-γ and Th17-type cytokine IL-17 in Tconv cells. Meanwhile, the expression of inhibitory receptor CD73 was also elevated on the surface of Tconv cells following the BCCs' supernatant treatment. Mechanistically, BCC-derived IL-35 exhausted Tconv cells and induced iTr35 by activating transcription factor STAT1/STAT3. Hence, our results indicate functions of BCC-derived IL-35 in promoting tumor progression through proliferation inhibition of tumor-infiltrating Tconv cells and induction of iTr35 cells in tumor microenvironment. This study highlights that IL-35 produced by BCCs are a potential therapeutic target for breast cancer.


Subject(s)
Breast Neoplasms/immunology , Interleukins/immunology , T-Lymphocytes, Regulatory/immunology , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation/physiology , Disease Progression , Female , HEK293 Cells , Humans , Interferon-gamma/immunology , Interleukin-17/immunology , Lymphocyte Activation/immunology , MCF-7 Cells , Middle Aged , STAT1 Transcription Factor/immunology , STAT3 Transcription Factor/immunology , T-Lymphocytes, Regulatory/pathology , Tumor Microenvironment/immunology
19.
Biochem Biophys Res Commun ; 505(2): 505-510, 2018 10 28.
Article in English | MEDLINE | ID: mdl-30268497

ABSTRACT

Age-related cataract, the most common cause of blindness worldwide, has been found closely associated with ß-crystallin B2 (ßB2 or CRYBB2). MicroRNAs (miRNAs) are the primary epigenetic regulators important for various biological processes. However, the role of miRNAs in the progression of lens cataract remains to be elucidated. In this study, we found a novel signal cascade miR-326-fibroblast growth factor 1 (FGF1)-ßB2 modulating the progression of lens cataract. In brief, miR-326 exacerbated but its antagomirs attenuated H2O2-induced apoptosis of HLEC-B3 human lens epithelial cells. Dual-luciferase reporter assay and Western blot showed that miR-326 inhibited FGF1 expression by directly targeting its mRNA 3'-UTR. Consistent with this result, miR-326 antagomir enhanced FGF1 protein level. In addition to FGF1, miR-326 antagomir also enhanced ßB2 expression and this enhancement was abolished by transfection of HLEC-B3 cells with FGF1 shRNA. These data demonstrated that miR-326 antagomir increased ßB2 expression via upregulating FGF1, which was further confirmed by the studies in a rat model of selenite-induced cataract. This work suggests that miR-326 antagomir might be a promising candidate to prevent progression of age-related cataract.


Subject(s)
Antagomirs/metabolism , Cataract/metabolism , Fibroblast Growth Factor 1/metabolism , MicroRNAs/antagonists & inhibitors , beta-Crystallin B Chain/metabolism , 3' Untranslated Regions , Age Factors , Animals , Apoptosis , Cataract/genetics , Cataract/pathology , Cataract/therapy , Cell Line , Disease Progression , Epithelial Cells/cytology , Fibroblast Growth Factor 1/genetics , Humans , Lens, Crystalline/cytology , Rats, Sprague-Dawley , Up-Regulation
20.
Cytokine ; 108: 43-52, 2018 08.
Article in English | MEDLINE | ID: mdl-29571039

ABSTRACT

IL-35, a novel IL-12 family member, is a potent inhibitory cytokine predominantly produced by regulatory T and B lymphocytes that exerts optimal suppression in immune response. However, it remains unclear whether IL-35 plays an inhibitory role on human dendritic cells. In the present study, we focused on the possible immunosuppressive effect of IL-35 on the differentiation, maturation and function of monocyte-derived DCs (MoDCs). Addition of exogenous IL-35 was able to partially suppress MoDCs differentiation in vitro. Subsequently, LPS was used for the maturation of MoDCs and IL-35 was found to mainly restrain the maturation of MoDCs, characterized by the remarkable down-regulation of costimulatory molecules, CD83 and HLA-DR as well as a reduced production of pro-inflammatory cytokines (IL-12p70, IFN-γ, and TNF-α). Furthermore, IL-35-treated MoDCs exhibited strong inhibition in the proliferation of allogeneic CD4+/CD8+ T lymphocytes. Meanwhile, IL-35-treated MoDCs also suppressed the polarization of naïve CD4+ T lymphocytes towards Th1 phenotype and impaired CD8+ T cells allogeneic responses. And the foregoing suppression of MoDCs maturation and function by IL-35 might be due to the aberrant activation of STAT1/STAT3 and inhibition of p38 MAPK/NF-κB signaling pathway. Our results demonstrated for the first time that IL-35 played a critical role in modulating not only adaptive immune response, but also innate immune response. The inhibitory effect of IL-35 on MoDCs maturation and function may facilitate the development of promising therapeutic interventions in tumors and other diseases.


Subject(s)
Cell Differentiation/drug effects , Dendritic Cells/physiology , Interleukins/genetics , Interleukins/immunology , Lymphocyte Activation/drug effects , Adaptive Immunity/drug effects , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cells, Cultured , Coculture Techniques , Cytokines/analysis , Cytokines/immunology , Dendritic Cells/drug effects , Humans , Immunity, Innate/drug effects , Interleukins/pharmacology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/immunology , Monocytes/immunology , Monocytes/physiology , Th1 Cells/immunology , Th1 Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL