1
|
Shi L, Liu S, Chen J, Wang H, Wang Z. Microglial polarization pathways and therapeutic drugs targeting activated microglia in traumatic brain injury. Neural Regen Res 2026; 21:39-56. [PMID: 39665832 PMCID: PMC12094552 DOI: 10.4103/nrr.nrr-d-24-00810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/03/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024] Open
Abstract
Traumatic brain injury can be categorized into primary and secondary injuries. Secondary injuries are the main cause of disability following traumatic brain injury, which involves a complex multicellular cascade. Microglia play an important role in secondary injury and can be activated in response to traumatic brain injury. In this article, we review the origin and classification of microglia as well as the dynamic changes of microglia in traumatic brain injury. We also clarify the microglial polarization pathways and the therapeutic drugs targeting activated microglia. We found that regulating the signaling pathways involved in pro-inflammatory and anti-inflammatory microglia, such as the Toll-like receptor 4 /nuclear factor-kappa B, mitogen-activated protein kinase, Janus kinase/signal transducer and activator of transcription, phosphoinositide 3-kinase/protein kinase B, Notch, and high mobility group box 1 pathways, can alleviate the inflammatory response triggered by microglia in traumatic brain injury, thereby exerting neuroprotective effects. We also reviewed the strategies developed on the basis of these pathways, such as drug and cell replacement therapies. Drugs that modulate inflammatory factors, such as rosuvastatin, have been shown to promote the polarization of anti-inflammatory microglia and reduce the inflammatory response caused by traumatic brain injury. Mesenchymal stem cells possess anti-inflammatory properties, and clinical studies have confirmed their significant efficacy and safety in patients with traumatic brain injury. Additionally, advancements in mesenchymal stem cell-delivery methods-such as combinations of novel biomaterials, genetic engineering, and mesenchymal stem cell exosome therapy-have greatly enhanced the efficiency and therapeutic effects of mesenchymal stem cells in animal models. However, numerous challenges in the application of drug and mesenchymal stem cell treatment strategies remain to be addressed. In the future, new technologies, such as single-cell RNA sequencing and transcriptome analysis, can facilitate further experimental studies. Moreover, research involving non-human primates can help translate these treatment strategies to clinical practice.
Collapse
Affiliation(s)
- Liping Shi
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Shuyi Liu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Jialing Chen
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Hong Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Zhengbo Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| |
Collapse
|
2
|
Liu S, Chen L, Shang Y. CEACAM5 exacerbates asthma by inducing ferroptosis and autophagy in airway epithelial cells through the JAK/STAT6-dependent pathway. Redox Rep 2025; 30:2444755. [PMID: 39844719 PMCID: PMC11758806 DOI: 10.1080/13510002.2024.2444755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
OBJECTIVES Asthma, a prevalent chronic disease, poses significant health threats and burdens healthcare systems. This study focused on the role of bronchial epithelial cells in asthma pathophysiology. METHODS Bioinformatics was used to identify key asthmarelated genes. An ovalbumin-sensitized mouse model and an IL-13-stimulated Beas-2B cell model were established for further investigation. RESULTS Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) was identified as a crucial gene in asthma. CEACAM5 expression was elevated in asthmatic mouse lung tissues and IL-13-stimulated Beas-2B cells, primarily in bronchial epithelial cells. CEACAM5 induced reactive oxygen species (ROS), lipid peroxidation, and ferroptosis. Interfering with CEACAM5 reduced ROS, malondialdehyde levels, and enhanced antioxidant capacity, while inhibiting iron accumulation and autophagy. Overexpression of CEACAM5 in IL-13-stimulated cells activated the JAK/STAT6 pathway, which was necessary for CEACAM5-induced autophagy, ROS accumulation, lipid peroxidation, and ferroptosis. CONCLUSION CEACAM5 promotes ferroptosis and autophagy in airway epithelial cells via the JAK/STAT6 pathway, exacerbating asthma symptoms. It represents a potential target for clinical treatment.
Collapse
Affiliation(s)
- Si Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Li Chen
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Yunxiao Shang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
3
|
Liang H, Zhou B, Li P, Zhang X, Zhang S, Zhang Y, Yao S, Qu S, Chen J. Stemness regulation in prostate cancer: prostate cancer stem cells and targeted therapy. Ann Med 2025; 57:2442067. [PMID: 39711287 DOI: 10.1080/07853890.2024.2442067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Increasing evidence indicates that cancer stem cells (CSCs) and cancer stem-like cells form a special subpopulation of cells that are ubiquitous in tumors. These cells exhibit similar characteristics to those of normal stem cells in tissues; moreover, they are capable of self-renewal and differentiation, as well as high tumorigenicity and drug resistance. In prostate cancer (PCa), it is difficult to kill these cells using androgen signaling inhibitors and chemotherapy drugs. Consequently, the residual prostate cancer stem cells (PCSCs) mediate tumor recurrence and progression. OBJECTIVE This review aims to provide a comprehensive and up-to-date overview of PCSCs, with a particular emphasis on potential therapeutic strategies targeting these cells. METHODS After searching in PubMed and Embase databases using 'prostate cancer' and 'cancer stem cells' as keywords, studies related were compiled and examined. RESULTS In this review, we detail the origin and characteristics of PCSCs, introduce the regulatory pathways closely related to CSC survival and stemness maintenance, and discuss the link between epithelial-mesenchymal transition, tumor microenvironment and tumor stemness. Furthermore, we introduce the currently available therapeutic strategies targeting CSCs, including signaling pathway inhibitors, anti-apoptotic protein inhibitors, microRNAs, nanomedicine, and immunotherapy. Lastly, we summarize the limitations of current CSC research and mention future research directions. CONCLUSION A deeper understanding of the regulatory network and molecular markers of PCSCs could facilitate the development of novel therapeutic strategies targeting these cells. Previous preclinical studies have demonstrated the potential of this treatment approach. In the future, this may offer alternative treatment options for PCa patients.
Collapse
Affiliation(s)
- Hao Liang
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Bin Zhou
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Peixin Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoyi Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shijie Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Yaozhong Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shengwen Yao
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Sifeng Qu
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Jun Chen
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| |
Collapse
|
4
|
Peng B, Yan MY, Chen YR, Sun F, Xiang XD, Liu D. The methyl-CpG binding domain 2 regulates peptidylarginine deiminase 4 expression and promotes neutrophil extracellular trap formation via the Janus kinase 2 signaling pathway in experimental severe asthma. Ann Med 2025; 57:2458207. [PMID: 39865866 PMCID: PMC11774153 DOI: 10.1080/07853890.2025.2458207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/28/2025] Open
Abstract
OBJECTIVE The prognosis for severe asthma is poor, and the current treatment options are limited. The methyl-CpG binding domain protein 2 (MBD2) participates in neutrophil-mediated severe asthma through epigenetic regulation. Neutrophil extracellular traps (NETs) play a critical role in the pathogenesis of severe asthma. This study aims to detect if MBD2 can reduce NETs formation and the potential mechanism in severe asthma. METHODS A severe asthma model was established in C57BL/6 wild-type mice exposure to house dust mite (HDM), ovalbumin (OVA), and lipopolysaccharide (LPS). Enzyme-linked immunosorbent assay was used to measure the concentrations of IL-4, IL-17A, and IFN-γ in lung tissues. Flow cytometry was employed to determine the percentages of Th2, Th17, and Treg cells in lung tissues. Quantitative real-time polymerase chain reaction was utilized to assess the mRNA expression levels of MBD2, JAK2, and PAD4. Western blotting and immunofluorescence were conducted to detect the protein of MBD2, JAK2, PAD4, and CitH3. HL-60 cells were differentiated into neutrophil-like cells by culturing in a medium containing dimethyl sulfoxide and then stimulated with LPS. KCC-07, Ruxolitinib, and Cl-amidine were used to inhibit the expressions of MBD2, JAK2, and PAD4, respectively. RESULTS Severe asthma mice were characterized by pulmonary neutrophilic inflammation and increased formation of neutrophil extracellular traps (NETs). The expression of MBD2, JAK2, and PAD4 was elevated in severe asthma mice. Inhibiting the expression of MBD2, JAK2, and PAD4 reduced NETs formation and decreased airway inflammation scores, total cell counts and neutrophil counts in BALF, and percentage of Th2 and Th17 cell in lung tissues, whereas increasing Treg cell counts. In both severe asthma mice and HL-60-differentiated neutrophil-like cells in vitro, inhibiting MBD2 reduced the mRNA and protein expression of JAK2 and PAD4, and inhibiting JAK2 reduced the expression of PAD4 mRNA and protein. CONCLUSION MBD2 regulates PAD4 expression through the JAK2 signaling pathway to promote NETs formation in mice with severe asthma. Further bench-based and bedside-based studies targeting the MBD2, PAD4, and JAK2 signaling pathways will help open new avenues for drug development of severe asthma.
Collapse
Affiliation(s)
- Biao Peng
- Department of Pulmonary and Critical Care Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Mu-Yun Yan
- Department of Pulmonary and Critical Care Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Yun-Rong Chen
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People’s Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Fei Sun
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Xu-Dong Xiang
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Da Liu
- Department of Pulmonary and Critical Care Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| |
Collapse
|
5
|
Bick F, Blanchetot C, Lambrecht BN, Schuijs MJ. Targeting γc family cytokines with biologics: current status and future prospects. MAbs 2025; 17:2468312. [PMID: 39967341 PMCID: PMC11845063 DOI: 10.1080/19420862.2025.2468312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
Over the recent decades the market potential of biologics has substantially expanded, and many of the top-selling drugs worldwide are now monoclonal antibodies or antibody-like molecules. The common gamma chain (γc) cytokines, Interleukin (IL-)2, IL-4, IL-7, IL-9, IL-15, and IL-21, play pivotal roles in regulating immune responses, from innate to adaptive immunity. Dysregulation of cell signaling by these cytokines is strongly associated with a range of immunological disorders, which includes cancer as well as autoimmune and inflammatory diseases. Given the essential role of γc cytokines in maintaining immune homeostasis, the development of therapeutic interventions targeting these molecules poses unique challenges. Here, we provide an overview of current biologics targeting either single or multiple γc cytokines or their respective receptor subunits across a spectrum of diseases, primarily focusing on antibodies, antibody-like constructs, and antibody-cytokine fusions. We summarize therapeutic biologics currently in clinical trials, highlighting how they may offer advantages over existing therapies and standard of care, and discuss recent advances in this field. Finally, we explore future directions and the potential of novel therapeutic intervention strategies targeting this cytokine family.
Collapse
Affiliation(s)
- Fabian Bick
- Argenx BV, Zwijnaarde, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | | | - Bart N. Lambrecht
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Martijn J. Schuijs
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
6
|
Liang J, Yang F, Li Z, Li Q. Epigenetic regulation of the inflammatory response in stroke. Neural Regen Res 2025; 20:3045-3062. [PMID: 39589183 PMCID: PMC11881735 DOI: 10.4103/nrr.nrr-d-24-00672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/15/2024] [Accepted: 09/20/2024] [Indexed: 11/27/2024] Open
Abstract
Stroke is classified as ischemic or hemorrhagic, and there are few effective treatments for either type. Immunologic mechanisms play a critical role in secondary brain injury following a stroke, which manifests as cytokine release, blood-brain barrier disruption, neuronal cell death, and ultimately behavioral impairment. Suppressing the inflammatory response has been shown to mitigate this cascade of events in experimental stroke models. However, in clinical trials of anti-inflammatory agents, long-term immunosuppression has not demonstrated significant clinical benefits for patients. This may be attributable to the dichotomous roles of inflammation in both tissue injury and repair, as well as the complex pathophysiologic inflammatory processes in stroke. Inhibiting acute harmful inflammatory responses or inducing a phenotypic shift from a pro-inflammatory to an anti-inflammatory state at specific time points after a stroke are alternative and promising therapeutic strategies. Identifying agents that can modulate inflammation requires a detailed understanding of the inflammatory processes of stroke. Furthermore, epigenetic reprogramming plays a crucial role in modulating post-stroke inflammation and can potentially be exploited for stroke management. In this review, we summarize current findings on the epigenetic regulation of the inflammatory response in stroke, focusing on key signaling pathways including nuclear factor-kappa B, Janus kinase/signal transducer and activator of transcription, and mitogen-activated protein kinase as well as inflammasome activation. We also discuss promising molecular targets for stroke treatment. The evidence to date indicates that therapeutic targeting of the epigenetic regulation of inflammation can shift the balance from inflammation-induced tissue injury to repair following stroke, leading to improved post-stroke outcomes.
Collapse
Affiliation(s)
- Jingyi Liang
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Fei Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Zixiao Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Center for Healthcare Quality Management in Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Engineering Research Center of Digital Healthcare for Neurological Diseases, Beijing, China
| | - Qian Li
- Laboratory for Clinical Medicine, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Tabaa MME, Harty MEE, Mohsen M, Rashad E, Negm WA, Elmorshedy K, Abu-Risha SE. Integrating network pharmacology and in vivo pharmacological validation to explore the gastroprotective mechanism of Sotetsuflavone against indomethacin-induced gastric ulcer in rats: Involvement of JAK2/STAT3 pathway. J Nutr Biochem 2025; 142:109934. [PMID: 40258498 DOI: 10.1016/j.jnutbio.2025.109934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/23/2025]
Abstract
Sotetsuflavone (SF) is an antioxidant flavonoid derived from the Cycas thouarsii R.Br. plant. Although SF regulates numerous cellular pathways influencing inflammation, its antiinflammatory benefits against gastric ulcers are less well-studied. Hence, it is imperative to thoroughly understand the potential gastroprotective mechanisms of SF. This study aimed to explore the effectiveness of SF against indomethacin (IND)-induced gastric ulcers. Network analysis and molecular docking were used to identify the specific targets and pathways related to SF and stomach ulcers. To validate the in vivo pharmacological action of SF, 36 rats were divided into six groups. Ulcer index (UI), protective percentage (PP), gastric mucosal mediators, oxidant/antioxidant status, and inflammatory markers (MIF, M-CSF, and AIF-1) were assessed. Additionally, the expression of PI3K, Akt, Siah2, SOCS3, JAK2, and STAT3 was determined. Stomach histopathology and immunohistochemistry were done. Network pharmacology detected 46 overlapping targets between SF and stomach ulcers, with HIF1A as the primary target among the top hubs. The network also revealed that JAK/STAT, PI3K/Akt, and HIF-1A signaling are among the top 50 markedly enriched KEGG pathways. Furthermore, docking results confirmed that SF has a strong binding affinity towards SOCS3, JAK2, STAT3, M-CSF (CSF-1), and AIF-1. Therefore, we hypothesized that the JAK2/STAT3 pathway may be primarily responsible for SF antiinflammatory action. Through up-regulating SOCS3, SF altered the PI3K/Akt pathway, mitigating oxidative stress, blocking the outflow of inflammatory mediators, and impeding gastric ulcer development. Overall, SF, by the SOCS3-mediated JAK2/STAT3 suppression, might considerably reduce oxidative stress, inflammation, and ulceration caused by indomethacin in the stomach.
Collapse
Affiliation(s)
- Manar M El Tabaa
- Pharmacology & Environmental Toxicology, Environmental Studies & Research Institute (ESRI), University of Sadat City, Sadat City, Menoufia, Egypt.
| | - Mohammed E El Harty
- Pharmacology & Environmental Toxicology, Environmental Studies & Research Institute (ESRI), University of Sadat City, Sadat City, Menoufia, Egypt
| | - Mohamed Mohsen
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Eman Rashad
- Cytology and Histology Department, Faculty of Veterinary Medicine, Cairo University, Egypt
| | - Walaa A Negm
- Pharmacognosy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | | | - Sally E Abu-Risha
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
8
|
Mahjoubin-Tehran M, Rezaei S, Kesharwani P, Karav S, Sahebkar A. Decoy oligodeoxynucleotides targeting STATs in non-cancer gene therapy. Gene 2025; 957:149482. [PMID: 40216342 DOI: 10.1016/j.gene.2025.149482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
The Signal Transducer and Activator of Transcription (STAT) protein family is crucial for organizing the epigenetic configuration of immune cells and controlling various fundamental cell physiological functions including apoptosis, development, inflammation, immunological responses, and cell proliferation and differentiation. The human genome has seven known STAT genes, named 1, 2, 3, 4, 5a, 5b, and 6. Aberrant activation of STAT signaling pathways is associated with many human disorders, particularly cardiovascular diseases (CVDs), making these proteins promising therapeutic targets. Improved understanding of altered and pathological gene expression and its role in the pathophysiology of various hereditary and acquired disorders has enabled the development of novel treatment approaches based on gene expression modulation. One such promising development is the oligodeoxynucleotide decoy method, which may allow researchers to specifically influence gene activation or repression. Various oligodeoxynucleotide decoys target STATs and affect the expression of its downstream genes. We summarized cell culture and preclinical research, which evaluated the effects of oligodeoxynucleotide decoys target STATs in different types of non-cancer illnesses.
Collapse
Affiliation(s)
- Maryam Mahjoubin-Tehran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Rezaei
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact and Outcome, Chitkara University, Rajpura 140417, Punjab, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Zhang M, Yuan L, Yang X, Zhao X, Xie J, Liu X, Wang F. TRAF1 promotes the progression of Helicobacter pylori-associated gastric cancer through EGFR/STAT/OAS signalling. Life Sci 2025; 373:123656. [PMID: 40287055 DOI: 10.1016/j.lfs.2025.123656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/11/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
AIMS Helicobacter pylori (H. pylori) is associated with various gastric diseases and is one of the pathogenic factors of gastric cancer (GC). We found that H. pylori induce the expression of TRAF1, but its mechanism of action is still unclear. Therefore, we wanted to determine whether TRAF1 is involved in the mechanism of H. pylori-related GC progression. MATERIALS AND METHODS In this study, we analysed TRAF1 expression and its prognostic significance using clinical specimens, performed functional studies involving TRAF1 overexpression or knockdown in cellular models, identified downstream signalling pathways regulated via RNA-seq, validated these mechanisms through pathway blockade and rescue experiments, and further confirmed the findings in an H. pylori-infected gastritis mouse model. KEY FINDINGS TRAF1 expression was significantly elevated in GC tissues and served as a poor prognostic biomarker. TRAF1 promoted GC cell proliferation, migration and invasion. RNA-seq analysis revealed that TRAF1 activated the EGFR/STAT/OAS signalling axis, upregulated STAT3 expression and increased the transcription of the OAS gene family. Pharmacological inhibition with ruxolitinib and AG490 effectively blocked EGFR/STAT/OAS signalling. In H. pylori-treated cell models, H. pylori infection activated the EGFR/STAT/OAS signalling axis. In vivo, we established an H. pylori-induced gastritis mouse model to validate the activation of this signalling pathway during the gastritis-carcinoma transition. SIGNIFICANCE TRAF1 may promote the proliferation, migration and invasion of H. pylori-associated GC by activating the EGFR/STAT/OAS signalling axis, suggesting that TRAF1 is a promising novel prognostic biomarker and therapeutic target for this malignancy.
Collapse
Affiliation(s)
- Minglin Zhang
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China
| | - Lingzhi Yuan
- Department of Digestive Nutrition, Hunan Children's Hospital, Central South University Affiliated Children's Hospital, Changsha, China
| | - Xueer Yang
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China
| | - Xuelin Zhao
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China
| | - Jie Xie
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China
| | - Xiaoming Liu
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China.
| | - Fen Wang
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China.
| |
Collapse
|
10
|
Cai XX, Huang YH, Lin YCD, Huang HY, Chen YG, Zhang DP, Zhang T, Liu Y, Zuo HL, Huang HD. A comprehensive review of small molecules, targets, and pathways in ulcerative colitis treatment. Eur J Med Chem 2025; 291:117645. [PMID: 40279769 DOI: 10.1016/j.ejmech.2025.117645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/06/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
Ulcerative colitis (UC), a chronic inflammatory bowel disease (IBD), poses significant clinical challenges because of its complex pathophysiology, long-term nature, and the limited efficacy of existing treatments. Small-molecule compounds, particularly those that are able to modulate inflammation-related signaling pathways and, in many cases, occur in nature, offer a promising alternative or supplement to conventional therapies. Studies on molecules for UC therapeutics reported in 1394 publications over the past 30 years were collected from the Web of Science (WOS) database. Only studies that verified therapeutic efficacy through animal experiments were included. Through an analysis of the molecular classes, structures, common targets, and pathways using network pharmacology, we identified 14 classes of compounds, 5 direct-target modules, and 3 crucial downstream pathways. Alkaloids, phenylpropanoids, flavonoids, and terpenes (and their derivatives) appeared most frequently and mainly targeted lipid metabolism, oxidative stress, immune regulation, signaling transduction, and cancer-related pathways. Notably, there has been an increasing trend of applying naturally sourced compounds in both preclinical and clinical trials, especially flavonoids, over the last five years. Although progress in UC research has been made, the majority of studies have focused on the overall therapeutic effects and biomarker alterations, with limited emphasis on the direct targets and underlying mechanisms. These findings highlight the need to explore novel small-molecule therapeutic strategies for UC, focusing on clearly defined targets and precise modes of action.
Collapse
Affiliation(s)
- Xiao-Xuan Cai
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Yi-Han Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Yang-Chi-Dung Lin
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Hsi-Yuan Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Yi-Gang Chen
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Da-Peng Zhang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China
| | - Tao Zhang
- R&D center, Better Way (Shanghai) Cosmetics Co., Ltd., Shanghai, 201103, PR China
| | - Yue Liu
- R&D center, Better Way (Shanghai) Cosmetics Co., Ltd., Shanghai, 201103, PR China
| | - Hua-Li Zuo
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China.
| | - Hsien-Da Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, PR China; Department of Endocrinology, Key Laboratory of Endocrinology of National Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China.
| |
Collapse
|
11
|
Reuss D, Brown JC, Sukhova K, Furnon W, Cowton V, Patel AH, Palmarini M, Thompson C, Barclay WS. Interference between SARS-CoV-2 and influenza B virus during coinfection is mediated by induction of specific interferon responses in the lung epithelium. Virology 2025; 608:110556. [PMID: 40318419 DOI: 10.1016/j.virol.2025.110556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/15/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Coinfections with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza virus have represented a major health concern since the beginning of the COVID-19 pandemic. The continued spread and constant emergence of new SARS-CoV-2 variants mean that cocirculation and coinfection with seasonal respiratory viruses will continue. Despite the considerable contribution of influenza B virus (IBV) infections to global disease burdens, its interactions with SARS-CoV-2 remain largely unstudied. In this study, we sequentially coinfected lung epithelial cells with representative SARS-CoV-2 variants and IBV strains. We found that prior infection with IBV impaired SARS-CoV-2 D614G, Delta and Omicron BA.1 replication, but did not affect replication of the more recent Omicron EG.5.1 variant. We additionally show that pre-infection with SARS-CoV-2 reduces live attenuated influenza vaccine (LAIV) replication, suggesting vaccine effectiveness in children carrying SARS-CoV-2 pre-infections can be negatively impacted in coinfection. Both SARS-CoV-2 and IBV induced strong type III interferon (IFN) responses, whereas SARS-CoV-2 drove type I IFN production not seen in IBV infection, suggesting viral interference through specific IFN responses. Treatment with innate immune response inhibitors BX795 and Ruxolitinib abrogated viral interference between IBV and SARS-CoV-2 in coinfection, demonstrating that IFN-stimulated gene (ISG) responses play a vital role in viral interference. More specifically, we show that the magnitude and timing of ISG expression, triggered by the primary infecting virus in sequential coinfection, facilitates viral interference between IBV and SARS-CoV-2.
Collapse
Affiliation(s)
- Dorothee Reuss
- Department of Infectious Disease, Imperial College London, UK.
| | | | - Ksenia Sukhova
- Department of Infectious Disease, Imperial College London, UK
| | - Wilhelm Furnon
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Vanessa Cowton
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Arvind H Patel
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | | | | | - Wendy S Barclay
- Department of Infectious Disease, Imperial College London, UK.
| |
Collapse
|
12
|
Weng D, Shi W, Hu Y, Su Y, Li A, Wei S, Guo S. Neutralization of IL-33 ameliorates septic myocardial injury through anti-inflammatory, anti-oxidative, and anti-apoptotic by regulating the NF-κB/STAT3/SOCS3 signaling pathway. Biochem Pharmacol 2025; 237:116954. [PMID: 40258576 DOI: 10.1016/j.bcp.2025.116954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/18/2025] [Accepted: 04/17/2025] [Indexed: 04/23/2025]
Abstract
Septic myocardial injury, a severe sepsis complication linked to high morbidity and mortality, remains a major global clinical challenge. Interleukin-33 (IL-33), a damage-associated pro-inflammatory factor, has been implicated in regulating immune responses and inflammation, but its specific role in septic myocardial injury has not been fully elucidated. This study examined IL-33's role in septic myocardial injury using Gene Expression Omnibus (GEO) database datasets, alongside in vitro and in vivo experiments. Our results indicated a significant upregulation of IL-33 in septic myocardial injury, as demonstrated in both clinical and experimental settings. Blocking IL-33 significantly enhanced cardiac function and alleviated cardiomyocyte damage. Mechanistic investigations revealed that neutralizing IL-33 mitigates inflammation, oxidative stress, and apoptosis in cardiomyocytes by regulating the nuclear factor kappa B (NF-κB)/signal transducer and activator of transcription 3 (STAT3)/suppressors of cytokine signaling 3 (SOCS3) signaling pathway. Peritoneal macrophages are recognized as a potential origin of IL-33, and targeting IL-33 derived from these cells further reduced cardiomyocyte injury. The study underscores IL-33's crucial involvement in septic myocardial injury pathogenesis, indicating that IL-33 may serve as a promising therapeutic target.
Collapse
Affiliation(s)
- Danlei Weng
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing 100020, China
| | - Wei Shi
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing 100020, China
| | - Yue Hu
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing 100020, China
| | - Yanqian Su
- Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing 100020, China
| | - Andong Li
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing 100020, China
| | - Shuxing Wei
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing 100020, China
| | - Shubin Guo
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing 100020, China.
| |
Collapse
|
13
|
Liu H, Xue H, Guo Q, Xue X, Yang L, Zhao K, Liu Y. Ferroptosis meets inflammation: A new frontier in cancer therapy. Cancer Lett 2025; 620:217696. [PMID: 40189012 DOI: 10.1016/j.canlet.2025.217696] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a critical player in cancer pathogenesis. Concurrently, inflammation, a key biological response to tissue injury or infection, significantly influences cancer development and progression. The interplay between ferroptosis and inflammation represents a promising yet underexplored area of research. This review synthesizes recent advances in understanding the molecular mechanisms governing their interaction, emphasizing how ferroptosis triggers inflammatory responses and how inflammatory mediators, such as TNF-α, regulate ferroptosis through iron metabolism and lipid peroxidation pathways. Key molecular targets within the ferroptosis-inflammation axis, including GPX4, ACSL4, and the NF-κB signaling pathway, offer therapeutic potential for cancer treatment. By modulating these targets, it may be possible to enhance ferroptosis and fine-tune inflammatory responses, thereby improving therapeutic outcomes. Additionally, this review explores the broader implications of targeting the ferroptosis-inflammation interplay in disease treatment, highlighting opportunities for developing innovative strategies to combat cancer. By bridging the gap in current knowledge, this review provides a comprehensive resource for researchers and clinicians, offering insights into the therapeutic potential of this intricate biological relationship.
Collapse
Affiliation(s)
- Hu Liu
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Hui Xue
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Qian Guo
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xutong Xue
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Lixue Yang
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China.
| | - Kaijun Zhao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Yu'e Liu
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
14
|
Zhang L, Song Z, Huang X, Jiang B, Shen Y, Li X, Jiang X, Wan J, Xu Q, Liu Q, He Z, Zhao B, Li J, Yan J, Zhang Z, Wang Z. Research progress on the regulation of interstitial cell of Cajal autophagy and apoptosis crosstalk by traditional Chinese medicine in gastrointestinal motility disorders. JOURNAL OF ETHNOPHARMACOLOGY 2025:120128. [PMID: 40513924 DOI: 10.1016/j.jep.2025.120128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 05/15/2025] [Accepted: 06/09/2025] [Indexed: 06/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gastrointestinal motility disorders (GMD) severely impact quality of life, with rising global prevalence linked to modern dietary and lifestyle changes. Traditional Chinese medicine (TCM), rooted in the "holistic regulation" philosophy and centuries of empirical application, demonstrates unique advantages in restoring gastrointestinal homeostasis. Historical records and modern clinical practices validate the efficacy of herbal compounds, bioactive phytochemicals, and external therapies in modulating intestinal pacemaker systems. AIM OF THE STUDY This study aims to systematically review the pathogenesis of GMD mediated by autophagy-apoptosis imbalance in interstitial cells of Cajal (ICCs), examine the therapeutic application of traditional Chinese medicine (TCM) interventions, and identify the bioactive components and molecular mechanisms underlying TCM's regulatory effects on ICCs homeostasis. METHODS Utilize PubMed and NCBI databases to conduct a comprehensive search on GMD , focusing on diseases such as slow transit constipation (STC), functional dyspepsia (FD), diabetic gastroparesis (DGP), gastroesophageal reflux disease (GERD), irritable bowel syndrome (IBS), functional constipation (FC) and opioid-induced constipation (OIC). Additionally, explore the underlying mechanisms involving autophagy, apoptosis, molecular crosstalk, and ICC dynamics. Investigate the potential therapeutic effects of herbal medicine, TCM compounds, bioactive phytocompounds, and external TCM therapeutic modalities on these GMD. RESULTS The study systematically identified 24 TCM compound formulations, 3 bioactive herbal extracts, and 10 specific active components, along with external therapeutic modalities including electroacupuncture (EA) and acupuncture. These therapeutic agents demonstrated multi-pathway regulatory effects by modulating autophagy-apoptosis dynamics in ICCs, with mechanistic analyses revealing their capacity to coordinate multiple signaling pathways for restoring gastrointestinal (GI) motility homeostasis. CONCLUSION TCM and external therapies demonstrate significant therapeutic efficacy in ameliorating GMD. The underlying molecular mechanisms may involve the coordinated modulation of a multi-target regulatory network that restores autophagy-apoptosis homeostasis in ICCs. Given the specificity and adaptability of this mechanistic framework, future research should prioritize the development of active constituents and their corresponding molecular targets as novel therapeutic agents and intervention points. These findings provide both a theoretical foundation and translational directions for advancing precision-targeted strategies in GI motility regulation.
Collapse
Affiliation(s)
- Lumei Zhang
- Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Zhongyang Song
- Gansu University of Traditional Chinese Medicine Affiliated Hospital, Lanzhou, Gansu 730000, China
| | - Xixi Huang
- Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Bing Jiang
- Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Yanyun Shen
- Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Xinyu Li
- Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Xiaoxue Jiang
- Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Jiayi Wan
- Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Qian Xu
- Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Qian Liu
- Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Zhaxicao He
- Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Bing Zhao
- Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Jingwei Li
- Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Jingnan Yan
- Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Zhiming Zhang
- Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Zhigang Wang
- Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Tianshui City Hospital of Traditional Chinese Medicine, Tianshui, Gansu, 743000, China.
| |
Collapse
|
15
|
Senapati S, Bertolini TB, Minnier MA, Yazicioglu MN, Markusic DM, Zhang R, Wicks J, Nahvi A, Herzog RW, Walsh MC, Cejas PJ, Armour SM. Inhibition of IFNAR-JAK signaling enhances tolerability and transgene expression of systemic non-viral DNA delivery. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102502. [PMID: 40206655 PMCID: PMC11979999 DOI: 10.1016/j.omtn.2025.102502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 02/28/2025] [Indexed: 04/11/2025]
Abstract
Lipid nanoparticles (LNPs) have demonstrated significant therapeutic value for non-viral delivery of mRNA and siRNA. While there is considerable interest in utilizing LNPs for delivering DNA (DNA-LNPs) to address a broad range of genetic disorders, acute inflammatory responses pose significant safety concerns and limit transgene expression below therapeutically relevant levels. However, the mechanisms and immune signaling pathways underlying DNA-LNP-triggered inflammatory responses are not well characterized. Through the use of gene-targeted mouse models, we have identified cGAS-STING and interferon-α/β receptor (IFNAR) pathways as major mediators of acute inflammation triggered by systemic delivery of DNA-LNPs. cGAS-STING activation induces expression of numerous JAK-STAT-activating cytokines, and we show that treatment of mice with the JAK inhibitors ruxolitinib or baricitinib significantly improves tolerability to systemically delivered DNA-LNPs. Furthermore, specific inhibition of IFNAR signaling enhances both DNA-LNP tolerability and transgene expression. Utilization of JAK inhibitors or IFNAR blockade represent promising strategies for enhancing the safety and efficacy of non-viral DNA delivery for gene therapy.
Collapse
Affiliation(s)
| | - Thais B. Bertolini
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - David M. Markusic
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rui Zhang
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| | - Joan Wicks
- Gene Therapy Research, Spark Therapeutics, Philadelphia, PA, USA
| | - Ali Nahvi
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| | - Roland W. Herzog
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Pedro J. Cejas
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| | - Sean M. Armour
- Discovery Group, Spark Therapeutics, Philadelphia, PA, USA
| |
Collapse
|
16
|
Verdonk JDJ, Piet B, Ter Heine R, van den Heuvel MM, Smeets RL, Koenen HJPM. Ex vivo pembrolizumab pharmacology for personalized PD-1 inhibitor therapy reveals a critical gap between receptor occupancy and T cell functionality. Int Immunopharmacol 2025; 157:114754. [PMID: 40318274 DOI: 10.1016/j.intimp.2025.114754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025]
Abstract
PURPOSE Targeting the programmed death (ligand)-1 (PD-1/PD-L1) axis with immune checkpoint inhibitors (ICIs), like pembrolizumab, has improved cancer survival. Unfortunately, the optimal dose remains unknown and less than 50 % of patients respond. Understanding PD-1 receptor pharmacology and developing early-response biomarkers are crucial to personalize therapy. We hypothesize that characterizing individual pre-treatment variability in immune responses to pembrolizumab will enhance PD-1 receptor pharmacology insights and improve treatment response prediction. Hence, this study evaluates the performance of a newly developed ex vivo immunopharmacological bioassay under healthy and pathological states. METHODS Peripheral blood mononuclear cells from healthy individuals and non-small cell lung cancer (NSCLC) patients were stimulated in vitro in the presence of pembrolizumab. PD-1 expression, interleukin-2 (IL-2) secretion, T cell differentiation, expression of activation markers and phosphorylation of T cell signalling molecules were analysed. RESULTS In healthy individuals, receptor saturation occurred at >0.025 μg/mL pembrolizumab. Yet IL-2 production still increased significantly beyond this concentration. NSCLC patients showed significantly higher PD-1 expression and IL-2 production than healthy individuals. Nevertheless, pembrolizumab induced IL-2 production similarly in both cohorts. In NSCLC patients, pembrolizumab inhibited differentiation towards CD4 central memory T cells. Remarkably, phosphorylation of proximal phospho-markers in response to pembrolizumab varied between NSCLC patients, potentially discriminating responders from non-responders. CONCLUSIONS We highlight the importance of evaluating T cell functionality alongside PD-1 receptor occupancy. We identified PD-1-induced modulation of phosphorylation of proximal signalling molecules as potential predictors for ICI treatment response in NSCLC. We recommend further development of this immunopharmacological bioassay for personalization of ICI treatment.
Collapse
Affiliation(s)
- Judith D J Verdonk
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Berber Piet
- Department of Pulmonary Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Rob Ter Heine
- Department of Pharmacy, Radboudumc, Nijmegen, The Netherlands
| | | | - Ruben L Smeets
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Hans J P M Koenen
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboudumc, Nijmegen, The Netherlands.
| |
Collapse
|
17
|
Talpaz M, Gerds AT, Lyons R, Langmuir P, Hunter D, Lamothe B, Hou K, McMahon B. A phase 2 study of itacitinib alone or in combination with low-dose ruxolitinib in patients with myelofibrosis. Leuk Res 2025; 155:107732. [PMID: 40516236 DOI: 10.1016/j.leukres.2025.107732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 05/30/2025] [Accepted: 06/04/2025] [Indexed: 06/16/2025]
Abstract
BACKGROUND The Janus kinase (JAK) 1/JAK2 inhibitor ruxolitinib has demonstrated efficacy/safety in patients with myelofibrosis; however, not all patients experience optimal and/or stable response, in part owing to dose-limiting toxicities. This phase 2 study evaluated itacitinib (JAK1-selective inhibitor) efficacy/safety alone or combined with low-dose ruxolitinib. METHODS Cohort A received itacitinib 200 mg once daily (QD) plus ruxolitinib at a previous stable dose (≤15 mg total daily dose). Cohort B (previously ruxolitinib-treated) received itacitinib 600 mg QD alone. The primary endpoint was baseline-to-week 24 spleen volume reduction (SVR). RESULTS Twenty-three patients were enrolled (median age, 71.0 years; intermediate-1/-2 risk, 73.9 %; Cohort A, n = 13; Cohort B, n = 10). Mean (standard deviation) percentage SVR from baseline was +6.9 % (27.5 %) and -3.0 % (34.7 %) in Cohorts A and B at week 24 (primary endpoint), and -1.6 % (14.7 %) and -24.6 % (21.7 %) in Cohorts A and B at week 12. SVR from baseline was achieved by 5 and 3 patients in Cohorts A and B at week 24, and by 9 and 7 patients in Cohorts A and B at week 12. Most common treatment-emergent adverse events (TEAEs) were anemia, diarrhea, and fatigue (each n = 8); most common grade ≥ 3 TEAEs were anemia (n = 6), thrombocytopenia (n = 5), fatigue (n = 3), and diarrhea (n = 2). CONCLUSIONS Overall, 8 of 23 patients enrolled achieved SVR at week 24; larger average changes in SVR at week 12 were observed for itacitinib monotherapy vs. the combination. No unexpected safety signals were observed.
Collapse
Affiliation(s)
- Moshe Talpaz
- Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA.
| | - Aaron T Gerds
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA
| | - Roger Lyons
- Texas Oncology and US Oncology Research, San Antonio, TX, USA
| | | | | | | | - Kevin Hou
- Incyte Corporation, Wilmington, DE, USA
| | - Brandon McMahon
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
18
|
Mao L, Xiong L, Deng Y, Xiong X. A Case of Intense Pulsed Light Aggravated Rosacea Successfully Treated by Abrocitinib. Clin Cosmet Investig Dermatol 2025; 18:1417-1421. [PMID: 40491845 PMCID: PMC12147815 DOI: 10.2147/ccid.s522317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 05/20/2025] [Indexed: 06/11/2025]
Abstract
Rosacea is a chronic inflammatory skin condition characterized by facial erythema, papules, pustules, telangiectasia, and flushing. Currently, various treatment options are available, but no definitive cure has been established. Phototherapy is primarily effective for treating telangiectatic rosacea because it helps alleviate erythema and telangiectasia. However, it can also pose risks; when applied inappropriately, phototherapy may worsen rosacea symptoms, making the condition more difficult to manage. This case report presents a patient with rosacea who experienced acute exacerbation after intense pulsed light therapy, characterized by persistent erythema, edema, pustules, exudation, and a burning sensation with pain. Subsequent treatment with oral abrocitinib for 12 weeks led to a gradual resolution of the patient's facial symptoms. Therefore, we hypothesized that the oral JAK-1 inhibitor abrocitinib not only serves as a promising new treatment option for rosacea but also offers therapeutic benefits in cases of inappropriate phototherapy.
Collapse
Affiliation(s)
- Lan Mao
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Ling Xiong
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yongqiong Deng
- Department of Dermatology, Chengdu Integrated Traditional Chinese and Western Medicine Hospital, Chengdu, Sichuan, People’s Republic of China
| | - Xia Xiong
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| |
Collapse
|
19
|
Su J, Lartey DA, Zanella G, Hawinkels LJAC, Matteoli G, Löwenberg M, Barnhoorn MC. Therapeutic potential of Janus kinase inhibitors for the management of fibrosis in inflammatory bowel disease. J Crohns Colitis 2025; 19:jjaf087. [PMID: 40411492 DOI: 10.1093/ecco-jcc/jjaf087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Indexed: 05/26/2025]
Abstract
Intestinal fibrosis in inflammatory bowel disease (IBD) is caused by uncontrolled accumulation of extracellular matrix deposited by fibroblasts. This may result in stricture formation, especially in Crohn's disease. Since there are no anti-fibrotic drugs available, endoscopic or surgical interventions are the only options to treat intestinal strictures. The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway plays a crucial role in intestinal homeostasis and inflammation. JAK inhibition represents a relatively novel therapeutic strategy in IBD by simultaneously blocking multiple cytokines across various inflammatory pathways. Interestingly, JAK inhibitors extend their benefits beyond anti-inflammatory effects, as they have been shown to interfere with fibrotic processes in various diseases, including IBD. We here summarize the current understanding of the role of the JAK-STAT pathway in the pathogenesis of intestinal fibrosis and the application of JAK inhibitors for IBD. In addition, we discuss the use of JAK inhibitors in other fibrotic-related diseases to postulate how these agents might be applied for future treatment of intestinal fibrosis.
Collapse
Affiliation(s)
- Jie Su
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dalia A Lartey
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Gaia Zanella
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Lukas J A C Hawinkels
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gianluca Matteoli
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Mark Löwenberg
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Marieke C Barnhoorn
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
20
|
Lee SH, Lee H, Kwon YJ, Kim SK, Seo EB, Sohn JO, Kim BH, Park JY, Ye SK. Chalcone-9: a novel inhibitor of the JAK-STAT pathway with potent anti-cancer effects in triple-negative breast cancer cells. Pharmacol Rep 2025; 77:761-774. [PMID: 40199813 PMCID: PMC12066378 DOI: 10.1007/s43440-025-00721-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/25/2025] [Accepted: 03/25/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND Breast cancer remains the leading cause of cancer incidence and mortality among women worldwide, with triple-negative breast cancer (TNBC) posing significant treatment challenges. The dysregulation of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway contributes to tumor progression, making it a potential therapeutic target. Chalcones, known for their diverse biological activities, including anti-cancer effects, hold promise for drug development. This study explores the anti-cancer activity of (E)-4-(3-(2-(benzyloxy)-6-hydroxyphenyl)-3-oxoprop-1-en-1-yl)benzoic acid (chalcone-9), a novel chalcone derivative. METHODS The cytotoxic effects of chalcone-9 were evaluated in breast cancer cell lines, including TNBC lines MDA-MB-231 and MDA-MB-468. Western blotting and qRT-PCR were used to analyze the impact on JAK1, JAK2, STAT1, and STAT3 activation and their downstream gene expression. In silico molecular docking analysis was conducted to determine whether chalcone-9 can interact with JAK1 and JAK2. A wound healing assay was used to observe the effect of chalcone-9 on tumor cell migration, and flow cytometry was employed to analyze whether chalcone-9 inhibits tumor cell cycle progression and induces apoptosis. The expression of apoptosis markers was also assessed. RESULTS Chalcone-9 exhibited dose-dependent cytotoxicity in breast cancer cell lines, with TNBC cells showing higher sensitivity. Chalcone-9 effectively inhibited the activation of JAK1, JAK2, STAT1, and STAT3, outperforming conventional JAK/STAT inhibitors. The structure of chalcone-9 was confirmed to stably interact with JAK1 and JAK2 proteins. It also suppressed STAT1 and STAT3 target gene expression, reduced tumor cell migration, and induced apoptosis, as evidenced by PARP and caspase cleavage and decreased survivin levels. CONCLUSIONS Chalcone-9 demonstrates significant anti-cancer activity, particularly against TNBC. By targeting the JAK/STAT pathway and promoting apoptosis, chalcone-9 emerges as a promising therapeutic candidate for aggressive breast cancers.
Collapse
Affiliation(s)
- Song-Hee Lee
- Department of Biomedical Sciences and Pharmacology, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
| | - Haeri Lee
- Department of Biomedical Sciences and Pharmacology, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Yong-Jin Kwon
- Department of Cosmetic Science, Kyungsung University, Busan, 48434, Republic of Korea
| | - Seul-Ki Kim
- Department of Biomedical Sciences and Pharmacology, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
| | - Eun-Bi Seo
- Department of Biomedical Sciences and Pharmacology, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jie Ohn Sohn
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Gangwon-Do, 25159, Republic of Korea
| | - Byung-Hak Kim
- Medience Co. Ltd., Chuncheon, Gangwon-Do, 24232, Republic of Korea
| | - Jung-Youl Park
- Glocal University Project Group, Andong National University, Andong, Gyeongsangbuk-Do, 36729, Republic of Korea
| | - Sang-Kyu Ye
- Department of Biomedical Sciences and Pharmacology, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea.
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Gangwon-Do, 25159, Republic of Korea.
- Biomedical Science Project (BK21PLUS), Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Neuro-Immune Information Storage Network Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
21
|
Abdel-Megeed RM, Abdel-Hamid AHZ, Kadry MO. Titanium nanostructure mitigating doxorubicin-induced testicular toxicity in rats via regulating major autophagy signaling pathways. Toxicol Rep 2025; 14:101869. [PMID: 39811821 PMCID: PMC11731616 DOI: 10.1016/j.toxrep.2024.101869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Doxorubicin (DOX) is a powerful antineoplastic FDA-approved anthracycline-derived antibiotic and is considered as the most suitable intervention for solid tumors and hematological cancers therapy. However, its therapeutic application is highly limited due to acute and chronic renal, hematological and testicular toxicity. Oxidative stress, lipid peroxidation and apoptosis in germ cells as well as low sperm count, motility and disturbing steroidogenesis are the principal machineries of DOX-induced testicular toxicity. Nevertheless, the comprehensive molecular pathways responsible for DOX-induced testicular damage are not yet fully understood. The current study aims to clarify the role of autophagy and apoptotic signaling pathways in testicular toxicity induced by DOX in the rat model. The study also investigates the potential role of both titanium dioxide nanoparticles (TiO2NP) loaded with DOX and Lactoferrin in combination with DOX in mitigating testicular toxicity induced by DOX the standard antitumor drug. In the present study, male Wister albino rats were intoxicated with a total cumulative dose of DOX (18 mg/kg) via intra-peritoneal injection and served as positive control group. The other two groups administered either TiO2NP-DOX or lactoferrin-DOX. Furthermore, biochemical and molecular analyses were then performed. DOX intoxication induced testicular toxicity, revealing mineral imbalance as indicated by an increase in both calcium and magnesium concentrations. Administration of either TiO2NP-DOX or lactoferrin-DOX resulted in a significant modulation of disrupted mineral concentrations, with TiO2NP-DOX showing superiority in modulating both magnesium and calcium concentrations. Acid Phosphatase level significantly increased upon DOX-induced testicular damage. Molecular analysis of EGFR and K-RAS gene expression showed significant overexpression, while p53 and JAK-2 gene expression was significantly reduced post-DOX intoxication. Protein expression of both AKT and PI3K significantly increased upon DOX administration. Results showed a remarkable modulation of all disrupted gene and protein expressions upon treatment with TiO2NP-DOX or Lactoferrin-DOX with the superiority of TiO2NP-DOX in modulating these parameters. In conclusion, TiO2NP-DOX could be a promising drug delivery system to improve bioavailability and drug release, as well as reducing DOX's adverse effects particularly on testicular function.
Collapse
Affiliation(s)
- Rehab M. Abdel-Megeed
- Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, El Buhouth St., Dokki, Cairo 12622, Egypt
| | - Abdel-Hamid Z. Abdel-Hamid
- Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, El Buhouth St., Dokki, Cairo 12622, Egypt
| | - Mai O. Kadry
- Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, El Buhouth St., Dokki, Cairo 12622, Egypt
| |
Collapse
|
22
|
Shammas T, Peiris MN, Meyer AN, Donoghue DJ. BCR-ABL: The molecular mastermind behind chronic myeloid leukemia. Cytokine Growth Factor Rev 2025; 83:45-58. [PMID: 40360311 DOI: 10.1016/j.cytogfr.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025]
Abstract
The chromosomal translocation t(9;22)(q34;q11), known as the Philadelphia (Ph) chromosome, results in the BCR-ABL gene fusion which gives rise to Chronic Myeloid Leukemia (CML), a slowly progressing hematopoietic cancer that begins in the bone marrow of the patient. Making up about 15 % of all new leukemia cases, CML remains a critical focus of cancer research and treatment due to its distinctive genetic hallmark, the BCR-ABL fusion gene. The BCR-ABL fusion protein is a constitutively active tyrosine kinase which signals to multiple pathways including the Ras/MAPK, PI3K/AKT, JAK/STAT and NF-kappaB pathways which promote uncontrolled cell proliferation and survival. While multiple tyrosine kinase inhibitors (TKIs) are used to specifically target the fusion in the treatment of CML, new therapies are becoming available to overcome the resistance that occurs during TKI treatments of the disease. The discovery of the Philadelphia chromosome and the subsequent elucidation of the BCR-ABL fusion protein have since become a paradigm for understanding the genetic basis of cancer and developing precision medicine approaches. This review highlights the etiology and historical discovery of the BCR-ABL fusion, recent advances in understanding its regulatory mechanisms, and emerging strategies for its therapeutic targeting.
Collapse
MESH Headings
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Protein Kinase Inhibitors/therapeutic use
- Animals
- Signal Transduction
- Philadelphia Chromosome
Collapse
Affiliation(s)
- Tara Shammas
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093-0367, USA
| | - Malalage N Peiris
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA
| | - April N Meyer
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093-0367, USA
| | - Daniel J Donoghue
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093-0367, USA; UCSD Moores Cancer Center, University of California San Diego, La Jolla, CA 92093-0367, USA.
| |
Collapse
|
23
|
Han L, Sun Y, Yang K, Long C. Inhibition of JAK2/STAT3 by pacritinib synergizes with chemotherapy in esophageal carcinoma. Toxicol In Vitro 2025; 106:106056. [PMID: 40112935 DOI: 10.1016/j.tiv.2025.106056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
The poor outcomes associated with esophageal carcinoma, particularly in advanced stages, necessitate the development of new treatment strategies. This study examines the efficacy of pacritinib, a multi-kinase inhibitor, both alone and in combination with carboplatin, in preclinical esophageal carcinoma models. Six esophageal carcinoma cell lines (KYSE-70, OE33, FLO-1, KYAE-1, ESO 26, and HCE-6) were treated with pacritinib, resulting in a dose-dependent reduction in cell viability. Combination index (CI) analysis demonstrated strong synergy between pacritinib and carboplatin across this panel of cell lines. In in vivo esophageal carcinoma xenograft model, pacritinib alone significantly reduced tumor growth and improved survival rates compared to control. Notably, the combination of pacritinib and carboplatin further reduced tumor growth and improved survival rates compared to either treatment alone. Toxicity assessment showed that neither single-agent nor combination treatment resulted in significantly altered levels of body weight and serum markers, supporting the safety profile of pacritinib in combination with carboplatin. Mechanistic studies revealed that while pacritinib inhibited the phosphorylation of JAK, STAT3, and IRAK1 in esophageal carcinoma cells, it is the suppression of the JAK/STAT3 pathway, rather than IRAK1, that is responsible for the synergistic effect with carboplatin. Our findings indicate that pacritinib possesses potent anti-tumor activity in esophageal carcinoma and enhances the efficacy of carboplatin through the suppression of JAK/STAT3 signaling, warranting further clinical investigation.
Collapse
Affiliation(s)
- Lan Han
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Yinyin Sun
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Kai Yang
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China.
| | - Cheng Long
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China.
| |
Collapse
|
24
|
Nene S, Devabattula G, Vambhurkar G, Tryphena KP, Khatri DK, Godugu C, Singh PK, Srivastava S. Topical delivery of baricitinib-impregnated nanoemulgel: a promising platform for inhibition of JAK -STAT pathway for the effective management of atopic dermatitis. Drug Deliv Transl Res 2025; 15:2200-2219. [PMID: 39467941 DOI: 10.1007/s13346-024-01732-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2024] [Indexed: 10/30/2024]
Abstract
Baricitinib, an inhibitor of Janus kinase 1/2 receptors majorly involved in the dysregulation of immune responses in atopic dermatitis, is currently approved for managing atopic dermatitis in Europe. The delivery of baricitinib through oral route is associated to several adverse effects due to off-target effects. Therefore, the current study is aimed at formulation of baricitinib loaded nanoemulgel for evaluation of topical delivery potential in the treatment of atopic dermatitis. The baricitinib-loaded nanoemulsions (0.05 and 0.1% w/w) revealed an average globule size of 162.86 ± 0.37 and 173.66 ± 4.88 nm respectively with narrow PDI. The optimized batch of baricitinib nanoemulsion was converted to nanoemulgel by the addition of the mixture of gel bases SEPINEO™ DERM and SEPINEO™ P 600 along with propylene glycol, resulting in pseudoplastic shear thinning behaviour. The optimized nanoemulgels have shown prominent retention of baricitinib in the skin along with permeation. The skin distribution study of coumarin-6 loaded nanoemulgel demonstrated high fluorescence in the epidermal layer. The western blot analysis revealed significant inhibition of phosphorylated signal transducers and activators of transcriptions 1 (##p < 0.01) and 3 (#p < 0.05) by application of 0.05 and 0.1% baricitinib nanoemulgel. The baricitinib nanoemulgels have shown anti-inflammatory activity by significantly reducing expressions of various inflammatory markers. Histopathological analysis of skin tissues treated with baricitinib nanoemulgel has demonstrated a marked reduction in acanthosis, hyperkeratosis, and intact outer epidermis. These results supported the potential role of baricitinib-loaded nanoemulgel in reducing the inflammation and disease severity associated with atopic dermatitis.
Collapse
Affiliation(s)
- Shweta Nene
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Geetanjali Devabattula
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Ganesh Vambhurkar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Kamatham Pushpa Tryphena
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Dharmendra Kumar Khatri
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Chandraiah Godugu
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Pankaj Kumar Singh
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India.
| |
Collapse
|
25
|
Severino P, D'Amato A, Prosperi S, Myftari V, Germanò R, Marek-Iannucci S, De Prisco A, Mariani MV, Marchiori L, Battaglia C, Tabacco L, Segato C, Mancone M, Fedele F, Vizza CD. Coronary microcirculation in myocardial ischemia: A genetic perspective. J Mol Cell Cardiol 2025; 203:67-75. [PMID: 40220989 DOI: 10.1016/j.yjmcc.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Coronary microvascular dysfunction (CMD) is a major contributor to ischemic heart disease (IHD), acting both independently and together with atherosclerosis. CMD encompasses structural and functional microcirculatory changes that result in dysregulated coronary blood flow. Structural abnormalities include microvascular remodeling, resulting in arteriolar and capillary narrowing, perivascular fibrosis and capillary rarefaction. Endothelial dysfunction and smooth muscle cell hyperactivity further impair microcirculation. Genetic factors may play a crucial role in the pathophysiology of CMD, mainly due to single nucleotide polymorphisms (SNPs) in genes that regulate coronary blood flow and microcirculation structural modifications. This manuscript aims to review the genetic determinants of CMD, with particular focus on ion channels, microRNAs (miRNAs), and proteins involved in the endothelial environment. The improving knowledge about genetic aspects of CMD opens the possibility to have new biomarkers, improving diagnosis and the development of targeted treatments in light of an even more patient-tailored approach.
Collapse
Affiliation(s)
- Paolo Severino
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Andrea D'Amato
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Silvia Prosperi
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Vincenzo Myftari
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Rosanna Germanò
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Stefanie Marek-Iannucci
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Andrea De Prisco
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Marco Valerio Mariani
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Ludovica Marchiori
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Corinne Battaglia
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Leonardo Tabacco
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Camilla Segato
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Massimo Mancone
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | | | - Carmine Dario Vizza
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| |
Collapse
|
26
|
Kumari S, Peela S, Srilatha M, Girish BP, Nagaraju GP. Adiponectin: its role in diabetic and pancreatic cancer. Mol Aspects Med 2025; 103:101370. [PMID: 40403652 DOI: 10.1016/j.mam.2025.101370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 04/24/2025] [Accepted: 05/18/2025] [Indexed: 05/24/2025]
Abstract
Adiponectin (ApN) is an antidiabetic and anti-inflammatory protein synthesized by adipose tissue. It is essential in regulating insulin sensitivity, glucose, and lipid metabolism by controlling AMPK, PPARα, and MAPK signals. It is an anti-inflammatory property that protects pancreatic β-cells. Often, low levels of ApN are linked to obesity, type II diabetes and the development of PDAC. However, changes in lifestyle and the use of certain drugs can improve ApN function and insulin sensitivity. PDAC is a highly aggressive cancer linked to obesity, type II diabetes, and insulin resistance. ApN plays a complex role in PDAC progression and can suppress PDAC development by weakening β-catenin signaling. Decreases in ApN levels are associated with increased PDAC risk in diabetic patients. PDAC and diabetes are interconnected through the development of insulin resistance, islet dysfunction, change in immunological response, inflammation, oxidative stress, and altered hormone secretion. Genetic studies highlight specific genes like HNF4G and PDX1 that influence both conditions and miRNAs such as miR-19a promote tumor progression through the PI3K/AKT pathway. This review discusses the role of ApN in diabetes and PDAC and the interrelation between diabetes and PDAC.
Collapse
Affiliation(s)
- Seema Kumari
- Department of Biotechnology, Dr.B.R. Ambedkar University, Srikakulam, 532410, AP, India
| | - Sujatha Peela
- Department of Biotechnology, Dr.B.R. Ambedkar University, Srikakulam, 532410, AP, India
| | - Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati, Andhra Pradesh, 517502, India
| | - Bala Prabhakar Girish
- Regional Agricultural Research Station, Institute of Frontier Technology, Acharya N G Ranga Agricultural University, Tirupati, India
| | - Ganji Purnachandra Nagaraju
- School of Medicine, Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| |
Collapse
|
27
|
Zhou M, Yin L, Siu CW. Comment on: Long-term outcomes of childhood-onset systemic lupus erythematosus. Rheumatology (Oxford) 2025; 64:4084-4085. [PMID: 39250729 DOI: 10.1093/rheumatology/keae482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/11/2024] Open
Affiliation(s)
- Mi Zhou
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong SAR, China
- Institute of Ultrasound in Medicine, Sichuan Provincial People's Hospital Wenjiang Hospital, Chengdu, China
| | - Lixue Yin
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chung-Wah Siu
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
28
|
Hamdy H, Aly WA, Elkord E. Investigating the functional role of BUB1B in aflatoxin B1-associated hepatocarcinogenesis. Toxicology 2025; 514:154127. [PMID: 40147685 DOI: 10.1016/j.tox.2025.154127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide, stemming from a complex interplay of genetic, environmental, and lifestyle factors. Aflatoxin B1 (AFB1), a prevalent food contaminant, is a known HCC risk factor, but its molecular mechanisms remain incompletely understood. This study investigated the contribution of BUB1B, a crucial spindle assembly checkpoint regulator, in AFB1-induced hepatocyte malignant transformation, we assessed AFB1's impact on cell proliferation, viability, cell cycle regulation, and BUB1B expression. BUB1B knockdown via siRNA revealed its role in epithelial-mesenchymal transition (EMT), cell motility, and proliferation. AFB1 exposure significantly altered cell proliferation and cell cycle dynamics, correlating with increased BUB1B expression. Furthermore, we identified a significant interaction between BUB1B and the IL12A-JAK2/STAT4 signaling pathway, suggesting a mechanism for immune evasion and tumor progression. These findings highlight BUB1B's critical role in AFB1-induced hepatocarcinogenesis and establish its potential target for HCC. Further research is needed to fully elucidate the underlying molecular mechanisms and explore the therapeutic implications of BUB1B inhibition in HCC treatment.
Collapse
Affiliation(s)
- Hayam Hamdy
- Center for Life Sciences, School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Medicine, Yunnan University, Kunming, China; Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, New Valley University, New Valley, Egypt
| | - Wafaa A Aly
- Department of Environmental Health, Institute of Environmental Studies, Arish University, Egypt
| | - Eyad Elkord
- Department of Biosciences and Bioinformatics & Suzhou Municipal Key Lab of Biomedical Sciences and Translational Immunology, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China; College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates; Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, Manchester, UK.
| |
Collapse
|
29
|
Abe GL, Xiao L, Sasaki JI, Sakai H, Kitagawa H, Kohno T, Funayama N, Imazato S. Fast Lithium-Releasing Phosphate Glasses Promote the Resolution of Inflammation. ACS APPLIED BIO MATERIALS 2025. [PMID: 40448634 DOI: 10.1021/acsabm.5c00128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Abstract
Delay in the resolution of inflammation has disastrous implications for tissue regeneration, but local anti-inflammatory signals could accelerate this process. To test this, phosphate-based bioactive glasses were fabricated to release lithium, an anti-inflammatory ion, at a range of ionic concentrations. Lithium release was contingent on the aluminum molar ratio within the glass formulation. Glasses that released lithium faster exerted greater anti-inflammatory effects on activated macrophages. These effects resulted from the inhibition of GSK3β activity and the promotion of CD206 expression. This study demonstrates the therapeutic potential of anti-inflammatory phosphate glasses in resolving inflammation in the regenerative environment.
Collapse
Affiliation(s)
- Gabriela L Abe
- Joint Research Laboratory of Advanced Functional Materials Science, Osaka University Graduate School of Dentistry, Suita 565-0871, Japan
| | - Linghao Xiao
- Joint Research Laboratory of Advanced Functional Materials Science, Osaka University Graduate School of Dentistry, Suita 565-0871, Japan
| | - Jun-Ichi Sasaki
- Joint Research Laboratory of Advanced Functional Materials Science, Osaka University Graduate School of Dentistry, Suita 565-0871, Japan
- Department of Dental Biomaterials, Osaka University Graduate School of Dentistry, Suita 565-0871, Japan
| | - Hirohiko Sakai
- Department of Dental Biomaterials, Osaka University Graduate School of Dentistry, Suita 565-0871, Japan
| | - Haruaki Kitagawa
- Joint Research Laboratory of Advanced Functional Materials Science, Osaka University Graduate School of Dentistry, Suita 565-0871, Japan
- Department of Dental Biomaterials, Osaka University Graduate School of Dentistry, Suita 565-0871, Japan
| | - Tomoki Kohno
- Joint Research Laboratory of Advanced Functional Materials Science, Osaka University Graduate School of Dentistry, Suita 565-0871, Japan
| | - Naoya Funayama
- Joint Research Laboratory of Advanced Functional Materials Science, Osaka University Graduate School of Dentistry, Suita 565-0871, Japan
| | - Satoshi Imazato
- Joint Research Laboratory of Advanced Functional Materials Science, Osaka University Graduate School of Dentistry, Suita 565-0871, Japan
- Department of Dental Biomaterials, Osaka University Graduate School of Dentistry, Suita 565-0871, Japan
| |
Collapse
|
30
|
Battilotti C, Azzella G, Dattola A, Rossi A, Svara F, Chello C, Del Duca E, Paolino G, Nisticò SP, Pellacani G, Grieco T. Efficacy of Upadacitinib in Treating Alopecia Areata, Atopic Dermatitis, and Th1 Comorbidities in Pediatric Patients: A Comprehensive Case Series and Literature Review. J Clin Med 2025; 14:3881. [PMID: 40507644 PMCID: PMC12155964 DOI: 10.3390/jcm14113881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2025] [Revised: 05/23/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025] Open
Abstract
Alopecia areata (AA) and atopic dermatitis (AD) are complex immune-mediated conditions that frequently coexist in pediatric patients, complicating treatment approaches. Upadacitinib, a selective JAK1 inhibitor, modulates both Th1 and Th2 pathways and is approved for AD in adolescents and adults. This study presents a case series of three adolescent patients with refractory AA and AD treated with upadacitinib 15 mg/day for 12 months, alongside a comprehensive literature review. All patients demonstrated rapid remission of AD symptoms within the first month and progressive hair regrowth, with SALT scores significantly improving at six and twelve months. No severe adverse events were reported. Notably, one patient achieved complete regrowth despite the presence of ophiasis, a pattern typically associated with poor prognosis. Our literature review identified only four previous pediatric cases successfully treated with upadacitinib, highlighting the novelty of our findings. These cases, together with our experience, suggest that upadacitinib offers a safe and effective therapeutic option for pediatric patients with concomitant AA and AD, including those who failed conventional or biologic therapies such as dupilumab. Larger, controlled studies are needed to confirm long-term efficacy and safety. Our results also support the potential role of upadacitinib in managing multiple Th1/Th2-mediated comorbidities in pediatric populations.
Collapse
Affiliation(s)
- Chiara Battilotti
- Dermatologic Clinic, Sapienza University of Rome, 00185 Rome, Italy; (C.B.); (G.A.); (A.D.); (A.R.); (C.C.); (E.D.D.); (S.P.N.); (G.P.); (T.G.)
| | - Giulia Azzella
- Dermatologic Clinic, Sapienza University of Rome, 00185 Rome, Italy; (C.B.); (G.A.); (A.D.); (A.R.); (C.C.); (E.D.D.); (S.P.N.); (G.P.); (T.G.)
| | - Annunziata Dattola
- Dermatologic Clinic, Sapienza University of Rome, 00185 Rome, Italy; (C.B.); (G.A.); (A.D.); (A.R.); (C.C.); (E.D.D.); (S.P.N.); (G.P.); (T.G.)
| | - Alfredo Rossi
- Dermatologic Clinic, Sapienza University of Rome, 00185 Rome, Italy; (C.B.); (G.A.); (A.D.); (A.R.); (C.C.); (E.D.D.); (S.P.N.); (G.P.); (T.G.)
| | - Francesca Svara
- Dermatologic Clinic, Sapienza University of Rome, 00185 Rome, Italy; (C.B.); (G.A.); (A.D.); (A.R.); (C.C.); (E.D.D.); (S.P.N.); (G.P.); (T.G.)
| | - Camilla Chello
- Dermatologic Clinic, Sapienza University of Rome, 00185 Rome, Italy; (C.B.); (G.A.); (A.D.); (A.R.); (C.C.); (E.D.D.); (S.P.N.); (G.P.); (T.G.)
| | - Ester Del Duca
- Dermatologic Clinic, Sapienza University of Rome, 00185 Rome, Italy; (C.B.); (G.A.); (A.D.); (A.R.); (C.C.); (E.D.D.); (S.P.N.); (G.P.); (T.G.)
| | - Giovanni Paolino
- Unit of Dermatology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy;
- Unit of Clinical Dermatology, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Steven P. Nisticò
- Dermatologic Clinic, Sapienza University of Rome, 00185 Rome, Italy; (C.B.); (G.A.); (A.D.); (A.R.); (C.C.); (E.D.D.); (S.P.N.); (G.P.); (T.G.)
| | - Giovanni Pellacani
- Dermatologic Clinic, Sapienza University of Rome, 00185 Rome, Italy; (C.B.); (G.A.); (A.D.); (A.R.); (C.C.); (E.D.D.); (S.P.N.); (G.P.); (T.G.)
| | - Teresa Grieco
- Dermatologic Clinic, Sapienza University of Rome, 00185 Rome, Italy; (C.B.); (G.A.); (A.D.); (A.R.); (C.C.); (E.D.D.); (S.P.N.); (G.P.); (T.G.)
| |
Collapse
|
31
|
Sakuludomkan C, Khowsathit J, Thippraphan P, Koonrungsesomboon N, Takuathung MN, Taychaworaditsakul W. Network Pharmacology and Molecular Docking-Based Approach to Explore Potential Bioactive Compounds from Kaempferia parviflora on Chemokine Signaling Pathways in the Treatment of Psoriasis Disease. Int J Mol Sci 2025; 26:5243. [PMID: 40508056 PMCID: PMC12154073 DOI: 10.3390/ijms26115243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Revised: 05/23/2025] [Accepted: 05/28/2025] [Indexed: 06/16/2025] Open
Abstract
Psoriasis is a chronic inflammatory skin disorder characterized by keratinocyte hyperproliferation and dysregulated chemokine signaling. Kaempferia parviflora (KP) has long been valued for its medicinal properties; however, its specific role in psoriasis treatment remains unclear. This study investigates the anti-psoriatic potential of methoxyflavones derived from KP through an integrated approach combining network pharmacology, molecular docking, and experimental validation. A total of 232 target genes were identified as being associated with KP bioactive compounds, of which 64 overlapped with psoriasis-related genes implicated in chemokine signaling pathways. Molecular docking analyses revealed that key methoxyflavones interact with pivotal proteins such as protein kinase B (AKT1 or AKT), proto-oncogene tyrosine-protein kinase (SRC), and phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1), suggesting their potential involvement in modulating inflammation. Experimental results confirmed that 5,7,4'-trimethoxyflavone and 3,5,7-trimethoxyflavone significantly inhibited keratinocyte proliferation, migration, and macrophage activation, key processes in psoriasis progression. Additionally, both compounds reduced nitric oxide production, supporting their anti-inflammatory effects. Western blot analysis further demonstrated that these compounds tended to decrease the phosphorylation levels of AKT and SRC, supporting their role in influencing inflammatory signaling pathways. These findings suggest that methoxyflavones from KP act through multi-target mechanisms, offering potential as natural therapeutic agents for psoriasis. Further, in vivo studies are needed to validate their efficacy and explore their clinical applications.
Collapse
Affiliation(s)
- Chotiwit Sakuludomkan
- Clinical Research Center for Food and Herbal Product Trials and Development (CR-FAH), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.S.); (N.K.); (M.N.T.)
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jittasak Khowsathit
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (J.K.); (P.T.)
| | - Pilaiporn Thippraphan
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (J.K.); (P.T.)
| | - Nut Koonrungsesomboon
- Clinical Research Center for Food and Herbal Product Trials and Development (CR-FAH), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.S.); (N.K.); (M.N.T.)
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Mingkwan Na Takuathung
- Clinical Research Center for Food and Herbal Product Trials and Development (CR-FAH), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.S.); (N.K.); (M.N.T.)
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Weerakit Taychaworaditsakul
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (J.K.); (P.T.)
| |
Collapse
|
32
|
Huang P, Yu X, Gao Z, Yang Q, Lin Y, Tu J, Cao Y, Liu Y, Su W, Cao G. Shenfu injection ameliorates hepatic ischemia-reperfusion injury through induction of ferroptosis via JAK2/STAT3 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119889. [PMID: 40316154 DOI: 10.1016/j.jep.2025.119889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/04/2025] [Accepted: 04/25/2025] [Indexed: 05/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shenfu injection (SF) is a traditional Chinese medicine (TCM) compound preparation developed from the Shenfu decoction, which is described in the ancient Chinese medical book "Ji Sheng Fang" from the Song Dynasty. It is composed of two traditional Chinese medicines: Ginseng Radix et Rhizoma Rubra (G. Radix) and Aconiti Lateralis Radix Preparata (A. Lateralis). SF is renowned for its effects of restoring yang, rescuing adverse conditions, replenishing qi and consolidating deficiency. Research indicated that SF may enhance the recovery from Hepatic ischemia-reperfusion injury (HIRI), although its potential pharmacological mechanisms remain to be clearly defined. AIM OF THE STUDY To explore the pharmacological effects and mechanisms of SF in the treatment of HIRI. MATERIALS AND METHODS This study employed network pharmacology alongside both in vivo and in vitro experimental validation. It involved retrieving drug ingredients and targets from a database, constructing networks of chemical composition-targets- pathways and protein-protein interactions to pinpoint key targets. To evaluate the binding affinity between active ingredients and their respective targets, molecular docking was employed. Further, the study predicted potential targets and signaling pathways influenced by SF through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analyses. Finally, in vivo and in vitro experimental validation were performed using the HIRI mouse model and the oxygen-glucose deprivation/reperfusion (OGD/R)-induced AML12 cells model. RESULTS Findings from in vivo experiments indicated that SF could markedly lower serum glutamic pyruvic transaminase (ALT) and glutamic oxaloacetic transaminase (AST) levels, improve hepatic histopathological damage, reduce the count of myeloperoxidase (MPO) positive cells, and decrease the levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) in HIRI mice. Moreover, we investigated ferroptosis-related biomarkers and found that pretreatment with SF could significantly reduce the levels of reactive oxygen species (ROS), malondialdehyde (MDA), and iron in both HIRI mice and OGD/R-induced AML12 cells. Furthermore, it boosted the activity of superoxide dismutase (SOD) and glutathione (GSH), upregulated the protein expression of glutathione Peroxidase 4 (GPX4), and elevated the mRNA expression of GPX4, solute carrier family 7 member 11 (SLC7A11), and prostaglandin-endoperoxide synthase 2 (PTGS2) in both in vivo and in vitro. The findings from the network pharmacology analysis showed that 51 active components of SF were effective against HIRI and 257 potential intersecting target points. Further screening identified five key targets: AKT1, IL-1β, TNF, STAT3, and PTGS2. KEGG pathway analysis enriched for signaling pathways such as JAK2/STAT3. Additionally, the outcomes of molecular docking revealed a significant binding affinity among the four primary active components of SF and their respective targets. Western blotting results indicated that SF could inhibit the activation of the JAK2/STAT3 pathway in HIRI mice and OGD/R-induced AML12 cells. CONCLUSION Through network pharmacology, molecular docking and in vivo and in vitro experiments, it was preliminarily demonstrated that SF attenuates HIRI through the induction of ferroptosis via JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Peng Huang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China.
| | - Xuecheng Yu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China.
| | - Zengxiang Gao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China.
| | - Qingsong Yang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China.
| | - Yunya Lin
- Hubei Shizhen Labortatory, Wuhan, People's Republic of China.
| | - Jiyuan Tu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China; Hubei Shizhen Labortatory, Wuhan, People's Republic of China.
| | - Yan Cao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China; Hubei Shizhen Labortatory, Wuhan, People's Republic of China.
| | - Yanju Liu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China; Hubei Shizhen Labortatory, Wuhan, People's Republic of China.
| | - Wenlong Su
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China; Hubei Shizhen Labortatory, Wuhan, People's Republic of China.
| | - Guosheng Cao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China; Hubei Shizhen Labortatory, Wuhan, People's Republic of China; Center of Traditional Chinese Medicine Modernization for Liver Diseases, Hubei University of Chinese Medicine, Wuhan, People's Republic of China.
| |
Collapse
|
33
|
Fernández-Cabero JJ, Lasa-Teja C, San Segundo D, Comins-Boo A, Irure-Ventura J, Walias Rivera D, Martín-Varillas JL, Mata C, Santos M, Aurrecoechea E, Blanco R, López-Hoyos M. Changes in NK Cells and Exhausted Th Cell Phenotype in RA Patients Treated with Janus Kinase Inhibitors: Implications for Adverse Effects. Int J Mol Sci 2025; 26:5160. [PMID: 40507968 PMCID: PMC12155405 DOI: 10.3390/ijms26115160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/19/2025] [Accepted: 05/21/2025] [Indexed: 06/16/2025] Open
Abstract
Recent concerns regarding the safety of Janus kinase inhibitors (JAKis) have prompted investigation into their impact on immune cell subsets in rheumatoid arthritis (RA) patients. This study aims to analyse alterations in immune cell populations induced by JAKis that may contribute to adverse events, such as infections or malignancies. This study included 78 RA patients meeting ACR/EULAR criteria with an established treatment with JAKis (tofacitinib, baricitinib, upadacitinib, or filgotinib), 20 healthy donors, and 20 RA patients treated with biological disease-modifying antirheumatic drugs (bDMARDs). Peripheral blood mononuclear cells (PBMCs) were immunophenotyped directly after isolation using multiparametric flow cytometry to characterise innate and adaptive immune-cell subsets. JAKi-treated patients showed a significant reduction in cytotoxic NK Dim (CD3-CD56+CD16+) cells and in the percentage of NK Dim cells expressing the activation marker Nkp30. In CD4+ T cells, the percentage of Th17 (CD3+CD4+CD45RA+CCR6+CXCR3-), Th1-17 (CD3+CD4+CD45RA+CCR6+CXCR3+), and central memory (CM, CD3+CD4+CD45RA+CD62L+) cells was lower in the JAKi group, while effector memory (EM, CD3+CD4+CD45RA-CD62L-) and terminally differentiated CD45RA (TEMRA, CD3+CD4+CD45RA+CD62L-) T helper cells were increased compared to healthy and bDMARD-treated controls. The reduction in NK Dim and Th1-17 cells and the increase in exhausted Th subsets suggest a potential compromise in antiviral immunity and balanced immune responses in JAKi-treated RA patients. These alterations may contribute to an increased risk of infections or malignancies.
Collapse
Affiliation(s)
- Juan José Fernández-Cabero
- Immunopathology Group, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain; (J.J.F.-C.); (C.L.-T.); (D.S.S.); (A.C.-B.); (J.I.-V.); (R.B.)
| | - Carmen Lasa-Teja
- Immunopathology Group, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain; (J.J.F.-C.); (C.L.-T.); (D.S.S.); (A.C.-B.); (J.I.-V.); (R.B.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - David San Segundo
- Immunopathology Group, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain; (J.J.F.-C.); (C.L.-T.); (D.S.S.); (A.C.-B.); (J.I.-V.); (R.B.)
- Department of Immunology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - Alejandra Comins-Boo
- Immunopathology Group, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain; (J.J.F.-C.); (C.L.-T.); (D.S.S.); (A.C.-B.); (J.I.-V.); (R.B.)
- Department of Immunology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - Juan Irure-Ventura
- Immunopathology Group, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain; (J.J.F.-C.); (C.L.-T.); (D.S.S.); (A.C.-B.); (J.I.-V.); (R.B.)
- Department of Immunology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - David Walias Rivera
- Cantabria Tissue and Blood Bank, Fundación Marqués de Valdecilla, 39008 Santander, Spain
| | - Jose Luis Martín-Varillas
- Immunopathology Group, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain; (J.J.F.-C.); (C.L.-T.); (D.S.S.); (A.C.-B.); (J.I.-V.); (R.B.)
- Department of Rheumatology, Hospital de Laredo, 39770 Laredo, Spain
| | - Cristina Mata
- Immunopathology Group, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain; (J.J.F.-C.); (C.L.-T.); (D.S.S.); (A.C.-B.); (J.I.-V.); (R.B.)
- Department of Rheumatology, Hospital de Laredo, 39770 Laredo, Spain
| | - Montserrat Santos
- Immunopathology Group, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain; (J.J.F.-C.); (C.L.-T.); (D.S.S.); (A.C.-B.); (J.I.-V.); (R.B.)
- Department of Rheumatology, Hospital Sierrallana, 39300 Torrelavega, Spain
| | - Elena Aurrecoechea
- Immunopathology Group, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain; (J.J.F.-C.); (C.L.-T.); (D.S.S.); (A.C.-B.); (J.I.-V.); (R.B.)
- Department of Rheumatology, Hospital Sierrallana, 39300 Torrelavega, Spain
| | - Ricardo Blanco
- Immunopathology Group, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain; (J.J.F.-C.); (C.L.-T.); (D.S.S.); (A.C.-B.); (J.I.-V.); (R.B.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - Marcos López-Hoyos
- Immunopathology Group, Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain; (J.J.F.-C.); (C.L.-T.); (D.S.S.); (A.C.-B.); (J.I.-V.); (R.B.)
- Department of Immunology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
- Department of Molecular Biology, University of Cantabria, 39005 Santander, Spain
| |
Collapse
|
34
|
Wang P, Liu J, Zhang M, Yang J, Lian P, Cheng X, Qin J. Radiation Exposure Induced Blood-Brain Barrier Injury via Mitochondria-Mediated Sterile Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e02356. [PMID: 40433769 DOI: 10.1002/advs.202502356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 05/07/2025] [Indexed: 05/29/2025]
Abstract
Radiation-induced brain injury (RIBI) is caused by exposure to high doses of ionizing radiation and characterized by severe cognitive dysfunction and brain necrosis. However, the pathogenesis of RIBI is not fully understood, and no effective intervention is available. This work describes a blood-brain barrier (BBB) microphysiological system (MPS), that allowed to explore the responses of BBB and distinct brain cells to radiation exposure. Following acute exposure to radiation of X-ray or γ-ray, characteristic RIBI-associated pathological responses are observed, including BBB compromise, DNA breaks, inhibited cell proliferation, cell hypertrophy, and proinflammatory cytokine release. Among the distinctive types of cells, brain endothelial cells show the highest radiosensitivity as compared to other cells in the MPS. Intriguingly, X-ray and γ-ray radiation consistently induce prominent sterile inflammation responses, especially type I interferon response, in the BBB MPS. These responses are mediated by radiation-induced mitochondrial DNA release and subsequent activation of cGAS-STING signaling pathway. Furthermore, it is found abrocitinib (JAK1 inhibitor) and idebenone (mitochondrial protectant) can attenuate radiation-induced inflammation and ameliorate injuries in the BBB MPS. These findings reveal the involvement of mitochondria-mediated sterile inflammation in RIBI pathogenesis, identifying mitochondria as a potential target for new radioprotective measures.
Collapse
Affiliation(s)
- Peng Wang
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
| | - Jiayue Liu
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
| | - Min Zhang
- Disvision of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P. R. China
| | - Juan Yang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, P. R. China
| | - Peihan Lian
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
| | - Xiu Cheng
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
| | - Jianhua Qin
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
- Disvision of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P. R. China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100000, China
| |
Collapse
|
35
|
Sun Q, Wang H, Xie J, Wang L, Mu J, Li J, Ren Y, Lai L. Computer-Aided Drug Discovery for Undruggable Targets. Chem Rev 2025. [PMID: 40423592 DOI: 10.1021/acs.chemrev.4c00969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Undruggable targets are those of therapeutical significance but challenging for conventional drug design approaches. Such targets often exhibit unique features, including highly dynamic structures, a lack of well-defined ligand-binding pockets, the presence of highly conserved active sites, and functional modulation by protein-protein interactions. Recent advances in computational simulations and artificial intelligence have revolutionized the drug design landscape, giving rise to innovative strategies for overcoming these obstacles. In this review, we highlight the latest progress in computational approaches for drug design against undruggable targets, present several successful case studies, and discuss remaining challenges and future directions. Special emphasis is placed on four primary target categories: intrinsically disordered proteins, protein allosteric regulation, protein-protein interactions, and protein degradation, along with discussion of emerging target types. We also examine how AI-driven methodologies have transformed the field, from applications in protein-ligand complex structure prediction and virtual screening to de novo ligand generation for undruggable targets. Integration of computational methods with experimental techniques is expected to bring further breakthroughs to overcome the hurdles of undruggable targets. As the field continues to evolve, these advancements hold great promise to expand the druggable space, offering new therapeutic opportunities for previously untreatable diseases.
Collapse
Affiliation(s)
- Qi Sun
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan 610213, China
| | - Hanping Wang
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Juan Xie
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Liying Wang
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Junxi Mu
- Peking-Tsinghua Center for Life Science, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Junren Li
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yuhao Ren
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Luhua Lai
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Science, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan 610213, China
- Research Unit of Drug Design Method, Chinese Academy of Medical Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
36
|
Lv K, Gong Z, Fu Y, Zhao S, Song Y, Wang H, Lin Z. A novel loss-of-function variant in STAT1 causes Mendelian susceptibility to mycobacterial disease. Front Cell Infect Microbiol 2025; 15:1595389. [PMID: 40491431 PMCID: PMC12146302 DOI: 10.3389/fcimb.2025.1595389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 05/05/2025] [Indexed: 06/11/2025] Open
Abstract
Introduction Mendelian Susceptibility to mycobacterial disease (MSMD) is a rare inherited immunodeficiency disorder characterized by increased susceptibility to atypical mycobacterial infections induced by defective IFN-γ pathway. Methods We report three patients from a family presenting with multiple osteolytic lesions and cutaneous granulomas due to Mycobacterium marinum infections. Functional studies, including Western blotting and immunofluorescence, assessed phosphorylation and nuclear translocation of the mutant STAT1-Ile707Thr in eukaryotic overexpression systems. A luciferase reporter assay evaluated its transcriptional activity. Additionally, structural analysis using AlphaFold3 predicted the variant's functional impact. Results A novel STAT1 variant (c.2120T>C, p.Ile707Thr) was identified. The STAT1-Ile707Thr mutant exhibited reduced phosphorylation and impaired nuclear translocation compared to wild-type STAT1. The luciferase assay confirmed decreased transcriptional activity. AlphaFold3-based cluster analysis supported a loss-of-function effect of the mutant. Discussion This study expands the spectrum of STAT1 variants and microbial pathogens associated with MSMD.
Collapse
Affiliation(s)
- Kunlun Lv
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Zhuoqing Gong
- Dermatology Hospital, Southern Medical University, Guangzhou, China
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Yiting Fu
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Sisi Zhao
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Yinggai Song
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Huijun Wang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Zhimiao Lin
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
37
|
Tao Y, Shen L, Wang P. Advances and perspectives on isolation, structural characterization, structure-property relationships, and pharmacological mechanisms of tumor immune glucans: A review. Int J Biol Macromol 2025:144621. [PMID: 40414394 DOI: 10.1016/j.ijbiomac.2025.144621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 05/14/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Tumor immune glucans (TIGs) are emerging as promising anticancer agents capable of enhancing immune responses and mitigating the side effects of chemotherapy. This review analyzes 59 TIGs published between 2015 and 2025, focusing on their isolation, structural characterization, structure-activity relationships, and pharmacological mechanisms. Literature searches were conducted in databases such as Web of Science, PubMed, Google Scholar, Embase, Cochrane Library, ClinicalTrials.gov and CNKI using keywords including "glucans", "tumor immunity", and "polysaccharide structure," covering studies in both English and Chinese. The review highlights the relationship between the structural features of TIGs and their antitumor activities.1,3-linked β-glucans surpassed 1,4-linked or 1,6-linked configurations in antitumor immunostimulatory activity. Elevated molecular weight enhances the structural robustness of TIGs chains, facilitating their selective recognition by cellular receptors and amplifying their anti-tumor immune responses. TIGs exert antitumor effects through mechanisms such as immunomodulation, inhibition of tumor immune escape, and remodeling of the tumor microenvironment, involving signaling pathways such as TLRs/NF-κB, JAK/STAT, MAPK, Bax/Bcl-2, PI3K/Akt, and VEGF/VEGFR. Despite their promising therapeutic potential, challenges remain, including incomplete structural characterization, insufficient IC50 value data, and a lack of reference drugs. Many studies focus on cytokine levels and cellular activity, but deeper molecular mechanistic insights are needed. Further research into the mechanisms of action and targets of TIGs may pave the way for new cancer treatment strategies.
Collapse
Affiliation(s)
- Yi Tao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, PR China; Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products, PR China.
| | - Lisha Shen
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, PR China; Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products, PR China
| | - Ping Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, PR China; Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products, PR China.
| |
Collapse
|
38
|
Tuz AA, Ghosh S, Karsch L, Antler M, Lakovic V, Lohmann S, Lehmann AH, Beer A, Nagel D, Jung M, Hörenbaum N, Kaygusuz V, Qefalia A, Alshaar B, Amookazemi N, Bolsega S, Basic M, Siveke JT, Heiles S, Grüneboom A, Lueong S, Herz J, Sickmann A, Hagemann N, Hasenberg A, Hermann DM, Gunzer M, Singh V. Gut microbiota deficiency reduces neutrophil activation and is protective after ischemic stroke. J Neuroinflammation 2025; 22:137. [PMID: 40410847 PMCID: PMC12100894 DOI: 10.1186/s12974-025-03448-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 04/15/2025] [Indexed: 05/25/2025] Open
Abstract
Neutrophils are readily activated immune cells after ischemic stroke in mice and patients. Still, the impact of gut microbiota on neutrophil activation and its influence on inflammatory brain injury remain undefined. We report that natural microbiota colonization of germ-free (GF) mice induces substantial neutrophil activation and deteriorates stroke pathology. The colonized Ex-GF stroke mice had considerably larger infarct sizes and higher sensorimotor deficits than GF littermates. Furthermore, employing an antibiotic-based mouse model of microbiota deficiency, we demonstrate that gut microbiota depletion induces a juvenile neutrophil phenotype characterized by the upregulation of resting state surface receptors, reduced inflammatory proteins, and levels of circulating NETs. This disarming of neutrophil responses was associated with decreased expression of brain inflammatory genes, vascular thrombus formation, reduced infarct size, and alleviated behavioral deficits. We conclude that gut microbes strongly influence neutrophil activation after stroke and thus directly contribute to stroke severity.
Collapse
Affiliation(s)
- Ali A Tuz
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
- Leibniz-Institut Für Analytische Wissenschaften - ISAS - E.V., 44139, Dortmund, Germany
| | - Susmita Ghosh
- Leibniz-Institut Für Analytische Wissenschaften - ISAS - E.V., 44139, Dortmund, Germany
| | - Laura Karsch
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Medina Antler
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Vivian Lakovic
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Sabrina Lohmann
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Amber Hope Lehmann
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Alexander Beer
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Dennis Nagel
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Marcel Jung
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Nils Hörenbaum
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Viola Kaygusuz
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Altea Qefalia
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Belal Alshaar
- Leibniz-Institut Für Analytische Wissenschaften - ISAS - E.V., 44139, Dortmund, Germany
| | - Niloufar Amookazemi
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Silvia Bolsega
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, 30625, Hannover, Lower-Saxony, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, 30625, Hannover, Lower-Saxony, Germany
| | - Jens T Siveke
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, Partner Site Essen), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sven Heiles
- Leibniz-Institut Für Analytische Wissenschaften - ISAS - E.V., 44139, Dortmund, Germany
- Lipidomics, Faculty of Chemistry, University of Duisburg-Essen, 45141, Essen, Germany
| | - Anika Grüneboom
- Leibniz-Institut Für Analytische Wissenschaften - ISAS - E.V., 44139, Dortmund, Germany
| | - Smiths Lueong
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, Partner Site Essen), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Josephine Herz
- Department of Pediatrics I, Neonatology and Experimental Perinatal Neurosciences, and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Albert Sickmann
- Leibniz-Institut Für Analytische Wissenschaften - ISAS - E.V., 44139, Dortmund, Germany
- Medical Faculty of the Ruhr-University Bochum, Universitätsstraße 150, Bochum, 44801, Germany
| | - Nina Hagemann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Anja Hasenberg
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany.
- Leibniz-Institut Für Analytische Wissenschaften - ISAS - E.V., 44139, Dortmund, Germany.
| | - Vikramjeet Singh
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany.
| |
Collapse
|
39
|
Kiriyama Y, Tokumaru H, Sadamoto H, Nochi H. Biological Actions of Bile Acids via Cell Surface Receptors. Int J Mol Sci 2025; 26:5004. [PMID: 40507815 PMCID: PMC12154436 DOI: 10.3390/ijms26115004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/16/2025] [Accepted: 05/21/2025] [Indexed: 06/16/2025] Open
Abstract
Bile acids (BAs) are synthesized in the liver from cholesterol and are subsequently conjugated with glycine and taurine. In the intestine, bile acids undergo various modifications, such as deconjugation, dehydrogenation, oxidation, and epimerization by the gut microbiota. These bile acids are absorbed in the intestine and transported to the liver as well as the systemic circulation. BAs can activate many types of receptors, including nuclear receptors and cell surface receptors. By activating these receptors, BAs can exert various effects on the metabolic, immune, and nervous systems. Recently, the detailed structure of TGR5, the major plasma membrane receptor for BAs, was elucidated, revealing a putative second BA binding site along with the orthosteric binding site. Furthermore, BAs act as ligands for bitter taste receptors and the Leukemia inhibitory factor receptor. In addition, the Mas-related, G-protein-coupled receptor X4 interacts with receptor activity-modifying proteins. Thus, a variety of cell surface receptors are associated with BAs, and BAs are thought to have very complex activities. This review focuses on recent advances regarding cell surface receptors for bile acids and the biological actions they mediate.
Collapse
Affiliation(s)
- Yoshimitsu Kiriyama
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Takamatsu 760-8542, Japan; (H.T.); (H.S.); (H.N.)
- Institute of Neuroscience, Tokushima Bunri University, Takamatsu 760-8542, Japan
| | - Hiroshi Tokumaru
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Takamatsu 760-8542, Japan; (H.T.); (H.S.); (H.N.)
| | - Hisayo Sadamoto
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Takamatsu 760-8542, Japan; (H.T.); (H.S.); (H.N.)
| | - Hiromi Nochi
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Takamatsu 760-8542, Japan; (H.T.); (H.S.); (H.N.)
| |
Collapse
|
40
|
Wang S, Cheng W, Wang X, Wu Z, Su J. Progress of microneedle targeted modulation technology in the reconstruction of immune microenvironment in diabetic wounds. Eur J Med Res 2025; 30:405. [PMID: 40394697 PMCID: PMC12090542 DOI: 10.1186/s40001-025-02667-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025] Open
Abstract
Wound healing in diabetic patients is mainly hindered by a combination of long-term glycosylation, persistent inflammatory response, and immunosuppressive state. The interaction of these factors not only results in considerable prolongation of the wound healing process but also elevates the likelihood of recurrent ulcer development, profoundly affecting patients' quality of life. Traditional treatments, including surgical debridement, anti-infection, dressing application, vascular intervention, and glycaemic control, can only relieve some symptoms. However, they are often ineffective in addressing the underlying cause of impaired wound healing. It is of concern that the importance of the immune microenvironment in diabetic wound healing has not yet been fully appreciated and investigated, and the homeostasis of the immune microenvironment is crucial for promoting cell proliferation, angiogenesis, and tissue repair. However, this microenvironment is often dysregulated in the diabetic state. This paper reviews the key factors leading to dysregulation of the immune microenvironment, including immune cell dysfunction, abnormal cytokine expression, and disruption of key signalling pathways, and introduces an innovative silicone-based microneedle drug delivery method, which takes advantage of microneedle's precise targeting and highly efficient drug loading capacity to deliver drugs with immunomodulatory functions directly to the wound in a sustained manner, activate the corresponding signalling pathways, promote the polarization of M1 macrophages into the M2 phenotype, and stimulate neovascularization, providing a low inflammatory and pro-angiogenic immune microenvironment for diabetic wound healing, which provides a new therapeutic idea and means for diabetic wound healing.
Collapse
Affiliation(s)
- Shunsheng Wang
- Department of Burn and Plastic Surgery, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, 215000, China
| | - Wei Cheng
- Jiangsu Key Laboratory of Thin Films, School of Physical Science and Technology, Soochow University, Suzhou, 215006, China
| | - Xue Wang
- Department of Burn and Plastic Surgery, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, 215000, China
| | - Zhuofan Wu
- Department of Burn and Plastic Surgery, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, 215000, China
| | - Jiandong Su
- Department of Burn and Plastic Surgery, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, 215000, China.
| |
Collapse
|
41
|
Yasir M, Choe J, Hassan M, Kloczkowski A, Chun W. Recent advances and future perspectives in small molecule JAK2 inhibitors. Future Med Chem 2025:1-17. [PMID: 40392133 DOI: 10.1080/17568919.2025.2507564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Accepted: 05/08/2025] [Indexed: 05/22/2025] Open
Abstract
The Janus kinase (JAK)/Signal Transducer and Activator of Transcription (STAT) signaling pathway is essential for controlling immune function, blood cell formation, and cell growth. Dysregulation of this pathway is implicated in various diseases, including hematologic malignancies, autoimmune disorders, and chronic inflammatory conditions. This review provides a comprehensive overview of the structural and functional aspects of JAK/STAT signaling, with a particular focus on the role of JAK2. This manuscript explores the primary regulators of the JAK/STAT pathway, such as Suppressors Of Cytokine Signaling (SOCS), Protein Inhibitors of Activated STATs (PIAS), and Protein Tyrosine Phosphatases (PTPs), which play a crucial role in maintaining cellular balance and stability. Additionally, the therapeutic landscape of JAK2 inhibitors is explored, covering both approved and investigational drugs, including their mechanisms of action, efficacy, and safety profiles. Emerging strategies such as drug repositioning using computational approaches and experimental validation are also highlighted. By integrating insights from molecular docking studies, machine learning models, and kinase assays, this review emphasizes the potential of JAK2 inhibitors in disease management.
Collapse
Affiliation(s)
- Muhammad Yasir
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - Jongseon Choe
- Department of Microbiology and Immunology, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - Mubashir Hassan
- The Steve and Cindy Rasmussen Institute for Genomic Medicine at Nationwide Children's Hospital, Columbus, OH, USA
| | - Andrzej Kloczkowski
- The Steve and Cindy Rasmussen Institute for Genomic Medicine at Nationwide Children's Hospital, Columbus, OH, USA
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| |
Collapse
|
42
|
Bellocchi C, Favalli EG, Maioli G, Agape E, Rossato M, Paini M, Severino A, Vigone B, Biggioggero M, Trombetta E, Caporali R, Beretta L. Whole-Blood RNA Sequencing Profiling of Patients With Rheumatoid Arthritis Treated With Tofacitinib. ACR Open Rheumatol 2025. [PMID: 40388487 DOI: 10.1002/acr2.11761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 05/21/2025] Open
Abstract
OBJECTIVE Patients with rheumatoid arthritis (RA) often fail to respond to therapies, including JAK inhibitors (JAKi), and treatment allocation is made via a trial-and-error strategy. A comprehensive analysis of responses to JAKi, including tofacitinib, by RNA sequencing (RNAseq) would allow the discovery of transcriptomic markers with a two-fold meaning: (1) an improved knowledge about the mechanisms of response to treatment (inference modeling) and (2) the definition of features that may be useful in treatment optimization and assignment (predictive modeling). METHODS Thirty-three patients with active RA were treated with a tofacitinib dose of 5 mg twice a day for 24 weeks and evaluated for EULAR Disease Activity Score in 28 joints using the C-reactive protein level response. Whole-blood RNA was collected before and after treatment to perform RNAseq transcriptome analysis. Linear models were used to determine differentially expressed genes (DEGs) (1) at baseline according to clinical responses and (2) in the pre-post comparison after tofacitinib treatment and in relation to EULAR responses. The capability of DEGs to predict a successful treatment was tested via machine learning modeling after extensive internal validation. RESULTS Of 26 patients who completed the study (per-protocol analysis), 15 (57.7%) achieved good responses, and 7 (26.9%) and 4 (15.3%) had moderate and no responses, respectively. Overall, 273 baseline genes were significantly associated with the attainment of good responses, contributing to several pathways linked to the immune system or RA pathogenesis (eg, citrullination processes and the negative regulation of natural killer function). The expression of several molecules was reverted by tofacitinib when good responses were reached, including AKT3, GK5, KLF12, FCRL3, BIRC3, TSPOAP1, and P2RY10. Finally, we isolated 14 markers that singularly were capable of predicting the attainment of good responses, including, NKG2D, CD226, CLEC2D, and CD52. CONCLUSION Whole-blood transcriptome analysis of patients with RA treated with tofacitinib identified genes whose expression may be relevant in prognostication and understanding the mechanisms of responses to therapy.
Collapse
Affiliation(s)
- Chiara Bellocchi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico and University of Milan, Milan, Italy
| | | | - Gabriella Maioli
- University of Milan and ASST PiniCTO - Presidio Gaetano Pini, Milan, Italy
| | | | | | | | - Adriana Severino
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Barbara Vigone
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Elena Trombetta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Roberto Caporali
- University of Milan and ASST PiniCTO - Presidio Gaetano Pini, Milan, Italy
| | - Lorenzo Beretta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
43
|
Dimitrion PM, Krevh R, Veenstra J, Ge J, Siddiqui A, Ferguson D, Hans A, Zuniga B, Sidhu K, Daveluy S, Hamzavi I, Zhou L, Adrianto I, Mi QS. High-throughput proteomics identifies inflammatory proteins associated with disease severity and genetic ancestry in patients with hidradenitis suppurativa. Br J Dermatol 2025; 192:1063-1071. [PMID: 39778115 DOI: 10.1093/bjd/ljaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/21/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Hidradenitis suppurativa (HS) is a chronic inflammatory skin condition with a greater prevalence and disease burden in patients who identify as African American and those with a family history of HS, suggesting a strong genetic component to its pathogenesis. OBJECTIVES To evaluate the relationship between plasma inflammatory protein expression, HS disease severity and genetic ancestry in a diverse cohort of patients with HS. METHODS We performed a case-control, single-centre study of patients with HS and age-, sex- and ethnicity-matched healthy control participants. We profiled circulating inflammatory proteins using Olink® high-throughput proteomics and determined genetic ancestry from whole-genome sequencing data. RESULTS Using linear regression, we identified novel proteins associated with HS, after adjusting for age, sex and ethnicity. Our analysis also revealed differences in the inflammatory proteome linked to disease severity. Specifically, we found that plasma levels of interleukin (IL)-6 can distinguish between different Hurley stages, indicating that IL-6 may serve as a marker of disease severity. Additionally, we found variations in inflammatory protein levels based on genetic ancestry: patients with predominantly African ancestry exhibited higher levels of inflammatory proteins associated with neutrophilic inflammation, while those with predominantly European ancestry showed increased levels of T helper 1-related inflammatory proteins. CONCLUSIONS Although we were unable to account for treatment status or comorbidities that may influence the level of inflammatory cytokines, genetic ancestry and disease severity may influence the plasma inflammatory profile in patients with HS.
Collapse
Affiliation(s)
- Peter M Dimitrion
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA
- Cancer Biology Graduate Program, School of Medicine, Wayne State University, Detroit, MI, USA
- Department of Dermatology, Henry Ford Health, Detroit, MI, USA
| | - Rachel Krevh
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA
- Department of Dermatology, Henry Ford Health, Detroit, MI, USA
| | - Jesse Veenstra
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA
- Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Center for Bioinformatics, Department of Public Health Sciences, Henry Ford Health, Detroit, MI, USA
| | - James Ge
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA
- Department of Dermatology, Henry Ford Health, Detroit, MI, USA
| | - Aamir Siddiqui
- Department of Plastic Surgery, Henry Ford Health, Detroit, MI, USA
| | | | - Aakash Hans
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA
- Department of Dermatology, Henry Ford Health, Detroit, MI, USA
| | - Bobby Zuniga
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA
- Department of Dermatology, Henry Ford Health, Detroit, MI, USA
| | - Kermanjot Sidhu
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA
- Department of Dermatology, Henry Ford Health, Detroit, MI, USA
| | - Steven Daveluy
- Department of Dermatology, Wayne State University, Detroit, MI, USA
| | - Iltefat Hamzavi
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Department of Dermatology, Henry Ford Health, Detroit, MI, USA
| | - Li Zhou
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA
- Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Henry Ford Health + Michigan State University Health Sciences, Detroit, MI, USA
| | - Indra Adrianto
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA
- Center for Bioinformatics, Department of Public Health Sciences, Henry Ford Health, Detroit, MI, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Henry Ford Health + Michigan State University Health Sciences, Detroit, MI, USA
| | - Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA
- Cancer Biology Graduate Program, School of Medicine, Wayne State University, Detroit, MI, USA
- Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Henry Ford Health + Michigan State University Health Sciences, Detroit, MI, USA
| |
Collapse
|
44
|
Han JY, Rhee WJ, Shin JS. Cytoplasmic HMGB1 promotes the activation of JAK2-STAT3 signaling and PD-L1 expression in breast cancer. Mol Med 2025; 31:197. [PMID: 40389855 PMCID: PMC12090602 DOI: 10.1186/s10020-025-01235-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 04/28/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND High-mobility group box 1 (HMGB1) plays various roles depending on its subcellular localization. Extracellular HMGB1 interacts with receptors, such as toll-like receptor 4 and receptor for advanced glycation end products (RAGE), promoting cell proliferation, survival, and migration in cancer cells. It also increases the expression of programmed death-ligand 1 (PD-L1) in cancer cells by binding to RAGE. However, the effect of intracellular HMGB1 on the regulation of immune checkpoints such as PD-L1 has not been well characterized. In this study, we aimed to investigate the effects of intracellular HMGB1 on PD-L1 expression in breast cancer cells. METHODS Human and mouse triple-negative breast cancer cells, MDA-MB-231 and 4T1, along with HMGB1-deficient mouse embryonic fibroblast cells, were cultured. HMGB1 overexpression was achieved using a Myc-tagged plasmid, while siHMGB1 constructs were used for gene silencing. Quantitative reverse-transcriptase PCR and western blot analysis were performed to assess gene and protein expressions. Confocal imaging, immunoprecipitation, and proximity ligation assays were used to investigate HMGB1 localization and Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) interactions. In vivo experiments were performed using tumor-bearing mice treated with STAT3 and HMGB1 inhibitors. Statistical analyses were performed using Student's t-tests, one-way analysis of variance, Pearson's correlation, and Kaplan-Meier survival analysis, with significance set at p < 0.05. RESULTS In breast cancer cells, HMGB1 translocation from the nucleus to the cytoplasm increased the JAK2-STAT3 interaction and induced STAT3 phosphorylation, leading to increased STAT3 target signaling, including the epithelial-mesenchymal transition (EMT) phenotype and PD-L1 expression. Inhibition of nucleo-cytoplasmic translocation of HMGB1 decreased STAT3 phosphorylation and PD-L1 expression. Furthermore, HMGB1 enhanced breast cancer cell migration, invasion, and EMT, contributing to tumor growth in an in vivo mouse model that were mitigated by the HMGB1-targeted approach. CONCLUSIONS These findings underscore the critical role of intracellular HMGB1 in modulating PD-L1 expression via the JAK2-STAT3 signaling pathways in breast cancer and suggest that targeting HMGB1 translocation is a promising strategy for breast cancer treatment.
Collapse
Affiliation(s)
- Ju-Young Han
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, South Korea
- Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Woo Joong Rhee
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, South Korea.
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| | - Jeon-Soo Shin
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, South Korea.
- Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea.
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
45
|
Arunachalam AK, Gilmour CK, Melenhorst JJ. Optimizing Chimeric Antigen Receptor T-Cell Therapy for Mantle Cell Lymphoma. J Clin Oncol 2025:JCO2500568. [PMID: 40388673 DOI: 10.1200/jco-25-00568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 04/11/2025] [Indexed: 05/21/2025] Open
Affiliation(s)
- Arun Kumar Arunachalam
- Cell Therapy & Immuno-Engineering Program, Cancer Sciences & Translational Oncology, Cleveland Clinic Research, Cleveland Clinic, Cleveland, OH
| | - Cassandra K Gilmour
- Cell Therapy & Immuno-Engineering Program, Cancer Sciences & Translational Oncology, Cleveland Clinic Research, Cleveland Clinic, Cleveland, OH
| | - Jan Joseph Melenhorst
- Cell Therapy & Immuno-Engineering Program, Cancer Sciences & Translational Oncology, Cleveland Clinic Research, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
46
|
Yamagiwa H, Hashimoto R, Arakane K, Murakami K, Soeda S, Oyama M, Zhu Y, Okada M, Shimodaira H. Predicting drug-gene relations via analogy tasks with word embeddings. Sci Rep 2025; 15:17240. [PMID: 40383732 PMCID: PMC12086191 DOI: 10.1038/s41598-025-01418-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 05/06/2025] [Indexed: 05/20/2025] Open
Abstract
Natural language processing is utilized in a wide range of fields, where words in text are typically transformed into feature vectors called embeddings. BioConceptVec is a specific example of embeddings tailored for biology, trained on approximately 30 million PubMed abstracts using models such as skip-gram. Generally, word embeddings are known to solve analogy tasks through simple vector arithmetic. For example, subtracting the vector for man from that of king and then adding the vector for woman yields a point that lies closer to queen in the embedding space. In this study, we demonstrate that BioConceptVec embeddings, along with our own embeddings trained on PubMed abstracts, contain information about drug-gene relations and can predict target genes from a given drug through analogy computations. We also show that categorizing drugs and genes using biological pathways improves performance. Furthermore, we illustrate that vectors derived from known relations in the past can predict unknown future relations in datasets divided by year. Despite the simplicity of implementing analogy tasks as vector additions, our approach demonstrated performance comparable to that of large language models such as GPT-4 in predicting drug-gene relations.
Collapse
Affiliation(s)
| | | | - Kiwamu Arakane
- Institute for Protein Research, Osaka University, Osaka, Japan
| | - Ken Murakami
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Shou Soeda
- Institute for Protein Research, Osaka University, Osaka, Japan
| | - Momose Oyama
- Kyoto University, Kyoto, Japan
- RIKEN, Tokyo, Japan
| | | | - Mariko Okada
- Institute for Protein Research, Osaka University, Osaka, Japan
| | | |
Collapse
|
47
|
Halvorsen S, Thomas M, Mino-Kenudson M, Kinowaki Y, Burke KE, Morgan D, Miller KC, Williams KM, Gurung J, McGoldrick J, Hopton M, Hoppe B, Samanta N, Martin S, Tirard A, Arnold BY, Tantivit J, Yarze J, Staller K, Chung DC, Villani AC, Sassi S, Khalili H. Single-cell transcriptomic characterization of microscopic colitis. Nat Commun 2025; 16:4618. [PMID: 40383833 PMCID: PMC12086216 DOI: 10.1038/s41467-025-59648-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 04/30/2025] [Indexed: 05/20/2025] Open
Abstract
Microscopic colitis (MC) is a chronic inflammatory disease of the large intestine and a common cause of chronic diarrhea in older adults. Here, we use single-cell RNA sequencing analysis of colonic mucosal tissue to build a cellular and molecular model for MC. Our results show that in MC, there is a substantial expansion of tissue CD8+ T cells, likely arising from local expansion following T cell receptor engagement. Within the T cell compartment, MC is characterized by a shift in CD8 tissue-resident memory T cells towards a highly cytotoxic and inflammatory phenotype and expansion of CD4+ T regulatory cells. These results provide insight into inflammatory cytokines shaping MC pathogenesis and highlight notable similarities and differences with other immune-mediated intestinal diseases, including a common upregulation of IL26 and an MC-specific upregulation of IL10. These data help identify targets against enteric T cell subsets as an effective strategy for treatment of MC.
Collapse
Affiliation(s)
- Stefan Halvorsen
- Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
| | - Molly Thomas
- Center for Immunology and Inflammatory Diseases, Department of Medicine, MGH, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Harvard Medical School (HMS), Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, MGH, Boston, MA, USA
| | - Mari Mino-Kenudson
- Harvard Medical School (HMS), Boston, MA, USA
- Department of Pathology, HMS, MGH, Boston, MA, USA
| | | | - Kristin E Burke
- Division of Gastroenterology, Department of Medicine, MGH, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, MGH, Boston, MA, USA
| | - David Morgan
- Clinical and Translational Epidemiology Unit, MGH, Boston, MA, USA
| | - Kaia C Miller
- Clinical and Translational Epidemiology Unit, MGH, Boston, MA, USA
- Department of Medicine, Duke University Health System, NC, Durham, USA
| | | | - Jenny Gurung
- Clinical and Translational Epidemiology Unit, MGH, Boston, MA, USA
| | | | - Megan Hopton
- Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
| | - Brooke Hoppe
- Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
| | - Nandini Samanta
- Center for Immunology and Inflammatory Diseases, Department of Medicine, MGH, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Sidney Martin
- Center for Immunology and Inflammatory Diseases, Department of Medicine, MGH, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Alice Tirard
- Center for Immunology and Inflammatory Diseases, Department of Medicine, MGH, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Benjamin Y Arnold
- Center for Immunology and Inflammatory Diseases, Department of Medicine, MGH, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Jessica Tantivit
- Center for Immunology and Inflammatory Diseases, Department of Medicine, MGH, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Joseph Yarze
- Division of Gastroenterology, Department of Medicine, MGH, Boston, MA, USA
| | - Kyle Staller
- Division of Gastroenterology, Department of Medicine, MGH, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, MGH, Boston, MA, USA
| | - Daniel C Chung
- Division of Gastroenterology, Department of Medicine, MGH, Boston, MA, USA
| | - Alexandra-Chloé Villani
- Center for Immunology and Inflammatory Diseases, Department of Medicine, MGH, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Harvard Medical School (HMS), Boston, MA, USA
| | - Slim Sassi
- Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Harvard Medical School (HMS), Boston, MA, USA
- Department of Orthopedic Surgery, MGH, Boston, MA, USA
| | - Hamed Khalili
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA.
- Division of Gastroenterology, Department of Medicine, MGH, Boston, MA, USA.
- Clinical and Translational Epidemiology Unit, MGH, Boston, MA, USA.
- Institute of Environmental Medicine, Nutrition Epidemiology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
48
|
Choi Y, Lee S, Kim HJ, Park T, Kwack WG, Yang S, Chung EK. State-of-the-Art Evidence for Clinical Outcomes and Therapeutic Implications of Janus Kinase Inhibitors in Moderate-to-Severe Ulcerative Colitis: A Narrative Review. Pharmaceuticals (Basel) 2025; 18:740. [PMID: 40430558 PMCID: PMC12114625 DOI: 10.3390/ph18050740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2025] [Revised: 05/07/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by relapsing inflammation and incomplete response to conventional therapies. Although biologics have advanced UC management, many patients with moderate-to-severe disease experience treatment failure, relapse, or adverse effects. This review evaluates the pharmacology, efficacy, and safety of oral Janus kinase (JAK) inhibitors-tofacitinib, upadacitinib, and filgotinib-to guide their clinical use in UC. Methods: A comprehensive literature review was conducted using the PubMed, Embase, Cochrane, and Web of Science databases to identify relevant studies on JAK inhibitors in UC. The review included Phase 3 randomized controlled trials (RCTs), real-world observational studies, and recent network meta-analyses. We assessed pharmacologic profiles, clinical efficacy, and safety data for tofacitinib, upadacitinib, and filgotinib. Additionally, we reviewed emerging pipeline agents and future directions in oral immunomodulatory therapy for UC. Results: All three agents demonstrated efficacy in the induction and maintenance of remission. Upadacitinib showed superior performance, including rapid symptom control, high clinical remission rates, and favorable long-term outcomes in both biologic-naïve and -experienced patients. Tofacitinib offered strong efficacy, particularly in early response, but was associated with higher risks of herpes zoster and thromboembolic events. Filgotinib provided moderate efficacy with a favorable safety profile, making it suitable for risk-averse populations. Meta-analyses consistently ranked upadacitinib highest in clinical efficacy and onset of action. Conclusions: JAK inhibitors offer effective and convenient oral treatment options for moderate-to-severe UC. Upadacitinib emerges as a high-efficacy agent; tofacitinib and filgotinib remain valuable based on patient-specific risk profiles. Future studies are needed to clarify optimal sequencing, long-term safety, and the role of emerging agents or combination therapies.
Collapse
Affiliation(s)
- Yunseok Choi
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.C.); (S.L.); (H.J.K.); (T.P.)
| | - Suhyun Lee
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.C.); (S.L.); (H.J.K.); (T.P.)
- Department of Pharmacy, College of Pharmacy, Woosuk University, Wanju 55338, Republic of Korea
| | - Hyeon Ji Kim
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.C.); (S.L.); (H.J.K.); (T.P.)
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Institute of Regulatory Innovation through Science, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Taemin Park
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.C.); (S.L.); (H.J.K.); (T.P.)
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Institute of Regulatory Innovation through Science, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Won Gun Kwack
- Division of Pulmonary, Allergy and Critical Care Medicine, Kyung Hee University Hospital, Seoul 02447, Republic of Korea;
| | - Seungwon Yang
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.C.); (S.L.); (H.J.K.); (T.P.)
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Institute of Regulatory Innovation through Science, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Eun Kyoung Chung
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.C.); (S.L.); (H.J.K.); (T.P.)
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Pharmacy, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
- Kyung Hee East-West Pharmaceutical Research Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
49
|
Elsisi WI, George RF, Syam YM, Abd-Ellatef GEF, Abd El-Karim SS. Recent achievements in molecular insights, anticancer activities, and comparative structure activity relationships of thiazolidin-4-one derivatives as EGFR inhibitors (2019-present). Bioorg Med Chem 2025; 128:118244. [PMID: 40513224 DOI: 10.1016/j.bmc.2025.118244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 05/01/2025] [Accepted: 05/16/2025] [Indexed: 06/16/2025]
Abstract
The EGFR family is considered one of the most important receptor tyrosine kinases. The family consists of four main members: EGFR, HER2, ErbB3, and ErbB4. They have a crucial role in cancer progression and growth, especially in solid tumors such as lung, breast, colon, neck, and brain tumors. Since the first member, EGFR, is highly incorporated in several pathways regarding cancer, we provide a comparative understanding of all FDA-approved drugs targeting EGFR, demonstrating their structures' developments to provide a good illustration of possible EGFR binding interactions. From the recently approved drugs, we selected our nucleus for study, which is the thiazolidin-4-one nucleus. The review aims to provide an overview of all recent EGFR TK inhibitors bearing this nucleus with additional demonstrations of their structure-activity relationships gathering all those SARs into a general one. A comprehensive understanding of SARs of recent thiazolidin-4-one derivatives may inspire others to develop new, potent, and selective drugs targeting EGFR.
Collapse
Affiliation(s)
- Wessam I Elsisi
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Center, 33 El-Bohouth Street, Dokki, Cairo, P.O Box 12622, Egypt.
| | - Riham F George
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Yasmin M Syam
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Center, 33 El-Bohouth Street, Dokki, Cairo, P.O Box 12622, Egypt
| | - Gamal E F Abd-Ellatef
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Center, 33 El-Bohouth Street, Dokki, Cairo, P.O Box 12622, Egypt
| | - Somaia S Abd El-Karim
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Center, 33 El-Bohouth Street, Dokki, Cairo, P.O Box 12622, Egypt.
| |
Collapse
|
50
|
Zuo H, Liao A, Shen S, Wang Y, Yang X, Weng S, He J, Xu X. microRNA-34 mediates a negative feedback loop in the JAK-STAT pathway to attenuate immune overactivation in an invertebrate. Cell Commun Signal 2025; 23:228. [PMID: 40382609 PMCID: PMC12085071 DOI: 10.1186/s12964-025-02230-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 05/04/2025] [Indexed: 05/20/2025] Open
Abstract
The JAK-STAT pathway is an essential signaling mechanism that initiates immune responses against pathogen infections. The intrinsic homeostatic regulation of JAK-STAT signaling is critical for maintaining immune homeostasis. Previous studies have shown that aberrant activation of the invertebrate JAK-STAT pathway leads to inflammation-like symptoms. Understanding the homeostatic mechanisms of this pathway in invertebrates is of significant interest. Pacific white shrimp (Penaeus vannamei) is one of the most extensively studied invertebrates in immune system research. In this study, we demonstrate that the shrimp microRNA-34 (miR-34) attenuates JAK-STAT pathway activation by targeting JAK, thereby inhibiting STAT phosphorylation and nuclear translocation. Interestingly, miR-34 expression is directly regulated by STAT, forming a negative feedback regulatory loop in the JAK-STAT pathway. Disrupting this loop results in excessive JAK-STAT pathway activation and immune overactivation, exacerbating inflammation caused by Vibrio parahaemolyticus infection in shrimp. This study provides new insights into the regulatory mechanism of the JAK-STAT pathway and its roles in maintaining immune homeostasis in invertebrates.
Collapse
Affiliation(s)
- Hongliang Zuo
- State Key Laboratory of Biocontrol, School of Life Sciences/ Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
| | - Alian Liao
- State Key Laboratory of Biocontrol, School of Life Sciences/ Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
| | - Sihui Shen
- State Key Laboratory of Biocontrol, School of Life Sciences/ Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
| | - Youxi Wang
- State Key Laboratory of Biocontrol, School of Life Sciences/ Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
| | - Xiya Yang
- State Key Laboratory of Biocontrol, School of Life Sciences/ Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
| | - Shaoping Weng
- State Key Laboratory of Biocontrol, School of Life Sciences/ Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
| | - Jianguo He
- State Key Laboratory of Biocontrol, School of Life Sciences/ Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China.
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China.
| | - Xiaopeng Xu
- State Key Laboratory of Biocontrol, School of Life Sciences/ Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/ China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China.
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China.
| |
Collapse
|