1
|
Wu J, Xu S, Li Z, Cong B, Yang Z, Yang Z, Gao W, Liu S, Yu Z, Xu S, Li N, Hou J, Wang G, Cao X, Liu S. SARS-CoV-2 enhances complement-mediated endothelial injury via the suppression of membrane complement regulatory proteins. Emerg Microbes Infect 2025; 14:2467781. [PMID: 39945674 PMCID: PMC11873982 DOI: 10.1080/22221751.2025.2467781] [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: 10/30/2024] [Revised: 01/29/2025] [Accepted: 02/11/2025] [Indexed: 03/01/2025]
Abstract
Complement hyperactivation and thrombotic microangiopathy are closely associated with severe COVID-19. Endothelial dysfunction is a key mechanism underlying thrombotic microangiopathy. To address the relationship between endothelial injury, complement activation and thrombotic microangiopathy of severe COVID-19, we wonder whether, and if so, what and how SARS-CoV-2 factors make endothelial cells (ECs) sensitive to complement-mediated cytotoxicity. We revealed that multiple SARS-CoV-2 proteins enhanced complement-mediated cytotoxicity to ECs by inhibiting membrane complement regulatory proteins (CRPs) and enhancing the deposition of complement-recognizing component FCN1. By screening with CRISPR/Cas9-gRNA libraries, we identified that ADAMTS9, SYAP1, and HIGD1A as intrinsic regulators of CD59 on ECs, which were inhibited by the SARS-CoV-2 M, NSP16, and ORF9b proteins. IFN-γ, GM-CSF, and IFN-α upregulated CD55 and CD59, while IFN-γ antagonized the inhibition of CD59 by the three SARS-CoV-2 proteins. So, the deficiency of IFN-γ weakened the protection of ECs by CRPs against complement-mediated injury which may be enhanced during infection. Our findings illustrated the regulation of protection against complement-mediated attack on self-cells by SARS-CoV-2 infection and immune responses, providing insights into endothelial injury, thrombotic microangiopathy, and potential targets for treating severe COVID-19.
Collapse
Affiliation(s)
- Jian Wu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
| | - Sanpeng Xu
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, People’s Republic of China
| | - Zhiqing Li
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
| | - Boyi Cong
- Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, People’s Republic of China
| | - Zongheng Yang
- Department of Immunology, Center for Immunotherapy, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Zhichao Yang
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
| | - Wanfeng Gao
- Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, People’s Republic of China
| | - Shuo Liu
- Department of Immunology, Center for Immunotherapy, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Zhou Yu
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
| | - Sheng Xu
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
| | - Nan Li
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
| | - Jin Hou
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
| | - Guoping Wang
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, People’s Republic of China
| | - Xuetao Cao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
- Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, People’s Republic of China
- Department of Immunology, Center for Immunotherapy, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Shuxun Liu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
2
|
Yang L, Yang W, Shen Y, Zhou Y. Advance in candidate genes in mandibular retrognathism: A systematic review. Arch Oral Biol 2025; 174:106234. [PMID: 40132276 DOI: 10.1016/j.archoralbio.2025.106234] [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] [Revised: 02/15/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025]
Abstract
OBJECTIVE This research aims to dissect the polygenic nature of non-syndromic mandibular retrognathism (MR) and to better understand the genetic underpinnings of MR, with a particular focus on the role of ethnic diversity in influencing genetic predispositions. METHODS A comprehensive systematic review was conducted on MR. Electronic databases such as PubMed and Google Scholar were employed, utilizing terms like 'mandibular', 'retrognathism', 'gene', and 'genetic'. This study strictly adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) framework. RESULTS Ten genetic studies were identified that satisfied the eligibility criteria, involving 1010 participants. Variations in candidate genes were reported across different populations, including myosin 1 H (MYO1H), matrilin 1 (MATN1), a disintegrin and metalloproteinase with thrombospondin motifs 9 (ADAMTS9), bone morphogenetic protein 2 (BMP2), parathyroid hormone (PTH), the vitamin-D related genes: vitamin D receptor (VDR), cytochrome P450 family 24 subfamily A member 1 (CYP24A1), and cytochrome P450 family 27 subfamily B member 1 (CYP27B1), collagen type II alpha 1 chain (COL2A1), transforming growth factor-β (TGF-β), TGF-β receptor 2 (TGFBR2), epidermal growth factor (EGF), and EGF receptor gene (EGFR). CONCLUSION These findings shed light on the role of genetic factors in MR. Future studies should adopt a multicentric approach to expand sample sizes and enhance the analysis of genetic variants associated with MR.
Collapse
Affiliation(s)
- Li Yang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weiping Yang
- Department of Orthodontics, The First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Yining Shen
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yu Zhou
- Department of Orthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
3
|
Eubanks E, VanderSleen K, Mody J, Patel N, Sacks B, Farahani MD, Wang J, Elliott J, Jaber N, Akçimen F, Bandres-Ciga S, Helweh F, Liu J, Archakam S, Kimelman R, Sharma B, Socha P, Guntur A, Huang Y, Ramalingam N, Guadagno E, Bartels T, Dettmer U, Mouradian MM, Bahrami AH, Dai W, Baum J, Shi Z, Hardy J, Kara E. Increased burden of rare risk variants across gene expression networks predisposes to sporadic Parkinson's disease. Cell Rep 2025; 44:115636. [PMID: 40317721 DOI: 10.1016/j.celrep.2025.115636] [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: 09/12/2024] [Revised: 02/04/2025] [Accepted: 04/10/2025] [Indexed: 05/07/2025] Open
Abstract
Alpha-synuclein (αSyn) is an intrinsically disordered protein that accumulates in the brains of patients with Parkinson's disease (PD). Through a high-throughput screen, we recently identified 38 genes whose knockdown modulates αSyn propagation. Here, we show that, among those, TAX1BP1 regulates how αSyn interacts with lipids, and ADAMTS19 modulates how αSyn phase separates into inclusions, adding to the growing body of evidence implicating those processes in PD. Through RNA sequencing, we identify several genes that are differentially expressed after knockdown of TAX1BP1 or ADAMTS19 and carry an increased frequency of rare risk variants in patients with PD versus healthy controls. Those differentially expressed genes cluster within modules in regions of the brain that develop high degrees of αSyn pathology. We propose a model for the genetic architecture of sporadic PD: increased burden of risk variants across genetic networks dysregulates pathways underlying αSyn homeostasis and leads to pathology and neurodegeneration.
Collapse
Affiliation(s)
- Elena Eubanks
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Katelyn VanderSleen
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Jiya Mody
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Neha Patel
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Benjamin Sacks
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | | | - Jinying Wang
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Jordan Elliott
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Nora Jaber
- Department of Cell Biology and Neuroscience & Institute for Quantitative Biomedicine, Rutgers University, Piscataway, NJ 08854, USA
| | - Fulya Akçimen
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sara Bandres-Ciga
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Fadel Helweh
- UNAM-National Nanotechnology Research Center and Institute of Materials Science & Nanotechnology, Bilkent University, Ankara 06800, Turkey
| | - Jun Liu
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Sanjana Archakam
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Robert Kimelman
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Bineet Sharma
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Philip Socha
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Ananya Guntur
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Yiming Huang
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Nagendran Ramalingam
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Elyse Guadagno
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Tim Bartels
- UK Dementia Research Institute, University College London, London W1T 7NF, UK
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - M Maral Mouradian
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Amir Houshang Bahrami
- UNAM-National Nanotechnology Research Center and Institute of Materials Science & Nanotechnology, Bilkent University, Ankara 06800, Turkey
| | - Wei Dai
- Department of Cell Biology and Neuroscience & Institute for Quantitative Biomedicine, Rutgers University, Piscataway, NJ 08854, USA
| | - Jean Baum
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Zheng Shi
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - John Hardy
- UK Dementia Research Institute, University College London, London W1T 7NF, UK; Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK; Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK; National Institute for Health Research (NIHR) University College London Hospitals Biomedical Research Centre, London W1T 7DN, UK; Institute for Advanced Study, The Hong Kong University of Science and Technology, SAR, Hong Kong, China
| | - Eleanna Kara
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA.
| |
Collapse
|
4
|
Zhu Z, Liu H, Feng L, Lu L, Zhu J, Liang Q, Lan Z, Ye Y, Wang S, Chen A, Yan J. Loss of ADAMTS5 promotes vascular calcification via versican/integrin β1/FAK signal. Atherosclerosis 2025; 404:119190. [PMID: 40215897 DOI: 10.1016/j.atherosclerosis.2025.119190] [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: 12/02/2024] [Revised: 03/29/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
INTRODUCTION Extracellular matrix (ECM) proteases have been closely linked to the pathogenesis of vascular calcification. A disintegrin and metalloprotease with thrombospondin motifs-5 (ADAMTS5) is an ECM-degrading enzyme involved in ECM remodeling. Versican, a critical ECM component in the arteries, can be proteolytically cleaved by ADAMTS5 and activates integrin β1. However, whether ADAMTS5 is involved in the regulation of the pathogenesis of vascular calcification remains unclear. This study investigates the regulatory role of ADAMTS5 in vascular calcification and its mechanistic link to versican-integrin β1/FAK signaling. METHODS AND RESULTS Western blot, immunofluorescence, and immunohistochemistry analysis revealed that ADAMTS5 expression was significantly downregulated in rat and human vascular smooth muscle cells (VSMCs), as well as in rat and human arteries during vascular calcification. In addition, both pharmacological inhibition of ADAMTS5 and knockdown of ADAMTS5 by siRNA significantly aggravated mineral deposition in rat and human VSMCs under osteogenic conditions. Moreover, adenovirus-mediated ADAMTS5 overexpression markedly attenuated calcification of VSMCs and aortic calcification in rats with chronic kidney disease. Furthermore, inhibition of ADAMTS5 promoted aortic calcification in VitD3-overloaded mice. Mechanistically, overexpression of ADAMTS5 significantly reduced versican protein levels, and inhibited integrin β1 and FAK phosphorylation in rat VSMCs, but increased versikine protein levels. Moreover, either knockdown of versican or pharmacological inhibition of FAK phosphorylation repressed VSMC calcification mediated by loss of ADAMTS5. CONCLUSIONS We have demonstrated for the first time that ADAMTS5 deficiency promotes versican accumulation and activates integrin β1/FAK signaling. These findings suggest ADAMTS5 as a potential therapeutic target for vascular calcification.
Collapse
MESH Headings
- Animals
- Versicans/metabolism
- Humans
- ADAMTS5 Protein/genetics
- ADAMTS5 Protein/metabolism
- ADAMTS5 Protein/deficiency
- Signal Transduction
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Vascular Calcification/enzymology
- Vascular Calcification/pathology
- Vascular Calcification/genetics
- Integrin beta1/metabolism
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Male
- Cells, Cultured
- Disease Models, Animal
- Focal Adhesion Kinase 1/metabolism
- Mice, Inbred C57BL
- Rats, Sprague-Dawley
- Rats
- Phosphorylation
- Mice
- Aortic Diseases/enzymology
- Aortic Diseases/genetics
- Aortic Diseases/pathology
Collapse
Affiliation(s)
- Zhenyu Zhu
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China; Department of Cardiology, Tongde Hospital of Zhejiang Province, PR China
| | - Hao Liu
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, PR China
| | - Liyun Feng
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China
| | - Lihe Lu
- Department of Pathophysiolgy, Zhongshan Medical School, Sun Yat-Sen University, Guangzhou, 510080, PR China
| | - Jiahui Zhu
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China
| | - Qingchun Liang
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510665, PR China
| | - Zirong Lan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China
| | - Yuanzhi Ye
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China
| | - Siyi Wang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China
| | - An Chen
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China.
| | - Jianyun Yan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China.
| |
Collapse
|
5
|
Naidoo P, Naicker T. A Disintegrin and Metalloprotease with Thrombospondin Motif, Member 13, and Von Willebrand Factor in Relation to the Duality of Preeclampsia and HIV Infection. Int J Mol Sci 2025; 26:4103. [PMID: 40362344 PMCID: PMC12071684 DOI: 10.3390/ijms26094103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Normal pregnancy is associated with multiple changes in the coagulation and the fibrinolytic system. In contrast to a non-pregnant state, pregnancy is a hypercoagulable state where the level of VWF increases by 200-375%, affecting coagulation activity. Moreover, in this hypercoagulable state of pregnancy, preeclampsia is exacerbated. ADAMTS13 cleaves the bond between Tyr1605 and Met1606 in the A2 domain of VWF, thereby reducing its molecular weight. A deficiency of ADAMTS13 originates from mutations in gene or autoantibodies formed against the protease, leading to defective enzyme production. Von Willebrand protein is critical for hemostasis and thrombosis, promoting thrombus formation by mediating the adhesion of platelets and aggregation at high shear stress conditions within the vessel wall. Mutations in VWF disrupts multimer assembly, secretion and/or catabolism, thereby influencing bleeding. VWF is the primary regulator of plasma ADAMTS13 levels since even minute amounts of active ADAMTS13 protease have a significant inhibitory effect on inflammation and thrombosis. VWF is released as a result of endothelial activation brought on by HIV infection. The SARS-CoV-2 infection promotes circulating proinflammatory cytokines, increasing endothelial secretion of ultra large VWF that causes an imbalance in VWF/ADAMTS13. Raised VWF levels corresponds with greater platelet adhesiveness, promoting a thrombotic tendency in stenotic vessels, leading to increased shear stress conditions.
Collapse
Affiliation(s)
| | - Thajasvarie Naicker
- Optics & Imaging Centre, Doris Duke Medical Research Institute, University of KwaZulu-Natal, 719 Umbilo Road, Congella, Durban 4013, South Africa
| |
Collapse
|
6
|
Pineda-Suazo D, Guillén-Chable F, Escobedo-Hinojosa WI, Galindo-Sánchez CE, Rosas C. Insights into Octopus maya cathepsins from metatranscriptome and genome: structure evolutionary relationships and functional role prediction in digestive processes. Biol Open 2025; 14:bio061778. [PMID: 40106538 PMCID: PMC12032550 DOI: 10.1242/bio.061778] [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: 10/22/2024] [Accepted: 03/06/2025] [Indexed: 03/22/2025] Open
Abstract
Physiological response to feeding is crucial for various production factors such as feed catabolism and growth. Despite growing significance in red Octopus maya aquaculture, large-scale commercial production is limited by not sufficiently knowing their nutritional needs, especially their digestive physiology. Since this species is carnivorous, one of the main feeding aspects is directed to protein digestion, but its enzymatic digestive repertoire has not been studied yet at genomic and transcriptomic levels. This study searched for protease enzymes encoded in O. maya genome and expressed in the transcriptome, allowing an initial annotation of genes involved in protein catabolism; 117 amino acid sequences related to 'octopus digestive enzymes' were retrieved from 66 available-species' genomes in the NCBI database, coding for cathepsins, papilins, and metalloproteases. Homology analysis identified 36 homologous sequences from O. maya transcriptome and three from its genome. Phylogenetic analysis grouped 37 of 39 sequences into 11 of 14 main clades, offering new insights into the evolutionary relationships and functional roles of these proteases. Phylogenetic and motif analyses resulted in selecting 19 amino acid O. maya sequences using multiple sequence alignment that were used to generate three-dimensional protein models. The obtained models revealed a diverse structural architecture among 16 modelled cathepsins; however, their catalytic potential to fully clarify their role in protein hydrolysis and cellular processes remains to be determined. Foundational data provides insights into biochemistry and physiology behind O. maya protein digestion. Further complementation of these results with enzymatic characterization of the identified proteases should allow for improved diet formulation in order to foster this species aquaculture.
Collapse
Affiliation(s)
- Daisy Pineda-Suazo
- Unidad Multidisciplinaria de Docencia e Investigación, Facultad de Ciencias UNAM, Puerto de abrigo s/n Sisal, Mpio, Hunucmá, Yucatán 97356, México
| | - Francisco Guillén-Chable
- Unidad Multidisciplinaria de Docencia e Investigación, Facultad de Ciencias UNAM, Puerto de abrigo s/n Sisal, Mpio, Hunucmá, Yucatán 97356, México
| | - Wendy Itzel Escobedo-Hinojosa
- Unidad de Química en Sisal, Facultad de Química, Universidad Nacional Autónoma de México, Puerto de abrigo s/n, Sisal, Yucatán 97356, México
| | - Clara E. Galindo-Sánchez
- Departamento de Biotecnología Marina, Laboratorio de Genómica Funcional, CICESE, Ensenada, Baja California 22860, México
| | - Carlos Rosas
- Unidad Multidisciplinaria de Docencia e Investigación, Facultad de Ciencias UNAM, Puerto de abrigo s/n Sisal, Mpio, Hunucmá, Yucatán 97356, México
| |
Collapse
|
7
|
Al-Awadhi A, Alwehaidah MS, Al-Sabaan K, Al-Ajmi N. Assessment of ferritin and hepcidin levels in splenectomised and non-splenectomised β-thalassemia major patients and exploring a potential correlation with von Willebrand factor and ADAMTS-13. Ann Hematol 2025; 104:2229-2238. [PMID: 40232405 PMCID: PMC12053081 DOI: 10.1007/s00277-025-06360-8] [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: 12/05/2024] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
Iron overload is a major complication in β-thalassemia major (β-TM) patients, resulting from ineffective erythropoiesis, increased gastrointestinal iron absorption and multiple blood transfusions. Excess iron accumulates in various organs, leading to organ dysfunction, and increased risk of thrombotic events. In this study we aim to determine levels of ferritin and its regulation hormone hepcidin in multi-transfused splenectomised and non-splenectomised β-thalassemia major patients and assess a possible correlation with the coagulation protein von Willebrand factor (vWF) and its cleaving protease ADAMTS-13. The study was conducted on 80 β-thalassemia major patients and 80 age- and sex-matched healthy controls. Plasma levels of vWF, ADAMTS-13, and hepcidin were assessed using the ELISA method. All patients presented with significantly higher levels of ferritin compared to normal controls (p < 0.001), while hepcidin levels were barely higher in patients (p = 0.05). Ferritin had a positive correlation with vWF antigen levels (r = 0.222, p = 0.05), ADAMTS-13 antigen levels (r = 0.334, p = 0.002) and ADAMTS-13 activity levels (r = 0.353, p = 0.001) in patients. Splenectomised patients had significantly higher levels of white blood cell counts, platelet counts and vWF antigen levels compared to non-splenectomised patients (p < 0.05), but ferritin and hepcidin levels were comparable between the two groups (p > 0.05). Hepcidin was not found to be correlated with any of the measured parameters in patients (p > 0.05). Iron overload is well manifested in our study group despite continuous chelation therapy. Unlike hepcidin, ferritin appeared to be associated with increased secretion of vWF and ADAMTS-13 in patients, while splenectomy had no effect on ferritin or hepcidin levels. These findings highlight the importance of proper iron monitoring in β-TM and recognition of thrombotic risks in managing this anemia.
Collapse
Affiliation(s)
- Anwar Al-Awadhi
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center, Kuwait University, Jabriya, Kuwait.
| | - Materah Salem Alwehaidah
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center, Kuwait University, Jabriya, Kuwait
| | - Kefayah Al-Sabaan
- Department of Hematology, Al-Farwaniya Hospital, Ministry of Health, Farwaniya, Kuwait
| | - Nouf Al-Ajmi
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center, Kuwait University, Jabriya, Kuwait
| |
Collapse
|
8
|
Bitsadze V, Lazarchuk A, Vorobev A, Khizroeva J, Tretyakova M, Makatsariya N, Gashimova N, Grigoreva K, Tatarintseva A, Karpova A, Mostovoi A, Zainulina M, Kapanadze D, Blbulyan A, Kuneshko N, Gris JC, Elalamy I, Gerotziafas G, Makatsariya A. Systemic Inflammatory Response Syndrome, Thromboinflammation, and Septic Shock in Fetuses and Neonates. Int J Mol Sci 2025; 26:3259. [PMID: 40244141 PMCID: PMC11989690 DOI: 10.3390/ijms26073259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
This article explores systemic inflammatory response syndrome (SIRS), thromboinflammation, and septic shock in fetuses and neonates, offering a comprehensive examination of their pathophysiology, diagnostic criteria, and clinical implications. It identifies SIRS as an exaggerated response to external stress, disrupting the balance between inflammation and adaptive mechanisms, driven by cytokines such as TNF-α and IL-1. The fetal inflammatory response syndrome (FIRS), a subset of SIRS, is noted for its role in adverse neonatal outcomes, including organ damage, inflammation, and long-term developmental disorders. The article discusses the extensive effects of FIRS on critical systems, including the blood, lungs, central nervous system, and kidneys. It highlights the challenges in diagnosing and managing septic shock in neonates, focusing on the relationship between inflammation and the hemostatic system. Additionally, the paper points out recent advancements, such as the convergent model of coagulation and emerging biomarkers like microRNAs for early detection. Despite this progress, gaps remain in understanding the molecular mechanisms underlying these conditions and in developing effective therapeutic strategies. This highlights the necessity for targeted research to mitigate the morbidity and mortality associated with septic shock in neonates.
Collapse
Affiliation(s)
- Victoria Bitsadze
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Arina Lazarchuk
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Alexander Vorobev
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Jamilya Khizroeva
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Maria Tretyakova
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Natalia Makatsariya
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Nilufar Gashimova
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Kristina Grigoreva
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Alena Tatarintseva
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| | - Anna Karpova
- Vorokhobov City Clinical Hospital No 67, Moscow Healthcare Department, 2/44 Salyama Adilya Str., Moscow 123423, Russia; (A.K.); (A.M.)
- Russian Medical Academy of Continuous Professional Education, Health Ministry of Russian Federation, 2/1 bldg. 1, Barrikadnaya Str., Moscow 123993, Russia
- Department of Polyclinic Therapy, Clinical Laboratory Diagnostics and Medical Biochemistry of Institute of Postgraduate Education of Yaroslavl State Medical University, Yaroslavl State Medical University, Health Ministry of Russian Federation, 5 Revolutsionnaya Str., Yaroslavl 150000, Russia
| | - Aleksei Mostovoi
- Vorokhobov City Clinical Hospital No 67, Moscow Healthcare Department, 2/44 Salyama Adilya Str., Moscow 123423, Russia; (A.K.); (A.M.)
- Russian Medical Academy of Continuous Professional Education, Health Ministry of Russian Federation, 2/1 bldg. 1, Barrikadnaya Str., Moscow 123993, Russia
- Department of Polyclinic Therapy, Clinical Laboratory Diagnostics and Medical Biochemistry of Institute of Postgraduate Education of Yaroslavl State Medical University, Yaroslavl State Medical University, Health Ministry of Russian Federation, 5 Revolutsionnaya Str., Yaroslavl 150000, Russia
| | - Marina Zainulina
- Snegirev Maternity Hospital No 6, 5 Mayakovskogo Str., Saint Petersburg 192014, Russia;
- Department of Obstetrics, Gynecology and Reproductology of Pavlov First Saint Petersburg State Medical University, Pavlov First Saint Petersburg State Medical University, Health Ministry of Russian Federation, 6/8 Lev Tolstoy Str., Saint Petersburg 197022, Russia
| | - Daredzhan Kapanadze
- Center of Pathology of Pregnancy and Hemostasis «Medlabi», 340112 Tbilisi, Georgia;
| | - Armen Blbulyan
- Research Center of Maternal and Child Health Protection, 22 Mashtots Avenue, Yerevan 0002, Armenia;
| | - Nart Kuneshko
- Moscow’s Region Odintsovo Maternity Hospital, Odintsovo 143003, Russia;
| | - Jean-Christophe Gris
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
- Faculty of Pharmaceutical and Biological Sciences, Montpellier University, 34093 Montpellier, France
| | - Ismail Elalamy
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
- Faculté Privee de Médecine de Marrakech (FPMM), Route Amizmiz, Marrakech 42312, Morocco
- Hopital Americain de Paris, 55 rue du Château, Neuilly Sur Seine, 92200 Paris, France
| | - Grigoris Gerotziafas
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
- INSERM UMR_S_938, Saint-Antoine Research Center (CRSA), Team “Cancer Biology and Therapeutics”, Group “Cancer—Angiogenesis—Thrombosis”, University Institute of Cancerology (UIC), Sorbonne University, 34 Rue du Crozatier, 75012 Paris, France
- Thrombosis Center, Tenon—Saint Antoine University Hospital, Hôpitaux Universitaires Est Parisien, Assitance Publique Hôpitaix de Paris (AP-HP), 4 Rue de la Chine, 75020 Paris, France
| | - Alexander Makatsariya
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, Moscow 119435, Russia; (V.B.); (A.L.); (A.V.); (M.T.); (N.M.); (N.G.); (K.G.); (A.T.); (J.-C.G.); (I.E.); (G.G.); (A.M.)
| |
Collapse
|
9
|
Van der Stede T, Van de Loock A, Turiel G, Hansen C, Tamariz-Ellemann A, Ullrich M, Lievens E, Spaas J, Yigit N, Anckaert J, Nuytens J, De Baere S, Van Thienen R, Weyns A, De Wilde L, Van Eenoo P, Croubels S, Halliwill JR, Mestdagh P, Richter EA, Gliemann L, Hellsten Y, Vandesompele J, De Bock K, Derave W. Cellular deconstruction of the human skeletal muscle microenvironment identifies an exercise-induced histaminergic crosstalk. Cell Metab 2025; 37:842-856.e7. [PMID: 39919738 DOI: 10.1016/j.cmet.2024.12.011] [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: 02/11/2024] [Revised: 10/14/2024] [Accepted: 12/18/2024] [Indexed: 02/09/2025]
Abstract
Plasticity of skeletal muscle is induced by transcriptional and translational events in response to exercise, leading to multiple health and performance benefits. The skeletal muscle microenvironment harbors myofibers and mononuclear cells, but the rich cell diversity has been largely ignored in relation to exercise adaptations. Using our workflow of transcriptome profiling of individual myofibers, we observed that their exercise-induced transcriptional response was surprisingly modest compared with the bulk muscle tissue response. Through the integration of single-cell data, we identified a small mast cell population likely responsible for histamine secretion during exercise and for targeting myeloid and vascular cells rather than myofibers. We demonstrated through histamine H1 or H2 receptor blockade in humans that this paracrine histamine signaling cascade drives muscle glycogen resynthesis and coordinates the transcriptional exercise response. Altogether, our cellular deconstruction of the human skeletal muscle microenvironment uncovers a histamine-driven intercellular communication network steering muscle recovery and adaptation to exercise.
Collapse
Affiliation(s)
- Thibaux Van der Stede
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium; Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Alexia Van de Loock
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium
| | - Guillermo Turiel
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Camilla Hansen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | | | - Max Ullrich
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium
| | - Eline Lievens
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium
| | - Jan Spaas
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium; BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Nurten Yigit
- OncoRNALab, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jasper Anckaert
- OncoRNALab, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Justine Nuytens
- OncoRNALab, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Siegrid De Baere
- Laboratory of Pharmacology and Toxicology, Department of Pathobiology, Pharmacology and Zoological Medicine, Ghent University, Merelbeke, Belgium
| | - Ruud Van Thienen
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium
| | - Anneleen Weyns
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium
| | - Laurie De Wilde
- Department of Diagnostic Sciences, Doping Control Laboratory, Ghent University, Ghent, Belgium
| | - Peter Van Eenoo
- Department of Diagnostic Sciences, Doping Control Laboratory, Ghent University, Ghent, Belgium
| | - Siska Croubels
- Laboratory of Pharmacology and Toxicology, Department of Pathobiology, Pharmacology and Zoological Medicine, Ghent University, Merelbeke, Belgium
| | - John R Halliwill
- Bowerman Sports Science Center, Department of Human Physiology, University of Oregon, Eugene, OR, USA
| | - Pieter Mestdagh
- OncoRNALab, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Erik A Richter
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Lasse Gliemann
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Ylva Hellsten
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jo Vandesompele
- OncoRNALab, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Wim Derave
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium.
| |
Collapse
|
10
|
Yuan S, Bacchetti R, Adams J, Cuffaro D, Rossello A, Nuti E, Santamaria S, Rainero E. The protease ADAMTS5 controls ovarian cancer cell invasion, downstream of Rab25. FEBS J 2025. [PMID: 40164572 DOI: 10.1111/febs.70080] [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: 09/26/2024] [Revised: 12/18/2024] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Ovarian cancer is the 3rd most common gynaecological malignancy worldwide, with a 5-year survival rate of < 30% in the presence of metastasis. Metastatic progression is characterised by extensive remodelling of the extracellular matrix, primarily mediated by secreted proteases, including members of the 'a disintegrin and metalloprotease with thrombospondin motif' (ADAMTS) family. In particular, ADAMTS5 has been reported to be upregulated in ovarian malignant tumours compared to borderline and benign lesions, suggesting it might play a role in metastatic progression. Furthermore, it has been suggested that Rab25, a small GTPase of the Ras family, might upregulate ADAMTS5 expression in ovarian cancer cells. Here we demonstrated that Rab25 promotes ADAMTS5 expression through the activation of the nuclear factor κB (NF-κB) signalling pathway. Furthermore, ADAMTS5 was necessary and sufficient to stimulate ovarian cancer cell migration through complex fibroblast-secreted matrices, while selective ADAMTS5 inhibition prevented ovarian cancer spheroid invasion in 3D systems. Finally, in ovarian cancer patients, high ADAMTS5 expression correlated with poor prognosis. Altogether, these data identify ADAMTS5 as a novel regulator of ovarian cancer cell migration and invasion, suggesting it might represent a previously undescribed therapeutic target to prevent ovarian cancer metastasis.
Collapse
Affiliation(s)
| | | | - Jamie Adams
- School of Biosciences, University of Sheffield, UK
- Clinical Medicine, School of Medicine & Population Health, University of Sheffield, UK
| | | | | | - Elisa Nuti
- Department of Pharmacy, University of Pisa, Italy
| | - Salvatore Santamaria
- Department of Biochemical Sciences, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | | |
Collapse
|
11
|
Hao H, Eberand BM, Larance M, Haltiwanger RS. Protein O-Fucosyltransferases: Biological Functions and Molecular Mechanisms in Mammals. Molecules 2025; 30:1470. [PMID: 40286076 PMCID: PMC11990869 DOI: 10.3390/molecules30071470] [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: 02/22/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Domain-specific O-fucosylation is an unusual type of glycosylation, where the fucose is directly attached to the serine or threonine residues in specific protein domains via an O-linkage. O-fucosylated proteins play critical roles in a wide variety of biological events and hold important therapeutic values, with the most studied being the Notch receptors and ADAMTS proteins. O-fucose glycans modulate the function of the proteins they modify and are closely associated with various diseases including cancer. In mammals, alongside the well-documented protein O-fucosyltransferase (POFUT) 1-mediated O-fucosylation of epidermal growth factor-like (EGF) repeats and POFUT2-mediated O-fucosylation of thrombospondin type 1 repeats (TSRs), a new type of O-fucosylation was recently identified on elastin microfibril interface (EMI) domains, mediated by POFUT3 and POFUT4 (formerly FUT10 and FUT11). In this review, we present an overview of our current knowledge of O-fucosylation, integrating the latest findings and with a particular focus on its biological functions and molecular mechanisms.
Collapse
Affiliation(s)
- Huilin Hao
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30605, USA;
| | - Benjamin M. Eberand
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; (B.M.E.); (M.L.)
| | - Mark Larance
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; (B.M.E.); (M.L.)
| | | |
Collapse
|
12
|
Divakar A, Kandasamy N, Varghese RM, Jeyachandran S, Thomas LR. Unlocking ADAMTS-5: In Silico insights into TMJ proteomics and docking dynamics. J Orthod Sci 2025; 14:11. [PMID: 40302877 PMCID: PMC12036764 DOI: 10.4103/jos.jos_89_24] [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: 08/13/2024] [Revised: 11/18/2024] [Accepted: 12/13/2024] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Temporomandibular joint (TMJ) disorder refers to a condition involving dysfunction or pain in the jaw joint and the muscles that control jaw movement. It can affect one or both sides of the jaw and can cause various symptoms, including Jaw pain or tenderness; Difficulty or discomfort when chewing; Clicking, popping, or grating sounds in the jaw joint; Jaw locking or limited movement; Earache or pain around the ear; Headaches or migraines; Neck and shoulder pain; Swelling on the side of the face. TMJ disorder can have various causes, including injury to the jaw joint, teeth grinding or clenching (bruxism), arthritis, stress, misalignment of the jaw or teeth, and excessive gum chewing. Computer-aided drug design (CADD) comprises a range of theoretical and computational strategies employed in contemporary drug discovery. Molecular docking stands out as a key technique within CADD, aiding in the comprehension of drug-molecule interactions for rational drug design, mechanistic investigations, and the creation of stable complexes with heightened specificity and potential effectiveness. Through the docking process, valuable information regarding binding energy, free energy, and predictions of complex stability is obtained, offering significant insights into drug development endeavors. AIM The objective of this research was to employ docking methodology to identify potential ADAMTS-5 protein for TMJ. Four ADAMTS-5 protein inhibitors previously reported in the literature were selected, and their compound structures were obtained from the Zinc15 database. The ADAMTS-5 protein was designated as the target and optimized utilizing the RCSB Protein Data Bank. Following pharmacophore modeling, 20 novel compounds were identified, and SwissDock was utilized to dock these compounds with the target protein. A comparison was made between the binding energies of the newly discovered compounds and those of previously published molecules with the target. RESULTS The results indicated that among the 20 ZINC1846088 and ZINC33606904 exhibited the highest binding energy and displayed superior properties compared to the other molecules. CONCLUSION The study concluded that ZINC1846088 and ZINC33606904 exhibited greater binding affinity than the reported inhibitors of ADAMTS-5 protein. Therefore, these two molecules can be used as a potential and promising lead for the treatment of TMJ and could be employed in targeted drug therapy. CATEGORIES Dentistry, TMJ.
Collapse
Affiliation(s)
- Anjusha Divakar
- Department of Orthodontics and Dentofacial Orthopaedics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Nagachandran Kandasamy
- Department of Orthodontics and Dentofacial Orthopaedics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Remmiya Mary Varghese
- Department of Orthodontics and Dentofacial Orthopaedics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Sivakamavalli Jeyachandran
- Department of Orthodontics and Dentofacial Orthopaedics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Lincy Rachel Thomas
- Department of Orthodontics and Dentofacial Orthopaedics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| |
Collapse
|
13
|
Han Y, Wang G, Han E, Yang S, Zhao R, Lan Y, Zhao M, Li Y, Ren L. SERPINI1 serves as a biomarker promoting cell proliferation and invasion in hepatocellular carcinoma. Cancer Cell Int 2025; 25:88. [PMID: 40082896 PMCID: PMC11908049 DOI: 10.1186/s12935-025-03716-y] [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: 09/23/2024] [Accepted: 02/25/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND SERPINI1 is a protein-coding gene, which has been reported to be related to malignancies, and the encoding protein is a secreted protein. Nevertheless, the specific effect of SERPINI1 on Hepatocellular carcinoma (HCC) remains unclear. METHODS The expression level of SERPINI1 in cancers was detected by the Gene Expression Omnibus (GEO) database, the Gene Expression Profiling Interactive Analysis (GEPIA) database and the collected serum of HCC patients. The receiver operating characteristic (ROC) curve and area under curve (AUC) were used to evaluate the diagnostic effectiveness of serum SERPINI1 and the combination of AFP and SERPINI1 for HCC. The Kaplan-Meier (KM) survival was used to evaluate the prognostic capacity of SERPINI1 for HCC in GEPIA database. Furthermore, the correlations between clinicopathological characteristics and the level of serum SERPINI1 were analyzed. Besides, we detected the expression of SERPINI1 in HepG2 by qPCR and western blot, and confirmed the biological function of SERPINI1 through MTT, EdU, wound healing and transwell invasion assay. RESULTS The results indicated that the level of SERPINI1 was significantly increased in tissue and serum of HCC patients. ROC analysis displayed that SERPINI1 had a significantly diagnostic value for HCC, the combination of AFP and SERPINI1 gained the higher specificity and sensitivity. The KM survival curves indicated that patients with SERPINI1 overexpression had worse overall survival. Furthermore, we found the positive correlations between serum SERPINI1 level and some clinicopathological characteristics, such as tumor size, differentiation degrees and so on. In addition, in vitro experiments revealed that SERPINI1 could promote the proliferation and invasion of HCC. CONCLUSIONS Taken together, our study demonstrates that SERPINI1, which is highly expressed in HCC and closely related to cell proliferation and invasion, may serve as a novel biomarker for diagnosis and prognosis of HCC.
Collapse
Affiliation(s)
- Yawei Han
- Department of Laboratory, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Huanhuxi Road, Hexi District, Tianjin, 300060, PR China
| | - Gaoyv Wang
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin, China
| | - Erwei Han
- Severe Medical Department, Gaocheng People's Hospital, Shijiazhuang City, Hebei Province, China
| | - Shuting Yang
- Department of Laboratory, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Huanhuxi Road, Hexi District, Tianjin, 300060, PR China
| | - Ran Zhao
- Department of Laboratory, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Huanhuxi Road, Hexi District, Tianjin, 300060, PR China
| | - Yvying Lan
- Department of Laboratory, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Huanhuxi Road, Hexi District, Tianjin, 300060, PR China
- Clinical Medical College, Tianjin Medical University, Tianjin, China
| | - Meng Zhao
- Department of Laboratory, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Huanhuxi Road, Hexi District, Tianjin, 300060, PR China.
| | - Yueguo Li
- Department of Laboratory, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Huanhuxi Road, Hexi District, Tianjin, 300060, PR China.
| | - Li Ren
- Department of Laboratory, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Huanhuxi Road, Hexi District, Tianjin, 300060, PR China.
| |
Collapse
|
14
|
Wang M, Liu H, Zhang M, Niu X, Sun M, Wang F, Ni Y, Hong T, Zhang W, Dang S. The secreted protease ADAMTS18 links early isoform transformation and maturation of glomerular basement membrane macromolecules to the integrity of the glomerular filtration barrier. Biochem Biophys Res Commun 2025; 750:151386. [PMID: 39879696 DOI: 10.1016/j.bbrc.2025.151386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
The glomerular filtration barrier (GFB) has a unique spatial structure, including porous capillary endothelial cells, glomerular basal membrane (GBM) and highly specialized podocytes. This special structure is essential for the hemofiltration process of nephrons. GBM is the central meshwork structure of GFB formed by the assembly and fusion of various extracellular matrix (ECM) macromolecules, such as laminins and collagens, which undergo isoform transformation and maturation that may require precise regulation by metalloproteinases. However, the role of metalloproteinase in GFB integrity remains elusive. A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTSs) gene family members are known for their roles in ECM remodeling. In this study, we found that ADAMTS18 was secreted by capillary endothelial cell within the glomeruli of human fetal kidney and mouse kidney. Adamts18 knockout (Adamts18-/-) mice exhibited early proteinuria with GFB dysplasia, including podocyte invagination surrounded by glomerular capillary network and microvillus of podocytes. Mechanistically, ADAMTS18 regulated the isoform transformation and maturation of GBM macromolecules. The levels of mature LAMA5 isoform and fibronectin were significantly lower in Adamts18-/- glomeruli than in Adamts18+/+ glomeruli. Co-immunoprecipitation (IP) results showed that the LAMA5 fragment (5XAU) was a novel interacting protein of ADAMTS18 and could be pulled down by ADAMTS18. These new findings shed light on the biological role of metalloproteinase in GFB integrity and related kidney diseases.
Collapse
Affiliation(s)
- Min Wang
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Science, East China Normal University, China
| | - Hanlin Liu
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Science, East China Normal University, China
| | - Mengxi Zhang
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Science, East China Normal University, China
| | - Xiaohan Niu
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Science, East China Normal University, China
| | - Min Sun
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Science, East China Normal University, China
| | - Fang Wang
- Core Facility of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingyin Ni
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Hong
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Science, East China Normal University, China
| | - Wei Zhang
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Science, East China Normal University, China.
| | - Suying Dang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
15
|
Auer S, Schicht M, Hoffmann L, Budday S, Frischknecht R, Blümcke I, Paulsen F. The Role of Perineuronal Nets in Physiology and Disease: Insights from Recent Studies. Cells 2025; 14:321. [PMID: 40072050 PMCID: PMC11898492 DOI: 10.3390/cells14050321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 03/15/2025] Open
Abstract
Perineuronal nets (PNNs) are specialized extracellular matrix structures that predominantly surround inhibitory neurons in the central nervous system (CNS). They have been identified as crucial regulators of synaptic plasticity and neuronal excitability. This literature review aims to summarize the current state of knowledge about PNNs, their molecular composition and structure, as well as their functional roles and involvement in neurological diseases. Furthermore, future directions in PNN research are proposed, and the therapeutic potential of targeting PNNs to develop novel treatment options for various neurological disorders is explored. This review emphasizes the importance of PNNs in CNS physiology and pathology and underscores the need for further research in this area.
Collapse
Affiliation(s)
- Sophia Auer
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Martin Schicht
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Lucas Hoffmann
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Partner of the European Reference Network (ERN) EpiCARE, 91054 Erlangen, Germany; (L.H.); (I.B.)
| | - Silvia Budday
- Institute of Continuum Mechanics and Biomechanics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany;
| | - Renato Frischknecht
- Department of Biology, Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany;
| | - Ingmar Blümcke
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Partner of the European Reference Network (ERN) EpiCARE, 91054 Erlangen, Germany; (L.H.); (I.B.)
| | - Friedrich Paulsen
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
| |
Collapse
|
16
|
NADOUR M, LEATIS RIVALETTEREVENO, BIARD M, FRÉBAULT N, RIVOLLET L, ST-LOUIS P, BLANCHETTE CR, THACKERAY A, PERRAT P, BEVILACQUA C, PREVEDEL R, CAPPADOCIA L, RAPTI G, DOITSIDOU M, BÉNARD CY. Remodeling of extracellular matrix collagen IV by MIG-6/papilin regulates neuronal architecture. RESEARCH SQUARE 2025:rs.3.rs-5962240. [PMID: 39989960 PMCID: PMC11844652 DOI: 10.21203/rs.3.rs-5962240/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Neuronal architecture established embryonically must persist lifelong to ensure normal brain function. However, little is understood about the mechanisms behind the long-term maintenance of neuronal organization. To uncover maintenance mechanisms, we performed a suppressor screen in sax-7/L1CAM mutants, which exhibit progressive disorganization with age. We identified the conserved extracellular matrix protein MIG-6/papilin as a key regulator of neuronal maintenance. Combining incisive molecular genetics, structural predictions, in vivo quantitative imaging, and cutting-edge Brillouin microscopy, we show that MIG-6/papilin remodels extracellular matrix collagen IV, working in concert with the secreted enzymes MIG-17/ADAMTS and PXN-2/peroxidasin. This remodeling impacts tissue biomechanics and ensures neuronal stability, even under increased mechanical stress. Our findings highlight an extracellular mechanism by which MIG-6/papilin supports the integrity of neuronal architecture throughout life. This work provides critical insights into the molecular basis of sustaining neuronal architecture and offers a foundation for understanding age-related and neurodegenerative disorders.
Collapse
Affiliation(s)
- Malika NADOUR
- Université du Québec à Montréal, Department of Biological Sciences, Montreal, QC, Canada
- Centre d’Excellence en Recherche sur les Maladies Orphelines – Fondation Courtois (CERMO-FC Research Center), Université du Québec à Montréal, Montreal, QC, Canada
| | - Robert I. VALETTE REVENO LEATIS
- Université du Québec à Montréal, Department of Biological Sciences, Montreal, QC, Canada
- Centre d’Excellence en Recherche sur les Maladies Orphelines – Fondation Courtois (CERMO-FC Research Center), Université du Québec à Montréal, Montreal, QC, Canada
| | - Marie BIARD
- Université du Québec à Montréal, Department of Biological Sciences, Montreal, QC, Canada
- Centre d’Excellence en Recherche sur les Maladies Orphelines – Fondation Courtois (CERMO-FC Research Center), Université du Québec à Montréal, Montreal, QC, Canada
| | - Noémie FRÉBAULT
- Université du Québec à Montréal, Department of Biological Sciences, Montreal, QC, Canada
- Centre d’Excellence en Recherche sur les Maladies Orphelines – Fondation Courtois (CERMO-FC Research Center), Université du Québec à Montréal, Montreal, QC, Canada
| | - Lise RIVOLLET
- Université du Québec à Montréal, Department of Biological Sciences, Montreal, QC, Canada
- Centre d’Excellence en Recherche sur les Maladies Orphelines – Fondation Courtois (CERMO-FC Research Center), Université du Québec à Montréal, Montreal, QC, Canada
| | - Philippe ST-LOUIS
- Université du Québec à Montréal, Department of Biological Sciences, Montreal, QC, Canada
- Centre d’Excellence en Recherche sur les Maladies Orphelines – Fondation Courtois (CERMO-FC Research Center), Université du Québec à Montréal, Montreal, QC, Canada
| | | | - Andrea THACKERAY
- University of Massachusetts Chan Medical School, Department of Neurobiology, MA, USA
| | - Paola PERRAT
- University of Massachusetts Chan Medical School, Department of Neurobiology, MA, USA
| | - Carlo BEVILACQUA
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Robert PREVEDEL
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Epigenetics and Neurobiology Unit, European Molecular Biology Laboratory, Rome, Italy
- Interdisciplinary Center of Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Laurent CAPPADOCIA
- Centre d’Excellence en Recherche sur les Maladies Orphelines – Fondation Courtois (CERMO-FC Research Center), Université du Québec à Montréal, Montreal, QC, Canada
- Université du Québec à Montréal, Department of Chemistry, Montreal, QC, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), QC, Canada
| | - Georgia RAPTI
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Epigenetics and Neurobiology Unit, European Molecular Biology Laboratory, Rome, Italy
- Interdisciplinary Center of Neurosciences, Heidelberg University, Heidelberg, Germany
- FENS-KAVLI Network of Excellence, Brussels, Belgium
| | - Maria DOITSIDOU
- University of Edinburgh, Centre for Discovery Brain Sciences, Edinburgh, Scotland
| | - Claire Y. BÉNARD
- Université du Québec à Montréal, Department of Biological Sciences, Montreal, QC, Canada
- Centre d’Excellence en Recherche sur les Maladies Orphelines – Fondation Courtois (CERMO-FC Research Center), Université du Québec à Montréal, Montreal, QC, Canada
- University of Massachusetts Chan Medical School, Department of Neurobiology, MA, USA
| |
Collapse
|
17
|
Long D, Zhan J, Tu C, Yu S, Rao J, Luo Q, Huang Z, Li J. Analysis of expression and its clinical significance of the ADAMTS-2 in systemic lupus erythematosus. Clin Rheumatol 2025; 44:681-691. [PMID: 39806074 DOI: 10.1007/s10067-025-07303-4] [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: 09/04/2024] [Revised: 12/05/2024] [Accepted: 12/26/2024] [Indexed: 01/16/2025]
Abstract
OBJECTIVES ADAMTS-2 is a procollagen N-proteinase that plays an important role in inflammation regulation. The objective of our research is to explore the expression of ADAMTS-2 in Systemic Lupus Erythematosus (SLE), and analyze its relationship with clinical features of SLE, and evaluate the potential value of ADAMTS-2 as a diagnostic biomarker in SLE. METHODS ADAMTS-2 expression in PBMCs was detected by RT-qPCR in SLE patients, RA patients, and healthy controls (HC). The diagnostic value of ADAMTS-2 for SLE was evaluated by ROC curve, and the correlation between ADAMTS-2 and the clinical characteristics of SLE was analyzed by Spearman's rank correlation coefficient. The expression profiles of GSE8650 and GSE82221 were downloaded from the GEO database. We performed GSEA to further understand the functions of ADAMTS-2 in SLE. CIBERSORT was utilized for immune cell infiltration analysis. RESULTS RT-qPCR results validated that the expression of ADAMTS-2 in PBMCs was significantly increased in SLE patients than RA patients and HC. ROC anaylsis suggested that ADAMTS-2 has significant value in distinguishing new-onset SLE patients from RA patients and HC (AUC = 0.805, p < 0.0001). The expression of ADAMTS-2 was negatively correlated with C3, WBC, PLT, neutrophil, and monocyte level. PBMCs samples with high ADAMTS-2 expression were enriched in TNFA_SIGNALING_VIA_NFKB pathway. We found that ADAMTS-2 was positively correlated with neutrophils, M0 macrophages and M2 macrophages. CONCLUSION ADAMTS-2 may be a potential biomarker of SLE patients and closely related to the occurrence and development of SLE. ADAMTS-2 is expected to be a new target for SLE treatment. Key Points • ADAMTS-2 is a potential biomarker of disease activity in SLE patients that develop a flare. • Samples with high ADAMTS-2 expression are enriched in TNFA_SIGNALING_VIA_NFKB pathway in SLE. • ADAMTS-2 expression is positively correlated with neutrophils, M0 macrophages and M2 macrophages.
Collapse
Affiliation(s)
- Dan Long
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, 330000, Jiangxi, China
| | - Jiahuan Zhan
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, 330000, Jiangxi, China
| | - Chaofei Tu
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, 330000, Jiangxi, China
| | - Shujiao Yu
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, 330000, Jiangxi, China
| | - Jiayue Rao
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, 330000, Jiangxi, China
| | - Qing Luo
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, 330000, Jiangxi, China
| | - Zikun Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, 330000, Jiangxi, China
| | - Junming Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, 330000, Jiangxi, China.
| |
Collapse
|
18
|
Xiao JH, Xu LZ, Ning JZ, Cheng F. Unveiling ADAMTS12: A key driver of bladder cancer progression via COL3A1-Mediated activation of the FAK/PI3K/AKT signaling pathway. J Biol Chem 2025; 301:108155. [PMID: 39761856 PMCID: PMC11795591 DOI: 10.1016/j.jbc.2025.108155] [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: 08/21/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/26/2025] Open
Abstract
Bladder cancer (BCa) is a common and lethal disease characterized by high recurrence rates and limited treatment options. Understanding the molecular pathways of BCa progress is crucial for investigating more effective targeted therapies. While ADAMTS12 is known to contribute to cancer progression and treatment resistance, its prognostic significance and underlying mechanisms in BCa remain poorly understood. To elucidate the molecular pathways and functions of ADAMTS12 in BCa, we employed various experimental approaches, including Transwell invasion assays, flow cytometry analysis, wound-healing assays, CCK-8 assays, and a xenograft tumor model. Our results demonstrated that overexpression of ADAMTS12 significantly enhanced cell growth, migration, and invasion while inhibiting apoptosis through the activation of the FAK/PI3K/AKT signaling pathway. Conversely, the knockdown of ADAMTS12 produced the opposite effects. In vivo studies further confirmed that the inhibition of ADAMTS12 effectively suppressed tumor progression. Comprehensive bioinformatics analysis of the TCGA-BLCA dataset and protein-protein interaction networks revealed a strong positive correlation between COL3A1 and ADAMTS12, identifying COL3A1 as a potential downstream target of ADAMTS12. Additionally, we observed a significant increase in the expression levels of ADAMTS12 and COL3A1 in BCa tissues compared to healthy tissues, as confirmed by Western blotting and qRT-PCR analysis. Notably, inhibition of COL3A1 reversed the enhanced cell growth and invasion associated with ADAMTS12 overexpression and suppressed cell apoptosis. Our findings suggest that ADAMTS12 promotes BCa progression through the FAK/PI3K/AKT signaling pathway by regulating COL3A1, highlighting its potential as a valuable marker for diagnosis and prognosis in BCa.
Collapse
Affiliation(s)
- Jian-Hua Xiao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, P.R. China; Department of Urology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, Hubei Province, P.R. China
| | - Li-Zhe Xu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, P.R. China
| | - Jin-Zhuo Ning
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, P.R. China.
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, P.R. China.
| |
Collapse
|
19
|
Kustiati U, Nugrahaningsih DAA, Kusindarta DL, Wihadmadyatami H. Lung cancer: Animal model of lung cancer, molecular carcinogenesis of lung cancer, and antitumor effect of Ocimum sanctum against lung cancer. Open Vet J 2025; 15:482-503. [PMID: 40201854 PMCID: PMC11974298 DOI: 10.5455/ovj.2025.v15.i2.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 12/03/2024] [Indexed: 04/10/2025] Open
Abstract
Lung cancer is the leading cause of fatalities related to cancer globally. There are numerous ways to treat lung cancer, including surgery, chemotherapy, and radiation. Since these treatments have not yet shown satisfactory results, more research into the underlying mechanisms and different approaches to therapy and prevention are needed. Animal models are essential to the study of lung cancer because they offer priceless information about the etiology, course, and possible treatments for the illness. The therapeutic application of phytochemicals and medicinal plants to treat cancer-related compounds has gained attention subsequently. In addition to discussing the molecular carcinogenic and antitumor effects of the herbal treatment Ocimum sanctum (OS) in connection to lung cancer, this review will address the current awareness regarding lung cancer in animal models. The multitude of animal models used in lung cancer research-such as genetically modified mice, carcinogen-induced models, and xenograft induction-provides a solid foundation for understanding the illness. By easing the examination of the environmental and genetic factors involved and enhancing the analysis of possibilities for treatment, these models eventually assist in the further development of lung cancer therapy. Additionally, using the herb plant OS is essential for both treating and preventing lung cancer. Standardizing dosages and enforcing laws on the use of herbal medications require more in-depth investigation.
Collapse
Affiliation(s)
- Ulayatul Kustiati
- Post-Graduate School of Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Laboratory of Pharmacology, Faculty of Veterinary Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Dwi Aris Agung Nugrahaningsih
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Dwi Liliek Kusindarta
- Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Hevi Wihadmadyatami
- Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
20
|
Szmajda-Krygier D, Nocoń Z, Pietrzak J, Krygier A, Balcerczak E. Assessment of Methylation in Selected ADAMTS Family Genes in Non-Small-Cell Lung Cancer. Int J Mol Sci 2025; 26:934. [PMID: 39940703 PMCID: PMC11816904 DOI: 10.3390/ijms26030934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Alterations in the methylation of genetic material can influence carcinogenesis by the downregulation or overexpression of ADAMTS (a disintegrin-like and metalloprotease with thrombospondin motifs) protease genes. Through their proteolytic activity, these enzymes are also capable of promoting angiogenesis. Consequently, ADAMTS proteases can either facilitate or inhibit cancer progression. This study aimed to evaluate the methylation levels of the ADAMTS6, ADAMTS9, and ADAMTS12 genes in non-small-cell lung cancer (NSCLC) using data from bioinformatics databases. The focus was on differences between lung adenocarcinoma (LUAD) and lung squamous-cell carcinoma (LUSC) subtypes and their impact on patient overall survival (OS). ADAMTS6 gene expression is significantly reduced in LUSC, and analysis of ADAMTS9 gene expression showed a significantly reduced gene transcript level in LUAD and LUSC, while both NSCLC subtypes demonstrated ADAMTS12 upregulation. In LUSC, significantly elevated promoter methylation was found in all of the aforementioned genes, while in LUAD, higher promoter methylation was observed only for ADAMTS9 and ADAMTS12. The differential methylation region (DMR) pattern demonstrated by ADAMTS6, ADAMTS9, and ADAMTS12 is a useful tool for distinguishing normal from cancer cells. The areas under the curve (AUCs) ranged from 0.86 to 0.99 for both LUAD and LUSC subtypes. The methylation level of different CpG sites among selected ADAMTS members is related to patient survival, suggesting it may have value as a prognostic marker. The methylation degree of promoter regions in genes encoding ADAMTS family proteins could significantly influence LUSC and LUAD. Increased promoter methylation could also reduce certain gene expression, contributing to cancer progression. The expression levels and specific DMRs of ADAMTS genes may serve as prognostic markers correlating with patient OS. Assessing ADAMTS gene methylation could become a diagnostic tool for differentiating NSCLC subtypes and potentially guide therapeutic strategies. Further research is needed to fully understand the activity and mechanisms of ADAMTS family proteins.
Collapse
Affiliation(s)
- Dagmara Szmajda-Krygier
- Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Czechoslowacka 4, 92-216 Lodz, Poland
| | - Zuzanna Nocoń
- Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| | - Jacek Pietrzak
- Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Czechoslowacka 4, 92-216 Lodz, Poland
| | - Adrian Krygier
- Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Czechoslowacka 4, 92-216 Lodz, Poland
| | - Ewa Balcerczak
- Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Czechoslowacka 4, 92-216 Lodz, Poland
| |
Collapse
|
21
|
Mangarova DB, Kaufmann JO, Brangsch J, Kader A, Möckel J, Heyl JL, Verlemann C, Adams LC, Ludwig A, Reimann C, Poller WC, Niehaus P, Karst U, Taupitz M, Hamm B, Weller MG, Makowski MR. ADAMTS4-Specific MR Peptide Probe for the Assessment of Atherosclerotic Plaque Burden in a Mouse Model. Invest Radiol 2025:00004424-990000000-00282. [PMID: 39804796 DOI: 10.1097/rli.0000000000001152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
INTRODUCTION Atherosclerosis is the underlying cause of multiple cardiovascular pathologies. The present-day clinical imaging modalities do not offer sufficient information on plaque composition or rupture risk. A disintegrin and metalloproteinase with thrombospondin motifs 4 (ADAMTS4) is a strongly upregulated proteoglycan-cleaving enzyme that is specific to cardiovascular diseases, inter alia, atherosclerosis. MATERIALS AND METHODS Male apolipoprotein E-deficient mice received a high-fat diet for 2 (n = 11) or 4 months (n = 11). Additionally, a group (n = 11) receiving pravastatin by drinking water for 4 months alongside the high-fat diet was examined. The control group (n = 10) consisted of C57BL/6J mice on standard chow. Molecular magnetic resonance imaging was performed prior to and after administration of the gadolinium (Gd)-based ADAMTS4-specific probe, followed by ex vivo analyses of the aortic arch, brachiocephalic arteries, and carotid arteries. A P value <0.05 was considered to indicate a statistically significant difference. RESULTS With advancing atherosclerosis, a significant increase in the contrast-to-noise ratio was measured after intravenous application of the probe (mean precontrast = 2.25; mean postcontrast = 11.47, P < 0.001 in the 4-month group). The pravastatin group presented decreased ADAMTS4 expression. A strong correlation between ADAMTS4 content measured via immunofluorescence staining and an increase in the contrast-to-noise ratio was detected ( R2 = 0.69). Microdissection analysis revealed that ADAMTS4 gene expression in the plaque area was significantly greater than that in the arterial wall of a control mouse ( P < 0.001). Laser ablation-inductively coupled plasma-mass spectrometry confirmed strong colocalization of areas positive for ADAMTS4 and Gd. CONCLUSIONS Magnetic resonance imaging using an ADAMTS4-specific agent is a promising method for characterizing atherosclerotic plaques and could improve plaque assessment in the diagnosis and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Dilyana B Mangarova
- From the Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany (D.B.M., J.O.K., J.B., A.K., J.M., J.L.H., C.R., M.T., B.H., M.R.M.); Department of Diagnostic and Interventional Radiology, Technical University of Munich, Munich, Germany (D.B.M., J.O.K., J.B., A.K., L.C.A., M.R.M.); Department of Chemistry, Humboldt-Universität zu Berlin, Berlin, Germany (J.O.K.); Division 1.5 Protein Analysis, Federal Institute for Materials Research and Testing, Berlin, Germany (J.O.K., M.G.W.); Department of Biology, Chemistry, and Pharmacy, Institute of Biology, Freie Universität Berlin, Berlin, Germany (A.K.); Department of Veterinary Medicine, Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Berlin, Germany (J.L.H.); Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany (C.V., P.N., U.K.); Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany (A.L.); DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany (A.L.); and Division of Cardiology, Massachusetts General Hospital, Harvard University, Boston, MA (W.C.P.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Xie MQ, Wang LJ, Xiao HM, Wei SJ. Regulatory networks of mRNAs and miRNAs involved in the immune response of diamondback moth, Plutella xylostella to fungal infection. BMC Genomics 2025; 26:15. [PMID: 39762741 PMCID: PMC11706182 DOI: 10.1186/s12864-024-11192-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND The entomopathogenic fungus, Isaria fumosorosea, shows promise as a biological control agent in managing the diamondback moth (DBM) Plutella xylostella, a highly destructive global pest of cruciferous vegetables. To date, the miRNA-mRNA regulatory networks underlying the immune response of DBM to I. fumosorosea infection are still poorly understood. Here, we characterize the expression profiles of miRNA and mRNA, and construct the miRNA-gene regulatory network in DBM infected with I. fumosorosea. RESULTS We identified 580 differentially expressed genes (DEGs) and 55 differentially expressed miRNAs (DEMs) in I. fumosorosea-infected DBM. Among these DEGs, we found 28 immunity-related genes, which mainly include pattern recognition receptors, signal modulators, and immune effectors. Integrated analysis discovered 87 negative correlation pairs between miRNA and mRNA, involving 40 DEMs and 62 DEGs in infected DBM. Additionally, 13 miRNAs and 10 corresponding mRNAs were identified as candidate miRNA-mRNA pairs for DBM immunity against fungal infection. Gene functional enrichment analysis indicated that these miRNAs could target genes associated with various pathways, such as the immune system, infectious diseases, digestive system, endocrine system, nervous system, and signal transduction. Finally, the regulatory relationships of six miRNA-mRNA pairs were validated using quantitative reverse transcription PCR. CONCLUSIONS For the first time, we present integrated miRNA and mRNA data to elucidate the immune response of the DBM to fungal infection. Our findings enhance the understanding of the immune response of the DBM to entomopathogenic fungi infection.
Collapse
Affiliation(s)
- Mei-Qiong Xie
- College of Life Sciences and Resources and Environment, Yichun University, Yichun, 336000, China
| | - Long-Jiang Wang
- College of Chemistry and Bioengineering, Yichun University, Yichun, 336000, China.
| | - Hua-Mei Xiao
- College of Life Sciences and Resources and Environment, Yichun University, Yichun, 336000, China
| | - Shu-Jun Wei
- Institute of Plant Protection, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China.
| |
Collapse
|
23
|
Janečková E, Juarez-Balarezo J, Tucker AS, Matalová E, Holomková K, Gaete M. Metalloproteinases are involved in the regulation of prenatal tooth morphogenesis. Am J Physiol Cell Physiol 2025; 328:C323-C333. [PMID: 39510136 DOI: 10.1152/ajpcell.00656.2024] [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: 09/09/2024] [Revised: 10/14/2024] [Accepted: 11/03/2024] [Indexed: 11/15/2024]
Abstract
During development, tooth germs undergo various morphological changes resulting from interactions between the oral epithelium and ectomesenchyme. These processes are influenced by the extracellular matrix, the composition of which, along with cell adhesion and signaling, is regulated by metalloproteinases. Notably, these include matrix metalloproteinases (MMPs), a disintegrin and metalloproteinases (ADAMs), and a disintegrin and metalloproteinases with thrombospondin motifs (ADAMTSs). Our analysis of previously published scRNAseq datasets highlight that these metalloproteinases show dynamic expression patterns during tooth development, with expression in a wide range of cell types, suggesting multiple roles in tooth morphogenesis. To investigate this, Marimastat, a broad-spectrum inhibitor of MMPs, ADAMs, and ADAMTSs, was applied to ex vivo cultures of mouse molar tooth germs. The treated samples exhibited significant changes in tooth germ size and morphology, including an overall reduction in size and an inversion of the typical bell shape. The cervical loop failed to extend, and the central area of the inner enamel epithelium protruded. Marimastat treatment also disrupted proliferation, cell polarization, and organization compared with control tooth germs. In addition, a decrease in laminin expression was observed, leading to a disruption in continuity of the basement membrane at the epithelial-mesenchymal junction. Elevated hypoxia-inducible factor 1-alpha gene (Hif-1α) expression correlated with a disruption to blood vessel development around the tooth germs. These results reveal the crucial role of metalloproteinases in tooth growth, shape, cervical loop elongation, and the regulation of blood vessel formation during prenatal tooth development.NEW & NOTEWORTHY Inhibition of metalloproteinases during tooth development had a wide-ranging impact on molar growth affecting proliferation, cell migration, and vascularization, highlighting the diverse role of these proteins in controlling development.
Collapse
Affiliation(s)
- Eva Janečková
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czech Republic
- Division of Biology, Glendale Community College, Glendale, California, United States
| | - Jesus Juarez-Balarezo
- Department of Anatomy, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Abigail S Tucker
- Department of Craniofacial Development and Stem Cell Biology, King's College London, London, United Kingdom
- 1st Faculty of Medicine, Institute of Histology and Embryology, Charles University, Prague, Czech Republic
| | - Eva Matalová
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czech Republic
| | - Kateřina Holomková
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czech Republic
| | - Marcia Gaete
- Department of Anatomy, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Center for Studies and Innovation in Dentistry, Faculty of Dentistry, Universidad Finis Terrae, Santiago, Chile
| |
Collapse
|
24
|
Wang J, Gao R, Qi J, Xing Y, Hong B, Wang H, Nie J. A comprehensive analysis of vasculogenic mimicry related genes to predict the survival rate of HCC and its influence on the tumor microenvironment. Front Genet 2024; 15:1437715. [PMID: 39748947 PMCID: PMC11693674 DOI: 10.3389/fgene.2024.1437715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 12/04/2024] [Indexed: 01/04/2025] Open
Abstract
Objectives Investigate the predictive value of Vasculogenic mimicry (VM) related genes for the survival and prognosis of Hepatocellular carcinoma (HCC) patients and its role in the tumor microenvironment (TME). Methods VM-related genes were obtained from previous literature, the expression profiles, single-cell data and clinical information of HCC patients were downloaded from public databases. The HCC patients were divided into different clusters by unsupervised clustering, the differences in prognosis and immune characteristics of VM-related clusters were analyzed. A prognostic model related to VM (VM Score) was constructed based on LASSO regression and univariate and multivariate Cox regression, the correlation between this model and chemotherapy drugs and immunotherapy was studied. Seurat package was used to standardize single-cell data for single-cell level analysis. The expression of risk factors in VM Score was verified by RT-qPCR. Results VM Score composed of SPP1, ADAMTS5 and ZBP1 was constructed and validated. VM Score was an independent prognostic factor for HCC. Through the analysis of single cell data further reveals the VM Score influence on TME. In addition, VM Score could provide ideas for the selection of immunotherapy and chemotherapy drugs. RT-qPCR showed that the expression of risk factors was different in HCC cell lines. Conclusion Our results suggest that VM Score may serve as a promising prognostic biomarker for HCC and provide new ideas for immunotherapy in HCC patients.
Collapse
Affiliation(s)
- Jingyun Wang
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Rong Gao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Jian Qi
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Yingru Xing
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Department of Clinical Laboratory, Anhui Zhongke Gengjiu Hospital, Hefei, China
| | - Bo Hong
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Hefei Cancer Hospital of CAS, Hefei, China
| | - Hongzhi Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Hefei Cancer Hospital of CAS, Hefei, China
| | - Jinfu Nie
- School of Medicine, Anhui University of Science and Technology, Huainan, China
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Hefei Cancer Hospital of CAS, Hefei, China
| |
Collapse
|
25
|
Wong CA, Sanchez-Rodriguez G, Ethier CR, Wood LB, Feola AJ. Ovariectomy drives increase of an ECM transcription signature in the posterior eye and retina. Vision Res 2024; 225:108507. [PMID: 39476526 PMCID: PMC11771480 DOI: 10.1016/j.visres.2024.108507] [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: 07/18/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/12/2024]
Abstract
Increased risk of developing glaucoma has recently been associated with early age of menopause. Here, we examined how age and surgically-induced menopause via ovariectomy (OVX) impacted gene expression in gene pathways previously linked to glaucoma, such as extracellular matrix (ECM) remodeling and TGF-β signaling. Using bulk RNA sequencing, we analyzed changes in young (3-4 months) and middle-aged (9-10 months) Long-Evans rats. We focused on posterior pole tissues (sclera and optic nerve head) but also examined the retina to compare observed changes across different tissue regions. Our results demonstrated that aging and OVX significantly alter gene expression in the sclera and optic nerve head. Generally, OVX triggered the enrichment of immune-related processes. However, OVX in young rats also led to significant enrichment of ECM and TGF-β gene sets. At the same time, these effects were diminished in middle-aged rats, indicating an age dependency of the effects of OVX on matrix-related pathways. Notably, the transcriptional factor Fos was downregulated in the posterior eye and retina in aged and OVX animals. Fos is a major regulator of cell proliferation and survival, and its dysregulation may play an important role in aging and menopause for women. These findings underscore the important role of menopause timing in modulating molecular pathways associated with glaucoma, which is consistent with clinical studies showing that early menopause may heighten the risk of developing this condition. This study also highlights the importance of considering women's health factors, such as menopause, in understanding and managing glaucoma risk.
Collapse
Affiliation(s)
- Cydney A Wong
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Gabriela Sanchez-Rodriguez
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States; Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, United States; Department of Ophthalmology, Emory University, Atlanta, GA, United States
| | - C Ross Ethier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States; Department of Ophthalmology, Emory University, Atlanta, GA, United States; George W. Woodruff School of Mechanical Engineering and Paker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Levi B Wood
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States; George W. Woodruff School of Mechanical Engineering and Paker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Andrew J Feola
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States; Department of Ophthalmology, Emory University, Atlanta, GA, United States; Center for Visual and Neurocognitive Rehabilitation, Joseph Maxwell Cleland Atlanta VA Medical Center, Atlanta, GA, United States.
| |
Collapse
|
26
|
Zhao D, Dou Y, Zeng L, Han Y, Lin F, Xu N, Liu J, Zeng Y. The effects of extracellular matrix-degrading enzymes polymorphisms on intervertebral disc degeneration. JOR Spine 2024; 7:e70012. [PMID: 39568776 PMCID: PMC11576917 DOI: 10.1002/jsp2.70012] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/22/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024] Open
Abstract
OBJECTIVE The objective of the current study was to investigate the correlation between polymorphisms in extracellular matrix-degrading enzymes and the risk of intervertebral disc degeneration (IDD) diseases. METHODS The databases PubMed, Embase, and Cochrane Database were systematically queried from the inception until March 2023 to ascertain studies that meet the eligibility criteria. Utilizing a standardized data collection form to extract data from individual studies. The data were quantified using odds ratio (OR) along with its corresponding 95% confidence interval (95% CI), following an allelic model of inheritance. RESULTS The study included a total of nine studies and indicated that the presence of rs17576 in the MMP9 gene was significantly associated with an increased risk of IDD diseases (GG: 1.30, 95% CI [1.09-1.55], p = 0.004). The presence of other polymorphisms in extracellular matrix-degrading enzymes did not exhibit a significant association with the susceptibility to IDD. CONCLUSION The current study demonstrated a noteworthy correlation between the GG genotype of MMP-9 rs17576 and susceptibility to IDD. The available evidence is insufficient to substantiate the correlation between other extracellular matrix-degrading enzymes and susceptibility to IDD. The constraints of this analysis necessitate further research involving larger sample sizes across diverse ethnicities to provide a comprehensive understanding of the true impact of these polymorphisms on susceptibility to IDD.
Collapse
Affiliation(s)
- Di Zhao
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine(Guangdong Provincial Hospital of Chinese Medicine)GuangzhouChina
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouChina
| | - Yao‐xing Dou
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouChina
| | - Ling‐feng Zeng
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine(Guangdong Provincial Hospital of Chinese Medicine)GuangzhouChina
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouChina
| | - Yan‐hong Han
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine(Guangdong Provincial Hospital of Chinese Medicine)GuangzhouChina
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouChina
| | - Fang‐zheng Lin
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouChina
| | - Nan‐jun Xu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine(Guangdong Provincial Hospital of Chinese Medicine)GuangzhouChina
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouChina
| | - Jun Liu
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical SciencesGuangzhouChina
- Guangdong Second Traditional Chinese Medicine Hospital (Guangdong Province Enginering Technology Research Institute of Traditional Chinese Medicine)GuangzhouChina
- The Fifth Clinical Medical College of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Yu‐ping Zeng
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine(Guangdong Provincial Hospital of Chinese Medicine)GuangzhouChina
| |
Collapse
|
27
|
Ding H, Shi X, Ma J, Cao C, Liu Y, Lu J, Bai L, Li X, Li H. Integrative transcriptomic analysis reveals Cd72 as a novel pro-inflammatory factor in microglia following experimental ischemic stroke. Exp Neurol 2024; 382:114974. [PMID: 39326825 DOI: 10.1016/j.expneurol.2024.114974] [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: 06/29/2024] [Revised: 09/07/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
Ischemic stroke remains a leading cause of global mortality and disability, with neuroinflammation playing a critical role in determining patient outcomes. Microglia, the brain's resident immune cells, can both exacerbate neuroinflammation and neuronal damage by releasing neurotoxic mediators and engaging in excessive phagocytosis, while also aiding recovery through the production of anti-inflammatory cytokines and debris clearance. However, the molecular mechanisms governing microglial activation and polarization after ischemic stroke are not well elucidated. In this study, we combined integrative transcriptomic analyses with experimental validation in a murine model of middle cerebral artery occlusion/reperfusion (MCAO/R) to explore microglial heterogeneity and identify key regulatory factors in ischemic stroke. Bioinformatics analysis identified Cd72 as a novel pro-inflammatory modulator within ischemia-associated microglial phenotypes. We observed significant upregulation of Cd72 in microglia following MCAO/R, and selective knockdown of Cd72 using CX3CR1Cre/ERT2 mice and Cre recombinase-dependent adeno-associated virus reduced MCAO/R-induced infarct volume, neuronal apoptosis, and neurological deficits. Furthermore, Cd72 expression in microglia was positively correlated with pro-inflammatory pathways and cytokines, including TNF-α, IL-1β, and IL-6. Knockdown of Cd72 significantly reduced these pro-inflammatory factors, highlighting its potential as a therapeutic target for mitigating inflammation in ischemic stroke. In conclusion, this study identifies Cd72 as a critical pro-inflammatory regulator in microglia following ischemic stroke, with its knockdown effectively reducing neuroinflammation and associated brain injury, highlighting Cd72 as a promising therapeutic target.
Collapse
Affiliation(s)
- Haojie Ding
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xuan Shi
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Junwei Ma
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Chang Cao
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Yangyang Liu
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Jinxin Lu
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Lei Bai
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xiang Li
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| | - Haiying Li
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| |
Collapse
|
28
|
Hassan HM, Hamdan AM, Alattar A, Alshaman R, Bahattab O, Al-Gayyar MMH. Evaluating anticancer activity of emodin by enhancing antioxidant activities and affecting PKC/ADAMTS4 pathway in thioacetamide-induced hepatocellular carcinoma in rats. Redox Rep 2024; 29:2365590. [PMID: 38861483 PMCID: PMC11168332 DOI: 10.1080/13510002.2024.2365590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024] Open
Abstract
Emodin is a naturally occurring anthraquinone derivative with a wide range of pharmacological activities, including neuroprotective and anti-inflammatory activities. We aim to assess the anticancer activity of emodin against hepatocellular carcinoma (HCC) in rat models using the proliferation, invasion, and angiogenesis biomarkers. After induction of HCC, assessment of the liver impairment and the histopathology of liver sections were investigated. Hepatic expression of both mRNA and protein of the oxidative stress biomarkers, HO-1, Nrf2; the mitogenic activation biomarkers, ERK5, PKCδ; the tissue destruction biomarker, ADAMTS4; the tissue homeostasis biomarker, aggregan; the cellular fibrinolytic biomarker, MMP3; and of the cellular angiogenesis biomarker, VEGF were measured. Emodin increased the survival percentage and reduced the number of hepatic nodules compared to the HCC group. Besides, emodin reduced the elevated expression of both mRNA and proteins of all PKC, ERK5, ADAMTS4, MMP3, and VEGF compared with the HCC group. On the other hand, emodin increased the expression of mRNA and proteins of Nrf2, HO-1, and aggrecan compared with the HCC group. Therefore, emodin is a promising anticancer agent against HCC preventing the cancer prognosis and infiltration. It works through many mechanisms of action, such as blocking oxidative stress, proliferation, invasion, and angiogenesis.
Collapse
Affiliation(s)
- Hanan M. Hassan
- Dept. of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Ahmed M. Hamdan
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Abdullah Alattar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Reem Alshaman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Omar Bahattab
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Mohammed M. H. Al-Gayyar
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| |
Collapse
|
29
|
Carver JJ, Amato CM, Hung-Chang Yao H, Zhu Y. Adamts9 is required for the development of primary ovarian follicles and maintenance of female sex in zebrafish†. Biol Reprod 2024; 111:1107-1128. [PMID: 39180722 PMCID: PMC11565245 DOI: 10.1093/biolre/ioae127] [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: 04/15/2024] [Revised: 07/15/2024] [Accepted: 08/23/2024] [Indexed: 08/26/2024] Open
Abstract
Previous studies have suggested that adamts9 (a disintegrin and metalloprotease with thrombospondin type-1 motifs, member 9), an extracellular matrix (ECM) metalloprotease, participates in primordial germ cell (PGC) migration and is necessary for female fertility. In this study, we found that adamts9 knockout (KO) led to reduced body size, and female-to-male sex conversion in late juvenile or adult zebrafish; however, primary sex determination was not affected in early juveniles of adamts9 KO. Overfeeding and lowering the rearing density rescued growth defects in female adamts9 KO fish but did not rescue defects in ovarian development in adamts9 KO. Delayed PGC proliferation, significantly reduced number and size of Stage IB follicles (equivalent to primary follicles) in early juveniles of adamts9 KO, and arrested development at Stage IB follicles in mid- or late-juveniles of adamts9 KO are likely causes of female infertility and sex conversion. Via RNAseq, we found significant enrichment of differentially expressed genes involved in ECM organization during sexual maturation in ovaries of wildtype fish; and significant dysregulation of these genes in adamts9 KO ovaries. RNAseq analysis also showed enrichment of inflammatory transcriptomic signatures in adult ovaries of these adamts9 KO. Taken together, our results indicate that adamts9 is critical for development of primary ovarian follicles and maintenance of female sex, and loss of adamts9 leads to defects in ovarian follicle development, female infertility, and sex conversion in late juveniles and mature adults. These results show that the ECM and extracellular metalloproteases play major roles in maintaining ovarian follicle development in zebrafish.
Collapse
Affiliation(s)
- Jonathan J Carver
- Department of Biology, East Carolina University, Greenville, NC, USA
| | - Ciro M Amato
- National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, USA
| | | | - Yong Zhu
- Department of Biology, East Carolina University, Greenville, NC, USA
| |
Collapse
|
30
|
Chang MY, Chan CK, Brune JE, Manicone AM, Bomsztyk K, Frevert CW, Altemeier WA. Regulation of versican expression in macrophages is mediated by canonical type I interferon signaling via ISGF3. Am J Physiol Cell Physiol 2024; 327:C1274-C1288. [PMID: 39400584 PMCID: PMC11559644 DOI: 10.1152/ajpcell.00174.2024] [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: 03/18/2024] [Revised: 09/09/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024]
Abstract
Growing evidence supports a role for versican as an important component of the inflammatory response, with both pro- and anti-inflammatory roles depending on the specific context of the system or disease under investigation. Our goal is to understand the regulation of macrophage-derived versican and the role it plays in innate immunity. In previous work, we showed that LPS triggers a signaling cascade involving Toll-like receptor (TLR)4, the Trif adaptor, type I interferons, and the type I interferon receptor, leading to increased versican expression by macrophages. In the present study, we used a combination of chromatin immunoprecipitation, siRNA, chemical inhibitors, and mouse model approaches to investigate the regulatory events downstream of the type I interferon receptor to better define the mechanism controlling versican expression. Results indicate that transcriptional regulation by canonical type I interferon signaling via interferon-stimulated gene factor 3 (ISGF3), the heterotrimeric transcription factor complex of Irf9, Stat1, and Stat2, controls versican expression in macrophages exposed to LPS. This pathway is not dependent on MAPK signaling, which has been shown to regulate versican expression in other cell types. The stability of versican mRNA may also contribute to prolonged versican expression in macrophages. These findings strongly support a role for macrophage-derived versican as a type I interferon-stimulated gene and further our understanding of versican's role in regulating inflammation.NEW & NOTEWORTHY We report the novel finding that versican expression is regulated by the interferon-stimulated gene factor 3 (ISGF3) arm of canonical type I Ifn signaling in LPS-stimulated macrophages. This pathway is distinct from mechanisms that control versican expression in other cell types. This suggests that macrophage-derived versican may play a role in limiting a potentially excessive inflammatory response. The detailed understanding of how versican expression is regulated in different cells could lead to unique approaches for enhancing its anti-inflammatory properties.
Collapse
Affiliation(s)
- Mary Y Chang
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States
- Center for Lung Biology, University of Washington at South Lake Union, Seattle, Washington, United States
| | - Christina K Chan
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States
- Center for Lung Biology, University of Washington at South Lake Union, Seattle, Washington, United States
| | - Jourdan E Brune
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States
- Center for Lung Biology, University of Washington at South Lake Union, Seattle, Washington, United States
| | - Anne M Manicone
- Center for Lung Biology, University of Washington at South Lake Union, Seattle, Washington, United States
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States
| | - Karol Bomsztyk
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, United States
| | - Charles W Frevert
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States
- Center for Lung Biology, University of Washington at South Lake Union, Seattle, Washington, United States
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States
| | - William A Altemeier
- Center for Lung Biology, University of Washington at South Lake Union, Seattle, Washington, United States
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States
| |
Collapse
|
31
|
Hu H, Wang Y, Liu Y, Zhang C, Li G, Zhang T, Dong B. Comprehensive Analysis of ADAMTS Gene Family in Renal Clear Cell Carcinoma and ADAMTS10 Research Combining Magnetic Resonance Imaging. Mol Biotechnol 2024; 66:3136-3149. [PMID: 37861954 DOI: 10.1007/s12033-023-00915-8] [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: 08/03/2023] [Accepted: 09/19/2023] [Indexed: 10/21/2023]
Abstract
Clear cell renal carcinoma (ccRCC) is one of the cancers that posed a severe threat to human life on a global scale. The ADAMTS family has been proven to be involved in a number of tumor types, although it is yet unknown how they relate to ccRCC. The mRNA expression matrix and other clinically relevant information of 607 ccRCC were sourced from TCGA database. The role of ADAMTS family genes in ccRCC was determined by differential gene expression analysis and gene set enrichment analysis (GSEA). Employing stage grading, gene mutation, and survival analysis, the genes most linked to the prognosis of ccRCC were identified. The influence of genes on the pathway was determined by Kyoto Encyclopedia of Genes and Genes (KEGG) analysis. Following that, the gene's impact on ccRCC was verified by qRT-PCR, WB, MTT, Transwell detection, and a wound healing assay. Bioinformatics analysis showed that ADAMTS10 was overexpressed in cancerous tissues of people with ccRCC and its expression increased with tumor grade. Mutation analysis showed that the main cause of mutation in the ADAMTS family gene was amplification. The prognosis and survival of the ADAMTS10 elevated expression group were lower than those of the poorly expressed group, as demonstrated by a survival analysis. On the basis of the findings of MRI, we examined 60 clinical patients and collected their cancer along with the surrounding tissues. The results of qPCR detection showed that the expression of ADAMTS10 was considerably higher in cancerous regions of 60 clinical users than it was in the tissues nearby. Inhibiting ADAMTS10 development prevents cancer cells from proliferating, invading, and migrating. The KEGG analysis links ADAMTS10 to the NF-κB signal pathway. WB experiment confirmed that inhibiting ADAMTS10 expression can inhibit the activation of the NF-κB signal pathway. ADAMTS10 may be a promising prognostic marker for ccRCC that can be employed independently.
Collapse
Affiliation(s)
- Haifeng Hu
- Department of Magnetic Resonance Imaging, The Second Affiliated Hospital of Qiqihar Medical College, 64 Zhonghua Xi Lu, Jianhua District, Qiqihar City, Heilongjiang Province, China.
| | - Ying Wang
- Department of Ultrasound, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar City, China
| | - Ying Liu
- Department of Magnetic Resonance Imaging, The Second Affiliated Hospital of Qiqihar Medical College, 64 Zhonghua Xi Lu, Jianhua District, Qiqihar City, Heilongjiang Province, China
| | - Chunyu Zhang
- Department of Magnetic Resonance Imaging, The Second Affiliated Hospital of Qiqihar Medical College, 64 Zhonghua Xi Lu, Jianhua District, Qiqihar City, Heilongjiang Province, China
| | - Guoan Li
- Department of Magnetic Resonance Imaging, The Second Affiliated Hospital of Qiqihar Medical College, 64 Zhonghua Xi Lu, Jianhua District, Qiqihar City, Heilongjiang Province, China
| | - Tianyu Zhang
- Department of Imaging, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar City, China
| | - Bo Dong
- Department of Urology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar City, China
| |
Collapse
|
32
|
Zhai W, Yang W, Ge J, Xiao X, Wu K, She K, Zhou Y, Kong Y, Wu L, Luo S, Pu X. ADAMTS4 exacerbates lung cancer progression via regulating c-Myc protein stability and activating MAPK signaling pathway. Biol Direct 2024; 19:94. [PMID: 39415271 PMCID: PMC11483991 DOI: 10.1186/s13062-024-00512-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/08/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Lung cancer is one of the most frequent cancers and the leading cause of cancer-related deaths worldwide with poor prognosis. A disintegrin and metalloproteinase with thrombospondin motifs 4 (ADAMTS4) is crucial in the regulation of the extracellular matrix (ECM), impacting its formation, homeostasis and remodeling, and has been linked to cancer progression. However, the specific involvement of ADAMTS4 in the development of lung cancer remains unclear. METHODS ADAMTS4 expression was identified in human lung cancer samples by immunohistochemical (IHC) staining and the correlation of ADAMTS4 with clinical outcome was determined. The functional impact of ADAMTS4 on malignant phenotypes of lung cancer cells was explored both in vitro and in vivo. The mechanisms underlying ADAMTS4-mediated lung cancer progression were explored by ubiquitination-related assays. RESULTS Our study revealed a significant upregulation of ADAMTS4 at the protein level in lung cancer tissues compared to para-carcinoma normal tissues. High ADAMTS4 expression inversely correlated with the prognosis of lung cancer patients. Knockdown of ADAMTS4 inhibited the proliferation and migration of lung cancer cells, as well as the tubule formation of HUVECs, while ADAMTS4 overexpression exerted opposite effects. Mechanistically, we found that ADAMTS4 stabilized c-Myc by inhibiting its ubiquitination, thereby promoting the in vitro and in vivo growth of lung cancer cells and inducing HUVECs tubule formation in lung cancer. In addition, our results suggested that ADAMTS4 overexpression activated MAPK signaling pathway. CONCLUSIONS We highlighted the promoting role of ADAMTS4 in lung cancer progression and proposed ADAMTS4 as a promising therapeutic target for lung cancer patients.
Collapse
Affiliation(s)
- Wei Zhai
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Road, Wuhan, 430030, Hubei, China
| | - Wensheng Yang
- Department of Thoracic Surgery, The Affiliated Shaoyang Hospital, Hengyang Medical School, University of South China, No. 36, Hongqi Road, Daxiang District, Shaoyang, 422000, Hunan, China
| | - Jing Ge
- Department of Geriatrics and Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Road, Wuhan, 430030, Hubei, China
| | - Xuelian Xiao
- Department of Medical Administration, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Kang Wu
- Sansure Biotech Inc.,, No. 680, Lusong Road, Yuelu District, Changsha, 410205, Hunan, China
| | - Kelin She
- Department of Thoracic Surgery, Hunan Provincial Pecople's Hospital, The First Affiliated Hospital of Huan Nomal University, No. 61, Jiefang West Road, Furong District, Changsha, 410013, Hunan, China
| | - Yu Zhou
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Yi Kong
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Lin Wu
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Shiya Luo
- Sansure Biotech Inc.,, No. 680, Lusong Road, Yuelu District, Changsha, 410205, Hunan, China
| | - Xingxiang Pu
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China.
| |
Collapse
|
33
|
Li R, Kuang Y, Niu Y, Zhang S, Chen S, Su F, Wang J, Lin S, Liu D, Shen C, Liang L, Zheng SG, Jie L, Xiao Y, Xu H. FTO-mediated RNA m 6A methylation regulates synovial aggression and inflammation in rheumatoid arthritis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167341. [PMID: 39025373 DOI: 10.1016/j.bbadis.2024.167341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/25/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024]
Abstract
Fibroblast-like synoviocytes (FLS) plays an important role in synovial inflammation and joint damage in rheumatoid arthritis (RA). As the most abundant mRNA modification, N6-methyladenosine (m6A) is involved in the development of various diseases; however, its role in RA remains to be defined. In this study, we reported the elevated expression of the m6A demethylase fat mass and obesity-associated protein (FTO) in FLS and synovium from RA patients. Functionally, FTO knockdown or treatment with FB23-2, an inhibitor of the mRNA m6A demethylase FTO, inhibited the migration, invasion and inflammatory response of RA FLS, however, FTO-overexpressed RA FLS exhibited increased migration, invasion and inflammatory response. We further demonstrated that FTO promoted ADAMTS15 mRNA stability in an m6A-IGF2BP1 dependent manner. Notably, the severity of arthritis was significantly reduced in CIA mice with FB23-2 administration or CIA rats with intra-articular injection of FTO shRNA. Our results illustrate the contribution of FTO-mediated m6A modification to joint damage and inflammation in RA and suggest that FTO might be a potential therapeutic target in RA.
Collapse
Affiliation(s)
- Ruiru Li
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Yu Kuang
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Yuanyuan Niu
- Department of General Practice, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Shuoyang Zhang
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Simin Chen
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Fan Su
- Department of Geriatrics, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Jingnan Wang
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Shuibin Lin
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Di Liu
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Chuyu Shen
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Liuqin Liang
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Song Guo Zheng
- Department of Immunology, School of Cell and Gene Therapy, Song Jiang Research Institutes, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Ligang Jie
- Department of Rheumatology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Youjun Xiao
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China.
| | - Hanshi Xu
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China.
| |
Collapse
|
34
|
Aswani SS, Aparna NS, Mohan MS, Boban PT, Saja K. Sesame oil downregulates the expression of ADAMTS-4 in high-fat diet-induced atherosclerosis. Prostaglandins Other Lipid Mediat 2024; 174:106862. [PMID: 38936541 DOI: 10.1016/j.prostaglandins.2024.106862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/06/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease forming plaques in medium and large-sized arteries. ADAMTS-4 (a disintegrin and metalloproteinase with thrombospondin motifs-4) is an extracellular-matrix remodelling enzyme involved in the degradation of versican in the arterial wall. Recent reports indicated that increased expression of ADAMTS-4 is associated with plaque progression and vulnerability. Bioactive components of dietary oil, like sesame oil, are reported to have anti-inflammatory and antioxidant properties. Here, we studied the effect of sesame oil on regulating ADAMTS-4 in high-fat diet-induced atherosclerosis rat model. Our results indicated that sesame oil supplementation improved the anti-inflammatory and anti-oxidative status of the body. It also reduced atherosclerotic plaque formation in high-fat diet-fed rats. Our results showed that the sesame oil supplementation significantly down-regulated the expression of ADAMTS-4 in serum and aortic samples. The versican, the large proteoglycan substrate of ADAMTS-4 in the aorta, was downregulated to normal control level on sesame oil supplementation. This study, for the first time, reveals that sesame oil could down-regulate the expression of ADAMTS-4 in high-fat diet-induced atherosclerosis, imparting a new therapeutic potential for sesame oil in the management of atherosclerosis.
Collapse
Affiliation(s)
- S S Aswani
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala 695581, India
| | - N S Aparna
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala 695581, India
| | - Mithra S Mohan
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala 695581, India
| | - P T Boban
- Department of Biochemistry, Government College Kariavattom, Thiruvananthapuram, Kerala 695581, India
| | - K Saja
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala 695581, India.
| |
Collapse
|
35
|
Gaddala P, Choudhary S, Sethi S, Sainaga Jyothi VG, Katta C, Bahuguna D, Singh PK, Pandey M, Madan J. Etodolac utility in osteoarthritis: drug delivery challenges, topical nanotherapeutic strategies and potential synergies. Ther Deliv 2024; 15:977-995. [PMID: 39345034 PMCID: PMC11583675 DOI: 10.1080/20415990.2024.2405456] [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: 06/29/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
Osteoarthritis (OSA) is a prevalent joint disorder characterized by losing articular cartilage, primarily affecting the hip, knee and spine joints. The impact of OSA offers a major challenge to health systems globally. Therapeutic approaches encompass surgical interventions, non-pharmacological therapies (exercise, rehabilitation, behavioral interventions) and pharmacological treatments. Inflammatory processes within OSA joints are regulated by pro-inflammatory and anti-inflammatory cytokines. Etodolac, a COX-2-selective inhibitor, is the gold standard for OSA management and uniquely does not inhibit gastric prostaglandins. This comprehensive review offers insights into OSA's pathophysiology, genetic factors and biological determinants influencing disease progression. Emphasis is placed on the pivotal role of etodolac in OSA management, supported by both preclinical and clinical evidences in topical drug delivery. Notably, in-silico docking studies suggested potential synergies between etodolac and baicalein, considering ADAMTS-4, COX-2, MMP-3 and MMP-13 as essential therapeutic targets. Integration of artificial neural network (ANN) techniques with nanotechnology approaches emerges as a promising strategy for optimizing and personalizing topical etodolac delivery. Furthermore, the synergistic potential of etodolac and baicalein warrants in-depth exploration. Hence, by embracing cutting-edge technologies like ANN and nanomedicine, the optimization of topical etodolac delivery could guide a new era of OSA treatment.
Collapse
Affiliation(s)
- Pavani Gaddala
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research, Hyderabad, Telangana, India
| | - Shalki Choudhary
- Department of Pharmaceutical Sciences & Drug Research, Punjabi University, Patiala, Punjab, India
| | - Sheshank Sethi
- Department of Pharmaceutical Sciences & Drug Research, Punjabi University, Patiala, Punjab, India
| | - Vaskuri Gs Sainaga Jyothi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research, Hyderabad, Telangana, India
| | - Chantibabu Katta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research, Hyderabad, Telangana, India
| | - Deepankar Bahuguna
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research, Hyderabad, Telangana, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research, Hyderabad, Telangana, India
| | - Manisha Pandey
- Department of Pharmaceutical Sciences, Central University of Haryana, SSH 17, Jant, Haryana, 123031, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research, Hyderabad, Telangana, India
| |
Collapse
|
36
|
Ochman B, Limanówka P, Mielcarska S, Kula A, Dawidowicz M, Wagner W, Hudy D, Szrot M, Piecuch JZ, Piecuch J, Czuba Z, Świętochowska E. Associations of SEMA7A, SEMA4D, ADAMTS10, and ADAM8 with KRAS, NRAS, BRAF, PIK3CA, and AKT Gene Mutations, Microsatellite Instability Status, and Cytokine Expression in Colorectal Cancer Tissue. Curr Issues Mol Biol 2024; 46:10218-10248. [PMID: 39329961 PMCID: PMC11431007 DOI: 10.3390/cimb46090609] [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: 08/25/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Semaphorins (SEMAs), ADAM, and ADAMTS family members are implicated in various cancer progression events within the tumor microenvironment across different cancers. In this study, we aimed to evaluate the expression of SEMA7A, SEMA4D, ADAM8, and ADAMTS10 in colorectal cancer (CRC) in relation to the mutational landscape of KRAS, NRAS, BRAF, PIK3CA, and AKT genes, microsatellite instability (MSI) status, and clinicopathological features. We also examined the associations between the expression of these proteins and selected cytokines, chemokines, and growth factors, assessed using a multiplex assay. Protein concentrations were quantified using ELISA in CRC tumors and tumor-free surgical margin tissue homogenates. Gene mutations were evaluated via RT-PCR, and MSI status was determined using immunohistochemistry (IHC). GSEA and statistical analyses were performed using R Studio. We observed a significantly elevated expression of SEMA7A in BRAF-mutant CRC tumors and an overexpression of ADAM8 in KRAS 12/13-mutant tumors. The expression of ADAMTS10 was decreased in PIK3CA-mutant CRC tumors. No significant differences in the expression of the examined proteins were observed based on MSI status. The SEMA7A and SEMA4D expressions were correlated with the expression of numerous cytokines associated with various immune processes. The potential immunomodulatory functions of these molecules and their suitability as therapeutic targets require further investigation.
Collapse
Affiliation(s)
- Błażej Ochman
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-808 Zabrze, Poland; (B.O.); (P.L.); (S.M.); (W.W.); (D.H.)
| | - Piotr Limanówka
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-808 Zabrze, Poland; (B.O.); (P.L.); (S.M.); (W.W.); (D.H.)
| | - Sylwia Mielcarska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-808 Zabrze, Poland; (B.O.); (P.L.); (S.M.); (W.W.); (D.H.)
| | - Agnieszka Kula
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-808 Katowice, Poland; (A.K.); (M.D.)
| | - Miriam Dawidowicz
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-808 Katowice, Poland; (A.K.); (M.D.)
| | - Wiktor Wagner
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-808 Zabrze, Poland; (B.O.); (P.L.); (S.M.); (W.W.); (D.H.)
| | - Dorota Hudy
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-808 Zabrze, Poland; (B.O.); (P.L.); (S.M.); (W.W.); (D.H.)
| | - Monika Szrot
- Department of General and Bariatric Surgery and Emergency Medicine in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 10 Marii Curie-Skłodowskiej, 41-800 Zabrze, Poland; (M.S.); (J.P.)
| | - Jerzy Zbigniew Piecuch
- Department of General and Bariatric Surgery and Emergency Medicine in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 10 Marii Curie-Skłodowskiej, 41-800 Zabrze, Poland; (M.S.); (J.P.)
| | - Jerzy Piecuch
- Department of General and Bariatric Surgery and Emergency Medicine in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 10 Marii Curie-Skłodowskiej, 41-800 Zabrze, Poland; (M.S.); (J.P.)
| | - Zenon Czuba
- Department of Microbiology and Immunology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-808 Zabrze, Poland;
| | - Elżbieta Świętochowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-808 Zabrze, Poland; (B.O.); (P.L.); (S.M.); (W.W.); (D.H.)
| |
Collapse
|
37
|
Aswani SS, Jayan SG, Mohan MS, Aparna NS, Boban PT, Saja K. Chrysin downregulates the expression of ADAMTS-4 in foam cells. Mol Biol Rep 2024; 51:968. [PMID: 39249599 DOI: 10.1007/s11033-024-09896-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Chrysin, a polyphenolic compound, possesses antioxidant and anti-inflammatory properties. In this study, we investigated the effect of chrysin on the expression of A disintegrin and metalloproteinase with thrombospondin motifs-4 (ADAMTS-4), a protease enzyme involved in degrading extracellular matrix associated with atherosclerosis. METHODS AND RESULTS We have studied the cell viability by MTT assay and foam cell formation by oil red O staining. The mRNA and protein expression of ADAMTS-4 was studied using quantitative polymerase chain reaction (qPCR) and Western blotting, respectively. Our study showed that chrysin significantly downregulates the expression of ADAMTS-4 in foam cells. CONCLUSION Chrysin's ability to downregulate the expression of ADAMTS-4, a protease involved in degrading the extracellular matrix, bestows upon it a new therapeutic potential for managing atherosclerosis.
Collapse
Affiliation(s)
- S S Aswani
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India
| | - Sreelekshmi G Jayan
- Department of Biotechnology, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India
| | - Mithra S Mohan
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India
| | - N S Aparna
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India
| | - P T Boban
- Department of Biochemistry, Government College Kariavattom, Thiruvananthapuram, Kerala, 695581, India
| | - K Saja
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India.
| |
Collapse
|
38
|
Gu X, Wei S, Lv X. Circulating tumor cells: from new biological insights to clinical practice. Signal Transduct Target Ther 2024; 9:226. [PMID: 39218931 PMCID: PMC11366768 DOI: 10.1038/s41392-024-01938-6] [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: 11/02/2023] [Revised: 05/31/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
The primary reason for high mortality rates among cancer patients is metastasis, where tumor cells migrate through the bloodstream from the original site to other parts of the body. Recent advancements in technology have significantly enhanced our comprehension of the mechanisms behind the bloodborne spread of circulating tumor cells (CTCs). One critical process, DNA methylation, regulates gene expression and chromosome stability, thus maintaining dynamic equilibrium in the body. Global hypomethylation and locus-specific hypermethylation are examples of changes in DNA methylation patterns that are pivotal to carcinogenesis. This comprehensive review first provides an overview of the various processes that contribute to the formation of CTCs, including epithelial-mesenchymal transition (EMT), immune surveillance, and colonization. We then conduct an in-depth analysis of how modifications in DNA methylation within CTCs impact each of these critical stages during CTC dissemination. Furthermore, we explored potential clinical implications of changes in DNA methylation in CTCs for patients with cancer. By understanding these epigenetic modifications, we can gain insights into the metastatic process and identify new biomarkers for early detection, prognosis, and targeted therapies. This review aims to bridge the gap between basic research and clinical application, highlighting the significance of DNA methylation in the context of cancer metastasis and offering new avenues for improving patient outcomes.
Collapse
Affiliation(s)
- Xuyu Gu
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shiyou Wei
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
39
|
Eubanks E, VanderSleen K, Mody J, Patel N, Sacks B, Farahani MD, Wang J, Elliott J, Jaber N, Akçimen F, Bandres-Ciga S, Helweh F, Liu J, Archakam S, Kimelman R, Sharma B, Socha P, Guntur A, Bartels T, Dettmer U, Mouradian MM, Bahrami AH, Dai W, Baum J, Shi Z, Hardy J, Kara E. Increased burden of rare risk variants across gene expression networks predisposes to sporadic Parkinson's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610195. [PMID: 39257816 PMCID: PMC11384021 DOI: 10.1101/2024.08.30.610195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Alpha-synuclein (αSyn) is an intrinsically disordered protein that accumulates in the brains of patients with Parkinson's disease and forms intraneuronal inclusions called Lewy Bodies. While the mechanism underlying the dysregulation of αSyn in Parkinson's disease is unclear, it is thought that prionoid cell-to-cell propagation of αSyn has an important role. Through a high throughput screen, we recently identified 38 genes whose knock down modulates αSyn propagation. Follow up experiments were undertaken for two of those genes, TAX1BP1 and ADAMTS19, to study the mechanism with which they regulate αSyn homeostasis. We used a recently developed M17D neuroblastoma cell line expressing triple mutant (E35K+E46K+E61K) "3K" αSyn under doxycycline induction. 3K αSyn spontaneously forms inclusions that show ultrastructural similarities to Lewy Bodies. Experiments using that cell line showed that TAX1BP1 and ADAMTS19 regulate how αSyn interacts with lipids and phase separates into inclusions, respectively, adding to the growing body of evidence implicating those processes in Parkinson's disease. Through RNA sequencing, we identified several genes that are differentially expressed after knock-down of TAX1BP1 or ADAMTS19. Burden analysis revealed that those differentially expressed genes (DEGs) carry an increased frequency of rare risk variants in Parkinson's disease patients versus healthy controls, an effect that was independently replicated across two separate cohorts (GP2 and AMP-PD). Weighted gene co-expression network analysis (WGCNA) showed that the DEGs cluster within modules in regions of the brain that develop high degrees of αSyn pathology (basal ganglia, cortex). We propose a novel model for the genetic architecture of sporadic Parkinson's disease: increased burden of risk variants across genetic networks dysregulates pathways underlying αSyn homeostasis, thereby leading to pathology and neurodegeneration.
Collapse
Affiliation(s)
- Elena Eubanks
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Katelyn VanderSleen
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Jiya Mody
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Neha Patel
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Benjamin Sacks
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | | | - Jinying Wang
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Jordan Elliott
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Nora Jaber
- Department of Cell Biology and Neuroscience & Institute for Quantitative Biomedicine, Rutgers University, Piscataway, NJ 08854, USA
| | - Fulya Akçimen
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Sara Bandres-Ciga
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Fadel Helweh
- UNAM-National Nanotechnology Research Center and Institute of Materials Science & Nanotechnology, Bilkent University, Ankara, Turkey
| | - Jun Liu
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Sanjana Archakam
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Robert Kimelman
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Bineet Sharma
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Philip Socha
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Ananya Guntur
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Tim Bartels
- UK Dementia Research Institute, University College London, London W1T 7NF, United Kingdom
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - M. Maral Mouradian
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| | - Amir Houshang Bahrami
- UNAM-National Nanotechnology Research Center and Institute of Materials Science & Nanotechnology, Bilkent University, Ankara, Turkey
- Living Matter Physics, Max Planck Institute for Dynamics and Self-Organization, 37077 Göttingen, Germany
| | - Wei Dai
- Department of Cell Biology and Neuroscience & Institute for Quantitative Biomedicine, Rutgers University, Piscataway, NJ 08854, USA
| | - Jean Baum
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Zheng Shi
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - John Hardy
- UK Dementia Research Institute, University College London, London W1T 7NF, United Kingdom
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
- National Institute for Health Research (NIHR) University College London Hospitals Biomedical Research Centre, London W1T 7DN, UK
- Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Eleanna Kara
- Department of Neurology, Robert Wood Johnson Medical School, Institute for Neurological Therapeutics at Rutgers, Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| |
Collapse
|
40
|
Cuffaro D, Burkhard T, Bernardoni BL, Di Leo R, Zhang X, Galati S, Tuccinardi T, Macchia M, Rossello A, Santamaria S, de Groot R, Nuti E. Design, synthesis and biological evaluation of arylsulfonamides as ADAMTS7 inhibitors. RSC Med Chem 2024; 15:2806-2825. [PMID: 39149096 PMCID: PMC11324053 DOI: 10.1039/d4md00149d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/19/2024] [Indexed: 08/17/2024] Open
Abstract
The proteolytic activity of the enzyme ADAMTS7 was recently shown to enhance the progression of atherosclerosis, in line with human genetic findings suggesting that ADAMTS7 has a role in the pathophysiology of coronary heart disease. Targeting the active site of ADAMTS7 with a small molecule inhibitor, therefore, has therapeutic potential. Here, we report the design and synthesis of a novel hydroxamate-based arylsulfonamide that is a potent and selective ADAMTS7 inhibitor. In silico studies guided the hit optimization process aiming to improve selectivity of the previously reported (non-selective) inhibitor EDV33. Our lead compound is a p-trifluoromethyl biphenyl sulfonamide, which displayed a 12-fold selectivity for ADAMTS7 (K i = 9 nM) over ADAMTS5 (K i = 110 nM) and an 8-fold increase in inhibition of ADAMTS7 compared to EDV33 (K i = 70 nM). The substitutions switched selectivity and produced a new potent ADAMTS7 inhibitor that can be taken forward for further characterisation.
Collapse
Affiliation(s)
- Doretta Cuffaro
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy +39 0502219551
| | - Tina Burkhard
- Department of Biochemical and Physiological Sciences, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey Edward Jenner Building Guildford GU2 7XH UK
| | | | - Riccardo Di Leo
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy +39 0502219551
| | - Xiaohan Zhang
- Institute of Cardiovascular Science, University College London 51 Chenies Mews London WC1E 6HX UK +44 (0) 20 3108 1423
| | - Salvatore Galati
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy +39 0502219551
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy +39 0502219551
| | - Marco Macchia
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy +39 0502219551
| | - Armando Rossello
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy +39 0502219551
| | - Salvatore Santamaria
- Department of Biochemical and Physiological Sciences, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey Edward Jenner Building Guildford GU2 7XH UK
| | - Rens de Groot
- Institute of Cardiovascular Science, University College London 51 Chenies Mews London WC1E 6HX UK +44 (0) 20 3108 1423
| | - Elisa Nuti
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy +39 0502219551
| |
Collapse
|
41
|
Chen WF, Chuang JMJ, Yang SN, Chen NF, Bhattacharya M, Liu HT, Dhama K, Chakraborty C, Wen ZH. Gene expression profiling and the isocitrate dehydrogenase mutational landscape of temozolomide‑resistant glioblastoma. Oncol Lett 2024; 28:378. [PMID: 38939621 PMCID: PMC11209862 DOI: 10.3892/ol.2024.14511] [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: 09/21/2023] [Accepted: 04/09/2024] [Indexed: 06/29/2024] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain cancer that occurs more frequently than other brain tumors. The present study aimed to reveal a novel mechanism of temozolomide resistance in GBM using bioinformatics and wet lab analyses, including meta-Z analysis, Kaplan-Meier survival analysis, protein-protein interaction (PPI) network establishment, cluster analysis of co-expressed gene networks, and hierarchical clustering of upregulated and downregulated genes. Next-generation sequencing and quantitative PCR analyses revealed downregulated [tyrosine kinase with immunoglobulin and epidermal growth factor homology domains 1 (TIE1), calcium voltage-gated channel auxiliary subunit α2Δ1 (CACNA2D1), calpain 6 (CAPN6) and a disintegrin and metalloproteinase with thrombospondin motifs 6 (ADAMTS6)] and upregulated [serum amyloid (SA)A1, SAA2, growth differentiation factor 15 (GDF15) and ubiquitin specific peptidase 26 (USP26)] genes. Different statistical models were developed for these genes using the Z-score for P-value conversion, and Kaplan-Meier plots were constructed using several patient cohorts with brain tumors. The highest number of nodes was observed in the PPI network was for ADAMTS6 and TIE1. The PPI network model for all genes contained 35 nodes and 241 edges. Immunohistochemical staining was performed using isocitrate dehydrogenase (IDH)-wild-type or IDH-mutant GBM samples from patients and a significant upregulation of TIE1 (P<0.001) and CAPN6 (P<0.05) protein expression was demonstrated in IDH-mutant GBM in comparison with IDH-wild-type GBM. Structural analysis revealed an IDH-mutant model demonstrating the mutant residues (R132, R140 and R172). The findings of the present study will help the future development of novel biomarkers and therapeutics for brain tumors.
Collapse
Affiliation(s)
- Wu-Fu Chen
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan, R.O.C
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, R.O.C
| | - Jimmy Ming-Jung Chuang
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan, R.O.C
| | - San-Nan Yang
- Department of Pediatrics, E-DA Hospital, School of Medicine, College of Medicine I-Shou University, Kaohsiung 82445, Taiwan, R.O.C
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan, R.O.C
| | - Nan-Fu Chen
- Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan, R.O.C
- Center for General Education, Cheng Shiu University, Kaohsiung 833301, Taiwan, R.O.C
| | | | - Hsin-Tzu Liu
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970374, Taiwan, R.O.C
| | - Kuldeep Dhama
- Division of Pathology, Indian Council of Agriculture Research-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, R.O.C
| |
Collapse
|
42
|
Sirois JP, Heinz A. Matrikines in the skin: Origin, effects, and therapeutic potential. Pharmacol Ther 2024; 260:108682. [PMID: 38917886 DOI: 10.1016/j.pharmthera.2024.108682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/31/2024] [Accepted: 06/21/2024] [Indexed: 06/27/2024]
Abstract
The extracellular matrix (ECM) represents a complex multi-component environment that has a decisive influence on the biomechanical properties of tissues and organs. Depending on the tissue, ECM components are subject to a homeostasis of synthesis and degradation, a subtle interplay that is influenced by external factors and the intrinsic aging process and is often disturbed in pathologies. Upon proteolytic cleavage of ECM proteins, small bioactive peptides termed matrikines can be formed. These bioactive peptides play a crucial role in cell signaling and contribute to the dynamic regulation of both physiological and pathological processes such as tissue remodeling and repair as well as inflammatory responses. In the skin, matrikines exert an influence for instance on cell adhesion, migration, and proliferation as well as vasodilation, angiogenesis and protein expression. Due to their manifold functions, matrikines represent promising leads for developing new therapeutic options for the treatment of skin diseases. This review article gives a comprehensive overview on matrikines in the skin, including their origin in the dermal ECM, their biological effects and therapeutic potential for the treatment of skin pathologies such as melanoma, chronic wounds and inflammatory skin diseases or for their use in anti-aging cosmeceuticals.
Collapse
Affiliation(s)
- Jonathan P Sirois
- Department of Pharmacy, LEO Foundation Center for Cutaneous Drug Delivery, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Heinz
- Department of Pharmacy, LEO Foundation Center for Cutaneous Drug Delivery, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
43
|
Töpfer U, Ryu J, Guerra Santillán KY, Schulze J, Fischer-Friedrich E, Tanentzapf G, Dahmann C. AdamTS proteases control basement membrane heterogeneity and organ shape in Drosophila. Cell Rep 2024; 43:114399. [PMID: 38944833 DOI: 10.1016/j.celrep.2024.114399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/07/2024] [Accepted: 06/10/2024] [Indexed: 07/02/2024] Open
Abstract
The basement membrane (BM) is an extracellular matrix that plays important roles in animal development. A spatial heterogeneity in composition and structural properties of the BM provide cells with vital cues for morphogenetic processes such as cell migration or cell polarization. Here, using the Drosophila egg chamber as a model system, we show that the BM becomes heterogeneous during development, with a reduction in Collagen IV density at the posterior pole and differences in the micropattern of aligned fiber-like structures. We identified two AdamTS matrix proteases required for the proper elongated shape of the egg chamber, yet the molecular mechanisms by which they act are different. Stall is required to establish BM heterogeneity by locally limiting Collagen IV protein density, whereas AdamTS-A alters the micropattern of fiber-like structures within the BM at the posterior pole. Our results suggest that AdamTS proteases control BM heterogeneity required for organ shape.
Collapse
Affiliation(s)
- Uwe Töpfer
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; School of Science, Technische Universität Dresden, 01062 Dresden, Germany.
| | - Jinhee Ryu
- School of Science, Technische Universität Dresden, 01062 Dresden, Germany
| | - Karla Yanín Guerra Santillán
- School of Science, Technische Universität Dresden, 01062 Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062 Dresden, Germany
| | - Jana Schulze
- School of Science, Technische Universität Dresden, 01062 Dresden, Germany
| | - Elisabeth Fischer-Friedrich
- Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062 Dresden, Germany; Biotechnology Center, Technische Universität Dresden, 01062 Dresden, Germany
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Christian Dahmann
- School of Science, Technische Universität Dresden, 01062 Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062 Dresden, Germany.
| |
Collapse
|
44
|
Wang L, He L, Yi W, Wang M, Xu F, Liu H, Nie J, Pan YH, Dang S, Zhang W. ADAMTS18-fibronectin interaction regulates the morphology of liver sinusoidal endothelial cells. iScience 2024; 27:110273. [PMID: 39040056 PMCID: PMC11261151 DOI: 10.1016/j.isci.2024.110273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/12/2024] [Accepted: 06/12/2024] [Indexed: 07/24/2024] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) have a unique morphological structure known as "fenestra" that plays a crucial role in liver substance exchange and homeostasis maintenance. In this study, we demonstrate that ADAMTS18 protease is primarily secreted by fetal liver endothelial cells. ADAMTS18 deficiency leads to enlarged fenestrae and increased porosity of LSECs, microthrombus formation in liver vessels, and an imbalance of liver oxidative stress. These defects worsen carbon tetrachloride (CCl4)-induced liver fibrosis and diethylnitrosamine (DEN)/high-fat-induced hepatocellular carcinoma (HCC) in adult Adamts18-deficient mice. Mechanically, ADAMTS18 functions as a modifier of fibronectin (FN) to regulate the morphological acquisition of LSECs via the vascular endothelial growth factor A (VEGFA) signaling pathways. Collectively, a mechanism is proposed for LSEC morphogenesis and liver homeostasis maintenance via ADAMTS18-FN-VEGFA niches.
Collapse
Affiliation(s)
- Liya Wang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Li He
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Weijia Yi
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Min Wang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Fangmin Xu
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Hanlin Liu
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Jiahui Nie
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Yi-Hsuan Pan
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Suying Dang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zhang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
45
|
Shibata Y, Huang Y, Yoshida M, Nishiwaki K. Mutations in fibulin-1 and collagen IV suppress the short healthspan of mig-17/ADAMTS mutants in Caenorhabditis elegans. PLoS One 2024; 19:e0305396. [PMID: 38980840 PMCID: PMC11232982 DOI: 10.1371/journal.pone.0305396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/29/2024] [Indexed: 07/11/2024] Open
Abstract
The ADAMTS (a disintegrin and metalloprotease with thrombospondin motifs) family metalloprotease MIG-17 plays a crucial role in the migration of gonadal distal tip cells (DTCs) in Caenorhabditis elegans. MIG-17 is secreted from the body wall muscle cells and localizes to the basement membranes (BMs) of various tissues including the gonadal BM where it regulates DTC migration through its catalytic activity. Missense mutations in the BM protein genes, let-2/collagen IV a2 and fbl-1/fibulin-1, have been identified as suppressors of the gonadal defects observed in mig-17 mutants. Genetic analyses indicate that LET-2 and FBL-1 act downstream of MIG-17 to regulate DTC migration. In addition to the control of DTC migration, MIG-17 also plays a role in healthspan, but not in lifespan. Here, we examined whether let-2 and fbl-1 alleles can suppress the age-related phenotypes of mig-17 mutants. let-2(k196) fully and fbl-1(k201) partly, but not let-2(k193) and fbl-1(k206), suppressed the senescence defects of mig-17. Interestingly, fbl-1(k206), but not fbl-1(k201) or let-2 alleles, exhibited an extended lifespan compared to the wild type when combined with mig-17. These results reveal allele specific interactions between let-2 or fbl-1 and mig-17 in age-related phenotypes, indicating that basement membrane physiology plays an important role in organismal aging.
Collapse
Affiliation(s)
- Yukimasa Shibata
- Department of Bioscience, Kwansei Gakuin University, Sanda, Japan
| | - Yijing Huang
- Department of Bioscience, Kwansei Gakuin University, Sanda, Japan
| | - Moeka Yoshida
- Department of Bioscience, Kwansei Gakuin University, Sanda, Japan
| | - Kiyoji Nishiwaki
- Department of Bioscience, Kwansei Gakuin University, Sanda, Japan
| |
Collapse
|
46
|
Revert-Ros F, Ventura I, Prieto-Ruiz JA, Hernández-Andreu JM, Revert F. The Versatility of Collagen in Pharmacology: Targeting Collagen, Targeting with Collagen. Int J Mol Sci 2024; 25:6523. [PMID: 38928229 PMCID: PMC11203716 DOI: 10.3390/ijms25126523] [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: 05/08/2024] [Revised: 06/01/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Collagen, a versatile family of proteins with 28 members and 44 genes, is pivotal in maintaining tissue integrity and function. It plays a crucial role in physiological processes like wound healing, hemostasis, and pathological conditions such as fibrosis and cancer. Collagen is a target in these processes. Direct methods for collagen modulation include enzymatic breakdown and molecular binding approaches. For instance, Clostridium histolyticum collagenase is effective in treating localized fibrosis. Polypeptides like collagen-binding domains offer promising avenues for tumor-specific immunotherapy and drug delivery. Indirect targeting of collagen involves regulating cellular processes essential for its synthesis and maturation, such as translation regulation and microRNA activity. Enzymes involved in collagen modification, such as prolyl-hydroxylases or lysyl-oxidases, are also indirect therapeutic targets. From another perspective, collagen is also a natural source of drugs. Enzymatic degradation of collagen generates bioactive fragments known as matrikines and matricryptins, which exhibit diverse pharmacological activities. Overall, collagen-derived peptides present significant therapeutic potential beyond tissue repair, offering various strategies for treating fibrosis, cancer, and genetic disorders. Continued research into specific collagen targeting and the application of collagen and its derivatives may lead to the development of novel treatments for a range of pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | - Fernando Revert
- Mitochondrial and Molecular Medicine Research Group, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain; (F.R.-R.); (I.V.); (J.A.P.-R.); (J.M.H.-A.)
| |
Collapse
|
47
|
Radisky ES. Extracellular proteolysis in cancer: Proteases, substrates, and mechanisms in tumor progression and metastasis. J Biol Chem 2024; 300:107347. [PMID: 38718867 PMCID: PMC11170211 DOI: 10.1016/j.jbc.2024.107347] [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: 02/09/2024] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 06/02/2024] Open
Abstract
A vast ensemble of extracellular proteins influences the development and progression of cancer, shaped and reshaped by a complex network of extracellular proteases. These proteases, belonging to the distinct classes of metalloproteases, serine proteases, cysteine proteases, and aspartic proteases, play a critical role in cancer. They often become dysregulated in cancer, with increases in pathological protease activity frequently driven by the loss of normal latency controls, diminished regulation by endogenous protease inhibitors, and changes in localization. Dysregulated proteases accelerate tumor progression and metastasis by degrading protein barriers within the extracellular matrix (ECM), stimulating tumor growth, reactivating dormant tumor cells, facilitating tumor cell escape from immune surveillance, and shifting stromal cells toward cancer-promoting behaviors through the precise proteolysis of specific substrates to alter their functions. These crucial substrates include ECM proteins and proteoglycans, soluble proteins secreted by tumor and stromal cells, and extracellular domains of cell surface proteins, including membrane receptors and adhesion proteins. The complexity of the extracellular protease web presents a significant challenge to untangle. Nevertheless, technological strides in proteomics, chemical biology, and the development of new probes and reagents are enabling progress and advancing our understanding of the pivotal importance of extracellular proteolysis in cancer.
Collapse
Affiliation(s)
- Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida, USA.
| |
Collapse
|
48
|
Qi H, Tian D, Luan F, Yang R, Zeng N. Pathophysiological changes of muscle after ischemic stroke: a secondary consequence of stroke injury. Neural Regen Res 2024; 19:737-746. [PMID: 37843207 PMCID: PMC10664100 DOI: 10.4103/1673-5374.382221] [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: 01/06/2023] [Revised: 03/30/2023] [Accepted: 06/01/2023] [Indexed: 10/17/2023] Open
Abstract
Sufficient clinical evidence suggests that the damage caused by ischemic stroke to the body occurs not only in the acute phase but also during the recovery period, and that the latter has a greater impact on the long-term prognosis of the patient. However, current stroke studies have typically focused only on lesions in the central nervous system, ignoring secondary damage caused by this disease. Such a phenomenon arises from the slow progress of pathophysiological studies examining the central nervous system. Further, the appropriate therapeutic time window and benefits of thrombolytic therapy are still controversial, leading scholars to explore more pragmatic intervention strategies. As treatment measures targeting limb symptoms can greatly improve a patient's quality of life, they have become a critical intervention strategy. As the most vital component of the limbs, skeletal muscles have become potential points of concern. Despite this, to the best of our knowledge, there are no comprehensive reviews of pathophysiological changes and potential treatments for post-stroke skeletal muscle. The current review seeks to fill a gap in the current understanding of the pathological processes and mechanisms of muscle wasting atrophy, inflammation, neuroregeneration, mitochondrial changes, and nutritional dysregulation in stroke survivors. In addition, the challenges, as well as the optional solutions for individualized rehabilitation programs for stroke patients based on motor function are discussed.
Collapse
Affiliation(s)
- Hu Qi
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Dan Tian
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Fei Luan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Ruocong Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Nan Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| |
Collapse
|
49
|
Nian J, Zhu Y, Lv X, Zhang Y, Xue Z, Wu Z, Zhou Y, Liu Y. Expression levels of ADAMTS 5, 9, and 12 in endometrial polyps and their predictive value for the diagnosis and recurrence of endometrial polyps. Eur J Obstet Gynecol Reprod Biol 2024; 295:86-91. [PMID: 38340595 DOI: 10.1016/j.ejogrb.2024.02.008] [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: 12/25/2023] [Revised: 01/30/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024]
Abstract
PURPOSE Endometrial polyps (EPs) are common gynecological disorders for which no clear etiology has been found. ADAMTS have been associated with a variety of diseases. This study aimed to investigate the potential correlation between serologic levels of ADAMTS 5, 9, and 12 in patients with EPs. METHODS A total of 88 patients were categorized into two groups: the EPs group, consisting of recurrent EPs and first occurrence EPs, and a control group. The study compared the general information and serum levels of ADAMTS 5, 9, and 12 between the groups. RESULTS Regarding the general data, a statistically significant age difference (p < 0.05) was observed, while no significant differences were found in the other variables. After considering age as a confounding factor, the previously observed statistical significance in the differences of ADAMTS5 and 9 between the groups diminished. However, it was found that the concentrations of ADAMTS12 in both the EPs group and the recurrent EPs group were significantly higher compared to the control group and the first occurrence EPs group (p < 0.05). ROC curves were generated to determine the critical values of ADAMTS12 for predicting EPs and recurrent EPs, which were found to be 0.6962 ng/ml (sensitivity: 100 %, specificity: 39.5 %) and 0.8768 ng/ml (sensitivity: 75.0 %, specificity: 76.3 %), respectively. CONCLUSION Our findings revealed elevated serologic levels of ADAMTS12 in the EPs group, particularly in the recurrent EPs group. Furthermore, ADAMTS-12 was identified as a valuable biomarker for assisting in the diagnosis and prediction of EPs recurrence.
Collapse
Affiliation(s)
- Jiejie Nian
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Hefei Maternal and Child Health Hospital, Hefei, Anhui 230001, China; Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, Anhui 230032, China
| | - Yuqing Zhu
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Hefei Maternal and Child Health Hospital, Hefei, Anhui 230001, China; Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, Anhui 230032, China
| | - Xiaoli Lv
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Hefei Maternal and Child Health Hospital, Hefei, Anhui 230001, China; Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, Anhui 230032, China
| | - Yue Zhang
- Department of Gynecology and Obstetrics, Xuancheng City People's Hospital, Xuancheng, Anhui, 242000, China
| | - Zhongkai Xue
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Hefei Maternal and Child Health Hospital, Hefei, Anhui 230001, China; Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, Anhui 230032, China
| | - Zhongran Wu
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Hefei Maternal and Child Health Hospital, Hefei, Anhui 230001, China
| | - Yujia Zhou
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Hefei Maternal and Child Health Hospital, Hefei, Anhui 230001, China
| | - Yu Liu
- Department of Gynecology and Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Hefei Maternal and Child Health Hospital, Hefei, Anhui 230001, China; Department of Gynecology and Obstetrics, The Fifth Clinical College of Anhui Medical University, Hefei, Anhui 230032, China.
| |
Collapse
|
50
|
Chang MY, Chan CK, Brune JE, Manicone AM, Bomsztyk K, Frevert CW, Altemeier WA. Regulation of Versican Expression in Macrophages is Mediated by Canonical Type I Interferon Signaling via ISGF3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.14.585097. [PMID: 38559011 PMCID: PMC10980001 DOI: 10.1101/2024.03.14.585097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Growing evidence supports a role for versican as an important component of the inflammatory response, with both pro- and anti-inflammatory roles depending on the specific context of the system or disease under investigation. Our goal is to understand the regulation of macrophage-derived versican and the role it plays in innate immunity. In previous work, we showed that LPS triggers a signaling cascade involving TLR4, the Trif adaptor, type I interferons, and the type I interferon receptor, leading to increased versican expression by macrophages. In the present study, we used a combination of chromatin immunoprecipitation, siRNA, chemical inhibitors, and mouse model approaches to investigate the regulatory events downstream of the type I interferon receptor to better define the mechanism controlling versican expression. Results indicate that transcriptional regulation by canonical type I interferon signaling via the heterotrimeric transcription factor, ISGF3, controls versican expression in macrophages exposed to LPS. This pathway is not dependent on MAPK signaling, which has been shown to regulate versican expression in other cell types. The stability of versican mRNA may also contribute to prolonged versican expression in macrophages. These findings strongly support a role for macrophage-derived versican as a type I interferon-stimulated gene and further our understanding of versican's role in regulating inflammation.
Collapse
Affiliation(s)
- Mary Y. Chang
- Department of Comparative Medicine, University of Washington, Seattle, WA
- Center for Lung Biology, University of Washington at South Lake Union, Seattle, WA
| | - Christina K. Chan
- Department of Comparative Medicine, University of Washington, Seattle, WA
- Center for Lung Biology, University of Washington at South Lake Union, Seattle, WA
| | - Jourdan E. Brune
- Department of Comparative Medicine, University of Washington, Seattle, WA
- Center for Lung Biology, University of Washington at South Lake Union, Seattle, WA
| | - Anne M. Manicone
- Center for Lung Biology, University of Washington at South Lake Union, Seattle, WA
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA
| | - Karol Bomsztyk
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA
| | - Charles W. Frevert
- Department of Comparative Medicine, University of Washington, Seattle, WA
- Center for Lung Biology, University of Washington at South Lake Union, Seattle, WA
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA
| | - William A. Altemeier
- Center for Lung Biology, University of Washington at South Lake Union, Seattle, WA
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|