v
Search
Advanced

Publications > Journals > Latest Articles

Results per page:
v
Research Letter Open Access
Chong Yang, Xinyu You, Donghui Cheng, Wenbin Cao, Tao Lu, Wenjun Jiang, Jipeng Jiang, Bangyou Zuo, Yu Zhang
Published online July 22, 2025
Journal of Clinical and Translational Hepatology. doi:10.14218/JCTH.2025.00107
Review Article Open Access
Himani Kumar, Akansha Deshwal, Sneha Datwani, Zaibo Li
Published online July 21, 2025
Journal of Clinical and Translational Pathology. doi:10.14218/JCTP.2025.00021
Abstract
High-grade endometrial carcinoma (HGEC) is an aggressive tumor with increasing incidence and mortality. Traditional classifications, such as Bokhman’s dualistic model and the World [...] Read more.

High-grade endometrial carcinoma (HGEC) is an aggressive tumor with increasing incidence and mortality. Traditional classifications, such as Bokhman’s dualistic model and the World Health Organization histopathological system, have limitations due to tumor heterogeneity and interobserver variability. This review provides a comprehensive understanding of how integrating histopathological and molecular data, particularly The Cancer Genome Atlas (TCGA) classification, advances risk stratification and personalized treatment in HGEC. It highlights current challenges and identifies future directions to improve diagnostic accuracy and patient outcomes through precision medicine.

A literature review was conducted focusing on the epidemiology, histopathology, and molecular profiling of HGEC, with an emphasis on TCGA and next-generation sequencing studies.

TCGA molecular classification stratifies HGEC into four subgroups with distinct prognoses which includes POLE-ultramutated (POLE), microsatellite instability hypermutated, copy number high and copy number low. The next-generation sequencing enhances diagnostic precision and guides personalized treatment. However, diagnostic challenges persist in clinical practice.

Integrating histopathology with TCGA-based molecular profiling refines HGEC classification, enabling improved risk stratification and targeted therapies. Continued efforts to improve diagnostic accuracy are essential to advance patient care.

Full article
Review Article Open Access
Narmadhaa Sivagurunathan, Latchoumycandane Calivarathan
Published online July 21, 2025
Gene Expression. doi:10.14218/GE.2025.00001
Abstract
Acetaminophen (APAP) is one of the most commonly used analgesic and antipyretic medications and is generally considered safe at therapeutic doses. However, overdose remains a leading [...] Read more.

Acetaminophen (APAP) is one of the most commonly used analgesic and antipyretic medications and is generally considered safe at therapeutic doses. However, overdose remains a leading cause of acute liver failure, primarily characterized by centrilobular (zone 3) hepatic necrosis, oxidative stress, mitochondrial dysfunction, and sterile inflammation. The hepatotoxic effects of APAP are localized to the centrilobular region, where cytochrome P450 2E1 is highly expressed. Cytochrome P450 2E1 catalyzes the conversion of APAP to a toxic metabolite, N-acetyl-p-benzoquinone imine. During overdose, the liver’s detoxification capacity is overwhelmed and excess N-acetyl-p-benzoquinone imine binds to cellular proteins, initiating oxidative stress and mitochondrial injury that culminate in hepatocyte death. A central component of APAP-induced hepatotoxicity is the activation of innate immune responses, particularly via inflammasome pathways. Inflammasomes are cytosolic multiprotein complexes that detect cellular damage and trigger inflammation. Among these, the NOD-, LRR-, and pyrin domain-containing 3 (NLRP3) inflammasome plays a significant role in APAP-induced liver injury. Upon activation, the NLRP3 inflammasome promotes autocatalytic cleavage of procaspase-1 into its active form, caspase-1, which subsequently processes the pro-inflammatory cytokines pro-interleukin-1β and pro-interleukin-18 into their mature forms. These cytokines recruit additional immune cells and amplify liver inflammation, exacerbating tissue injury. Thus, the NLRP3 inflammasome serves as a key mechanistic link between the initial toxic insult and the ensuing inflammatory response in APAP hepatotoxicity. This review aimed to explore the molecular mechanisms underlying APAP-induced liver injury, particularly inflammasome activation, and evaluate the current and emerging therapeutic strategies.

Full article
Original Article Open Access
Qian Zhang, Jiajun Wang, Yifan He, Kun Zhang, Wei Hong, Tao Han
Published online July 18, 2025
Journal of Clinical and Translational Hepatology. doi:10.14218/JCTH.2025.00112
Abstract
Autoimmune hepatitis (AIH) is a severe immune-mediated liver disease with limited treatment options beyond immunosuppressants, which carry significant side effects. Existing evidence [...] Read more.

Autoimmune hepatitis (AIH) is a severe immune-mediated liver disease with limited treatment options beyond immunosuppressants, which carry significant side effects. Existing evidence suggests that mesaconate (MSA) possesses immunomodulatory properties and may offer advantages over itaconate derivatives by avoiding succinate dehydrogenase inhibition. However, its specific role in AIH remains unclear. This study aimed to investigate the therapeutic effects of MSA on AIH and to elucidate its underlying mechanisms of action.

A murine AIH model was established via tail vein injection of concanavalin A (ConA, 20 mg/kg). MSA (250 mg/kg) was administered intraperitoneally 6 h before ConA exposure. Liver histology, serum transaminase levels, apoptosis markers, oxidative stress markers, and inflammatory cytokines were analyzed to assess the therapeutic efficacy of MSA. Additionally, RNA sequencing and Western blotting were performed to explore the mechanisms of MSA action. In vitro validation was conducted using RAW264.7 macrophages pretreated with MSA (1 mM) followed by interferon-gamma (IFN-γ, 50 ng/mL) stimulation.

MSA pretreatment effectively mitigated ConA-induced AIH by reducing inflammatory responses, oxidative stress, and apoptosis both in vivo and in vitro. The underlying protective mechanism involved MSA-mediated downregulation of IFN-γ expression and subsequent inhibition of the Janus tyrosine kinase 1/2–signal transducer and activator of transcription 1 signaling pathway. The involvement of this pathway in human AIH was also confirmed.

This study provides the first evidence that MSA ameliorates AIH by suppressing the IFN-γ–Janus tyrosine kinase 1/2–signal transducer and activator of transcription 1 signaling pathway, offering novel mechanistic insights and a promising therapeutic candidate for the future treatment of autoimmune disorders.

Full article
PrevPage 8 of 8 1245678Next
Back to Top