1.Genomic variant surveillance of SARS-CoV-2 positive specimens using a direct PCR product sequencing surveillance (DPPSS) method
Nicole Ann L. Tuberon ; Francisco M. Heralde III ; Catherine C. Reportoso ; Arturo L. Gaitano III ; Wilmar Jun O. Elopre ; Kim Claudette J. Fernandez
Acta Medica Philippina 2025;59(Early Access 2025):1-12
BACKGROUND AND OBJECTIVE
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as the causative agent of COVID-19 has significantly challenged the public health landscape in late 2019. After almost 3 years of the first ever SARS-CoV-2 case, the World Health Organization (WHO) declared the end of this global health emergency in May 2023. Although, despite the subsequent drop of COVID-19 cases, the SARS-CoV-2 infection still exhibited multiple waves of infection, primarily attributed to the appearance of new variants. Five of these variants have been classified as Variants of Concern (VOC): Alpha, Beta, Gamma, Delta, and the most recent, Omicron. Therefore, the development of methods for the timely and accurate detection of viral variants remains fundamental, ensuring an ongoing and effective response to the disease. This study aims to evaluate the feasibility of the application of an in-house approach in genomic surveillance for the detection of SARS-CoV-2 variants using in silico designed primers.
METHODSThe primers used for the study were particularly designed based on conserved regions of certain genes in the virus, targeting distinct mutations found in known variants of SARS-CoV-2. Viral RNA extracts from nasopharyngeal samples (n=14) were subjected to quantitative and qualitative tests (Nanodrop and AGE). Selected samples were then analyzed by RT-PCR and amplicons were submitted for sequencing. Sequence alignment analysis was carried out to identify the prevailing COVID-19 variant present in the sample population.
RESULTSThe study findings demonstrated that the in-house method was able to successfully amplify conserved sequences (spike, envelope, membrane, ORF1ab) and enabled identification of the circulating SARS-CoV-2 variant among the samples. Majority of the samples were identified as Omicron variant. Three out of four designed primers effectively bound into the conserved sequence of target genes present in the sample, revealing the specific SARSCoV-2 variant. The detected mutations characterized for Omicron found in the identified lineages included K417N, S477N, and P681H which were also identified as mutations of interest. Furthermore, identification of the B.1.448 lineage which was not classified in any known variant also provided the potential of the developed in-house method in detecting unknown variants of COVID-19.
CONCLUSIONAmong the five VOCs, Omicron is the most prevalent and dominant variant. The in-house direct PCR product sequencing surveillance (DPPSS) method provided an alternative platform for SAR-CoV-2 variant analysis which is accessible and affordable than the conventional diagnostic surveillance methods and the whole genome sequencing. Further evaluation and improvements on the oligonucleotide primers may offer significant contribution to the development of a specific and direct PCRbased detection of new emerging COVID-19 variants.
Sars-cov-2 ; Polymerase Chain Reaction ; Dna Primers ; Oligonucleotide Primers
2.Effect of electroacupuncture at "Hegu" (LI4) and "Taichong" (LR3) on DNA methylation of the SLC6A4 gene promoter in the hippocampus of depressed rats.
Xi ZHANG ; Shengyong SU ; Xin LI ; Tian WANG
Chinese Acupuncture & Moxibustion 2025;45(11):1609-1616
OBJECTIVE:
To observe the effect of electroacupuncture (EA) at "Hegu" (LI4) and "Taichong" (LR3) on DNA methylation of the solute carrier family 6 member 4 (SLC6A4) gene promoter region in the hippocampus of depressed rats, and to explore the potential antidepressant mechanism of EA.
METHODS:
Thirty male Sprague-Dawley rats were randomly divided into a blank group, a model group, a medication group, a 5-Azacytidine (5-AZA) group, and an EA group, 6 rats in each group. Depression models were established in the model group, the medication group, the 5-AZA group, and the EA group using chronic unpredictable mild stress (CUMS) combined with solitary housing. The medication group was treated with intragastric administration of fluoxetine hydrochloride capsules; the 5-AZA group was treated with intraperitoneal injection of 5-AZA; the EA group was treated with EA at bilateral "Hegu" (LI4) and "Taichong" (LR3), with disperse-dense wave, frequency of 2 Hz/100 Hz, and intensity of 1-1.2 mA, 20 min each session. All the treatment was given in three groups once daily for 21 consecutive days. Behavioral changes were evaluated by sucrose preference test, open field test, and novelty-suppressed feeding test. Serum levels of serotonin (5-HT), dopamine (DA), and norepinephrine (NE) were measured by ELISA. The expression of SLC6A4 and 5-HT1AR protein and mRNA in hippocampus was detected by Western blot and real-time quantitative PCR, respectively. DNA methylation status of the SLC6A4 promoter region in hippocampal tissue was analyzed by bisulfite sequencing PCR (BSP).
RESULTS:
Compared with the blank group, the model group showed decreased sucrose preference, reduced total locomotor distance, and prolonged latency to feeding (P<0.05), decreased serum 5-HT, DA, and NE levels (P<0.05), downregulated hippocampal SLC6A4 and 5-HT1AR protein and mRNA expression (P<0.05), and increased CpG site methylation rate of the SLC6A4 promoter region (P<0.05). Compared with the model group, the medication group, the 5-AZA group, and the EA group exhibited increased sucrose preference, increased total locomotor distance, shortened latency to feeding (P<0.05), elevated serum 5-HT, DA, and NE levels (P<0.05), upregulated hippocampal SLC6A4 and 5-HT1AR protein and mRNA expression (P<0.05), and reduced CpG site methylation rate of the SLC6A4 promoter (P<0.05). Compared with the medication group and the 5-AZA group, the EA group showed higher sucrose preference, greater total locomotor distance, shorter latency to feeding (P<0.05), and increased serum DA and NE levels (P<0.05).
CONCLUSION
EA could improve depressive behaviors in depressed rat models. The underlying mechanism may involve inhibition of SLC6A4 hypermethylation in the hippocampus on the serotonergic system, upregulation of SLC6A4 and 5-HT1AR protein and mRNA expression, and elevation of monoamine neurotransmitters such as 5-HT.
Animals
;
Electroacupuncture
;
Male
;
Hippocampus/metabolism*
;
Rats, Sprague-Dawley
;
Rats
;
Acupuncture Points
;
DNA Methylation
;
Depression/metabolism*
;
Promoter Regions, Genetic
;
Serotonin Plasma Membrane Transport Proteins/metabolism*
;
Humans
3.Driving effect of P16 methylation on telomerase reverse transcriptase-mediated immortalization and transformation of normal human fibroblasts.
Xuehong ZHANG ; Paiyun LI ; Ying GAN ; Shengyan XIANG ; Liankun GU ; Jing ZHOU ; Xiaorui ZHOU ; Peihuang WU ; Baozhen ZHANG ; Dajun DENG
Chinese Medical Journal 2025;138(3):332-342
BACKGROUND:
P16 inactivation is frequently accompanied by telomerase reverse transcriptase ( TERT ) amplification in human cancer genomes. P16 inactivation by DNA methylation often occurs automatically during immortalization of normal cells by TERT . However, direct evidence remains to be obtained to support the causal effect of epigenetic changes, such as P16 methylation, on cancer development. This study aimed to provide experimental evidence that P16 methylation directly drives cancer development.
METHODS:
A zinc finger protein-based P16 -specific DNA methyltransferase (P16-Dnmt) vector containing a "Tet-On" switch was used to induce extensive methylation of P16 CpG islands in normal human fibroblast CCD-18Co cells. Battery assays were used to evaluate cell immortalization and transformation throughout their lifespan. Cell subcloning and DNA barcoding were used to track the diversity of cell evolution.
RESULTS:
Leaking P16-Dnmt expression (without doxycycline-induction) could specifically inactivate P16 expression by DNA methylation. P16 methylation only promoted proliferation and prolonged lifespan but did not induce immortalization of CCD-18Co cells. Notably, cell immortalization, loss of contact inhibition, and anchorage-independent growth were always prevalent in P16-Dnmt&TERT cells, indicating cell transformation. In contrast, almost all TERT cells died in the replicative crisis. Only a few TERT cells recovered from the crisis, in which spontaneous P16 inactivation by DNA methylation occurred. Furthermore, the subclone formation capacity of P16-Dnmt&TERT cells was two-fold that of TERT cells. DNA barcoding analysis showed that the diversity of the P16-Dnmt&TERT cell population was much greater than that of the TERT cell population.
CONCLUSION
P16 methylation drives TERT -mediated immortalization and transformation of normal human cells that may contribute to cancer development.
Humans
;
Telomerase/genetics*
;
DNA Methylation/physiology*
;
Fibroblasts/cytology*
;
Cyclin-Dependent Kinase Inhibitor p16/metabolism*
;
Cell Line
;
Cell Transformation, Neoplastic/genetics*
4.Circulating tumor DNA- and cancer tissue-based next-generation sequencing reveals comparable consistency in targeted gene mutations for advanced or metastatic non-small cell lung cancer.
Weijia HUANG ; Kai XU ; Zhenkun LIU ; Yifeng WANG ; Zijia CHEN ; Yanyun GAO ; Renwang PENG ; Qinghua ZHOU
Chinese Medical Journal 2025;138(7):851-858
BACKGROUND:
Molecular subtyping is an essential complementarity after pathological analyses for targeted therapy. This study aimed to investigate the consistency of next-generation sequencing (NGS) results between circulating tumor DNA (ctDNA)-based and tissue-based in non-small cell lung cancer (NSCLC) and identify the patient characteristics that favor ctDNA testing.
METHODS:
Patients who diagnosed with NSCLC and received both ctDNA- and cancer tissue-based NGS before surgery or systemic treatment in Lung Cancer Center, Sichuan University West China Hospital between December 2017 and August 2022 were enrolled. A 425-cancer panel with a HiSeq 4000 NGS platform was used for NGS. The unweighted Cohen's kappa coefficient was employed to discriminate the high-concordance group from the low-concordance group with a cutoff value of 0.6. Six machine learning models were used to identify patient characteristics that relate to high concordance between ctDNA-based and tissue-based NGS.
RESULTS:
A total of 85 patients were enrolled, of which 22.4% (19/85) had stage III disease and 56.5% (48/85) had stage IV disease. Forty-four patients (51.8%) showed consistent gene mutation types between ctDNA-based and tissue-based NGS, while one patient (1.2%) tested negative in both approaches. Patients with advanced diseases and metastases to other organs would be suitable for the ctDNA-based NGS, and the generalized linear model showed that T stage, M stage, and tumor mutation burden were the critical discriminators to predict the consistency of results between ctDNA-based and tissue-based NGS.
CONCLUSION
ctDNA-based NGS showed comparable detection performance in the targeted gene mutations compared with tissue-based NGS, and it could be considered in advanced or metastatic NSCLC.
Humans
;
Carcinoma, Non-Small-Cell Lung/pathology*
;
Circulating Tumor DNA/blood*
;
High-Throughput Nucleotide Sequencing/methods*
;
Female
;
Male
;
Lung Neoplasms/pathology*
;
Middle Aged
;
Mutation/genetics*
;
Aged
;
Adult
;
Aged, 80 and over
5.C/EBPβ-Lin28a positive feedback loop triggered by C/EBPβ hypomethylation enhances the proliferation and migration of vascular smooth muscle cells in restenosis.
Xiaojun ZHOU ; Shan JIANG ; Siyi GUO ; Shuai YAO ; Qiqi SHENG ; Qian ZHANG ; Jianjun DONG ; Lin LIAO
Chinese Medical Journal 2025;138(4):419-429
BACKGROUND:
The main cause of restenosis after percutaneous transluminal angioplasty (PTA) is the excessive proliferation and migration of vascular smooth muscle cells (VSMCs). Lin28a has been reported to play critical regulatory roles in this process. However, whether CCAAT/enhancer-binding proteins β (C/EBPβ) binds to the Lin28a promoter and drives the progression of restenosis has not been clarified. Therefore, in the present study, we aim to clarify the role of C/EBPβ-Lin28a axis in restenosis.
METHODS:
Restenosis and atherosclerosis rat models of type 2 diabetes ( n = 20, for each group) were established by subjecting to PTA. Subsequently, the difference in DNA methylation status and expression of C/EBPβ between the two groups were assessed. EdU, Transwell, and rescue assays were performed to assess the effect of C/EBPβ on the proliferation and migration of VSMCs. DNA methylation status was further assessed using Methyltarget sequencing. The interaction between Lin28a and ten-eleven translocation 1 (TET1) was analysed using co-immunoprecipitation (Co-IP) assay. Student's t -test and one-way analysis of variance were used for statistical analysis.
RESULTS:
C/EBPβ expression was upregulated and accompanied by hypomethylation of its promoter in restenosis when compared with atherosclerosis. In vitroC/EBPβ overexpression facilitated the proliferation and migration of VSMCs and was associated with increased Lin28a expression. Conversely, C/EBPβ knockdown resulted in the opposite effects. Chromatin immunoprecipitation assays further demonstrated that C/EBPβ could directly bind to Lin28a promoter. Increased C/EBPβ expression and enhanced proliferation and migration of VSMCs were observed after decitabine treatment. Further, mechanical stretch promoted C/EBPβ and Lin28a expression accompanied by C/EBPβ hypomethylation. Additionally, Lin28a overexpression reduced C/EBPβ methylation via recruiting TET1 and enhanced C/EBPβ-mediated proliferation and migration of VSMCs. The opposite was noted in Lin28a knockdown cells.
CONCLUSION
Our findings suggest that the C/EBPβ-Lin28a axis is a driver of restenosis progression, and presents a promising therapeutic target for restenosis.
Animals
;
Cell Proliferation/genetics*
;
Cell Movement/genetics*
;
Muscle, Smooth, Vascular/metabolism*
;
Rats
;
DNA Methylation/physiology*
;
CCAAT-Enhancer-Binding Protein-beta/genetics*
;
Male
;
Myocytes, Smooth Muscle/cytology*
;
Rats, Sprague-Dawley
;
RNA-Binding Proteins/genetics*
;
Cells, Cultured
;
Coronary Restenosis/metabolism*
6.Poly (ADP-ribose) polymerase inhibitors in cancer therapy.
Chinese Medical Journal 2025;138(6):634-650
Poly(ADP-ribose) polymerase (PARP) inhibitors (PARPis) have emerged as critical agents for cancer therapy. By inhibiting the catalytic activity of PARP enzymes and trapping them in the DNA, PARPis disrupt DNA repair, ultimately leading to cell death, particularly in cancer cells with homologous recombination repair deficiencies, such as those harboring BRCA mutations. This review delves into the mechanisms of action of PARPis in anticancer treatments, including the inhibition of DNA repair, synthetic lethality, and replication stress. Furthermore, the clinical applications of PARPis in various cancers and their adverse effects as well as their combinations with other therapies and the mechanisms underlying resistance are summarized. This review provides comprehensive insights into the role and mechanisms of PARP and PARPis in DNA repair, with a particular focus on the potential of PARPi-based therapies in precision medicine for cancer treatment.
Humans
;
Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use*
;
Neoplasms/genetics*
;
DNA Repair/drug effects*
;
Animals
;
Antineoplastic Agents/therapeutic use*
7.Mitochondrial RNA metabolism, a potential therapeutic target for mitochondria-related diseases.
Tongyue DUAN ; Liya SUN ; Kaiyue DING ; Qing ZHAO ; Lujun XU ; Chongbin LIU ; Lin SUN
Chinese Medical Journal 2025;138(7):808-818
In recent years, the roles of mitochondrial RNA and its associated human diseases have been reported to increase significantly. Treatments based on mtRNA metabolic processes and nuclear gene mutations are thus discussed. The mitochondrial oxidative phosphorylation process is affected by mtRNA metabolism, including mtRNA production, maturation, stabilization, and degradation, which leads to a variety of inherited human mitochondrial diseases. Moreover, mitochondrial diseases are caused by mitochondrial messenger RNA, mitochondrial transfer RNA, and mitochondrial ribosomal RNA gene mutations. This review presents the molecular mechanisms of human mtRNA metabolism and pathological mutations in mtRNA metabolism-related nuclear-encoded/nonencoded genes and mitochondrial DNA mutations to highlight the importance of mitochondrial RNA-related diseases and treatments.
Humans
;
Mitochondrial Diseases/therapy*
;
RNA, Mitochondrial
;
RNA/genetics*
;
Mitochondria/genetics*
;
Mutation/genetics*
;
RNA, Transfer/genetics*
;
DNA, Mitochondrial/genetics*
8.SMUG1 promoted the progression of pancreatic cancer via AKT signaling pathway through binding with FOXQ1.
Zijian WU ; Wei WANG ; Jie HUA ; Jingyao ZHANG ; Jiang LIU ; Si SHI ; Bo ZHANG ; Xiaohui WANG ; Xianjun YU ; Jin XU
Chinese Medical Journal 2025;138(20):2640-2656
BACKGROUND:
Pancreatic cancer is a lethal malignancy prone to gemcitabine resistance. The single-strand selective monofunctional uracil DNA glycosylase (SMUG1), which is responsible for initiating base excision repair, has been reported to predict the outcomes of different cancer types. However, the function of SMUG1 in pancreatic cancer is still unclear.
METHODS:
Gene and protein expression of SMUG1 as well as survival outcomes were assessed by bioinformatic analysis and verified in a cohort from Fudan University Shanghai Cancer Center. Subsequently, the effect of SMUG1 on proliferation, cell cycle, and migration abilities of SMUG1 cells were detected in vitro . DNA damage repair, apoptosis, and gemcitabine resistance were also tested. RNA sequencing was performed to determine the differentially expressed genes and signaling pathways, followed by quantitative real-time polymerase chain reaction and Western blotting verification. The cancer-promoting effect of forkhead box Q1 (FOXQ1) and SMUG1 on the ubiquitylation of myelocytomatosis oncogene (c-Myc) was also evaluated. Finally, a xenograft model was established to verify the results.
RESULTS:
SMUG1 was highly expressed in pancreatic tumor tissues and cells, which also predicted a poor prognosis. Downregulation of SMUG1 inhibited the proliferation, G1 to S transition, migration, and DNA damage repair ability against gemcitabine in pancreatic cancer cells. SMUG1 exerted its function by binding with FOXQ1 to activate the Protein Kinase B (AKT)/p21 and p27 pathway. Moreover, SMUG1 also stabilized the c-Myc protein via AKT signaling in pancreatic cancer cells.
CONCLUSIONS
SMUG1 promotes proliferation, migration, gemcitabine resistance, and c-Myc protein stability in pancreatic cancer via protein kinase B signaling through binding with FOXQ1. Furthermore, SMUG1 may be a new potential prognostic and gemcitabine resistance predictor in pancreatic ductal adenocarcinoma.
Humans
;
Pancreatic Neoplasms/pathology*
;
Forkhead Transcription Factors/genetics*
;
Signal Transduction/genetics*
;
Animals
;
Cell Line, Tumor
;
Proto-Oncogene Proteins c-akt/metabolism*
;
Cell Proliferation/physiology*
;
Mice
;
Uracil-DNA Glycosidase/genetics*
;
Female
;
Male
;
Gemcitabine
;
Mice, Nude
;
Apoptosis/physiology*
;
Deoxycytidine/analogs & derivatives*
;
Cell Movement/genetics*
9.Transposable elements in health and disease: Molecular basis and clinical implications.
Chinese Medical Journal 2025;138(18):2220-2233
Transposable elements (TEs), once considered genomic "junk", are now recognized as critical regulators of genome function and human disease. These mobile genetic elements-including retrotransposons (long interspersed nuclear elements [LINE-1], Alu, short interspersed nuclear element-variable numbers of tandem repeats-Alu [SVA], and human endogenous retrovirus [HERV]) and DNA transposons-are tightly regulated by multilayered mechanisms that operate from transcription through to genomic integration. Although typically silenced in somatic cells, TEs are transiently activated during key developmental stages-such as zygotic genome activation and cell fate determination-where they influence chromatin architecture, transcriptional networks, RNA processing, and innate immune responses. Dysregulation of TEs, however, can lead to genomic instability, chronic inflammation, and various pathologies, including cancer, neurodegeneration, and aging. Paradoxically, their reactivation also presents new opportunities for clinical applications, particularly as diagnostic biomarkers and therapeutic targets. Understanding the dual role of TEs-and balancing their contributions to normal development and disease-is essential for advancing novel therapies and precision medicine.
Humans
;
DNA Transposable Elements/physiology*
;
Animals
;
Long Interspersed Nucleotide Elements/genetics*
;
Neoplasms/genetics*
;
Genomic Instability/genetics*
;
Endogenous Retroviruses/genetics*
10.Protein aggregation in neurodegenerative diseases.
Jiannan WANG ; Lijun DAI ; Zhentao ZHANG
Chinese Medical Journal 2025;138(21):2753-2768
Neurodegenerative diseases constitute a group of chronic disorders characterized by the progressive loss of neurons. Major neurodegenerative conditions include Alzheimer's disease, Parkinson's disease, Huntington's disease, frontotemporal lobar degeneration, and amyotrophic lateral sclerosis. Pathologically, these diseases are marked by the accumulation of aggregates formed by pathological proteins such as amyloid-β, tau, α-synuclein, and TAR DNA-binding protein 43. These proteins assemble into amyloid fibrils that undergo prion-like propagation and dissemination, ultimately inducing neurodegeneration. Understanding the biology of these protein aggregates is fundamental to elucidating the pathophysiology of neurodegenerative disorders. In this review, we summarize the molecular mechanisms underlying the aggregation and transmission of pathological proteins, the processes through which these protein aggregates trigger neurodegeneration, and the interactions between different pathological proteins. We also provide an overview of the current diagnostic approaches and therapeutic strategies targeting pathological protein aggregates.
Humans
;
Neurodegenerative Diseases/metabolism*
;
alpha-Synuclein/metabolism*
;
Amyloid beta-Peptides/metabolism*
;
tau Proteins/metabolism*
;
Protein Aggregation, Pathological/metabolism*
;
DNA-Binding Proteins/metabolism*
;
Animals
;
Protein Aggregates/physiology*


Result Analysis
Print
Save
E-mail