51
|
Gu L, Li S, Zhou L, Yuan F, Zhang T, Wang Y, Liu T, Li M, Zhang Z, Guo X. Ecophysiological and transcriptional landscapes of arbuscular mycorrhiza fungi enhancing yield, quality, and stalk rot resistance in Anoectochilus roxburghii. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2025; 223:109885. [PMID: 40220671 DOI: 10.1016/j.plaphy.2025.109885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 03/23/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Anoectochilus roxburghii (Wall.) Lindl. (A. roxburghii) is an increasingly popular medicinal herb. Arbuscular mycorrhiza (AM) fungi, known for their symbiotic relationships with plant roots, enhance nutrient uptake and disease resistance in host plants. However, their specific regulatory mechanisms in A. roxburghii are not fully understood. In this study, Fujian A. roxburghii was inoculated with the AM fungus Glomus intraradices, and successful root colonization was observed. Following AM fungal colonization, there was a significant upregulation of photosynthesis-related genes in the stems, accompanied by improved canopy phenotypes and root architecture. Consequently, AM-inoculated plants exhibited increased fresh and dry biomass, as well as elevated levels of polysaccharides and flavonoids. Additionally, the incidence of Fusarium oxysporum-induced stalk rot was reduced in AM-inoculated plants. Analysis of defense-related enzymes indicated that AM-inoculated plants exhibited a rapid and robust response to pathogen infection, mitigating oxidative stress. Transcriptomic analysis revealed significant upregulation of genes associated "Fatty acid degradation", "MAPK signaling pathway-plant", and "Plant-pathogen interaction", suggesting their involvement in enhanced disease resistance. A regulatory network centered on ACX1 and calmodulin, involving multiple transcription factors such as WRKY, bHLH, ERF, NAC, and HSF, was implicated in defense responses. These findings demonstrated the beneficial effects of AM fungi on yield, quality, and disease resistance in A. roxburghii, providing a theoretical foundation for its cultivation and genetic improvement.
Collapse
Affiliation(s)
- Li Gu
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Shurong Li
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Lichun Zhou
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Feiyue Yuan
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Tingting Zhang
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Yankun Wang
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Tiedong Liu
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Mingjie Li
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Zhongyi Zhang
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Xiaolei Guo
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| |
Collapse
|
52
|
Singh J, Pradhan P, Kataria A, Sinha S, Ehtesham NZ, Monk PN, Hasnain SE. Conservation of Putative Liquid-Liquid Phase Separating Proteins in Multiple Drug-Resistant Mycobacterium tuberculosis: Role in Host-Pathogen Interactions? ACS Infect Dis 2025. [PMID: 40183374 DOI: 10.1021/acsinfecdis.4c00722] [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: 04/05/2025]
Abstract
We observed a high proportion of proteins in pathogenic Mycobacterium species that can potentially undergo liquid-liquid phase separation (LLPS) mediated biomolecular condensate formation, compared to nonpathogenic species. These proteins mainly include the PE-PPE and PE-PGRS families of proteins that have nucleic acid and protein-protein binding functions, typical of LLPS proteins. We also mapped identified LLPS proteins in M. tuberculosis (M.tb) drug-resistant databases PubMLST and TBProfiler, based upon the WHO 2023 catalogue of resistance-associated mutations. High sequence conservation of LLPS-associated proteins in various multiple drug-resistant M.tb isolates points to their potentially important role in virulence and host-pathogen interactions during pathogenic evolution. This analysis provides a perspective on the role of protein phase separation in the evaluation of M.tb pathogenesis and offers avenues for future research aimed at developing innovative strategies to combat M.tb infection.
Collapse
Affiliation(s)
- Jasdeep Singh
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology-Delhi, New Delhi 110016, India
| | - Prashant Pradhan
- Laboratory of Nuclear Organization, Cecil H. and Ida Green Center for Reproductive Biology Sciences, Division of Basic Research, Department of Obstetrics and Gynecology, Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9096, United States
| | - Arti Kataria
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Hamilton, Montana 59840, United States
| | - Sanjeev Sinha
- Department of Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Nasreen Z Ehtesham
- Department of Life Science, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh 201310, India
| | - Peter N Monk
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, U.K
| | - Seyed E Hasnain
- Department of Life Science, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh 201310, India
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology-Delhi, New Delhi 110016, India
| |
Collapse
|
53
|
Li K, Zhang Y, Diao Y, Fan S. Paeonol regulates the DDIT4-mTOR signaling pathway in macrophages to promote diabetic wound healing. Int Immunopharmacol 2025; 151:114347. [PMID: 39987633 DOI: 10.1016/j.intimp.2025.114347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Diabetic foot ulcers are a common complication in people with diabetes, and patients with severe disease are at risk of amputation. Current studies have found that one of the reasons for the difficulty in healing diabetic foot ulcers is the Abnormal polarization of the M1/M2 phenotype of macrophages, which leads to a prolonged inflammatory period of the wound. The aim of this study was to investigate whether paeonol can promote the polarization of macrophages towards the M2 type and whether M2 type macrophages can regulate the DDIT4-mTOR signaling pathway and slow down the inflammatory response of diabetic foot ulcers. METHODS C57BL/6 mice were used to establish an animal model of diabetic foot ulcers and the effect of paeonol on wound healing was investigated. The effects of paeonol on wound healing of foot ulcer in diabetic mice were evaluated using histological staining and immunohistochemistry. The molecular mechanism of refractory healing of foot ulcers was speculated through network pharmacology. The effects of Paeonol on phenotypic polarization of macrophages and the mechanism of inhibiting inflammation were studied by q-PCR, ELISA, immunofluorescence and Western. RESULTS Paeonol can effectively promote wound healing in diabetic mice. HE staining showed that paeonol could improve the inflammatory infiltration in the ulcer wound of diabetic mice; Masson trichromatic staining showed that paeonol could increase the increase of muscle fibers and collagen in the wound tissue of diabetic mice; immunofluorescence results showed that paeonol could increase the angiogenesis in the wound tissue of diabetic mice. Network pharmacological analysis showed that the molecular mechanism of paeonol in treating diabetic wound healing may be through DDIT4-mTOR signaling pathway. q-PCR, ELISA, immunofluorescence and Western blot showed that paeonol could reduce the expression of the signature protein CD86 and inflammatory factors in M1 macrophages, and promote the phenotypic polarization of M2 macrophages, which is the mechanism of inhibiting inflammation by activating DDIT4-mTOR signaling pathway. CONCLUSION Paeonol can promote the polarization of macrophages towards M2 type, reduce inflammatory response and accelerate wound surface healing through DDIT4-mTOR signaling pathway, providing a new therapeutic strategy for the treatment of diabetic foot ulcers.
Collapse
Affiliation(s)
- Kun Li
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China.
| | - Yingying Zhang
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Yunpeng Diao
- College of Pharmacy, Dalian Medical University, Dalian 116044, China; Dalian anti-infective traditional Chinese Medicine Development Engineering Technology Research Center, China
| | - Shuyuan Fan
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China.
| |
Collapse
|
54
|
Huo Z, Fan C, Li K, Xu C, Niu Y, Wang F. Identification and validation of hub m7G-related genes and infiltrating immune cells in osteoarthritis based on integrated computational and bioinformatics analysis. BMC Musculoskelet Disord 2025; 26:333. [PMID: 40186163 PMCID: PMC11971809 DOI: 10.1186/s12891-025-08539-6] [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: 02/09/2024] [Accepted: 03/17/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a joint disease closely associated with synovial tissue inflammation, with the severity of synovitis impacting disease progression. m7G RNA methylation is critical in RNA processing, metabolism, and function, but its role in OA synovial tissue is not well understood. This study explores the relationship between m7G methylation and immune infiltration in OA. METHODS Data were obtained from the GEO database. Hub genes related to m7G were identified using differential expression and LASSO-Cox regression analysis, and a diagnostic model was developed. Functional enrichment, drug target prediction, and target gene-related miRNA prediction were performed for these genes. Immune cell infiltration was analyzed using the CIBERSORT algorithm, and unsupervised clustering analysis was conducted to examine immune infiltration patterns. RT-qPCR was used to validate hub gene expression. RESULTS Seven m7G hub genes (SNUPN, RNMT, NUDT1, LSM1, LARP1, CYFIP2, and CYFIP1) were identified and used to develop a nomogram for OA risk prediction. Functional enrichment indicated involvement in mRNA metabolism and RNA transport. Differences in macrophage and T-cell infiltration were observed between OA and normal groups. Two distinct m7G immune infiltration patterns were identified, with significant microenvironment differences between clusters. RT-qPCR confirmed differential hub gene expression. CONCLUSION A diagnostic model based on seven m7G hub genes was developed, highlighting these genes as potential biomarkers and significant players in OA pathogenesis.
Collapse
Affiliation(s)
- Zhenhui Huo
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| | - Chongyi Fan
- Department of Orthopedics, Aerospace Central Hospital, Beijing, 100049, China
| | - Kehan Li
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| | - Chenyue Xu
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| | - Yingzhen Niu
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China.
| | - Fei Wang
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China.
| |
Collapse
|
55
|
Nagarajan P, Winkler TW, Bentley AR, Miller CL, Kraja AT, Schwander K, Lee S, Wang W, Brown MR, Morrison JL, Giri A, O'Connell JR, Bartz TM, de Las Fuentes L, Gudmundsdottir V, Guo X, Harris SE, Huang Z, Kals M, Kho M, Lefevre C, Luan J, Lyytikäinen LP, Mangino M, Milaneschi Y, Palmer ND, Rao V, Rauramaa R, Shen B, Stadler S, Sun Q, Tang J, Thériault S, van der Graaf A, van der Most PJ, Wang Y, Weiss S, Westerman KE, Yang Q, Yasuharu T, Zhao W, Zhu W, Altschul D, Ansari MAY, Anugu P, Argoty-Pantoja AD, Arzt M, Aschard H, Attia JR, Bazzanno L, Breyer MA, Brody JA, Cade BE, Chen HH, Chen YDI, Chen Z, de Vries PS, Dimitrov LM, Do A, Du J, Dupont CT, Edwards TL, Evans MK, Faquih T, Felix SB, Fisher-Hoch SP, Floyd JS, Graff M, Gu C, Gu D, Hairston KG, Hanley AJ, Heid IM, Heikkinen S, Highland HM, Hood MM, Kähönen M, Karvonen-Gutierrez CA, Kawaguchi T, Kazuya S, Kelly TN, Komulainen P, Levy D, Lin HJ, Liu PY, Marques-Vidal P, McCormick JB, Mei H, Meigs JB, Menni C, Nam K, Nolte IM, Pacheco NL, Petty LE, Polikowsky HG, Province MA, Psaty BM, Raffield LM, Raitakari OT, Rich SS, Riha RL, Risch L, Risch M, Ruiz-Narvaez EA, Scott RJ, Sitlani CM, Smith JA, Sofer T, Teder-Laving M, Völker U, Vollenweider P, Wang G, Willems van Dijk K, Wilson OD, Xia R, Yao J, Young KL, Zhang R, Zhu X, Below JE, Böger CA, Conen D, Cox SR, Dörr M, Feitosa MF, Fox ER, Franceschini N, Gharib SA, Gudnason V, Harlow SD, He J, Holliday EG, Kutalik Z, Lakka TA, Lawlor DA, Lee S, Lehtimäki T, Li C, Liu CT, Mägi R, Matsuda F, Morrison AC, Penninx BW, Peyser PA, Rotter JI, Snieder H, Spector TD, Wagenknecht LE, Wareham NJ, Zonderman AB, North KE, Fornage M, Hung AM, Manning AK, Gauderman J, Chen H, Munroe PB, Rao DC, van Heemst D, Redline S, Noordam R, Wang H. A large-scale genome-wide study of gene-sleep duration interactions for blood pressure in 811,405 individuals from diverse populations. Mol Psychiatry 2025:10.1038/s41380-025-02954-w. [PMID: 40181193 DOI: 10.1038/s41380-025-02954-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 03/11/2025] [Indexed: 04/05/2025]
Abstract
Although both short and long sleep duration are associated with elevated hypertension risk, our understanding of their interplay with biological pathways governing blood pressure remains limited. To address this, we carried out genome-wide cross-population gene-by-short-sleep and long-sleep duration interaction analyses for three blood pressure traits (systolic, diastolic, and pulse pressure) in 811,405 individuals from diverse population groups. We discovered 22 novel gene-sleep duration interaction loci for blood pressure, mapped to 23 genes. Investigating these genes' functional implications shed light on neurological, thyroidal, bone metabolism, and hematopoietic pathways that necessitate future investigation for blood pressure management that caters to sleep health lifestyle. Non-overlap between short sleep (12) and long sleep (10) interactions underscores the plausible nature of distinct influences of both sleep duration extremes in cardiovascular health. Several of our loci are specific towards a particular population background or sex, emphasizing the importance of addressing heterogeneity entangled in gene-environment interactions, when considering precision medicine design approaches for blood pressure management.
Collapse
Affiliation(s)
- Pavithra Nagarajan
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Thomas W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Clint L Miller
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Aldi T Kraja
- University of Mississippi Medical Center, Jackson, MS, USA
| | - Karen Schwander
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Songmi Lee
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Wenyi Wang
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - John L Morrison
- Division of Biostatistics, Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Ayush Giri
- Division of Quantitative and Clinical Sciences, Department of Obstetrics & Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626), Department of Veterans Affairs, Nashville, TN, USA
| | - Jeffrey R O'Connell
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Lisa de Las Fuentes
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Center for Biostatistics and Data Science, Institute for Informatics, Data Science, and Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Valborg Gudmundsdottir
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, Department of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Sarah E Harris
- Department of Psychology, The University of Edinburgh, Edinburgh, UK
| | - Zhijie Huang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Mart Kals
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Minjung Kho
- Graduate School of Data Science, Seoul National University, Seoul, South Korea
| | - Christophe Lefevre
- Department of Data Sciences, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere, Finland
| | - Massimo Mangino
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
- National Heart & Lung Institute, Cardiovascular Genomics and Precision Medicine, Imperial College London, London, UK
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam UMC/Vrije universiteit, Amsterdam, Netherlands
- GGZ inGeest, Amsterdam, Netherlands
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Varun Rao
- Division of Nephrology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Rainer Rauramaa
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Botong Shen
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Stefan Stadler
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Quan Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jingxian Tang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Sébastien Thériault
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec City, QC, Canada
| | - Adriaan van der Graaf
- Statistical Genetics Group, Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Peter J van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Yujie Wang
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stefan Weiss
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Kenneth E Westerman
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Qian Yang
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Tabara Yasuharu
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Wei Zhao
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Wanying Zhu
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Drew Altschul
- Department of Psychology, The University of Edinburgh, Edinburgh, UK
- School of Psychology, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Md Abu Yusuf Ansari
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - Pramod Anugu
- Jackson Heart Study, University of Mississippi Medical Center, Jackson, MS, USA
| | - Anna D Argoty-Pantoja
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Michael Arzt
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Hugues Aschard
- Department of Computational Biology, F-75015 Paris, France Institut Pasteur, Université Paris Cité, Paris, France
- Department of Epidemiology, Harvard TH School of Public Health, Boston, MA, USA
| | - John R Attia
- School of Medicine and Public Health, College of Health Medicine and Wellbeing, University of Newcastle, New Lambton Heights, NSW, Australia
| | - Lydia Bazzanno
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Max A Breyer
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Brian E Cade
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Hung-Hsin Chen
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Zekai Chen
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Latchezar M Dimitrov
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Anh Do
- Center for Biostatistics and Data Science, Institute for Informatics, Data Science, and Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Jiawen Du
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Charles T Dupont
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Todd L Edwards
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626), Department of Veterans Affairs, Nashville, TN, USA
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Tariq Faquih
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Stephan B Felix
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Cardiology, Pneumology, Infectious Diseases, Intensive Care Medicine, Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Susan P Fisher-Hoch
- School of Public Health, The University of Texas Health Science Center at Houston (UTHealth), Brownsville, TX, USA
| | - James S Floyd
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Mariaelisa Graff
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Charles Gu
- Center for Biostatistics and Data Science, Institute for Informatics, Data Science, and Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Dongfeng Gu
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Kristen G Hairston
- Department of Endocrinology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Anthony J Hanley
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Iris M Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Sami Heikkinen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Heather M Highland
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michelle M Hood
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Mika Kähönen
- Department of Clinical Physiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere, Finland
| | | | - Takahisa Kawaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Setoh Kazuya
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan
| | - Tanika N Kelly
- Division of Nephrology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | | | - Daniel Levy
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Henry J Lin
- The Institute for Translational Genomics and Population Sciences, Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Peter Y Liu
- The Institute for Translational Genomics and Population Sciences, Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Joseph B McCormick
- School of Public Health, The University of Texas Health Science Center at Houston (UTHealth), Brownsville, TX, USA
| | - Hao Mei
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - James B Meigs
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Cristina Menni
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Kisung Nam
- Graduate School of Data Science, Seoul National University, Seoul, South Korea
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Natasha L Pacheco
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Lauren E Petty
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hannah G Polikowsky
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Olli T Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, and Department of Clinical Physiology and Nuclear Medicine, University of Turku, and Turku University Hospital, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Stephen S Rich
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
| | - Renata L Riha
- Department of Sleep Medicine, The University of Edinburgh, Edinburgh, UK
| | - Lorenz Risch
- Faculty of Medical Sciences, Institute for Laboratory Medicine, Private University in the Principality of Liechtenstein, Vaduz, Liechtenstein
- Center of Laboratory Medicine, Institute of Clinical Chemistry, University of Bern and Inselspital, Bern, Switzerland
| | - Martin Risch
- Central Laboratory, Cantonal Hospital Graubünden, Chur, Switzerland
- Medical Laboratory, Dr. Risch Anstalt, Vaduz, Liechtenstein
| | | | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, College of Health Medicine and Wellbeing, University of Newcastle, New Lambton Heights, NSW, Australia
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer A Smith
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- CardioVascular Institute (CVI), Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Maris Teder-Laving
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Peter Vollenweider
- Department of Medicine, Internal Medicine, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Guanchao Wang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
- Department of Internal Medicine, Division of Endocrinology, Leiden, Netherlands
| | - Otis D Wilson
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626), Department of Veterans Affairs, Nashville, TN, USA
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rui Xia
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kristin L Young
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ruiyuan Zhang
- Department of Epidemiology, O'Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jennifer E Below
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Carsten A Böger
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
- Department of Nephrology and Rheumatology, Kliniken Südostbayern, Traunstein, Germany
- KfH Kidney Centre Traunstein, Traunstein, Germany
| | - David Conen
- Population Health Research Institute, Medicine, McMaster University, Hamilton, ON, Canada
| | - Simon R Cox
- Department of Psychology, The University of Edinburgh, Edinburgh, UK
| | - Marcus Dörr
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Cardiology, Pneumology, Infectious Diseases, Intensive Care Medicine, Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Ervin R Fox
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Nora Franceschini
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sina A Gharib
- Pulmonary, Critical Care and Sleep Medicine, Medicine, University of Washington, Seattle, WA, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, Department of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Sioban D Harlow
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Jiang He
- Department of Epidemiology, O'Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elizabeth G Holliday
- School of Medicine and Public Health, College of Health Medicine and Wellbeing, University of Newcastle, New Lambton Heights, NSW, Australia
| | - Zoltan Kutalik
- Statistical Genetics Group, Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Center for Primary Care and Public Health, University of Lausanne, Lausanne, Switzerland
| | - Timo A Lakka
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Seunggeun Lee
- Graduate School of Data Science, Seoul National University, Seoul, South Korea
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere, Finland
| | - Changwei Li
- Department of Epidemiology, O'Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Brenda Wjh Penninx
- Department of Psychiatry, Amsterdam UMC/Vrije universiteit, Amsterdam, Netherlands
- GGZ inGeest, Amsterdam, Netherlands
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Tim D Spector
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Lynne E Wagenknecht
- Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Kari E North
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Adriana M Hung
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626), Department of Veterans Affairs, Nashville, TN, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Alisa K Manning
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Metabolism Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - James Gauderman
- Division of Biostatistics, Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Patricia B Munroe
- Clinical Pharmacology and Precision Medicine, Queen Mary University of London, London, UK
| | - Dabeeru C Rao
- Center for Biostatistics and Data Science, Institute for Informatics, Data Science, and Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Heming Wang
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
56
|
Ju F, Wang J, Xu K, Xu Q, Liu X, Tian T, Du Z, Wang J, Liao Z, Wang B, Zhang H. Genome-wide insights into the nomenclature, evolution and expression of tobacco TIFY/JAZ genes. PLANTA 2025; 261:103. [PMID: 40183817 DOI: 10.1007/s00425-025-04676-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/20/2025] [Indexed: 04/05/2025]
Abstract
MAIN CONCLUSION A systematic nomenclature for tobacco TIFY/JAZ proteins was established via genome-wide analysis, and the gene transcription patterns and potential functions of these proteins were analyzed as well. Intensive studies focused on the plant-specific JAZ regulators of jasmonate (JA) signaling in tobacco due to their critical roles in regulating JA-mediated development, secondary metabolism, and stress responses. JAZs comprise a subfamily of the TIFY proteins, yet the reported TIFY/JAZ regulators of tobacco spp. are tangled in naming confusion, which resulted in nomenclature chaos. Here, we identified 32 TIFY/JAZ proteins via genome-wide analysis of tobacco cultivar TN90 and obtained their homologues in Nicotiana sylvestris and Nicotiana tomentosiformis. By bioinformatic analysis, these TIFY/JAZ regulators were classified into 4 subfamilies (i.e., 21 JAZs, 5 ZIM & ZMLs, 2 TIFY8s, and 4 PPDs) based on their phylogenetic relationship to establish a systematic nomenclature, which indicated gene loss or genomic rearrangement during the formation of common tobacco. Analysis of JA-induced expression revealed that these TIFY/JAZ genes displayed distinct expression patterns in the leaves and roots upon JA treatment. Further microarray and metabolomics assays observed that 5 TIFY/JAZ genes were differentially expressed in the plants with dysfunction of COI1, the receptor protein of JA hormone and that the abundance of a series of primary and secondary metabolites was altered as well. A predicted protein interaction network of tobacco TIFY/JAZ proteins was also constructed, and it indicated that 120 proteins may interact with these regulators. Findings of this work provide valuable information about TIFY/JAZ proteins in regulating JA responses and metabolic processes in tobacco and may contribute greatly to future studies on tobacco TIFY/JAZ proteins.
Collapse
Affiliation(s)
- Fuzhu Ju
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
| | - Jiahao Wang
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
| | - Ke Xu
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
| | - Qing Xu
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
| | - Xiaofeng Liu
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
| | - Tian Tian
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
| | - Zaifeng Du
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
| | - Jialin Wang
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
| | - Zhihua Liao
- SWU-TAAHC Medicinal Plant Joint R&D Centre, School of Life Sciences, Southwest University, Chongqing, 400716, China
| | - Bingwu Wang
- Tobacco Breeding and Biotechnology Research Center, Yunnan Academy of Tobacco Agricultural Sciences, Kunming, 650021, China.
| | - Hongbo Zhang
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China.
| |
Collapse
|
57
|
Gomes FP, Durbin KR, Schauer K, Nwachukwu JC, R Kobylski R, Njeri JW, Seath CP, Saviola AJ, McClatchy DB, Diedrich JK, Garrett PT, Papa AB, Ciolacu I, Kelleher NL, Nettles KW, Yates JR. Native top-down proteomics enables discovery in endocrine-resistant breast cancer. Nat Chem Biol 2025:10.1038/s41589-025-01866-8. [PMID: 40186031 DOI: 10.1038/s41589-025-01866-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 02/24/2025] [Indexed: 04/07/2025]
Abstract
Oligomerization of proteoforms produces functional protein complexes. Characterization of these assemblies within cells is critical to understanding the molecular mechanisms involved in disease and to designing effective drugs. Here we present a native top-down proteomics (nTDP) strategy to identify protein assemblies (≤70 kDa) in breast cancer cells and in cells that overexpress epidermal growth factor receptor (EGFR), which serves as a resistance model of estrogen receptor-alpha (ER)-targeted therapies. This nTDP approach identified ~104 complexoforms from 17 protein complexes, which revealed several molecular features of the breast cancer proteome, including EGFR-induced dissociation of nuclear transport factor 2 (NUTF2) assemblies that modulate ER activity. We found that the K4 and K55 post-translational modification sites discovered with nTDP differentially impact the effects of NUTF2 on the inhibition of the ER signaling pathway. The characterization of endogenous proteoform-proteoform/ligand interactions revealed the molecular diversity of complexoforms and their role in breast cancer growth.
Collapse
Affiliation(s)
| | | | | | - Jerome C Nwachukwu
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
| | - Robin R Kobylski
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, USA
| | - Jacqline W Njeri
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, USA
| | - Ciaran P Seath
- Department of Chemistry, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
| | | | | | | | | | - Alexandra B Papa
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
- Florida Atlantic University, Jupiter, FL, USA
| | - Ianis Ciolacu
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
- Florida Atlantic University, Jupiter, FL, USA
| | - Neil L Kelleher
- Proteinaceous, Evanston, IL, USA
- Northwestern University, National Resource for Translational and Developmental Proteomics, Evanston, IL, USA
| | - Kendall W Nettles
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA.
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, USA.
| | | |
Collapse
|
58
|
Loomis S, Silva DG, Savopoulos R, Cilia J, Li J, Davis MD, Virley D, Foley A, Loro E, McCreary AC. Behavioral and transcriptomic effects of a novel cannabinoid on a rat valproic acid model of autism. Neuropharmacology 2025; 273:110450. [PMID: 40187640 DOI: 10.1016/j.neuropharm.2025.110450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition characterized by impaired social communication, restricted interests, repetitive behavior and irritability. Exposure to valproic acid (VPA) during pregnancy has been shown to increase the risk of autism in children and has led to the development of the in-utero VPA rat model that elicits neurodevelopmental autistic-like features. Offspring exhibit behavioral and neurobiological alterations modelling ASD symptoms. We performed a behavioral and molecular assessment in a rat in-utero VPA model treated with a novel botanical cannabinoid, JZP541. Male offspring from dams treated with VPA were tested acutely and sub-chronically with JZP541 (10, 30, or 100 mg/kg, intraperitoneally). A behavioral testing battery was performed, and brain frontal cortex and hippocampus used for RNA sequencing. In utero exposure to VPA resulted in progeny showing behavioral phenotypes characteristic of ASD. JZP541 attenuated these deficits in social, stereotypic, hyperactivity and irritability behavior in a dose-dependent fashion. VPA exposure was associated with a substantial transcriptional dysregulation impacting multiple key biological processes in a tissue-dependent manner. The expression profiles were integrated with publicly available datasets of autism-associated genes to support the validity of the model used and to focus on the effects of treatment on known autism-relevant transcriptional targets. This approach indicated a strong and dose-dependent reduction of the autism-associated gene expression signature in brain samples from animals dosed with JZP541. Our findings demonstrate JZP541 was able to ameliorate ASD associated behavioral deficits, and this was supported by improvements in putative transcriptional biomarkers of ASD.
Collapse
Affiliation(s)
- Sally Loomis
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK.
| | - Diogo G Silva
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK
| | | | - Jackie Cilia
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK
| | - Jennifer Li
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK
| | - Mat D Davis
- Jazz Pharmaceuticals Inc., Palo Alto, CA, USA
| | - David Virley
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK
| | | | - Emanuele Loro
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK
| | | |
Collapse
|
59
|
Kianpoor S, Ehsani A, Torshizi RV, Masoudi AA, Bakhtiarizadeh MR. Unlocking the genetic code: a comprehensive Genome-Wide association study and gene set enrichment analysis of cell-mediated immunity in chickens. BMC Genomics 2025; 26:337. [PMID: 40181279 PMCID: PMC11970016 DOI: 10.1186/s12864-025-11538-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/27/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND The poultry immune system is essential for protecting against infectious diseases and maintaining health and productivity. Cell-mediated immune responses (CMIs) protect organisms against intracellular pathogens. This study aimed to enrich the findings of genome-wide association studies (GWAS) by including several systematic gene set enrichment analyses (GSEA) related to cell-mediated immune responses in chickens. METHODS To investigate the function of the cellular immune system, phenotypic data were collected based on the differences in skin thickness before and after impregnation with dinitrochlorobenzene (DNCB) solution. Additionally, 312 hybrid birds of the F2 generation of Arian broiler chickens and Urmia native chickens were genotyped using the Illumina 60k SNP BeadChip. A general linear model (GLM) with an FDR < 5% was used for the association analysis. Functional enrichment analysis of the identified candidate genes was performed using the Enrichr database. A protein‒protein interaction (PPI) network was constructed using the STRING database. In addition, colocalization analysis was applied to identify QTLs related to the immune system. RESULTS GWAS revealed 147 SNPs associated with the CMI trait, which were related to 1363 genes. These genes were significantly enriched in eight KEGG pathways, 22 Reactome pathways, and 18 biological processes. PPI network analysis led to the identification of 26 hub genes. The three hub genes PSMA3, PSMC2 and PSMB4 were enriched in almost all pathways related to cellular immunity, including the proteasome, interleukin-1 signaling, and programmed cell death pathways, which makes them important candidates involved in CMI. In addition, the MAP3K8, NLRC5, UBB, CASP6, DAPK2, TNFRSF6B, TNFSF15, and PIK3CD genes were identified as key genes in several functional pathways. A total of 10 SNPs were found in interferon-gamma QTLs, and two SNPs were found in the cell-mediated immune response QTL region, leading to the identification of 12 cellular immune response-related genes that were reported as important candidates in previous studies. CONCLUSION The post-GWAS analysis in this study led to the identification of key genes that regulate the biological processes of cellular immunity in chickens. Therefore, selecting birds that excel in expressing such genes can improve immunity in chickens.
Collapse
Affiliation(s)
- Somayeh Kianpoor
- Department of Animal Science, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Alireza Ehsani
- Department of Animal Science, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran.
| | - Rasoul Vaez Torshizi
- Department of Animal Science, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Ali Akbar Masoudi
- Department of Animal Science, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | | |
Collapse
|
60
|
Kitani A, Matsui Y. Integrative network analysis reveals novel moderators of Aβ-Tau interaction in Alzheimer's disease. Alzheimers Res Ther 2025; 17:70. [PMID: 40176187 PMCID: PMC11967117 DOI: 10.1186/s13195-025-01705-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 02/25/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Although interactions between amyloid-beta and tau proteins have been implicated in Alzheimer's disease (AD), the precise mechanisms by which these interactions contribute to disease progression are not yet fully understood. Moreover, despite the growing application of deep learning in various biomedical fields, its application in integrating networks to analyze disease mechanisms in AD research remains limited. In this study, we employed BIONIC, a deep learning-based network integration method, to integrate proteomics and protein-protein interaction data, with an aim to uncover factors that moderate the effects of the Aβ-tau interaction on mild cognitive impairment (MCI) and early-stage AD. METHODS Proteomic data from the ROSMAP cohort were integrated with protein-protein interaction (PPI) data using a Deep Learning-based model. Linear regression analysis was applied to histopathological and gene expression data, and mutual information was used to detect moderating factors. Statistical significance was determined using the Benjamini-Hochberg correction (p < 0.05). RESULTS Our results suggested that astrocytes and GPNMB + microglia moderate the Aβ-tau interaction. Based on linear regression with histopathological and gene expression data, GFAP and IBA1 levels and GPNMB gene expression positively contributed to the interaction of tau with Aβ in non-dementia cases, replicating the results of the network analysis. CONCLUSIONS These findings suggest that GPNMB + microglia moderate the Aβ-tau interaction in early AD and therefore are a novel therapeutic target. To facilitate further research, we have made the integrated network available as a visualization tool for the scientific community (URL: https://igcore.cloud/GerOmics/AlzPPMap ).
Collapse
Affiliation(s)
- Akihiro Kitani
- Department of Integrated Health Science, Biomedical and Health Informatics Unit, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yusuke Matsui
- Department of Integrated Health Science, Biomedical and Health Informatics Unit, Nagoya University Graduate School of Medicine, Nagoya, Japan.
- Institute for Glyco-Core Research (Igcore), Nagoya University, Nagoya, Aichi, 461-8673, Japan.
| |
Collapse
|
61
|
Yin J, Hu Y, Yan K, Xu Y, Li D. Uncovering the bioactive constituents and their mechanisms of the Forsythiae Fructus against hyperpigmentation using a combined strategy integrating cell-specific extraction, plasma pharmaceutical chemistry and network pharmacology. J Pharm Biomed Anal 2025; 262:116865. [PMID: 40194472 DOI: 10.1016/j.jpba.2025.116865] [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: 01/24/2025] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025]
Abstract
Skin hyperpigmentation is a type of difficult-to-treat disease that frequently results from the improper metabolism of facial pigment. The herb Forsythiae Fructus (FF) possesses whitening and freckle-removal properties. Its probable active components and anti-hyperpigmentation mechanism, however, are still unknown. In the current investigation, the active components were initially identified and screened by UHPLC-Q-Orbitrap HRMS/MS employing B16 cell-specific extraction and plasma pharmaceutical chemistry, respectively. Subsequently, the component-target-disease network and protein-protein interaction (PPI) network of FF were built by using a network pharmacology approach. The probable targets and pathways were found using gene ontology (GO) and KEGG enrichment analysis. The essential elements and core genes causing illnesses were identified through molecular docking. Lastly, based on network analysis, cell experiments were carried out to further explore the efficacy of the main active ingredients in the treatment of abnormal melanosis. As a result, 37 ingredients were identified in FF extract, 22 compounds in the decoction had a specific affinity with B16 mouse melanoma cells, and a total of 10 prototype compounds and 11 metabolites were detected in rat plasma. Through in vitro and in vivo screening methods, 16 potential active ingredients were obtained, and 229 biological targets and 1515 disease-related targets were predicted by network pharmacology. In addition, in vitro cell experiments revealed that kaempferol, luteolin, and wogonin all exhibited inhibition of melanin production and tyrosinase activity. The proposed combination method of rapid screening of active ingredients in vivo and integrated network pharmacology in vitro could explore the therapeutic mechanism of FF against hyper-pigmentation, and provide a theoretical evidence for the development and utilization of FF.
Collapse
Affiliation(s)
- Jintuo Yin
- Department of Pharmacy, the Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Yalin Hu
- Department of Pharmacy, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Kai Yan
- Hebei Institute for Drug and Medical Device Control, Shijiazhuang 050299, China
| | - Yanmei Xu
- Hebei Institute for Drug and Medical Device Control, Shijiazhuang 050299, China
| | - Deqiang Li
- Department of Pharmacy, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China.
| |
Collapse
|
62
|
Chen J, Wen F, Zhou J, Tan M. Evaluating the Mechanism Underlying Multi-Compound Synergy of Banxia Decoction in the Treatment of Hashimoto's Thyroiditis Based on Network Pharmacology and Molecular Docking. Int J Gen Med 2025; 18:1887-1902. [PMID: 40196382 PMCID: PMC11972970 DOI: 10.2147/ijgm.s502321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/09/2025] [Indexed: 04/09/2025] Open
Abstract
Objective We aimed to utilize network pharmacological analysis and molecular docking to elucidate the potential mechanisms of Banxia Decoction (BD) action in the treatment of Hashimoto's thyroiditis (HT). Materials and Methods Active compounds and HT-related targets were predicted using databases and the intersection of the results was taken. STRING and DAVID 6.8 tools were used to obtain the protein-protein interaction (PPI) network and perform GO and KEGG evaluations, respectively. Discovery Studio 2017 R2 was utilized to perform molecular docking and RT-qPCR was conducted to confirm hub gene expressions in clinical samples. Results A total of 136 active compounds in BD were screened, and 74 potential targets related to HT were identified in BD. Further, 17 key targets in the PPI network were identified and HIF1A, EP300, PRKCA, and TERT were included for subnet analysis. Next, a network of "Chinese medicine-active compound-potential target-signal pathway" was obtained and the HIF-1 signaling pathway was identified as the key pathway. Finally, 8 active compounds and their stable binding to target proteins were confirmed by molecular docking; MAPK3, SRC, TERT, and HIF1A were upregulated in HT relative to the goiter samples. Conclusion The integration of network pharmacology and molecular docking provides a systematic framework for exploring the multi-component and multi-target characteristics of BD in HT, underscores the therapeutic potential of BD in HT by targeting genes and pathways involved in immune regulation and oxidative stress. These findings not only enhance our understanding of BD's pharmacological mechanisms but also lay the groundwork for the development of novel therapeutic strategies for HT.
Collapse
Affiliation(s)
- Jian Chen
- Department of Gastroenterology Medical Center and Thyroid Gastrointestinal Hernia Surgery, Digestive Disease Medical Center, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, 412000, People’s Republic of China
| | - Fang Wen
- Department of Intensive Care Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, 412000, People’s Republic of China
| | - Juan Zhou
- Department of Respiratory and Critical Care Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine Central South University, Zhuzhou, Hunan, 412000, People’s Republic of China
| | - Miduo Tan
- Department of Breast Surgery, Digestive Disease Medical Center, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, 412000, People’s Republic of China
| |
Collapse
|
63
|
Dong K, Ye Z, Hu F, Shan C, Wen D, Cao J. An evolutionary dynamics analysis of the plant DEK gene family reveals the role of BnaA02g08940D in drought tolerance. Int J Biol Macromol 2025; 298:140053. [PMID: 39828179 DOI: 10.1016/j.ijbiomac.2025.140053] [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: 10/13/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
DEK is a chromatin protein that interacts with DNA to influence chromatin formation, thereby affecting plant growth, development, and stress response. This study investigates the molecular evolution of the DEK family in plants, with a particular focus on the Brassica species. A total of 127 DEK genes were identified in 34 plants and classified into seven groups based on the phylogenetic analysis. The distribution of motifs and gene structure is similar within each group, indicating a high degree of conservation. The results of the collinearity analysis indicated that the DEK protein has undergone a certain degree of evolutionary conservation. The expansion of the DEK family is primarily attributable to whole-genome duplication (WGD) or segmental duplication events. The DEK protein has undergone purification during its evolutionary history, and several positively selected sites have been identified. Moreover, the examination of cis-acting elements and expression patterns revealed that the BnDEKs play a significant role in plant growth and stress response. The protein-protein interaction network identified several noteworthy proteins that interact with DEK. These analyses enhance our comprehension of the DEK gene family and establish the foundation for additional validation of its function. Further research demonstrated that the overexpression of one DEK family member, BnaA02g08940D, enhanced the transgenic Arabidopsis tolerance to drought and osmosis. This indicates that the DEK family may respond when plants are subjected to drought stress, thereby strengthening the plant's resilience.
Collapse
Affiliation(s)
- Kui Dong
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Ziyi Ye
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Fei Hu
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Chaofan Shan
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Dongyu Wen
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Jun Cao
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, Jiangsu, China.
| |
Collapse
|
64
|
Govta N, Govta L, Sela H, Peleg G, Distelfeld A, Fahima T, Beckles DM, Krugman T. Plasticity of Root System Architecture and Whole Transcriptome Responses Underlying Nitrogen Deficiency Tolerance Conferred by a Wild Emmer Wheat QTL. PLANT, CELL & ENVIRONMENT 2025; 48:2835-2855. [PMID: 39887777 PMCID: PMC11893928 DOI: 10.1111/pce.15416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/16/2025] [Accepted: 01/19/2025] [Indexed: 02/01/2025]
Abstract
Our aim was to elucidate mechanisms underlying nitrogen (N)-deficiency tolerance in bread wheat (cultivar Ruta), conferred by a wild emmer wheat QTL (WEW; IL99). We hypothesised that the tolerance in IL99 is driven by enhanced N-uptake through modification of root system architecture (RSA) underscored by transcriptome modifications. Severe N-deficiency (0.1 N for 26 days) triggered significantly higher plasticity in IL99 compared to Ruta by modifying 16 RSA traits; nine of which were IL99-specific. The change in root growth in IL99 was collectively characterised by a transition in root orientation from shallow to steep, increased root number and length, and denser networks, enabling nutrient acquisition from a larger volume and deeper soil layers. Gene ontology and KEGG-enrichment analyses highlighted IL99-specific pathways and candidate genes elevated under N-deficiency. This included Jasmonic acid metabolism, a key hormone mediating RSA plasticity (AOS1, TIFY, MTB2, MYC2), and lignification-mediated root strengthening (CYP73A, 4CL). 'N-metabolism' was identified as a main shared pathway to IL99 and Ruta, with enhanced nitrate uptake predominant in IL99 (NRT2.4), while remobilisation was the main strategy in Ruta (NRT2.3). These findings provide novel insights into wheat plasticity response underlying tolerance to N-deficiency and demonstrate the potential of WEW for improving N-uptake under suboptimal conditions.
Collapse
Affiliation(s)
- Nikolai Govta
- Department of Evolutionary and Environmental Biology, and Institute of EvolutionUniversity of HaifaHaifaIsrael
| | - Liubov Govta
- Department of Evolutionary and Environmental Biology, and Institute of EvolutionUniversity of HaifaHaifaIsrael
| | - Hanan Sela
- Department of Evolutionary and Environmental Biology, and Institute of EvolutionUniversity of HaifaHaifaIsrael
| | | | - Assaf Distelfeld
- Department of Evolutionary and Environmental Biology, and Institute of EvolutionUniversity of HaifaHaifaIsrael
| | - Tzion Fahima
- Department of Evolutionary and Environmental Biology, and Institute of EvolutionUniversity of HaifaHaifaIsrael
| | - Diane M. Beckles
- Department of Plant SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | - Tamar Krugman
- Department of Evolutionary and Environmental Biology, and Institute of EvolutionUniversity of HaifaHaifaIsrael
| |
Collapse
|
65
|
Wang X, Liu Y, Zhang S, Zhang J, Lin X, Liang Y, Zong M, Hanley KL, Lee J, Karin M, Feng GS. Genomic and transcriptomic analyses of chemical hepatocarcinogenesis aggravated by oncoprotein loss. Hepatology 2025; 81:1181-1196. [PMID: 39397357 DOI: 10.1097/hep.0000000000001037] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 07/10/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND AND AIMS The chemical carcinogen diethylnitrosamine (DEN) is often used to induce HCC in mice. Curiously, several labs have reported that the removal of oncoproteins from hepatocytes exacerbated DEN-induced HCC, with mechanisms unknown. This study aimed at deciphering molecular mechanisms underlying the tumor suppressive effect of oncoproteins. APPROACH AND RESULTS We generated mutant mouse lines with hepatocyte-specific deletions of Met , Ptpn11 / Shp2 , Ikkβ , or Ctnnb1/β-catenin and assessed DEN-induced tumorigenesis in the wild-type and mutant mice. To systematically examine genetic and molecular signaling alterations, we performed whole exome and RNA-sequencing on liver samples collected at the pre-cancer and established cancer stages. Although the mutational profiles of DEN-induced tumors were barely different in wild-type and mutant mice, oncoprotein ablation increased DEN-induced mutational burdens, especially in Shp2-deficient tumors. RNA-sequencing revealed multiple changes in signaling pathways, in particular, upregulated epithelial-mesenchymal transition, cell migration, and tumor metastasis, as well as downregulated small molecule metabolism that was affected by oncoprotein ablation. We identified key molecules and pathways that are associated with hepatic innate immunity and implicated in liver tumorigenesis. In addition, we unveiled markedly changed expression of a few miRNAs in the human HCC database. CONCLUSIONS The aggravation of DEN-induced HCC progression seen on oncoprotein ablation could be caused by common and distinct genomic and signaling alterations. This study reveals a new level of complexity in hepatocarcinogenesis and elucidates molecular mechanisms underlying tumor evolution and recurrence.
Collapse
Affiliation(s)
- Xinyi Wang
- Department of Pathology, School of Medicine, La Jolla, California, USA
- Department of Molecular Biology, School of Biological Sciences, La Jolla, California, USA
- Moores Cancer Center, University of California at San Diego, La Jolla, California, USA
| | - Yingluo Liu
- Department of Pathology, School of Medicine, La Jolla, California, USA
- Department of Molecular Biology, School of Biological Sciences, La Jolla, California, USA
- Moores Cancer Center, University of California at San Diego, La Jolla, California, USA
| | - Shuo Zhang
- Department of Pathology, School of Medicine, La Jolla, California, USA
- Department of Molecular Biology, School of Biological Sciences, La Jolla, California, USA
- Moores Cancer Center, University of California at San Diego, La Jolla, California, USA
| | - Jiemeng Zhang
- Department of Pathology, School of Medicine, La Jolla, California, USA
- Department of Molecular Biology, School of Biological Sciences, La Jolla, California, USA
- Moores Cancer Center, University of California at San Diego, La Jolla, California, USA
| | - Xiaoxue Lin
- Department of Pathology, School of Medicine, La Jolla, California, USA
- Department of Molecular Biology, School of Biological Sciences, La Jolla, California, USA
- Moores Cancer Center, University of California at San Diego, La Jolla, California, USA
| | - Yan Liang
- Department of Pathology, School of Medicine, La Jolla, California, USA
- Department of Molecular Biology, School of Biological Sciences, La Jolla, California, USA
- Moores Cancer Center, University of California at San Diego, La Jolla, California, USA
| | - Min Zong
- Department of Pathology, School of Medicine, La Jolla, California, USA
- Department of Molecular Biology, School of Biological Sciences, La Jolla, California, USA
- Moores Cancer Center, University of California at San Diego, La Jolla, California, USA
| | - Kaisa L Hanley
- Department of Pathology, School of Medicine, La Jolla, California, USA
- Department of Molecular Biology, School of Biological Sciences, La Jolla, California, USA
- Moores Cancer Center, University of California at San Diego, La Jolla, California, USA
| | - Jin Lee
- Department of Pathology, School of Medicine, La Jolla, California, USA
- Department of Molecular Biology, School of Biological Sciences, La Jolla, California, USA
- Moores Cancer Center, University of California at San Diego, La Jolla, California, USA
| | - Michael Karin
- Departments of Pharmacology and Pathology, University of California at San Diego, La Jolla, California, USA
| | - Gen-Sheng Feng
- Department of Pathology, School of Medicine, La Jolla, California, USA
- Department of Molecular Biology, School of Biological Sciences, La Jolla, California, USA
- Moores Cancer Center, University of California at San Diego, La Jolla, California, USA
| |
Collapse
|
66
|
Ban Z, Lin Y, Yan Y, Dawson KA. Unraveling biomolecular and community grammars of RNA granules via machine learning. PNAS NEXUS 2025; 4:pgaf093. [PMID: 40160533 PMCID: PMC11952899 DOI: 10.1093/pnasnexus/pgaf093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/06/2025] [Indexed: 04/02/2025]
Abstract
Membraneless RNA granules are essential for posttranscriptional gene regulation, influencing cellular functions and contributing to neurodegenerative diseases. However, a comprehensive understanding of their compositions and organization has been challenging due to their complex nature. In this study, we develop robust machine learning models to reliably identify RNA granule proteomes within the human proteome, capturing central RNA granule characteristics despite the heterogeneity across diverse in vitro conditions. Furthermore, we uncover protein-protein interaction (PPI) community grammars within the RNA granule proteome, highlighting PPIs as key stabilizers of RNA granule structure and function. Dense PPI clusters serve as stable "cores," forming key functional subunits across heterogeneous RNA granules. We introduce a state-of-the-art framework for understanding RNA granule biology and underscore the critical role of PPIs in maintaining RNA granule integrity.
Collapse
Affiliation(s)
- Zhan Ban
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Dublin 4, D04 N2E5, Ireland
- School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, D04 N2E5, Ireland
| | - Yuchen Lin
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Dublin 4, D04 N2E5, Ireland
- School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, D04 N2E5, Ireland
| | - Yan Yan
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Dublin 4, D04 N2E5, Ireland
- School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, D04 N2E5, Ireland
| | - Kenneth A Dawson
- Centre for BioNano Interactions, School of Chemistry, University College Dublin, Dublin 4, D04 N2E5, Ireland
- School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, D04 N2E5, Ireland
| |
Collapse
|
67
|
Asada N, Ginsberg P, Paust HJ, Song N, Riedel JH, Turner JE, Peters A, Kaffke A, Engesser J, Wang H, Zhao Y, Khatri R, Gild P, Dahlem R, Diercks BP, Das S, Ignatova Z, Huber TB, Prinz I, Gagliani N, Mittrücker HW, Krebs CF, Panzer U. The integrated stress response pathway controls cytokine production in tissue-resident memory CD4 + T cells. Nat Immunol 2025; 26:557-566. [PMID: 40050432 PMCID: PMC11957990 DOI: 10.1038/s41590-025-02105-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 02/04/2025] [Indexed: 03/12/2025]
Abstract
Tissue-resident memory T (TRM) cells are a specialized T cell population that reside in tissues and provide a rapid protective response upon activation. Here, we showed that human and mouse CD4+ TRM cells existed in a poised state and stored messenger RNAs encoding proinflammatory cytokines without protein production. At steady state, cytokine mRNA translation in TRM cells was suppressed by the integrated stress response (ISR) pathway. Upon activation, the central ISR regulator, eIF2α, was dephosphorylated and stored cytokine mRNA was translated for immediate cytokine production. Genetic or pharmacological activation of the ISR-eIF2α pathway reduced cytokine production and ameliorated autoimmune kidney disease in mice. Consistent with these results, the ISR pathway in CD4+ TRM cells was downregulated in patients with immune-mediated diseases of the kidney and the intestine compared to healthy controls. Our results indicated that stored cytokine mRNA and translational regulation in CD4+ TRM cells facilitate rapid cytokine production during local immune response.
Collapse
Affiliation(s)
- Nariaki Asada
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pauline Ginsberg
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Joachim Paust
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ning Song
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan-Hendrik Riedel
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan-Eric Turner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anett Peters
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Kaffke
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonas Engesser
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Huiying Wang
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yu Zhao
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, Center for Biomedical AI, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Robin Khatri
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, Center for Biomedical AI, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Philipp Gild
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roland Dahlem
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn-Philipp Diercks
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarada Das
- Institute of Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany
| | - Zoya Ignatova
- Institute of Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Immo Prinz
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Systems Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Gagliani
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Willi Mittrücker
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute for Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian F Krebs
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulf Panzer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
68
|
Dharshan SS, Ramamurthy K, Kaliraj S, Manikandan K, Chitra V, Rajagopal R, Alfarhan A, Namasivayam SKR, Kathiravan MK, Arockiaraj J. Combined effects of vitamin D3 and dioxopiperidinamide derivative on lipid homeostasis, inflammatory pathways, and redox imbalance in non-alcoholic fatty liver disease in vivo zebrafish model. Biotechnol Appl Biochem 2025; 72:320-339. [PMID: 39252166 DOI: 10.1002/bab.2666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/24/2024] [Indexed: 09/11/2024]
Abstract
Liver damage and metabolic dysfunctions, the defining features of non-alcoholic fatty liver disease (NAFLD), are marked by inflammation, oxidative stress, and excessive hepatic fat accumulation. The current therapeutic approaches for NAFLD are limited, necessitating exploring novel treatment strategies. Dioxopiperidinamide derivatives, particularly DOPA-33, have shown effective anti-inflammatory and antioxidant properties, potentially offering therapeutic benefits against NAFLD. This study investigated the combined potential of vitamin D3 (Vit D3) and DOPA-33 in treating NAFLD. The network pharmacology analysis identified key NAFLD targets modulated by Vit D3 and DOPA-33, emphasizing their potential mechanisms of action. In NAFLD-induced zebrafish models, Vit D3 and DOPA-33 significantly reduced hepatic lipid accumulation, oxidative stress, and apoptosis, demonstrating superior efficacy over individual treatments. The treatment also lowered reactive oxygen species (ROS) levels, decreased liver damage, and enhanced antioxidant defense mechanisms. Moreover, behavioral analyses showed improved locomotion and reduced weight gain in treated zebrafish. Biochemical analyses revealed lower triglycerides (TG) and glucose levels with improved oxidative markers. Furthermore, histological analyses indicated reduced hepatic steatosis and inflammation, with decreased expression of lipogenesis-related genes and inflammatory mediators. Finally, high-performance liquid chromatography (HPLC) confirmed a significant reduction in hepatic cholesterol levels, indicating the effectiveness of the combination therapy in addressing key NAFLD-related dyslipidemias. These findings suggest that Vit D3 + DOPA-33 targets pathways involved in lipid metabolism, inflammation, and oxidative stress by offering a promising therapeutic approach for NAFLD.
Collapse
Affiliation(s)
- Santhanam Sanjai Dharshan
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, Tamil Nadu, India
| | - Karthikeyan Ramamurthy
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, Tamil Nadu, India
| | - Salamuthu Kaliraj
- Department of Chemistry, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Krishnan Manikandan
- Department of Pharmaceutical Analysis, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Vellapandian Chitra
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Rajakrishnan Rajagopal
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Alfarhan
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - S Karthick Raja Namasivayam
- Centre for Applied Research, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, India
| | - Muthu Kumaradoss Kathiravan
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, Tamil Nadu, India
| |
Collapse
|
69
|
Ryu T, Adler BL, Jeong SJ, Lee DC, Hoke A, Na CH, Chung T. Quantitative serum proteomic analysis for biomarker discovery in post-COVID-19 postural orthostatic tachycardia syndrome (PC-POTS) patients. Auton Neurosci 2025; 258:103247. [PMID: 40022872 DOI: 10.1016/j.autneu.2025.103247] [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: 10/07/2024] [Revised: 12/18/2024] [Accepted: 01/26/2025] [Indexed: 03/04/2025]
Abstract
Postural orthostatic tachycardia syndrome (POTS) is a chronic, debilitating condition that is characterized by an excessive increase in heart rate upon orthostatic challenge. Before the COVID-19 pandemic, POTS affected 0.5 % to 1 % of the U.S. population. Since the pandemic, the incidence has risen sharply, adding an estimated 6-7 million new cases in the U.S. Despite its importance, there is currently no reliable biomarker for POTS, leading to significant diagnostic delays. A major hurdle in identifying biomarkers is the heterogeneous nature of the syndrome. To address this, we focused on a homogeneous subgroup of post-COVID-19 POTS (PC-POTS) patients. We conducted quantitative proteomics on sera from 9 PC-POTS patients and 9 healthy controls, identifying 31 proteins with significantly different abundances in PC-POTS patients. Most elevated proteins were linked to actin filaments or immune functions/inflammation. Weighted Gene Co-Expression Network Analysis revealed module 7 (M7) correlated strongly with PC-POTS diagnosis and related traits. The key proteins in M7 included MTPN, TAGLN2, ADP-ribosylation factor 1, PDLIM1, PPIA, CNN2, LGALSL, TXN, TLN1, TUBA4A, IL4, TREML1, GP1BA, and, all highly correlated with these traits. Cell-type enrichment analysis revealed that M7 was highly associated with immune and neuronal cells. The main pathways identified in M7 included the integrin signaling pathway, blood coagulation, and glycolysis. These findings suggest that the key proteins in M7 could serve as biomarkers for PC-POTS. This study uses quantitative proteomics to identify potential biomarkers that differentiate PC-POTS patients from healthy controls, establishing a foundation for further research and validation.
Collapse
Affiliation(s)
- Taekyung Ryu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brittany L Adler
- Department of Medicine, Rheumatology Division, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seeun Judy Jeong
- Department of Physical Medicine and Rehabilitation, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David C Lee
- Department of Emergency Medicine and Department of Population Health, NYU Grossman School of Medicine, New York, USA
| | - Ahmet Hoke
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chan Hyun Na
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Tae Chung
- Department of Physical Medicine and Rehabilitation, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
70
|
Shanthikumar S, Gubbels L, Davies K, Walker H, Wong ATC, Levi E, Saffery R, Ranganathan S, Neeland MR. Highly multiplexed cytokine analysis of bronchoalveolar lavage and plasma reveals age-related dynamics and correlates of inflammation in children. Mucosal Immunol 2025; 18:380-389. [PMID: 39675726 DOI: 10.1016/j.mucimm.2024.12.008] [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: 07/24/2024] [Revised: 10/08/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
Despite the central role of cytokines in mediating inflammation that underlies a range of childhood diseases, cytokine testing remains primarily limited to research settings and surrogate markers of inflammation are often used to inform clinical diagnostic and treatment decisions. There are currently no reference ranges available for cytokines in healthy children, either systemically (in blood) or at sites of disease (such as the lung). In our study, we aimed to develop an openly accessible dataset of cytokines in the airways and blood of healthy children spanning 1 to 16 years of age. We examined how cytokine concentration changes during childhood and assessed whether a core set of cytokine markers could be used to indirectly evaluate the response of a broad spectrum of inflammatory analytes. To develop our dataset, a total of 65 unique analytes were quantified in cell-free bronchoalveolar lavage (BAL) and plasma from 78 children. We showed that age profoundly impacts soluble immune analyte concentration in both sample types and identified a highly correlative core set of 10 analytes in BAL and 11 analytes in plasma capable of indirectly evaluating the response of up to 44 inflammatory mediators. This study addresses an urgent need to develop reference ranges for cytokines in healthy children to aid in diagnosis of disease, to determine eligibility for, and to monitor the effects of, cytokine-targeted monoclonal antibody therapy.
Collapse
Affiliation(s)
- Shivanthan Shanthikumar
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia; Respiratory and Sleep Medicine, Royal Children's Hospital, Parkville, VIC, Australia
| | - Liam Gubbels
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Karen Davies
- Otolaryngology, Royal Children's Hospital, Parkville, VIC, Australia
| | - Hannah Walker
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia; Children's Cancer Centre, Royal Children's Hospital, Parkville, VIC, Australia
| | - Anson Tsz Chun Wong
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Eric Levi
- Otolaryngology, Royal Children's Hospital, Parkville, VIC, Australia; Clinical Sciences, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Richard Saffery
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Sarath Ranganathan
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia; Respiratory and Sleep Medicine, Royal Children's Hospital, Parkville, VIC, Australia
| | - Melanie R Neeland
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
71
|
Yu X, Yao Y, Zhou H, Zhu J, Zhang N, Sang S, Zhou H. Integrating network pharmacology and experimental validation to explore the potential mechanism by which resveratrol acts on osimertinib resistance in lung cancer. Oncol Lett 2025; 29:192. [PMID: 40041411 PMCID: PMC11877012 DOI: 10.3892/ol.2025.14938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/16/2024] [Indexed: 03/06/2025] Open
Abstract
Globally, osimertinib resistance has been a long-term challenge. Resveratrol, a naturally occurring polyphenolic compound found in various plants, has the potential to modulate multidrug resistance mechanisms. However, the specific role of resveratrol in delaying osimertinib resistance in lung cancer is still unclear. The present study aimed to investigate the therapeutic effects and underlying mechanisms of resveratrol in delaying osimertinib resistance. Accordingly, the corresponding targets of resveratrol were screened through the Traditional Chinese Medicine Systems Pharmacology database. Similarly, the corresponding targets for osimertinib resistance were mined from the GeneCards database. A protein-protein interaction network was subsequently constructed to pinpoint key hub genes that resveratrol may target to delay resistance. Molecular docking analysis was then employed to assess the binding energy between the predicted key targets and resveratrol. Finally, in vitro experiments were performed to validate the results. Ultimately, 13 potential therapeutic targets of resveratrol related to delaying osimertinib resistance were identified. Kyoto Encyclopedia of Genes and Genomes analysis suggested that the effects of resveratrol may be associated with the apoptotic pathway. Molecular docking revealed that resveratrol has good binding affinities with MCL1 and BCL2L11. In vitro experiments confirmed that resveratrol inhibited the proliferation of osimertinib-resistant cells and upregulated the expression of BCL2L11. In conclusion, resveratrol may promote apoptosis by targeting BCL2L11 to delay osimertinib resistance.
Collapse
Affiliation(s)
- Xin Yu
- Department of Respiratory Medicine, Traditional Chinese Medical Hospital of Zhuji, Zhuji, Zhejiang 311800, P.R. China
| | - Yuan Yao
- Department of TCM, Shimen Er Lu Community Health Service Center of Jing'an District, Shanghai 200041, P.R. China
- Department of General Practice, Shanghai Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai 200433, P.R. China
| | - Haiwen Zhou
- Department of Respiratory Medicine, Traditional Chinese Medical Hospital of Zhuji, Zhuji, Zhejiang 311800, P.R. China
| | - Jintao Zhu
- Department of Respiratory Medicine, Traditional Chinese Medical Hospital of Zhuji, Zhuji, Zhejiang 311800, P.R. China
| | - Nini Zhang
- Department of Respiratory Medicine, Traditional Chinese Medical Hospital of Zhuji, Zhuji, Zhejiang 311800, P.R. China
| | - Shuliu Sang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
| | - Hailun Zhou
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
| |
Collapse
|
72
|
Cooper CL, Morrow G, Yuan M, Postler TS, Neal ML, Cross RW, Woolsey C, Agans KN, Borisevich V, McNamara RP, Atyeo C, Roy V, Germosen D, Hou F, Li SL, Reiserova L, Choi Y, Wilson A, Wagner D, Wallace-Selman O, Carpov A, Geng F, Frederick DJ, DeStefano J, Ercolini AM, Enriquez AS, Hastie KM, Ramos da Silva S, Sayeed E, Coleman JW, Kilianski A, Alter G, Saphire EO, Aitchison JD, Geisbert TW, Gupta SB, Feinberg MB, Parks CL. Preclinical development of a replication-competent vesicular stomatitis virus-based Lassa virus vaccine candidate advanced into human clinical trials. EBioMedicine 2025; 114:105647. [PMID: 40157130 DOI: 10.1016/j.ebiom.2025.105647] [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: 05/24/2024] [Revised: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Lassa fever (LF) is a zoonotic haemorrhagic disease caused by Lassa virus (LASV), which is endemic in West African countries. The multimammate rat is the main animal reservoir and its geographic range is expected to expand due to influences like climate change and land usage, and this will place larger parts of Africa at risk. We conducted preclinical development on a promising experimental vaccine that allowed its advancement into human trials. METHODS The LF vaccine is based on a vesicular stomatitis virus (VSV) vector in which the VSV glycoprotein (G) was replaced with the LASV glycoprotein complex (GPC). Earlier studies showed that this vaccine (VSVΔG-LASV-GPC) was efficacious in macaques, thus we regenerated VSVΔG-LASV-GPC using laboratory and documentation practices required to support vaccine manufacturing and human trials. The efficacy of the clinical vaccine candidate was assessed in cynomolgus macaques and more extensive immunologic analysis was performed than previously to investigate immune responses associated with protection. FINDINGS A single VSVΔG-LASV-GPC vaccination elicited innate, humoural and cellular immune responses, prevented development of substantial LASV viraemia, and protected animals from disease. Vaccinated macaques developed polyfunctional antibodies and serum was shown to neutralize virus expressing GPCs representative of geographically diverse LASV lineages. INTERPRETATION The VSVΔG-LASV-GPC clinical candidate elicited immunity that protected 10 of 10 vaccinated macaques from disease supporting its use in a clinical development program, which recently progressed to phase 2 clinical trials. Moreover, immunologic analysis showed that virus-neutralizing serum antibodies likely played a role in preventing LASV disease in vaccinated macaques. FUNDING This work was supported by the Coalition for Epidemic Preparedness Innovations (CEPI), The National Institute of Allergy and Infectious Diseases (NIAID)/National Institutes of Health (NIH), The Bill and Melinda Gates Global Vaccine Accelerator Program, the Burroughs Wellcome Fund, and financial gifts and support by Nancy Zimmerman, Mark and Lisa Schwartz, and Terry and Susan Ragon.
Collapse
Affiliation(s)
| | - Gavin Morrow
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Maoli Yuan
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Thomas S Postler
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Maxwell L Neal
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Robert W Cross
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Courtney Woolsey
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Krystle N Agans
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Viktoriya Borisevich
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ryan P McNamara
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA 02139, USA
| | - Caroline Atyeo
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA 02139, USA
| | - Vicky Roy
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA 02139, USA
| | - Daritza Germosen
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA 02139, USA
| | - Fuxiang Hou
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Shui L Li
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Lucia Reiserova
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Yesle Choi
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Aaron Wilson
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Denise Wagner
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | | | - Alexei Carpov
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Fuqiang Geng
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | | | - Joanne DeStefano
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Anne M Ercolini
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Adrian S Enriquez
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Kathryn M Hastie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | | | | | - John W Coleman
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | | | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA 02139, USA
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - John D Aitchison
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA; Departments of Pediatrics and Biochemistry, University of Washington, Seattle, WA 98109, USA
| | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | | | - Christopher L Parks
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA.
| |
Collapse
|
73
|
冯 苗, 徐 小, 李 宁, 杨 铭, 翟 远. [Mechanism of Cnidii Fructus in the treatment of periodontitis with osteoporosis based on network pharmacology, molecular docking, and molecular dynamics simulation]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2025; 43:249-261. [PMID: 40132971 PMCID: PMC11960409 DOI: 10.7518/hxkq.2025.2024275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 11/05/2024] [Indexed: 03/27/2025]
Abstract
OBJECTIVES This study aimed to explore the active components, potential targets, and mechanism of Cnidii Fructus in the treatment of periodontitis with osteoprosis through network pharmacology, molecular docking, and molecular dynamics simulation technology. METHODS The main chemical constituents and targets of Cnidii Fructus were screened using the TCMSP and SwissTargetPrediction databases, as well as literature reports. Targets of periodontitis and osteoporosis were predicted using different databases. The intersection targets of Cnidii Fructus, periodontitis, and osteoporosis were obtained using Venny 2.1. The protein-protein interaction network was formed on the STRING platform. Cytoscape 3.9.1 was used to construct the active component-intersection target interaction network, perform the topological analysis, and screen key targets and core active components. Furthermore, the Metascape database was used to perform gene ontology (GO) function and Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analysis on the intersection targets. The top five key targets and core active components were selected as receptor proteins and ligand small molecules. Discovery Studio 2019 was used to dock ligands and receptors and visualize the docking results. Molecular dynamics simulation was conducted using Gromacs2022.3 to assess the stability of the interactions between the core active components and the main targets. RESULTS A total of 20 potential active ingredients of Cnidii Fructus were screened, and 116 targets of Cnidii Fructus were obtained for treating periodontitis and osteoporosis. GO and KEGG analyses of the 116 targets showed that Cnidii Fructus may play a therapeutic role through the phosphoinositide 3-kinase-protein kinase B (PI3K-Akt) and advanced glycation end products-receptor for advanced glycation end products (AGE-RAGE) signaling pathways. Molecular docking showed that the core constituents were well bound to the main targets. Molecular dynamics simulations confirmed the stability of the Diosmetin-AKT1 complex system. CONCLUSIONS The preliminary discovery of the potential molecular pharmacological mechanism of Cnidii Fructus extract in the targeted treatment of periodontitis with osteoporosis through a multi-component, multitarget, and multi-pathway approach can serve as a theoretical foundation for future drug-development research and clinical application.
Collapse
|
74
|
Kaur B, Rana D, Sharma R, Konar M, Dhillon MS, Chouhan DK, Saini UC, Prakash M, Arora A, Verma I, Kaur J, Sharma S. Proteomic Insights Into Early Detection and Progression of Knee Osteoarthritis: Unveiling Molecular Signatures. Arch Med Res 2025; 56:103206. [PMID: 40174302 DOI: 10.1016/j.arcmed.2025.103206] [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: 05/16/2024] [Revised: 02/20/2025] [Accepted: 03/05/2025] [Indexed: 04/04/2025]
Abstract
AIM Osteoarthritis (OA) is the eleventh most disabling condition, with radiographic classification based on the Kellgren-Lawrence (KL) grading system. Early detection is critical to implement interventions to slow disease progression and improve patient outcomes. Proteomics, as a powerful strategy, could contribute to a better understanding of the disease pathophysiology and its early detection. OBJECTIVES The study aims to identify and confirm proteins associated with early detection and their role in the progression of knee OA. METHODOLOGY Synovial fluid (SF) and serum samples from the Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, were categorized by KL classification and subjected to SWATHTM analysis in the discovery phase. Seven samples of each OA grade were analyzed. A mass dynamics tool was used for data analysis and visualization. Significant protein expression level was defined as -1≤ log2FC ≥1 with p-value < 0.05, and ELISA was used for validation in a greater number of patients. RESULTS 29 significantly modulated proteins were observed in osteoarthritis grade comparisons. Cathepsin G (CTSG) and angiotensinogen (AGT) were upregulated, whereas fumarylacetoacetase (FAH) and neural cell adhesion molecule 1 (NCAM1) were downregulated with radiographic disease progression, as validated by ELISA. CTSG, AGT, and NCAM1 showed good sensitivity and specificity in discriminating between early and late OA grades. Notably, serum and synovial fluid levels of AGT and NCAM1 exhibited significant correlation. CONCLUSION This is one of the first studies to comprehensively analyze proteins associated with OA progression. Additionally, the identified protein signatures have great potential for OA progression and differential diagnosis of early and late-stage OA.
Collapse
Affiliation(s)
- Bhavneet Kaur
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Diksha Rana
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rinkle Sharma
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Monidipa Konar
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Mandeep S Dhillon
- Department of Orthopaedics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Devendra K Chouhan
- Department of Orthopaedics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Uttam Chand Saini
- Department of Orthopaedics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Mahesh Prakash
- Department of Radio Diagnosis and Imaging, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amit Arora
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Indu Verma
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Jyotdeep Kaur
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sadhna Sharma
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
75
|
Niu C, Meng X, Wang T. Identification of Ferroptosis-Inflammation Related Hub Genes and the Disease Subtypes in Idiopathic Pulmonary Fibrosis via System Biology Approaches. Mol Biotechnol 2025; 67:1720-1733. [PMID: 38734841 DOI: 10.1007/s12033-024-01158-x] [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: 12/26/2023] [Accepted: 03/24/2024] [Indexed: 05/13/2024]
Abstract
We aim to screen and analyze the ferroptosis inflammation-related hub genes associated with idiopathic pulmonary fibrosis (IPF). The GSE52463 and GSE110147 datasets were obtained from the GEO database and merged. The DEGs were selected by differential analysis and intersected with inflammation-related genes and ferroptosis-related genes to acquire the ferroptosis-related differentially expressed genes (FRDEGs). GO, KEGG, GSEA, and GSVA were performed to investigate the features of FRDEGs. The key module genes were selected by WGCNA and employed to generate the PPI network using Cytoscape. Subsequently, the hub genes were identified using cytoHubba and validated by ROC curves generated by survivalROC. Finally, the correlations of hub genes were analyzed through Spearman and the subtypes of IPF were constructed using ConsensusClusterPlus. A total of 1814 DEGs were screened out and 18 FRDEGs were acquired from the intersection of DEGs, ferroptosis-related genes, and inflammation-related genes. GO and KEGG analysis revealed that FRDEGs were primarily involved in bacterial-origin molecular, response infectious disease, and iron ion transport. GSEA results suggested a predominant association with autoimmune diseases and GSVA identified ten different pathways between PF and control. Through WGCNA, three highly correlated modules were identified and ten key module genes were obtained by intersecting genes in the three modules with FRDEGs. Finally, employing three algorithms within the cytoHubba led to the identification of eight hub genes: CCND1, TP53, STAT3, CTNNB1 CDH1, ESR1, HSP90AA1, and EP300. Eventually, two distinct subtypes of IPF were identified. The present research successfully identified the hub genes associated with ferroptosis and inflammation and their biological effects on IPF. Furthermore, two disease subtypes of IPF were constructed.
Collapse
Affiliation(s)
- Chongyang Niu
- Changchun University of Chinese Medicine, No. 1035 Boshuo Road, Jingyue Economic Development district, Changchun, Jilin Province, 130000, China
- Lung Disease Center, The Affiliated Hospital of Changchun University of Chinese Medicine, No. 1478 Gongnong Road, Chaoyang District, Changchun, Jilin Province, 130000, China
| | - Xiaoyu Meng
- Pediatric Center, The third affiliated hospital of Changchun university Chinese medicine, No. 1643 Jingyue Street, Nanguan District, Changchun, Jilin Province, 130000, China
| | - Tan Wang
- Lung Disease Center, The Affiliated Hospital of Changchun University of Chinese Medicine, No. 1478 Gongnong Road, Chaoyang District, Changchun, Jilin Province, 130000, China.
| |
Collapse
|
76
|
Yan H, Wu X, Li H, Yu Z, Jin X. Pan-Cancer Analysis Identifies BCLAF1 as a Potential Biomarker for Renal Cell Carcinoma. Biochem Genet 2025; 63:1479-1508. [PMID: 38573525 DOI: 10.1007/s10528-024-10773-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 03/04/2024] [Indexed: 04/05/2024]
Abstract
B-cell lymphoma-2-associated transcription factor 1 (BCLAF1) is a versatile protein involved in the regulation of gene transcription and post-transcriptional processing. Although BCLAF1 exerts a broad tumor suppressor effect or tumor promoter effect in many cancer types, the specific roles concerning its expression levels, and its impact on tumorigenesis in Renal cell carcinoma (RCC) remain unclear. Here, we utilized the Cancer Genome Atlas (TCGA) and Genotype Tissue Expression (GTEx) datasets alongside R software and online tools to unravel the specific roles of BCLAF1 in 33 cancer types, including its expression levels, tumor immune and molecular subtypes, and its correlation with prognosis, diagnosis, DNA methylation, and immune microenvironment. Additionally, we carried out cell biology experiments to independently investigate the expression of BCLAF1 in RCC and its effects on tumor progression. BCLAF1 was differentially expressed in tumor tissues compared to normal tissues across various cancer types and was also differentially expressed in different immune and molecular subtypes. In RCC, patients with high BCLAF1 expression had a better prognosis and BCLAF1 was tightly correlated with the stage, gender, and histological grade of patients. Furthermore, BCLAF1 had higher DNA methylation levels and higher immune infiltration levels in tumor tissues. Additionally, cell functional experiments confirmed the low expression of BCLAF1 in RCC and that BCLAF1 significantly inhibited the proliferation, migration, and invasion, while inducing apoptosis and cell cycle arrest in RCC cells in vitro. Our study under-scored the potential of BCLAF1 as an important actor in tumorigenesis, especially concerning RCC where it may serve as an effective prognostic marker.
Collapse
Affiliation(s)
- Huan Yan
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiang Wu
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Hong Li
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Zongdong Yu
- Department of Neurosurgery, Shangrao People's Hospital, Shangrao, 334099, China.
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
77
|
Kjærsgaard Andersen R, Stefansdottir L, Riis PT, Halldorsson G, Ferkingstad E, Oddsson A, Walters B, Olafsdottir TA, Rutsdottir G, Zachariae C, Thomsen SF, Brodersen T, Dinh KM, Knowlton KU, Knight S, Nadauld LD, Banasik K, Brunak S, Hansen TF, Hjalgrim H, Sørensen E, Mikkelsen C, Ullum H, Nyegaard M, Bruun MT, Erikstrup C, Ostrowski SR, Eidsmo L, Saunte DML, Sigurgeirsson B, Orvar KB, Saemundsdottir J, Melsted P, Norddahl GL, Sulem P, Stefansson H, Holm H, Gudbjartsson D, Thorleifsson G, Jonsdottir I, Pedersen OBV, Jemec GBE, Stefansson K. A genome-wide association meta-analysis links hidradenitis suppurativa to common and rare sequence variants causing disruption of the Notch and Wnt/β-catenin signaling pathways. J Am Acad Dermatol 2025; 92:761-772. [PMID: 39645042 DOI: 10.1016/j.jaad.2024.11.050] [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: 02/16/2024] [Revised: 10/15/2024] [Accepted: 11/15/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND The contributions of genetic and environmental risk factors to hidradenitis suppurativa (HS) are both poorly understood. OBJECTIVE To identify sequence variants that associate with HS and determine the contribution of environmental risk factors and inflammatory diseases to HS pathogenesis. METHODS A genome-wide association meta-analysis of 4814 HS cases (Denmark: 1977; Iceland: 1266; Finland: 800; UK: 569; and US: 202) and 1.2 million controls, searching for sequence variants associated with HS. RESULTS We found 8 independent sequence variants associating with HS, 6 common and 2 rare (frequency <1%). Four associations point to candidate causal genes, NCSTN, PSENEN, WNT10A, and TMED10, that all map to the Notch and Wnt/β-catenin signaling pathways, involved in epidermal keratinization. LIMITATIONS Limited racial diversity may prevent identification of sequence variants of particular importance in non-Caucasian populations. CONCLUSIONS These findings demonstrate that genes and pathways involved in epidermal keratinization are the genetic backbone of HS pathology.
Collapse
Affiliation(s)
- Rune Kjærsgaard Andersen
- Department of Dermatology, Zealand University Hospital, Roskilde, Denmark; Department of Immunology and Microbiology, Leo Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark.
| | | | - Peter Theut Riis
- Department of Dermatology, Zealand University Hospital, Roskilde, Denmark
| | - Gisli Halldorsson
- deCODE Genetics/Amgen, Inc, Reykjavik, Iceland; School of Engineering and Natural Sciences, University of Iceland, Reykjavík, Iceland
| | | | | | | | - Thorunn A Olafsdottir
- deCODE Genetics/Amgen, Inc, Reykjavik, Iceland; Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | | | - Claus Zachariae
- Department of Dermatology and Allergy, Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Simon Francis Thomsen
- Department of Dermatology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Thortsen Brodersen
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
| | - Khoa Manh Dinh
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Kirk U Knowlton
- Intermountain Medical Center, Intermountain Heart Institute, Salt Lake City, Utah; University of Utah, School of Medicine, Salt Lake City, Utah
| | - Stacey Knight
- Intermountain Medical Center, Intermountain Heart Institute, Salt Lake City, Utah; University of Utah, School of Medicine, Salt Lake City, Utah
| | | | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Folkmann Hansen
- Danish Headache Center, Department of Neurology, Copenhagen University Hospital, Rigshospitalet-Glostrup, Copenhagen, Denmark
| | - Henrik Hjalgrim
- Danish Cancer Society Research Center, Danish Cancer Society, Copenhagen, Denmark; Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark; Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Chirstina Mikkelsen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Science, Copenhagen University, Copenhagen, Denmark
| | | | - Mette Nyegaard
- Department of Health Science and Technology, Aalborg University, Gistrup, Denmark
| | - Mie Topholm Bruun
- Clinical Immunology Research Unit, Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Liv Eidsmo
- Department of Immunology and Microbiology, Leo Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Ditte Marie Lindhardt Saunte
- Department of Dermatology, Zealand University Hospital, Roskilde, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Bárdur Sigurgeirsson
- Department of Dermatology, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Kjartar B Orvar
- Department of Medicine, Landspitali, The National University Hospital of Iceland, Reykjavík, Iceland
| | | | - Pall Melsted
- deCODE Genetics/Amgen, Inc, Reykjavik, Iceland; School of Engineering and Natural Sciences, University of Iceland, Reykjavík, Iceland
| | | | | | | | - Hilma Holm
- deCODE Genetics/Amgen, Inc, Reykjavik, Iceland
| | - Daniel Gudbjartsson
- deCODE Genetics/Amgen, Inc, Reykjavik, Iceland; School of Engineering and Natural Sciences, University of Iceland, Reykjavík, Iceland
| | | | - Ingileif Jonsdottir
- deCODE Genetics/Amgen, Inc, Reykjavik, Iceland; Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Ole Birger Vesterager Pedersen
- Department of Dermatology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Gregor Borut Ernst Jemec
- Department of Dermatology, Zealand University Hospital, Roskilde, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Kari Stefansson
- deCODE Genetics/Amgen, Inc, Reykjavik, Iceland; Faculty of Medicine, University of Iceland, Reykjavík, Iceland.
| |
Collapse
|
78
|
Zhang X, Ma S, Naz SI, Soderblom EJ, Aliferis C, Kraus VB. Plasma extracellular vesicles carry immune system-related peptides that predict human longevity. GeroScience 2025; 47:1455-1469. [PMID: 39695065 PMCID: PMC11979029 DOI: 10.1007/s11357-024-01454-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: 09/15/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024] Open
Abstract
Extracellular vesicles (EVs) play crucial roles in aging. In this National Institutes on Aging-funded study, we sought to identify circulating extracellular vesicle (EV) biomarkers indicative of longevity. The plasma EV proteome of 48 older adults (mean age 77.2 ± 1.7 years [range 72-80]; 50% female, 50% Black, 50% < 2-year survival, 50% ≥ 10-year survival) was analyzed by high-resolution mass spectrometry and flow cytometry. The ability of EV peptides to predict longevity was evaluated in discovery (n = 32) and validation (n = 16) datasets with areas under receiver operating characteristic curves (AUCs). Longevity-associated large EV (LEV) plasma subpopulations were mainly related to immune cells (HLA-ABC+, CD9+, and CD31+) and muscle cells (MCAD+ and RyR2+). Of 7960 identified plasma EV peptides (519 proteins), 46.4% were related to the immune system and 10.1% to muscle. Compared with short-lived older adults, 756 EV peptides (131 proteins) had a higher abundance, and 130 EV peptides (78 proteins) had a lower abundance in long-lived adults. Among longevity-associated peptides, 437 (58 proteins) were immune system related, and 12 (2 proteins) were muscle related. Using just three to five plasma EV peptides (mainly complement components C2-C6), we achieved high predictive accuracy for longevity (AUC range 0.91-1 in a hold-out validation dataset). Our findings suggest that immune cells produce longevity-associated plasma EVs and elucidate fundamental mechanisms regulating aging and longevity. EV longevity predictors suggest there may be merit in targeting complement pathways to extend lifespan, for instance, with any one of the multiple complement inhibitors currently available or in clinical development.
Collapse
Affiliation(s)
- Xin Zhang
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, 27701, USA.
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, 27701, USA.
| | - Sisi Ma
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN, USA
| | - Syeda Iffat Naz
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN, USA
| | - Erik J Soderblom
- Duke Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Durham, NC, USA
| | - Constantin Aliferis
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN, USA
| | - Virginia Byers Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, 27701, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, 27701, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
79
|
Andersen MD, Wolter K, Enemark MH, Lauridsen KL, Hamilton-Dutoit SJ, Starklint J, d'Amore F, Ludvigsen M, Honoré B, Kamper P. Proteomic profiling identifies classic Hodgkin lymphoma patients at risk of bleomycin pulmonary toxicity. Leuk Lymphoma 2025; 66:656-667. [PMID: 39625996 DOI: 10.1080/10428194.2024.2434170] [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: 08/13/2024] [Revised: 10/21/2024] [Accepted: 11/20/2024] [Indexed: 03/29/2025]
Abstract
Advances in treating classic Hodgkin lymphoma (cHL) have improved cure rates, with overall survival exceeding 80%, resulting in a growing population of survivors at risk of long-term complications, particularly cardiac and pulmonary toxicity. Bleomycin, a key component of combination chemotherapy, is associated with bleomycin-induced pulmonary toxicity (BPT). Using label-free quantification nano liquid chromatography-tandem mass spectrometry, protein expression in diagnostic lymphoma samples from patients with and without BPT was compared. Results showed differential protein expression and disrupted cellular pathways, suggesting biological differences in BPT risk. Immunohistochemical analysis revealed higher expression of JAK3, BID, and MMP9, and lower expression of CD20, TPD52, and PIK3R4 in patients with BPT. High BID and low CD20 expression were associated with inferior overall survival, while high BID and low JAK3 and CD20 expression were linked to poorer progression-free survival. These findings highlight altered protein profiles in pretreatment cHL biopsies associated with BPT development.
Collapse
Affiliation(s)
- Maja Dam Andersen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Katharina Wolter
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Marie Hairing Enemark
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | | | - Jørn Starklint
- Department of Medicine, Regional Hospital Goedstrup, Herning, Denmark
| | - Francesco d'Amore
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Maja Ludvigsen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Bent Honoré
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Peter Kamper
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
80
|
Huo P, Li Z, Jin S, Wang S, Luo Y, Zhu L, Jin Z. Mechanism of β‑sitosterol in treating keloids: Network pharmacology, molecular docking and experimental verification. Mol Med Rep 2025; 31:95. [PMID: 39981895 PMCID: PMC11868730 DOI: 10.3892/mmr.2025.13460] [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: 10/03/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
β‑sitosterol (SIT) has anti‑inflammatory, anti‑tumor and anti‑fibrotic effects. However, the precise mechanisms underlying its efficacy in keloid treatment remain elusive. The present study aimed to elucidate the therapeutic effect of SIT on keloids. The active components of Fructus arctii, target molecules of these components and disease‑associated target molecules were identified and retrieved from various databases. Molecular docking was employed to evaluate the binding affinity of the active compounds for key targets. Cell viability and proliferation were evaluated via CCK‑8 and EdU assays, while cell migration capacity was assessed via wound healing assays and cell migration and invasion abilities were determined via Transwell assays. A rescue study involving YS‑49 was conducted. Western blot analysis was performed to assess the expression levels of proteins associated with EMT and proteins involved in the PI3K/AKT signaling pathway. A subcutaneous keloid fibroproliferative model was established in nude mice and immunohistochemical staining was performed on tissue sections. By intersecting the keloid targets, 29 targets were identified, with 10 core targets revealed by protein-protein interaction analysis. Molecular docking revealed a robust binding affinity between SIT and PTEN. In addition to inhibiting cell viability, invasion and migration, SIT significantly decreased the levels of phosphorylated (p‑)PI3K and p‑AKT, downregulated the protein expression of Vimentin and Snail proteins and increased the protein expression of Zonula Occludens‑1 and E‑cadherin. YS‑49 reversed the inhibitory effect of SIT on keloid in SIT‑treated cells. In vivo experiments demonstrated that SIT suppressed the growth of a keloid model in nude mice and increased PTEN expression. The present study provided the first evidence that SIT inhibits keloid proliferation, migration and invasion by modulating the PTEN/PI3K/AKT signaling pathway, suggesting its potential as a novel therapeutic approach for keloid treatment.
Collapse
Affiliation(s)
- Pingping Huo
- Keloid Research Center, Department of Dermatology, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China
- Department of Medicine, Yanbian University Medical College, Yanji, Jilin 133000, P.R. China
- Department of Medical Cosmetology, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China
| | - Zhouna Li
- Department of Medical Cosmetology, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China
| | - Shan Jin
- Keloid Research Center, Department of Dermatology, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China
| | - Sujie Wang
- Keloid Research Center, Department of Dermatology, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China
- Department of Medicine, Yanbian University Medical College, Yanji, Jilin 133000, P.R. China
| | - Yinli Luo
- Keloid Research Center, Department of Dermatology, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China
- Department of Medicine, Yanbian University Medical College, Yanji, Jilin 133000, P.R. China
| | - Lianhua Zhu
- Department of Medicine, Yanbian University Medical College, Yanji, Jilin 133000, P.R. China
- Department of Dermatology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong 511518, P.R. China
| | - Zhehu Jin
- Keloid Research Center, Department of Dermatology, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China
- Department of Medicine, Yanbian University Medical College, Yanji, Jilin 133000, P.R. China
- Department of Medical Cosmetology, Yanbian University Hospital, Yanji, Jilin 133000, P.R. China
| |
Collapse
|
81
|
Gonçalves-Carneiro D, Mastrocola E, Lei X, Bieniasz PD. Modulation of host gene expression by the zinc finger antiviral protein. Proc Natl Acad Sci U S A 2025; 122:e2420819122. [PMID: 40146858 DOI: 10.1073/pnas.2420819122] [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: 10/09/2024] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
The zinc finger antiviral protein (ZAP) depletes nonself RNAs through recognition of their elevated CpG dinucleotide content. CpG dinucleotides are sparse in most endogenous mammalian mRNAs, but a subset might potentially be modulated by ZAP. While CpG frequency alone is insufficient to predict ZAP-regulation, we developed an algorithm using experimentally determined compositional features to predict which endogenous mRNAs may be ZAP-regulated. Using ZAP-knockout mice, we demonstrate that levels of many host mRNAs that are algorithmically predicted ZAP targets are indeed increased when ZAP is absent. ZAP is interferon-inducible, and we also identify genes that are downregulated by ZAP during an innate immune response. Many ZAP-regulated gene products are extracellular matrix or of nucleosome components, whose ZAP-mediated control is conserved in human cells. Overall, we provide a tool for the prediction of ZAP target genes and reveal host mRNAs that are ZAP-regulated.
Collapse
Affiliation(s)
- Daniel Gonçalves-Carneiro
- Department of Infectious Disease, St Mary's Medical School, Imperial College London, London W2 1NY, United Kingdom
- Laboratory of Retrovirology, The Rockefeller University, New York, NY 10065
| | - Emily Mastrocola
- Laboratory of Retrovirology, The Rockefeller University, New York, NY 10065
| | - Xiao Lei
- Laboratory of Retrovirology, The Rockefeller University, New York, NY 10065
| | - Paul D Bieniasz
- Laboratory of Retrovirology, The Rockefeller University, New York, NY 10065
- HHMI, The Rockefeller University, New York, NY 10065
| |
Collapse
|
82
|
Xiao T, Rahhal O, Wang L, Deng Z, Wang R, Xu X, Qi L, Tang Z. TPPP3, a Good Prognostic Indicator, Suppresses Cell Proliferation and Migration in OSCC. Int Dent J 2025; 75:970-983. [PMID: 39814636 PMCID: PMC11976587 DOI: 10.1016/j.identj.2024.09.035] [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: 08/26/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 01/18/2025] Open
Abstract
INTRODUCTION AND AIMS Oral squamous cell carcinoma (OSCC) is one of the most prevalent malignancy of the head and neck. Early diagnosis of OSCC is difficult and the prognosis has not improved significantly. This study aims to explore the role of tubulin polymerisation promoting protein 3 (TPPP3) in the occurrence and development of OSCC and discover new diagnostic and prognostic markers for OSCC. METHODS Using UALCAN, GEPIA, western blot, and quantitative real-time polymerase chain reaction, we studied TPPP3 expression and its relationship with tumour stage. Then, we detected the effect of TPPP3 on OSCC biological functions by CCK-8 and cell scratch assays, as well as correlations between TPPP3 expression and survival of different kinds of head and neck squamous cell carcinoma (HNSC) patients through Kaplan-Meier plotter. Besides, we explored coexpressed genes associated with TPPP3 in HNSC using LinkedOmics and protein-protein interaction networks of TPPP3 using STRING and Cytoscape. Furthermore, we explored possible molecular mechanisms that TPPP3 functions in HNSC using UALCAN, Kaplan-Meier plotter, and TIMER. Finally, we analysed promoter methylation level by UALCAN and mutation by cBioPortal of TPPP3 in HNSC. RESULTS TPPP3 was less expressed in OSCC. The TPPP3 expression level was negatively correlated with tumour stage. Furthermore, TPPP3 significantly inhibited OSCC proliferation and migration. Besides, TPPP3 high expression was significantly associated with good prognosis in different kinds of HNSC patients. Additionally, TPPP3 may regulate the occurrence and development of OSCC through the PALMD/PI3K pathway. TPPP3 methylation level in HNSC decreased. Finally, we found that TPPP3 genetic alteration was involved in TPPP3 mRNA expression change in HNSC. CONCLUSION TPPP3 functions as a tumour suppressor in OSCC and is associated with good prognosis in HNSC patients. TPPP3 can be used as a potential biomarker for prognosis and diagnosis of OSCC. CLINICAL RELEVANCE TPPP3 can be used as a potential biomarker for prognosis and diagnosis of OSCC in clinical practice.
Collapse
Affiliation(s)
- Ting Xiao
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Omar Rahhal
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Liping Wang
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Zhiyuan Deng
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Ran Wang
- Beijing Nuclear Industry Hospital, Beijing, China
| | - Xinghuanyu Xu
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Lu Qi
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China.
| | - Zhangui Tang
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China.
| |
Collapse
|
83
|
Ismail NZ, Khairuddean M, Abubakar S, Arsad H. Network pharmacology, molecular docking and molecular dynamics simulation of chalcone scaffold-based compounds targeting breast cancer receptors. J Biomol Struct Dyn 2025; 43:3242-3257. [PMID: 38149857 DOI: 10.1080/07391102.2023.2296606] [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: 08/01/2023] [Accepted: 12/12/2023] [Indexed: 12/28/2023]
Abstract
Compounds with a chalcone scaffold-based structure have demonstrated promising anticancer biological activity. However, the molecular interactions between chalcone scaffold-based compounds and breast cancer-associated proteins remain unclear. Through network pharmacology, molecular docking, and molecular dynamics (MD) simulation analyses, compounds with a chalcone scaffold-based structure were evaluated for their interaction with potential breast cancer targets. The compounds were retrieved from the ASINEX database, resulting in 575,302 compounds. A total of 342 compounds with chalcone scaffold-based structures were discovered. From the 342 compounds that was analysed, ten were chosen due to their adherence to Lipinski's rule, having an appropriate range of lipophilicity (LOGP), and topological polar surface area (TPSA), and absence of any toxicity. Based on target intersection, 50 target genes were found and subjected to protein-protein interaction (PPI), gene ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Four target genes were found to be involved in the breast cancer pathway. Consequently, molecular docking was utilised to analyse the molecular interactions between the compounds and four target protein receptors. Compound 211 exhibited the highest binding affinities for the epidermal growth factor receptor (EGFR), fibroblast growth factor receptor 1 (FGFR1), oestrogen receptor (ESR1), and cyclin dependent kinase 6 (CDK6) with values of -8.95 kcal/mol, -8.60 kcal/mol, -10.33 kcal/mol, and -9.90 kcal/mol, respectively. During MD simulation, compound 211 and its respective proteins were stable, compact, and had minimal flexibility. The findings provide foundations for future studies into the interaction underlying the anti-breast cancer potential of compounds with chalcone-based scaffold structures.
Collapse
Affiliation(s)
| | - Melati Khairuddean
- School of Chemical Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | - Sadiq Abubakar
- School of Chemical Sciences, Universiti Sains Malaysia, Penang, Malaysia
- Department of Pure and Industrial Chemistry, Bayero University Kano, Kano, Nigeria
| | - Hasni Arsad
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| |
Collapse
|
84
|
Sun T, Hazra A, Lui A, Zeng S, Wang X, Rao S, Owens LA, Fei Z, Zhao Y, Mazourek M, Giovannoni JG, Li L. GLKs directly regulate carotenoid biosynthesis via interacting with GBFs in plants. THE NEW PHYTOLOGIST 2025; 246:645-665. [PMID: 39953697 DOI: 10.1111/nph.20457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/18/2025] [Indexed: 02/17/2025]
Abstract
Carotenoids are vital photosynthetic pigments for plants. Golden2-like transcription factors (GLKs) are widely recognized as major regulators of Chl biosynthesis and chloroplast development. However, despite GLKs being subjected to intensive investigations, whether GLKs directly regulate carotenoid biosynthesis and the molecular mechanisms by which GLKs transcriptionally activate their target genes remain unclear. Here, we report that GLKs directly regulate carotenoid biosynthesis and activate their target genes in a G-box binding factor (GBF)-dependent manner in Arabidopsis. Both in vitro and in vivo studies reveal that GLKs physically interact with GBFs to activate transcription of phytoene synthase (PSY), the gene encoding a rate-limiting enzyme for carotenoid biosynthesis. While GLKs possess transactivation activity, they depend on GBFs to directly bind to the G-box motif to modulate PSY expression. Loss of GBFs impairs GLK function in regulating carotenoid and Chl biosynthesis. Since the G-box motif is an enriched motif in the promoters of GLK-regulated genes, the GLK-GBF regulatory module likely serves as a common mechanism underlying GLK-regulated photosynthetic pigment biosynthesis and chloroplast development. Our findings uncover a novel regulatory machinery of carotenoid biosynthesis, discover a molecular mechanism of transcriptional regulation by GLKs, and divulge GLKs as important regulators to coordinate photosynthetic pigment synthesis in plants.
Collapse
Affiliation(s)
- Tianhu Sun
- Robert W. Holley Center for Agriculture and Health, USDA-ARS, Cornell University, Ithaca, NY, 14853, USA
- Plant Breeding and Genetics Section, School of Integrative Plant Science, Cornell University, Ithaca, NY, 14853, USA
- Department of Biological Sciences, College of Arts and Sciences, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Abhijit Hazra
- Robert W. Holley Center for Agriculture and Health, USDA-ARS, Cornell University, Ithaca, NY, 14853, USA
- Plant Breeding and Genetics Section, School of Integrative Plant Science, Cornell University, Ithaca, NY, 14853, USA
| | - Andy Lui
- Robert W. Holley Center for Agriculture and Health, USDA-ARS, Cornell University, Ithaca, NY, 14853, USA
- Plant Breeding and Genetics Section, School of Integrative Plant Science, Cornell University, Ithaca, NY, 14853, USA
| | - Shaohua Zeng
- Robert W. Holley Center for Agriculture and Health, USDA-ARS, Cornell University, Ithaca, NY, 14853, USA
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic improvement, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, China
| | - Xin Wang
- Boyce Thompson Institute, Ithaca, NY, 14853, USA
| | - Sombir Rao
- Robert W. Holley Center for Agriculture and Health, USDA-ARS, Cornell University, Ithaca, NY, 14853, USA
- Plant Breeding and Genetics Section, School of Integrative Plant Science, Cornell University, Ithaca, NY, 14853, USA
| | - Lauren A Owens
- Robert W. Holley Center for Agriculture and Health, USDA-ARS, Cornell University, Ithaca, NY, 14853, USA
| | - Zhangjun Fei
- Boyce Thompson Institute, Ithaca, NY, 14853, USA
| | - Yunde Zhao
- Section of Cell and Developmental Biology, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Michael Mazourek
- Plant Breeding and Genetics Section, School of Integrative Plant Science, Cornell University, Ithaca, NY, 14853, USA
| | - James G Giovannoni
- Robert W. Holley Center for Agriculture and Health, USDA-ARS, Cornell University, Ithaca, NY, 14853, USA
- Boyce Thompson Institute, Ithaca, NY, 14853, USA
| | - Li Li
- Robert W. Holley Center for Agriculture and Health, USDA-ARS, Cornell University, Ithaca, NY, 14853, USA
- Plant Breeding and Genetics Section, School of Integrative Plant Science, Cornell University, Ithaca, NY, 14853, USA
| |
Collapse
|
85
|
Sun R, Wang Y, Zhu R, Li L, Xi Q, Dai Y, Li J, Cao Y, Guo X, Pan X, Wang Q, Zhang B. Genome-wide identification of CA genes in cotton and the functional analysis of GhαCA4-D, GhβCA6-D and GhγCA2-D in response to drought and salt stresses. Int J Biol Macromol 2025; 304:140872. [PMID: 39938833 DOI: 10.1016/j.ijbiomac.2025.140872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 01/25/2025] [Accepted: 02/08/2025] [Indexed: 02/14/2025]
Abstract
Carbonic anhydrases (CAs) are critical metalloenzymes, widely exist in organisms, which involve in many physiological processes, including response to adverse environmental conditions. Although CA genes have been comprehensive identified and analyzed in numerous plants, there are a few of reports in cotton. Therefore, we conducted an exhaustive research for CA genes from two tetraploid cotton species and their ancestral species. A total of 138 CA genes were found, and 45 of them belonged to Gossypium hirsutum. Phylogenetic relationships and sequences analysis showed that CA genes were categorized into three distinct subtypes: α-type, β-type and γ-type. The exon numbers of β-type members were highly variable. Various types of cis-elements, including drought inducibility, were identified in CA genes, suggesting that CA genes might be involved in the regulation of drought stress response. qRT-PCR was applied to assess the gene expression level in various tissues under drought stress. The results indicated that the expression levels of GhαCA4-D, GhβCA1-A, GhβCA1-D, GhβCA3-D and GhβCA6-D were significantly higher in leaves than that in stems and roots. The expression of GhαCA4-A, GhαCA8-A, GhαCA4-D, GhβCA3-D, GhβCA6-D and GhγCAL1-D was significantly upregulated in roots at severe drought treatment. The functions of GhαCA4-D, GhβCA6-D and GhγCA2-D were analyzed using virus-induced gene silencing (VIGS) technology. Compared to the controls, GhγCA2-D-silenced upland cotton seedlings were more sensitive to salt stress. However, the drought tolerance of GhαCA4-D and GhβCA6-D silenced plants was significantly decreased. Stomatal density, width and area were significantly higher in TRV:GhβCA6-D compared to TRV:00 inoculated plants. GhαCA4-D silenced plants were susceptible to oxidative stress, and silencing GhαCA4-D induced leave cell death. Our results will assist to make clear the regulatory mechanism of CA genes under abiotic stress.
Collapse
Affiliation(s)
- Runrun Sun
- Henan International Joint Laboratory of Functional Genomics and Molecular Breeding of Cotton, Henan Collaborative Innovation Center of Modern Biological Breeding, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Yuanyuan Wang
- Henan International Joint Laboratory of Functional Genomics and Molecular Breeding of Cotton, Henan Collaborative Innovation Center of Modern Biological Breeding, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Ruihao Zhu
- Henan International Joint Laboratory of Functional Genomics and Molecular Breeding of Cotton, Henan Collaborative Innovation Center of Modern Biological Breeding, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Lijie Li
- Henan International Joint Laboratory of Functional Genomics and Molecular Breeding of Cotton, Henan Collaborative Innovation Center of Modern Biological Breeding, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China; Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Qianhui Xi
- Henan International Joint Laboratory of Functional Genomics and Molecular Breeding of Cotton, Henan Collaborative Innovation Center of Modern Biological Breeding, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Yunpeng Dai
- Henan International Joint Laboratory of Functional Genomics and Molecular Breeding of Cotton, Henan Collaborative Innovation Center of Modern Biological Breeding, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Jiahui Li
- Henan International Joint Laboratory of Functional Genomics and Molecular Breeding of Cotton, Henan Collaborative Innovation Center of Modern Biological Breeding, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Yuanyuan Cao
- Henan International Joint Laboratory of Functional Genomics and Molecular Breeding of Cotton, Henan Collaborative Innovation Center of Modern Biological Breeding, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Xinlei Guo
- Henan International Joint Laboratory of Functional Genomics and Molecular Breeding of Cotton, Henan Collaborative Innovation Center of Modern Biological Breeding, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Xiaoping Pan
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Qinglian Wang
- Henan International Joint Laboratory of Functional Genomics and Molecular Breeding of Cotton, Henan Collaborative Innovation Center of Modern Biological Breeding, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China.
| | - Baohong Zhang
- Department of Biology, East Carolina University, Greenville, NC 27858, USA.
| |
Collapse
|
86
|
Sayedyahossein S, Huang K, Zhang C, Karimi M, Bahmani M, O’Donnell BL, Wakefield B, Li Z, Johnston D, Leighton SE, Huver MS, Dagnino L, Sacks DB, Penuela S. Pannexin 1 crosstalk with the Hippo pathway in malignant melanoma. FEBS J 2025; 292:1633-1653. [PMID: 39786847 PMCID: PMC11971028 DOI: 10.1111/febs.17396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/19/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
In this study, we explored the intricate relationship between Pannexin 1 (PANX1) and the Hippo signaling pathway effector, Yes-associated protein (YAP). Analysis of The Cancer Genome Atlas (TCGA) data revealed a significant positive correlation between PANX1 mRNA and core Hippo components, Yes-associated protein 1 [YAP], Transcriptional coactivator with PDZ-binding motif [TAZ], and Hippo scaffold, Ras GTPase-activating-like protein IQGAP1 [IQGAP1], in invasive cutaneous melanoma and breast carcinoma. Furthermore, we demonstrated that PANX1 expression is upregulated in invasive melanoma cell lines and is associated with increased YAP protein levels. Notably, our investigations uncovered a previously unrecognized interaction between endogenous PANX1 and the Hippo scaffold protein IQGAP1 in melanoma cells. Moreover, our findings revealed that IQGAP1 exhibits differential expression in melanoma cells and plays a regulatory role in cellular morphology. Functional studies involving PANX1 knockdown provided compelling evidence that PANX1 modulates YAP protein levels and its cotranscriptional activity in melanoma and breast carcinoma cells. Importantly, our study highlights the potential therapeutic significance of targeting PANX1. Pharmacological inhibition of PANX1 using selective FDA-approved inhibitors or PANX1 knockdown reduced YAP levels in melanoma cells. Furthermore, our Clariom™ S analysis unveiled key genes implicated in cell proliferation, such as neuroglin1 (NRG1), β-galactoside binding protein and galectin-3 (LGALS3), that are affected in PANX1-deficient cells. In summary, our investigation delves into the intricate interplay between PANX1 and YAP in the context of invasive melanoma, offering valuable insights into potential therapeutic strategies for effective treatment.
Collapse
Affiliation(s)
- Samar Sayedyahossein
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland, USA, 20892
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Kenneth Huang
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Christopher Zhang
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Mehdi Karimi
- Department of Mathematics, Illinois State University, Normal, Illinois, USA, 61790
| | | | - Brooke L. O’Donnell
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Brent Wakefield
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Zhigang Li
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland, USA, 20892
| | - Danielle Johnston
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Stephanie E. Leighton
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Matthew S. Huver
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Lina Dagnino
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - David B. Sacks
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, Maryland, USA, 20892
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1
| |
Collapse
|
87
|
Sarfraz M, Bakht MA, Alshammari MS, Alrofaidi M, Alzahrani AR, Eltaib L, Asdaq SMB, Aba Alkhayl FF, Abida, Mohd Imran. Beyond traditional medications: exploring novel and potential inhibitors of trypanothione reductase (LmTr) of Leishmania parasites. J Biomol Struct Dyn 2025; 43:3130-3143. [PMID: 38213287 DOI: 10.1080/07391102.2023.2300062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/07/2023] [Indexed: 01/13/2024]
Abstract
The trypanothione reductase enzyme, which neutralizes the reactive oxygen species produced inside the macrophages to kill the parasites, is one of the evasion strategies Leishmania uses to survive inside the cells. The vitality of the parasite depends on Leishmania major trypanothione reductase (LmTr), a NADPH-dependent flavoprotein oxidoreductase essential for thiol metabolism. Since this enzyme is distinct and lacking in humans, we focused on it in our study to screen for new inhibitors to combat leishmaniasis. Using the I-TASSER server, a three-dimensional model of LmTr was generated. The Autodock vina program was used in high-throughput virtual screening of the ZINC database. The top seven molecules were ranked according to their binding affinity. The compounds with the highest binding affinities and the right number of hydrogen bonds were chosen. These compounds may be effective at inhibiting the target enzyme's (LmTr) activity, making them new leishmaniasis treatments. These compounds may serve as a useful starting point for a hit-to-lead approach in the quest for new anti-Leishmania drugs that are more efficient and less cytotoxic. The average node degree is 5.09, the average local clustering coefficient is 0.868, and the PPI enrichment p-value is 8.9e-06, indicating that it is sufficiently connected to regulate the network. TRYR (LmTr protein) also interacts physically with ten additional proteins in the pathogenesis network. The findings of the study indicated that successfully suppressing the LmTr protein in vitro and in vivo may finally result in regulating the L. major pathogenesis.
Collapse
Affiliation(s)
- Muhammad Sarfraz
- College of Pharmacy, Al Ain University, Al Ain, United Arab Emirates
| | - M Afroz Bakht
- Department of Chemistry, College of Science and Humanity Studies, Prince Sattam Bin, Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Mohammed Sanad Alshammari
- Department of Computer Science, Faculty of Computing and Information Technology, Northern Border University, Rafha, Saudi Arabia
| | - Mohammad Alrofaidi
- Department of Pharmaceutical Chemistry, Faculty of Clinical Pharmacy, Al-Baha University, Al-Baha, Saudi Arabia
| | - Abdullah R Alzahrani
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Al-Abidiyah, Makkah, Saudi Arabia
| | - Lina Eltaib
- Department of Pharmaceutics, Faculty of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | | | - Faris F Aba Alkhayl
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Abida
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| |
Collapse
|
88
|
Dong M, Liu X, Zhao C, Fang Z, Wang Z, Guo X, Wang Y, Li Y, Ye M, Jia L. Temporal resolved multi-proteomic analysis enabled the systematic characterization of N-glycosylation pattern changes during Jurkat T cell activation. Anal Bioanal Chem 2025; 417:2169-2183. [PMID: 39998645 DOI: 10.1007/s00216-025-05805-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025]
Abstract
Protein glycosylation plays essential roles in regulating innate and adaptive immune response. Previous studies only focused on individual protein-glycan interactions or specific glycoform changes during T cell activation, yet the systematic characterization of protein glycosylation alterations remains insufficiently elucidated. To address these limitations, we conducted temporally resolved quantitative analysis of glycoforms, site-specific glycans, glycoproteins, and glycosylation enzymes in activated Jurkat T cells, and successfully portrayed the dynamic landscape of protein glycosylation during Jurkat T cell activation. We found the heterogeneity and number of significantly upregulated glycopeptides increased along with activation. For most glycopeptides, their alteration patterns did not correlate with the abundance of their glycoprotein substrates. However, functional molecules including CD69, CD28, and PTPRC demonstrated co-upregulation at both the protein and glycosylation levels. Correlation analysis between glycopeptides and glycotransferases indicated that sialylated or fucosylated peptides were well correlated with enzymes involved in glycan branching and capping. Comparative analysis of global peptides, glycopeptides, and phosphopeptides revealed their distinctive changing patterns along Jurkat T cell activation, and only glycosylation demonstrated a steady increase trend with a large proportion of upregulated glycopeptides. Collectively, this integrated multi-proteomics characterization of activated Jurkat T cells provided insights for the development of novel therapeutic strategy targeting glycosylation.
Collapse
Affiliation(s)
- Mingming Dong
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, 116000, Liaoning, China.
| | - Xiaoyan Liu
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Changrui Zhao
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, 116000, Liaoning, China
| | - Zheng Fang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Zhongyu Wang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xin Guo
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, 116000, Liaoning, China
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Yan Wang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Yanan Li
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Mingliang Ye
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lingyun Jia
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, 116000, Liaoning, China
| |
Collapse
|
89
|
Försti A, Ambrozkiewicz F, Marciniak M, Lubinski J, Hemminki K. Search for germline gene variants in colorectal cancer families presenting with multiple primary colorectal cancers. Int J Cancer 2025; 156:1393-1403. [PMID: 39654522 PMCID: PMC11789446 DOI: 10.1002/ijc.35283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/15/2024] [Accepted: 11/22/2024] [Indexed: 02/04/2025]
Abstract
A double primary colorectal cancer (CRC) in a familial setting signals a high risk of CRC. In order to identify novel CRC susceptibility genes, we whole-exome sequenced germline DNA from nine persons with a double primary CRC and a family history of CRC. The detected variants were processed by bioinformatics filtering and prioritization, including STRING protein-protein interaction and pathway analysis. A total of 150 missense, 19 stop-gain, 22 frameshift and 13 canonical splice site variants fulfilled our filtering criteria. The STRING analysis identified 20 DNA repair/cell cycle proteins as the main cluster, related to genes CHEK2, EXO1, FAAP24, FANCI, MCPH1, POLL, PRC1, RECQL, RECQL5, RRM2, SHCBP1, SMC2, XRCC1, in addition to CDK18, ENDOV, ZW10 and the known mismatch repair genes. Another STRING network included extracellular matrix genes and TGFβ signaling genes. In the nine whole-exome sequenced patients, eight harbored at least two candidate DNA repair/cell cycle/TGFβ signaling gene variants. The number of families is too small to provide evidence for individual variants but, considering the known role of DNA repair/cell cycle genes in CRC, the clustering of multiple deleterious variants in the present families suggests that these, perhaps jointly, contributed to CRC development in these families.
Collapse
Affiliation(s)
- Asta Försti
- Hopp Children's Cancer Center (KiTZ)HeidelbergGermany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ)German Cancer Consortium (DKTK)HeidelbergGermany
| | - Filip Ambrozkiewicz
- Biomedical Center, Faculty of MedicineCharles University PilsenPilsenCzech Republic
| | - Magdalena Marciniak
- Department of Genetics and Pathology, International Hereditary Cancer CenterPomeranian Medical University in SzczecinSzczecinPoland
| | - Jan Lubinski
- Department of Genetics and Pathology, International Hereditary Cancer CenterPomeranian Medical University in SzczecinSzczecinPoland
| | - Kari Hemminki
- Biomedical Center, Faculty of MedicineCharles University PilsenPilsenCzech Republic
- Division of Cancer EpidemiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| |
Collapse
|
90
|
Shen R, Wang J, Zhao Y, Dang Z, Zhang K, Li M, Yang Q, Gao LN. Polysaccharides from Scrophularia ningpoensis Hemsl. improve reserpine-induced depression-like behavior by inhibiting HTR2A/HTR2C mediated AKT/GSK3β/β-catenin/CBP/BDNF signalling. Int J Biol Macromol 2025; 301:140445. [PMID: 39884598 DOI: 10.1016/j.ijbiomac.2025.140445] [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: 07/15/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Scrophularia ningpoensis Hemsl. is a traditional Chinese medicine used to regulate blood sugar levels, immunity, etc. We previously isolated polysaccharides from S. ningpoensis Hemsl. (SNPS) and innovatively observed that SNPS exhibit antidepressant properties; however, the underlying mechanism is still unclear. Here, we employed network pharmacology to predict the potential targets and antidepressant mechanism of SNPS. Accordingly, we detected the effects of SNPS on monoamine neurotransmitter synthesis, metabolism, receptor expression and signal transduction in reserpine (RES)-treated mice using ELISA, HPLC-electrochemistry, metabonomics, Golgi-Cox staining and Western blotting. Finally, the mechanism of SNPS on key targets (HTR2A and HTR2C) was verified in vivo and in vitro. Results showed that SNPS ameliorated depression by restoring monoamine neurotransmitter homeostasis and hippocampal neurogenesis. SNPS reversed the depletion of 5-HT, NE and DA by activating the tryptophan (Trp)/5-HT and tyrosine (Tyr)/DA/NE metabolic pathways. SNPS decreased HTR2A and HTR2C contents, leading to the phosphorylation of AKT and GSK3β, followed by increases in β-catenin, CBP and BDNF levels. Mechanistically, SNPS reduced the levels of HTR2A and HTR2C proteins by inhibiting their mRNA transcription, rather than inducing protein degradation. In conclusion, by inhibiting the transcription of HTR2A and HTR2C, SNPS activated the AKT/GSK3β/β-catenin/CBP/BDNF pathway, thereby exerting dose-dependent antidepressant effects.
Collapse
Affiliation(s)
- Ruhui Shen
- College of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, PR China; College of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, PR China; Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai 20080, PR China
| | - Jian'an Wang
- College of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, PR China
| | - Yijin Zhao
- College of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, PR China
| | - Zhaojin Dang
- College of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, PR China
| | - Ke Zhang
- College of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, PR China
| | - Ming Li
- College of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, PR China
| | - Qian Yang
- College of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, PR China
| | - Li-Na Gao
- College of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, PR China; College of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, PR China; Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, Shandong 272067, PR China; Jining Key Laboratory of Depression Prevention and Treatment, Jining Medical University, Jining, Shandong 272067, PR China.
| |
Collapse
|
91
|
Martino RA, Volke DC, Tenaglia AH, Tribelli PM, Nikel PI, Smania AM. Genetic Dissection of Cyclic di-GMP Signalling in Pseudomonas aeruginosa via Systematic Diguanylate Cyclase Disruption. Microb Biotechnol 2025; 18:e70137. [PMID: 40172309 PMCID: PMC11963287 DOI: 10.1111/1751-7915.70137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 04/04/2025] Open
Abstract
The second messenger bis-(3' → 5')-cyclic dimeric guanosine monophosphate (c-di-GMP) governs adaptive responses in the opportunistic pathogen Pseudomonas aeruginosa, including biofilm formation and the transition from acute to chronic infections. Understanding the intricate c-di-GMP signalling network remains challenging due to the overlapping activities of numerous diguanylate cyclases (DGCs). In this study, we employed a CRISPR-based multiplex genome-editing tool to disrupt all 32 GGDEF domain-containing proteins (GCPs) implicated in c-di-GMP signalling in P. aeruginosa PA14. Phenotypic and physiological analyses revealed that the resulting mutant was unable to form biofilms and had attenuated virulence. Residual c-di-GMP levels were still detected despite the extensive GCP disruption, underscoring the robustness of this regulatory network. Taken together, these findings provide insights into the complex c-di-GMP metabolism and showcase the importance of functional overlapping in bacterial signalling. Moreover, our approach overcomes the native redundancy in c-di-GMP synthesis, providing a framework to dissect individual DGC functions and paving the way for targeted strategies to address bacterial adaptation and pathogenesis.
Collapse
Affiliation(s)
- Román A. Martino
- Universidad Nacional de CórdobaFacultad de Ciencias Químicas, Departamento de Química Biológica Ranwel CaputtoCórdobaArgentina
- CONICET, Universidad Nacional de CórdobaCentro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC)CórdobaArgentina
| | - Daniel C. Volke
- The Novo Nordisk Foundation Center for BiosustainabilityTechnical University of DenmarkKongens LyngbyDenmark
| | - Albano H. Tenaglia
- Universidad Nacional de CórdobaFacultad de Ciencias Químicas, Departamento de Química Biológica Ranwel CaputtoCórdobaArgentina
- CONICET, Universidad Nacional de CórdobaCentro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC)CórdobaArgentina
| | - Paula M. Tribelli
- Universidad de Buenos AiresFacultad de Ciencias Exactas y Naturales, Departamento de Química BiológicaBuenos AiresArgentina
- CONICET, Universidad de Buenos AiresInstituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)Buenos AiresArgentina
| | - Pablo I. Nikel
- The Novo Nordisk Foundation Center for BiosustainabilityTechnical University of DenmarkKongens LyngbyDenmark
| | - Andrea M. Smania
- Universidad Nacional de CórdobaFacultad de Ciencias Químicas, Departamento de Química Biológica Ranwel CaputtoCórdobaArgentina
- CONICET, Universidad Nacional de CórdobaCentro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC)CórdobaArgentina
| |
Collapse
|
92
|
Padam KSR, Pereira SD, Kumar NAN, Radhakrishnan R. Natural Antisense Transcript-Mediated Regulation of HOXA10-AS in Oral Squamous Cell Carcinoma. J Oral Pathol Med 2025; 54:217-231. [PMID: 40038044 PMCID: PMC11986565 DOI: 10.1111/jop.13613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/29/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND The oncogenic role of HOXA10-AS and HOXA10 in cancer has been well documented. However, the epigenetic role of HOXA10 and the natural antisense-mediated regulation of HOXA10-AS in oral squamous cell carcinoma progression is not understood. METHODS A total of 35 oral squamous cell carcinoma specimens and 35 adjacent normal clinical specimens were collected and categorized on the basis of their lymph node status. HOXA10-AS and HOXA10 expression were analyzed using RT-qPCR. Methyl-capture sequencing was performed using lymph node-negative (n = 6) and lymph node-positive (n = 5) matched cases. The promoter activity of HOXA10 was determined using a luciferase assay. ChIP-qPCR was performed to determine histone mark localization in the distal promoter region of HOXA10. A protein-protein interaction network of genome-wide antisense targets was constructed using StringDB, and functional enrichment was performed using the R package ClusterProfiler. Transient siRNA-mediated transfection was performed to target specific exons of the HOXA10-AS gene, followed by subsequent cell proliferation, cell cycle, and cell migration assays and validation of cancer signaling pathways through western blotting. RESULTS HOXA10-AS and its antisense target HOXA10 were significantly overexpressed in the lymph node-positive samples. The transcriptionally active distal promoter of HOXA10 consists of a constitutively unmethylated CpG island region (CUR). H3K4me3, H3K27ac, and H3K27me3 histone mark deposition at the adjacent methylated loci of the distal promoter suggest the nature of euchromatin-driven regulation. Genome-wide mapping revealed 11 potential targets of HOXA10-AS. Targeted specific knockdown of HOXA10-AS exons significantly reduced the expression of HOXA10 and deregulated its downstream targets, contributing to decreased cell cycle progression and epithelial-to-mesenchymal transition. CONCLUSION HOXA10-AS regulates the expression of HOXA10 through a natural antisense-mediated mechanism and is epigenetically regulated by constitutively unmethylated marks in the distally enhancing promoter of HOXA10.
Collapse
Affiliation(s)
- Kanaka Sai Ram Padam
- Department of Cell and Molecular Biology, Manipal School of Life SciencesManipal Academy of Higher EducationManipalKarnatakaIndia
| | - Satyajit Dey Pereira
- Department of Cell and Molecular Biology, Manipal School of Life SciencesManipal Academy of Higher EducationManipalKarnatakaIndia
| | - Naveena A. N. Kumar
- Department of Surgical Oncology, Kasturba Medical College and HospitalManipal Academy of Higher EducationManipalKarnatakaIndia
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, ManipalManipal Academy of Higher EducationManipalKarnatakaIndia
- Academic Unit of Oral and Maxillofacial Pathology, School of Clinical DentistryUniversity of SheffieldSheffieldUK
- Academic Unit of Oral Biology and Oral PathologyMuscatWattayahSultanate of Oman
| |
Collapse
|
93
|
Lotfi MS, Jamali H, B Rassouli F. Network pharmacology and in silico study of quercetin and structurally similar flavonoids as osteogenesis inducers that interact with oestrogen receptors. Arch Physiol Biochem 2025:1-12. [PMID: 40160020 DOI: 10.1080/13813455.2025.2483910] [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: 04/08/2024] [Revised: 09/04/2024] [Accepted: 03/18/2025] [Indexed: 04/02/2025]
Abstract
Background: Osteoporosis poses a global health challenge, particularly with an ageing population. Quercetin, isorhamnetin, avicularin, isoquercetin, quercitrin, and taxifolin are natural flavonoids with similar structure that induce ontogenesis. Methods: In the present study, proteins in oestrogen signalling and bone morphogenesis were analysed, and hub genes were identified with Cytoscape, followed by pathway analysis. Then, molecular targets of flavonoids and osteoporosis-related targets were identified, and overlaps were detected. Molecular docking and dynamics simulations assessed flavonoid interactions with ERs. Results: The study identified 14 gene products linked to osteoporosis, including ESR1 and ESR2. Enrichment analyses confirmed ESR involvement in various biological processes. SwissTargetPrediction highlighted quercetin and isorhamnetin as favourable targets for ESR1 and ESR2. Molecular docking and dynamics revealed favourable and stable binding of flavonoids to ERα and ERβ. Conclusion: These interactions suggest therapeutic potential of natural flavonoids for osteoporosis treatment by targeting ERs, laying a foundation for future research in preclinical and clinical settings.
Collapse
Affiliation(s)
- Mohammad-Sadegh Lotfi
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hamidreza Jamali
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Fatemeh B Rassouli
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
94
|
Akçay S, Gurkok-Tan T, Ekici S. Identification of key genes in immune-response post-endurance run in horses. J Equine Vet Sci 2025; 149:105418. [PMID: 40174711 DOI: 10.1016/j.jevs.2025.105418] [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: 09/09/2024] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 04/04/2025]
Abstract
Intense physical activity in endurance horses triggers complex immune and inflammatory responses, yet the molecular mechanisms underlying these adaptations remain unclear. This study investigated immune-related transcriptomic changes following a 160 km endurance ride, focusing on sex-based differences. Using a bioinformatics approach, differentially expressed genes (DEGs), pathways, microRNAs (miRNAs), and transcription factors (TFs) were analyzed before (T0) and after (T1) the ride. A protein-protein interaction (PPI) analysis was conducted to identify key regulatory genes. Pathway enrichment analysis revealed significant activation of immune-regulatory and ribosomal pathways. Notably, TLR4, CXCL8, and CCL5 were identified as key hub genes involved in immune modulation post-exercise. Comparisons between female (FT1 vs FT0) and gelding (GT1 vs GT0) horses revealed distinct molecular responses. Female horses exhibited upregulation of ribosomal protein genes, suggesting enhanced protein synthesis and muscle recovery. In contrast, geldings showed increased expression of inflammatory and stress-related genes, indicating a heightened immune response. Notably, sex-based differences were observed, with FT1 vs FT0 and GT1 vs GT0 comparisons revealing distinct KEGG pathway enrichments. Additionally, miRNA and TF analyses revealed regulatory elements influencing endurance-related immune responses. Our findings demonstrated sex-specific molecular mechanisms underlying endurance exercise adaptation, with females prioritizing protein synthesis and recovery, while geldings exhibit stronger inflammatory responses and stress-related pathways. This study provides critical insights into how sex influences exercise physiology at the transcriptomic level, with potential applications in training and recovery strategies for endurance horses.
Collapse
Affiliation(s)
- S Akçay
- Department of Molecular Biology of Genetics, Kırşehir Ahi Evran University, Bagbaşı, 40100, Kırşehir Turkey
| | - T Gurkok-Tan
- Department of Field Crops, Food and Agriculture Vocational School, Cankiri Karatekin University, Merkez, 18100, Çankırı, Turkey
| | - S Ekici
- Veterinary Control Central Research Institute, Keçiören, 06100, Ankara, Turkey.
| |
Collapse
|
95
|
Akter NN, Uddin MM, Uddin N, Asha IJ, Uddin MS, Hossain MA, Alam F, Shifat SK, Zihad MA, Rahman MH. Structural and Functional Characterization of a Putative Type VI Secretion System Protein in Cronobacter sakazakii as a Potential Therapeutic Target: A Computational Study. Evol Bioinform Online 2025; 21:11769343251327660. [PMID: 40171225 PMCID: PMC11960190 DOI: 10.1177/11769343251327660] [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: 10/03/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Background Cronobacter sakazakii, a foodborne pathogen with a fatality rate of 33%, is a rod-shaped, Gram-negative, non-spore-forming bacterium responsible for causing meningitis, bacteremia, and necrotizing enterocolitis. Despite many unknown functions of hypothetical proteins in bacterial genomes, bioinformatic techniques have successfully annotated their roles in various pathogens. Objectives The aim of this investigation is to identify and annotate the structural and functional properties of a hypothetical protein (HP) from Cronobacter sakazakii 7G strain (accession no. WP_004386962.1, 277 residues) using computational tools. Methods Multiple bioinformatic tools were used to identify the homologous protein and to construct and validate its 3D structure. A 3D model was generated using SWISS-MODEL and validated using tools, developing a reliable 3D structure. The STRING and CASTp servers provided information on protein-protein interactions and active sites, identifying functional partners. Results The putative protein was soluble, stable, and localized in the cytoplasmic membranes, indicating its biological activity. Functional annotation identified TagJ (HsiE1) within the protein, a member of the ImpE superfamily involved in the transport of toxins and a part of the bacterial type VI secretion system (T6SS). The 3-dimensional structure of this protein was validated through molecular docking involving 6 different compounds. Among these, ceforanide demonstrated the strongest binding scores, -7.5 kcal/mol for the hypothetical protein and -7.2 kcal/mol for its main template protein (PDB ID: 4UQX.1). Conclusion Comparative genomics study suggests that the protein found in C. sakazakii may be a viable therapeutic target because it seems distinctive and different from human proteins. The results of multiple sequence alignment (MSA) and molecular docking supported HP's potential involvement as a T6SS. These in silico results represent that the examined HP could be valuable for studying C. sakazakii infections and creating medicines to treat C. sakazakii-mediated disorders.
Collapse
Affiliation(s)
- Nurun Nahar Akter
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Md. Moin Uddin
- Department of Biotechnology, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Nesar Uddin
- Department of Apparel Engineering, Textile Engineering College, Noakhali, Bangladesh
| | - Israt Jahan Asha
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Md Soyeb Uddin
- Department of Botany, Cox’s Bazar Government College, Cox’s Bazar, Bangladesh
| | - Md. Arju Hossain
- Department of Biochemistry and Biotechnology, Khwaja Yunus Ali University, Sirajganj, Bangladesh
| | - Fahadul Alam
- Department of Biological Sciences, Texas Tech University, TX, USA
| | - Siratul Kubra Shifat
- Department of Bioprocess and Genetic Engineering, Chattogram Veterinary and Animal Sciences University, Chattogram, Bangladesh
| | - Md. Abu Zihad
- Department of Microbiology, Primeasia University, Dhaka, Bangladesh
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Islamic University, Kushtia, Bangladesh
- Center for Advanced Bioinformatics and Artificial Intelligence Research, Islamic University, Kushtia, Bangladesh
| |
Collapse
|
96
|
Gao J, Sun L, Liao R, Lyu Y, Zhang S, Xu J, He M, Wu C, Zhang D, Lin Y, Dai J. Genomic Dissection of Chinese Yangtze River Delta White Goat Based on Whole Genome Sequencing. Animals (Basel) 2025; 15:979. [PMID: 40218372 DOI: 10.3390/ani15070979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/16/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
The conservation and utilization of livestock genetic resources is essential for the maintenance of biodiversity and breed innovation. Whole genome sequencing (WGS) was performed on 90 samples from Chinese Yangtze River Delta White goats (YRD), sourced from two populations of Chongming island white goats and Haimen white goats, aiming to dissect their genomic characteristics. In addition, 262 WGS data from nine other breeds of goats were downloaded from the NCBI database. These WGS data obtained were used to identify and analyze genetic variation with the goat reference genome, and the genetic structure of goat populations was analyzed. Through selective sweep analysis, the selection-signature genes and their polymorphic features were identified. It was found that the most significant genomic selection region in YRD goats was in the region of 62.9-64.6 Mb on chromosome 13, which contained genes related to the coat color and muscle growth of the goats. Nucleotide diversity of MYH7B, a gene related to the development of the goat's skeletal muscle, within the Yangtze River Delta white goat population was significantly lower than in other domestic and foreign goat breeds, suggesting that the gene was subject to selection. In addition, the IGF2BP2 gene, reported to be associated with litter size in goats, showed clear selection-signature characteristics in the Boer goats compared to the YRD goats.
Collapse
Affiliation(s)
- Jun Gao
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
- Shanghai Municipal Key Laboratory of Agri-Genetics and Breeding, Shanghai 201106, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
| | - Lingwei Sun
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
- Shanghai Municipal Key Laboratory of Agri-Genetics and Breeding, Shanghai 201106, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
| | - Rongrong Liao
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
| | - Yuhua Lyu
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
| | - Shushan Zhang
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
- Shanghai Municipal Key Laboratory of Agri-Genetics and Breeding, Shanghai 201106, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
| | - Jiehuan Xu
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
- Shanghai Municipal Key Laboratory of Agri-Genetics and Breeding, Shanghai 201106, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
| | - Mengqian He
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
- Shanghai Municipal Key Laboratory of Agri-Genetics and Breeding, Shanghai 201106, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
| | - Caifeng Wu
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
- Shanghai Municipal Key Laboratory of Agri-Genetics and Breeding, Shanghai 201106, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
| | - Defu Zhang
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
- Shanghai Municipal Key Laboratory of Agri-Genetics and Breeding, Shanghai 201106, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
| | - Yuexia Lin
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
| | - Jianjun Dai
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
- Shanghai Municipal Key Laboratory of Agri-Genetics and Breeding, Shanghai 201106, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
| |
Collapse
|
97
|
Iacovacci J, Brough R, Moughari FA, Alexander J, Kemp H, Tutt ANJ, Natrajan R, Lord CJ, Haider S. Proteogenomic discovery of RB1-defective phenocopy in cancer predicts disease outcome, response to treatment, and therapeutic targets. SCIENCE ADVANCES 2025; 11:eadq9495. [PMID: 40138429 PMCID: PMC11939072 DOI: 10.1126/sciadv.adq9495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 02/11/2025] [Indexed: 03/29/2025]
Abstract
Genomic defects caused by truncating mutations or deletions in the Retinoblastoma tumor suppressor gene (RB1) are frequently observed in many cancer types leading to dysregulation of the RB pathway. Here, we propose an integrative proteogenomic approach that predicts cancers with dysregulation in the RB pathway. A subset of these cancers, which we term as "RBness," lack RB1 genomic defects and yet phenocopy the transcriptional profile of RB1-defective cancers. We report RBness as a pan-cancer phenomenon, associated with patient outcome and chemotherapy response in multiple cancer types, and predictive of CDK4/6 inhibitor response in estrogen-positive breast cancer. Using RNA interference and a CRISPR-Cas9 screen in isogenic models, we find that RBness cancers also phenocopy synthetic lethal vulnerabilities of cells with RB1 genomic defects. In summary, our findings suggest that dysregulation of the RB pathway in cancers lacking RB1 genomic defects provides a molecular rationale for how these cancers could be treated.
Collapse
Affiliation(s)
- Jacopo Iacovacci
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- Data Science Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano 20133, Italy
| | - Rachel Brough
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Fatemeh Ahmadi Moughari
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - John Alexander
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Harriet Kemp
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Andrew N. J. Tutt
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Rachael Natrajan
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Christopher J. Lord
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Syed Haider
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| |
Collapse
|
98
|
Jiang H, Zhou L, Zhang H, Yu Z. Bioinformatic analysis of glycolysis and lactate metabolism genes in head and neck squamous cell carcinoma. Sci Rep 2025; 15:10781. [PMID: 40155682 PMCID: PMC11953422 DOI: 10.1038/s41598-025-94843-z] [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: 12/19/2024] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a heterogeneous cancer with significant global incidence. This study investigates glycolysis- and lactate metabolism-related genes (GALMRGs) in HNSCC, focusing on their impact on prognosis, the tumor immune microenvironment, and their potential as therapeutic biomarkers. Analysis of data from the Cancer Genome Atlas and Gene Expression Omnibus identified 16 GALMRGs that were differentially expressed in HNSCC compared to normal tissues. Functional analysis revealed the involvement of lactate and pyruvate metabolism and HIF-1 signaling pathways. Weighted gene co-expression network analysis identified two module genes, CDKN3 and SLC2A1. Five key genes (CAV1, CDKN3, LDHA, MB, and PER2) were identified through univariate, multivariate, and LASSO regression analyses and used to construct a prognostic model. This model demonstrated strong predictive accuracy for overall survival, stratifying patients into high- and low-risk groups. Immune cell infiltration analysis showed a negative correlation between resting and activated mast cells, and low-risk patients had higher tumor mutational burden, suggesting a better response to immunotherapy. Consensus clustering classified HNSCC into two distinct molecular subtypes with differing expression of the key genes. This GALMRG-based prognostic model is a promising biomarker for predicting HNSCC outcomes and immunotherapy responses, providing valuable insights for personalized treatment strategies.
Collapse
Affiliation(s)
- Huanyu Jiang
- School of Medicine, Southeast University, 87 Dingjiaqiao, Hunan Road, Nanjing, 210009, Jiangsu, China
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Benq Hospital of Nanjing Medical University, Nanjing, 210019, Jiangsu, China
| | - Lijuan Zhou
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Benq Hospital of Nanjing Medical University, Nanjing, 210019, Jiangsu, China
| | - Haidong Zhang
- School of Medicine, Southeast University, 87 Dingjiaqiao, Hunan Road, Nanjing, 210009, Jiangsu, China
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Benq Hospital of Nanjing Medical University, Nanjing, 210019, Jiangsu, China
| | - Zhenkun Yu
- School of Medicine, Southeast University, 87 Dingjiaqiao, Hunan Road, Nanjing, 210009, Jiangsu, China.
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Benq Hospital of Nanjing Medical University, Nanjing, 210019, Jiangsu, China.
| |
Collapse
|
99
|
Zhu M, Hong J, Liu X, Wang H, Lou L. Comprehensive analysis of GDFs as therapeutic targets and prognosis biomarkers in gastric cancer. Medicine (Baltimore) 2025; 104:e41976. [PMID: 40153751 PMCID: PMC11957613 DOI: 10.1097/md.0000000000041976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/10/2025] [Indexed: 03/30/2025] Open
Abstract
Growth/differentiation factors (GDFs, GDF1-3, GDF5-7, GDF9-11, and GDF15) belong to a subfamily of the transforming growth factor-β. GDFs play an important role in morphogenetic and developmental activities in many tissues. And many GDFs family numbers have been observed to be correlated with various types of tumors. However, the diverse expression patterns and prognostic values of ten GDFs in gastric cancer (GC) have yet to be analyzed. Herein we investigated the transcriptional and survival data of GDFs in patients with GC from the Gene Expression Profiling Interactive Analysis, The Cancer Genome Atlas, cBioPortal, Tumor Immune Estimation Resource, Tumor Immune Syngeneic Mouse, UALCAN, Human Protein Atlas Gene Expression Omnibus and The Database for Annotation, Visualization and Integrated Discovery databases. We found that multiple GDF family members are highly expressed in GC, which can prompt diagnosis and evaluate prognosis, and can be used as target points for GC immunotherapy.
Collapse
Affiliation(s)
- Minjie Zhu
- Department of General Surgery, Hangzhou Third People’s Hospital, Hangzhou, Zhejiang, China
| | - Jiawei Hong
- Department of Hepatobiliary and Pancreatic Surgery, Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan (Hangzhou) Hospital, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, P.R. China
| | - Xianfang Liu
- Department of Cardiology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Haiming Wang
- Department of General Surgery, Hangzhou Third People’s Hospital, Hangzhou, Zhejiang, China
| | - Longquan Lou
- Department of General Surgery, Hangzhou Third People’s Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
100
|
Fernández-Olivares A, Orellana VP, Llanquinao J, Nuñez G, Pérez-Moreno P, Contreras-Riquelme S, Martin AJ, Mammano F, Alfaro IE, Calderón JF, Stehberg J, Sáez MA, Retamal MA. Connexin46 in the nucleus of cancer cells: a possible role as transcription modulator. Cell Commun Signal 2025; 23:153. [PMID: 40148950 PMCID: PMC11948717 DOI: 10.1186/s12964-025-02151-w] [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: 11/13/2024] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Oncogenes drive cancer progression, but few are active exclusively in tumor cells. Connexins (Cxs), traditionally recognized as ion channel proteins, can localize to the nucleus and regulate gene expression, playing key roles in both physiological and pathological processes. Cx46, once thought to be restricted to the eye lens, has been implicated in tumor growth, though its underlying mechanisms remain unclear. This study investigates the nuclear presence of Cx46 in cancer cells and its potential role as a transcriptional modulator. METHODS We employed ChIP-Seq, confocal immunofluorescence, and nuclear protein purification to assess Cx46 localization and DNA interactions. Functional assays were conducted to evaluate its effects on invasion, division, spheroid formation, and mesenchymal marker expression. Single-point mutations and molecular dynamics simulations were used to explore potential Cx46-DNA interactions. RESULTS Cx46 mRNA upregulation was found in a variety of tumors compared to adjacent healthy tissue. In HeLa cells, which do not express Cx46, its transfection promoted proliferation, invasion and self-renewal capacity, cancer stem cell traits and mesenchymal features. Consistently, in Sk-Mel-2, which naturally express Cx46, reduced Cx46 expression led to a decrease in the similar parameters. In HeLa cells, nuclear Cx46 was detected in two forms, full length 46 kDa and a 30 kDa fragment (GJA3-30 k), ChIP-Seq experiments revealed that Cx46 binds to the DNA at intergenic and promoter regions, leading to the activation of oncogenic pathways. Molecular dynamics simulations suggest that GJA3-30 k dimerizes in a RAD50-like structure, forming stable DNA complexes. Cx46 and in some cases GJA3-30 k were detected in the nuclei of multiple cancer cell lines, including prostate, breast and skin cancers. CONCLUSIONS Our findings reveal a novel nuclear role for Cx46 in cancer, demonstrating its function as a transcriptional regulator and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Ainoa Fernández-Olivares
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Viviana P Orellana
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Jesús Llanquinao
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
- Translational Medicine Laboratory, Instituto Oncológico Fundación Arturo López Pérez, Santiago 7500691, Chile
| | - Gonzalo Nuñez
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Pablo Pérez-Moreno
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Sebastián Contreras-Riquelme
- Plant Genome Regulation Lab, Centro de Biotecnología Vegetal, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Alberto Jm Martin
- Laboratorio de Redes Biológicas, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago, Chile
- Escuela de Ingeniería, Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Santiago, Chile
| | - Fabio Mammano
- Department of Physics and Astronomy "G. Galilei", University of Padova, Institute of Biochemistry and Cell Biology, Italian National Research Council, 00015 Monterotondo, Padova, 35131, Italy
| | - Ivan E Alfaro
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
- Centro Ciencia & Vida, Fundación Ciencia & Vida. Nuñoa, Santiago, Chile
| | - Juan F Calderón
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Jimmy Stehberg
- Laboratorio de Neurobiología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad Andres Bello, Santiago, 8370146, Chile
| | - Mauricio A Sáez
- Departamento de Procesos Diagnósticos y Evaluación, Laboratorio de Investigación en Salud de Precisión, Facultad de Ciencias de la Salud, Universidad Católica de Temuco, Temuco, Chile.
| | - Mauricio A Retamal
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile.
| |
Collapse
|