51
|
Alemu BK, Tommasi S, Hulin JA, Meyers J, Mangoni AA. Current knowledge on the mechanisms underpinning vasculogenic mimicry in triple negative breast cancer and the emerging role of nitric oxide. Biomed Pharmacother 2025; 186:118013. [PMID: 40147105 DOI: 10.1016/j.biopha.2025.118013] [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: 12/16/2024] [Revised: 03/13/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025] Open
Abstract
Vasculogenic mimicry (VM) is the process by which cancer cells form vascular-like channels to support their growth and dissemination. These channels lack endothelial cells and are instead lined by the tumour cells themselves. VM was first reported in uveal melanomas but has since been associated with other aggressive solid tumours, such as triple-negative breast cancer (TNBC). In TNBC patients, VM is associated with tumour aggressiveness, drug resistance, metastatic burden, and poor prognosis. The lack of effective targeted therapies for TNBC has stimulated research on the mechanisms underpinning VM in order to identify novel druggable targets. In recent years, studies have highlighted the role of nitric oxide (NO), the NO synthesis inhibitor, asymmetric dimethylarginine (ADMA), and dimethylarginine dimethylaminohydrolase 1 (DDAH1), the key enzyme responsible for ADMA metabolism, in regulating VM. Specifically, NO inhibition through downregulation of DDAH1 and consequent accumulation of ADMA appears to be a promising strategy to suppress VM in TNBC. This review discusses the current knowledge regarding the molecular pathways underpinning VM in TNBC, anti-VM therapies under investigation, and the emerging role of NO regulation in VM.
Collapse
Affiliation(s)
- Belete Kassa Alemu
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia; Injibara University, College of Medicine and Health Sciences, Department of Pharmacy, Injibara, Ethiopia
| | - Sara Tommasi
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia; Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia
| | - Julie-Ann Hulin
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Jai Meyers
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Arduino A Mangoni
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia; Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia.
| |
Collapse
|
52
|
Liu P, Fu M, Liu D, Chao T, Zhang J. Mechanisms of Radiation-induced Brain Injury in Mice Based on Bioinformatics Analysis. Radiat Res 2025; 203:321-332. [PMID: 40133766 DOI: 10.1667/rade-24-00204.1] [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: 08/16/2024] [Accepted: 03/04/2025] [Indexed: 03/27/2025]
Abstract
Radiation therapy is a crucial adjunct treatment for head and neck tumors, as well as primary or metastatic brain tumors. Radiation-induced brain injury is one of the most severe complications, postirradiation, in patients with head and neck tumors, and significantly impacts their quality of life. Currently, there are no effective treatments for radiation-induced brain injury, making the study of radiation-induced molecular mechanisms and the identification of early damage biomarkers critical for the early diagnosis and treatment of such injuries. In this study, twelve male C57 mice aged 6-8 weeks were randomly divided into a control group, a 15 Gy irradiation group, and a 30 Gy irradiation group. Mice were exposed to 6 MV X rays. The control group underwent the same anesthesia procedure as the irradiated groups but did not receive radiation. General health and weight changes were monitored and recorded. Four months postirradiation, mice were subjected to intracranial magnetic resonance imaging [T2-weighted imaging (T2WI)], open field test (OFT), novel object recognition (NOR), followed by a collection of brain tissues for immunofluorescence, SA-β-gal staining, and transcriptomic and metabolomic analyses. Compared to the control group, the 15 Gy and 30 Gy irradiated mice showed reduced activity and weight loss. The irradiated mice exhibited impaired recognition memory in the NOR test and decreased body weight, but radiation had no significant effect on weight or performance in the OFT. Electron microscopy reveals significant demyelination of mouse cortex after irradiation, and MRI T2-weighted imaging demonstrated varying degrees of brain atrophy and ventricular enlargement in irradiated mice compared to the control group. Immunofluorescence staining showed a significant increase in astrocytes and microglia activated after irradiation. SA-β-gal staining revealed significant increases in the numbers of β-gal+ cells in irradiated mice compared to those in untreated control mice. Bioinformatics analysis identified enriched pathways primarily related to lipid metabolism and neuroinflammatory responses; associated metabolites and genes were variously upregulated or downregulated. The findings suggest that radiation-induced brain injury involves complex biological processes, with lipid metabolism disorders and neuroinflammation being the predominant pathological changes observed. Further studies on these metabolic pathways and genes could enhance our understanding of the pathogenic mechanisms underlying radiation-induced brain injury and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Peiquan Liu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Fu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Liu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tengfei Chao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaxuan Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
53
|
Gong Y, Wu M, Huang Y, He X, Yuan J, Dang B. Research developments in the neurovascular unit and the blood‑brain barrier (Review). Biomed Rep 2025; 22:88. [PMID: 40166412 PMCID: PMC11956146 DOI: 10.3892/br.2025.1966] [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: 11/18/2024] [Accepted: 03/05/2025] [Indexed: 04/02/2025] Open
Abstract
The neurovascular unit (NVU) is composed of neurons, glial cells, brain microvascular endothelial cells (BMECs), pericytes, and the extracellular matrix. The NVU controls the permeability of the blood-brain barrier (BBB) and protects the brain from harmful blood-borne and endogenous and exogenous substances. Among these, neurons transmit signals, astrocytes provide nutrients, microglia regulate inflammation, and BMECs and pericytes strengthen barrier tightness and coverage. These cells, due to their physical structure, anatomical location, or physiological function, maintain the microenvironment required for normal brain function. In this review, the BBB structure and mechanisms are examined to obtain a better understanding of the factors that influence BBB permeability. The findings may aid in safeguarding the BBB and provide potential therapeutic targets for drugs affecting the central nervous system.
Collapse
Affiliation(s)
- Yating Gong
- Department of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215600, P.R. China
| | - Muyao Wu
- Department of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215600, P.R. China
| | - Yaqian Huang
- Department of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215600, P.R. China
| | - Xiaoyi He
- Department of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215600, P.R. China
| | - Jiaqi Yuan
- Department of Neurosurgery, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215600, P.R. China
| | - Baoqi Dang
- Department of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215600, P.R. China
| |
Collapse
|
54
|
Engelmann C. Peripheral fat mobilization and mitochondrial fat metabolism: Fueling the energy demands of liver regeneration. J Hepatol 2025; 82:942-944. [PMID: 40016070 DOI: 10.1016/j.jhep.2025.01.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 03/01/2025]
Affiliation(s)
- Cornelius Engelmann
- Charité - Universitaetsmedizin Berlin; Campus Virchow Klinikum; Department of Hepatology and Gastroenterology, Berlin, Germany; University College London, Institute of Liver and Digestive Health, Royal Free Campus, London, United Kingdom
| |
Collapse
|
55
|
Lee SG, Kim Y, Park SW, Kim MW, Oh JS, Park S, Lee S, Lee YH, Jeong Y, Park JH, Lee M, Shin H, Kim S, Bae YM, Kim CY, Chung HM. Evaluation of phthalates induced cardiotoxicity using human iPSCs-derived cardiomyocyte and dual-cardiotoxicity evaluation methods. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 296:118196. [PMID: 40262243 DOI: 10.1016/j.ecoenv.2025.118196] [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: 11/12/2024] [Revised: 03/26/2025] [Accepted: 04/12/2025] [Indexed: 04/24/2025]
Abstract
Phthalates, known as plasticizers, are endocrine disruptor, and their risks are being highlighted as their use increases worldwide. Di-2-ethylhexyl phthalate (DEHP), the most prevalent of the phthalates, is known to be toxic to humans, and it has recently been reported to be linked to cardiotoxicity. Although many other phthalates are also widely used, data on their cardiotoxic effects are yet to be well established. In this study, we assessed the cardiotoxic potential of various phthalates using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and a microelectrode array-based dual-cardiotoxicity evaluation method previously reported. Cytotoxicity results showed that acute exposure to DEHP, dibutyl phthalate (DBP), benzyl butyl phthalate (BBP), and di-n-octyl phthalate (DnOP) did not affect the viability of hiPSC-CMs. Before examining the functional changes in hiPSC-CMs caused by exposure to these four phthalates, we present changes in field potential (FP) and contractility based on the blocking of major ions for reference. Contrary to concerns, FP results showed a dramatic decrease in spike amplitude, beat period, and FP duration (FPD) at high doses of DBP and BBP rather than DEHP. Interestingly, DnOP resulted in a prolonged FPD, unlike the others. Furthermore, contractility results indicated that, unlike DEHP and DnOP, high doses of DBP and BBP caused beating arrest along with decreased beat amplitude. Overall, this study demonstrated that phthalates other than DEHP can also induce cardiotoxicity, even with acute exposure. It is expected that the application of the established evaluation method will facilitate the development of safe alternatives.
Collapse
Affiliation(s)
- Seul-Gi Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Yoonseo Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Sang Woong Park
- Department of Emergency Medical Services, Eulji University, 553 Sanseong-daero, Seongnam-si, Gyeonggi-do 13135, South Korea
| | - Min Woo Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Jeong-Seop Oh
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, 1, Gwanak-Ro, Gwanak-Gu, Seoul 08826, South Korea
| | - Shinhye Park
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Suemin Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Yun Hyeong Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Youngin Jeong
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Jeong Hwan Park
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Myeonghee Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Hyewon Shin
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Seeun Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea
| | - Young Min Bae
- Department of Physiology, Institute of Functional Genomics and Research Institute of Medical Science, Konkuk University School of Medicine, 268 Chungwon-daero, Chungju 27478, South Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, South Korea.
| | - Hyung Min Chung
- Department of Stem Cell Biology, Konkuk University School of Medicine, 268 Chungwon-daero, Chungju 27478, South Korea.
| |
Collapse
|
56
|
Zhang H, Yu Y, Qian C. Oligonucleotide-Based Modulation of Macrophage Polarization: Emerging Strategies in Immunotherapy. Immun Inflamm Dis 2025; 13:e70200. [PMID: 40325939 PMCID: PMC12053320 DOI: 10.1002/iid3.70200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 03/10/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Recent advances in immunotherapy have spotlighted macrophages as central mediators of disease treatment. Their polarization into pro‑inflammatory (M1) or anti‑inflammatory (M2) states critically influences outcomes in cancer, autoimmunity, and chronic inflammation. Oligonucleotides have emerged as highly specific, scalable, and cost‑effective agents for reprogramming macrophage phenotypes. OBJECTIVE To review oligonucleotide strategies-including ASOs, siRNAs, miRNA mimics/inhibitors, and aptamers-for directing macrophage polarization and their therapeutic implications. REVIEW SCOPE We examine key signaling pathways governing M1/M2 phenotypes, describe four classes of oligonucleotides and their mechanisms, and highlight representative preclinical and clinical applications. KEY INSIGHTS Agents such as AZD9150, MRX34, and AS1411 demonstrate macrophage reprogramming in cancer, inflammation, and infection models. Advances in ligand‑conjugated nanoparticles and chemical modifications improve delivery and stability, yet immunogenicity, off‑target effects, and formulation challenges remain significant barriers. FUTURE PERSPECTIVES Optimizing delivery platforms, enhancing molecular stability, and rigorous safety profiling are critical. Integration with emerging modalities-such as engineered CAR‑macrophages-will enable precise, disease‑specific interventions, and advance oligonucleotide‑guided macrophage modulation toward clinical translation.
Collapse
Affiliation(s)
- Hanfu Zhang
- National Key Laboratory of Immunity & Inflammation, Institute of ImmunologyNaval Medical UniversityShanghaiChina
- School of Molecular SciencesUniversity of Western AustraliaCrawleyWAAustralia
| | - Yizhi Yu
- National Key Laboratory of Immunity & Inflammation, Institute of ImmunologyNaval Medical UniversityShanghaiChina
| | - Cheng Qian
- National Key Laboratory of Immunity & Inflammation, Institute of ImmunologyNaval Medical UniversityShanghaiChina
| |
Collapse
|
57
|
Jiang J, Duo K, Zhu S, Wang Y, Xue H, Piao C, Ren Y, Lei X, Zhang Y, Liu J, Yang L, Zhang N. Investigation of the mechanism of Buyang Huanwu decoction in improving learning and memory impairment in Alzheimer's disease mice based on lipidomics. J Nat Med 2025; 79:568-590. [PMID: 40195204 DOI: 10.1007/s11418-025-01890-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 02/26/2025] [Indexed: 04/09/2025]
Abstract
In this study, a lipid disorder Alzheimer's disease (AD) model was developed with high-fat diet and D-galactose injected intraperitoneally (HFD & D-gal) to evaluate the activities of Buyang Huanwu Decoction (BYHWD) compared with donepezil hydrochloride. The learning and memory abilities of BYHWD were evaluated by Morris water maze test (MWM). The lipid levels in serum, histopathology, and immunohistochemistry of hyperphosphorylated tau protein in hippocampal neurons were conducted to prove the therapy effects of BYHWD. After the identification of constituents absorbed into the brain using LC-MS, UPLC-TQ-MS was employed to analyze endogenous lipid metabolites in the hippocampi of mice. Based on the validated differential markers identified through lipidomics analysis, we further substantiated potential therapeutic pathway of BYHWD through the application of molecular docking technology. The mechanism underlying BYHWD was subsequently confirmed by palmitic acid-injured HT22 cells. The results showed that BYHWD significantly improved the cognitive deficits and regulated the lipid levels of HFD & D-gal mice. BYHWD also protected the neuronal cell condition of hippocampal neurons, increased the density of dendritic spines, and reduced the expression of P-tau. Lipidomics revealed that 41 differential lipid metabolites were retuned after BYHWD administration, and this change may be related to the PPARγ pathway. Calycosin-7-glucoside showed good interaction with PPARγ in vivo composition analysis. Calycosin-7-glucoside increased the mRNA expression levels of lipid metabolism-related enzymes and PPARγ, as well as the expression of PPARγ protein in vitro study. BYHWD activated the PPARγ pathway to induce peroxisome proliferation and regulated lipid metabolism disorders in the AD mice brain.
Collapse
Affiliation(s)
- Jing Jiang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Heilongjiang Institute for Drug Control, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Kai Duo
- Heilongjiang Institute for Drug Control, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Siyu Zhu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yitong Wang
- Heilongjiang Institute for Drug Control, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Hui Xue
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Chengyu Piao
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yifan Ren
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xia Lei
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, China
| | - Yafeng Zhang
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, China.
| | - Jianxin Liu
- School of Pharmaceutical Sciences, China-Pakistan, International Science and Technology Innovation Cooperation Base for Ethnic Medicine Development in Hunan Province, Hunan University of Medicine, Huaihua, Hunan, China
| | - Lihong Yang
- Heilongjiang Institute for Drug Control, NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Harbin, Heilongjiang, China.
| | - Ning Zhang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| |
Collapse
|
58
|
Tabtimmai L, Phonchan T, Thongprik N, Kaennakam S, Yodsin N, Choowongkomon K, Sonklin C, Jadsadajerm S, Wisetsai A. New oxepin and dihydrobenzofuran derivatives from Bauhinia saccocalyx roots and their anti-inflammatory, cytotoxic, and antioxidant activities. J Nat Med 2025; 79:543-555. [PMID: 40085400 DOI: 10.1007/s11418-025-01888-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 02/19/2025] [Indexed: 03/16/2025]
Abstract
Four new oxepin and dihydrobenzofuran derivatives, saccoxepins A-C (1-3) and saccobenzofurin A (4), along with one known compound, bauhinoxepin A (5), were isolated from the roots of Bauhinia saccocalyx. The structures were elucidated by extensive analysis of spectroscopic data in combination with ECD analysis. The EtOAc extract exhibited significant NO inhibition (94.4 ± 0.35%, 50 μg/mL), and saccoxepin A and bauhinoxepin A demonstrated strong NO suppression, with IC50 values of 49.35 µM and 30.28 µM, respectively, alongside notable antioxidant activity. Saccoxepin A and bauhinoxepin A selectively reduced interleukin-6 (IL-6) levels, while bauhinoxepin A slightly lowered tumor necrosis factor-alpha (TNF-α) at a low dose. Furthermore, bauhinoxepin A exhibited cytotoxicity against HCT-116 cells, with an IC50 of 8.88 µM. These findings suggest that the roots of B. saccocalyx possess potent antioxidant, anti-inflammatory, and anticancer activities, supporting its traditional medicinal applications and highlighting its potential as a source of therapeutic agents.
Collapse
Affiliation(s)
- Lueacha Tabtimmai
- Department of Biotechnology, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bangkok, 10800, Thailand
- Food and Agro-Industrial Research Center, King Mongkut's University of Technology North Bangkok, Bangkok, 10800, Thailand
| | - Thanyathon Phonchan
- Department of Industrial Chemistry, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bangkok, 10800, Thailand
| | - Natrinee Thongprik
- Department of Industrial Chemistry, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bangkok, 10800, Thailand
| | - Sutin Kaennakam
- Department of Agro-Industrial, Food, and Environmental Technology, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok (KMUTNB), Bangkok, 10800, Thailand
| | - Nuttapon Yodsin
- Department of Chemistry, Faculty of Science, Silpakorn University, Nakhon Pathom, 73000, Thailand
| | - Kiattawee Choowongkomon
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand
| | - Chanikan Sonklin
- Department of Industrial Chemistry, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bangkok, 10800, Thailand
| | - Supachai Jadsadajerm
- Department of Industrial Chemistry, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bangkok, 10800, Thailand
| | - Awat Wisetsai
- Department of Industrial Chemistry, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bangkok, 10800, Thailand.
| |
Collapse
|
59
|
Li Y, Pan AP, Yu AY. Recent Progression of Pathogenesis and Treatment for Diabetic Cataracts. Semin Ophthalmol 2025; 40:275-282. [PMID: 39530428 DOI: 10.1080/08820538.2024.2427789] [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: 05/20/2024] [Revised: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Background: Cataracts are still the main cause of blindness worldwide. The incidence of cataracts in the diabetic population is 3-5 times higher than in healthy people. With the increasing incidence of diabetes and the development of aging, as well as the higher risk of surgical and postoperative complications of diabetic patients undergoing surgery, it is still necessary to study the occurrence and development mechanism of diabetic cataracts as well as potential therapeutic targets and therapeutic drugs.Methods: A retrospective review of the literature from PubMed (2017-2024).Results: We summarized the the current literature on the molecular mechanism and prevention and treatment of diabetic cataracts.Conclusions: The aqueous humor metabolism changes, oxidative stress, reactive oxygen species generation increase, the conversion of polyol pathway, as well as non-coding RNA expression changes play important roles in diabetic cataract and these processes is closely linked with each other. Inhibitors or drugs target to these processes, such as aldose reductase inhibitors, antioxidants, natural flavonoid compounds, as well as nanotechnology-based therapeutic product, have shown promising prospects in the prevention and treatment of diabetic cataract.
Collapse
Affiliation(s)
- Yike Li
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang P. R. China
- National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, P. R. China
| | - An-Peng Pan
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang P. R. China
- National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, P. R. China
| | - A-Yong Yu
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang P. R. China
- National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, P. R. China
| |
Collapse
|
60
|
Ding J, Zhang S, Li Q, Xia B, Wu J, Lu X, Huang C, Yuan X, You Q. Geraniin attenuates isoproterenol-induced cardiac hypertrophy by inhibiting inflammation, oxidative stress and cellular apoptosis. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2025; 29:307-319. [PMID: 39572368 PMCID: PMC12012319 DOI: 10.4196/kjpp.24.200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/08/2024] [Accepted: 10/13/2024] [Indexed: 04/22/2025]
Abstract
Geraniin, a polyphenol derived from the fruit peel of Nephelium lappaceum L., has been shown to possess anti-inflammatory and antioxidant properties in the cardiovascular system. The present study explored whether geraniin could protect against an isoproterenol (ISO)-induced cardiac hypertrophy model. Mice in the ISO group received an intraperitoneal injection of ISO (5 mg/kg) once daily for 9 days, and the administration group were injected with ISO after 5 days of treatment with geraniin or spironolactone. Potential therapeutic effects and related mechanisms analysed by anatomical coefficients, histopathology, blood biochemical indices, reverse transcription-PCR and immunoblotting. Geraniin decreased the cardiac pathologic remodeling and myocardial fibrosis induced by ISO, as evidenced by the modifications to anatomical coefficients, as well as the reduction in collagen I/III á1mRNA and protein expression and cross-sectional area in hypertrophic cardiac tissue. In addition, geraniin treatment reduced ISO-induced increase in the mRNA and protein expression levels of interleukin (IL)-6, IL-1β and tumor necrosis factor-α, whereas ISO-induced IL-10 showed the opposite behaviour in hypertrophic cardiac tissue. Further analysis showed that geraniin partially reversed the ISO-induced increase in malondialdehyde and nitric oxide, and the ISO-induced decrease in glutathione, superoxide dismutase and glutathione. Furthermore, it suppressed the ISO-induced cellular apoptosis of hypertrophic cardiac tissue, as evidenced by the decrease in B-cell lymphoma-2 (Bcl-2)-associated X/caspase-3/caspase-9 expression, increase in Bcl-2 expression, and decrease in TdT-mediated dUTP nick-end labeling-positive cells. These findings suggest that geraniin can attenuate ISO-induced cardiac hypertrophy by inhibiting inflammation, oxidative stress and cellular apoptosis.
Collapse
Affiliation(s)
- Jiaqi Ding
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Shenjie Zhang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Qi Li
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Boyu Xia
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Jingjing Wu
- Department of Cardiology, Suzhou Kowloon Hospital of Shanghai Jiaotong University School of Medicine, Suzhou 215027, Jiangsu, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Xiaomei Yuan
- Department of Cardiology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Qingsheng You
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| |
Collapse
|
61
|
Zhang W, Cai Q, You L, Zhang W, Zheng X, Jiang C, Chen C. Study on the common mechanisms of gastroesophageal reflux disease and interstitial lung disease. Hum Immunol 2025; 86:111300. [PMID: 40209518 DOI: 10.1016/j.humimm.2025.111300] [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: 06/03/2024] [Revised: 03/05/2025] [Accepted: 03/19/2025] [Indexed: 04/12/2025]
Abstract
OBJECTIVE Interstitial lung disease (ILD) and gastroesophageal reflux disease (GERD) have complex interactions and can exacerbate severity of each other. This study aimed to screen shared genes between ILD and GERD and explore their common mechanisms and clinical value. METHODS We obtained microarray datasets of ILD and GERD from public databases. Shared genes were screened by differential expression analysis and Venn analysis. Hub genes were screened from shared genes using the protein-protein interaction (PPI) network analysis. The ssGSEA algorithm was utilized to estimate immune infiltration level of ILD and GERD, and correlation of hub genes with immune cell infiltration was studied. Finally, potential drugs that may act on hub genes were screened using DSigDB. RESULTS 52 shared genes were obtained through Venn analysis. PPI network analysis identified 10 hub genes (BMP4, NT5E, PPARG, EPCAM, DPP4, KLF2, MMP1, AGR2, ADAMTS1, GATA6) that may have diagnostic performance (p < 0.05). The results of immune infiltration showed that hub genes were highly linked to multiple immune cell infiltrations (p < 0.05). In addition, we identified 5 potential drugs. Notably, thioridazine may target 5 hub genes (MMP1, AGR2, KLF2, ADAMTS1, and PPARG) simultaneously (p < 0.05) and had the potential to be a novel therapeutic drug. CONCLUSION In summary, we have screened out the hub genes with diagnostic value in ILD and GERD, and also revealed the close relationship between the hub genes and the disease immune microenvironment, providing new research directions for the common mechanism and interaction of the two diseases.
Collapse
Affiliation(s)
- Wen Zhang
- Ningde Clinical Medical College of Fujian Medical University, Ningde City 352100 Fujian Province, China; Department of Pulmonary and Critical Care Medicine, Ningde Municipal Hospital of Ningde Normal University, Ningde City 352100 Fujian Province, China
| | - Qizhi Cai
- Ningde Clinical Medical College of Fujian Medical University, Ningde City 352100 Fujian Province, China; Department of Gastroenterology Medicine, Ningde Municipal Hospital of Ningde Normal University, Ningde City 352100 Fujian Province, China
| | - Liusheng You
- Ningde Clinical Medical College of Fujian Medical University, Ningde City 352100 Fujian Province, China; Department of Gastroenterology Medicine, Ningde Municipal Hospital of Ningde Normal University, Ningde City 352100 Fujian Province, China
| | - Wei Zhang
- Ningde Clinical Medical College of Fujian Medical University, Ningde City 352100 Fujian Province, China; Department of Gastroenterology Medicine, Ningde Municipal Hospital of Ningde Normal University, Ningde City 352100 Fujian Province, China
| | - Xiujin Zheng
- Ningde Clinical Medical College of Fujian Medical University, Ningde City 352100 Fujian Province, China; Department of Gastroenterology Medicine, Ningde Municipal Hospital of Ningde Normal University, Ningde City 352100 Fujian Province, China
| | - Chenglin Jiang
- Ningde Clinical Medical College of Fujian Medical University, Ningde City 352100 Fujian Province, China; Department of Gastroenterology Medicine, Ningde Municipal Hospital of Ningde Normal University, Ningde City 352100 Fujian Province, China
| | - Changdan Chen
- Ningde Clinical Medical College of Fujian Medical University, Ningde City 352100 Fujian Province, China; Department of Gastroenterology Medicine, Ningde Municipal Hospital of Ningde Normal University, Ningde City 352100 Fujian Province, China.
| |
Collapse
|
62
|
Tulsian K, Thakker D, Vyas VK. Overcoming chimeric antigen receptor-T (CAR-T) resistance with checkpoint inhibitors: Existing methods, challenges, clinical success, and future prospects : A comprehensive review. Int J Biol Macromol 2025; 306:141364. [PMID: 39988153 DOI: 10.1016/j.ijbiomac.2025.141364] [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: 08/09/2024] [Revised: 11/20/2024] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Immune checkpoint blockade is, as of today, the most successful form of cancer immunotherapy, with more than 43 % of cancer patients in the US eligible to receive it; however, only up to 12.5 % of patients respond to it. Similarly, adoptive cell therapy using bioengineered chimeric antigen receptorT (CAR-T) cells and T-cell receptor (TCR) cells has provided excellent responses against liquid tumours, but both forms of immunotherapy have encountered challenges within a tumour microenvironment that is both lacking in tumour-specific T-cells and is strongly immunosuppressive toward externally administered CAR-T and TCR cells. This review focuses on understanding approved checkpoint blockade and adoptive cell therapy at both biological and clinical levels before delving into how and why their combination holds significant promise in overcoming their individual shortcomings. The advent of next-generation checkpoint inhibitors has further strengthened the immune checkpoint field, and a special section explores how these inhibitors can address existing hurdles in combining checkpoint blockade with adoptive cell therapy and homing in on our cancer target for long-term immunity.
Collapse
Affiliation(s)
- Kartik Tulsian
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Dhinal Thakker
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India.
| |
Collapse
|
63
|
Zhong H, Zhou S, Yin S, Qiu Y, Liu B, Yu H. Tumor microenvironment as niche constructed by cancer stem cells: Breaking the ecosystem to combat cancer. J Adv Res 2025; 71:279-296. [PMID: 38866179 DOI: 10.1016/j.jare.2024.06.014] [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: 04/08/2024] [Revised: 05/27/2024] [Accepted: 06/09/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are a distinct subpopulation of cancer cells with the capacity to constantly self-renew and differentiate, and they are the main driver in the progression of cancer resistance and relapse. The tumor microenvironment (TME) constructed by CSCs is the "soil" adapted to tumor growth, helping CSCs evade immune killing, enhance their chemical resistance, and promote cancer progression. AIM OF REVIEW We aim to elaborate the tight connection between CSCs and immunosuppressive components of the TME. We attempt to summarize and provide a therapeutic strategy to eradicate CSCs based on the destruction of the tumor ecological niche. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three main key concepts. First, we highlight that CSCs recruit and transform normal cells to construct the TME, which further provides ecological niche support for CSCs. Second, we describe the main characteristics of the immunosuppressive components of the TME, targeting strategies and summarize the progress of corresponding drugs in clinical trials. Third, we explore the multilevel insights of the TME to serve as an ecological niche for CSCs.
Collapse
Affiliation(s)
- Hao Zhong
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Shiyue Zhou
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Shuangshuang Yin
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin, China.
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.
| | - Haiyang Yu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China.
| |
Collapse
|
64
|
Meanti R, Bresciani E, Rizzi L, Molteni L, Coco S, Omeljaniuk RJ, Torsello A. Cannabinoid Receptor 2 (CB2R) as potential target for the pharmacological treatment of neurodegenerative diseases. Biomed Pharmacother 2025; 186:118044. [PMID: 40209306 DOI: 10.1016/j.biopha.2025.118044] [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: 12/28/2024] [Revised: 03/25/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025] Open
Abstract
The endocannabinoid system (ECS) is a ubiquitous physiological system that plays a crucial role in maintaining CNS homeostasis and regulating its functions. It includes cannabinoid receptors (CBRs), endogenous cannabinoids (eCBs), and the enzymes responsible for their synthesis and degradation. In recent years, growing evidence has highlighted the therapeutic potential of the ECS and CBRs, in a wide range of severe diseases and pathological conditions, including Alzheimer's and Parkinson's diseases, Amyotrophic Lateral Sclerosis, Multiple Sclerosis, Huntington's Disease, HIV-1 associated neurocognitive disorders, neuropathic pain and migraine. Targeting the cannabinoid type 2 receptor (CB2R) has gained attention due to its ability to (i) mitigate neuroinflammatory responses, (ii) regulate mitochondrial function and (iii) provide trophic support, all without eliciting the psychotropic actions associated with CB1R activation. This review aims to explore the potential of CB2R modulation as a strategy for the prevention and treatment of neurologic disorders, exploring both preclinical and clinical findings.
Collapse
Affiliation(s)
- Ramona Meanti
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Laura Molteni
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Silvia Coco
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Robert J Omeljaniuk
- Department of Biology, Lakehead University, 955 Oliver Rd, Thunder Bay, Ontario P7B 5E1, Canada.
| | - Antonio Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| |
Collapse
|
65
|
Yafang H, Chenxi L, Haoran Z, Yuan L, Feifei R, Xia D, Zhiyong C. Study on the Molecular Mechanism of XiaoXianXiong Decoction in the Treatment of Atherosclerosis Based on UHPLC-Q Exactive Focus MS/MS, Network Pharmacology, and Experimental Validation. Biomed Chromatogr 2025; 39:e70062. [PMID: 40107965 DOI: 10.1002/bmc.70062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 02/24/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Atherosclerosis (AS), a leading pathological basis of severe cardiovascular diseases, poses a significant threat to human health. XiaoXianXiong Decoction (XXXD), a classical traditional Chinese medicine (TCM) prescription, has demonstrated promising effects in the treatment of AS. To investigate the underlying mechanism of XXXD in treatment with AS, we used UHPLC-Q Focus MS/MS, network pharmacology and in vivo validation methods. The results showed that 59 chemical components of XXXD were identified. Network pharmacology showed that 11 key compounds, 10 key targets and five key signaling pathways involved in the therapeutic effects of XXXD on AS. Experimental verification confirmed that XXXD significantly improved dyslipidemia, lipid accumulation and pathological changes in the aorta during AS. These effects were linked to the inhibition of the PI3K/AKT signaling pathway, down-regulation of PI3K, HRAS, EGF, CREB and up-regulation of NOS3 expression. This work may provide a theoretical basis for further research on the molecular mechanisms for XXXD in AS treatment.
Collapse
Affiliation(s)
- Hou Yafang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Lu Chenxi
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China
| | - Zhang Haoran
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China
| | - Liu Yuan
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China
| | - Ren Feifei
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China
| | - Du Xia
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chen Zhiyong
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China
| |
Collapse
|
66
|
Chaves MM. Neutrophils and purinergic signaling: Partners in the crime against Leishmania parasites? Biochimie 2025; 232:43-53. [PMID: 39855456 DOI: 10.1016/j.biochi.2025.01.004] [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: 06/30/2024] [Revised: 12/18/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
The parasite of the genus Leishmania is the causative agent of diseases that affect humans called leishmaniasis. These diseases affect millions of people worldwide and the currently existing drugs are either very toxic or the parasites acquire resistance. Therefore, new elimination mechanisms need to be elucidated so that new therapeutic strategies can be developed. Much has already been discussed about the role of neutrophils in Leishmania infection, and their participation is still controversial. A recent study showed that receptors present in the neutrophil membrane, the purinergic receptors, can control the infection when activated, but the triggering mechanism has not been elucidated. In this review, we will address the possible participation of purinergic receptors expressed in the neutrophil extracellular membrane that may be participating in the detection of Leishmania infection and their possible effects during parasitism.
Collapse
Affiliation(s)
- Mariana M Chaves
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Bio-Manguinhos, Oswaldo Cruz Foundation, Brazilian Ministry of Health, Rio de Janeiro, Brazil.
| |
Collapse
|
67
|
Wu H, Song Z, Chen Q, Yan R, Zhao H, Li H. Disrupting reconsolidation by systemic inhibition of Thioredoxin-1 attenuates cocaine and morphine relapse. Biomed Pharmacother 2025; 186:118037. [PMID: 40199134 DOI: 10.1016/j.biopha.2025.118037] [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: 12/31/2024] [Revised: 03/30/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025] Open
Abstract
The enduring nature of drug-associated memories is an essential factor contributing to the relapse. Drug-related cues can activate drug memories, making them enter reconsolidation, during which interventions can effectively disrupt these memories. Interventions targeting memory reconsolidation present a promising therapeutic strategy for addressing substance use disorders (SUDs). Oxidative stress can disrupt neural function and impair memory. Thioredoxin-1 (Trx-1) effectively alleviates oxidative stress and reduces inflammation levels. However, few studies have connected Trx-1 to drug memory or explored its specific role in reconsolidation. This research employed the conditioned place preference (CPP) model to investigate the effects of Trx-1 inhibitors on the reconsolidation of morphine- and cocaine-related memories. Results show that immediate administration of PX-12, a Trx-1 inhibitor, after retrieval significantly attenuated the reinstatement of cocaine and morphine CPP induced by both cues and the drug itself, with the effect lasting for at least 14 days. In contrast, the inhibition of Trx-1, either 6 hours following retrieval or in the absence of retrieval, does not influence drug-seeking behaviors associated with cocaine or morphine. Furthermore, Trx-1 inhibitor itself did not produce any preferences. In summary, our results indicate that Trx-1 activity is crucial for cocaine- and morphine-related memories, and that the Trx-1 inhibitor may serve as a potential treatment for drug abuse.
Collapse
Affiliation(s)
- Hao Wu
- Department of Anesthesiology, Peking Union Medical College Hospital, Peking Union Medical College, and Chinese Academy of Medical Science, Beijing, China
| | - Zhihao Song
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center for Skull Base Surgery and Neurooncology in Hunan Province, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qijun Chen
- Department of Clinical Laboratory, The Affiliated Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou 510620, China
| | - Ruyu Yan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center for Skull Base Surgery and Neurooncology in Hunan Province, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Haiting Zhao
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Haoyu Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center for Skull Base Surgery and Neurooncology in Hunan Province, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
68
|
Thakur MR, Tupe RS. l-Arginine: A multifaceted regulator of diabetic cardiomyopathy. Biochem Biophys Res Commun 2025; 761:151720. [PMID: 40186920 DOI: 10.1016/j.bbrc.2025.151720] [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: 02/14/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
In diabetes mellitus, dysregulated glucose and lipid metabolism lead to diabetic cardiomyopathy (DCM) by imparting pathological myocardial remodeling and cellular injury. Accelerated glycation, oxidative stress, and activated inflammatory pathways culminate in cardiac fibrosis and hypertrophy in DCM. The regulatory effects of l-Arginine (L-Arg) have been elucidated in the pathological changes of DCM, including myocardial fibrosis, hypertrophy, and apoptosis, by inhibiting glycation and oxidative stress-induced inflammation. Disturbed L-Arg metabolism and decreased intracellular L-Arg pool are correlated with the progression of DCM; therefore, L-Arg supplementation has been prescribed for various cardiovascular dysfunctions. This review expands the therapeutic potential of L-Arg supplementation in DCM by elucidating its molecular mechanism of action and exploring potential clinical outcomes.
Collapse
Affiliation(s)
- Muskan R Thakur
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, 412115, Maharashtra, India
| | - Rashmi S Tupe
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, 412115, Maharashtra, India.
| |
Collapse
|
69
|
Gauthier T, Lim YJ, Jin W, Liu N, Patiño LC, Chen W, Warren J, Martin D, Morell RJ, Dveksler G, Su GH, Chen W. Activin A activation of Smad3 mitigates innate inflammation in mouse models of psoriasis and sepsis. J Clin Invest 2025; 135:e187063. [PMID: 40067393 PMCID: PMC12043092 DOI: 10.1172/jci187063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/05/2025] [Indexed: 05/02/2025] Open
Abstract
Phosphorylation of Smad3 is a critical mediator of TGF-β signaling, which plays an important role in regulating innate immune responses. However, whether Smad3 activation can be regulated in innate immune cells in TGF-β-independent contexts remains poorly understood. Here, we show that Smad3 is activated through the phosphorylation of its C-terminal residues (pSmad3C) in murine and human macrophages in response to bacterial and viral ligands, and this activation is mediated by activin A in a TGF-β-independent manner. Specifically, infectious ligands, such as LPS, induced secretion of activin A through the transcription factor STAT5 in macrophages, and activin A signaling in turn activated pSmad3C. This activin A/Smad3 axis controlled mitochondrial ATP production and ATP conversion into adenosine by CD73 in macrophages, enforcing an antiinflammatory mechanism. Consequently, mice with a deletion of activin A receptor 1b specifically in macrophages (Acvr1bfl/fl-Lyz2cre) succumbed more to sepsis as a result of uncontrolled inflammation and exhibited exacerbated skin disease in a mouse model of imiquimod-induced psoriasis. Thus, we have revealed a previously unrecognized natural brake to inflammation in macrophages that occurs through the activation of Smad3 in an activin A-dependent manner.
Collapse
Affiliation(s)
- Thierry Gauthier
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Yun-Ji Lim
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Wenwen Jin
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Na Liu
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Liliana C. Patiño
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Weiwei Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - James Warren
- Department of Pathology, Uniformed Services University, Bethesda, Maryland, USA
| | - Daniel Martin
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Robert J. Morell
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Gabriela Dveksler
- Department of Pathology, Uniformed Services University, Bethesda, Maryland, USA
| | - Gloria H. Su
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - WanJun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
70
|
Tihăuan BM, Onisei T, Slootweg W, Gună D, Iliescu C, Chifiriuc MC. Cannabidiol-A friend or a foe? Eur J Pharm Sci 2025; 208:107036. [PMID: 39929375 DOI: 10.1016/j.ejps.2025.107036] [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: 12/20/2024] [Revised: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 03/23/2025]
Abstract
Cannabidiol (CBD), one of the main actives from Cannabis sativa has been perpetually explored lately for its therapeutic effects. Its main attributes, such as anti-inflammatory and antioxidant effects, snowball into pain management, epilepsy and seizure alleviation, anxiety relief, as well as numerous other implications through the entire metabolism. However, conventional administration routes challenge its therapeutic potential, with reported poor water solubility, hepatic degradation, gastric instability and erratic bioavailability observed in oral administration. As a result, the transdermal delivery systems have emerged as a promising alternative to oral or inhaled routes, offering improved bioavailability and targeted effects. The medical use of CBD throughout Europe, UK, USA or Australia is extensive and usually represented by pharmaceutical preparations recommended after conventional treatment routs fail. The non-medical use is limited by each country's own legislation, a wider range of products being available, but the irregular regulatory landscape coupled with the growing market of cannabinoid-infused products, emphasizes the need for standardized formulations and further clinical research. The present work critically examines the transdermal administration of cannabidiol, explores the skin's potential as a route and the strategies involved in using it for systemic targeting. We highlighted key challenges and provided insights into CBD`s variable bioavailability based on different administration routes and methods, thus compiling a literature-based absorption, distribution, metabolism, and excretion (ADME) study. We also explore the role of the endocannabinoid system, its function in various medical conditions, and the therapeutic effects associated with CBD, particularly in light of the varying legislation across countries. While the breadth of potential benefits is compelling, it is essential to emphasize the ongoing nature of CBD research as individual responses to it can vary significantly.
Collapse
Affiliation(s)
- Bianca-Maria Tihăuan
- Research Institute of the University of Bucharest-ICUB, 91-95 Spl. Independentei, 50567 Bucharest, Romania; National Institute for Research and Development in Food Bioresources, Dinu Vintilă Street, No.6, 021102 Bucharest, Romania; eBio-hub Research Centre, National University for Science and Technology Politehnica Bucharest, Bucharest, Romania; Academy of Romanian Scientists, Bucharest, Romania
| | - Tatiana Onisei
- National Institute for Research and Development in Food Bioresources, Dinu Vintilă Street, No.6, 021102 Bucharest, Romania
| | - Walter Slootweg
- QB3 Research & Development, Spaarndammerstraaat 4d, 1013SV Amsterdam, Netherlands
| | - Daniel Gună
- S.C. Absolute Essential Oils Ltd. (AEO), Adunații Copăceni Village, Giurgiu County, 38 Troitei Street, 087005, Romania
| | - Ciprian Iliescu
- eBio-hub Research Centre, National University for Science and Technology Politehnica Bucharest, Bucharest, Romania; Academy of Romanian Scientists, Bucharest, Romania; National Institute for Microtechnologies, 126A Erou Iancu Nicolae Street, Voluntari 077190, Romania.
| | - Mariana-Carmen Chifiriuc
- Research Institute of the University of Bucharest-ICUB, 91-95 Spl. Independentei, 50567 Bucharest, Romania.
| |
Collapse
|
71
|
Sá-Pessoa J, Calderón-González R, Lee A, Bengoechea JA. Klebsiella pneumoniae emerging anti-immunology paradigms: from stealth to evasion. Trends Microbiol 2025; 33:533-545. [PMID: 39884872 DOI: 10.1016/j.tim.2025.01.003] [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: 12/27/2024] [Accepted: 01/13/2025] [Indexed: 02/01/2025]
Abstract
Klebsiella pneumoniae (KP) is a global threat to human health due to the isolation of multidrug-resistant strains. Despite advancements in understanding KP's population structure, antibiotic resistance mechanisms, and transmission patterns, a gap remains in how KP evades defenses, allowing the pathogen to flourish in tissues despite an activated immune system. KP infection biology has been shaped by the notion that the pathogen has evolved to shield from defenses more than actively suppress them. This review describes new paradigms of how KP exploits the coevolution with the innate immune system to hijack immune effectors and receptors to ablate signaling pathways and to counteract cell-intrinsic immunity, making apparent that KP can no longer be considered only as a stealth pathogen.
Collapse
Affiliation(s)
- Joana Sá-Pessoa
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT7 1NN, UK
| | - Ricardo Calderón-González
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT7 1NN, UK
| | - Alix Lee
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT7 1NN, UK
| | - José A Bengoechea
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT7 1NN, UK.
| |
Collapse
|
72
|
Wu H, Liu J, Zhang XH, Jin S, Li P, Liu H, Zhao L, Wang J, Zhao S, Tian HD, Lai JR, Hao Y, Liu GR, Hou K, Yan M, Liu SL, Pang D. The combination of flaxseed lignans and PD-1/ PD-L1 inhibitor inhibits breast cancer growth via modulating gut microbiome and host immunity. Drug Resist Updat 2025; 80:101222. [PMID: 40048957 DOI: 10.1016/j.drup.2025.101222] [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: 12/29/2024] [Revised: 02/22/2025] [Accepted: 02/22/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Patients with breast cancer (BC) who benefit from the PD-1/PD-L1 inhibitor (PDi) is limited, necessitating novel strategies to improve immunotherapy efficacy of BC. Here we aimed to investigate the inhibitory effects of flaxseed lignans (FL) on the biological behaviors of BC and evaluate the roles of FL in enhancing the anticancer effects of PDi. METHODS HPLC was used to detect the content of enterolactone (ENL), the bacterial transformation product of FL. Transcript sequencing was performed and identified CD38 as a downstream target gene of ENL. CD38-overexpressing cells were constructed and cell proliferation, colony formation, wound healing and transwell assays were used to assess the function of ENL/CD38 axis on BC cells in vitro. Multiplexed immunohistochemistry (mIHC) and CyTOF were used to detect the changes of the tumor immune microenvironment (TIM). 16S rDNA sequencing was used to explore the changes of gut microbiota in mice. A series of in vivo experiments were conducted to investigate the anticancer effects and mechanisms of FL and PDi. RESULTS FL was converted to ENL by gut microbiota and FL administration inhibited the progression of BC. ENL inhibited the malignant behaviors of BC by downregulating CD38, a key gene associated with immunosuppression and PD-1/PD-L1 blockade resistance. The mIHC assay revealed that FL administration enhanced CD3+, CD4+ and CD8+ cells and reduced F4/80+ cells in TIM. CyTOF confirmed the regulatory effects of FL and FL in combination with PDi (FLcPDi) on TIM. In addition, 16S rDNA analysis demonstrated that FLcPDi treatment significantly elevated the abundance of Akkermansia and, importantly, Akkermansia administration enhanced the response to PDi in mice treated with antibiotics. CONCLUSIONS The FL/ENL/CD38 axis inhibited BC progression. FL enhanced the anticancer effects of PDi by modulating gut microbiota and host immunity.
Collapse
Affiliation(s)
- Hao Wu
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Jiena Liu
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xing-Hua Zhang
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Shengye Jin
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ping Li
- The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Huidi Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Liuying Zhao
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jianyu Wang
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shilu Zhao
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hong-Da Tian
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Jin-Ru Lai
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Yi Hao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Gui-Rong Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Kaijian Hou
- School of Public Health, Shantou University, Shantou, China; Longhu People's Hospital, Shantou, China.
| | - Meisi Yan
- Department of Pathology, Harbin Medical University, Harbin, China.
| | - Shu-Lin Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China; Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
| | - Da Pang
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
73
|
Kim M, Naik SD, Kim SW, Joung M, Yum YA, Aswar VR, Jeong LS. Design, synthesis, and biological evaluation of 5'-deoxy (N)-methanocarbanucleoside derivatives as A 3 adenosine receptor ligands. Bioorg Med Chem Lett 2025; 120:130134. [PMID: 39947351 DOI: 10.1016/j.bmcl.2025.130134] [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: 12/13/2024] [Revised: 01/20/2025] [Accepted: 02/08/2025] [Indexed: 02/18/2025]
Abstract
Based on the potent and selective antagonism exhibited by truncated North (N)-methanocarba adenosine analogs, we synthesized a series of 5'-deoxy (N)-methanocarba nucleosides to explore their structure-activity relationships (SAR). The stereoselective synthesis of the North cyclopropyl-fused alcohol was achieved from d-ribose using ring-closing metathesis, oxidative rearrangement, and cyclopropanation as key steps. Mitsunobu reactions were employed to condense nucleobases with glycosyl donors, followed by N6 functionalization with various amines. Despite their innovative design, all synthesized analogs exhibited lower binding affinity compared to the 4'-thio series and fully truncated (N)-methanocarba adenosine. Docking studies revealed that the 4'-methyl group of the rigid North conformational sugar introduces steric clashes, which likely contribute to the reduced affinity. These findings underscore the critical role of sugar conformation and steric effects in receptor interactions, providing valuable insights for the development of potent and selective A3AR ligands.
Collapse
Affiliation(s)
- Minjae Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Siddhi D Naik
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea; Government College of Arts Science and Commerce, Khandola, Marcela, Goa 403107, India
| | - Seung Woo Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Misuk Joung
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Yun A Yum
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Vikas R Aswar
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Lak Shin Jeong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea; Future Medicine Co., Ltd, 54 Changup-ro, Sujeong-gu, Seongnam, Gyeonggi-do 13449, Republic of Korea.
| |
Collapse
|
74
|
Liang J, Wu H, Song Z, Li G, Zhang J, Ding W. Machine learning‑based construction of damage‑associated molecular patterns related score identifies subtypes of pancreatic adenocarcinoma with distinct prognosis. Oncol Lett 2025; 29:246. [PMID: 40177138 PMCID: PMC11962577 DOI: 10.3892/ol.2025.14992] [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: 12/16/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
The present study aimed to assess the prognostic significance of Damage-Associated Molecular Pattern (DAMP)-related gene expression in pancreatic adenocarcinoma (PAAD) and to develop a scoring system based on these genes. Consensus clustering was performed on patients with PAAD using data from The Cancer Genome Atlas (TCGA) and Meta-cohort datasets, identifying three distinct clusters: C1 (pro-DAMP), C2 (intermediate) and C3 (anti-DAMP). Differential gene expression analysis between clusters C1 and C3 identified 141 significant genes. Least Absolute Shrinkage and Selection Operator Cox regression was utilized to derive an optimal predictor set, leading to the identification of six hub genes associated with the DAMP status, which were then employed to calculate the DAMPscore. Weighted Gene Co-expression Network Analysis revealed a strong correlation between these eight hub genes and the DAMPscore. The functionality of these hub genes in PAAD was validated using a Cell Counting Kit-8 assay and Transwell assays. The results indicated that patients with PAAD with elevated DAMPscores exhibited significantly reduced survival times. Receiver operating characteristic (ROC) curve analysis indicated that the DAMPscore has robust prognostic capabilities. In the Meta-cohort, the area under the ROC curve (AUC) values for the DAMPscore to predict overall survival at 1, 3 and 5 years were 0.65, 0.70 and 0.77, respectively, while the AUC values for the TCGA-PAAD cohort were 0.71, 0.73 and 0.72, respectively. Additional cohorts, such as E-MTAB-6134 and ICGC-AU, corroborated the predictive power of the DAMPscore. A comparison of the DAMPscore with other prognostic models revealed that it consistently exhibited a superior C-index across most PAAD cohorts. Furthermore, in vitro experiments demonstrated that PLEK2, a hub gene related to the DAMPscore, is involved in critical biological processes such as cell proliferation, migration and invasion. In conclusion, the DAMPscore is a promising prognostic biomarker for PAAD, surpassing traditional models in various datasets. This study emphasizes the role of DAMP-related pathways in influencing tumor biology and highlights the importance of immune modulation in PAAD prognosis, suggesting that therapeutic strategies targeting DAMP signaling could improve patient outcomes.
Collapse
Affiliation(s)
- Jing Liang
- Department of Oncology, Xiangxi Autonomous Prefecture People's Hospital, Ji Shou University, Jishou, Hunan 416000, P.R. China
| | - Hui Wu
- Department of Oncology, Xiangxi Autonomous Prefecture People's Hospital, Ji Shou University, Jishou, Hunan 416000, P.R. China
| | - Zewen Song
- Department of Oncology, Xiangxi Autonomous Prefecture People's Hospital, Ji Shou University, Jishou, Hunan 416000, P.R. China
| | - Guoyin Li
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan 466001, P.R. China
| | - Jianfeng Zhang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan 410000, P.R. China
| | - Wenxin Ding
- Department of Oncology, Xiangxi Autonomous Prefecture People's Hospital, Ji Shou University, Jishou, Hunan 416000, P.R. China
| |
Collapse
|
75
|
Yousef DA, Abdalla MS, Elshopakey GE, Al-Olayan E, Abdel Moneim AE, Ramadan SS. Diosmin-loaded chitosan nanoparticles mitigate doxorubicin-evoked cardiotoxicity in rats by featuring oxidative imbalance mechanism, NF-κB, and Bcl-2/Bax pathways. Int J Biol Macromol 2025; 305:140991. [PMID: 39952491 DOI: 10.1016/j.ijbiomac.2025.140991] [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: 08/05/2024] [Revised: 01/12/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Cardiotoxicity is doxorubicin's primary side effect. Its cardiac toxicity has been attributed to the generation of free radicals. The present work was designed to understand the potential underlying pathways behind the cardioprotective action of diosmin (Dio) and Dio-loaded chitosan nanoparticles (DCNPs) against doxorubicin (Dox)-mediated cardiotoxicity. Male rats were allocated into five groups: control, Dio (100 mg/kg), Dox (12 mg/kg), Dio + Dox (100 mg/kg + 12 mg/kg), and DCNPs+Dox (100 mg/kg DCNPs/orally+12 mg/kg Dox/IP). Notably, in response to Dox, a significant increase of cardiac biomarkers with a decrease in Na+/K+-ATPase activity was detected. The cardiac inflammatory and pro-apoptotic protein levels were elevated with decreased cardiac interleukin-10 and Bcl-2 levels when the rats were subjected to Dox. Also, the cardiac expression of the fibrotic marker MMP-9 was increased. Moreover, Dox raised malondialdehyde and nitric oxide levels, accompanied by minimizing antioxidant status. Also, Dox-treated rats showed cardiac histopathological impairment compared to the control. The oral administration of Dio or DCNPs enhanced the activity of antioxidant enzymes and diminished inflammatory cytokines and apoptotic markers in the Dox-exposed rats. In summary, these findings indicate that DCNPs exhibit significant cardioprotective effectiveness against Dox-mediated toxicity by suppressing various mechanisms, such as redox status, the NF-κB pathway, and apoptosis.
Collapse
Affiliation(s)
- Doaa A Yousef
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo 11795, Egypt
| | - Mohga S Abdalla
- Biochemistry Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo 11795, Egypt
| | - Gehad E Elshopakey
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, 35516, Egypt; Department of Veterinary Diseases, Faculty of Veterinary Medicine, Delta University for Science and Technology, 35712 Gamasa, Egypt
| | - Ebtesam Al-Olayan
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Ahmed E Abdel Moneim
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo 11795, Egypt; Al-Ayen Scientific Research Center, Al-Ayen Iraqi University, AUIQ, P.O. Box: 64004, An Nasiriyah, Thi Qar, Iraq.
| | - Shimaa S Ramadan
- Biochemistry Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo 11795, Egypt
| |
Collapse
|
76
|
Liu Y, Xue R. Pancreatic stellate cell: Update on molecular investigations and clinical translation in pancreatic cancer. Int J Cancer 2025; 156:1672-1685. [PMID: 39825771 DOI: 10.1002/ijc.35326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025]
Abstract
Pancreatic cancer is a particularly aggressive tumor, distinguished by the presence of a prominent collagenous stroma and desmoplasia that envelops the tumor cells. Pancreatic stellate cell (PSC) contributes to the formation of a dense fibrotic stroma and has been demonstrated to facilitate tumor progression. As the significance of PSCs is increasingly revealed, more explorations are focused on the complex molecular mechanisms and tumor-stromal crosstalk in order to guide potential therapeutic approaches through deactivating or reprogramming PSCs. Nevertheless, significant challenges persist in translating preclinical discoveries into clinical applications. In this review, we expect to offer a comprehensive overview of the latest molecular advancements in PSCs, along with new insights into the clinical therapeutic strategies targeting PSCs.
Collapse
Affiliation(s)
- Yawei Liu
- School of Basic Medicine Sciences, Capital Medical University, Beijing, China
- Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Ran Xue
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
77
|
Gupta J, Mohammed MH, Alghazali T, Uthirapathy S, R R, Thakur V, Kaur M, Naidu KS, Kubaev A, Al-Mukhtar MM. Inflammasomes and autophagy in cancer: unlocking targeted therapies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04184-x. [PMID: 40310530 DOI: 10.1007/s00210-025-04184-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/13/2025] [Indexed: 05/02/2025]
Abstract
This study clarifies the interaction between autophagy and inflammasome within the cancer framework. The inflammasome generates pro-inflammatory cytokines to direct the immune response to pathogens and cellular stressors. Autophagy maintains cellular homeostasis and can either promote or inhibit cancer. These pathways interact to affect tumorigenesis, immune responses, and therapy. Autophagy controls inflammasome activity by affecting cancer pathogenesis and tumor microenvironment inflammation, highlighting novel cancer therapeutic approaches. Recent studies indicate that modulating autophagy and inflammasome pathways can boost anti-cancer immunity, reduce drug-resistance, and improve therapeutic efficacy. Recent studies indicate modulating inflammasome and autophagy pathways can augment anti-cancer immunity, mitigate therapy resistance, and improve treatment efficacy. Cancer research relies on understanding the inflammasome-autophagy relationship to develop targeted therapies that enhance anti-tumor efficacy and reduce inflammatory symptoms. Customized therapies may improve outcomes based on autophagy gene variations and inflammasome polymorphisms. This study investigates autophagy pathways and the inflammasome in tumor immunopathogenesis, cytokine function, and cancer therapeutic strategies, highlighting their significance in cancer biology and treatment.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Pin Code 281406, U.P., India.
| | - Mohammed Hashim Mohammed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, Al-Maarif University, Anbar, Iraq.
| | | | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Roopashree R
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Vishal Thakur
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Manpreet Kaur
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra, Pradesh- 531162, India
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan
| | - Mahmoud Mussleh Al-Mukhtar
- Anesthesia Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| |
Collapse
|
78
|
Srisomboon Y, Tojima I, Iijima K, Kita H, O'Grady SM. Allergen-induced activation of epithelial P2Y 2 receptors promotes adenosine triphosphate exocytosis and type 2 immunity in airways. J Allergy Clin Immunol 2025; 155:1607-1622. [PMID: 39863058 DOI: 10.1016/j.jaci.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/29/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Environmental allergens induce the release of danger signals from the airway epithelium that trigger type 2 immune responses and promote airway inflammation. OBJECTIVE We investigated the role of allergen-stimulated P2Y2 receptor activation in regulating adenosine triphosphate (ATP), IL-33, and DNA release by human bronchial epithelial (hBE) cells and mouse airways. METHODS The hBE cells were exposed to Alternaria alternata extract and secretion of ATP, IL-33, and DNA were studied in vitro. Molecular and cellular mechanisms were examined by biochemical and genetic approaches. Mice were treated intranasally with pharmacologic agents and exposed to Alternaria extract. RESULTS Exposure of hBE cells to Alternaria extract stimulated P2Y2 receptors coupled to phospholipase C β3, leading to activation of multiple protein kinase C (PKC) isoforms and an increase in intracellular Ca2+ concentration. Small interfering RNAs targeting PKC δ or inhibiting PKC δ activity with delcasertib blocked exocytosis of ATP and reduced IL-33 and DNA secretion. Moreover, a peptide antagonist for myristoylated alanine-rich C-kinase substrate (MARCKS) reduced vesicular ATP release. A proximity ligand assay showed that Alternaria extract stimulated MARCKS desorption from the plasma membrane and delcasertib prevented the response. Finally, the P2Y2 receptor antagonist AR-C118925XX and delcasertib blocked IL-33, DNA, and type 2 cytokine secretion in vivo in mice exposed to Alternaria. CONCLUSION P2Y2 receptor stimulation after allergen exposure promoted activation of PLC β3, PKC δ, and MARCKS protein desorption from the apical membrane, which facilitated ATP exocytosis and subsequent secretion of IL-33 and DNA. Epithelial P2Y2 receptors serve as primary sensors for aeroallergen-induced alarmin release by airway epithelial cells.
Collapse
Affiliation(s)
- Yotesawee Srisomboon
- Departments of Animal Science, Integrative Biology, and Physiology, University of Minnesota, St Paul, Minn
| | - Ichiro Tojima
- Division of Allergy, Asthma and Clinical Immunology, Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Koji Iijima
- Division of Allergy, Asthma and Clinical Immunology, Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Hirohito Kita
- Division of Allergy, Asthma and Clinical Immunology, Department of Medicine, Mayo Clinic, Scottsdale, Ariz.
| | - Scott M O'Grady
- Departments of Animal Science, Integrative Biology, and Physiology, University of Minnesota, St Paul, Minn.
| |
Collapse
|
79
|
Zuo J, Kang J, Hong J, Li J, Fang Y, Liu C, Xie M, Chen Z. Synthesis and Preliminary Evaluation of an In Vivo Stable 131I-Labeled Deuterated Tropane for Mapping Dopamine Transporter. J Labelled Comp Radiopharm 2025; 68:e4147. [PMID: 40323699 DOI: 10.1002/jlcr.4147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/02/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025]
Abstract
Dopamine transporter (DAT) is closely associated with neurodegenerative diseases such as Parkinson's disease (PD). To develop an in vivo stable radioligand targeting DAT, we synthesized a novel iodine-131-labeled tropane analog [131I]1 with deuteration on both the N-fluoropropyl and 2β-carbomethoxy positions of the tropane scaffold and then biologically compared with the previously reported analog [131I]FP-CIT-d6 with deuteration only on the N-fluoropropyl group. The radioligand [131I]1 was obtained via a radioiodine-destannylation reaction with a radiochemical yield of ~95%, a radiochemical purity of > 99% and a molar activity of 12.72 GBq/μmol. [131I]1 exhibited a high in vitro binding affinity for DAT with an IC50 value of 2.2 nM. Metabolic stability studies demonstrated that [131I]1 displayed superior in vivo stability compared with [131I]FP-CIT-d6. Biodistribution studies revealed that [131I]1 had better DAT affinity, specificity, and a higher target-to-background ratio than [131I]FP-CIT-d6. Ex vivo autoradiography and blocking experiments validated the selective and reversible binding of [131I]1 to DAT and superiority to [131I]FP-CIT-d6. These preliminary results suggest that deuterated radioligand [131I]1, with enhanced in vivo stability and higher target-to-background ratio, is a promising DAT probe, warranting the development and application of 123I-labeled counterpart ([123I]1) for SPECT imaging in DAT-related disorders.
Collapse
Affiliation(s)
- Jiaojiao Zuo
- School of Pharmaceutical Science, Inner Mongolia Medical University, Hohhot, China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Jing Kang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Jingjing Hong
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Jingwen Li
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yi Fang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Chunyi Liu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Minhao Xie
- School of Pharmaceutical Science, Inner Mongolia Medical University, Hohhot, China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Zhengping Chen
- School of Pharmaceutical Science, Inner Mongolia Medical University, Hohhot, China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
80
|
Bahriz HA, Abdelaziz RR, El-Kashef DH. Allopurinol abates hepatocellular carcinoma in rats via modulation of NLRP3 inflammasome and NF-κB pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6043-6058. [PMID: 39636403 DOI: 10.1007/s00210-024-03666-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024]
Abstract
The present research was performed to examine the possible capability of allopurinol to prevent developing hepatocellular carcinoma (HCC) and to explore the fundamental mechanisms that control the hepatoprotective effect considering the enormous impact of HCC on patients' quality of life. Male Sprague Dawely rats were given i.p. injection of thioacetamide (TAA) (200 mg/kg) twice a week for 16 weeks in order to induce HCC. The histological analysis and assessment of the serum levels of liver function indicators verified the development of HCC. Two regimens of allopurinol (100 mg/kg, p.o.) were used; the first involved giving it concurrently with TAA from week 13 to week 16, and the second regimen started from week 9 to week 16. Chronic TAA damage was associated with considerable overexpression of the profibrogenic cytokine TGF-β, degradation and nuclear translocation of NF-κB, which released a number of inflammatory mediators, and upregulation of the NLRP3/caspase1 pathway. Administration of allopurinol demonstrated considerable enhancements in liver function and oxidative balance. Moreover, pathological characteristics like cirrhosis, dysplastic changes, and HCC nodules were greatly diminished. Allopurinol via suppressing TGF-β expression, inhibiting NF-κB nuclear translocation, and restricting inflammatory NLRP3/caspase1/IL-1β pathway was able to protect against TAA-induced liver damage, and it could be a promising therapy for HCC.
Collapse
MESH Headings
- Animals
- Allopurinol/pharmacology
- Allopurinol/therapeutic use
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Male
- NF-kappa B/metabolism
- Carcinoma, Hepatocellular/chemically induced
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/prevention & control
- Inflammasomes/metabolism
- Rats, Sprague-Dawley
- Thioacetamide
- Signal Transduction/drug effects
- Rats
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Liver Neoplasms/chemically induced
- Liver Neoplasms/drug therapy
- Liver Neoplasms, Experimental/pathology
- Liver Neoplasms, Experimental/chemically induced
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/prevention & control
- Liver Neoplasms, Experimental/drug therapy
- Liver/drug effects
- Liver/pathology
- Liver/metabolism
Collapse
Affiliation(s)
- Heba A Bahriz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| | - Rania R Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Dalia H El-Kashef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
81
|
Ehlers L, Meyts I. Getting to know adenosine deaminase 2 deficiency inside and out. J Allergy Clin Immunol 2025; 155:1451-1463. [PMID: 39956283 DOI: 10.1016/j.jaci.2025.01.040] [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: 11/22/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/18/2025]
Abstract
Ten years after the description of the first cohorts of patients with adenosine deaminase (ADA2) deficiency (DADA2), the pathomechanisms underlying the disease on a cellular level remain poorly understood. With the establishment of the lysosomal localization of the ADA2 protein and its involvement in nucleic acid sensing, the pathophysiologic focus has shifted to the inside of the cell. At the same time, extracellular (serum) ADA2 enzyme activity continues to be the diagnostic reference standard in patients with suspected DADA2. The diverse clinical phenotype and weak genotype-phenotype correlations further complicate the identification of shared cellular mechanisms that cause inflammation, immunodeficiency, and bone marrow failure in the absence of functional ADA2. This review inspects the characteristics of the ADA2 protein and its proposed function. The latter is discussed in the context of possible mechanisms driving the clinical phenotype in patients lacking functional ADA2. We discuss established processes and introduce unexplored pathways in the pathogenesis of DADA2.
Collapse
Affiliation(s)
- Lisa Ehlers
- Department of Microbiology, Immunology and Transplantation, Laboratory for Inborn Errors of Immunity, Katholieke Universiteit (KU) Leuven, Leuven, Belgium; Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany; German Center for Child and Adolescent Health (DZKJ), partner site Berlin, Berlin, Germany; Deutsches Rheuma-Forschungszentrum, an Institute of the Leibniz Association, Berlin, Germany
| | - Isabelle Meyts
- Department of Microbiology, Immunology and Transplantation, Laboratory for Inborn Errors of Immunity, Katholieke Universiteit (KU) Leuven, Leuven, Belgium; Department of Pediatrics, University Hospitals Leuven, KU Leuven, Leuven, Belgium.
| |
Collapse
|
82
|
Liu J, Deng L, Yao B, Zhang Y, Huang J, Huang S, Liang C, Shen Y, Wang X. Carboxylesterase 2A gene knockout or enzyme inhibition alleviates steatohepatitis in rats by regulating PPARγ and endoplasmic reticulum stress. Free Radic Biol Med 2025; 232:279-291. [PMID: 40089078 DOI: 10.1016/j.freeradbiomed.2025.03.021] [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: 01/14/2025] [Revised: 03/09/2025] [Accepted: 03/13/2025] [Indexed: 03/17/2025]
Abstract
Metabolic dysfunction associated steatotic liver disease (MASLD) is a widespread liver disease that progresses from simple steatosis to severe steatohepatitis stage. Despite the recognized importance of carboxylesterase 2 (CES2) in hepatic lipid metabolism, the role of CES2 in hepatic inflammation remains unclear. The rat genome encodes six Ces2 genes and Ces2a shows high expression in the liver and intestine. Lipid metabolism, inflammation, fibrosis, and endoplasmic reticulum (ER) stress were investigated in Ces2a knockout (KO) rats. KO rats showed spontaneous liver lipid accumulation due to increased lipogenesis and reduced fatty acid oxidation. Non-targeted lipidomic analysis revealed enhanced lysophosphatidylcholines (LPCs) and phosphatidylcholines (PCs) in KO rats and increased concentrations of ligands, thus activating the expression of PPARγ. Although there was simple lipid accumulation in the liver of KO rats, Ces2a deficiency showed a significant protective effect against LPS and diet-induced hepatic steatohepatitis by inhibiting ER stress regulated by PPARγ activation. In line with this, treatment with tanshinone IIA, a CES2 inhibitor, significantly alleviated the progression of steatohepatitis induced by the MCD diet. In conclusion, the increased PPARγ expression in Ces2a deficiency may counteract liver inflammation and ER stress despite the presence of simple steatosis. Therefore, CES2 inhibition represents a potential therapeutic approach for steatohepatitis.
Collapse
Affiliation(s)
- Jie Liu
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Luyao Deng
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Bingyi Yao
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Yuanjin Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Junze Huang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Shengbo Huang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Chenmeizi Liang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Yifei Shen
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Xin Wang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China.
| |
Collapse
|
83
|
d'Ovidio D, Adami C. Pain Management for Gastrointestinal Conditions in Exotic Animals. Vet Clin North Am Exot Anim Pract 2025; 28:347-363. [PMID: 39730216 DOI: 10.1016/j.cvex.2024.11.006] [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] [Indexed: 12/29/2024]
Abstract
Gastrointestinal (GI) disorders are very common in exotic animals, such as reptiles, birds, mammals, and can be extremely painful. This review aims to provide the reader with a better understanding of the different pain mechanisms and manifestations across orders and species in order to provide the most updated information on pain recognition and management for GI conditions in exotic animals.
Collapse
Affiliation(s)
- Dario d'Ovidio
- European College of Zoological Medicine (Small Mammals); Private practitioner, Via C. Colombo 118, 80022 Arzano, Naples, Italy.
| | - Chiara Adami
- Department of Veterinary Medicine, University of Cambridge, CB3 0ES, Cambridge, UK
| |
Collapse
|
84
|
Dong RF, Qin CJ, Yin Y, Han LL, Xiao CM, Wang KD, Wei RY, Xia YZ, Kong LY. Discovery of a potent inhibitor of chaperone-mediated autophagy that targets the HSC70-LAMP2A interaction in non-small cell lung cancer cells. Br J Pharmacol 2025; 182:2287-2309. [PMID: 37311689 DOI: 10.1111/bph.16165] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/26/2023] [Accepted: 05/27/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND AND PURPOSE Chaperone-mediated autophagy (CMA) is a selective type of autophagy targeting protein degradation and maintains high activity in many malignancies. Inhibition of the combination of HSC70 and LAMP2A can potently block CMA. At present, knockdown of LAMP2A remains the most specific method for inhibiting CMA and chemical inhibitors against CMA have not yet been discovered. EXPERIMENTAL APPROACH Levels of CMA in non-small cell lung cancer (NSCLC) tissue samples were confirmed by tyramide signal amplification dual immunofluorescence assay. High-content screening was performed based on CMA activity, to identify potential inhibitors of CMA. Inhibitor targets were determined by drug affinity responsive target stability-mass spectrum and confirmed by protein mass spectrometry. CMA was inhibited and activated to elucidate the molecular mechanism of the CMA inhibitor. KEY RESULTS Suppression of interactions between HSC70 and LAMP2A blocked CMA in NSCLC, restraining tumour growth. Polyphyllin D (PPD) was identified as a targeted CMA small-molecule inhibitor through disrupting HSC70-LAMP2A interactions. The binding sites for PPD were E129 and T278 at the nucleotide-binding domain of HSC70 and C-terminal of LAMP2A, respectively. PPD accelerated unfolded protein generation to induce reactive oxygen species (ROS) accumulation by inhibiting HSC70-LAMP2A-eIF2α signalling axis. Also, PPD prevented regulatory compensation of macroautophagy induced by CMA inhibition via blocking the STX17-SNAP29-VAMP8 signalling axis. CONCLUSIONS AND IMPLICATIONS PPD is a targeted CMA inhibitor that blocked both HSC70-LAMP2A interactions and LAMP2A homo-multimerization. CMA suppression without increasing the regulatory compensation from macroautophagy is a good strategy for NSCLC therapy. LINKED ARTICLES This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc.
Collapse
Affiliation(s)
- Rui-Fang Dong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Cheng-Jiao Qin
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yong Yin
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Liang-Liang Han
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Cheng-Mei Xiao
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Kai-Di Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Rong-Yuan Wei
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yuan-Zheng Xia
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ling-Yi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
85
|
Cortes-Justo E, Ortiz-Butrón R, Vilches-Flores A. Cannabidiol oil delays pancreatic islet dysfunction in Wistar rats under hypercaloric diet. Biomed Pharmacother 2025; 186:117993. [PMID: 40106970 DOI: 10.1016/j.biopha.2025.117993] [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: 11/06/2024] [Revised: 03/09/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Hypercaloric diet (HCD) intake can lead to metabolic alterations, such as metabolic syndrome and type-2 diabetes mellitus. Phytocannabinoid cannabidiol (CBD) is a GPR55 receptor antagonist involved in insulin secretion and other functions in pancreatic islet. The therapeutic use of CBD has been suggested for diabetes, but little is known regarding its effects on pancreatic islet physiology. Our aim was to evaluate the effects of CBD oil on pancreatic islets, from Wistar rats under HCD. Male rats were divided in 4 groups: Normal diet vehicle-treated (control) and CBD-treated group. Rats under HCD were subdivided in treated with vehicle (HCD) and with CBD oil administered 21 mg/Kg orally, 0.5 ml in 3 days per week; controls received coconut oil as vehicle. Body weight, food intake, and water consumption were recorded. After 20 weeks, glucose tolerance curve was performed; serum insulin was determined by ELISA, and pancreas was removed for histological and gene expression analysis for insulin, glucagon, PDX-1, MafA and GPR55 receptor. CBD treatment reduced body weight and food intake but increased fluid consumption, independently of diets. In control group, CBD did not alter blood glucose and serum insulin, but modified expression for GPR55 receptor, glucagon, insulin and MafA. Rats under HCD and treated with CBD decreased glycaemia, insulinaemia, islets relative area, GPR55-positive cells, PDX-1 and MafA gene expression, meanwhile insulin and glucagon expression was increased. In conclusion, CBD ameliorated HCD effects through changes in insulin, glucagon and GPR55 receptor expressions. We assume CBD interacts with other receptors beside GPR55.
Collapse
Affiliation(s)
- Edgardo Cortes-Justo
- Posgrado e Investigación, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Rocío Ortiz-Butrón
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas. Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Alonso Vilches-Flores
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico.
| |
Collapse
|
86
|
Si Z, Zhao S, Zhang Z, Chen T, Wang R, Dong C, Wang K, Sun C, Song Z, Shen Z, Gao W. Bone marrow mesenchymal stem cells alleviate liver fibrosis after rat liver transplantation through JAK1/STAT5 pathway. Stem Cell Res Ther 2025; 16:217. [PMID: 40312752 PMCID: PMC12044927 DOI: 10.1186/s13287-025-04353-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/17/2025] [Indexed: 05/03/2025] Open
Abstract
OBJECTIVE The effectiveness of bone marrow mesenchymal stem cells (BMSCs) in post-transplantation liver fibrosis has not been studied. The aim of this study was to investigate the effect of BMSCs on liver fibrosis and their role in the Janus-activated kinase (JAK) 1/ signal transducer and activator of transcription (STAT) 5 pathway after liver transplantation (LT). METHODS A rat model of post-LT liver fibrosis induced by cold ischemia injury was successfully established. BMSCs were injected into the rats through the portal vein. Hepatic stellate cell (HSC)-T6 were co-cultured with BMSCs in vitro after hypoxia-reoxygenation. JAK1 inhibitor Abrocitinib and JAK1 agonist RO8191 were used to study the JAK1/STAT5 signaling pathway. RESULTS BMSCs significantly alleviated liver fibrosis caused by cold ischemia-reperfusion injury after rat LT in vivo. After BMSCs transplantation, the levels of JAK1 and p-STAT5 in rat liver were significantly reduced. After using Abrocitinib, the stage of liver fibrosis and the levels of collagen type I alpha 1 chain (COL1A1) and actin alpha 2 (ACTA2) decreased. After using RO8191, the stage of liver fibrosis and the levels of COL1A1 and ACTA2 increased. BMSCs significantly reduced the activation of HSC-T6 after hypoxia-reoxygenation in vitro. After co-culturing with BMSCs after HSC-T6 hypoxia-reoxygenation, the levels of JAK1 and p-STAT5 were significantly reduced. After the addition of Abrocitinib, the levels of COL1A1 and ACTA2 decreased in HSC-T6; in contrast, after adding RO8191, the levels of COL1A1 and ACTA2 increased in HSC-T6 after hypoxia-reoxygenation. After using anti-IL7 antibody or anti-IL7Rα in vivo and in vitro, the stage of liver fibrosis and the levels of COL1A1 and ACTA2 decreased as well as the phosphorylation level of STAT5. CONCLUSIONS BMSCs alleviate hepatic cell damage, reduce hepatic cell-derived IL7, downregulate IL7R/JAK1/STAT5 in HSCs, thereby reducing HSCs' activation and ultimately alleviating liver fibrosis after liver transplantation.
Collapse
Affiliation(s)
- Zhuyuan Si
- Department of Organ Transplantation, Qilu Hospital of Shandong University, Jinan, China
- Department of Liver Transplantation, Organ Transplantation Center, Tianjin First Central Hospital, Tianjin, China
| | - Shengqiao Zhao
- Department of Liver Transplantation, Organ Transplantation Center, Tianjin First Central Hospital, Tianjin, China
| | - Zhixin Zhang
- Department of Liver Transplantation, Organ Transplantation Center, Tianjin First Central Hospital, Tianjin, China
| | - Tianran Chen
- Department of Liver Transplantation, Organ Transplantation Center, Tianjin First Central Hospital, Tianjin, China
| | - Ruofan Wang
- Department of Liver Transplantation, Organ Transplantation Center, Tianjin First Central Hospital, Tianjin, China
| | - Chong Dong
- Department of Liver Transplantation, Organ Transplantation Center, Tianjin First Central Hospital, Tianjin, China
| | - Kai Wang
- Department of Liver Transplantation, Organ Transplantation Center, Tianjin First Central Hospital, Tianjin, China
| | - Chao Sun
- Department of Liver Transplantation, Organ Transplantation Center, Tianjin First Central Hospital, Tianjin, China
| | - Zhuolun Song
- Department of Liver Transplantation, Organ Transplantation Center, Tianjin First Central Hospital, Tianjin, China
- Tianjin Key Laboratory of Organ Transplantation, Tianjin First Central Hospital, Tianjin, China
| | - Zhongyang Shen
- Department of Liver Transplantation, Organ Transplantation Center, Tianjin First Central Hospital, Tianjin, China.
- Tianjin Key Laboratory of Organ Transplantation, Tianjin First Central Hospital, Tianjin, China.
| | - Wei Gao
- Department of Liver Transplantation, Organ Transplantation Center, Tianjin First Central Hospital, Tianjin, China.
- Tianjin Key Laboratory of Organ Transplantation, Tianjin First Central Hospital, Tianjin, China.
| |
Collapse
|
87
|
Kuchay MS, Choudhary NS, Ramos-Molina B. Pathophysiological underpinnings of metabolic dysfunction-associated steatotic liver disease. Am J Physiol Cell Physiol 2025; 328:C1637-C1666. [PMID: 40244183 DOI: 10.1152/ajpcell.00951.2024] [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: 01/22/2025] [Revised: 01/31/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is emerging as the leading cause of chronic liver disease worldwide, reflecting the global epidemics of obesity, metabolic syndrome, and type 2 diabetes. Beyond its strong association with excess adiposity, MASLD encompasses a heterogeneous population that includes individuals with normal body weight ("lean MASLD") highlighting the complexity of its pathogenesis. This disease results from a complex interplay between genetic susceptibility, epigenetic modifications, and environmental factors, which converge to disrupt metabolic homeostasis. Adipose tissue dysfunction and insulin resistance trigger an overflow of lipids to the liver, leading to mitochondrial dysfunction, oxidative stress, and hepatocellular injury. These processes promote hepatic inflammation and fibrogenesis, driven by cross talk among hepatocytes, immune cells, and hepatic stellate cells, with key contributions from gut-liver axis perturbations. Recent advances have unraveled pivotal molecular pathways, such as transforming growth factor-β signaling, Notch-induced osteopontin, and sphingosine kinase 1-mediated responses, that orchestrate fibrogenic activation. Understanding these interconnected mechanisms is crucial for developing targeted therapies. This review integrates current knowledge on the pathophysiology of MASLD, emphasizing emerging concepts such as lean metabolic dysfunction-associated steatohepatitis (MASH), epigenetic alterations, hepatic extracellular vesicles, and the relevance of extrahepatic signals. It also discusses novel therapeutic strategies under investigation, aiming to provide a comprehensive and structured overview of the evolving MASLD landscape for both basic scientists and clinicians.
Collapse
Affiliation(s)
| | - Narendra Singh Choudhary
- Institute of Digestive and Hepatobiliary Sciences, Medanta-The Medicity Hospital, Gurugram, India
| | - Bruno Ramos-Molina
- Group of Obesity, Diabetes & Metabolism, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| |
Collapse
|
88
|
Prete MM, Feitosa GTB, Ribeiro MAT, Fidalgo TM, Sanchez ZM. Adverse clinical effects associated with the use of synthetic cannabinoids: A systematic review. Drug Alcohol Depend 2025; 272:112698. [PMID: 40334326 DOI: 10.1016/j.drugalcdep.2025.112698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 05/09/2025]
Abstract
Synthetic cannabinoids (SCs) are potent agonists of CB1 and CB2 receptors, with affinities approximately 100 times greater than that of natural cannabis. This increased potency is associated with severe clinical outcomes, including psychosis, dependence, and various autonomic disturbances, such as seizures and rhabdomyolysis. The objective of this study was to synthesize the existing literature on the clinical effects of SCs, emphasizing health risks and identifying knowledge gaps. Following PRISMA guidelines, a comprehensive search was conducted across three databases-PubMed, Embase, and Lilacs-resulting in 944 studies. Eligible articles focused on clinical effects associated to SC use, while exclusion criteria encompassed studies unrelated to clinical outcomes, other reviews, animal studies, and those concentrating solely on psychiatric symptoms or therapeutic uses of SCs. In total, 49 studies published between 2010 and 2022 were included, representing diverse populations and study designs. Participants were predominantly young adult males, although ages ranged from 12 to 72 years. SCs' clinical effects predominantly affected the neurological and cardiovascular systems, with common symptoms including seizures, altered consciousness, tachycardia, and hypertension. Hospital and ICU admissions varied, reflecting the complex nature of SC toxicity. Compared to cannabis, SC use was linked to more severe cardiovascular and neurological complications. Additional rare complications included thromboembolic events, immune thrombocytopenic purpura, and psychiatric disturbances. This review highlights the urgent need for targeted public health policies to mitigate the risks associated with SC use and improve medical management. It also stresses the importance of further controlled studies to elucidate the underlying mechanisms of these clinical effects and their pharmacological basis.
Collapse
Affiliation(s)
- Mariana M Prete
- Departamento de Medicina Preventiva, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Gabriel T B Feitosa
- Departamento de Psiquiatria, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Maria A T Ribeiro
- Departamento de Psiquiatria, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - T M Fidalgo
- Departamento de Psiquiatria, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Zila M Sanchez
- Departamento de Medicina Preventiva, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.
| |
Collapse
|
89
|
Kim SK, Kim JH, Moon I, Min J, Park J, Kim MG. Cardioprotective Effects of PARP Inhibitors: A Meta-Analysis of Animal Studies. Transl Res 2025:S1931-5244(25)00049-0. [PMID: 40316011 DOI: 10.1016/j.trsl.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
Poly(adenosine diphosphate [ADP] ribose) polymerase (PARP) inhibitors are expected to provide benefits to the cardiovascular system. However, the cardioprotective effect of PARP inhibitors has not been systematically reviewed or quantitatively analyzed. This study aimed to assess the cardioprotective effects of PARP inhibitors through a meta-analysis of animal studies. Three databases PubMed, Web of Sciences, and Embase were searched until September 1, 2023. The risk of bias was assessed using SYRCLE's Risk of Bias. A total of 74 animal studies that investigated the cardiac function of PARP inhibitors compared to placebo or vehicle, were included. Outcome measures were hemodynamic indexes, cardiac contractility, and biomarkers of myocardial injury. Pooled effect size was estimated using a random-effects model with RevMan 5.4. PARP inhibitors were associated with enhanced hemodynamic indexes, including cardiac output (standardized mean difference, 0.86 [95% CI, 0.54 to 1.17]; p < 0.00001) and stroke volume (0.42 [0.07 to 0.76]; p = 0.02). PARP inhibitors were associated with increased cardiac contractility, including ejection fraction (0.71 [0.42 to 1.01]; p < 0.00001) and fractional shortening (0.96 [0.62 to 1.31]; p < 0.00001). PARP inhibitors were associated with decreased troponin І (-1.42 [-2.16 to -0.68]; p = 0.0002), plasma B-type natriuretic peptide (-0.95 [-1.56 to -0.33]; p = 0.003), creatine kinase (-1.81 [-2.63 to -0.99]; p < 0.0001), and infarct size (-1.58 [-2.01 to -1.14]; p < 0.00001). PARP inhibitors improve cardiac functions and attenuate myocardial injury in animals, which indicate the cardioprotective effects. Further human studies are necessary.
Collapse
Affiliation(s)
- Seong Kyung Kim
- College of Pharmacy, Ewha Womans University, Seoul, South Korea
| | - Jae Hyun Kim
- School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, Jeonju, South Korea
| | - Inyeong Moon
- College of Pharmacy, Ewha Womans University, Seoul, South Korea
| | - Jiwon Min
- College of Pharmacy, Ewha Womans University, Seoul, South Korea
| | - Jieun Park
- College of Pharmacy, Ewha Womans University, Seoul, South Korea
| | - Myeong Gyu Kim
- College of Pharmacy, Ewha Womans University, Seoul, South Korea; Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
90
|
Cheng Y, Yang Y, Su Y, Chen R, Qian D, Xu J. FAERS based disproportionality analysis and network pharmacology investigation of taxanes associated drug induced liver injury. Sci Rep 2025; 15:15137. [PMID: 40307389 PMCID: PMC12043806 DOI: 10.1038/s41598-025-99669-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 04/22/2025] [Indexed: 05/02/2025] Open
Abstract
Taxanes play a crucial role in cancer treatment, particularly for non-small cell lung cancer and breast cancer. However, real-world studies examining drug-induced liver injury (DILI) associated with these drugs remain limited. Our study investigates the association between taxanes and DILI through analysis of the Food and Drug Administration Adverse Event Reporting System (FAERS) database, alongside an exploration of potential hepatotoxicity mechanisms via network pharmacology. We collected DILI reports related to taxanes from the FAERS database between January 2004 and March 2024, employing disproportionality analyses with the reporting odds ratio (ROR) and 95% confidence intervals. Our findings revealed a significant association between paclitaxel (ROR = 2.35) and nab-paclitaxel (ROR = 3.14) with DILI, while docetaxel demonstrated no significant correlation (ROR = 0.68), although it was linked to higher mortality rates and earlier onset. Network pharmacology analysis uncovered that the mechanisms of liver injury induced by these two drugs may not be entirely congruent. Unique targets for docetaxel included BCL2, CNR2, and MAPK1, while the 'Regulation of lipolysis in adipocytes' pathway was specifically associated with docetaxel-induced DILI. Our findings indicate that taxanes exhibit differential hepatotoxic risks and hepatotoxicity mechanisms, emphasizing the need for enhanced drug safety monitoring strategies for cancer patients.
Collapse
Affiliation(s)
- Yijie Cheng
- Pharmacy Department, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu, China
- Central Laboratory, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu, China
- School of Biology and Food Engineering, Changshu Institute of Technology, Changshu, China
| | - Yuxin Yang
- School of Biology and Food Engineering, Changshu Institute of Technology, Changshu, China
| | - Yan Su
- School of Biology and Food Engineering, Changshu Institute of Technology, Changshu, China
| | - Ruihuan Chen
- Pharmacy Department, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu, China
| | - Da Qian
- Central Laboratory, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu, China.
| | - Jingyuan Xu
- Central Laboratory, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu, China.
- School of Biology and Food Engineering, Changshu Institute of Technology, Changshu, China.
| |
Collapse
|
91
|
Magesh K, Wu SP, Velmathi S. A near-infrared julolidine probe for visualization of mitochondrial peroxynitrite in living cells. Org Biomol Chem 2025; 23:4142-4151. [PMID: 40168033 DOI: 10.1039/d5ob00036j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The overproduction of peroxynitrite (ONOO-) in mitochondria has been associated with various pathophysiological conditions and disorders. However, the use of fluorescent probes to visualize mitochondrial ONOO- in biological systems is limited due to their low emission wavelengths and small Stokes shifts, which present significant challenges. In this study, we designed and synthesized julolidine-based near-infrared (NIR) fluorescent probes, named JQMe and JCN, specifically to monitor mitochondrial ONOO-. Comparative photophysical studies revealed that JQMe exhibits superior properties for sensing ONOO- compared to JCN. Initially, JQMe emitted fluorescence emission at 706 nm via an intramolecular charge transfer (ICT) mechanism. Upon the addition of ONOO-, the NIR fluorescence emission of JQMe at 706 nm was suppressed, resulting in a rapid on-off fluorescence response within 5 minutes. JQMe exhibited high specific selectivity towards ONOO- over other competing interferents, accompanied by a colorimetric change from deep blue to colorless. Additionally, JQMe exhibited a significant Stokes shift of 106 nm and a low detection limit of 6.5 nM. The proposed sensing mechanism was validated through ESI mass spectrometry and DFT studies. Furthermore, JQMe was successfully employed to monitor both endogenous and exogenous ONOO- in living cells using inducer and inhibitor tests. Remarkably, time-dependent colocalization experiments revealed that JQMe effectively targets and reacts with mitochondrial ONOO-.
Collapse
Affiliation(s)
- Kuppan Magesh
- Organic and Polymer Synthesis Laboratory, Department of Chemistry, National Institute of Technology, Tiruchirappalli - 620 015, India.
| | - Shu Pao Wu
- Department of Applied Chemistry, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan, China
| | - Sivan Velmathi
- Organic and Polymer Synthesis Laboratory, Department of Chemistry, National Institute of Technology, Tiruchirappalli - 620 015, India.
| |
Collapse
|
92
|
Yang J, des Rieux A, Malfanti A. Stimuli-Responsive Nanomedicines for the Treatment of Non-cancer Related Inflammatory Diseases. ACS NANO 2025; 19:15189-15219. [PMID: 40249331 PMCID: PMC12045021 DOI: 10.1021/acsnano.5c00700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
Nanomedicines offer a means to overcome the limitations associated with traditional drug dosage formulations by affording drug protection, enhanced drug bioavailability, and targeted drug delivery to affected sites. Inflamed tissues possess unique microenvironmental characteristics (including excessive reactive oxygen species, low pH levels, and hypoxia) that stimuli-responsive nanoparticles can employ as triggers to support on-demand delivery, enhanced accumulation, controlled release, and activation of anti-inflammatory drugs. Stimuli-responsive nanomedicines respond to physicochemical and pathological factors associated with diseased tissues to improve the specificity of drug delivery, overcome multidrug resistance, ensure accurate diagnosis and precision therapy, and control drug release to improve efficacy and safety. Current stimuli-responsive nanoparticles react to intracellular/microenvironmental stimuli such as pH, redox, hypoxia, or specific enzymes and exogenous stimuli such as temperature, magnetic fields, light, and ultrasound via bioresponsive moieties. This review summarizes the general strategies employed to produce stimuli-responsive nanoparticles tailored for inflammatory diseases and all recent advances, reports their applications in drug delivery, and illustrates the progress made toward clinical translation.
Collapse
Affiliation(s)
- Jingjing Yang
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Anne des Rieux
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Alessio Malfanti
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
- Department
of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| |
Collapse
|
93
|
Xie L, Peng YQ, Shen X. Identifying therapeutic target genes for diabetic retinopathy using systematic druggable genome-wide Mendelian randomization. Diabetol Metab Syndr 2025; 17:145. [PMID: 40301928 PMCID: PMC12039192 DOI: 10.1186/s13098-025-01710-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 04/22/2025] [Indexed: 05/01/2025] Open
Abstract
INTRODUCTION The treatment and prevention of diabetic retinopathy (DR) remain significant challenges. Mendelian randomization (MR) has been widely used to explore novel therapeutic targets. In this study, we conducted a systematic druggable genome-wide MR analysis to explore potential therapeutic targets for DR. METHODS We obtained data on druggable genes and screened for genes within blood expression quantitative trait loci (eQTL), which were then subjected to MR analysis and colocalization analysis with DR genome-wide association studies data to identify genes strongly associated with DR. Additionally, Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, protein-protein interaction (PPI) network construction, drug candidate prediction, and molecular docking were performed to provide valuable insights for the development of more effective and targeted therapeutic drugs. RESULTS MR analysis of blood eQTLs revealed 30 significant DR-associated druggable genes, with PRKAB1 (OR = 0.935, 95% CI: 0.892 to 0.980) and CNR1 (OR = 0.814, 95% CI: 0.696 to 0.951) being protective genes, whereas CACNA1E (OR = 1.282, 95% CI: 1.050 to 1.565), NME1 (OR = 1.198, 95% CI: 1.028 to 1.397), and CHRNA2 (OR = 1.192, 95% CI: 1.025 to 1.386) were associated with increased risk. KEGG analysis highlighted significant pathways, including adrenergic signaling in cardiomyocytes (hsa04261), the oxytocin signaling pathway (hsa04921), and arrhythmogenic right ventricular cardiomyopathy (hsa05412). PPI network analysis identified two key modules: one comprising BIN1, CDH2, ACTN1, EPAS1, CEBPA, and CTSD nodes, and the other consisting of CACNG6, CACNA1E, CACNA2D3, and RASGRP3 nodes. Drug candidate prediction suggested ethanol and isoflupredone as potential therapeutic interventions, and molecular docking revealed C5's strong protein binding affinity. CONCLUSIONS This study utilized MR and colocalization analysis to identify potential drug targets for DR. The findings provide promising leads for the treatments of DR, potentially reducing drug development costs.
Collapse
Affiliation(s)
- Long Xie
- Department of Orthopedics, The Fourth Hospital of Changsha (Integrated Traditional Chinese and Western Medicine Hospital of Changsha), Hunan Normal University, Changsha, Hunan Province, 410006, China.
| | - Yu Qin Peng
- Department of Ophthalmology, The Fourth Hospital of Changsha (Integrated Traditional Chinese and Western Medicine Hospital of Changsha), Hunan Normal University, Changsha, Hunan Province, 410006, China
| | - Xiang Shen
- Department of Orthopedics, The Fourth Hospital of Changsha (Integrated Traditional Chinese and Western Medicine Hospital of Changsha), Hunan Normal University, Changsha, Hunan Province, 410006, China
| |
Collapse
|
94
|
Ning Y, Li B, Chen W, Feng L, Huang X, Liu B. DNA Framework-Based Lysosome-Targeting Chimeras: Intracellular ATP-Facilitated Extracellular Protein Degradation. ACS NANO 2025; 19:15853-15862. [PMID: 40237339 DOI: 10.1021/acsnano.5c00745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Targeted protein degradation (TPD) offered a riveting therapeutic paradigm to eradicate pathogenesis-relevant proteins, especially those belonging to the once-considered undruggable proteome. Considering that adenosine triphosphate (ATP) is the primary energy source for cell activities and lysosomes are important ATP storage sites, herein, the first example of dual-function tetrahedral DNA framework-based lysosome-targeting chimeras (TDF-LYTACs) is proposed for elucidating the correlation between extracellular protein degradation via the lysosome pathway and the fluctuations in intracellular ATP levels. In our study, platelet-derived growth factor (PDGF), a driver of cancer invasion and metastasis, was chosen as the protein of interest. To achieve multifunctionality, we employed a tetrahedral DNA framework formed by an aptamer of PDGF, human apurinic/apyrimidinic endonuclease 1 (APE1)-triggered ATP probes, and a ligand of the cell-surface lysosome-shuttling receptor (IGFIIR). TDF-LYTACs efficiently and quickly shuttled PDGF proteins to lysosomes, degraded them through the lysosomal pathway, and further visualized the intracellular ATP level synchronously. Furthermore, we found a significant correlation between the degradation efficiency of PDGF and intracellular ATP levels over time; that is, a higher ATP level corresponded to higher degradation efficiency and vice versa. We anticipate that our versatile TDF-LYTACs will offer a perspective for degrading multifunctional extracellular proteins.
Collapse
Affiliation(s)
- Yujun Ning
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers and Institute of Biomedical Sciences, Fudan University, Shanghai 200433, People's Republic of China
| | - Bin Li
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers and Institute of Biomedical Sciences, Fudan University, Shanghai 200433, People's Republic of China
| | - Weishuai Chen
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers and Institute of Biomedical Sciences, Fudan University, Shanghai 200433, People's Republic of China
| | - Langxia Feng
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers and Institute of Biomedical Sciences, Fudan University, Shanghai 200433, People's Republic of China
| | - Xuedong Huang
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers and Institute of Biomedical Sciences, Fudan University, Shanghai 200433, People's Republic of China
| | - Baohong Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers and Institute of Biomedical Sciences, Fudan University, Shanghai 200433, People's Republic of China
| |
Collapse
|
95
|
Che Z, Cai M, Dong X, Yuan Y, Wang Y, Xiao L, Song Y, Zhong J, Luo P, Wang H, Lu G, Sun Y, Xiao J. Angiotensinogen inhibition concurrently mitigates alcohol-associated hepatic and muscle injury. Metabolism 2025:156275. [PMID: 40311841 DOI: 10.1016/j.metabol.2025.156275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 04/16/2025] [Accepted: 04/27/2025] [Indexed: 05/03/2025]
Abstract
AIMS The organ communication mechanisms driven by alcohol-associated liver disease (ALD) remain inadequately understood. This study explores the endocrine roles of the hepatokine angiotensinogen (AGT) and the renin-angiotensin system (RAS) in ALD. METHODS AND RESULTS Hepatokine screening tests revealed that chronic-binge ethanol consumption upregulates hepatic AGT production, triggering downstream RAS activation. Hepatocyte-specific knockout of Agt (AGTΔHep) significantly alleviated ALD-induced liver injury. In organ screening between AGTflox/flox (AGTf/f) and AGTΔHep mice, skeletal muscle exhibited the most pronounced improvement in alcoholic myopathy (AM)-related phenotypes, including reduced muscle mass, enhanced oxidative stress, and mitochondrial dysfunction post-ethanol administration. Mechanistically, the renin-angiotensin axis transmits damaging signals from AGT to their membrane receptor AGTR1 in both hepatocytes and myocytes. Pharmacological inhibition of AGT, renin, and angiotensin-converting enzyme, as well as specific knockdown of Agtr1 in hepatocytes or myocytes, effectively attenuated both conditions. Activation of the counteractive axis of the RAS-AGTR1 pathway, involving Ang (1-7) and its membrane receptor MAS1, ameliorated the alcoholic injury of both liver and muscle. Conversely, specific knockdown of Mas1 in hepatocytes and myocytes exacerbated these injuries. CONCLUSIONS Our work demonstrates that hepatokine AGT promotes ALD and AM through the activation of the RAS-AGTR1 axis and the inhibition of the Ang (1-7)-MAS1 axis, offering a foundation for concurrent therapeutic strategies for both diseases.
Collapse
Affiliation(s)
- Zhaodi Che
- Department of Anesthesiology and Clinical Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Mingxiang Cai
- Clinical Research Platform for Interdiscipline of Stomatology, The First Affiliated Hospital of Jinan University, Department of Stomatology, College of Stomatology, Jinan University, Guangzhou 510630, China
| | - Xiaowu Dong
- Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225009, China
| | - Yuan Yuan
- Department of Anesthesiology and Clinical Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Aier School of Ophthalmology, Central South University, Changsha 410083, China
| | - Yaodong Wang
- Kunshan Hospital of Chinese Medicine, Kunshan Affiliated Hospital of Yangzhou University, Suzhou 215000, China
| | - Lu Xiao
- Department of Anesthesiology and Clinical Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yali Song
- Department of Anesthesiology and Clinical Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jiajun Zhong
- Department of Anesthesiology and Clinical Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Pingping Luo
- Department of Anesthesiology and Clinical Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Hao Wang
- Department of Anesthesiology and Clinical Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Guotao Lu
- Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225009, China.
| | - Yao Sun
- Department of Oral Implantology, School of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200092, China.
| | - Jia Xiao
- Department of Anesthesiology and Clinical Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao 266000, China.
| |
Collapse
|
96
|
Weng CC, Chiang CC, Chung YH, Ho YP, Chang YC, Hsiao IT, Mach RH. Evaluation of a simplified radiolabeling method for a PARP inhibitor in an animal model of breast cancer. EJNMMI Res 2025; 15:50. [PMID: 40301197 PMCID: PMC12040802 DOI: 10.1186/s13550-025-01236-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 04/05/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Several poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitors were recently approved by the US Food and Drug Administration for use in cancer treatment. To facilitate the discovery of novel PARP-targeting ligands, a radioiodinated ligand, I-125-KX1, was developed and validated for its specificity to PARP-1; however, its preparation procedure is time-consuming. The present study employed a solid-phase extraction (SPE) method in the radiolabeling procedure of I-123/125-KX1 and evaluated its binding specificity by using receptor binding assays, autoradiography, and in vivo single photon emission computed tomography (SPECT) imaging technique. RESULTS Through the incorporation of the SPE purification method as the final step in the radioiodination procedure, the resultant product I-123/125-KX1 exhibited high radiochemical purity (> 99%) and an acceptable radiochemical yield (58.6% for I-123-KX1, 73.3% for I-125-KX1). The binding characteristics of this radiotracer were validated through saturation binding assays conducted on MDA-MB-231 and MCF-7 cells. The Kd values obtained for the tracer (~ 1.0 nM) was consistent with values reported in the literature, and the Bmax values of these two cell lines (2017 ± 178 fmol/mg on MDA-MB-231 vs. 1393 ± 105 fmol/mg on MCF-7) were in line with the results from Western blot analyses. To demonstrate the in vivo imaging ability of I-123-KX1 prepared in this study, an MDA-MB-231 tumor animal model was used and the tracer displayed a suitable uptake on the tumor tissues (6.9 ± 0.8%ID/mL). The binding specificity of the SPE-purified I-125-KX1 was further verified using in vitro autoradiography in conjunction with various PARP inhibitors. Additionally, an anti-PARP-1 immunohistochemistry experiment was conducted, which revealed that the autoradiograms of the radiotracer displayed a similar pattern. CONCLUSIONS This suggests that the I-123/125-KX1 prepared using the SPE method showed some comparable properties to those from the traditional method, indicating its potential suitability for future radioligand preparation in PARP studies. However, further characterization studies may be needed to confirm its efficacy.
Collapse
Affiliation(s)
- Chi-Chang Weng
- Department of Medical Imaging and Radiological Sciences, College of Medicine & Healthy Aging Center, Chang Gung University, Taoyuan, Taiwan.
- Department of Medical Research and Development, Research Division, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan.
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, Taiwan.
| | - Chao-Chih Chiang
- Department of Isotope Application Research, National Atomic Research Institute, Taoyuan, Taiwan
| | - Yi-Hsiu Chung
- Department of Medical Research and Development, Research Division, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan
| | - Yi-Pei Ho
- Department of Medical Imaging and Radiological Sciences, College of Medicine & Healthy Aging Center, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Chuan Chang
- Department of Medical Imaging and Radiological Sciences, College of Medicine & Healthy Aging Center, Chang Gung University, Taoyuan, Taiwan
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Ing-Tsung Hsiao
- Department of Medical Imaging and Radiological Sciences, College of Medicine & Healthy Aging Center, Chang Gung University, Taoyuan, Taiwan
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Robert H Mach
- Division of Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
97
|
Umehara K, Parrott N, Günther A, Bogman K. Prospective DDI Risk Assessment of Vicasinabin with PBPK Modeling by Integrating In Vitro Data. Clin Pharmacol Ther 2025. [PMID: 40294080 DOI: 10.1002/cpt.3686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 04/07/2025] [Indexed: 04/30/2025]
Abstract
Vicasinabin is an oral cannabinoid receptor 2 (CB2) agonist showing anti-inflammatory effects and was developed for the treatment of chronic inflammatory diseases such as diabetic retinopathy. Vicasinabin is mainly metabolized by CYP3A4, with minor contributions from CYP2C19 and UGTs. The drug shows in vitro induction of CYP3A4, as well as inhibition of hepatic and renal transporters. Translation of in vitro data to a clinical drug-drug interaction (DDI) risk assessment has been challenging, with a potential role of CYP2C19 genotypes in the pharmacokinetics to be considered. A physiologically based pharmacokinetic (PBPK) model of vicasinabin based on a bottom-up approach predicted a moderate systemic exposure reduction for the selective CYP3A4 substrate midazolam. Neither the OATP1B1/P-gp/CYP3A4 inhibition effect on atorvastatin nor the OCT2/MATE1 inhibition effect on metformin was predicted to be of clinical relevance by PBPK modeling, as was confirmed by clinical DDI study data. After successful PBPK model prediction of itraconazole DDI using an in vitro fm,CYP3A4 of 0.6, the model was applied to simulate weak or moderate exposure changes of vicasinabin after co-administration with perpetrators for CYP3A4 and CYP2C19 (erythromycin, fluconazole, fluvoxamine, efavirenz, and rifampicin). A strong effect of induction due to rifampicin was also indicated. The CYP2C19 genotypes did not result in a significant impact on the victim DDI prediction for vicasinabin due to a low fm,CYP2C19 (∼0.2). The case study illustrated the usefulness of prospective PBPK predictions of clinical drug-drug interactions using in vitro data.
Collapse
Affiliation(s)
- Kenichi Umehara
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Neil Parrott
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Andreas Günther
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Katrijn Bogman
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
98
|
Arif M, Lehoczki A, Haskó G, Lohoff FW, Ungvari Z, Pacher P. Global and tissue-specific transcriptomic dysregulation in human aging: Pathways and predictive biomarkers. GeroScience 2025:10.1007/s11357-025-01672-z. [PMID: 40295347 DOI: 10.1007/s11357-025-01672-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Aging is a universal biological process that impacts all tissues, leading to functional decline and increased susceptibility to age-related diseases, particularly cardiometabolic disorders. While aging is characterized by hallmarks such as mitochondrial dysfunction, chronic inflammation, and dysregulated metabolism, the molecular mechanisms driving these processes remain incompletely understood, particularly in a tissue-specific context. To address this gap, we conducted a comprehensive transcriptomic analysis across 40 human tissues using data from the Genotype-Tissue Expression (GTEx) project, comparing individuals younger than 40 years with those older than 65 years. We identified over 17,000 differentially expressed genes (DEGs) across tissues, with distinct patterns of up- and down-regulation. Enrichment analyses revealed that up-regulated DEGs were associated with inflammation, immune responses, and apoptosis, while down-regulated DEGs were linked to mitochondrial function, oxidative phosphorylation, and metabolic processes. Using gene co-expression network (GCN) analyses, we identified 1,099 genes as dysregulated nodes (DNs) shared across tissues, reflecting global aging-associated transcriptional shifts. Integrating machine learning approaches, we pinpointed key aging biomarkers, including GDF15 and EDA2R, which demonstrated strong predictive power for aging and were particularly relevant in cardiometabolic tissues such as the heart, liver, skeletal muscle, and adipose tissue. These genes were also validated in plasma proteomics studies and exhibited significant correlations with clinical cardiometabolic health indicators. This study provides a multi-tissue, integrative perspective on aging, uncovering both systemic and tissue-specific molecular signatures. Our findings advance understanding of the molecular underpinnings of aging and identify novel biomarkers that may serve as therapeutic targets for promoting healthy aging and mitigating age-related diseases.
Collapse
Affiliation(s)
- Muhammad Arif
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute On Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
- Section On Fibrotic Disorders, National Institute and Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
- Department of Molecular and Clinical Medicine, SciLifeLab, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Andrea Lehoczki
- Doctoral College/Institute of Preventive Medicine and Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Falk W Lohoff
- Section On Clinical Genomics and Experimental Therapeutics, National Institute On Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute On Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
99
|
Cheng Z, Wang H, Zhang Y, Ren B, Fu Z, Li Z, Tu C. Deciphering the role of liquid-liquid phase separation in sarcoma: Implications for pathogenesis and treatment. Cancer Lett 2025; 616:217585. [PMID: 39999920 DOI: 10.1016/j.canlet.2025.217585] [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: 12/10/2024] [Revised: 02/04/2025] [Accepted: 02/21/2025] [Indexed: 02/27/2025]
Abstract
Liquid-liquid phase separation (LLPS) is a significant reversible and dynamic process in organisms. Cells form droplets that are distinct from membrane-bound cell organelles by phase separation to keep biochemical processes in order. Nevertheless, the pathological state of LLPS contributes to the progression of a variety of tumor-related pathogenic issues. Sarcoma is one kind of highly malignant tumor characterized by aggressive metastatic potential and resistance to conventional therapeutic agents. Despite the significant clinical relevance, research on phase separation in sarcomas currently faces several major challenges. These include the limited availability of sarcoma samples, insufficient attention from the research community, and the complex genetic heterogeneity of sarcomas. Recently, emerging evidence have elaborated the specific effects and pathways of phase separation on different sarcoma subtypes, including the effect of sarcoma fusion proteins and other physicochemical factors on phase separation. This review aims to summarize the multiple roles of phase separation in sarcoma and novel molecular inhibitors that target phase separation. These insights will broaden the understanding of the mechanisms concerning sarcoma and offer new perspectives for future therapeutic strategies.
Collapse
Affiliation(s)
- Zehao Cheng
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Xiangya School of Medicine, Central South University, Changsha, Hunan, 410011, China
| | - Hua Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Yibo Zhang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Xiangya School of Medicine, Central South University, Changsha, Hunan, 410011, China
| | - Bolin Ren
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Zheng Fu
- Shanghai Xinyi Biomedical Technology Co., Ltd, Shanghai, 201306, China
| | - Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Changsha Medical University, Changsha, Hunan, 410219, China.
| |
Collapse
|
100
|
Cao P, Jaeschke H, Ni HM, Ding WX. The Ways to Die: Cell Death in Liver Pathophysiology. Semin Liver Dis 2025. [PMID: 40199509 DOI: 10.1055/a-2576-4332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Liver diseases are closely associated with various cell death mechanisms, including apoptosis, necroptosis, autophagy, pyroptosis, and ferroptosis. Each process contributes uniquely to the pathophysiology of liver injury and repair. Importantly, these mechanisms are not limited to hepatocytes; they also significantly involve nonparenchymal cells. This review examines the molecular pathways and regulatory mechanisms underlying these forms of cell death in hepatocytes, emphasizing their roles in several liver diseases, such as ischemia-reperfusion injury, metabolic dysfunction-associated steatotic liver disease, drug-induced liver injury, and alcohol-associated liver disease. Recent insights into ferroptosis and pyroptosis may reveal novel therapeutic targets for managing liver diseases. This review aims to provide a comprehensive overview of these cell death mechanisms in the context of liver diseases, detailing their molecular signaling pathways and implications for potential treatment strategies.
Collapse
Affiliation(s)
- Peng Cao
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
- Division of Gastroenterology, Hepatology and Mobility, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|