Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Int J Med Sci ; 21(6): 983-993, 2024.
Article in English | MEDLINE | ID: mdl-38774750

ABSTRACT

Previous studies have highlighted the protective effects of pyruvate kinase M2 (PKM2) overexpression in septic cardiomyopathy. In our study, we utilized cardiomyocyte-specific PKM2 knockout mice to further investigate the role of PKM2 in attenuating LPS-induced myocardial dysfunction, focusing on mitochondrial biogenesis and prohibitin 2 (PHB2). Our findings confirmed that the deletion of PKM2 in cardiomyocytes significantly exacerbated LPS-induced myocardial dysfunction, as evidenced by impaired contractile function and relaxation. Additionally, the deletion of PKM2 intensified LPS-induced myocardial inflammation. At the molecular level, LPS triggered mitochondrial dysfunction, characterized by reduced ATP production, compromised mitochondrial respiratory complex I/III activities, and increased ROS production. Intriguingly, the absence of PKM2 further worsened LPS-induced mitochondrial damage. Our molecular investigations revealed that LPS disrupted mitochondrial biogenesis in cardiomyocytes, a disruption that was exacerbated by the absence of PKM2. Given that PHB2 is known as a downstream effector of PKM2, we employed PHB2 adenovirus to restore PHB2 levels. The overexpression of PHB2 normalized mitochondrial biogenesis, restored mitochondrial integrity, and promoted mitochondrial function. Overall, our results underscore the critical role of PKM2 in regulating the progression of septic cardiomyopathy. PKM2 deficiency impeded mitochondrial biogenesis, leading to compromised mitochondrial integrity, increased myocardial inflammation, and impaired cardiac function. The overexpression of PHB2 mitigated the deleterious effects of PKM2 deletion. This discovery offers a novel insight into the molecular mechanisms underlying septic cardiomyopathy and suggests potential therapeutic targets for intervention.


Subject(s)
Cardiomyopathies , Mice, Knockout , Mitochondria, Heart , Myocytes, Cardiac , Prohibitins , Pyruvate Kinase , Sepsis , Animals , Cardiomyopathies/pathology , Cardiomyopathies/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/etiology , Mice , Myocytes, Cardiac/pathology , Myocytes, Cardiac/metabolism , Sepsis/metabolism , Sepsis/pathology , Sepsis/genetics , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Humans , Organelle Biogenesis , Lipopolysaccharides/toxicity , Male , Disease Models, Animal
2.
Int J Med Sci ; 21(5): 809-816, 2024.
Article in English | MEDLINE | ID: mdl-38617011

ABSTRACT

This comprehensive review delves into the pivotal role of mitochondria in doxorubicin-induced cardiotoxicity, a significant complication limiting the clinical use of this potent anthracycline chemotherapeutic agent. Doxorubicin, while effective against various malignancies, is associated with dose-dependent cardiotoxicity, potentially leading to irreversible cardiac damage. The review meticulously dissects the molecular mechanisms underpinning this cardiotoxicity, particularly focusing on mitochondrial dysfunction, a central player in this adverse effect. Central to the discussion is the concept of mitochondrial quality control (MQC), including mitochondrial dynamics (fusion/fission balance) and mitophagy. The review presents evidence linking aberrations in these processes to cardiotoxicity in doxorubicin-treated patients. It elucidates how doxorubicin disrupts mitochondrial dynamics, leading to an imbalance between mitochondrial fission and fusion, and impairs mitophagy, culminating in the accumulation of dysfunctional mitochondria and subsequent cardiac cell damage. Furthermore, the review explores emerging therapeutic strategies targeting mitochondrial dysfunction. It highlights the potential of modulating mitochondrial dynamics and enhancing mitophagy to mitigate doxorubicin-induced cardiac damage. These strategies include pharmacological interventions with mitochondrial fission inhibitors, fusion promoters, and agents that modulate mitophagy. The review underscores the promising results from preclinical studies while advocating for more extensive clinical trials to validate these approaches in human patients. In conclusion, this review offers valuable insights into the intricate relationship between mitochondrial dysfunction and doxorubicin-mediated cardiotoxicity. It underscores the need for continued research into targeted mitochondrial therapies as a means to improve the cardiac safety profile of doxorubicin, thereby enhancing the overall treatment outcomes for cancer patients.


Subject(s)
Cardiotoxicity , Mitochondrial Diseases , Humans , Cardiotoxicity/etiology , Doxorubicin/adverse effects , Mitochondria , Anthracyclines
3.
Cell Stress Chaperones ; 29(2): 349-357, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38485043

ABSTRACT

This comprehensive review delves into the pivotal role of mitochondria in doxorubicin-induced cardiotoxicity, a significant complication limiting the clinical use of this potent anthracycline chemotherapeutic agent. Doxorubicin, while effective against various malignancies, is associated with dose-dependent cardiotoxicity, potentially leading to irreversible cardiac damage. The review meticulously dissects the molecular mechanisms underpinning this cardiotoxicity, particularly focusing on mitochondrial dysfunction, a central player in this adverse effect. Central to the discussion is the concept of mitochondrial quality control, including mitochondrial dynamics (fusion/fission balance) and mitophagy. The review presents evidence linking aberrations in these processes to cardiotoxicity in doxorubicin-treated patients. It elucidates how doxorubicin disrupts mitochondrial dynamics, leading to an imbalance between mitochondrial fission and fusion, and impairs mitophagy, culminating in the accumulation of dysfunctional mitochondria and subsequent cardiac cell damage. Furthermore, the review explores emerging therapeutic strategies targeting mitochondrial dysfunction. It highlights the potential of modulating mitochondrial dynamics and enhancing mitophagy to mitigate doxorubicin-induced cardiac damage. These strategies include pharmacological interventions with mitochondrial fission inhibitors, fusion promoters, and agents that modulate mitophagy. The review underscores the promising results from preclinical studies while advocating for more extensive clinical trials to validate these approaches in human patients. In conclusion, this review offers valuable insights into the intricate relationship between mitochondrial dysfunction and doxorubicin-mediated cardiotoxicity. It underscores the need for continued research into targeted mitochondrial therapies as a means to improve the cardiac safety profile of doxorubicin, thereby enhancing the overall treatment outcomes for cancer patients.


Subject(s)
Cardiotoxicity , Mitochondrial Diseases , Humans , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Cardiotoxicity/pathology , Doxorubicin/adverse effects , Mitochondria , Antibiotics, Antineoplastic/adverse effects , Mitochondrial Diseases/complications , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Myocytes, Cardiac
4.
J Cell Physiol ; 239(4): e31192, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38284280

ABSTRACT

Obesity and metabolic diseases, such as insulin resistance, type 2 diabetes, nonalcoholic fatty liver disease, and cardiovascular ailments, represent formidable global health challenges, bearing considerable implications for both morbidity and mortality rates. It has become increasingly evident that chronic, low-grade inflammation plays a pivotal role in the genesis and advancement of these conditions. The involvement of C-C chemokine ligand 2 (CCL2) and its corresponding receptor, C-C chemokine receptor 2 (CCR2), has been extensively documented in numerous inflammatory maladies. Recent evidence indicates that the CCL2/CCR2 pathway extends beyond immune cell recruitment and inflammation, exerting a notable influence on the genesis and progression of metabolic syndrome. The present review seeks to furnish a comprehensive exposition of the CCL2-CCR2 signaling axis within the context of obesity and metabolic disorders, elucidating its molecular mechanisms, functional roles, and therapeutic implications.


Subject(s)
Metabolic Diseases , Obesity , Receptors, CCR2 , Humans , Chemokine CCL2/metabolism , Inflammation , Ligands , Metabolic Diseases/metabolism , Obesity/metabolism , Receptors, CCR2/metabolism , Signal Transduction , Animals , Mice
5.
Cell Signal ; 115: 111040, 2024 03.
Article in English | MEDLINE | ID: mdl-38199596

ABSTRACT

MBIP is a component of the Ada2A containing complex (ATAC) and has been identified as a susceptibility gene in several cancers. However, the role and molecular mechanism of MBIP in esophageal squamous cell carcinoma (ESCC) remain unclear. Our finding indicated that the expression level of MBIP in ESCC was higher than that in normal tissue (P < 0.05) based on the data from the Cancer Gene Atlas (TCGA) and Gene Expression Omnibus (GEO). Kaplan-Meier analysis showed that high MBIP expression was closely associated with deeper invasion and worse prognosis. Transwell assay and mouse xenograft assay demonstrated that MBIP overexpression promoted migration and invasion in vitro and in vivo, while MBIP knockdown played the opposite role. Furthermore, the results of RNA-seq, qRT-PCR, western blotting and rescue experiments revealed that MBIP promoted epithelial-mesenchymal transition (EMT) via the phosphorylation JNK/p38 in ESCC. Our study indicates that MBIP plays a significant role in the prognosis and metastasis of ESCC, suggesting that MBIP might serve as an ESCC prognostic biomarker.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Methylglycosides , Animals , Mice , Humans , Esophageal Squamous Cell Carcinoma/genetics , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/metabolism , Cell Proliferation/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Neoplasm Invasiveness/genetics , Intracellular Signaling Peptides and Proteins/metabolism
6.
Gene ; 896: 148038, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38036077

ABSTRACT

BACKGROUND: Hemophilia A is caused by a deficiency of coagulation factor VIII in the body due to a defect in the F8 gene. The emergence of CRISPR/Cas9 gene editing technology will make it possible to alter the expression of the F8 gene in hemophiliacs, while achieving a potential cure for the disease. METHODS: Initially, we identified high-activity variants of FVIII and constructed donor plasmids using enzymatic digestion and ligation techniques. Subsequently, the donor plasmids were co-transfected with sgRNA-Cas9 protein into mouse Neuro-2a cells, followed by flow cytometry-based cell sorting and puromycin selection. Finally, BDD-hF8 targeted to knock-in the mROSA26 genomic locus was identified and validated for FVIII expression. RESULTS: We identified the p18T-BDD-F8-V3 variant with high FVIII activity and detected the strongest pX458-mROSA26-int1-sgRNA1 targeted cleavage ability and no cleavage events were found at potential off-target sites. Targeted knock-in of BDD-hF8 cDNA at the mROSA26 locus was achieved based on both HDR/NHEJ gene repair approaches, and high level and stable FVIII expression was obtained, successfully realizing gene editing in vitro. CONCLUSIONS: Knock-in of exogenous genes based on the CRISPR/Cas9 system targeting genomic loci is promising for the research and treatment of a variety of single-gene diseases.


Subject(s)
CRISPR-Cas Systems , Factor VIII , Hemophilia A , Animals , Mice , CRISPR-Associated Protein 9/genetics , Gene Editing/methods , Hemophilia A/genetics , Hemophilia A/therapy , RNA, Guide, CRISPR-Cas Systems , Factor VIII/biosynthesis , Factor VIII/genetics
7.
Cell Stress Chaperones ; 28(6): 675-688, 2023 11.
Article in English | MEDLINE | ID: mdl-37755621

ABSTRACT

Myocardial microvessels are composed of a monolayer of endothelial cells, which play a crucial role in maintaining vascular barrier function, luminal latency, vascular tone, and myocardial perfusion. Endothelial dysfunction is a key factor in the development of cardiac microvascular injury and diabetic cardiomyopathy. In addition to their role in glucose oxidation and energy metabolism, mitochondria also participate in non-metabolic processes such as apoptosis, intracellular ion handling, and redox balancing. Mitochondrial dynamics and mitophagy are responsible for regulating the quality and quantity of mitochondria in response to hyperglycemia. However, these endogenous homeostatic mechanisms can both preserve and/or disrupt non-metabolic mitochondrial functions during diabetic endothelial damage and cardiac microvascular injury. This review provides an overview of the molecular features and regulatory mechanisms of mitochondrial dynamics and mitophagy. Furthermore, we summarize findings from various investigations that suggest abnormal mitochondrial dynamics and defective mitophagy contribute to the development of diabetic endothelial dysfunction and myocardial microvascular injury. Finally, we discuss different therapeutic strategies aimed at improving endothelial homeostasis and cardiac microvascular function through the enhancement of mitochondrial dynamics and mitophagy.


Subject(s)
Diabetes Mellitus , Mitophagy , Humans , Mitophagy/physiology , Endothelial Cells , Mitochondrial Dynamics , Myocardium
8.
Medicine (Baltimore) ; 102(32): e34540, 2023 Aug 11.
Article in English | MEDLINE | ID: mdl-37565908

ABSTRACT

Hyperuricemia nephropathy, also known as gouty nephropathy, refers to renal damage induced by hyperuricemia caused by excessive production of serum uric acid or low excretion of uric acid. the persistence of symptoms will lead to changes in renal tubular phenotype and accelerate the progress of renal fibrosis. The existence and progressive aggravation of symptoms will bring a heavy burden to patients, their families and society, affect their quality of life and reduce their well-being. With the increase of reports on hyperuricemia nephropathy, the importance of related signal pathways in the pathogenesis of hyperuricemia nephropathy is becoming more and more obvious, but most studies are limited to the upper and lower mediating relationship between 1 or 2 signal pathways. The research on the comprehensiveness of signal pathways and the breadth of crosstalk between signal pathways is limited. By synthesizing the research results of signal pathways related to hyperuricemia nephropathy in recent years, this paper will explore the specific mechanism of hyperuricemia nephropathy, and provide new ideas and methods for the treatment of hyperuricemia nephropathy based on a variety of signal pathway crosstalk and personal prospects.


Subject(s)
Hyperuricemia , Kidney Calculi , Humans , Uric Acid , Hyperuricemia/complications , Quality of Life , Signal Transduction , Kidney Calculi/complications
9.
Cell Stress Chaperones ; 28(3): 239-251, 2023 05.
Article in English | MEDLINE | ID: mdl-37093549

ABSTRACT

Myocardial ischemia reduces the supply of oxygen and nutrients to cardiomyocytes, leading to an energetic crisis or cell death. Mitochondrial dysfunction is a decisive contributor to the reception, transmission, and modification of cardiac ischemic signals. Cells with damaged mitochondria exhibit impaired mitochondrial metabolism and increased vulnerability to death stimuli due to disrupted mitochondrial respiration, reactive oxygen species overproduction, mitochondrial calcium overload, and mitochondrial genomic damage. Various intracellular and extracellular stress signaling pathways converge on mitochondria, so dysfunctional mitochondria tend to convert from energetic hubs to apoptotic centers. To interrupt the stress signal transduction resulting from lethal mitochondrial damage, cells can activate mitophagy (mitochondria-specific autophagy), which selectively eliminates dysfunctional mitochondria to preserve mitochondrial quality control. Different pharmacological and non-pharmacological strategies have been designed to augment the protective properties of mitophagy and have been validated in basic animal experiments and pre-clinical human trials. In this review, we describe the process of mitophagy in cardiomyocytes under ischemic stress, along with its regulatory mechanisms and downstream effects. Then, we discuss promising therapeutic approaches to preserve mitochondrial homeostasis and protect the myocardium against ischemic damage by inducing mitophagy.


Subject(s)
Mitochondria , Mitophagy , Animals , Humans , Mitophagy/genetics , Mitochondria/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Signal Transduction
10.
Clin Chim Acta ; 540: 117227, 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36640930

ABSTRACT

BACKGROUND: Early stratification of disease progression remains one of the major challenges towards the post-coronavirus disease 2019 (COVID-19) era. The clinical relevance of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) nucleic acid load is debated due to the heterogeneity in patients' underlying health conditions. We determined the prognostic value of nasopharyngeal viral load dynamic conversion for COVID-19. METHODS: The cycling threshold (Ct) values of 28,937 nasopharyngeal SARS-CoV-2 RT-PCRs were retrospectively collected from 3,364 COVID-19 patients during hospitalization and coordinated to the onset of disease progression. The ROC curve was utilized to determine the predictive performance of the rate of Ct value alteration between two consecutive RT-PCR runs within 48 h (ΔCt%) for disease transformation across patients with different COVID-19 severity and immune backgrounds, and further validated with 1,860 SARS-CoV-2 RT-PCR results from an independent validation cohort of 262 patients. For the 67 patients with severe COVID-19, Kaplan-Meier analysis was performed to evaluate the difference in survival between patients stratified by the magnitude of Ct value alteration between the late and early stages of hospitalization. RESULTS: The kinetics of viral nucleic acid conversion diversified across COVID-19 patients with different clinical characteristics and disease severities. The ΔCt% is a clinical characteristic- and host immune status-independent indicator for COVID-19 progression prediction (AUC = 0.79, 95 % CI = 0.76 to 0.81), which outperformed the canonical blood test markers, including c-reactive protein (AUC = 0.57, 95 % CI = 0.53 to 0.61), serum amyloid A (AUC = 0.61, 95 % CI = 0.54 to 0.68), lactate dehydrogenase (AUC = 0.61, 95 % CI = 0.56 to 0.67), d-dimer (AUC = 0.56, 95 % CI = 0.46 to 0.66), and lymphocyte count (AUC = 0.62, 95 % CI = 0.58 to 0.66). Patients with persistent high SARS-CoV-2 viral load (an increase of mean Ct value < 50 %) during the first 3 days of hospitalization demonstrated a significantly unfavorable survival (HR = 0.16, 95 % CI = 0.04 to 0.65, P = 2.41 × 10-3). CONCLUSIONS: Viral nucleic acid dynamics of SARS-CoV-2 eliminates the inter-patient variance of basic health conditions and therefore, can serve as a prognostic marker for COVID-19.


Subject(s)
COVID-19 , Humans , COVID-19/diagnosis , SARS-CoV-2 , Retrospective Studies , Prognosis , Time Factors , Viral Load , Disease Progression
11.
Cancer Cell ; 41(1): 181-195.e9, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36584672

ABSTRACT

Integrated molecular analysis of human cancer has yielded molecular classification for precise management of cancer patients. Here, we analyzed the whole genomic, epigenomic, transcriptomic, and proteomic data of 155 esophageal squamous cell carcinomas (ESCCs). Multi-omics analysis led to the classification of ESCCs into four subtypes: cell cycle pathway activation, NRF2 oncogenic activation, immune suppression (IS), and immune modulation (IM). IS and IM cases were highly immune infiltrated but differed in the type and distribution of immune cells. IM cases showed better response to immune checkpoint blockade therapy than other subtypes in a clinical trial. We further developed a classifier with 28 features to identify the IM subtype, which predicted anti-PD-1 therapy response with 85.7% sensitivity and 90% specificity. These results emphasize the clinical value of unbiased molecular classification based on multi-omics data and have the potential to further improve the understanding and treatment of ESCC.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Carcinoma, Squamous Cell/genetics , Multiomics , Proteomics
12.
J Clin Lab Anal ; 36(10): e24689, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36098043

ABSTRACT

BACKGROUND: Vitreoretinal lymphoma (VRL) can commonly masquerade as chronic idiopathic uveitis due to its nonspecific clinical presentation. Thus, its early diagnosis is difficult. In this study, new logistic regression models were used to classify VRL and uveitis. Additionally, the diagnostic performance of interleukin (IL)-10, the IL-10/IL-6, and the Interleukin Score for IntraOcular Lymphoma Diagnosis (ISOLD) are evaluated. METHODS: Sixty-nine aqueous humors (AH) (46 VRL, 23 uveitis) and 65 vitreous humors (VH) (49 VRL, 16 uveitis) were collected from a single-center retrospective cohort. Logistic regression models were conducted based on IL-6 and IL-10. The cut-off values, area under the receiver operating characteristic curve (ROC) curve (AUC), sensitivity and specificity of IL-10, the IL-10/IL-6, the ISOLD, and the models were calculated from the ROC. Furthermore, Spearman's rank correlation analysis was performed to determine cytokine levels in VH and AH. RESULTS: We redefined the cut-off values of IL-10, the IL-10/IL-6, the ISOLD, and the logistic regression models. In AH, the AUC values of IL-10, ISOLD, IL10/IL6, and the model were 0.91, 0.953, 0.952, and 0.967. In VH, they were 0.93, 0.95, 0.954, and 0.954, respectively. IL-6 (r = 0.7844) and IL-10 (r = 0.8506) in AH and VH showed a strong correlation. CONCLUSIONS: IL-6 and IL-10 levels were introduced into new logistic regression models. The diagnostic efficacy of the models improved compared to the indicators mentioned above among Chinese patients. Additionally, the models could predict the probability of VRL more accurately. A strong correlation of cytokine levels showed the great potential of AH as prioritized auxiliary diagnostic for VRL.


Subject(s)
Eye Neoplasms , Intraocular Lymphoma , Lymphoma, Non-Hodgkin , Retinal Neoplasms , Uveitis , Cytokines , Eye Neoplasms/diagnosis , Eye Neoplasms/pathology , Humans , Interleukin-10 , Interleukin-6 , Interleukins , Intraocular Lymphoma/diagnosis , Intraocular Lymphoma/pathology , Logistic Models , Retinal Neoplasms/diagnosis , Retinal Neoplasms/pathology , Retrospective Studies , Uveitis/diagnosis , Uveitis/pathology , Vitreous Body
13.
Small ; 18(37): e2203531, 2022 09.
Article in English | MEDLINE | ID: mdl-35962758

ABSTRACT

Activatable fluorescence imaging in the second near-infrared window (NIR-II FL, 1000-1700 nm) is of great significance for accurate tumor diagnosis and targeting therapy. However, the clinical translation of most stimulus-activated nanoprobes is severely restricted by insufficient tumor response and out-of-synchronization theranostic process. Herein, an intelligent nanofactory AUC-GOx/Cel that possesses the "external supply, internal promotion" dual H2 O2 -amplification strategy for homologous activated tumor theranostic is designed. This nanofactory is constructed via a two-step biomineralization method using Au-doped Ag2 S as a carrier for glucose oxidase (GOx) and celastrol, followed by the growing of CuS to "turn off" the NIR-II FL signal. In the overexpressed H2 O2 tumor-microenvironment, the CuS featuring a responsive-degradability behavior can effectively release Cu ions, resulting in the "ON" state of NIR-II FL and Fenton-like activity. The exposed GOx can realize the intratumoral H2 O2 supply (external supply) via the effective conversion of glucose, and mediating tumor-starvation therapy; the interaction of celastrol and mitochondria can offer a substantial increase in the endogenous H2 O2 level (internal promotion), thereby significantly promoting the chemodynamic therapy (CDT) efficacy. Meanwhile, the dual H2 O2 -enhancement performance will in turn accelerate the degradation of AUC-GOx/Cel, and achieve a positive feedback mechanism for self-reinforcing CDT.


Subject(s)
Nanoparticles , Neoplasms , Cell Line, Tumor , Copper , Glucose Oxidase/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/therapy , Pentacyclic Triterpenes/therapeutic use , Theranostic Nanomedicine/methods , Tumor Microenvironment
14.
Stem Cell Res Ther ; 13(1): 353, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35883203

ABSTRACT

BACKGROUND: Hemophilia B is a rare inherited genetic bleeding disorder caused by a deficiency or lack of coagulation factor IX, the gene for which (F9) is located on the X chromosome. Hemophilia B is currently incurable and the standard treatment is coagulation factor replacement therapy. Although gene therapy has the potential to cure hemophilia, significant barriers are still needed to be overcome, e.g., off-target effects and immunoreactivity, so new approaches must be explored. Nonsense mutations account for 8% of all the hemophilia B mutation types and can result in the development of coagulation factor inhibitors. In this study, CRISPR/Cas9 technology was used to construct a mouse embryonic stem cell model with a hemophilia B nonsense mutation (F9 c.223C > T) in humans to investigate the pathogenesis and treatment of nonsense mutations in hemophilia B. METHODS: First, a donor plasmid with a mutation (F9 c.223 C > T) and sgRNAs were constructed. Second, both the donor plasmid and the px330-sgRNA were electroporated into mouse embryonic stem cell, and the mutant cells were then screened using puromycin and red fluorescence. Third, the mutant cell lines were tested for pluripotency and the ability to differentiate into three layers. Finally, the effect of mutation on gene function was studied in the differentiation system. RESULTS: The mutant vector and effective sgRNA were constructed, and the mutant cell line was screened. This mutant cell line exhibited pluripotency and the ability to differentiate into three layers. This point mutation affects F9 expression at both the RNA and protein levels in the differentiation system. CONCLUSIONS: The mutant cell line obtained in the current study had a single-base mutation rather than a base deletion or insertion in the exon, which is more similar to clinical cases. In addition, the mutant has the characteristics of mouse embryonic stem cells, and this point mutation affects F9 gene transcription and translation, which can be used as a disease model for studying the pathogenesis and treatment of hemophilia at the stem cell level.


Subject(s)
Hemophilia A , Hemophilia B , Animals , CRISPR-Cas Systems/genetics , Codon, Nonsense/genetics , Factor IX/genetics , Factor IX/metabolism , Hemophilia A/genetics , Hemophilia B/genetics , Hemophilia B/therapy , Humans , Mice , Mouse Embryonic Stem Cells/metabolism , Mutation , Technology
15.
Front Comput Neurosci ; 16: 885091, 2022.
Article in English | MEDLINE | ID: mdl-35651590

ABSTRACT

To construct a prognostic model for preoperative prediction on computed tomography (CT) images of esophageal squamous cell carcinoma (ESCC), we created radiomics signature with high throughput radiomics features extracted from CT images of 272 patients (204 in training and 68 in validation cohort). Multivariable logistic regression was applied to build the radiomics signature and the predictive nomogram model, which was composed of radiomics signature, traditional TNM stage, and clinical features. A total of 21 radiomics features were selected from 954 to build a radiomics signature which was significantly associated with progression-free survival (p < 0.001). The area under the curve of performance was 0.878 (95% CI: 0.831-0.924) for the training cohort and 0.857 (95% CI: 0.767-0.947) for the validation cohort. The radscore of signatures' combination showed significant discrimination for survival status. Radiomics nomogram combined radscore with TNM staging and showed considerable improvement over TNM staging alone in the training cohort (C-index, 0.770 vs. 0.603; p < 0.05), and it is the same with clinical data (C-index, 0.792 vs. 0.680; p < 0.05), which were confirmed in the validation cohort. Decision curve analysis showed that the model would receive a benefit when the threshold probability was between 0 and 0.9. Collectively, multiparametric CT-based radiomics nomograms provided improved prognostic ability in ESCC.

16.
J Transl Med ; 20(1): 285, 2022 06 25.
Article in English | MEDLINE | ID: mdl-35752862

ABSTRACT

BACKGROUND: Aberrant glycosylation has been recognized as a hallmark of cancer and N-glycosylation is one of the main types of glycosylation in eukaryotes. Although N-glycoproteomics has made contributions to the discovery of biomarkers in a variety of cancers, less is known about the abnormal glycosylation signatures in esophageal squamous cell carcinoma (ESCC). METHODS: In this study, we reported the proteomics and N-glycoproteomic site-mapping analysis of eight pairs of ESCC tissues and adjacent normal tissues. With zic-HILIC enrichment, TMT-based isobaric labeling, LC-MS/MS analysis, differentially expressed N-glycosylation was quantitatively characterized. Lectin affinity enrichment combined with western blot was used to validate the potential biomarkers in ESCC. RESULTS: A series of differentially expressed glycoproteins (e.g., LAMP2, PLOD2) and enriched signaling pathways (e.g., metabolism-related pathway, ECM-receptor interaction, focal adhesion) were identified. Besides that, seven significantly enriched motifs were found from the identified N-glycosylation sites. Three clusters were identified after conducting the dynamic profiling analysis of glycoprotein change during lymph node metastasis progression. Further validation found that the elevated fucosylation level of ITGB1, CD276 contributed to the occurrence and development of ESCC, which might be the potential biomarkers in ESCC. CONCLUSION: In summary, we characterized the N-glycosylation and N-glycoprotein alterations associated with ESCC. The typical changes in glycoprotein expression and glycosylation occupancy identified in our study will not only be used as ESCC biomarkers but also improve the understanding of ESCC biology.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , B7 Antigens , Biomarkers , Biomarkers, Tumor/metabolism , Chromatography, Liquid , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Glycoproteins/metabolism , Humans , Tandem Mass Spectrometry
17.
Oncol Lett ; 24(2): 253, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35765282

ABSTRACT

Gastric cancer (GC) comprises the 3rd cause of cancer-related death worldwide. Increased expression of neural precursor cell expressed, developmentally downregulated 9 (NEDD9) is commonly observed in GC, however, its underlying molecular mechanism in GC remains unknown. The potential interaction between miR-4735-3p and NEDD9 was predicted by TargetScan 7.1. Expression profiles of miR-4735-3p and NEDD9 were examined between GC tissues and normal tissues by reverse transcription-quantitative (RT-q) PCR. The relationship between miR-4735-3p and NEDD9 was validated by RT-qPCR, western blotting, dual luciferase reporter assay and RNA immunoprecipitation assay. Biological relationship between miR-4735-3p and NEDD9 was evidenced by the cell invasion and cell migration assays. NEDD9 level was negatively associated with miR-4735-3p level in GC tissues. miR-4735-3p suppressed NEDD9 levels in GC cells. NEDD9 was revealed to be a target gene of miR-4735-3p. miR-4735-3p overexpression suppressed cell migration and invasion of GC cells, which were antagonized by overexpression of NEDD9. Moreover, miR-4735-3p mimic decreased the levels of matrix metalloproteinases 2/9, increased the level of E-cadherin, which were reversed by overexpression of NEDD9. Collectively, the present study provided a potential mechanism for the tumor suppressor role of miR-4735-3p in GC by targeting NEDD9.

18.
Article in English | MEDLINE | ID: mdl-35571730

ABSTRACT

Objective: Fetal growth restriction (FGR) affects 5% to 10% of newborns and is a major determinant of perinatal morbidity and mortality. Myocardial performance index (MPI), also known as the Tei index, is a useful, noninvasive, and Doppler-derived myocardial performance tool for fetal cardiac function evaluation. The purpose of the study is to evaluate ultrasonic prediction on FGR and postnatal outcomes using MPI and blood flow spectrum. Methods: This retrospective study included 240 pregnant women developing FGR and 240 healthy pregnant women. The blood flow spectrum of middle cerebral artery (MCA), umbilical artery (UA), and ductus venous including systolic to diastolic ratio (S/D), resistant index (RI), pulse index (PI), and peak ventricular systolic velocity/atrial contraction valley velocity (S/a) were examined using the GE Voluson E8 ultrasound system. Results: The MPI, S/D, RI, PI of UA, and S/a were all higher but S/D, RI, and PI of MCA were lower in the FGR group than those in the control group (P < 0.001). The MPI, S/D, RI, PI of UA, S/D, RI, PI of MCA, and ductus venous S/a yielded AUC of 0.813, 0.835, 0.791, 0.804, 0.789, 0.796, 0.803, and 0.784 when they were used to predict the incidence of FGR. Of note, the pregnant women with poor pregnancy outcomes exhibited higher values of MPI, S/D, RI, PI of UA, and S/a with lower scores of 1 min Apgar concomitant with lower values regarding S/D, RI, and PI of MCA than those with favorable pregnancy outcomes (P < 0.001). The MPI (r = -0.623), S/D (r = -0.660), RI (r = -0.601), PI (r = -630) of UA, and S/a (r = -0.573) shared negative correlations with 1 min Apgar scores (P < 0.001). Of note, the S/D (r = 0.562), RI (r = 0.597), and PI (r = 0.619) of MCA were positively correlated with 1 min Apgar scores (P < 0.001). It was revealed that the MPI, S/D, RI, PI of UA, S/D, RI, PI of MCA, and ductus venous S/a yielded AUC of 0.806, 0.833, 0.774, 0.788, 0.807, 0.729, 0.748, and 0.770 when they were used to predict the incidence of poor pregnancy outcomes for pregnant women developing FGR. Conclusion: Our study demonstrates good ultrasonic prediction on FGR and postnatal outcomes using MPI and blood flow spectrum.

19.
Stem Cell Res ; 60: 102682, 2022 04.
Article in English | MEDLINE | ID: mdl-35123343

ABSTRACT

The lack of coagulation factor VIII in patient with nonsense mutation hemophilia A leads to varying degrees of bleeding symptoms, and long-term use of alternative therapies can produce inhibitors that affect the efficacy. In this study, human induced pluripotent stem cells (iPSCs) of hemophilia A were generated by reprogramming of urine cells. Human urine cells (HUCs) were isolated by collecting patients' mid-stream urine, and cultured to good state in urine medium. Then, the HUCs were transfected with PEP4-EO2S-ET2K and pCEP4-M2L, and iPSCs were obtained in the medium without trophoblast cells and the composition was determined. Finally, alkaline phosphatase staining, karyotype analysis, immunofluorescence staining and teratoma were used to verify that we successfully reprogrammed hemophilia A-specific human induced pluripotent stem cells from patients' urine cells, providing a safe and effective cell model for the study of molecular mechanism and related treatment of hemophilia.


Subject(s)
Hemophilia A , Induced Pluripotent Stem Cells , Cell Differentiation , Factor VIII/genetics , Hemophilia A/genetics , Hemophilia A/therapy , Humans , Mutation/genetics
20.
Stem Cell Res ; 60: 102684, 2022 04.
Article in English | MEDLINE | ID: mdl-35121195

ABSTRACT

Hemophilia B (HB) is an X chromosome-linked recessive disorder caused by a quantitative or qualitative defect of coagulation zymogen factor IX. In this study, urine cells were collected from a patient with HB who carries variant F9 c.223C > T (p.R75X), and reprogrammed into induced pluripotent stem cells (iPSCs) using the reprogramming factors, OCT4, SOX2, m-MYC, and KLF4. The HB-iPSC line (SXMUi001-A) has characteristics similar to human embryonic stem cell, namely, pluripotency and the potential to differentiate into three germ layers. This cell line can be used as a disease model for exploring the molecular mechanism and readthrough treatment of HB.


Subject(s)
Hemophilia B , Induced Pluripotent Stem Cells , Cell Differentiation , Cell Line , Cellular Reprogramming , Factor IX/genetics , Factor IX/metabolism , Germ Layers , Hemophilia B/genetics , Humans , Induced Pluripotent Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...