1
|
Roy S, Deka D, Kondaveeti SB, Ayyadurai P, Siripragada S, Philip N, Pathak S, Duttaroy AK, Banerjee A. An overview of potential of natural compounds to regulate epigenetic modifications in colorectal cancer: a recent update. Epigenetics 2025; 20:2491316. [PMID: 40239010 PMCID: PMC12005453 DOI: 10.1080/15592294.2025.2491316] [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: 12/30/2024] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
Colorectal cancer (CRC) remains an alarming global health concern despite advancements in treatment modalities over recent decades. Among the various factors contributing to CRC, this review emphasizes the critical role of epigenetic mechanisms in its pathogenesis and progression. This review also describes the potential role of natural compounds in altering the epigenetic landscape, focused mainly on DNA methylation, histone modification, and non-coding RNAs. Publications from the previous five years were searched and retrieved using well-known search engines and databases like PubMed, Google Scholar, and ScienceDirect. Keywords like CRC/colorectal cancer, CAC/Colitis associated CRC, inflammasomes, epigenetic modulation, genistein, curcumin, quercetin, resveratrol, anthocyanins, sulforaphane, and epigallocatechin-3-gallate were used in various combinations during the search. These natural compounds predominantly affect pathways such as Wnt/β-catenin, NF-κB, and PI3K/AKT to suppress CRC cell proliferation and oxidative stress and enhance anti-inflammation and apoptosis. However, their clinical use is restricted due to their low bioavailability. However, multiple methods exist to overcome challenges like this, including but not limited to structural modifications, nanoparticle encapsulations, bio-enhancers, and novel advanced delivery systems. These methods improve their potential as supportive therapies that target CRC progression epigenetically with fewer side effects. Current research focuses on enhancing epigenetic targeting to control CRC progression while minimizing side effects, emphasizing improved specificity, bioavailability, and efficacy as standalone or synergistic therapies.
Collapse
Affiliation(s)
- Susmita Roy
- Medical Biotechnology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Dikshita Deka
- Medical Biotechnology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Suresh Babu Kondaveeti
- Department of Biochemistry, Symbiosis Medical College for Women, Symbiosis International (Deemed University), Pune, India
| | - Pavithra Ayyadurai
- Medical Biotechnology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Sravani Siripragada
- Medical Biotechnology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Neha Philip
- Medical Biotechnology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Surajit Pathak
- Medical Biotechnology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Antara Banerjee
- Medical Biotechnology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| |
Collapse
|
2
|
Takyi E, Nirmalkar K, Adams J, Krajmalnik-Brown R. Interventions targeting the gut microbiota and their possible effect on gastrointestinal and neurobehavioral symptoms in autism spectrum disorder. Gut Microbes 2025; 17:2499580. [PMID: 40376856 PMCID: PMC12087657 DOI: 10.1080/19490976.2025.2499580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 05/18/2025] Open
Abstract
Autism spectrum disorder (ASD) is a developmental disorder that is characterized by deficits in social communication and restricted, repetitive, and stereotyped behaviors. In addition to neurobehavioral symptoms, children with ASD often have gastrointestinal symptoms (e.g. constipation, diarrhea, gas, abdominal pain, reflux). Several studies have proposed the role of gut microbiota and metabolic disorders in gastrointestinal symptoms and neurodevelopmental dysfunction in ASD patients; these results offer promising avenues for novel treatments of this disorder. Interventions targeting the gut microbiota - such as fecal microbiota transplant (FMT), microbiota transplant therapy (MTT), probiotics, prebiotics, synbiotics, antibiotics, antifungals, and diet - promise to improve gut health and can potentially improve neurological symptoms. The modulation of the gut microbiota using MTT in ASD has shown beneficial and long-term effects on GI symptoms and core symptoms of autism. Also, the modulation of the gut microbiota to resemble that of typically developing individuals seems to be the most promising intervention. As most of the studies carried out with MTT are open-label studies, more extensive double-blinded randomized control trials are needed to confirm the efficacy of MTT as a therapeutic option for ASD. This review examines the current clinical research evidence for the use of interventions that target the microbiome - such as antibiotics, antifungals, probiotics/prebiotics, synbiotics, and MTT - and their effectiveness in changing the gut microbiota and improving gastrointestinal and neurobehavioral symptoms in ASD.
Collapse
Affiliation(s)
- Evelyn Takyi
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
| | - Khemlal Nirmalkar
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
| | - James Adams
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
- School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, USA
| | - Rosa Krajmalnik-Brown
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
- School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
3
|
Long Z, Ge C, Zhao Y, Liu Y, Zeng Q, Tang Q, Dong Z, He G. Enhanced autophagic clearance of amyloid-β via histone deacetylase 6-mediated V-ATPase assembly and lysosomal acidification protects against Alzheimer's disease in vitro and in vivo. Neural Regen Res 2025; 20:2633-2644. [PMID: 38993141 PMCID: PMC11801289 DOI: 10.4103/nrr.nrr-d-23-01633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/05/2024] [Accepted: 03/29/2024] [Indexed: 07/13/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202509000-00025/figure1/v/2024-11-05T132919Z/r/image-tiff Recent studies have suggested that abnormal acidification of lysosomes induces autophagic accumulation of amyloid-β in neurons, which is a key step in senile plaque formation. Therefore, restoring normal lysosomal function and rebalancing lysosomal acidification in neurons in the brain may be a new treatment strategy for Alzheimer's disease. Microtubule acetylation/deacetylation plays a central role in lysosomal acidification. Here, we show that inhibiting the classic microtubule deacetylase histone deacetylase 6 with an histone deacetylase 6 shRNA or thehistone deacetylase 6 inhibitor valproic acid promoted lysosomal reacidification by modulating V-ATPase assembly in Alzheimer's disease. Furthermore, we found that treatment with valproic acid markedly enhanced autophagy, promoted clearance of amyloid-β aggregates, and ameliorated cognitive deficits in a mouse model of Alzheimer's disease. Our findings demonstrate a previously unknown neuroprotective mechanism in Alzheimer's disease, in which histone deacetylase 6 inhibition by valproic acid increases V-ATPase assembly and lysosomal acidification.
Collapse
Affiliation(s)
- Zhimin Long
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Chuanhua Ge
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Yueyang Zhao
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Yuanjie Liu
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Qinghua Zeng
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Qing Tang
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Physiology, Chongqing Medical University, Chongqing, China
| | - Zhifang Dong
- Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Guiqiong He
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
Yu Z, Guo Y, Chen H, Wan W, Hu M, Li Y, Wei T, Chen Q. A phloem-limited unculturable bacterium induces mild xenophagy in insect vectors for persistent infection. Microbiol Res 2025; 297:128186. [PMID: 40262357 DOI: 10.1016/j.micres.2025.128186] [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: 01/17/2025] [Revised: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
Xenophagy is an important antibacterial defense mechanism that many organisms use to engulf intracellular pathogens. However, the mechanisms of xenophagy triggered by insect-borne plant bacteria are not well understood. Candidatus Liberibacter asiaticus (CLas) causes Huanglongbing, which poses a serious threat to citrus production. CLas is a phloem-limited unculturable bacterium that is transmitted by the Asian citrus psyllid in a persistent and propagative manner in nature. Here, we found that CLas infection in the gut of psyllids triggered a mild and anti-bacterial xenophagy. Xenophagy limited excessive propagation of CLas to maintain psyllid survival, because overload of CLas was detrimental to psyllid life. Furthermore, the outer membrane β-barrel protein (OMBB) of CLas is the key secreted protein that induces xenophagy in psyllids by interacting with ATG8 and ATG14. OMBB can independently induce autophagy in psyllid and non-host cells. Together, these results revealed that an insect-borne plant bacterium activates mild xenophagy to control its propagation, thereby achieving persistent infection in insect vectors.
Collapse
Affiliation(s)
- Zhongkai Yu
- State Key Laboratory of Agricultural and Forestry Biosecurity, Fujian Agriculture and Forestry University, Fujian, Fuzhou, China
| | - Yuxin Guo
- State Key Laboratory of Agricultural and Forestry Biosecurity, Fujian Agriculture and Forestry University, Fujian, Fuzhou, China
| | - Hongyan Chen
- State Key Laboratory of Agricultural and Forestry Biosecurity, Fujian Agriculture and Forestry University, Fujian, Fuzhou, China
| | - Wenqiang Wan
- State Key Laboratory of Agricultural and Forestry Biosecurity, Fujian Agriculture and Forestry University, Fujian, Fuzhou, China
| | - Mengting Hu
- State Key Laboratory of Agricultural and Forestry Biosecurity, Fujian Agriculture and Forestry University, Fujian, Fuzhou, China
| | - You Li
- State Key Laboratory of Agricultural and Forestry Biosecurity, Fujian Agriculture and Forestry University, Fujian, Fuzhou, China
| | - Taiyun Wei
- State Key Laboratory of Agricultural and Forestry Biosecurity, Fujian Agriculture and Forestry University, Fujian, Fuzhou, China.
| | - Qian Chen
- State Key Laboratory of Agricultural and Forestry Biosecurity, Fujian Agriculture and Forestry University, Fujian, Fuzhou, China.
| |
Collapse
|
5
|
Uppar PM, Kim NY, Harish KK, Beeraka NM, Gaonkar SL, Madegowda M, Sethi G, Rangappa KS, Nikolenko VN, Chinnathambi A, Alharbi SA, Ahn KS, Basappa B. Targeting breast cancer cells with 2-indolyl-1,3,4-oxadiazole compounds by inducing apoptosis, paraptosis and autophagy. Chem Biol Interact 2025; 415:111528. [PMID: 40288434 DOI: 10.1016/j.cbi.2025.111528] [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: 12/06/2024] [Revised: 04/09/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
While 2-Indolyl-1,3,4-oxadiazole derivatives are recognized for their antibacterial properties, their potential as anticancer agents remains underexplored. This study investigates the anti-breast cancer properties of a novel 2-Indolyl-1,3,4-oxadiazole compound, 5l, focusing on its ability to induce apoptosis, paraptosis, and autophagy, and targeting poly (ADP-ribose) polymerase (PARP1), a critical enzyme in DNA repair. A series of 1,3,4-oxadiazole derivatives (compounds 5a-5m) were synthesized using an optimized multi-step process, enhancing reaction efficiency and yield. In silico molecular docking was used to determine binding efficacy of these derivatives. Lead compound, 5l, underwent cytotoxicity assays against MDA-MB-231, MCF-7, BT-474, and SK-BR-3 breast cancer cell lines, as well as the non-cancerous MCF-10A cell line. Molecular docking assessed the interaction of 5l with the PARP1 active site. Frontier molecular orbital (FMO) and molecular electrostatic potential (MESP) analyses were conducted to map electron distribution and identify reactive regions within compound 5l. The effects of 5l on cellular processes such as apoptosis, autophagy, and endoplasmic reticulum (ER) integrity were evaluated using live and dead assays, Annexin V staining, ER-tracker dye staining, and acridine orange assays. Western blotting analyzed apoptosis, paraptosis, and autophagy-related genomic instability. The optimized synthesis yielded high-purity 1,3,4-oxadiazole derivatives. Compound 5l displayed significant anticancer activity, with IC50 values of 63.7 μM, 29.1 μM, 50.3 μM, and 39.8 μM for MDA-MB-231, MCF-7, BT-474, and SK-BR-3 cell lines respectively, demonstrating its cytotoxic efficacy. Molecular docking revealed that 5l binds to PARP1 active site with a binding energy of -11.7 kcal/mol, indicating a strong interaction supporting its role as a PARP1 inhibitor. Annexin V assays, ER-tracker dye staining, and Acridine orange assays were used to assess apoptosis, ER integrity, and autophagy. 5l induced upregulation of cleaved PARP and downregulation of Alix-loaded proteins, alongside increased LC3-II expression, indicating autophagy-mediated genomic instability. Compound 5l exhibits potent anti-breast cancer activity through paraptosis, apoptosis, and autophagy-mediated genomic instability and by PARP1 inhibition with typically a low IC50 values, highlighting its potential as a therapeutic agent.
Collapse
Affiliation(s)
- Pradeep M Uppar
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore, 570006, India; Sri Sri Shivalingeshwara Swamy Govt. First Grade College, Channagiri, 577213, India.
| | - Na Young Kim
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| | - Keshav Kumar Harish
- Department of Studies in Physics, University of Mysore, Manasagangotri, Mysore 570006, India.
| | - Narasimha M Beeraka
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Anantapuramu, Chiyyedu, Andhra Pradesh, 515721, India; Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow, 119991, Russia.
| | - Santhosh L Gaonkar
- Department of Chemistry, Manipal Institute of Technology, Manipal University, Manipal, 576104, India.
| | - Mahendra Madegowda
- Department of Studies in Physics, University of Mysore, Manasagangotri, Mysore 570006, India.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600.
| | | | - Vladimir N Nikolenko
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow, 119991, Russia.
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, PO Box -2455, Riyadh, 11451, Saudi Arabia.
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, PO Box -2455, Riyadh, 11451, Saudi Arabia.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| | - Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore, 570006, India.
| |
Collapse
|
6
|
Wang JC, Zhao L, Yu XY, Wu TP, Xia CF, Shi JF, He H, Chen ZQ, Shi D, Xue H, Ao Q, Liao SP, Zheng ZQ, Huang QF, Li L, Lin SL, Li YX, Hu WL, Peng J, Lei L, Cao MM, Yang F, Yan XX, He SY, Cao MD, Zhang SL, Teng Y, Li QR, Tan NP, Yu HY, Cheng HH, Wang XM, Wu WQ, Chen WQ. Diagnostic efficacy of fecal-based miR-92a for advanced colorectal neoplasia: a prospective multicenter screening trial. Mil Med Res 2025; 12:30. [PMID: 40506768 PMCID: PMC12164163 DOI: 10.1186/s40779-025-00613-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/29/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND More efficacious, noninvasive screening methods are needed for advanced colorectal neoplasia. miR-92a is a reliable and reproducible biomarker for early colorectal cancer detection in stool samples. We compared the diagnostic efficacies of miR-92a, immunochemical fecal occult blood testing (FIT), and their combination (FIT + miR-92a) in a prospective multicenter screening trial. METHODS Overall, 16,240 participants aged 30-75 years were enrolled between April 1, 2021, and December 31, 2023. A total of 15,586 participants returned samples available for both FIT and miR-92a tests. All those with positive, and a random selection of those with negative screening tests were recommended to undergo colonoscopy. Follow-ups were performed until participants completed the colonoscopic examination. A total of 1401 screen-positive and 2079 randomly selected screen-negative individuals completed colonoscopies. Primary outcomes included sensitivity, number needed to screen (NNS), Youden index and receiver operating characteristic area under the curve (AUC) for advanced adenomas and colorectal cancer [advanced neoplasia (AN)] for each screening modality in the diagnostic performance analysis. RESULTS Colonoscopy was performed in 3480 individuals. The colonoscopy compliance rate was 47.8% for screen-positive individuals. The sensitivity of miR-92a versus FIT for AN was 70.9% versus 54.3% (P < 0.001), NNS was 24.7 versus 32.2 (P = 0.001), Youden index was 47.9% versus 35.0% (P < 0.001), AUC was 0.74 versus 0.67 (P = 0.010). FIT + miR-92a had a sensitivity of 85.4%, an NNS of 20.5, a Youden index of 47.9% and an AUC of 0.74 for AN. CONCLUSIONS For AN screening, miR-92a demonstrated better sensitivity, NNS, Youden index and AUC as compared with FIT. Compared with FIT, using miR-92a appears to be more efficient for population-based screening programs. Screening sensitivity for AN can be further enhanced if conditionally used in combination with FIT. TRIAL REGISTRATION Chinese Clinical Trial Registration Number: ChiCTR2200065415.
Collapse
Affiliation(s)
- Jia-Chen Wang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Li Zhao
- Department of Health Management, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University/the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518053, Guangdong, China
| | - Xiang-Yang Yu
- Department of Gastrointestinal Surgery, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300000, China
| | - Ting-Ping Wu
- Department of Gastroenterology, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, 518020, Guangdong, China
| | - Chang-Fa Xia
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ju-Fang Shi
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hui He
- Department of Health Management, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University/the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518053, Guangdong, China
| | - Zhi-Qi Chen
- Department of Nursing, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300000, China
| | - Dan Shi
- Department of Gastrointestinal Surgery, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300000, China
| | - Han Xue
- Department of Health Management, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University/the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518053, Guangdong, China
| | - Qi Ao
- Department of Gastrointestinal Surgery, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300000, China
| | - Shu-Ping Liao
- Department of Health Management, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University/the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518053, Guangdong, China
| | - Zhang-Qiang Zheng
- Department of Gastrointestinal Surgery, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300000, China
| | - Qiong-Fang Huang
- Department of Health Management, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University/the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518053, Guangdong, China
| | - Lin Li
- Department of Gastrointestinal Surgery, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300000, China
| | - Sui-Ling Lin
- Department of Gastroenterology, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, 518020, Guangdong, China
| | - Ying-Xue Li
- Department of Gastroenterology, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University/the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518053, Guangdong, China
| | - Wen-Long Hu
- Clinical Medical Research Center, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University/the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518053, Guangdong, China
| | - Ji Peng
- Shenzhen Center for Chronic Disease Control, Shenzhen, 518081, Guangdong, China
| | - Lin Lei
- Shenzhen Center for Chronic Disease Control, Shenzhen, 518081, Guangdong, China
| | - Mao-Mao Cao
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Fan Yang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xin-Xin Yan
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Si-Yi He
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Meng-Di Cao
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shao-Li Zhang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yi Teng
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qian-Ru Li
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Nuo-Pei Tan
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hao-Yang Yu
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518000, Guangdong, China
| | - Hong-Hui Cheng
- Department of Gastroenterology, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, 518020, Guangdong, China.
| | - Xi-Mo Wang
- Tianjin Third Central Hospital, Tianjin Medical University, Tianjin, 300192, China.
- Tianjin Third Central Hospital, Nankai University, Tianjin, 300192, China.
- Tianjin Third Central Hospital, Tianjin University, Tianjin, 300192, China.
| | - Wei-Qing Wu
- Department of Health Management, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University/the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518053, Guangdong, China.
| | - Wan-Qing Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
7
|
Huang YH, Lin Q, Jin XY, Chou CY, Wei JJ, Xing J, Guo HM, Liu ZF, Lu Y. Classification of pediatric video capsule endoscopy images for small bowel abnormalities using deep learning models. World J Gastroenterol 2025; 31:107601. [DOI: 10.3748/wjg.v31.i21.107601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Revised: 04/14/2025] [Accepted: 05/19/2025] [Indexed: 06/06/2025] Open
Abstract
BACKGROUND Video capsule endoscopy (VCE) is a noninvasive technique used to examine small bowel abnormalities in both adults and children. However, manual review of VCE images is time-consuming and labor-intensive, making it crucial to develop deep learning methods to assist in image analysis.
AIM To employ deep learning models for the automatic classification of small bowel lesions using pediatric VCE images.
METHODS We retrospectively analyzed VCE images from 162 pediatric patients who underwent VCE between January 2021 and December 2023 at the Children's Hospital of Nanjing Medical University. A total of 2298 high-resolution images were extracted, including normal mucosa and lesions (erosions/erythema, ulcers, and polyps). The images were split into training and test datasets in a 4:1 ratio. Four deep learning models: DenseNet121, Visual geometry group-16, ResNet50, and vision transformer were trained using 5-fold cross-validation, with hyperparameters adjusted for optimal classification performance. The models were evaluated based on accuracy, precision, recall, F1-score, and area under the receiver operating curve (AU-ROC). Lesion visualization was performed using gradient-weighted class activation mapping.
RESULTS Abdominal pain was the most common indication for VCE, accounting for 62% of cases, followed by diarrhea, vomiting, and gastrointestinal bleeding. Abnormal lesions were detected in 93 children, with 38 diagnosed with inflammatory bowel disease. Among the deep learning models, DenseNet121 and ResNet50 demonstrated excellent classification performance, achieving accuracies of 90.6% [95% confidence interval (CI): 89.2-92.0] and 90.5% (95%CI: 89.9-91.2), respectively. The AU-ROC values for these models were 93.7% (95%CI: 92.9-94.5) for DenseNet121 and 93.4% (95%CI: 93.1-93.8) for ResNet50.
CONCLUSION Our deep learning-based diagnostic tool developed in this study effectively classified lesions in pediatric VCE images, contributing to more accurate diagnoses and increased diagnostic efficiency.
Collapse
Affiliation(s)
- Yi-Hsuan Huang
- Department of Gastroenterology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Qian Lin
- Department of Gastroenterology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Xin-Yan Jin
- School of Electronic Science and Engineering, Nanjing University, Nanjing 210023, Jiangsu Province, China
| | - Chih-Yi Chou
- College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Jia-Jie Wei
- Department of Gastroenterology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Jiao Xing
- Department of Gastroenterology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Hong-Mei Guo
- Department of Gastroenterology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Zhi-Feng Liu
- Department of Gastroenterology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Yan Lu
- Department of Gastroenterology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| |
Collapse
|
8
|
Ali A, Azmat U, Khatoon A, Akbar K, Murtaza B, Ji Z, Irshad U, Su Z. From gene editing to tumor eradication: The CRISPR revolution in cancer therapy. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 196:114-131. [PMID: 40250571 DOI: 10.1016/j.pbiomolbio.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
Cancer continues to be a significant worldwide health concern, characterized by high rates of occurrence and death. Unfortunately, existing treatments frequently fall short of delivering satisfying therapeutic outcomes. Immunotherapy has ushered in a new era in the treatment of solid tumors, yet its effectiveness is still constrained and comes with unwanted side effects. The advancement of cutting-edge technology, propelled by gene analysis and manipulation at the molecular scale, shows potential for enhancing these therapies. The advent of genome editing technologies, including CRISPR-Cas9, can greatly augment the efficacy of cancer immunotherapy. This review explores the mechanism of CRISPR-Cas9-mediated genome editing and its wide range of tools. The study focuses on analyzing the effects of CRISPR-induced double-strand breaks (DSBs) on cancer immunotherapy, specifically by gene knockdown or knockin. In addition, the study emphasizes the utilization of CRISPR-Cas9-based genome-wide screening to identify targets, the potential of spatial CRISPR genomics, and the extensive applications and difficulties of CRISPR-Cas9 in fundamental research, translational medicine, and clinical environments.
Collapse
Affiliation(s)
- Ashiq Ali
- Department of Histology and Embryology, Shantou University Medical College, Shantou, China.
| | - Urooj Azmat
- Department of Zoology, Wildlife and Fisheries, Faculty of Sciences, University of Agriculture, Faisalabad, 38040, Punjab, Pakistan
| | - Aisha Khatoon
- Department of Pathology, University of Agriculture, Faisalabad, Pakistan
| | - Kaynaat Akbar
- Department of Zoology, Wildlife and Fisheries, Faculty of Sciences, University of Agriculture, Faisalabad, 38040, Punjab, Pakistan
| | - Bilal Murtaza
- School of Bioengineering, Dalian University of Science and Technology Dalian, China
| | - Ziyi Ji
- Department of Histology and Embryology, Shantou University Medical College, Shantou, China
| | - Urooj Irshad
- Department of Zoology, Faculty of Sciences, Superior University, Lahore, Pakistan
| | - Zhongjing Su
- Department of Histology and Embryology, Shantou University Medical College, Shantou, China.
| |
Collapse
|
9
|
Ye CF, Wu JD, Li LR, Sun SG, Wang YG, Jiang TA, Long X, Zhao J. Co-inhibition of RAGE and TLR4 sensitizes pancreatic cancer to irreversible electroporation in mice by disrupting autophagy. Acta Pharmacol Sin 2025; 46:1757-1771. [PMID: 39953172 PMCID: PMC12098883 DOI: 10.1038/s41401-025-01487-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/16/2025] [Indexed: 02/17/2025]
Abstract
Irreversible electroporation (IRE) is a local ablative treatment for patients with pancreatic cancer. During the IRE procedure, high-intensity electric pulses are released intratumorally to disrupt plasma membranes and induce cell death. Since the intensity of the pulsed electric field (PEF) can be decreased by the tumor microenvironment, some cancer cells are subjected to a sublethal PEF and may survive to cause tumor recurrence later. Autophagy activation induced by anticancer therapies is known to promote treatment resistance. In this study, we investigated whether autophagy is activated in residual cancer cells after IRE and assessed the roles it plays during tumor recurrence. Subcutaneous KPC-A548 or Panc02 murine pancreatic cancer cell line xenograft mouse models were established; once the tumors reached 7 mm in one dimension, the tumor-bearing mice were subjected to IRE. For in vitro sublethal PEF treatment, the pancreatic cancer cell suspension was in direct contact with the electrodes and pulsed at room temperature. We showed that autophagy was activated in surviving residual cells, as evidenced by increased expression of LC3 and p62. Suppression of autophagy with hydroxychloroquine (60 mg/kg, daily intraperitoneal injection) markedly increased the efficacy of IRE. We demonstrated that autophagy activation can be attributed to increased expression of high-mobility group box 1 (HMGB1); co-inhibition of two HMGB1 receptors, receptor for advanced glycosylation end products (RAGE) and Toll-like receptor 4 (TLR4), suppressed autophagy activation by upregulating the PI3K/AKT/p70 ribosomal S6 protein kinase (p70S6K) axis and sensitized pancreatic cancer cells to PEF. We prepared a polymeric micelle formulation (M-R/T) encapsulating inhibitors of both RAGE and TLR4. The combination of IRE and M-R/T (equivalent to RAGE inhibitor at 10.4 mg/kg and TLR4 inhibitor at 5.7 mg/kg, intravenous or intraperitoneal injection every other day) significantly promoted tumor apoptosis, suppressed cell cycle progression, and prolonged animal survival in pancreatic tumor models. This study suggests that disruption of HMGB1-mediated autophagy with nanomedicine is a promising strategy to enhance the response of pancreatic cancer to IRE.
Collapse
Affiliation(s)
- Cui-Fang Ye
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia-di Wu
- Department of Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lin-Rong Li
- Department of Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shu-Guo Sun
- Department of Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu-Gang Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tian-An Jiang
- Department of Ultrasound Medicine, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Xin Long
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Jun Zhao
- Department of Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Nuclear Medicine and PET, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
10
|
Xiang Y, Liu Q, Liu K, Chen L, Chen F, Li T, Li S, Yu Q, Lv Q, Xiang Z. An exosome-based nanoplatform for siRNA delivery combined with starvation therapy promotes tumor cell death through autophagy, overcoming refractory KRAS-mutated tumors and restoring cetuximab chemosensitivity. Mater Today Bio 2025; 32:101732. [PMID: 40290881 PMCID: PMC12022660 DOI: 10.1016/j.mtbio.2025.101732] [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: 01/19/2025] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/30/2025] Open
Abstract
Multi-drug combination therapy is one of the most effective strategies for the treatment of drug-resistant and advanced tumors. Modern nanodrug delivery systems are crucial for multi-drug combination therapy and gene therapy. However, research on direct injection of RNAi has not yielded significant results. Artificial vectors are emerging as promising delivery systemts for RNA for gene therapy. In this study, a multi-drug therapy system was built based on a biodegradable exosome nano-platform exploiting the protective and low immunogenic properties of exosomes for RNA. This work aimed to accomplish the co-delivery of siRNA and 3-Bromopyruvic acid (3BP) on an exosome nanoplatform, enhancing targeting by coupling cetuximab (CTX) to exosome membranes, resulting in a new nanomedicine Exo@siRNA/3BP-CTX (ERBC) engineered exosomes. The synthesis conditions were optimized to obtain stable, safe, and effective nanomedicines. Successful targeting of tumors with CTX inhibited KRAS oncogene expression and significantly reduced glucose uptake by cancer cells. This enhanced the starvation therapy effect of the energy deprivation agent 3BP, thus promoting excessive autophagy activation in cells and doubling apoptosis. However, ERBC combined with CTX therapy restored cellular chemosensitivity to CTX. These findings indicate that engineered exosomes with dual therapeutic activities is a promising approach for treating refractory KRAS-mutant cancers.
Collapse
Affiliation(s)
- Yurong Xiang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, 400000, Chongqing, China
- Chongqing Key Laboratory of Department of General Surgery, The First Afiliated Hospital of Chongqing Medical University, No.1 Youyi Road, 400000, Chongqing, China
| | - Qiang Liu
- Department of Hepatobiliary Surgery, Suining First People's Hospital, 22 Youfang Street, 629000, Suining, China
| | - Kang Liu
- Department of Cardiovascular Surgery, Fuwai Yunnan Cardiovascular Hospital, 528 Shahe North Road, 400042, Kunming, China
| | - Liuxian Chen
- Department of Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, No.1 Medical College Road, 400016, Chongqing, China
| | - Fengjiao Chen
- Center for Clinical Molecular Medical Detection and Biobank, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, 400000, Chongqing, China
| | - Tao Li
- Department of Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, No.1 Medical College Road, 400016, Chongqing, China
| | - Siqi Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, 400000, Chongqing, China
- Chongqing Key Laboratory of Department of General Surgery, The First Afiliated Hospital of Chongqing Medical University, No.1 Youyi Road, 400000, Chongqing, China
| | - Qiang Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, 400000, Chongqing, China
- Chongqing Key Laboratory of Department of General Surgery, The First Afiliated Hospital of Chongqing Medical University, No.1 Youyi Road, 400000, Chongqing, China
| | - Quan Lv
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, 400000, Chongqing, China
- Chongqing Key Laboratory of Department of General Surgery, The First Afiliated Hospital of Chongqing Medical University, No.1 Youyi Road, 400000, Chongqing, China
| | - Zheng Xiang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, 400000, Chongqing, China
- Chongqing Key Laboratory of Department of General Surgery, The First Afiliated Hospital of Chongqing Medical University, No.1 Youyi Road, 400000, Chongqing, China
| |
Collapse
|
11
|
Gupta T, Murtaza M. Advancing targeted therapies in pancreatic cancer: Leveraging molecular abberrations for therapeutic success. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 196:19-32. [PMID: 39988056 DOI: 10.1016/j.pbiomolbio.2025.02.003] [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: 11/10/2024] [Revised: 02/03/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Pancreatic cancer is one of the most deadly with poor prognosis and overall survival rate due to the dense stroma in the tumors which often is challenging for the delivery of drug to penetrate deep inside the tumor bed and usually results in the progression of cancer. The conventional treatment such as chemotherapy, radiotherapy or surgery shows a minimal benefit in the survival due to the drug resistance, poor penetration, less radiosensitivity or recurrence of tumor. There is an urgent demand to develop molecular-level targeted therapies to achieve therapeutic efficacy in the pancreatic ductal adenocarcinoma (PDAC) patients. The precision oncology focuses on the unique attributes of the patient such as epigenome, proteome, genome, microbiome, lifestyle and diet habits which contributes to promote oncogenesis. The targeted therapy helps to target the mutated proteins responsible for controlling growth, division and metastasis of tumor in the cancer cells. It is very important to consider all the attributes of the patient to provide the suitable personalized treatment to avoid any severe side effects. In this review, we have laid emphasis on the precision medicine; the utmost priority is to improve the survival of cancer patients by targeting molecular mutations through transmembrane proteins, inhibitors, signaling pathways, immunotherapy, gene therapy or the use of nanocarriers for the delivery at the tumor site. It will become beneficial therapeutic window to be considered for the advanced stage pancreatic cancer patients to prolong their survival rate.
Collapse
Affiliation(s)
- Tanvi Gupta
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan.
| | - Mohd Murtaza
- Fermentation & Microbial Biotechnology Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180016, India.
| |
Collapse
|
12
|
Saeed A, Yousuf S, Noverati N, Chen A, Saleem N, Farooq U, Muniraj T, Persaud A, Xiao Y, Kumar A, Schlachterman A, Kowalski T, Adler D, Kamal F. Cold- versus hot-snare endoscopic mucosal resection of colorectal polyps: meta-analysis of randomized controlled trials. Gastrointest Endosc 2025; 101:1239-1243.e2. [PMID: 39725332 DOI: 10.1016/j.gie.2024.12.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND AND AIMS Interest in cold endoscopic mucosal resection (EMR) for colorectal polyps has been growing. We conducted a meta-analysis of randomized controlled trials to compare cold and hot EMR for colorectal polyps. METHODS We reviewed several databases from inception to October 6, 2024. Outcomes of interest were recurrent or residual neoplasia, en-bloc resection, incomplete resection, perforation, and intraprocedural and delayed bleeding. Pooled risk ratios (RRs) with 95% confidence intervals (CIs) were calculated by means of a random-effects model. RESULTS Rate of recurrent or residual neoplasia was significantly higher in the cold EMR group: RR, 2.03 (95% CI, 1.19-3.48). Rates of delayed bleeding RR, 0.42 (95% CI, 0.21-0.86) and perforation RR, 0.13 (95% CI, 0.03-0.59) were significantly lower with cold EMR. We found no significant difference in other outcomes between groups. CONCLUSIONS Cold EMR is associated with lower risk of delayed bleeding and perforation but higher risk of recurrent or residual neoplasia compared with hot EMR.
Collapse
Affiliation(s)
- Aamir Saeed
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Saira Yousuf
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Nicholas Noverati
- Division of Gastroenterology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Anna Chen
- Division of Gastroenterology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Nasir Saleem
- Division of Gastroenterology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Umer Farooq
- Division of Gastroenterology, St Louis University School of Medicine, St Louis, Missouri
| | - Thiruvengadam Muniraj
- Division of Gastroenterology, Yale University School of Medicine, New Haven, Connecticut
| | - Alana Persaud
- Division of Gastroenterology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Yasi Xiao
- Division of Gastroenterology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Anand Kumar
- Division of Gastroenterology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Alexander Schlachterman
- Division of Gastroenterology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Thomas Kowalski
- Division of Gastroenterology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Douglas Adler
- Center for Advanced Therapeutic Endoscopy, Porter Adventist Hospital, Denver, Colorado, USA
| | - Faisal Kamal
- Division of Gastroenterology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania.
| |
Collapse
|
13
|
Navidifar T, Meftah E, Baghsheikhi H, Kazemzadeh K, Karimi H, Rezaei N. Dual role of hepcidin in response to pathogens. Microb Pathog 2025; 203:107496. [PMID: 40118299 DOI: 10.1016/j.micpath.2025.107496] [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: 05/26/2024] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
Hepcidin is the primary regulator of vertebrate iron homeostasis. Its production is stimulated by systemic iron levels and inflammatory signals. Although the role of hepcidin in iron homeostasis is well characterized, its response to pathogenic agents is complex and diverse. In this review, we examine studies that investigate the role of hepcidin in response to infectious agents. Interleukin-6 (IL-6) is a key factor responsible for the induction of hepcidin expression. During infection, hepcidin-mediated depletion of extracellular iron serves as a protective mechanism against a variety of pathogens. However, accumulation of iron in macrophages through hepcidin-mediated pathways may increase susceptibility to intracellular pathogens such as Mycobacterium tuberculosis. Prolonged elevation of hepcidin production can lead to anemia due to reduced iron availability for erythropoiesis, a condition referred to as anemia of inflammation. In addition, we highlight the role of hepcidin upregulation in several infectious contexts, including HIV-associated anemia, iron deficiency anemia in Helicobacter pylori infection, and post-malarial anemia in pediatric patients. In addition, we show that certain infectious agents, such as hepatitis C virus (HCV), can suppress hepcidin production during both the acute and chronic phases of infection, while hepatitis B virus (HBV) exhibits similar suppression during the chronic phase.
Collapse
Affiliation(s)
- Tahereh Navidifar
- Department of Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran; Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Elahe Meftah
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hediyeh Baghsheikhi
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran; USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Kazemzadeh
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanie Karimi
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
14
|
Liu X, Gu J, Liu S, Huang J, Li L, Wang F, He S, Mi Z, Zhang Y, Wen J, Gao Q, Yang H, Feng Y, Luo H, Zhai X, Zhang L, Zheng Y, Luo Y, Niu T. The mitochondrial protease ClpP is a promising target for multiple myeloma treatment. Biochem Pharmacol 2025; 236:116855. [PMID: 40054785 DOI: 10.1016/j.bcp.2025.116855] [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: 11/07/2024] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025]
Abstract
Drug resistance and relapse are the major obstacles in multiple myeloma (MM) treatment, driving the search for novel therapeutics. The chemoactivation of mitochondrial caseinolytic protease P (ClpP) has shown to have anticancer effects on many tumors, but has seldom been elucidated in MM. Here we found that the CLPP expression was elevated in MM patients, and further increased in relapsed cases. After synthesizing and screening a panel of ClpP agonists, we identified a compound, 7b, as the most potent anti-MM agent in vitro. 7b activated ClpP protease activity, selectively degrading mitochondrial proteins, many of which are involved in oxidative phosphorylation (OXPHOS). As result, 7b treated MM had metabolic dysfunction, the mitochondrial membrane potential (MMP) collapse, reduced OXPHOS levels, and increased mitochondrial reactive oxygen species (ROS), leading to mitophagy-mediated MM cell death. Notably, 7b also showed efficacy against drug-resistant MM cell lines, including bortezomib- and lenalidomide-resistant cells. In vivo, 7b also exhibited remarkable anti-MM activity with tolerable side effects. In conclusion, targeting ClpP represents a promising therapeutic strategy for MM, with 7b serving as a potent anti-MM agent, especially for relapsed and refractory MM.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Jinlong Gu
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, China
| | - Song Liu
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, China
| | - Jingcao Huang
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Linfeng Li
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Fangfang Wang
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Siyao He
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Ziyue Mi
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Zhang
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Jingjing Wen
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China; Department of Hematology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Qianwen Gao
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China; College of Life Science, Sichuan University, Chengdu, China
| | - Haonan Yang
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Feng
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Hongmei Luo
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyu Zhai
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Li Zhang
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuhuan Zheng
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China.
| | - Youfu Luo
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, China.
| | - Ting Niu
- Department of Hematology/Institute of Hematology, West China Hospital, Sichuan University, Chengdu, China; State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, China; National Facility for Translational Medicine (Sichuan), West China Hospital, Sichuan University, China.
| |
Collapse
|
15
|
Ho MS. Clearance Pathways for α-Synuclein in Parkinson's Disease. J Neurochem 2025; 169:e70124. [PMID: 40509661 PMCID: PMC12163304 DOI: 10.1111/jnc.70124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2025] [Revised: 05/18/2025] [Accepted: 05/30/2025] [Indexed: 06/16/2025]
Abstract
Protein aggregation and accumulation are hallmark features of neurodegenerative diseases. In Parkinson's disease, the progressive formation and propagation of α-synuclein aggregates-found in Lewy bodies and Lewy neurites-are closely linked to widespread neuronal dysfunction, dopaminergic neuron loss, and the emergence of both motor and nonmotor symptoms, including anosmia, cognitive decline, and depression. Despite their pathological significance, the mechanisms underlying the formation, spread, and clearance of these aggregates remain incompletely understood. In this review, we examine the cellular and molecular pathways responsible for the elimination of protein aggregates in the diseased brain. We first summarize various experimental models of α-synuclein pathology, followed by a discussion of the degradation mechanisms in neurons and glial cells under pathological conditions. These findings offer new insights into cell type-specific clearance pathways and highlight potential therapeutic targets for mitigating α-synuclein-associated toxicity in Parkinson's disease.
Collapse
Affiliation(s)
- Margaret S. Ho
- Institute of NeuroscienceNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Brain Research CenterNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| |
Collapse
|
16
|
Sugawara Y, Morinaga H, Chen J, Kitagawa Y, Ogata H, Karim A, Kikuchi M, Khan M, Yasuhara E, Goto T, Martyn JAJ, Yasuhara S. Mito-Kaede photoactivation and chase experiment for mitophagy: mitophagy flux response toward various stimulations. Biotechniques 2025:1-13. [PMID: 40449520 DOI: 10.1080/07366205.2025.2505357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 05/09/2025] [Indexed: 06/03/2025] Open
Abstract
Mitophagy, a crucial mitochondrial quality control system for cellular stress adaptation, is a key focus in pathophysiology and drug discovery. Developing a simple and versatile mitophagy flux assay is vital for advancing our understanding of cellular responses. Addressing a gap in systematic methods, we employ the photoactivatable fluorescent protein mito-Kaede in C2C12 myocytes, demonstrating its remarkable versatility in quantifying mitophagy flux responses under various stimuli, including carbonyl cyanide m-chlorophenyl hydrazone (CCCP), TNF-α, lipopolysaccharide (LPS), and hypoxia. This study underscores the validity and distinctive advantages of the mito-Kaede assay through comparative analysis with conventional assays including Western blotting (WB), potentially providing valuable insights for both mitophagy flux analysis and drug development.
Collapse
Affiliation(s)
- Yoh Sugawara
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Hiroyuki Morinaga
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
- Department of Trauma and Critical Care Medicine, Kyorin University, Faculty of Medicine
| | - Jingyuan Chen
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yoshinori Kitagawa
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
| | - Hiroki Ogata
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
| | - Asiya Karim
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
| | - Miu Kikuchi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
| | - Maryam Khan
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
| | - Erica Yasuhara
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
| | - Takahisa Goto
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Joseph A Jeevendra Martyn
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
| | - Shingo Yasuhara
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
| |
Collapse
|
17
|
Xu Y, Xie R, Weng Y, Fang Y, Tao S, Zhang H, Chen H, Han A, Jiang Q, Liang W. Role and mechanism of gut microbiota-host interactions in the pathogenesis of Crohn's disease. Int J Colorectal Dis 2025; 40:130. [PMID: 40437310 PMCID: PMC12119691 DOI: 10.1007/s00384-025-04917-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/08/2025] [Indexed: 06/01/2025]
Abstract
BACKGROUND Crohn's disease (CD) is a chronic, nonspecific inflammatory bowel disease with a poor prognosis. Despite its increasing incidence, curing CD remains challenging due to its complex etiology and unclear pathogenesis. METHODS A comprehensive PubMed and Web of Science search was conducted using the keywords Crohn's disease, gut microbiota, dysbiosis, pathogenesis and treatment, focusing on studies published between 2014 and 2024. RESULTS Recent studies have demonstrated a close relationship between gut microbiota dysbiosis and the development of CD. Although many dysbioses associated with CD have not yet been proven to be causal or consequential, it has been observed that the gut microbiota in CD patients exhibits reduced diversity, a decrease in beneficial bacteria, and an increase in pathogenic bacteria. These changes may lead to decreased intestinal barrier function, abnormal immune responses, and enhanced inflammatory reactions, which are related to the disease's activity, phenotype, drug treatment efficacy, and postoperative therapeutic outcomes. Therefore, further exploration of the microbiota-host interactions and the pathogenesis of CD, the identification of biomarkers, and the development of targeted strategies for modulating the gut microbiota could offer new avenues for the prevention and treatment of CD. CONCLUSIONS This review highlights the pivotal role of gut microbiota dysbiosis in driving CD pathogenesis and its progression, while underscoring its potential as a therapeutic target through dietary modulation, microbial interventions, and integrative strategies to improve clinical management and prognostic outcomes.
Collapse
Affiliation(s)
- Yao Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Health Science Center, Ningbo University, Ningbo, China
| | - Runxiang Xie
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yuqing Weng
- Health Science Center, Ningbo University, Ningbo, China
| | - Yewei Fang
- Department of Clinical Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Shuan Tao
- Department of Clinical Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - He Zhang
- Laboratory Medical School, Bengbu Medical University, Bengbu, China
| | - Huimin Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Axiang Han
- Department of Clinical Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Qi Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| | - Wei Liang
- Department of Clinical Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| |
Collapse
|
18
|
Zhang Y, Ma P, Wang S, Chen S, Deng H. Restoring calcium crosstalk between ER and mitochondria promotes intestinal stem cell rejuvenation through autophagy in aged Drosophila. Nat Commun 2025; 16:4909. [PMID: 40425608 PMCID: PMC12116733 DOI: 10.1038/s41467-025-60196-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
Breakdown of calcium network is closely associated with cellular aging. Previously, we found that cytosolic calcium (CytoCa2+) levels were elevated while mitochondrial calcium (MitoCa2+) levels were decreased and associated with metabolic shift in aged intestinal stem cells (ISCs) of Drosophila. How MitoCa2+ was decoupled from the intracellular calcium network and whether the reduction of MitoCa2+ drives ISC aging, however, remains unresolved. Here, we show that genetically restoring MitoCa2+ can reverse ISC functional decline and promote intestinal homeostasis by activating autophagy in aged flies. Further studies indicate that MitoCa2+ and Mitochondria-ER contacts (MERCs) form a positive feedback loop via IP3R to regulate autophagy independent of AMPK. Breakdown of this loop is responsible for MitoCa2+ reduction and ISC dysfunction in aged flies. Our results identify a regulatory module for autophagy initiation involving calcium crosstalk between the ER and mitochondria, providing a strategy to treat aging and age-related diseases.
Collapse
Affiliation(s)
- Yao Zhang
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 20092, China
| | - Peng Ma
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 20092, China
| | - Saifei Wang
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 20092, China
| | - Shuxin Chen
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 20092, China
| | - Hansong Deng
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 20092, China.
| |
Collapse
|
19
|
Gong B, Liu Y, Yan W, Cheng C, Yang H, Huang J, Liu Q, Liu Y, Guo J, Deng X, Zhou B, Zheng D, Liu X, Liu Z, Fang W. NAP1L1 degradation by FBXW7 reduces the deubiquitination of HDGF-p62 signaling to stimulate autophagy and induce primary cisplatin chemosensitivity in nasopharyngeal carcinoma. Mol Cancer 2025; 24:152. [PMID: 40414865 DOI: 10.1186/s12943-025-02349-z] [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: 10/10/2024] [Accepted: 05/05/2025] [Indexed: 05/27/2025] Open
Abstract
Nucleosome assembly protein 1-like 1 (NAP1L1) has been implicated in promoting tumor cell proliferation. However, its role in regulating autophagy in tumors, including nasopharyngeal carcinoma (NPC), remains unclear. In this study, we observed that autophagy-inducing agents reduced NAP1L1 protein levels without affecting its mRNA expression. Reduced NAP1L1 enhanced autophagosome formation and maturation, thereby promoting cisplatin (DDP) chemosensitivity in both in vitro and in vivo NPC models. Mechanistically, reduced NAP1L1 impaired the recruitment of ubiquitin-specific protease 14 (USP14), limiting the deubiquitination of heparin-binding growth factor (HDGF) and decreasing HDGF protein levels. In turn, reduced HDGF suppressed USP14-mediated p62 deubiquitination, leading to further declines in p62 protein levels. Notably, the F-box and WD repeat domain-containing protein 7 (FBXW7), an inhibitory E3 ubiquitin ligase, directly interacted with and ubiquitinated NAP1L1, promoting its degradation. This degradation triggered NPC autophagy and enhanced DDP chemosensitivity by disrupting NAP1L1-induced HDGF/p62 signaling. Clinically, NAP1L1 protein expression was inversely correlated with FBXW7 levels in NPC tissue samples. Patients exhibiting high NAP1L1 and low FBXW7 levels had the poorest DDP chemosensitivity and survival outcomes. Our findings demonstrate that FBXW7-mediated NAP1L1 degradation suppresses HDGF-p62 signaling, thereby inducing autophagy and enhancing DDP chemosensitivity. These results underscore the potential of NAP1L1 and FBXW7 as therapeutic targets for NPC treatment.
Collapse
Affiliation(s)
- Bin Gong
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yahui Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Weiwei Yan
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Chao Cheng
- Department of Otolaryngology, Shenzhen Longgang Otolaryngology Hospital, Shenzhen, Guangdong, China
| | - Huiling Yang
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Jiyu Huang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Qing Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yuyan Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jiankang Guo
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiaojie Deng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Beixian Zhou
- Center of Stem Cell and Regenerative Medicine, The People's Hospital of Gaozhou, Gaozhou, China.
| | - Dayong Zheng
- Department of Oncology, Shunde Hospital of Southern Medical Universtiy, Foshan, China.
| | - Xiong Liu
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Zhen Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University Guangzhou, Guangdong, China.
- Central Laboratory, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.
| | - Weiyi Fang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
- Central Laboratory, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
20
|
Galindo-Cabello N, Sobas-Abad EM, Lapresa R, Agulla J, Almeida Á, López A, Pastor JC, Pastor-Idoate S, Usategui-Martín R. The TP53 Arg72Pro polymorphism predicts visual and neurodegenerative outcomes in retinal detachment. Cell Death Dis 2025; 16:415. [PMID: 40419469 DOI: 10.1038/s41419-025-07739-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 05/03/2025] [Accepted: 05/15/2025] [Indexed: 05/28/2025]
Abstract
Retinal detachment (RD) separates the retina from the retinal epithelium, causing photoreceptor apoptosis and irreversible vision loss. Even with successful surgical reattachment, complete visual recovery is not guaranteed. The TP53 Arg72Pro polymorphism, implicated in apoptosis, has emerged as a potential predictor of RD outcomes. We investigated the impact of the Arg72Pro polymorphism on retinal neurodegeneration and functional recovery in patients. The underlying mechanisms were analyzed in a humanized TP53 Arg72Pro RD mouse model. In a cohort of 180 patients, carriers of the Pro allele exhibited decreased apoptotic gene expression and improved visual recovery. Complementary findings in mice revealed that the Pro variant preserved photoreceptor integrity and reduced apoptosis rates following RD. Our findings highlight the potential of this TP53 polymorphism as a biomarker for RD outcomes and a tool for tailoring therapies. This study underscores the importance of integrating genetic profiling into personalized medicine approaches to improve recovery of RD patients' visual outcomes.
Collapse
Affiliation(s)
- Nadia Galindo-Cabello
- Unit of Excellence Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, Valladolid, Spain
- Department of Cell Biology, Genetics, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, Valladolid, Spain
- Network of Inflammatory Diseases- Networks of Cooperative Research Oriented to Health Results (RICORS), Carlos III National Institute of Health, Madrid, Spain
| | - Eva M Sobas-Abad
- Unit of Excellence Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, Valladolid, Spain
- Network of Inflammatory Diseases- Networks of Cooperative Research Oriented to Health Results (RICORS), Carlos III National Institute of Health, Madrid, Spain
- School of Nursing, University of Valladolid, Valladolid, Spain
| | - Rebeca Lapresa
- Institute of Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain
- Institute of Functional Biology and Genomics, CSIC, University of Salamanca, Salamanca, Spain
| | - Jesús Agulla
- Institute of Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain
- Institute of Functional Biology and Genomics, CSIC, University of Salamanca, Salamanca, Spain
| | - Ángeles Almeida
- Institute of Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain
- Institute of Functional Biology and Genomics, CSIC, University of Salamanca, Salamanca, Spain
| | - Antonio López
- Unit of Excellence Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, Valladolid, Spain
| | - José Carlos Pastor
- Unit of Excellence Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, Valladolid, Spain
| | - Salvador Pastor-Idoate
- Unit of Excellence Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, Valladolid, Spain.
- Network of Inflammatory Diseases- Networks of Cooperative Research Oriented to Health Results (RICORS), Carlos III National Institute of Health, Madrid, Spain.
- Department of Ophthalmology, University Clinical Hospital of Valladolid, Valladolid, Spain.
| | - Ricardo Usategui-Martín
- Unit of Excellence Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, Valladolid, Spain.
- Department of Cell Biology, Genetics, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, Valladolid, Spain.
- Network of Inflammatory Diseases- Networks of Cooperative Research Oriented to Health Results (RICORS), Carlos III National Institute of Health, Madrid, Spain.
| |
Collapse
|
21
|
Hong L, Zhao C, Wu T, Hu X, Li X, Li L. The role of programmed cell death in renal cancer: a bibliometric perspective (1998-2024). Discov Oncol 2025; 16:889. [PMID: 40410435 PMCID: PMC12102035 DOI: 10.1007/s12672-025-02610-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 05/07/2025] [Indexed: 05/25/2025] Open
Abstract
OBJECTIVE This bibliometric study aimed to map the global research landscape of programmed cell death (PCD) in renal cancer, delineating publication trends, influential authors, contributing regions, and thematic shifts between 1998 and 2023 year. METHODS We retrieved 5, 134 records from the Web of Science Core Collection (1998-2023) using comprehensive keywords encompassing "renal cancer," "programmed cell death," and related synonyms. After applying inclusion and exclusion criteria, we conducted co-citation, keyword, and cluster analyses with CiteSpace (v.6.3.R2) and VOSviewer (v.1.6.20) to identify major research fronts, collaboration networks, and thematic clusters. RESULTS Findings revealed a progressive increase in publications, notably accelerating after 2010 in tandem with the rise of immunotherapeutic strategies and targeted molecular interventions. China and the United States emerged as leading contributors, while journals such as Cancer Research and Clinical Cancer Research dominated in both publication frequency and citation impact. Authors including Kwon Taeg Kyu and Dahiya Rajvir significantly shaped foundational apoptosis research. Keyword and cluster analyses demonstrated a shift from earlier apoptosis- and angiogenesis-focused studies toward intersections of metabolic reprogramming, immune infiltration, and newer cell death modalities (e.g., ferroptosis, pyroptosis). High-impact papers underscored immunotherapy's pivotal role in modulating cell death pathways and informing novel combination regimens. CONCLUSION PCD research in renal cancer has evolved into a dynamic, interdisciplinary domain integrating immunology, molecular targeting, and multi-omic profiling. Future development of the field aimed at refining precision therapies that exploit diverse cell death mechanisms and thereby improve clinical outcomes.
Collapse
Affiliation(s)
- Liang Hong
- Department of Medical Laboratory, The People's Hospital of Aba Tibetan and Qiang Autonomous Prefecture, Aba Tibetan and Qiang Autonomous Prefecture, Barkam, 624000, China
| | - Chun Zhao
- Department of General Practice, The People's Hospital of Dazu, Chongqing, The Affiliated Dazu's Hospital of Chongqing Medical University, Chongqing, 402360, China
| | - Tingping Wu
- Department of General Practice, The People's Hospital of Dazu, Chongqing, The Affiliated Dazu's Hospital of Chongqing Medical University, Chongqing, 402360, China
| | - Xiaorong Hu
- Department of Nephrology, The People's Hospital of Dazu, Chongqing, The Affiliated Dazu's Hospital of Chongqing Medical University, Chongqing, 402360, China
| | - Xueyao Li
- Department of Nephrology, The People's Hospital of Dazu, Chongqing, The Affiliated Dazu's Hospital of Chongqing Medical University, Chongqing, 402360, China.
| | - Lu Li
- Department of Nephrology, The People's Hospital of Dazu, Chongqing, The Affiliated Dazu's Hospital of Chongqing Medical University, Chongqing, 402360, China.
| |
Collapse
|
22
|
Gebauer N, Wang SS. The Role of EBV in the Pathogenesis of Diffuse Large B-Cell Lymphoma. Curr Top Microbiol Immunol 2025. [PMID: 40399571 DOI: 10.1007/82_2025_296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
There are multiple established risk factors for DLBCL; these risk factors share an underlying biology, which generally cause immune dysfunction, spanning immunosuppression to chronic inflammation. EBV is an established risk factor for DLBCL and approximately 10% of DLBCLs are EBV-positive. EBV is a ubiquitous infection, and it is thus among populations that are immunocompromised, by age or medically defined, where EBV-positive DLBCLs arise. In this chapter, we review the current classification, epidemiology, clinical, pathology, and molecular characteristics of EBV-positive DLBCL, and discuss the role of EBV in lymphoma tumorigenesis. We further discuss current and novel treatments aimed at the NFκB pathway and other targets.
Collapse
Affiliation(s)
- Niklas Gebauer
- Klinik für Hämatologie und Onkologie, UKSH Campus Lübeck, Lübeck, Germany
| | - Sophia S Wang
- Division of Computational and Quantitative Medicine, City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| |
Collapse
|
23
|
Peng H, Cheng Q, Chen J, Wang Y, Du M, Lin X, Zhao Q, Chen S, Zhang J, Wang X. Green Tea Epigallocatechin-3-gallate Ameliorates Lipid Accumulation and Obesity-Associated Metabolic Syndrome via Regulating Autophagy and Lipolysis in Preadipocytes and Adipose Tissue. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12272-12291. [PMID: 40347183 DOI: 10.1021/acs.jafc.5c00973] [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: 05/12/2025]
Abstract
Previous studies have shown that epigallocatechin-3-gallate (EGCG), the most abundant catechin in green tea, demonstrates promising antiobesity effects. While autophagy mediates obesity via preadipocyte differentiation and lipogenesis, EGCG's potential autophagy-dependent antiobesity mechanism remains unclear. We used 3T3-L1 cells and high-fat-diet (HFD)-fed mice to examine how EGCG inhibits adipogenesis and lipogenesis via autophagy. EGCG (50 or 100 mg/kg) significantly attenuated HFD-induced weight gain, fat accumulation, hyperlipidemia, and glucose intolerance in mice. It also enhanced autophagy and lipolysis in white adipose tissue (WAT). EGCG profoundly inhibited terminal preadipocyte differentiation and lipid droplet formation in 3T3-L1 cells accompanied by reduced PPARγ, C/EBPα, and FASN expressions. Mechanistically, EGCG inhibited autophagy during the early stage of preadipocyte differentiation, as evidenced by increased autophagosome accumulation and impaired autophagic flux. The antiadipogenic effect of EGCG was further aggravated by the autophagy inhibitor chloroquine. Meanwhile, EGCG increased p38 and AMPK/ACC phosphorylation while inhibiting JNK phosphorylation in 3T3-L1 cells at an early stage of adipogenesis. Interestingly, EGCG reduced the expression of lipolytic enzymes HSL and ATGL, and it decreased glycerol contents in differentiated 3T3-L1 cells. These findings provide novel insights into the mechanism of using green tea EGCG in functional foods to combat obesity by targeting autophagy and lipolysis.
Collapse
Affiliation(s)
- He Peng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, P. R. China
| | - Qi Cheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, P. R. China
| | - Jiajun Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, P. R. China
| | - Ying Wang
- College of Food and Health, Zhejiang A&F University, Hangzhou 311300, P. R. China
| | - Menghao Du
- Research Institute of Subtropical Forestry, Chinese Academy of Forestry, Hangzhou 311400, P. R. China
| | - Xiaojian Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, P. R. China
| | - Qian Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, P. R. China
| | - Shengjia Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, P. R. China
| | - Jingsa Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, P. R. China
| | - Xingya Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, P. R. China
- School of Pharmaceutical Sciences & Institute of Advanced Studies, Taizhou University, Taizhou 318000, P. R. China
| |
Collapse
|
24
|
Le Floch M, Wolf F, McIntyre L, Weinert C, Palm A, Volk K, Herzog P, Kirk SH, Steinhäuser JL, Stopp C, Geissler ME, Herzog M, Sulk S, Kather JN, Meining A, Hann A, Palm S, Hampe J, Herzog N, Brinkmann F. Galar - a large multi-label video capsule endoscopy dataset. Sci Data 2025; 12:828. [PMID: 40394033 DOI: 10.1038/s41597-025-05112-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 04/29/2025] [Indexed: 05/22/2025] Open
Abstract
Video capsule endoscopy (VCE) is an important technology with many advantages (non-invasive, representation of small bowel), but faces many limitations as well (time-consuming analysis, short battery lifetime, and poor image quality). Artificial intelligence (AI) holds potential to address every one of these challenges, however the progression of machine learning methods is limited by the avaibility of extensive data. We propose Galar, the most comprehensive dataset of VCE to date. Galar consists of 80 videos, culminating in 3,513,539 annotated frames covering functional, anatomical, and pathological aspects and introducing a selection of 29 distinct labels. The multisystem and multicenter VCE data from two centers in Saxony (Germany), was annotated framewise and cross-validated by five annotators. The vast scope of annotation and size of Galar make the dataset a valuable resource for the use of AI models in VCE, thereby facilitating research in diagnostic methods, patient care workflow, and the development of predictive analytics in the field.
Collapse
Affiliation(s)
- Maxime Le Floch
- Else Kröner Fresenius Center for Digital Health, Technische Universität Dresden (TU Dresden), Dresden, Germany.
- Department of Medicine I, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany.
| | - Fabian Wolf
- Else Kröner Fresenius Center for Digital Health, Technische Universität Dresden (TU Dresden), Dresden, Germany
- Institute of Computer Science, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Lucian McIntyre
- Else Kröner Fresenius Center for Digital Health, Technische Universität Dresden (TU Dresden), Dresden, Germany
- Institute of Computer Science, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | | | - Albrecht Palm
- Else Kröner Fresenius Center for Digital Health, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Konrad Volk
- Else Kröner Fresenius Center for Digital Health, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Paul Herzog
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Sophie Helene Kirk
- Else Kröner Fresenius Center for Digital Health, Technische Universität Dresden (TU Dresden), Dresden, Germany
- Department of Medicine I, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Jonas L Steinhäuser
- Else Kröner Fresenius Center for Digital Health, Technische Universität Dresden (TU Dresden), Dresden, Germany
- Department of Medicine I, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Catrein Stopp
- Else Kröner Fresenius Center for Digital Health, Technische Universität Dresden (TU Dresden), Dresden, Germany
- Department of Medicine I, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Mark Enrik Geissler
- Department of Medicine I, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Moritz Herzog
- Else Kröner Fresenius Center for Digital Health, Technische Universität Dresden (TU Dresden), Dresden, Germany
- Department of Medicine I, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Stefan Sulk
- Department of Medicine I, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Jakob Nikolas Kather
- Else Kröner Fresenius Center for Digital Health, Technische Universität Dresden (TU Dresden), Dresden, Germany
- Department of Medicine I, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Alexander Meining
- Interventional and Experimental Endoscopy (InExEn), Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Alexander Hann
- Interventional and Experimental Endoscopy (InExEn), Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Steffen Palm
- Medical Office for Gastroenterology and Internal Medicine, Dippoldiswalde, Germany
| | - Jochen Hampe
- Else Kröner Fresenius Center for Digital Health, Technische Universität Dresden (TU Dresden), Dresden, Germany
- Department of Medicine I, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Nora Herzog
- Else Kröner Fresenius Center for Digital Health, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Franz Brinkmann
- Else Kröner Fresenius Center for Digital Health, Technische Universität Dresden (TU Dresden), Dresden, Germany
- Department of Medicine I, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
| |
Collapse
|
25
|
ALKhemeiri N, Eljack S, Saber-Ayad MM. Perspectives of Targeting Autophagy as an Adjuvant to Anti-PD-1/PD-L1 Therapy for Colorectal Cancer Treatment. Cells 2025; 14:745. [PMID: 40422248 DOI: 10.3390/cells14100745] [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: 02/13/2025] [Revised: 05/07/2025] [Accepted: 05/12/2025] [Indexed: 05/28/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in the world, with increasing incidence and mortality rates. Standard conventional treatments for CRC are surgery, chemotherapy, and radiotherapy. Recently, immunotherapy has been introduced as a promising alternative to CRC treatment that utilizes patients' immune system to combat cancer cells. The beneficial effect of immune checkpoint inhibitors, specifically anti-PD-1/ PD-L1, has been ascribed to the abundance of DNA replication errors that result in the formation of neoantigens. Such neoantigens serve as distinct flags that amplify the immune response when checkpoint inhibitors (ICIs) are administered. DNA replication errors in CRC patients are expressed as two statuses: the first is the deficient mismatch repair (MSI-H/dMMR) with a higher overall immune response and survival rate than the second status of patients with proficient mismatch repair (MSS/pMMR). There is a limitation to using anti-PD-1/PD-L1 as it is only confined to MSI-H/dMMR, where there is an abundance of T-cell inhibitory ligands (PD-L1). This calls for investigating new therapeutic interventions to widen the scope of ICIs' role in the treatment of CRC. Autophagy modulation provides a good example. Autophagy is a cellular process that plays a crucial role in maintaining cellular homeostasis and has been studied for its impact on tumor development, progression, and response to treatment. In this review, we aim to highlight autophagy as a potential determinant in tumor immune response and to study the impact of autophagy on the tumor immune microenvironment. Moreover, we aim to investigate the value of a combination of anti-PD-1/PD-L1 agents with autophagy modulators as an adjuvant therapeutic approach for CRC treatment.
Collapse
Affiliation(s)
- Nasrah ALKhemeiri
- College of Graduate Studies, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Sahar Eljack
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Department of Pharmaceutics, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Maha Mohamed Saber-Ayad
- College of Graduate Studies, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Department of Pharmacology, Faculty of Medicine, Cairo University, Cairo 12211, Egypt
| |
Collapse
|
26
|
Usategui-Martín R, Esteban-López V, Chantre-Fortes E, Sánchez-Martín M, Riancho JA, López DE, González-Sarmiento R. The p.R321C mutation in the p62 protein is associated with abnormalities in the central nervous system. Sci Rep 2025; 15:16929. [PMID: 40374720 PMCID: PMC12081919 DOI: 10.1038/s41598-025-00764-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 04/30/2025] [Indexed: 05/17/2025] Open
Abstract
SQSTM1/p62 has an essential role in autophagy, a catabolic pathway that is vital for maintaining cell homeostasis. p62 alterations have been observed in multiple pathological conditions, including neurodegenerative diseases and bone metabolism alterations. The p.R321C p62 protein mutation has been described in patients with amyotrophic lateral sclerosis, frontotemporal lobar degeneration, and Paget's disease of bone. In vitro studies associated the p62-321C variant with a blockade of autophagy and with the activation of the NF-kB pathway. We aimed to provide a deeper understating of the pathophysiological consequences of the p.R321C p62 mutation using a humanized mouse model. Micro-computed tomography, immunohistochemistry, and western blot analysis studied the functional consequences of the p. R321C p62 mutation. Statistical analyses were performed using SPSS software. The results showed that the p62-321C mice developed seizures after tactile-vestibular stimulation, probably associated with a blockage of the autophagy and NF-kB activation. Changes in expression of cFos and p62 were found in the amygdala, hypothalamic nuclei, and hippocampi nuclei. In addition, numerous degenerating motor neurons were observed in the spinal cord of the p62-321C mice. We report that the blockage of the autophagy, caused by p.R321C p62 mutation, is associated with abnormalities in the central nervous system, mainly seizures after tactile-vestibular stimulation and degeneration of the motor neurons of the spinal cord but not with bone abnormalities in a humanized mouse model.
Collapse
Affiliation(s)
- Ricardo Usategui-Martín
- Molecular Medicine Unit, Department of Medicine, Faculty of Medicine, University of Salamanca, Campus Miguel Unamuno, 37007, Salamanca, Spain.
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain.
| | - Vega Esteban-López
- Institute of Neuroscience of Castilla y León (INCYL), University of Salamanca, 37007, Salamanca, Spain
| | - Estefanía Chantre-Fortes
- Institute of Neuroscience of Castilla y León (INCYL), University of Salamanca, 37007, Salamanca, Spain
| | - Manuel Sánchez-Martín
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
- Transgenic Facility, University of Salamanca, 37007, Salamanca, Spain
| | - José A Riancho
- Department of Medicine and Psychiatry, Faculty of Medicine, University of Cantabria, IDIVAL, 39011, Santander, Spain
- Internal Medicine Department, Marqués de Valdecilla University Hospital, 39008, Santander, Spain
| | - Dolores E López
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
- Institute of Neuroscience of Castilla y León (INCYL), University of Salamanca, 37007, Salamanca, Spain
- Department of Cell Biology and Pathology, Faculty of Medicine, University of Salamanca, 37007, Salamanca, Spain
| | - Rogelio González-Sarmiento
- Molecular Medicine Unit, Department of Medicine, Faculty of Medicine, University of Salamanca, Campus Miguel Unamuno, 37007, Salamanca, Spain.
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain.
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, 37007, Salamanca, Spain.
| |
Collapse
|
27
|
Ma JX, Li XJ, Li YL, Liu MC, Du RH, Cheng Y, Li LJ, Ai ZY, Jiang JT, Yan SY. Chaperonin-containing tailless complex polypeptide 1 subunit 6A negatively regulates autophagy and protects colorectal cancer cells from cisplatin-induced cytotoxicity. World J Gastroenterol 2025; 31:105729. [DOI: 10.3748/wjg.v31.i18.105729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND As a member of the chaperonin-containing tailless complex polypeptide 1 (TCP1) complex, which plays a pivotal role in ensuring the accurate folding of numerous proteins, chaperonin-containing TCP1 subunit 6A (CCT6A) participates in various physiological and pathological processes. However, its effects on cell death and cancer therapy and the underlying mechanisms need further exploration in colorectal cancer (CRC) cells.
AIM To explore the effects of CCT6A on cell death and cancer therapy and the underlying mechanisms in CRC.
METHODS Cell proliferation was evaluated using the MTS assay, EdU staining, and colony growth assays. The expression of CCT6A was monitored by immunoblotting and quantitative PCR. CCT6A was knocked out by CRISPR-Cas9, and overexpressed by transfecting plasmids. Autophagy was examined by immunoblotting and the mCherry-GFP-LC3 assay. To monitor apoptosis and necroptosis, immunoblotting, co-immunoprecipitation, and flow cytometry were employed.
RESULTS Cisplatin (DDP) exerted cytotoxic effects on CRC cells while simultaneously downregulating the expression of CCT6A. Depletion of CCT6A amplified the cytotoxic effects of DDP, whereas overexpression of CCT6A attenuated these adverse effects. CCT6A suppressed autophagy, apoptosis, and necroptosis under both basal and DDP-treated conditions. Autophagy inhibitors significantly enhanced the cytotoxic effects of DDP, whereas a necroptosis inhibitor partially reversed the cell viability loss induced by DDP. Furthermore, inhibiting autophagy enhanced both apoptosis and necroptosis induced by DDP.
CONCLUSION CCT6A negatively modulates autophagy, apoptosis, and necroptosis, and CCT6A confers resistance to DDP therapy in CRC, suggesting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Jian-Xing Ma
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining 272067, Shandong Province, China
| | - Xiao-Jun Li
- Department of General Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Ya-Long Li
- Department of General Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Ming-Chan Liu
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining 272067, Shandong Province, China
| | - Rui-Hang Du
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining 272067, Shandong Province, China
| | - Yi Cheng
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining 272067, Shandong Province, China
| | - Liang-Jie Li
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining 272067, Shandong Province, China
| | - Zhi-Ying Ai
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining 272067, Shandong Province, China
| | - Jian-Tao Jiang
- The Second Affiliated Hospital of Xi’an Jiaotong University, Xibei Hospital, Xi’an 710000, Shaanxi Province, China
| | - Si-Yuan Yan
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining 272067, Shandong Province, China
| |
Collapse
|
28
|
Hsu CY, Ahmad I, Maya RW, Abass MA, Gupta J, Singh A, Joshi KK, Premkumar J, Sahoo S, Khosravi M. The potential therapeutic approaches targeting gut health in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS): a narrative review. J Transl Med 2025; 23:530. [PMID: 40350437 PMCID: PMC12066075 DOI: 10.1186/s12967-025-06527-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Accepted: 04/21/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a complex disorder characterized by persistent fatigue and cognitive impairments, with emerging evidence highlighting the role of gut health in its pathophysiology. The main objective of this review was to synthesize qualitative and quantitative data from research examining the gut microbiota composition, inflammatory markers, and therapeutic outcomes of interventions targeting the microbiome in the context of ME/CFS. METHODS The data collection involved a detailed search of peer-reviewed English literature from January 1995 to January 2025, focusing on studies related to the microbiome and ME/CFS. This comprehensive search utilized databases such as PubMed, Scopus, and Web of Science, with keywords including "ME/CFS," "Gut-Brain Axis," "Gut Health," "Intestinal Dysbiosis," "Microbiome Dysbiosis," "Pathophysiology," and "Therapeutic Approaches." Where possible, insights from clinical trials and observational studies were included to enrich the findings. A narrative synthesis method was also employed to effectively organize and present these findings. RESULTS The study found notable changes in the gut microbiota diversity and composition in ME/CFS patients, contributing to systemic inflammation and worsening cognitive and physical impairments. As a result, various microbiome interventions like probiotics, prebiotics, specific diets, supplements, fecal microbiota transplantation, pharmacological interventions, improved sleep, and moderate exercise training are potential therapeutic strategies that merit further exploration. CONCLUSIONS Interventions focusing on the gut-brain axis may help reduce neuropsychiatric symptoms in ME/CFS by utilizing the benefits of the microbiome. Therefore, identifying beneficial microbiome elements and incorporating their assessments into clinical practice can enhance patient care through personalized treatments. Due to the complexity of ME/CFS, which involves genetic, environmental, and microbial factors, a multidisciplinary approach is also necessary. Since current research lacks comprehensive insights into how gut health might aid ME/CFS treatment, standardized diagnostics and longitudinal studies could foster innovative therapies, potentially improving quality of life and symptom management for those affected.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University, Tempe Campus, Phoenix, AZ, USA
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | | | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, India
- Graphic Era (Deemed to Be University), Dehradun, Uttarakhand, India
| | - J Premkumar
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Samir Sahoo
- Department of General Medicine, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, India
| | - Mohsen Khosravi
- Department of Psychiatry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
- Health Promotion Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
- Community Nursing Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| |
Collapse
|
29
|
Li J, Luo L, Liu Y, Li W, Wei X. Mesenteric lymphangioma in childhood: a case report and narrative literature review. Front Oncol 2025; 15:1541445. [PMID: 40416869 PMCID: PMC12098272 DOI: 10.3389/fonc.2025.1541445] [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: 12/13/2024] [Accepted: 04/15/2025] [Indexed: 05/27/2025] Open
Abstract
Lymphangioma is an uncommon benign neoplasm of the lymphatic system, predominantly observed in children and infants and infrequently in adults. Mesenteric lymphangioma is very rare. This article reports on the case of a 4-year-old female who was admitted to the hospital due to abdominal pain. Ultrasonography identified a cystic mass in the abdominal cavity, diagnosed as mesenteric lymphangioma, for which surgical intervention was advised. The patient underwent laparoscopic lesion resection with small bowel resection and anastomosis under general anesthesia. Pathological examination confirmed the diagnosis of mesenteric lymphangioma. In children, it frequently presents as an acute abdomen. Our comprehensive literature analysis strongly suggests that treatment decisions for pediatric mesenteric lymphangioma need to be guided by a careful assessment of individual patient presentations. Although exploratory laparotomy with tumor resection and involved bowel segment removal remains the gold-standard treatment, the advent of laparoscopic techniques and sclerotherapy has facilitated the evolution of personalized therapeutic strategies, potentially reducing dependence on conventional surgical approaches in the future.
Collapse
Affiliation(s)
- Jin Li
- Department of Ultrasound, Deyang People’s Hospital, Deyang, China
| | - Le Luo
- Medical Records Statistics Management Section, Deyang People’s Hospital, Deyang, China
| | - Yong Liu
- Department of Pediatric Surgery, Deyang People’s Hospital, Deyang, China
| | - Wenlan Li
- Department of Ultrasound, Deyang People’s Hospital, Deyang, China
| | - Xin Wei
- Department of Ultrasound, Deyang People’s Hospital, Deyang, China
| |
Collapse
|
30
|
Górecki M, Żbikowska A, Tokłowicz M, Sajdak S, Englert-Golon M, Andrusiewicz M. Hsa-miR-21-5p and Hsa-miR-145-5p Expression: From Normal Tissue to Malignant Changes-Context-Dependent Correlation with Estrogen- and Hypoxia-Vascularization-Related Pathways Genes: A Pilot Study. Int J Mol Sci 2025; 26:4461. [PMID: 40362695 PMCID: PMC12072406 DOI: 10.3390/ijms26094461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 05/04/2025] [Accepted: 05/06/2025] [Indexed: 05/15/2025] Open
Abstract
Ovarian cancer (OC) is a severe gynecological malignancy with a high mortality rate among women worldwide. It is often diagnosed at advanced stages due to the lack of effective screening methods. This study investigated the expression patterns of microRNAs (miRNAs) hsa-miR-21-5p and hsa-miR-145-5p as potential OC prognostic and diagnostic biomarkers and their correlation with estrogen-dependent (ESR1 & 2, PELP1 and c-SRC) and hypoxia-neovascularization-induced (HIF1A, EPAS1, and VEGFA) pathway genes. Tissue samples obtained from twenty patients with confirmed ovarian cancer and twenty controls were analyzed using quantitative polymerase chain reaction (qPCR) to examine miRNA and mRNA levels. The qPCR analysis revealed significantly higher hsa-miR-21-5p and lower hsa-miR-145-5p expression in OC tissues than controls. Moreover, a significant trend was observed in hsa-miR-21-5p and hsa-miR-145-5p expression levels across normal, non-cancerous changes and malignant ovarian tissues. The hsa-miR-21-5p showed better diagnostic potential than hsa-miR-145-5p. We also observed inconsistent correlations in hsa-miR-21-5p and hsa-mir-145-5p and estrogen-related and hypoxia-neovascularization-dependent genes in ovarian cancer across all groups. This suggests that the relationship between these miRNAs and the selected genes is context-specific. Our findings suggest that hsa-miR-21-5p and hsa-miR-145-5p expression levels may be prognostic or diagnostic markers for ovarian cancer patients.
Collapse
Affiliation(s)
- Mateusz Górecki
- Department of Cell Biology, Poznan University of Medical Sciences, Rokietnicka 5D, 60-806 Poznań, Poland; (M.G.); (A.Ż.); (M.T.)
- Cell Biology Research Group, Student Scientific Society, Poznan University of Medical Sciences, Rokietnicka 5E, 60-806 Poznań, Poland
| | - Aleksandra Żbikowska
- Department of Cell Biology, Poznan University of Medical Sciences, Rokietnicka 5D, 60-806 Poznań, Poland; (M.G.); (A.Ż.); (M.T.)
| | - Małgorzata Tokłowicz
- Department of Cell Biology, Poznan University of Medical Sciences, Rokietnicka 5D, 60-806 Poznań, Poland; (M.G.); (A.Ż.); (M.T.)
| | - Stefan Sajdak
- Division of Gynecology, Poznan University of Medical Sciences, 10 Fredry St., 61-701 Poznań, Poland;
- Department of Gynaecology and Obstetrics, Collegium Medicum University of Zielona Góra, Zyty 28, 65-046 Zielona Góra, Poland
| | - Monika Englert-Golon
- Division of Gynaecological Oncology, Department of Gynaecology, Gynecological and Obstetrics Clinical Hospital, Poznan University of Medical Sciences, Polna 33, 60-535 Poznań, Poland;
| | - Mirosław Andrusiewicz
- Department of Cell Biology, Poznan University of Medical Sciences, Rokietnicka 5D, 60-806 Poznań, Poland; (M.G.); (A.Ż.); (M.T.)
| |
Collapse
|
31
|
Azuma R, Shingu Y, Gao J, Wakasa S. Effects of Left Ventricular Unloading on Cardiac Function, Heart Failure Markers, and Autophagy in Rat Hearts with Acute Myocardial Infarction. Int J Mol Sci 2025; 26:4422. [PMID: 40362660 PMCID: PMC12072880 DOI: 10.3390/ijms26094422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 05/04/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
Percutaneous ventricular assist devices are utilized in cases of cardiogenic shock following acute myocardial infarction (AMI). However, the mechanism underlying the beneficial effects of LV unloading in AMI remains unclear. This study aimed to examine the impact of LV unloading on cardiac function, heart failure markers, and protein degradation (autophagy and ubiquitin-proteasome system: UPS) post AMI in rats. Nine-week-old male Lewis rats were randomized into non-AMI, AMI, non-AMI with LV unloading, and AMI with LV unloading groups. LV unloading was achieved through heterotopic heart-lung transplantation. Rats were euthanized 2 and 14 days after the procedure. Cardiac functional assessment was performed using Langendorff heart perfusion. RT-PCR and Western blot analyses were conducted using the LV myocardium. The rate pressure product was comparable between the non-AMI with LV unloading group and the AMI with LV unloading at 14 days. The atrial natriuretic factor tended to be suppressed by LV unloading. LV unloading had reducing effects on the expressions of p62, selectively degraded during autophagy, both 2 and 14 days after AMI. There was no effect on the parameters for the UPS. LV unloading has a mitigating effect on the deterioration of cardiac function following AMI. Autophagy, which was suppressed by AMI, was ameliorated by LV unloading.
Collapse
|
32
|
Cheng Y, Zhou L, Wang D, Li X, Lin R, Chen J, Tu F, Lin Y, Wu W, Liu M, Zhang H, Qiu H. Inhaled alone versus inhaled plus intravenous polymyxin B for the treatment of pneumonia due to carbapenem-resistant gram-negative bacteria: A prospective randomized controlled trial. Int J Antimicrob Agents 2025; 65:107483. [PMID: 40023452 DOI: 10.1016/j.ijantimicag.2025.107483] [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: 08/04/2024] [Revised: 02/12/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
OBJECTIVES Infections due to carbapenem-resistant Gram-negative bacteria (CR-GNB) are associated with considerable morbidity and mortality. Polymyxin B (PMB) is a first-line agent for CR-GNB-associated pneumonia, but limited data exist on the clinical use of inhaled (IH) PMB. METHODS A single-center, prospective randomized controlled trial was conducted in China to compare IH PMB alone with IH plus intravenous (IV) PMB between February 2022 and February 2024. The primary outcome was the clinical cure rate. RESULTS Twenty-two evaluable patients were assigned to the IH group, and 56 patients were included in the IH+IV group. Baseline characteristics were comparable between the two groups. No significant differences were observed in clinical cure rates, favorable clinical outcomes, microbiological outcomes, all-cause mortality, or pneumonia-related mortality. However, IH PMB alone was associated with a lower incidence of nephrotoxicity (P = 0.030). IH PMB demonstrated significantly higher drug concentrations in the epithelial lining fluid (ELF) compared to systemic administration. Patients with immunosuppressive therapy (OR, 0.066; 95% CI, 0.010-0.433; P = 0.005), malignancies (OR, 0.112; 95% CI, 0.016-0.797; P = 0.029), and higher SOFA scores (OR, 0.693; 95% CI, 0.518-0.929; P = 0.014) were less likely to achieve favorable clinical outcomes. Conversely, higher PMB ELF 1-hour concentrations (OR, 1.085; 95% CI, 1.026-1.148; P = 0.004) were associated with more favorable clinical outcomes. The combination of these four indicators demonstrated excellent diagnostic performance (AUC = 0.882). Plasma 1-hour PMB concentrations showed acceptable predictive performance for nephrotoxicity (AUC = 0.766). CONCLUSIONS The potential benefits of IH PMB outweigh the risks, making it an effective treatment for CR-GNB-associated pneumonia in combination with other empirical antimicrobial agents.
Collapse
Affiliation(s)
- Yu Cheng
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China; College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Lili Zhou
- Department of Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Danjie Wang
- Department of Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xueyong Li
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China; College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Rongqi Lin
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China; College of Pharmacy, Fujian Medical University, Fuzhou, China; Department of Pharmacy, Shanghang County Hospital, Shanghang, China
| | - Junnian Chen
- Department of Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Fuquan Tu
- Department of Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yiqin Lin
- Department of Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Wenwei Wu
- Department of Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Maobai Liu
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China; College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Hui Zhang
- Department of Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Hongqiang Qiu
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China; College of Pharmacy, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
33
|
Li Q, Zhang M, Gao Q, Chen X. High fat-induced the upregulation of LOX-1 in RF/6A cells under high glucose condition. J Diabetes Complications 2025; 39:109019. [PMID: 40168811 DOI: 10.1016/j.jdiacomp.2025.109019] [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: 02/08/2025] [Accepted: 03/25/2025] [Indexed: 04/03/2025]
Abstract
OBJECTIVE To investigate the effect of ox-LDL on the expression of lectin-like receptor of ox-LDL (LOX-1) and intercellular adhesion molecule-1 (ICAM-1) in RF/6A cells under high-glucose condition. METHODS RF/6A cells were cultured in normal or high-glucose medium for two days. Furthermore, RF/6A cells were cultured in medium with high glucose and ox-LDL or normal medium with ox-LDL. The concentrations of ox-LDL were determined by initial screening based on migration and immunofluorescence. The expressions of LOX-1 and ICAM-1 were determined by western blot. RESULTS The maximal effect of glucose on RF/6A cells was observed with the concentration of 25 mmol/l for 48 h. The LOX-1 expression was upregulated under high glucose condition than normal glucose (p < 0.05). There were significant LOX-1 overexpression and blocked ICAM-1 activation in RF/6A cells under high-glucose condition (p < 0.05). In the normal medium with ox-LDL groups, LOX-1 expression was both increased than in the normal medium group (p < 0.05). In the high glucose medium with ox-LDL groups, the expression levels of LOX-1 and ICAM-1 were increased than the high glucose medium group (p < 0.05). CONCLUSION A certain concentration of ox-LDL blocks high-glucose-induced retinal vascular endothelial injury by inhibiting the upregulation of ICAM-1 due to a high-glucose environment. Dyslipidemia may play an important role in the development of diabetic retinopathy, emphasizing the importance of active regulation of blood lipids in diabetic retinopathy therapy.
Collapse
Affiliation(s)
- Qin Li
- Department of Ophthalmology, Bright Gaoxin Eye Hospital, Urumqi, Xinjiang 830013, China
| | - Mingmei Zhang
- Department of Ophthalmology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Qianying Gao
- Department of Ophthalmology, Ophthalmological Center of Sun Yat-sen University, Guangzhou, Guangdong 510030, China
| | - Xueyi Chen
- Department of Ophthalmology, Lanzhou Aier Eye Hospital, Lanzhou, Gansu 730030, China.
| |
Collapse
|
34
|
Maaz M, Sultan MT, Noman AM, Zafar S, Tariq N, Hussain M, Imran M, Mujtaba A, Yehuala TF, Mostafa EM, Selim S, Al Jaouni SK, Alsagaby SA, Al Abdulmonem W. Anthocyanins: From Natural Colorants to Potent Anticancer Agents. Food Sci Nutr 2025; 13:e70232. [PMID: 40321606 PMCID: PMC12048707 DOI: 10.1002/fsn3.70232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/15/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Cancer is a prevalent global disease affecting ~20 million individuals, and this burden causes the death of ~9.7 million people in 2024. The prevalence rate is continuously increasing due to exposure to harmful environmental and occupational contaminants (toxins and chemicals), compromised immune response, genetic modifications, and poor lifestyle and dietary practices. The management of cancer is challenging and demands cost-effective and safe therapeutic strategies. This review accentuates the anticancer potential of anthocyanins and its associated underlying mechanism. Anthocyanins, the active components extracted from grapes, berries, black chokeberries, eggplants, black currants, sweet cherries, strawberries, black grapes, plums, and red onions, hold antioxidant and anti-inflammatory potential. The bioavailability of anthocyanins is a crucial factor in imposing their anticancer effect, and this bioavailability can be improved by microbial phenolic catabolites, provision of α-casein, and nano delivery systems. Anthocyanins hinder cell migration, invasion, and proliferation by inducing apoptosis, suppressing cell cycle at G0/G1, S, or G2/M stages, and modulating signaling pathways such as apoptotic cascades, PI3K/Akt, MAPK, and NF-κB. Moreover, anthocyanins downregulate oncogenes (Bcl-2, MYC, and HER2) and improve the activity of tumor suppressor genes (TP53, BRCA1, and RB1). Anthocyanins, particularly cyanidin-3-O-glucoside, suppress inflammation and production of pro-inflammatory cytokines (COX-2, TNF-α, and IL-6) in colorectal cancer and hepatocellular carcinoma. Moreover, it causes cell cycle inhibition and mitochondrial dysfunction in ovarian and cervical malignancies. Although pre-clinical studies have proved anticancer activities, further clinical trials are required to validate its therapeutic impact and standard dose regimens.
Collapse
Affiliation(s)
- Muhammad Maaz
- Department of Human Nutrition, Faculty of Food Science and NutritionBahauddin Zakariya UniversityMultanPakistan
| | - Muhammad Tauseef Sultan
- Department of Human Nutrition, Faculty of Food Science and NutritionBahauddin Zakariya UniversityMultanPakistan
| | - Ahmad Mujtaba Noman
- Department of Human Nutrition, Faculty of Food Science and NutritionBahauddin Zakariya UniversityMultanPakistan
- TIMES Institute MultanMultanPakistan
| | - Shehnshah Zafar
- Department of Human Nutrition, Faculty of Food Science and NutritionBahauddin Zakariya UniversityMultanPakistan
| | - Naima Tariq
- Department of Food Science and Technology, Faculty of Food Science and NutritionBahauddin Zakariya UniversityMultanPakistan
| | - Muzzamal Hussain
- Department of Food ScienceGovernment College University FaisalabadFaisalabadPakistan
| | - Muhammad Imran
- Department of Food Science and TechnologyUniversity of NarowalNarowalPakistan
| | - Ahmed Mujtaba
- Department of Food Sciences and Technology, Faculty of Engineering Sciences and TechnologyHamdard University Islamabad CampusIslamabadPakistan
| | - Tadesse Fenta Yehuala
- Faculty of Chemical and Food EngineeringBahir Dar Institute of Technology, Bahir Dar UniversityBahir DarEthiopia
| | - Ehab M. Mostafa
- Department of Pharmacognosy, College of PharmacyJouf UniversitySakakaSaudi Arabia
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical SciencesJouf UniversitySakakaSaudi Arabia
| | - Soad K. Al Jaouni
- Department of Hematology/Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of MedicineKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Suliman A. Alsagaby
- Department of Medical Laboratory Sciences, College of Applied Medical SciencesMajmaah UniversityAl‐MajmaahSaudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of MedicineQassim UniversityBuraidahSaudi Arabia
| |
Collapse
|
35
|
Capatina TF, Oatu A, Babasan C, Trifu S. Translating Molecular Psychiatry: From Biomarkers to Personalized Therapies-A Narrative Review. Int J Mol Sci 2025; 26:4285. [PMID: 40362522 PMCID: PMC12072283 DOI: 10.3390/ijms26094285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/10/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
In this review, we explore the biomarkers of different psychiatric disorders, such as major depressive disorder, generalized anxiety disorder, schizophrenia, and bipolar disorder. Moreover, we show the interplay between genetic and environmental factors. Novel techniques such as genome-wide association studies (GWASs) have identified numerous risk loci and single-nucleotide polymorphisms (SNPs) implicated in these conditions, contributing to a better understanding of their mechanisms. Moreover, the impact of genetic variations on drug metabolisms, particularly through cytochrome P450 (CYP450) enzymes, highlights the importance of pharmacogenomics in optimizing psychiatric treatment. This review also explores the role of neurotransmitter regulation, immune system interactions, and metabolic pathways in psychiatric disorders. As the technology advances, integrating genetic markers into clinical practice will be crucial in advancing precision psychiatry, improving diagnostic accuracy and therapeutic interventions for individual patients.
Collapse
Affiliation(s)
| | - Anamaria Oatu
- Department of Psychiatry, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.O.); (C.B.)
| | - Casandra Babasan
- Department of Psychiatry, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.O.); (C.B.)
| | - Simona Trifu
- Department of Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
36
|
Morgan MJ, Kim YS. RIPK3 in necroptosis and cancer. Mol Cells 2025; 48:100199. [PMID: 40010643 PMCID: PMC11938148 DOI: 10.1016/j.mocell.2025.100199] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025] Open
Abstract
Receptor-interacting protein kinase-3 is essential for the cell death pathway called necroptosis. Necroptosis is activated by the death receptor ligands and pattern recognition receptors of the innate immune system, leading to significant consequences in inflammation and in diseases, particularly cancer. Necroptosis is highly proinflammatory compared with other modes of cell death because cell membrane integrity is lost, resulting in releases of cytokines and damage-associated molecular patterns that potentiate inflammation and activate the immune system. We discuss various ways that necroptosis is triggered along with its potential role in cancer and therapy.
Collapse
Affiliation(s)
- Michael J Morgan
- Department of Natural Sciences, Northeastern State University, Tahlequah, OK 74464, USA.
| | - You-Sun Kim
- Department of Biochemistry, Ajou University School of Medicine, Ajou University, Suwon 16499, Korea; Department of Biomedical Sciences, Graduate School, Ajou University, Suwon 16499, Korea.
| |
Collapse
|
37
|
Michael FA, Jung M, Reimers M, Oschwald C, Mihm U, Welsch C, Walter D, Finkelmeier F, Masseli J, Pathil A, Dultz G, Puhlmann D, Diaz-Martinez L, Hack D, Lingwal N, Bojunga J, Kempf VA, Zeuzem S, Friedrich-Rust M. A New Drying Method of Thermolabile Flexible Endoscope Channels by Laminar and Turbulent Airflow: A Prospective, Randomized-Controlled, Single-Center, Proof-of-Concept Trial. Am J Gastroenterol 2025; 120:1027-1035. [PMID: 39311431 PMCID: PMC12043260 DOI: 10.14309/ajg.0000000000003093] [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: 02/15/2024] [Accepted: 09/12/2024] [Indexed: 11/02/2024]
Abstract
INTRODUCTION International guidelines suggest different possibilities for drying of endoscopes during reprocessing. Clinical results of these available drying methods are not satisfactory. The aim of this study was to compare the drying cycle of a standard endoscope washer-disinfector (EWD) (standard drying method [SD]) with a shortened mandatory drying by the EWD followed by a special drying device using laminar and turbulent air flow (novel drying method [ND]). METHODS Sixty endoscopes (duodenoscopes, colonoscocopes, and gastroscopes) from 3 different manufacturers underwent high-level disinfection and drying depending on the randomization group. Operational time of drying was measured for both groups. Residual fluid in the channels was measured using a laboratory scale. After a 14-day storage period, a sample of the endoscope channels was obtained to determine bacterial contamination. RESULTS ND had significantly fewer residual water in endoscope channels (SD: 90% vs ND: 0%; P < 0.001) after high-level disinfection and drying and less bacterial contamination after storage for 14 days (SD: 47% vs ND: 20%; P = 0.028). Time consumed for drying in ND was also significantly shorter (SD: 16 minutes 4 seconds vs ND: 5 minutes 59 seconds; P < 0.001). DISCUSSION Drying with a special automatic drying device was superior compared with an EWD's drying program as evidenced by no measurable residual water, reduced microbiological contamination, and a more than 2-fold decrease in operational time. Thus, drying by laminar and turbulent airflow may represent an attractive alternative to the currently used standard approach in the reprocessing process of flexible endoscopes.
Collapse
Affiliation(s)
- Florian A. Michael
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Germany;
| | - Michael Jung
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Germany;
| | - Mike Reimers
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Germany;
| | - Clara Oschwald
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Germany;
| | - Ulrike Mihm
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Germany;
| | - Christoph Welsch
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Germany;
| | - Dirk Walter
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Germany;
| | - Fabian Finkelmeier
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Germany;
| | - Johannes Masseli
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Germany;
| | - Anita Pathil
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Germany;
| | - Georg Dultz
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Germany;
| | - Danuta Puhlmann
- Goethe University Frankfurt, University Hospital, Institute for Medical Microbiology and Infection Control, Germany and and University Center of Competence for Infection Control of the State of Hessen, Frankfurt Main, Germany;
| | - Laura Diaz-Martinez
- Goethe University Frankfurt, University Hospital, Institute for Medical Microbiology and Infection Control, Germany and and University Center of Competence for Infection Control of the State of Hessen, Frankfurt Main, Germany;
| | - Daniel Hack
- Goethe University Frankfurt, University Hospital, Institute for Medical Microbiology and Infection Control, Germany and and University Center of Competence for Infection Control of the State of Hessen, Frankfurt Main, Germany;
| | - Neelam Lingwal
- Goethe University Frankfurt, University Hospital, Institute of Biostatistics and Mathematical Modeling, Germany.
| | - Jörg Bojunga
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Germany;
| | - Volkhard A.J. Kempf
- Goethe University Frankfurt, University Hospital, Institute for Medical Microbiology and Infection Control, Germany and and University Center of Competence for Infection Control of the State of Hessen, Frankfurt Main, Germany;
| | - Stefan Zeuzem
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Germany;
| | | |
Collapse
|
38
|
Zhao B, Shi G, Shi J, Li Z, Xiao Y, Qiu Y, He L, Xie F, Yu D, Cao H, Du H, Zhang J, Zhou Y, Jiang C, Li W, Li M, Wang Z. Research progress on the mechanism and treatment of cachexia based on tumor microenvironment. Nutrition 2025; 133:112697. [PMID: 39999652 DOI: 10.1016/j.nut.2025.112697] [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: 12/07/2024] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025]
Abstract
Cachexia is a prevalent multifactorial syndrome characterized by a substantial decrease in food intake, which results from processes such as proteolysis, lipolysis, inflammatory activation, and autophagy, ultimately leading to weight loss. In cancer patients, this condition is referred to as cancer-related cachexia (CRC) and affects over 50% of this population. A comprehensive understanding of the intricate interactions between tumors and the host organism is essential for the development of effective treatments for tumor cachexia. This review aims to elucidate the role of the tumor microenvironment (TME) in the pathogenesis of tumor-associated cachexia and to summarize the current evidence supporting treatment modalities that target the TME.
Collapse
Affiliation(s)
- Bochen Zhao
- School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an, China
| | - Gege Shi
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Jiaxin Shi
- School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an, China
| | - Zhaozhao Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Yang Xiao
- Department of Experiment Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yueyuan Qiu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Lei He
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Fei Xie
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Duo Yu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Haiyan Cao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Haichen Du
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Jieyu Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Yang Zhou
- School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an, China
| | - Caiyi Jiang
- School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an, China
| | - Weina Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Meng Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Zhaowei Wang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
39
|
He D, Chen S, Wang X, Wen X, Gong C, Liu L, He G. Icaritin Represses Autophagy to Promote Colorectal Cancer Cell Apoptosis and Sensitized Low-Temperature Photothermal Therapy via Targeting HSP90-TXNDC9 Interactions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412953. [PMID: 40184625 PMCID: PMC12120733 DOI: 10.1002/advs.202412953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/10/2025] [Indexed: 04/06/2025]
Abstract
Colorectal cancer (CRC) ranks among the leading causes of cancer-related dea ths worldwide, and the rising incidence and mortality of CRC underscores the urgent need for better understanding and management strategies. Icaritin (ICA) is the metabolites of icariin, a natural flavonoid glycoside compound derived from the stems and leaves of Epimedium. It has broad spectrum antitumor activity and inhibits the proliferation, migration, and invasion of CRC cells, and causes S phase cell cycle arrest. It exerts its antitumor effects against CRC through repressing autophagy to promote CRC cell apoptosis via interfering the HSP90-TXNDC9 interactions. The safety and efficacy of ICA are also affirmed in a mouse xenograft model. Additionally, to test whether ICA exerts synergistic effects with low-temperature photothermal therapy (LTPTT), a novel nanodrug delivery system, employing SiO2 nanocarriers, is designed aiming to load ICA with photothermal materials polydopamine (PDA), and folic acid (FA). This SiO2/Ica-PDA-FA multifunctional nanocomposite actively targets tumor tissues through the high affinity of FA for cancer cells. Once internalized, the acidic intracellular environment triggers the controlled release of ICA, inhibiting HSP90-TXNDC9 interactions. By LTPTT and ICA drug therapy under near-infrared illumination, a dual synergistic antitumor effect is achieved, holding promise for enhancing therapeutic outcomes in CRC treatment.
Collapse
Affiliation(s)
- Dan He
- Division of Head & Neck Tumor Multimodality TreatmentCancer Center and Department of Dermatology & VenerologyWest China HospitalSichuan UniversityChengdu610041China
- Department of OncologyThe Second Affiliated Hospital of Chengdu Medical CollegeNuclear Industry 416 HospitalChengdu610051China
| | - Siliang Chen
- Division of Head & Neck Tumor Multimodality TreatmentCancer Center and Department of Dermatology & VenerologyWest China HospitalSichuan UniversityChengdu610041China
- Laboratory of DermatologyClinical Institute of Inflammation and ImmunologyFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Xiaoyun Wang
- Division of Head & Neck Tumor Multimodality TreatmentCancer Center and Department of Dermatology & VenerologyWest China HospitalSichuan UniversityChengdu610041China
- Laboratory of DermatologyClinical Institute of Inflammation and ImmunologyFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Xiang Wen
- Division of Head & Neck Tumor Multimodality TreatmentCancer Center and Department of Dermatology & VenerologyWest China HospitalSichuan UniversityChengdu610041China
- Laboratory of DermatologyClinical Institute of Inflammation and ImmunologyFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Changyang Gong
- Division of Head & Neck Tumor Multimodality TreatmentCancer Center and Department of Dermatology & VenerologyWest China HospitalSichuan UniversityChengdu610041China
| | - Lei Liu
- Division of Head & Neck Tumor Multimodality TreatmentCancer Center and Department of Dermatology & VenerologyWest China HospitalSichuan UniversityChengdu610041China
| | - Gu He
- Division of Head & Neck Tumor Multimodality TreatmentCancer Center and Department of Dermatology & VenerologyWest China HospitalSichuan UniversityChengdu610041China
- Laboratory of DermatologyClinical Institute of Inflammation and ImmunologyFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
- Institute of Precision Drug Innovation and Cancer CenterThe Second Hospital of Dalian Medical UniversityDalian116023China
| |
Collapse
|
40
|
Zhang Y, Qian C, Chu C, Yang XZ, Wu Y, Cai L, Yao S, He W, Guo Z, Chen Y. Self-Assembly of Short Peptides Activates Specific ER-Phagy and Induces Pyroptosis for Enhanced Tumor Immunotherapy. Angew Chem Int Ed Engl 2025; 64:e202422874. [PMID: 40069115 DOI: 10.1002/anie.202422874] [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: 11/24/2024] [Revised: 02/21/2025] [Accepted: 03/11/2025] [Indexed: 03/25/2025]
Abstract
Developing specific endoplasmic reticulum-autophagy (ER-phagy) inducers is highly desirable for discovering new ER-phagy receptors and elucidating the detailed ER-phagy mechanism and potential cancer immunotherapy. However, most of the current ER-phagy-inducing methods cause nonselective autophagy of other organelles. In this work, we report the design and synthesis of simple and stable short peptides (D-FFxFFs) that could specifically trigger ER-phagy, which further induces pyroptosis and activates the immune response against tumor cells. D-FFxFFs locate preferentially in ER and readily self-assemble to form nanosized misfolded protein mimics, which lead to distinct upregulation of dedicated ER-phagy receptors with no obvious autophagy of other organelles. Significant unfolded protein response (UPR) is activated via IRE1-JNK and PERK-ATF4 pathways. Interestingly, the persistent ER-phagy triggers ER Ca2+ release and a surge in mitochondrial Ca2+ levels, resulting in GSDMD-mediated pyroptosis other than apoptosis. The ER-phagy induces pyroptosis and activates a distinct antitumor immune response without evolving the acquired drug resistance. This work not only provides a powerful tool for investigating the mechanism and function of ER-phagy but also offers an appealing strategy for anticancer immunotherapy.
Collapse
Affiliation(s)
- Yunhua Zhang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
| | - Chengyuan Qian
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
| | - Chengyan Chu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
| | - Xiu-Zhi Yang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
| | - Yanping Wu
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
| | - Linxiang Cai
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
| | - Shankun Yao
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
| | - Weijiang He
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
- Nanchuang (Jiangsu) Institute of Chemistry and Health, Nanjing, 210000, P.R. China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
- Nanchuang (Jiangsu) Institute of Chemistry and Health, Nanjing, 210000, P.R. China
| | - Yuncong Chen
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
- Nanchuang (Jiangsu) Institute of Chemistry and Health, Nanjing, 210000, P.R. China
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, Jiangsu, 210008, P.R. China
| |
Collapse
|
41
|
Dong Z, Li O, Li Y, Xiao Z, Li F, Xu S, Ji D. Dynamic adenoma detection rate influences the risk of metachronous advanced neoplasia after removal of low-risk findings in screening colonoscopy. Surg Endosc 2025; 39:3354-3363. [PMID: 40246761 PMCID: PMC12041121 DOI: 10.1007/s00464-025-11732-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 04/06/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND Endoscopists' adenoma detection rate (ADR) may change over time and dynamic ADR has been identified as a predictor for post-screening colorectal cancer. However, whether low-risk findings removed by an endoscopist with a lower dynamic ADR benefit from a shorter duration of follow-up requires further research. METHODS We conducted a two-center retrospective study of individuals who had low-risk findings removed and underwent subsequent surveillance colonoscopy. Endoscopists' dynamic ADR was the ADR of the previous 100 screening colonoscopies performed by the same endoscopist. A Cox-regression model and Kaplan-Meier survival analysis were used to explore the relationship between dynamic ADR and metachronous advanced colorectal neoplasia (ACRN). RESULTS Totally, 3471 individuals who had low-risk findings removed in baseline colonoscopy were included in analysis. Decreasing endoscopists' dynamic ADR was independently associated with metachronous ACRN. A 3.97-, 2.21-, and 2.67-fold increased risk for metachronous ACRN was observed in individuals of which baseline colonoscopy was performed by an endoscopist with a dynamic ADR of < 15%, 15-19%, and 20-24%, respectively, compared with those with the highest dynamic ADR (≥ 25%). The cumulative incidence of metachronous ACRN reached the 5% threshold at 4.5 years, 7.3 years, and 6.2 years in the dynamic ADR < 15%, 15-19%, and 20-24% group, respectively. CONCLUSION Endoscopists' dynamic ADR influences the risk of metachronous ACRN after removal of low-risk findings in screening colonoscopy. Individuals undergoing removal of low-risk findings in screening colonoscopy by an endoscopist with a dynamic ADR < 25% may benefit from a shorter duration of follow-up interval.
Collapse
Affiliation(s)
- Zhiyu Dong
- Department of Gastrointestinal Endoscopy, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Ouyang Li
- Department of Gastrointestinal Endoscopy, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yanglei Li
- Department of Gastrointestinal Endoscopy, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Zili Xiao
- Department of Gastrointestinal Endoscopy, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Feng Li
- Department of Gastrointestinal Endoscopy, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Shuchang Xu
- Department of Gastroenterology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Danian Ji
- Department of Gastrointestinal Endoscopy, Huadong Hospital Affiliated to Fudan University, Shanghai, China.
| |
Collapse
|
42
|
Jeon E, Kim SH, Kim S, Ryu DS, Kim JW, Kim K, Kim DH, Park J, Lee J. Hydrogel-Impregnated Robust Interlocking Nano Connector (HiRINC) for Noninvasive Anti-Migration of Esophageal Stent. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414944. [PMID: 40072298 PMCID: PMC12087745 DOI: 10.1002/adma.202414944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 02/18/2025] [Indexed: 05/20/2025]
Abstract
Migration of implanted self-expandable metallic stent (SEMS) in the malignant or benign esophageal stricture is a common complication but not yet resolved. Herein, this research develops a hydrogel-impregnated robust interlocking nano connector (HiRINC) to ensure adhesion and reduce the mechanical mismatch between SEMSs and esophageal tissues. Featuring a network-like porous layer, HiRINC significantly enhances adhesion and energy dissipation during esophageal peristalsis by utilizing mechanical interlocking and increasing hydrogen bonding sites, thereby securing SEMS to tissues. The anti-swelling property of HiRINC prevents excessive hydrogel expansion, avoiding esophageal blockage. Ex vivo and in vivo adhesion tests confirm that the HiRINC outperforms flat surfaces without RINC structures and effectively prevents stent migration. HiRINC-coated SEMS maintains its position and luminal patency, minimizing stent-induced tissue hyperplasia and inflammatory responses in rat and porcine esophageal models during the 4-week follow-up. This novel HiRINC-SEMS can ensure anti-migration and prolonged stent patency in the rat and porcine esophagus and seems to be expanded to other nonvascular luminal organs and various implantable metallic devices.
Collapse
Affiliation(s)
- Eunyoung Jeon
- Department of ChemistryHanyang UniversitySeoul04763Republic of Korea
- VN IncSeoul04763Republic of Korea
| | - Song Hee Kim
- Department of Convergence MedicineAsan Medical CenterUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gil, Songpa‐guSeoul05505Republic of Korea
- Biomedical Engineering Research CenterAsan Institute for Life SciencesAsan Medical Center88 Olympic‐ro 43‐gil, Songpa‐guSeoul05505Republic of Korea
| | - Sukyoung Kim
- Department of ChemistryHanyang UniversitySeoul04763Republic of Korea
| | - Dae Sung Ryu
- Department of Convergence MedicineAsan Medical CenterUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gil, Songpa‐guSeoul05505Republic of Korea
- Biomedical Engineering Research CenterAsan Institute for Life SciencesAsan Medical Center88 Olympic‐ro 43‐gil, Songpa‐guSeoul05505Republic of Korea
| | - Ji Won Kim
- Department of Convergence MedicineAsan Medical CenterUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gil, Songpa‐guSeoul05505Republic of Korea
- Biomedical Engineering Research CenterAsan Institute for Life SciencesAsan Medical Center88 Olympic‐ro 43‐gil, Songpa‐guSeoul05505Republic of Korea
| | - Kayoung Kim
- Department of Fiber Convergence Material EngineeringDankook UniversityGyeonggi‐Do16890Republic of Korea
| | - Do Hoon Kim
- Department of GastroenterologyAsan Medical CenterUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gil, Songpa‐guSeoul05505Republic of Korea
| | - Jung‐Hoon Park
- Department of Convergence MedicineAsan Medical CenterUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gil, Songpa‐guSeoul05505Republic of Korea
- Biomedical Engineering Research CenterAsan Institute for Life SciencesAsan Medical Center88 Olympic‐ro 43‐gil, Songpa‐guSeoul05505Republic of Korea
| | - Joonseok Lee
- Department of ChemistryHanyang UniversitySeoul04763Republic of Korea
- Research Institute for Convergence of Basic SciencesHanyang UniversitySeoul04763Republic of Korea
- VN IncSeoul04763Republic of Korea
| |
Collapse
|
43
|
Wach AA, Storman D, Więckowski K, Wojtaszek-Główka M, Żabicka W, Krupka K, Abadi AK, Celińska-Löwenhoff M, Bala MM. The efficacy of probiotics, prebiotics or synbiotics in scleroderma: A systematic review. Clin Nutr ESPEN 2025; 68:206-215. [PMID: 40311922 DOI: 10.1016/j.clnesp.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 05/03/2025]
Abstract
INTRODUCTION Pathogenesis of systemic scleroderma is a complex subject. Previous research has emphasized a possible contribution of the intestinal microbiome in developing symptoms. The use of probiotic formulas brings benefit in treatment of various autoimmune diseases, but the evidence for scleroderma is still not exhaustive. No official recommendations have been formulated on this topic, nor has the existing evidence been evaluated. OBJECTIVE We aimed to assess the efficacy of probiotics, prebiotics, or synbiotics in patients with scleroderma with a focus on the improvement of symptoms and change in the quality of life (QoL) both measured using validated scales, small intestinal bacterial overgrowth (SIBO) eradication, as well as change in the intensity of interstitial lung disease and occurrence of pulmonary hypertension. METHODS We performed a systematic search for randomized trials that assessed the beneficial and harmful effects of pro-, pre-, and synbiotics in patients with scleroderma. Pairs of authors independently selected studies for inclusion, extracted data, and assessed the risk of bias (RoB) of the included studies. We examined the certainty of evidence in accordance with the GRADE approach. RESULTS We screened 1801 references (after the removal of duplicates), and assessed 16 citations in full text. We identified four randomized controlled trials (RCTs) with 103 participants in intervention and 87 in control groups. The evidence suggests that probiotic intake may not affect gastrointestinal symptoms measured with Visual Analogue Scale for Gastrointestinal Tract (VAS-GIT) (change in the VAS-GIT: MD 0.04, 95% CI -0.31 to 0.40, low certainty, n = 56 probiotic group, n = 57 placebo group) and the evidence is very uncertain about the effect on gastrointestinal symptoms measured with The University of California Los Angeles Scleroderma Clinical Trial Consortium GIT 2.0 instrument (UCLA GIT 2.0) (change in the UCLA GIT 2.0 - total score: MD -0.18 95% CI -0.44, 0.07, very low certainty, n = 76 probiotic group, n = 74 placebo group). Meta-analysis showed that probiotic intake may slightly improve the social functioning measured with UCLA GIT 2.0 compared to placebo (MD -0.21, 95% CI -0.32 to -0.10, low certainty, n = 57 probiotic group, n = 53 placebo group), yet may result in little to no difference in physical functioning measured with Health Assessment Questionnaire-Disability Index (HAQ-DI) (MD 0.08, 95% CI -0.15 to 0.32, low certainty, n = 56 probiotic group, n = 57 placebo group) and frequency of adverse events, change in QoL and SIBO eradication. We did not find any studies addressing the effect of pro-, pre-, or synbiotics on interstitial lung disease or pulmonary hypertension. The certainty of evidence was low to very low mainly due to high RoB (lack of blinding, incomplete reporting) and imprecision. - CONCLUSIONS We were unable to definitively prove a positive or negative effect of probiotic intake in patients with systemic sclerosis. The results should be interpreted with caution due to low or very low certainty of evidence and would need more confirmation from strain and dose specific RCTs at a lower RoB.
Collapse
Affiliation(s)
- Adam A Wach
- Students' Scientific Research Group, Systematic Reviews Unit, Jagiellonian University Medical College, Krakow, Poland
| | - Dawid Storman
- Department of Hygiene and Dietetics, Chair of Epidemiology and Preventive Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Krzysztof Więckowski
- Students' Scientific Research Group, Systematic Reviews Unit, Jagiellonian University Medical College, Krakow, Poland
| | - Magdalena Wojtaszek-Główka
- Students' Scientific Research Group, Systematic Reviews Unit, Jagiellonian University Medical College, Krakow, Poland
| | - Wioletta Żabicka
- Students' Scientific Research Group, Systematic Reviews Unit, Jagiellonian University Medical College, Krakow, Poland
| | - Klaudia Krupka
- Students' Scientific Research Group, Systematic Reviews Unit, Jagiellonian University Medical College, Krakow, Poland
| | - Ahmad Kamal Abadi
- Department of Computer Engineering, Faculty of Engineering, University of Arak, Arak, Iran
| | - Magdalena Celińska-Löwenhoff
- II Chair of Internal Medicine of prof. A. Szczeklik, Department of Allergy, Autoimmunisation and Hypercoagulability, Faculty of Medicine Jagiellonian University Medical College, Krakow, Poland
| | - Malgorzata M Bala
- Department of Hygiene and Dietetics, Chair of Epidemiology and Preventive Medicine, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
44
|
Wu J, Zhou X, Tsang CY, Mei Q, Zhang Y. Bioengineered nanomaterials for dynamic diagnostics in vivo. Chem Soc Rev 2025. [PMID: 40289891 DOI: 10.1039/d5cs00136f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
In vivo diagnostics obtains real-time physiological information directly from the site of interest in a patient's body, providing more accurate disease diagnosis compared with ex vivo diagnostics. Particularly, in vivo dynamic diagnostics allows the continuous monitoring of physiological signals over a period of time, offering deeper insights into disease pathogenesis and progression. However, achieving in situ dynamic diagnostics in deep tissues presents challenges related to energy and signal penetration as well as dynamic monitoring. Bioengineered nanomaterials serve as an ideal platform for in vivo dynamic diagnostics, leveraging energy conversion and biofunctionalization to enable continuous acquisition of physiological information across temporal and spatial scales. In this review, with reference to the studies from the last five years, we summarize the fundamental components that are essential for dynamic diagnosis in vivo. Firstly, an input energy source with high tissue penetration is needed, such as near-infrared (NIR) light, X-rays, magnetic field and ultrasound. Secondly, a nanomaterial class that is responsive to such an energy source to provide a readable output signal is chosen. Thirdly, bioengineered nanoprobes are designed to exhibit spatial, temporal or spatiotemporal changes in the output signal. Finally, different methods are used to analyse the output signal of nanoprobes, such as detecting changes in optical, radiation, magnetic and ultrasound signals. This review also discusses the obstacles and potential solutions for advancing these bioengineered nanomaterials toward clinical translational applications.
Collapse
Affiliation(s)
- Jizhong Wu
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 117583, Singapore
| | - Xinyu Zhou
- Department of Biomedical Engineering, College of Biomedicine, The City University of Hong Kong, Kowloon 999077, Hong Kong.
| | - Chung Yin Tsang
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 117583, Singapore
| | - Qingsong Mei
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China.
| | - Yong Zhang
- Department of Biomedical Engineering, College of Biomedicine, The City University of Hong Kong, Kowloon 999077, Hong Kong.
| |
Collapse
|
45
|
Pu Y, Wang Q, Pan Y, Wang X, Guan Z, Zhu Y, Yang Z. Cytidinyl/Cationic Lipid Encapsulating Insulin-Like Growth Factor 1 Receptor siRNA for Hepatocellular Carcinoma Therapy. Mol Pharm 2025. [PMID: 40289542 DOI: 10.1021/acs.molpharmaceut.4c01318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent form of invasive liver cancer, representing over 90% of all liver cancer cases. Currently, there is a lack of targeted therapy for HCC. Insulin-like growth factor 1 receptor (IGF1R) is abnormally expressed in HCC, leading to the malignant proliferation and contributing to the antiapoptosis mechanisms in tumor cells. In this study, small interfering RNAs targeting IGF1R mRNA (siIGF1Rs) have been designed. Additionally, a full 2'-F/2'-OMe modification with partial phosphorothioation was applied to improve the biological properties of these siIGF1Rs. Based on previous research, stable lipid complexes with uniform particle sizes were constructed using cytidinyl lipid DNCA/cationic lipid CLD (Mix) supplemented with DSPE-PEG (siIGF1R/Mix/PEG). The complexes were formed through hydrogen-bonding, π-π stacking, and electrostatic interactions. The siIGF1R/Mix/PEG complex entered the cytoplasm and nucleus of HCC cells, reduced IGF1R mRNA and pre-mRNA levels by over 95% and 50% respectively, further arrested the cell cycle in the S phase, and promoted cell apoptosis. Importantly, siIGF1R/Mix/PEG (0.8 mg/kg, i.v.) selectively accumulated in the tumor, significantly inhibiting tumor growth by 91.31% compared to the naked siRNA group, with slower release and a more prolonged effect.
Collapse
Affiliation(s)
- Yang Pu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Quanxin Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yufei Pan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xixian Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhu Guan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yuejie Zhu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Genable (Beijing) Biotechnology Co. Ltd, #38 Yongda Road, Beijing 102609, China
| | - Zhenjun Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Genable (Beijing) Biotechnology Co. Ltd, #38 Yongda Road, Beijing 102609, China
| |
Collapse
|
46
|
Piccirelli S, Salvi D, Pugliano CL, Tettoni E, Facciorusso A, Rondonotti E, Mussetto A, Fuccio L, Cesaro P, Spada C. Unmet Needs of Artificial Intelligence in Small Bowel Capsule Endoscopy. Diagnostics (Basel) 2025; 15:1092. [PMID: 40361910 PMCID: PMC12071857 DOI: 10.3390/diagnostics15091092] [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: 03/31/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Small bowel capsule endoscopy (SBCE) has emerged in the past two decades as the cornerstone for assessing small bowel disorders, and its use is supported by several guidelines. However, there are several limitations, such as the considerable time required for gastroenterologists to review these videos and reach a diagnosis. To address these limitations, researchers have explored the integration of artificial intelligence in the interpretation of these videos. In our review, we explore the evolving and emerging role of artificial intelligence in SBCE and examine the latest advancements and ongoing studies in these areas, aiming at overcoming current limitations.
Collapse
Affiliation(s)
- Stefania Piccirelli
- Department of Gastroenterology and Endoscopy, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (S.P.); (C.L.P.); (E.T.); (P.C.)
| | - Daniele Salvi
- Department of Gastroenterology and Endoscopy, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (S.P.); (C.L.P.); (E.T.); (P.C.)
| | - Cecilia Lina Pugliano
- Department of Gastroenterology and Endoscopy, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (S.P.); (C.L.P.); (E.T.); (P.C.)
| | - Enrico Tettoni
- Department of Gastroenterology and Endoscopy, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (S.P.); (C.L.P.); (E.T.); (P.C.)
| | - Antonio Facciorusso
- Department of Experimental Medicine, Università del Salento, 73100 Lecce, Italy
| | | | - Alessandro Mussetto
- Gastroenterology Unit, Santa Maria delle Croci Hospital, 48121 Ravenna, Italy;
| | - Lorenzo Fuccio
- Gastroenterology Unit, University of Bologna, 40136 Bologna, Italy;
| | - Paola Cesaro
- Department of Gastroenterology and Endoscopy, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (S.P.); (C.L.P.); (E.T.); (P.C.)
| | - Cristiano Spada
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
47
|
Ge MW, Chen HL. Unveiling the Potential: Suggestions for Refining the Bowel Preparation Study on 1 L PEG-Asc Combinations. Am J Gastroenterol 2025:00000434-990000000-01715. [PMID: 40272010 DOI: 10.14309/ajg.0000000000003452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Affiliation(s)
- Meng-Wei Ge
- School of Nursing and Rehabilitation, Nantong University, Nantong, Jiangsu, PR China
| | | |
Collapse
|
48
|
Sruthi KK, Ummanni R. Valosin-Containing Protein (VCP/p97) Mediates Neuroendocrine Differentiation in Prostate Cancer Cells Through Pim1 Signaling Inducing Autophagy. Prostate 2025. [PMID: 40269472 DOI: 10.1002/pros.24900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/19/2025] [Accepted: 03/28/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Neuroendocrine Prostate Cancer (NEPC) is an aggressive type of androgen-independent prostate cancer (AIPC) associated with resistance to treatment. Valosin-containing protein (VCP/p97) has been found to be overexpressed in prostate cancer (PCa) cells undergoing neuroendocrine differentiation (NED) in response to interleukin-6 (IL-6). This study explores the molecular mechanisms through which VCP/p97 contributes to the progression of NEPC. METHODS To investigate the role of VCP/p97 in the NED of PCa, we overexpressed the VCP/p97 in PCa cells. The molecular mechanisms underlying VCP/p97 induced NED were assessed by using western blot analysis and RT-PCR. Morphological changes were analyzed by using both bright field and confocal microscope. Lysotracker staining was performed to identify autophagy in VCP positive PCa cells. RESULTS In the present study, we found that VCP/p97 expression was notably higher in neuroendocrine (NE) cells NCI-H660 and PC3 than in other PCa cells. IL-6 treatment led to significant VCP/p97 overexpression in LNCaP and VCaP cells, with a marked increase in NE markers NSE and CHR-A. Inhibition of VCP/p97 using NMS-873 attenuated NED features, suggesting that VCP/p97 is required for NED progression. Moreover, VCP's role in NED is linked to its regulation via Pim1 in differentiating cells. Exogenous expression of VCP/p97 enhanced Pim1 and c-Myc expression, which were diminished upon VCP/p97 inhibition which is corroborated by reduced NED markers. Pim1 inhibition using AZD1208 and c-Myc knockdown further supported Pim1's involvement in VCP mediated NED. To promote NED, VCP/p97 regulated autophagy, as evidenced by increased LC3B and decreased SQSTM1/p62 levels upon VCP overexpression. Inhibition of VCP/p97 or autophagy disrupted NED and autophagic flux, arresting NED of LNCaP cells. Lysotracker staining and autophagic flux assays confirmed VCP's role in enhancing lysosomal-mediated autophagy and autophagolysosome formation. Furthermore, we show that AMPK activation, via LKB1 is essential for VCP/p97 mediated NED and autophagy. CONCLUSION VCP drives NED in PCa cells through a complex interplay involving the Pim1 axis and autophagy pathways. These findings highlight the potential of targeting VCP/p97 and its associated mechanisms as therapeutic strategies to inhibit NED progression.
Collapse
Affiliation(s)
- K K Sruthi
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ramesh Ummanni
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
49
|
Michetti F, Cirone M, Strippoli R, D'Orazi G, Cordani M. Mechanistic insights and therapeutic strategies for targeting autophagy in pancreatic ductal adenocarcinoma. Discov Oncol 2025; 16:592. [PMID: 40266451 PMCID: PMC12018664 DOI: 10.1007/s12672-025-02400-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/15/2025] [Indexed: 04/24/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterised by early metastasis and resistance to anti-cancer therapy, leading to an overall poor prognosis. Macroautophagy (hereinafter referred to as autophagy) is a conserved cellular homeostasis mechanism that degrades various cargoes (e.g., proteins, organelles, and pathogens) mainly playing a role in promoting survival under environmental stress. Autophagy is an essential defense mechanism against PDAC initiation, acting on multiple levels to maintain cellular and tissue homeostasis. However, autophagy is also intimately involved in the molecular mechanisms driving PDAC progression, facilitating the adaptation of cancer cells to the tumor microenvironment's harsh conditions. In this review, we examine the complex role of autophagy in PDAC and assess the potential of modulating autophagy as a therapeutic strategy. By reviewing current research and clinical trials, we seek to elucidate how targeting autophagy can disrupt PDAC tumor survival mechanisms, enhance the efficacy of existing treatments, and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Federica Michetti
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, 00149, Rome, Italy
| | - Mara Cirone
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161, Rome, Italy
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, 00149, Rome, Italy.
| | - Gabriella D'Orazi
- UniCamillus-Saint Camillus International University of Health and Medical Sciences, Via di Sant'Alessandro 8, 00131, Rome, Italy.
- Department of Research and Advanced Technologies, Regina Elena National Cancer Institute IRCCS, Via Elio Chianesi 51, 00144, Rome, Italy.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040, Madrid, Spain.
| |
Collapse
|
50
|
Chen Y, Ballarò R, Sans M, Thege FI, Zuo M, Dou R, Min J, Yip-Schneider M, Zhang J, Wu R, Irajizad E, Makino Y, Rajapakshe KI, Rudsari HK, Hurd MW, León-Letelier RA, Katayama H, Ostrin E, Vykoukal J, Dennison JB, Do KA, Hanash SM, Wolff RA, Guerrero PA, Kim M, Schmidt CM, Maitra A, Fahrmann JF. Long-chain sulfatide enrichment is an actionable metabolic vulnerability in intraductal papillary mucinous neoplasm (IPMN)-associated pancreatic cancers. Gut 2025:gutjnl-2025-335220. [PMID: 40268349 DOI: 10.1136/gutjnl-2025-335220] [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: 03/04/2025] [Accepted: 04/05/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND We conducted an integrated cross-species spatial assessment of transcriptomic and metabolomic alterations associated with progression of intraductal papillary mucinous neoplasms (IPMNs), which are bona fide cystic precursors of pancreatic ductal adenocarcinoma (PDAC). OBJECTIVE We aimed to uncover biochemical and molecular drivers that underlie malignant progression of IPMNs to PDAC. DESIGN Matrix-assisted laser desorption/ionisation (MALDI) mass spectrometry (MS)-based spatial imaging and Visium spatial transcriptomics (ST) was performed on human resected IPMN/PDAC tissues (n=23) as well as pancreata from a mutant Kras;Gnas mouse model of IPMN/PDAC. Functional studies in murine IPMN/PDAC-derived Kras;Gnas cells were performed using CRISPR/cas9 technology, small interfering RNAs, and pharmacological inhibition. RESULTS MALDI-MS analyses of patient tissues revealed long-chain hydroxylated sulfatides to be selectively enriched in the neoplastic epithelium of IPMN/PDAC. Integrated ST analyses showed cognate transcripts involved in sulfatide biosynthesis, including UGT8, Gal3St1, and FA2H, to co-localise with areas of sulfatide enrichment. Genetic knockout or pharmacological inhibition of UGT8 in Kras;Gnas IPMN/PDAC cells decreased protein expression of FA2H and Gal3ST1 with consequent alterations in mitochondrial morphology and reduced mitochondrial respiration. Small molecule inhibition of UGT8 elicited anticancer effects via ceramide-mediated compensatory mitophagy and activation of intrinsic apoptosis pathways. In vivo, UGT8 inhibition suppressed tumour growth in allograft models of murine IPMN/PDAC cells derived from Kras;Gnas and Kras;Tp53;Gnas mice. CONCLUSION Our work identifies enhanced sulfatide metabolism as an early metabolic alteration in cystic precancerous lesions of the pancreas that persists through invasive neoplasia and a potential actionable vulnerability in IPMN-derived PDAC.
Collapse
Affiliation(s)
- Yihui Chen
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Riccardo Ballarò
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Marta Sans
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Fredrik Ivar Thege
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mingxin Zuo
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rongzhang Dou
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jimin Min
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michele Yip-Schneider
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - J Zhang
- Department of Epidemiology, Indiana University, Indianapolis, Indiana, USA
| | - Ranran Wu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ehsan Irajizad
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yuki Makino
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kimal I Rajapakshe
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hamid K Rudsari
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mark W Hurd
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ricardo A León-Letelier
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hiroyuki Katayama
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Edwin Ostrin
- Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jody Vykoukal
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennifer B Dennison
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Robert A Wolff
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Paolo A Guerrero
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - C Max Schmidt
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Anirban Maitra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Johannes F Fahrmann
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|