1
|
Zhou Q, Lei L, Cheng J, Chen J, Du Y, Zhang X, Li Q, Li C, Deng H, Wong CC, Zhuang B, Li G, Bai X. Microbiota-induced S100A11-RAGE axis underlies immune evasion in right-sided colon adenomas and is a therapeutic target to boost anti-PD1 efficacy. Gut 2025; 74:214-228. [PMID: 39251326 DOI: 10.1136/gutjnl-2024-332193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 08/19/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Tumourigenesis in right-sided and left-sided colons demonstrated distinct features. OBJECTIVE We aimed to characterise the differences between the left-sided and right-sided adenomas (ADs) representing the early stage of colonic tumourigenesis. DESIGN Single-cell and spatial transcriptomic datasets were analysed to reveal alterations between right-sided and left-sided colon ADs. Cells, animal experiments and clinical specimens were used to verify the results. RESULTS Single-cell analysis revealed that in right-sided ADs, there was a significant reduction of goblet cells, and these goblet cells were dysfunctional with attenuated mucin biosynthesis and defective antigen presentation. An impairment of the mucus barrier led to biofilm formation in crypts and subsequent bacteria invasion into right-sided ADs. The regions spatially surrounding the crypts with biofilm occupation underwent an inflammatory response by lipopolysaccharide (LPS) and an apoptosis process, as revealed by spatial transcriptomics. A distinct S100A11+ epithelial cell population in the right-sided ADs was identified, and its expression level was induced by bacterial LPS and peptidoglycan. S100A11 expression facilitated tumour growth in syngeneic immunocompetent mice with increased myeloid-derived suppressor cells (MDSC) but reduced cytotoxic CD8+ T cells. Targeting S100A11 with well-tolerated antagonists of its receptor for advanced glycation end product (RAGE) (Azeliragon) significantly impaired tumour growth and MDSC infiltration, thereby boosting the efficacy of anti-programmed cell death protein 1 therapy in colon cancer. CONCLUSION Our findings unravelled that dysfunctional goblet cells and consequential bacterial translocation activated the S100A11-RAGE axis in right-sided colon ADs, which recruits MDSCs to promote immune evasion. Targeting this axis by Azeliragon improves the efficacy of immunotherapy in colon cancer.
Collapse
Affiliation(s)
- Qiming Zhou
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Linhan Lei
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junhong Cheng
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junyou Chen
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuyang Du
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xuehua Zhang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qing Li
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuangen Li
- Institute of Chinese Medical Sciences, University of Macao, Taipa, Macao
| | - Haijun Deng
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chi Chun Wong
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Baoxiong Zhuang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guoxin Li
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Xiaowu Bai
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Yin L, Huang X, Zhang B, Zhu Q, Zhao H. Narciclasine attenuates sepsis-associated acute kidney injury through the ESR1/S100A11 axis. Funct Integr Genomics 2025; 25:13. [PMID: 39808340 DOI: 10.1007/s10142-024-01513-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/15/2024] [Accepted: 12/25/2024] [Indexed: 01/16/2025]
Abstract
Narciclasine (Ncs) was effective in sepsis management due to its antioxidant properties. The present study dissected the protective effects of Ncs against sepsis-associated acute kidney injury (SA-AKI) and the molecular mechanisms. The SA-AKI mice were developed using cecum ligation and puncture and pretreated with Ncs and adenoviruses. Human renal microvascular endothelial cells (RMECs) were induced with LPS and treated with Ncs. Ncs alleviated proximal tubular dilatation, interstitial widening, and necrosis in renal tissues and reduced the renal injury marker and pro-inflammatory cytokine levels in the serum of SA-AKI mice. Ncs promoted the expression of ZO-1, VE-cadherin, and CD31 and the activities of SOD, GSH-Px, and CAT, and inhibited the levels of pro-inflammatory cytokines, and apoptosis rate in LPS-treated RMECs. Estrogen receptor 1 (ESR1) was a target protein of Ncs, and S100 calcium-binding protein A11 (S100A11) was a target of the transcription factor ESR1. Ncs blocked transcription of S100A11 by inhibiting ESR1. Silencing of S100A11 overturned the deteriorating effects of ESR1 overexpression on SA-AKI progression in vivo and RMEC injury in vitro. These findings suggest that Ncs may ameliorate SA-AKI by repressing the ESR1/S100A11 signaling, providing a novel perspective for research on SA-AKI.
Collapse
Affiliation(s)
- Liping Yin
- Department of Emergency and Critical Care Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China
- Department of Emergency and Critical Care Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China
| | - Xiaofei Huang
- Department of Emergency and Critical Care Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China
- Department of Emergency and Critical Care Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China
| | - Beibei Zhang
- Department of Emergency and Critical Care Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China
- Department of Emergency and Critical Care Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China
| | - Qiyong Zhu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China
| | - Hui Zhao
- Department of Emergency and Critical Care Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China.
- Department of Emergency and Critical Care Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, People's Republic of China.
| |
Collapse
|
3
|
Chen G, Qi H, Jiang L, Sun S, Zhang J, Yu J, Liu F, Zhang Y, Du S. Integrating single-cell RNA-Seq and machine learning to dissect tryptophan metabolism in ulcerative colitis. J Transl Med 2024; 22:1121. [PMID: 39707393 DOI: 10.1186/s12967-024-05934-w] [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: 07/30/2024] [Accepted: 12/01/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is a persistent inflammatory bowels disease (IBD) characterized by immune response dysregulation and metabolic disruptions. Tryptophan metabolism has been believed as a significant factor in UC pathogenesis, with specific metabolites influencing immune modulation and gut microbiota interactions. However, the precise regulatory mechanisms and key genes involved remain unclear. METHODS AUCell, Ucell, and other functional enrichment algorithms were utilized to determine the activation patterns of tryptophan metabolism at the UC cell level. Differential analysis identified key genes associated with tryptophan metabolism. Five machine learning algorithms, including Random Forest, Boruta algorithm, LASSO, SVM-RFE, and GBM were integrated to identify and categorize disease-specific characteristic genes. RESULTS We observed significant heterogeneity in tryptophan metabolism activity across cell types in UC, with the highest activity levels in macrophages and fibroblasts. Among the key tryptophan metabolism-related genes, CTSS, S100A11, and TUBB were predominantly expressed in macrophages and significantly upregulated in UC, highlighting their involvement in immune dysregulation and inflammation. Cross-analysis with bulk RNA data confirmed the consistent upregulation of these genes in UC samples, highly indicating their relevance in UC pathology and potential as targets for therapeutic intervention. CONCLUSIONS This study is the first to reveal the heterogeneity of tryptophan metabolism at the single-cell level in UC, with macrophages emerging as key contributors to inflammatory processes. The identification of CTSS, S100A11, and TUBB as key regulators of tryptophan metabolism in UC underscores their potential as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Guorong Chen
- Department of Gastroenterology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, 100029, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Hongying Qi
- Department of Spleen and Stomach Diseases of Traditional Chinese Medicine, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, 100029, China
| | - Li Jiang
- Department of Endocrinology, Aviation General Hospital, Beijing, 100025, China
| | - Shijie Sun
- Department of Gastroenterology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, 100029, China
| | - Junhai Zhang
- Department of Gastroenterology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, 100029, China
| | - Jiali Yu
- Department of Gastroenterology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, 100029, China
| | - Fang Liu
- Department of Gastroenterology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, 100029, China
| | - Yanli Zhang
- Department of Gastroenterology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, 100029, China.
| | - Shiyu Du
- Department of Gastroenterology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, 100029, China.
| |
Collapse
|
4
|
Praveen J, Anusuyadevi M, Jayachandra KS. Unraveling the potential of Epicatechin gallate from crataegus oxyacantha in targeting aberrant cardiac Ca2+ signalling proteins: an in-depth in-silico investigation for heart failure therapy. J Biomol Struct Dyn 2024:1-15. [PMID: 39648361 DOI: 10.1080/07391102.2024.2435624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 03/25/2024] [Indexed: 12/10/2024]
Abstract
The cardiovascular sarcoplasmic reticulum (SR) calcium (Ca2+) ATPase is an imperative determinant of cardiac functionality. In addition, anomalies in Ca2+ handling protein and atypical energy metabolism are inherent in heart failure (HF). Moreover, Ca2+ overload in SR leads to mitochondrial matrix Ca2+ overload, which can trigger the generation of Reactive Oxygen Species (ROS), culminating in the triggering of the Permeability Transition Pore (PTP) and Cytochrome C release, resulting in apoptosis that leads to arrhythmias and numerous disorders. Although proteins involved in the molecular mechanism of Ca2+ dysfunction regarding mitochondrial dysfunction remains elusive, this study aims to assess the major Ca2+ handling proteins which may be involved in the Ca2+ malfunction that causes mitochondrial dysfunction and predicting the most effective drug by targeting the analyzed Ca2+ handling proteins through various insilico analyses. Thirteen proteins absorbed from interaction analysis were docked with four optimal phytochemicals from Crataegus oxyacantha (COC). Furthermore, The ADME profile of tyramine, vitexin, Epicatechin, and Epicatechin gallate was acclimated to evaluate potential drugability utilizing QikProp. So, molecular docking evaluations were performed using Glide (Maestro), autodock, and vina. Based on the results of 156 dockings by Maestro, auto-dock, and auto-dock vina, PKAC-a with Epicatechin gallate exhibits good interaction. Therefore, a 2000 ns molecular dynamics (MD) simulation was utilized to assess the feasible phytochemical Epicatechin gallate - PKAC-a complex binding stability utilizing Desmond and this study confirmed that Epicatechin gallate from COC has high possibilities to inhibit the aberrant cardiac Ca2+ signaling proteins due to its conformational rigidity.
Collapse
Affiliation(s)
- J Praveen
- Department of Bioinformatics, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | - M Anusuyadevi
- Department of Biochemistry, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| | - K S Jayachandra
- Department of Bioinformatics, Bharathidasan University, Tiruchirappalli, Tamilnadu, India
| |
Collapse
|
5
|
Zhou J, Murata H, Tomonobu N, Mizuta N, Yamakawa A, Yamamoto KI, Kinoshita R, Sakaguchi M. S100A11 is involved in the progression of colorectal cancer through the desmosome-catenin-TCF signaling pathway. In Vitro Cell Dev Biol Anim 2024; 60:1138-1149. [PMID: 38842658 PMCID: PMC11655616 DOI: 10.1007/s11626-024-00930-2] [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: 03/06/2024] [Accepted: 05/16/2024] [Indexed: 06/07/2024]
Abstract
Compiling evidence has indicated that S100A11 expression at high levels is closely associated with various cancer species. Consistent with the results reported elsewhere, we have also revealed that S100A11 is highly expressed in squamous cell carcinoma, mesothelioma, and pancreatic cancers and plays a crucial role in cancer progression when secreted into extracellular fluid. Those studies are all focused on the extracellular role of S100A11. However, most of S100A11 is still present within cancer cells, although the intracellular role of S100A11 in cancer cells has not been fully elucidated. Thus, we aimed to investigate S100A11 functions within cancer cells, primarily focusing on colorectal cancer cells, whose S100A11 is abundantly present in cells and still poorly studied cancer for the protein. Our efforts revealed that overexpression of S100A11 promotes proliferation and migration, and downregulation inversely dampens those cancer behaviors. To clarify how intracellular S100A11 aids cancer cell activation, we tried to identify S100A11 binding proteins, resulting in novel binding partners in the inner membrane, many of which are desmosome proteins. Our molecular approach defined that S100A11 regulates the expression level of DSG1, a component protein of desmosome, by which S100A11 activates the TCF pathway via promoting nuclear translocation of γ-catenin from the desmosome. The identified new pathway greatly helps to comprehend S100A11's nature in colorectal cancers and others.
Collapse
Affiliation(s)
- Jin Zhou
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
- Medical Oncology Department of Gastrointestinal Tumors, Liaoning Cancer Hospital & Institute, Cancer Hospital of the Dalian University of Technology, Shenyang, Liaoning, China
| | - Hitoshi Murata
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan.
| | - Nahoko Tomonobu
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Naoko Mizuta
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Atsuko Yamakawa
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Ken-Ichi Yamamoto
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Rie Kinoshita
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Masakiyo Sakaguchi
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| |
Collapse
|
6
|
Kapic A, Zaman K, Nguyen V, Prokai-Tatrai K, Prokai L. Identification of Estrogen-Responsive Proteins in Mouse Seminal Vesicles Through Mass Spectrometry-Based Proteomics. Pharmaceuticals (Basel) 2024; 17:1508. [PMID: 39598420 PMCID: PMC11597337 DOI: 10.3390/ph17111508] [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: 09/29/2024] [Revised: 11/02/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Although estrogenic compounds promise therapeutic potential in treating various conditions, concerns regarding their endocrine-disrupting effects have been raised. Current methodologies for screening estrogenicity in rodent models are limited to the female-specific uterotrophic bioassay. Studies have reported enlargement of the seminal vesicles in orchiectomized males treated with estrogens. However, identifying estrogenicity strictly through changes in wet weights is uninformative regarding the molecular mechanisms of these agents. Therefore, protein-based biomarkers can complement and improve the sensitivity of weight-based assessments. To this end, we present a discovery-driven proteomic analysis of 17β-estradiol's effects on the seminal vesicles. Methods: We treated orchidectomized mice with the hormone for five days and used the vehicle-treated group as a control. Seminal vesicles were analyzed by shotgun approach using data-dependent nanoflow liquid chromatography-tandem mass spectrometry and label-free quantification. Proteins found to be differentially expressed between the two groups were processed through a bioinformatics pipeline focusing on pathway analyses and assembly of protein interaction networks. Results: Out of 668 identified proteins that passed rigorous validation criteria, 133 were regulated significantly by 17β-estradiol. Ingenuity Pathway Analysis® linked them to several hormone-affected pathways, including those associated with immune function such as neutrophil degranulation. The altered protein interaction networks were also related to functions including endocrine disruption, abnormal metabolism, and therapeutic effects. Conclusions: We identified several potential biomarkers for estrogenicity in mouse seminal vesicles, many of them not previously linked with exogenous 17β-estradiol exposure.
Collapse
Affiliation(s)
| | | | | | | | - Laszlo Prokai
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (A.K.); (K.Z.); (V.N.); (K.P.-T.)
| |
Collapse
|
7
|
Lépine M, Robert MC, Sleno L. Discovery and Verification of Sjögren's Syndrome Protein Biomarkers in Tears by Targeted LC-MRM. J Proteome Res 2024. [PMID: 38682820 DOI: 10.1021/acs.jproteome.4c00163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Sjögren's syndrome (SS) is an autoimmune rheumatic disorder characterized by exocrine gland dysfunction, mainly from the lacrimal and salivary glands. The disease causes severe aqueous dry eye syndrome (DED) and is associated with high rates of complications, including corneal ulceration, scaring, and perforation. Systemic complications may occur as well as a higher risk of developing lymphoma. Diagnosis of SS-DED is often delayed and difficult to establish. With the aim of discovering biomarkers to help discriminate SS-DED patients, a combination of untargeted and targeted LC-MS/MS analyses were performed on tear samples collected on Schirmer strips and subjected to tryptic digestion. Following the analysis of three cohorts and the development of two targeted LC-sMRM methods for the verification of putative biomarkers found in the first cohort of samples, 64 proteins could be linked to Sjögren's syndrome, in the hopes of helping to confirm diagnoses as well as potentially stratifying the severity of disease in these patients. Proteins that were increased in SS-DED showed activation of the immune system and alterations in homeostasis. Several proteases and protease inhibitors were found to be significantly changing in SS-DED, as well as a consistent decrease in specific proteins known to be secreted by the lacrimal gland.
Collapse
Affiliation(s)
- Maggy Lépine
- University of Quebec in Montreal (UQAM), Chemistry Department, PO Box 8888, Downtown Station, Montreal, Quebec H3C 3P8, Canada
- CERMO-FC, Centre d'Excellence de Recherche sur les Maladies Orphelines-Fondation Courtois, 141 Avenue du President Kennedy, Montreal, Quebec H2X 3Y7, Canada
| | - Marie-Claude Robert
- Centre de Recherche du Centre Hospitalier Universitaire de (CR-CHUM), Ophthalmology Department, 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
- CERMO-FC, Centre d'Excellence de Recherche sur les Maladies Orphelines-Fondation Courtois, 141 Avenue du President Kennedy, Montreal, Quebec H2X 3Y7, Canada
| | - Lekha Sleno
- University of Quebec in Montreal (UQAM), Chemistry Department, PO Box 8888, Downtown Station, Montreal, Quebec H3C 3P8, Canada
- CERMO-FC, Centre d'Excellence de Recherche sur les Maladies Orphelines-Fondation Courtois, 141 Avenue du President Kennedy, Montreal, Quebec H2X 3Y7, Canada
| |
Collapse
|
8
|
Deepu V, Rai V, Agrawal DK. Quantitative Assessment of Intracellular Effectors and Cellular Response in RAGE Activation. ARCHIVES OF INTERNAL MEDICINE RESEARCH 2024; 7:80-103. [PMID: 38784044 PMCID: PMC11113086 DOI: 10.26502/aimr.0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
The review delves into the methods for the quantitative assessment of intracellular effectors and cellular response of Receptor for Advanced Glycation End products (RAGE), a vital transmembrane receptor involved in a range of physiological and pathological processes. RAGE bind to Advanced Glycation End products (AGEs) and other ligands, which in turn activate diverse downstream signaling pathways that impact cellular responses such as inflammation, oxidative stress, and immune reactions. The review article discusses the intracellular signaling pathways activated by RAGE followed by differential activation of RAGE signaling across various diseases. This will ultimately guide researchers in developing targeted and effective interventions for diseases associated with RAGE activation. Further, we have discussed how PCR, western blotting, and microscopic examination of various molecules involved in downstream signaling can be leveraged to monitor, diagnose, and explore diseases involving proteins with unique post-translational modifications. This review article underscores the pressing need for advancements in molecular approaches for disease detection and management involving RAGE.
Collapse
Affiliation(s)
- Vinitha Deepu
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91763, USA
| | - Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91763, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91763, USA
| |
Collapse
|
9
|
Melle C, Hoffmann B, Wiesenburg A, Biskup C. FLIM-FRET-based analysis of S100A11/annexin interactions in living cells. FEBS Open Bio 2024; 14:626-642. [PMID: 38408765 PMCID: PMC10988696 DOI: 10.1002/2211-5463.13782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 02/28/2024] Open
Abstract
Proteins achieve their biological functions in cells by cooperation in protein complexes. In this study, we employed fluorescence lifetime imaging microscopy (FLIM)-based Förster resonance energy transfer (FRET) measurements to investigate protein complexes comprising S100A11 and different members of the annexin (ANX) family, such as ANXA1, ANXA2, ANXA4, ANXA5, and AnxA6, in living cells. Using an S100A11 mutant without the capacity for Ca2+ binding, we found that Ca2+ binding of S100A11 is important for distinct S100A11/ANXA2 complex formation; however, ANXA1-containing complexes were unaffected by this mutant. An increase in the intracellular calcium concentration induced calcium ionophores, which strengthened the ANXA2/S100A11 interaction. Furthermore, we were able to show that S100A11 also interacts with ANXA4 in living cells. The FLIM-FRET approach used here can serve as a tool to analyze interactions between S100A11 and distinct annexins under physiological conditions in living cells.
Collapse
Affiliation(s)
- Christian Melle
- Biomolecular Photonics Group, Jena University HospitalFriedrich Schiller University JenaGermany
| | - Birgit Hoffmann
- Biomolecular Photonics Group, Jena University HospitalFriedrich Schiller University JenaGermany
| | - Annett Wiesenburg
- Biomolecular Photonics Group, Jena University HospitalFriedrich Schiller University JenaGermany
| | - Christoph Biskup
- Biomolecular Photonics Group, Jena University HospitalFriedrich Schiller University JenaGermany
| |
Collapse
|
10
|
Mattisson J, Halvardson J, Davies H, Bruhn-Olszewska B, Olszewski P, Danielsson M, Bjurling J, Lindberg A, Zaghlool A, Rychlicka-Buniowska E, Dumanski JP, Forsberg LA. Loss of chromosome Y in regulatory T cells. BMC Genomics 2024; 25:243. [PMID: 38443832 PMCID: PMC10913415 DOI: 10.1186/s12864-024-10168-7] [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: 07/27/2023] [Accepted: 02/28/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Mosaic loss of chromosome Y (LOY) in leukocytes is the most prevalent somatic aneuploidy in aging humans. Men with LOY have increased risks of all-cause mortality and the major causes of death, including many forms of cancer. It has been suggested that the association between LOY and disease risk depends on what type of leukocyte is affected with Y loss, with prostate cancer patients showing higher levels of LOY in CD4 + T lymphocytes. In previous studies, Y loss has however been observed at relatively low levels in this cell type. This motivated us to investigate whether specific subsets of CD4 + T lymphocytes are particularly affected by LOY. Publicly available, T lymphocyte enriched, single-cell RNA sequencing datasets from patients with liver, lung or colorectal cancer were used to study how LOY affects different subtypes of T lymphocyte. To validate the observations from the public data, we also generated a single-cell RNA sequencing dataset comprised of 23 PBMC samples and 32 CD4 + T lymphocytes enriched samples. RESULTS Regulatory T cells had significantly more LOY than any other studied T lymphocytes subtype. Furthermore, LOY in regulatory T cells increased the ratio of regulatory T cells compared with other T lymphocyte subtypes, indicating an effect of Y loss on lymphocyte differentiation. This was supported by developmental trajectory analysis of CD4 + T lymphocytes culminating in the regulatory T cells cluster most heavily affected by LOY. Finally, we identify dysregulation of 465 genes in regulatory T cells with Y loss, many involved in the immunosuppressive functions and development of regulatory T cells. CONCLUSIONS Here, we show that regulatory T cells are particularly affected by Y loss, resulting in an increased fraction of regulatory T cells and dysregulated immune functions. Considering that regulatory T cells plays a critical role in the process of immunosuppression; this enrichment for regulatory T cells with LOY might contribute to the increased risk for cancer observed among men with Y loss in leukocytes.
Collapse
Affiliation(s)
- Jonas Mattisson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jonatan Halvardson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Hanna Davies
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Bożena Bruhn-Olszewska
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Paweł Olszewski
- 3P-Medicine Laboratory, Medical University of Gdańsk, Gdańsk, Poland
| | - Marcus Danielsson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Josefin Bjurling
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Amanda Lindberg
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Ammar Zaghlool
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | - Jan P Dumanski
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- 3P-Medicine Laboratory, Medical University of Gdańsk, Gdańsk, Poland
| | - Lars A Forsberg
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- The Beijer Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
11
|
McCallum RT, Thériault RK, Manduca JD, Russell ISB, Culmer AM, Doost JS, Martino TA, Perreault ML. Nrf2 activation rescues stress-induced depression-like behaviour and inflammatory responses in male but not female rats. Biol Sex Differ 2024; 15:16. [PMID: 38350966 PMCID: PMC10863247 DOI: 10.1186/s13293-024-00589-0] [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: 11/26/2023] [Accepted: 01/31/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Major depressive disorder (MDD) is a recurring affective disorder that is two times more prevalent in females than males. Evidence supports immune system dysfunction as a major contributing factor to MDD, notably in a sexually dimorphic manner. Nuclear factor erythroid 2-related factor 2 (Nrf2), a regulator of antioxidant signalling during inflammation, is dysregulated in many chronic inflammatory disorders; however, its role in depression and the associated sex differences have yet to be explored. Here, we investigated the sex-specific antidepressant and immunomodulatory effects of the potent Nrf2 activator dimethyl fumarate (DMF), as well as the associated gene expression profiles. METHODS Male and female rats were treated with vehicle or DMF (25 mg/kg) whilst subjected to 8 weeks of chronic unpredictable stress. The effect of DMF treatment on stress-induced depression- and anxiety-like behaviours, as well as deficits in recognition and spatial learning and memory were then assessed. Sex differences in hippocampal (HIP) microglial activation and gene expression response were also evaluated. RESULTS DMF treatment during stress exposure had antidepressant effects in male but not female rats, with no anxiolytic effects in either sex. Recognition learning and memory and spatial learning and memory were impaired in chronically stressed males and females, respectively, and DMF treatment rescued these deficits. DMF treatment also prevented stress-induced HIP microglial activation in males. Conversely, females displayed no HIP microglial activation associated with stress exposure. Last, chronic stress elicited sex-specific alterations in HIP gene expression, many of which were normalized in animals treated with DMF. Of note, most of the differentially expressed genes in males normalized by DMF were related to antioxidant, inflammatory or immune responses. CONCLUSIONS Collectively, these findings support a greater role of immune processes in males than females in a rodent model of depression. This suggests that pharmacotherapies that target Nrf2 have the potential to be an effective sex-specific treatment for depression.
Collapse
Affiliation(s)
- Ryan T McCallum
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Rachel-Karson Thériault
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Joshua D Manduca
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Isaac S B Russell
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Angel M Culmer
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Janan Shoja Doost
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Tami A Martino
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Melissa L Perreault
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
12
|
Mohammed TO, Lin YR, Akter L, Weissenbruch K, Ngo KX, Zhang Y, Kodera N, Bastmeyer M, Miyanari Y, Taoka A, Franz CM. S100A11 promotes focal adhesion disassembly via myosin II-driven contractility and Piezo1-mediated Ca2+ entry. J Cell Sci 2024; 137:jcs261492. [PMID: 38277157 DOI: 10.1242/jcs.261492] [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: 07/18/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
S100A11 is a small Ca2+-activatable protein known to localize along stress fibers (SFs). Analyzing S100A11 localization in HeLa and U2OS cells further revealed S100A11 enrichment at focal adhesions (FAs). Strikingly, S100A11 levels at FAs increased sharply, yet transiently, just before FA disassembly. Elevating intracellular Ca2+ levels with ionomycin stimulated both S100A11 recruitment and subsequent FA disassembly. However, pre-incubation with the non-muscle myosin II (NMII) inhibitor blebbistatin or with an inhibitor of the stretch-activatable Ca2+ channel Piezo1 suppressed S100A11 recruitment, implicating S100A11 in an actomyosin-driven FA recruitment mechanism involving Piezo1-dependent Ca2+ influx. Applying external forces on peripheral FAs likewise recruited S100A11 to FAs even if NMII activity was inhibited, corroborating the mechanosensitive recruitment mechanism of S100A11. However, extracellular Ca2+ and Piezo1 function were indispensable, indicating that NMII contraction forces act upstream of Piezo1-mediated Ca2+ influx, in turn leading to S100A11 activation and FA recruitment. S100A11-knockout cells display enlarged FAs and had delayed FA disassembly during cell membrane retraction, consistent with impaired FA turnover in these cells. Our results thus demonstrate a novel function for S100A11 in promoting actomyosin contractility-driven FA disassembly.
Collapse
Affiliation(s)
- Tareg Omer Mohammed
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| | - You-Rong Lin
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Lucky Akter
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Kai Weissenbruch
- Cell and Neurobiology, Zoological Institute, Karlsruhe Institute of Technology, 76131, Karlsruhe, Germany
| | - Kien Xuan Ngo
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Yanjun Zhang
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Noriyuki Kodera
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Martin Bastmeyer
- Cell and Neurobiology, Zoological Institute, Karlsruhe Institute of Technology, 76131, Karlsruhe, Germany
- Institute for Biological and Chemical Systems - Biological Information Processing, Karlsruhe Institute of Technology, 76344, Eggenstein-Leopoldshafen, Germany
| | - Yusuke Miyanari
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa, 920-1192, Japan
- Cancer Research Institute, Kanazawa University, Kanazawa, 920-1162, Japan
| | - Azuma Taoka
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa, 920-1192, Japan
- Institute of Science and Engineering, Kanazawa University, Kanazawa, 920-1162, Japan
| | - Clemens M Franz
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| |
Collapse
|
13
|
He J, Lei Y, Li X, Wu B, Tang Y. Exploring the prognostic value of S100A11 and its association with immune infiltration in breast cancer. Sci Rep 2023; 13:22922. [PMID: 38129538 PMCID: PMC10739898 DOI: 10.1038/s41598-023-50160-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
Breast cancer (BC) is a severe danger to women's lives and health globally. S100A11 is aberrantly expressed in many carcinomas and serves a crucial function in cancer development. However, the role of S100A11 in BC is unclear. In this study, we utilized multiple databases and online tools, including the TCGA database, cBioPortal, and STRING, to evaluate the significance of S100A11 in BC prognosis and immune infiltration. We found that S100A11 was considerably more abundant in BC tissues. Survival analysis indicated that individuals with S100A11 high expression of BC had shorter overall survival. Multivariate Cox regression analysis revealed that high S100A11 expression independently influenced the poor outcome of patients with BC (HR = 1.738, 95%CI 1.197-2.524). Our nomogram incorporating five factors, including S100A11, age, clinical stage, N, and M, was developed to anticipate the survival probability in BC prognosis. The model demonstrated good consistency and accuracy. Furthermore, the mutation rete of S100A11 was 14%. Survival analysis suggested that breast cancer patients with S100A11 mutation had a worse prognosis. KEGG pathway enrichment analysis revealed that S100A11 may be mainly involved in the IL-17 signaling pathway. Finally, we discovered a correlation between S100A11 expression and immune cell infiltration on BC. S100A11 expression was positively associated with 17 immune checkpoint-related genes. In conclusion, this study indicates that S100A11 may contribute to a worse prognosis for BC and potentially has a significant impact through its influence on immune cell infiltration and the IL-17 signaling pathway.
Collapse
Affiliation(s)
- Junfang He
- School of Public Health, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yuxi Lei
- School of Public Health, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiabin Li
- Precision Pathology Diagnosis for Serious Diseases Key Laboratory of LuZhou, Luzhou, 646000, Sichuan, China
| | - Bin Wu
- Departments of Breast Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Yan Tang
- School of Public Health, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Institute of Cancer Medicine, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
14
|
Gounou C, Rouyer L, Siegfried G, Harté E, Bouvet F, d'Agata L, Darbo E, Lefeuvre M, Derieppe MA, Bouton L, Mélane M, Chapeau D, Martineau J, Prouzet-Mauleon V, Tan S, Souleyreau W, Saltel F, Argoul F, Khatib AM, Brisson AR, Iggo R, Bouter A. Inhibition of the membrane repair protein annexin-A2 prevents tumor invasion and metastasis. Cell Mol Life Sci 2023; 81:7. [PMID: 38092984 PMCID: PMC10719157 DOI: 10.1007/s00018-023-05049-3] [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: 07/24/2023] [Revised: 11/09/2023] [Accepted: 11/11/2023] [Indexed: 12/17/2023]
Abstract
Cancer cells are exposed to major compressive and shearing forces during invasion and metastasis, leading to extensive plasma membrane damage. To survive this mechanical stress, they need to repair membrane injury efficiently. Targeting the membrane repair machinery is thus potentially a new way to prevent invasion and metastasis. We show here that annexin-A2 (ANXA2) is required for membrane repair in invasive breast and pancreatic cancer cells. Mechanistically, we show by fluorescence and electron microscopy that cells fail to reseal shear-stress damaged membrane when ANXA2 is silenced or the protein is inhibited with neutralizing antibody. Silencing of ANXA2 has no effect on proliferation in vitro, and may even accelerate migration in wound healing assays, but reduces tumor cell dissemination in both mice and zebrafish. We expect that inhibiting membrane repair will be particularly effective in aggressive, poor prognosis tumors because they rely on the membrane repair machinery to survive membrane damage during tumor invasion and metastasis. This could be achieved either with anti-ANXA2 antibodies, which have been shown to inhibit metastasis of breast and pancreatic cancer cells, or with small molecule drugs.
Collapse
Affiliation(s)
- C Gounou
- CNRS, Bordeaux INP, CBMN, UMR 5248, University of Bordeaux, Bât. B14, Allée Geoffroy Saint Hilaire, 33600, Pessac, France
| | - L Rouyer
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
| | - G Siegfried
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
- XenoFish, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615, Pessac, France
| | - E Harté
- CNRS, LOMA, UMR 5798, University of Bordeaux, 33400, Talence, France
| | - F Bouvet
- CNRS, Bordeaux INP, CBMN, UMR 5248, University of Bordeaux, Bât. B14, Allée Geoffroy Saint Hilaire, 33600, Pessac, France
| | - L d'Agata
- CNRS, Bordeaux INP, CBMN, UMR 5248, University of Bordeaux, Bât. B14, Allée Geoffroy Saint Hilaire, 33600, Pessac, France
| | - E Darbo
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
| | - M Lefeuvre
- CNRS, Bordeaux INP, CBMN, UMR 5248, University of Bordeaux, Bât. B14, Allée Geoffroy Saint Hilaire, 33600, Pessac, France
| | - M A Derieppe
- Animalerie Mutualisée, Service Commun des Animaleries, University of Bordeaux, 33000, Bordeaux, France
| | - L Bouton
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
| | - M Mélane
- CNRS, LOMA, UMR 5798, University of Bordeaux, 33400, Talence, France
| | - D Chapeau
- CNRS, Bordeaux INP, CBMN, UMR 5248, University of Bordeaux, Bât. B14, Allée Geoffroy Saint Hilaire, 33600, Pessac, France
| | - J Martineau
- Animalerie Mutualisée, Service Commun des Animaleries, University of Bordeaux, 33000, Bordeaux, France
| | - V Prouzet-Mauleon
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
- CRISPRedit, TBMcore, UAR CNRS 3427, Inserm US 005, University of Bordeaux, Bordeaux, France
| | - S Tan
- CNRS, Bordeaux INP, CBMN, UMR 5248, University of Bordeaux, Bât. B14, Allée Geoffroy Saint Hilaire, 33600, Pessac, France
| | - W Souleyreau
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
| | - F Saltel
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
| | - F Argoul
- CNRS, LOMA, UMR 5798, University of Bordeaux, 33400, Talence, France
| | - A M Khatib
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
- XenoFish, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615, Pessac, France
- Bergonié Institute, Bordeaux, France
| | - A R Brisson
- CNRS, Bordeaux INP, CBMN, UMR 5248, University of Bordeaux, Bât. B14, Allée Geoffroy Saint Hilaire, 33600, Pessac, France
| | - R Iggo
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
| | - A Bouter
- CNRS, Bordeaux INP, CBMN, UMR 5248, University of Bordeaux, Bât. B14, Allée Geoffroy Saint Hilaire, 33600, Pessac, France.
| |
Collapse
|
15
|
Furutani M, Suganuma M, Akiyama S, Mitsumori R, Takemura M, Matsui Y, Satake S, Nakano Y, Niida S, Ozaki K, Hosoyama T, Shigemizu D. RNA-Sequencing Analysis Identification of Potential Biomarkers for Diagnosis of Sarcopenia. J Gerontol A Biol Sci Med Sci 2023; 78:1991-1998. [PMID: 37347997 DOI: 10.1093/gerona/glad150] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Indexed: 06/24/2023] Open
Abstract
Sarcopenia is a geriatric disease associated with increased mortality and disability. Early diagnosis and intervention are required to prevent it. This study investigated biomarkers for sarcopenia by using a combination of comprehensive clinical data and messenger RNA-sequencing (RNA-seq) analysis obtained from peripheral blood mononuclear cells. We enrolled a total of 114 older adults aged 66-94 years (52 sarcopenia diagnosed according to the Asian Working Group for Sarcopenia 2019 consensus and 62 normal older people). We used clinical data which were not included diagnosis criteria of sarcopenia, and stride length showed significance by logistic regression analysis (Bonferroni corrected p = .012, odds ratio = 0.14, 95% confidence interval [CI]: 0.05-0.40). RNA-seq analysis detected 6 differential expressed genes (FAR1, GNL2, HERC5, MRPL47, NUBP2, and S100A11). We also performed gene-set enrichment analysis and detected 2 functional modules (ie, hub genes, MYH9, and FLNA). By using any combination of the 9 candidates and basic information (age and sex), risk-prediction models were constructed. The best model by using a combination of stride length, HERC5, S100A11, and FLNA, achieved a high area under the curve (AUC) of 0.91 in a validation cohort (95% CI: 0.78-0.95). The quantitative PCR results of the 3 genes were consistent with the trend observed in the RNA-seq results. When BMI was added, the model achieved a high AUC of 0.95 (95% CI: 0.84-0.99). We have discovered potential biomarkers for the diagnosis of sarcopenia. Further refinement may lead to their future practical use in clinical use.
Collapse
Affiliation(s)
- Motoki Furutani
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Aichi, Japan
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Mutsumi Suganuma
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Shintaro Akiyama
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Risa Mitsumori
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Marie Takemura
- Center for Frailty and Locomotive Syndrome, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Yasumoto Matsui
- Center for Frailty and Locomotive Syndrome, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Shosuke Satake
- Center for Gerontology and Social Science, Research Institute, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Shumpei Niida
- Core Facility Administration, Research Institute, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Kouichi Ozaki
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Aichi, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Tohru Hosoyama
- Geroscience Research Center, Research Institute, National Center for Geriatrics and Gerontology, AichiJapan
| | - Daichi Shigemizu
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Aichi, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| |
Collapse
|
16
|
Meng Q, Chen C, Yang N, Gololobova O, Shi C, Dunn CA, Rossi M, Martindale JL, Basisty N, Ding J, Delannoy M, Basu S, Mazan-Mamczarz K, Shin CH, Yang JH, Johnson PF, Witwer KW, Biragyn A, Sen P, Abdelmohsen K, De S, Gorospe M. Surfaceome analysis of extracellular vesicles from senescent cells uncovers uptake repressor DPP4. Proc Natl Acad Sci U S A 2023; 120:e2219801120. [PMID: 37862381 PMCID: PMC10614838 DOI: 10.1073/pnas.2219801120] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 08/24/2023] [Indexed: 10/22/2023] Open
Abstract
Senescent cells are beneficial for repairing acute tissue damage, but they are harmful when they accumulate in tissues, as occurs with advancing age. Senescence-associated extracellular vesicles (S-EVs) can mediate cell-to-cell communication and export intracellular content to the microenvironment of aging tissues. Here, we studied the uptake of EVs from senescent cells (S-EVs) and proliferating cells (P-EVs) and found that P-EVs were readily taken up by proliferating cells (fibroblasts and cervical cancer cells) while S-EVs were not. We thus investigated the surface proteome (surfaceome) of P-EVs relative to S-EVs derived from cells that had reached senescence via replicative exhaustion, exposure to ionizing radiation, or treatment with etoposide. We found that relative to P-EVs, S-EVs from all senescence models were enriched in proteins DPP4, ANXA1, ANXA6, S10AB, AT1A1, and EPHB2. Among them, DPP4 was found to selectively prevent uptake by proliferating cells, as ectopic overexpression of DPP4 in HeLa cells rendered DPP4-expressing EVs that were no longer taken up by other proliferating cells. We propose that DPP4 on the surface of S-EVs makes these EVs refractory to internalization by proliferating cells, advancing our knowledge of the impact of senescent cells in aging-associated processes.
Collapse
Affiliation(s)
- Qiong Meng
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| | - Chen Chen
- Laboratory of Molecular Biology and Immunology, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| | - Na Yang
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| | - Olesia Gololobova
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Changyou Shi
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| | - Christopher A. Dunn
- Flow Cytometry Unit, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| | - Martina Rossi
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| | - Jennifer L. Martindale
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| | - Nathan Basisty
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| | - Jun Ding
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| | - Michael Delannoy
- Department of Cell Biology and Imaging Facility, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Srikanta Basu
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD21702
| | - Krystyna Mazan-Mamczarz
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| | - Chang Hoon Shin
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| | - Jen-Hao Yang
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| | - Peter F. Johnson
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD21702
| | - Kenneth W. Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Arya Biragyn
- Laboratory of Molecular Biology and Immunology, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| | - Payel Sen
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| | - Supriyo De
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD21224
| |
Collapse
|
17
|
Kalló G, Bertalan PM, Márton I, Kiss C, Csősz É. Salivary Chemical Barrier Proteins in Oral Squamous Cell Carcinoma-Alterations in the Defense Mechanism of the Oral Cavity. Int J Mol Sci 2023; 24:13657. [PMID: 37686462 PMCID: PMC10487546 DOI: 10.3390/ijms241713657] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most frequent types of head and neck cancer. Despite the genetic and environmental risk factors, OSCC is also associated with microbial infections and/or dysbiosis. The secreted saliva serves as the chemical barrier of the oral cavity and, since OSCC can alter the protein composition of saliva, our aim was to analyze the effect of OSCC on the salivary chemical barrier proteins. Publicly available datasets regarding the analysis of salivary proteins from patients with OSCC and controls were collected and examined in order to identify differentially expressed chemical barrier proteins. Network analysis and gene ontology (GO) classification of the differentially expressed chemical barrier proteins were performed as well. One hundred and twenty-seven proteins showing different expression pattern between the OSCC and control groups were found. Protein-protein interaction networks of up- and down-regulated proteins were constructed and analyzed. The main hub proteins (IL-6, IL-1B, IL-8, TNF, APOA1, APOA2, APOB, APOC3, APOE, and HP) were identified and the enriched GO terms were examined. Our study highlighted the importance of the chemical barrier of saliva in the development of OSCC.
Collapse
Affiliation(s)
- Gergő Kalló
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary; (P.M.B.); (I.M.); (É.C.)
- Biomarker Research Group, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
| | - Petra Magdolna Bertalan
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary; (P.M.B.); (I.M.); (É.C.)
- Biomarker Research Group, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
| | - Ildikó Márton
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary; (P.M.B.); (I.M.); (É.C.)
| | - Csongor Kiss
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary;
| | - Éva Csősz
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary; (P.M.B.); (I.M.); (É.C.)
- Biomarker Research Group, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
| |
Collapse
|
18
|
Peerapen P, Boonmark W, Thongboonkerd V. Characterizations of annexin A1-interacting proteins in apical membrane and cytosolic compartments of renal tubular epithelial cells. Comput Struct Biotechnol J 2023; 21:3796-3809. [PMID: 37560129 PMCID: PMC10407547 DOI: 10.1016/j.csbj.2023.07.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 07/27/2023] [Accepted: 07/27/2023] [Indexed: 08/11/2023] Open
Abstract
Annexin A1 (ANXA1) is a multifunctional calcium-binding protein that can bind to membrane phospholipids. Under high-calcium condition, ANXA1 expression increases on renal epithelial cell surface, leading to enhanced adhesion of calcium oxalate (CaOx) crystal (stone material) onto the cells. To regulate various cellular processes, ANXA1 interacts with many other intracellular protein partners. However, components of the ANXA1-interacting protein complex remain unclear. Herein, we characterized the interacting complexes of apical membrane (ApANXA1) and cytosolic (cyANXA1) forms of ANXA1 in apical membrane and cytosolic compartments, respectively, of renal epithelial cells under high-calcium condition using proteomic and bioinformatic approaches. After fractionation, the ApANXA1- and CyANXA1-interacting partners were identified by immunoprecipitation followed by nanoLC‑ESI‑Qq-TOF tandem mass spectrometry (IP-MS/MS). The ANXA1-interacting partners that were common in both apical membrane and cytosolic compartments and those unique in each compartment were then analyzed for their physico-chemical properties (molecular weight, isoelectric point, amino acid contents, instability index, aliphatic index, and grand average of hydropathicity), secondary structure (α-helix, β-turn, random coil, and extended strand), molecular functions, biological processes, reactome pathways and KEGG pathways. The data demonstrated that each set of these interacting proteins exhibited common and unique characteristics and properties. The knowledge from this study may lead to better understanding of the ApANXA1 and CyAXNA1 biochemistry and functions as well as the pathophysiology of CaOx kidney stone formation induced by high-calcium condition.
Collapse
Affiliation(s)
- Paleerath Peerapen
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Wanida Boonmark
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
19
|
Mandarino A, Thiyagarajan S, Martins ACF, Gomes RDS, Vetter SW, Leclerc E. S100s and HMGB1 Crosstalk in Pancreatic Cancer Tumors. Biomolecules 2023; 13:1175. [PMID: 37627239 PMCID: PMC10452588 DOI: 10.3390/biom13081175] [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: 04/13/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Pancreatic cancer remains a disease that is very difficult to treat. S100 proteins are small calcium binding proteins with diverse intra- and extracellular functions that modulate different aspects of tumorigenesis, including tumor growth and metastasis. High mobility group box 1 (HMGB1) protein is a multifaceted protein that also actively influences the development and progression of tumors. In this study, we investigate the possible correlations, at the transcript level, between S100s and HMGB1 in pancreatic cancer. For this purpose, we calculated Pearson's correlations between the transcript levels of 13 cancer-related S100 genes and HMGB1 in a cDNA array containing 19 pancreatic cancer tumor samples, and in 8 human pancreatic cancer cell lines. Statistically significant positive correlations were found in 5.5% (5 out of 91) and 37.4% (34 of 91) of the possible S100/S100 or S100/HMGB1 pairs in cells and tumors, respectively. Our data suggest that many S100 proteins crosstalk in pancreatic tumors either with other members of the S100 family, or with HMGB1. These newly observed interdependencies may be used to further the characterization of pancreatic tumors based on S100 and HMGB1 transcription profiles.
Collapse
Affiliation(s)
| | | | | | | | | | - Estelle Leclerc
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| |
Collapse
|
20
|
Kuo YS, Tsai YM. Exploring the Risk Factors and Possible Role of Calcium in Infective Endocarditis. Diagnostics (Basel) 2023; 13:2386. [PMID: 37510130 PMCID: PMC10378623 DOI: 10.3390/diagnostics13142386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/25/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Infective endocarditis (IE) is a severe and potentially life-threatening infection that affects the endocardium, the inner lining of the heart chambers and valves. Although rare, it is a potentially fatal condition, with an incidence of 3-10 cases per 100,000 people per year in developed countries and a mortality rate of up to 30% within 30 days. Early identification and diagnosis are critical for improving outcomes. The diagnosis of IE typically involves a combination of biomarkers, blood cultures, and echocardiography. However, currently, there are no specific biomarkers for the early detection of IE. Given the lack of specific biomarkers for IE, serum calcium levels have been suggested to play a unique role in IE. There have been few articles on the correlation between serum calcium and IE, suggesting that patients with endocarditis and lower levels of serum calcium may have a poorer prognosis. Therefore, in this article, we focus on biomarkers of endocarditis and discuss the evidence showing serum calcium as a potential indicator of IE.
Collapse
Affiliation(s)
- Yen-Shou Kuo
- Division of Thoracic Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei City 11490, Taiwan
| | - Yuan-Ming Tsai
- Division of Thoracic Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei City 11490, Taiwan
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
21
|
Halder A, Bhowmick C, Dutta PK, Mahadevappa M. Identification and Analysis of Imaging-Genomic Signatures to Study Recurrence in Breast Cancers. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38083388 DOI: 10.1109/embc40787.2023.10339965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
One of the main causes of breast cancer related death is its recurrence. In this study, we investigate the association of gene expression and pathological image features to understand breast cancer recurrence. A total of 172 breast cancer patient data was downloaded from the TCGA-BRCA database. The dataset contained diagnostic whole slide images and RNA-seq data of 80 recurrent and 92 disease-free breast cancer patients. We performed genomic analysis on RNA-seq data to obtain the hub genes related to recurrent breast cancer. We extracted relevant pathomic features from histopathology images. The discriminative ability of the hub genes and pathomic features were evaluated using machine learning classifiers. We used Spearman rank correlation analysis to find statistically significant association between gene expression and pathomic features. We identified that, genes which are related to breast cancer progression is significantly associated (adjusted p-value<0.05) with several pathomic features.Clinical Relevance- Histopathology is the gold standard for cancer detection. It provides us with cellular level information. A strong association between a pathomic feature and a gene expression will help clinicians understand the cellular and molecular mechanism of cancer for better prognosis.
Collapse
|
22
|
Cerón JJ, Ortín-Bustillo A, López-Martínez MJ, Martínez-Subiela S, Eckersall PD, Tecles F, Tvarijonaviciute A, Muñoz-Prieto A. S-100 Proteins: Basics and Applications as Biomarkers in Animals with Special Focus on Calgranulins (S100A8, A9, and A12). BIOLOGY 2023; 12:881. [PMID: 37372165 DOI: 10.3390/biology12060881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023]
Abstract
S100 proteins are a group of calcium-binding proteins which received this name because of their solubility in a 100% saturated solution of ammonium sulphate. They have a similar molecular mass of 10-12 KDa and share 25-65% similarity in their amino acid sequence. They are expressed in many tissues, and to date 25 different types of S100 proteins have been identified. This review aims to provide updated information about S100 proteins and their use as biomarkers in veterinary science, with special emphasis on the family of calgranulins that includes S100A8 (calgranulin A; myeloid-related protein 8, MRP8), S100A9 (calgranulin B; MRP14), and S100A12 (calgranulin C). The proteins SA100A8 and S100A9 can be linked, forming a heterodimer which is known as calprotectin. Calgranulins are related to the activation of inflammation and the immune system and increase in gastrointestinal diseases, inflammation and sepsis, immunomediated diseases, and obesity and endocrine disorders in different animal species. This review reflects the current knowledge about calgranulins in veterinary science, which should increase in the future to clarify their role in different diseases and potential as biomarkers and therapeutic targets, as well as the practical use of their measurement in non-invasive samples such as saliva or feces.
Collapse
Affiliation(s)
- José Joaquín Cerón
- Interdisciplinary Laboratory of Clinical Analysis (Interlab-UMU), Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, Campus de Espinardo s/n, 30100 Murcia, Spain
| | - Alba Ortín-Bustillo
- Interdisciplinary Laboratory of Clinical Analysis (Interlab-UMU), Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, Campus de Espinardo s/n, 30100 Murcia, Spain
| | - María José López-Martínez
- Interdisciplinary Laboratory of Clinical Analysis (Interlab-UMU), Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, Campus de Espinardo s/n, 30100 Murcia, Spain
| | - Silvia Martínez-Subiela
- Interdisciplinary Laboratory of Clinical Analysis (Interlab-UMU), Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, Campus de Espinardo s/n, 30100 Murcia, Spain
| | - Peter David Eckersall
- Interdisciplinary Laboratory of Clinical Analysis (Interlab-UMU), Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, Campus de Espinardo s/n, 30100 Murcia, Spain
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Garscube Estate, Glasgow G61 1QH, UK
| | - Fernando Tecles
- Interdisciplinary Laboratory of Clinical Analysis (Interlab-UMU), Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, Campus de Espinardo s/n, 30100 Murcia, Spain
| | - Asta Tvarijonaviciute
- Interdisciplinary Laboratory of Clinical Analysis (Interlab-UMU), Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, Campus de Espinardo s/n, 30100 Murcia, Spain
| | - Alberto Muñoz-Prieto
- Interdisciplinary Laboratory of Clinical Analysis (Interlab-UMU), Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, Campus de Espinardo s/n, 30100 Murcia, Spain
| |
Collapse
|
23
|
Wu Y, Wu S, Li F, Zeng T, Luo X. Association between serum S100A11 levels and glucose metabolism in diabetic process. Diabetol Metab Syndr 2023; 15:36. [PMID: 36872321 PMCID: PMC9987151 DOI: 10.1186/s13098-023-01004-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/19/2023] [Indexed: 03/07/2023] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is a prevalent non-communicable metabolic disease, and S100A11 is a newly identified gene closely related to metabolism. The association of S100A11 with diabetes is unclear. This study aimed to assess the relationship between S100A11 and markers of glucose metabolism in patients with different glucose tolerance and gender. METHODS This study included 97 participants. Baseline data were obtained, and the serum levels of S100A11 and metabolic markers (glycated hemoglobin [HbA1c], insulin release test, and oral glucose tolerance test) were measured. Linear and nonlinear correlations between serum S100A11 levels and HOMA-IR, HOMA of β, HbA1c, insulin sensitivity index (ISI), corrected insulin response (CIR), and oral disposition index (DIo) were analyzed. The expression of S100A11 was also detected in mice. RESULTS Serum S100A11 levels increased in patients with impaired glucose tolerance (IGT) of both genders. S100A11 mRNA and protein expression increased in obese mice. There were nonlinear correlations between S10011 levels and CIR, FPI, HOMA-IR, whole-body ISI in the IGT group. S100A11 was nonlinearly correlated with HOMA-IR, hepatic ISI, FPG, FPI, and HbA1c in the DM group. In the male group, S100A11 was linearly correlated with HOMA-IR and nonlinearly correlated with DIo (derived from hepatic ISI) and HbA1c. In the female population, S100A11 was nonlinearly correlated with CIR. CONCLUSIONS Serum S100A11 levels were highly expressed in patients with IGT and in the liver of obese mice. In addition, there were linear and nonlinear correlations between S100A11 and markers of glucose metabolism, demonstrating that S100A11 has a role in diabetes. Trial registration ChiCTR1900026990.
Collapse
Affiliation(s)
- Yao Wu
- Department of Laboratory Medicine, School of Medicine, Chongqing University Three Gorges Hospital, Chongqing University, No.165, Xincheng Avenue, Wanzhou District, Chongqing, 404000, China
| | - Shaobo Wu
- The Center of Clinical Research of Endocrinology and Metabolic Diseases in Chongqing, Chongqing University Three Gorges Hospital, Chongqing, 404100, China
- Department of Endocrinology, Chongqing University Three Gorges Hospital, Chongqing, 404100, China
| | - Fang Li
- Department of Laboratory Medicine, School of Medicine, Chongqing University Three Gorges Hospital, Chongqing University, No.165, Xincheng Avenue, Wanzhou District, Chongqing, 404000, China
| | - Ting Zeng
- Department of Laboratory Medicine, School of Medicine, Chongqing University Three Gorges Hospital, Chongqing University, No.165, Xincheng Avenue, Wanzhou District, Chongqing, 404000, China
| | - Xiaohe Luo
- Department of Laboratory Medicine, School of Medicine, Chongqing University Three Gorges Hospital, Chongqing University, No.165, Xincheng Avenue, Wanzhou District, Chongqing, 404000, China.
- The Center of Clinical Research of Endocrinology and Metabolic Diseases in Chongqing, Chongqing University Three Gorges Hospital, Chongqing, 404100, China.
| |
Collapse
|
24
|
Pan W, Jie W, Huang H. Vascular calcification: Molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2023; 4:e200. [PMID: 36620697 PMCID: PMC9811665 DOI: 10.1002/mco2.200] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 01/05/2023] Open
Abstract
Vascular calcification (VC) is recognized as a pathological vascular disorder associated with various diseases, such as atherosclerosis, hypertension, aortic valve stenosis, coronary artery disease, diabetes mellitus, as well as chronic kidney disease. Therefore, it is a life-threatening state for human health. There were several studies targeting mechanisms of VC that revealed the importance of vascular smooth muscle cells transdifferentiating, phosphorous and calcium milieu, as well as matrix vesicles on the progress of VC. However, the underlying molecular mechanisms of VC need to be elucidated. Though there is no acknowledged effective therapeutic strategy to reverse or cure VC clinically, recent evidence has proved that VC is not a passive irreversible comorbidity but an active process regulated by many factors. Some available approaches targeting the underlying molecular mechanism provide promising prospects for the therapy of VC. This review aims to summarize the novel findings on molecular mechanisms and therapeutic interventions of VC, including the role of inflammatory responses, endoplasmic reticulum stress, mitochondrial dysfunction, iron homeostasis, metabolic imbalance, and some related signaling pathways on VC progression. We also conclude some recent studies on controversial interventions in the clinical practice of VC, such as calcium channel blockers, renin-angiotensin system inhibitions, statins, bisphosphonates, denosumab, vitamins, and ion conditioning agents.
Collapse
Affiliation(s)
- Wei Pan
- Department of Cardiology, the Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Joint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic DiseaseSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Wei Jie
- Department of Cardiology, the Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Joint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic DiseaseSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Hui Huang
- Department of Cardiology, the Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Joint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic DiseaseSun Yat‐sen UniversityShenzhenGuangdongChina
| |
Collapse
|
25
|
Zheng M, Meng H, Li Y, Shi J, Han Y, Zhao C, Chen J, Han J, Liang J, Chen Y, Liu Q, Wang Y. S100A11 Promotes Metastasis via AKT and ERK Signaling Pathways and Has a Diagnostic Role in Hepatocellular Carcinoma. Int J Med Sci 2023; 20:318-328. [PMID: 36860671 PMCID: PMC9969497 DOI: 10.7150/ijms.80503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/10/2023] [Indexed: 02/04/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common and malignant liver tumor worldwide, although the treatment approaches for HCC continue to evolve, metastasis is the main reason for high mortality rates. S100 calcium-binding protein A11 (S100A11), an important member of the S100 family of small calcium-binding proteins, is overexpressed in various cells and regulates tumor development and metastasis. However, few studies report the role and underlying regulatory mechanisms of S100A11 in HCC development and metastasis. Herein, we discovered that S100A11 is overexpressed and associated with poor clinical outcomes in HCC cohorts, and we provided the first demonstration that S100A11 could serve as a novel diagnostic biomarker used in conjunction with AFP for HCC. Further analysis implied that S100A11 outperforms AFP in determining whether HCC patients have hematogenous metastasis or not. Using in vitro cell culture model, we demonstrated that S100A11 is overexpressed in metastatic hepatoma cells, knockdown of S100A11 decreases hepatoma cells proliferation, migration, invasion, and epithelial-mesenchymal transition process by inhibiting AKT and ERK signaling pathways. Altogether, our study provides new sights into the biological function and mechanisms underlying S100A11 in promoting metastasis of HCC and explores a novel target for HCC diagnosis and treatment.
Collapse
Affiliation(s)
- Mei Zheng
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Huan Meng
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Yunhui Li
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Jingren Shi
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Ying Han
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Changxu Zhao
- Beijing Institute of Microbiology and Epidemiology, Beijing, 100850, China
| | - Jin Chen
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Jinyu Han
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Jing Liang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Yuan Chen
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Qiqi Liu
- Beijing Institute of Microbiology and Epidemiology, Beijing, 100850, China
| | - Yajie Wang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| |
Collapse
|
26
|
Schwager SC, Young KM, Hapach LA, Carlson CM, Mosier JA, McArdle TJ, Wang W, Schunk C, Jayathilake AL, Bates ME, Bordeleau F, Antonyak MA, Cerione RA, Reinhart-King CA. Weakly migratory metastatic breast cancer cells activate fibroblasts via microvesicle-Tg2 to facilitate dissemination and metastasis. eLife 2022; 11:e74433. [PMID: 36475545 PMCID: PMC9767463 DOI: 10.7554/elife.74433] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer cell migration is highly heterogeneous, and the migratory capability of cancer cells is thought to be an indicator of metastatic potential. It is becoming clear that a cancer cell does not have to be inherently migratory to metastasize, with weakly migratory cancer cells often found to be highly metastatic. However, the mechanism through which weakly migratory cells escape from the primary tumor remains unclear. Here, utilizing phenotypically sorted highly and weakly migratory human breast cancer cells, we demonstrate that weakly migratory metastatic cells disseminate from the primary tumor via communication with stromal cells. While highly migratory cells are capable of single cell migration, weakly migratory cells rely on cell-cell signaling with fibroblasts to escape the primary tumor. Weakly migratory cells release microvesicles rich in tissue transglutaminase 2 (Tg2) which activate murine fibroblasts and lead weakly migratory cancer cell migration in vitro. These microvesicles also induce tumor stiffening and fibroblast activation in vivo and enhance the metastasis of weakly migratory cells. Our results identify microvesicles and Tg2 as potential therapeutic targets for metastasis and reveal a novel aspect of the metastatic cascade in which weakly migratory cells release microvesicles which activate fibroblasts to enhance cancer cell dissemination.
Collapse
Affiliation(s)
- Samantha C Schwager
- Department of Biomedical Engineering, Vanderbilt UniversityNashvilleUnited States
| | - Katherine M Young
- Department of Biomedical Engineering, Vanderbilt UniversityNashvilleUnited States
| | - Lauren A Hapach
- Department of Biomedical Engineering, Vanderbilt UniversityNashvilleUnited States
- Department of Biomedical Engineering, Cornell UniversityIthacaUnited States
| | - Caroline M Carlson
- Department of Biomedical Engineering, Vanderbilt UniversityNashvilleUnited States
| | - Jenna A Mosier
- Department of Biomedical Engineering, Vanderbilt UniversityNashvilleUnited States
| | | | - Wenjun Wang
- Department of Biomedical Engineering, Vanderbilt UniversityNashvilleUnited States
| | - Curtis Schunk
- Department of Biomedical Engineering, Vanderbilt UniversityNashvilleUnited States
| | | | - Madison E Bates
- Department of Biomedical Engineering, Vanderbilt UniversityNashvilleUnited States
| | - Francois Bordeleau
- CHU de Québec-Université Laval Research Center (Oncology division), UniversitéLaval Cancer Research Center and Faculty of Medicine, Université LavalQuébeccCanada
| | - Marc A Antonyak
- Department of Biomedical Science, Cornell UniversityIthacaUnited States
| | - Richard A Cerione
- Department of Biomedical Science, Cornell UniversityIthacaUnited States
| | | |
Collapse
|
27
|
Delangre E, Oppliger E, Berkcan S, Gjorgjieva M, Correia de Sousa M, Foti M. S100 Proteins in Fatty Liver Disease and Hepatocellular Carcinoma. Int J Mol Sci 2022; 23:ijms231911030. [PMID: 36232334 PMCID: PMC9570375 DOI: 10.3390/ijms231911030] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 01/27/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a highly prevalent and slow progressing hepatic pathology characterized by different stages of increasing severity which can ultimately give rise to the development of hepatocellular carcinoma (HCC). Besides drastic lifestyle changes, few drugs are effective to some extent alleviate NAFLD and HCC remains a poorly curable cancer. Among the deregulated molecular mechanisms promoting NAFLD and HCC, several members of the S100 proteins family appear to play an important role in the development of hepatic steatosis, non-alcoholic steatohepatitis (NASH) and HCC. Specific members of this Ca2+-binding protein family are indeed significantly overexpressed in either parenchymal or non-parenchymal liver cells, where they exert pleiotropic pathological functions driving NAFLD/NASH to severe stages and/or cancer development. The aberrant activity of S100 specific isoforms has also been reported to drive malignancy in liver cancers. Herein, we discuss the implication of several key members of this family, e.g., S100A4, S100A6, S100A8, S100A9 and S100A11, in NAFLD and HCC, with a particular focus on their intracellular versus extracellular functions in different hepatic cell types. Their clinical relevance as non-invasive diagnostic/prognostic biomarkers for the different stages of NAFLD and HCC, or their pharmacological targeting for therapeutic purpose, is further debated.
Collapse
|
28
|
Transcriptome Analyses Provide Insights into the Auditory Function in Trachemys scripta elegans. Animals (Basel) 2022; 12:ani12182410. [PMID: 36139269 PMCID: PMC9495000 DOI: 10.3390/ani12182410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/05/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
An auditory ability is essential for communication in vertebrates, and considerable attention has been paid to auditory sensitivity in mammals, birds, and frogs. Turtles were thought to be deaf for a long time; however, recent studies have confirmed the presence of an auditory ability in Trachemys scripta elegans as well as sex-related differences in hearing sensitivity. Earlier studies mainly focused on the morphological and physiological functions of the hearing organ in turtles; thus, the gene expression patterns remain unclear. In this study, 36 transcriptomes from six tissues (inner ear, tympanic membrane, brain, eye, lung, and muscle) were sequenced to explore the gene expression patterns of the hearing system in T. scripta elegans. A weighted gene co-expression network analysis revealed that hub genes related to the inner ear and tympanic membrane are involved in development and signal transduction. Moreover, we identified six differently expressed genes (GABRA1, GABRG2, GABBR2, GNAO1, SLC38A1, and SLC12A5) related to the GABAergic synapse pathway as candidate genes to explain the differences in sexually dimorphic hearing sensitivity. Collectively, this study provides a critical foundation for genetic research on auditory functions in turtles.
Collapse
|
29
|
Singh H, Agrawal DK. Therapeutic potential of targeting the receptor for advanced glycation end products (RAGE) by small molecule inhibitors. Drug Dev Res 2022; 83:1257-1269. [PMID: 35781678 PMCID: PMC9474610 DOI: 10.1002/ddr.21971] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/06/2022] [Accepted: 06/12/2022] [Indexed: 01/05/2023]
Abstract
Receptor for advanced glycation end products (RAGE) is a 45 kDa transmembrane receptor of immunoglobulin family that can bind to various endogenous and exogenous ligands and initiate the inflammatory downstream signaling pathways. RAGE is involved in various disorders including cardiovascular and neurodegenerative diseases, cancer, and diabetes. This review summarizes the structural features of RAGE and its various isoforms along with their pathological effects. Mainly, the article emphasized on the translational significance of antagonizing the interactions of RAGE with its ligands using small molecules reported in the last 5 years and discusses future approaches that could be employed to block the interactions in the treatment of chronic inflammatory ailments. The RAGE inhibitors described in this article could prove as a powerful approach in the management of immune‐inflammatory diseases. A critical review of the literature suggests that there is a dire need to dive deeper into the molecular mechanism of action to resolve critical issues that must be addressed to understand RAGE‐targeting therapy and long‐term blockade of RAGE in human diseases.
Collapse
Affiliation(s)
- Harbinder Singh
- Department of Translational Research, College of Osteopathic Medicine of the Pacific Western University of Health Sciences, Pomona, California, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
30
|
Zeng X, Guo H, Liu Z, Qin Z, Cong Y, Ren N, Zhang Y, Zhang N. S100A11 activates the pentose phosphate pathway to induce malignant biological behaviour of pancreatic ductal adenocarcinoma. Cell Death Dis 2022; 13:568. [PMID: 35752610 PMCID: PMC9233679 DOI: 10.1038/s41419-022-05004-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/22/2022] [Accepted: 06/07/2022] [Indexed: 01/21/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most refractory malignancies and has a poor prognosis. In recent years, increasing evidence has shown that an imbalance of metabolism may contribute to unrestricted pancreatic tumour progression and that the pentose phosphate pathway (PPP) plays a pivotal role in cellular metabolism. S100A11 has been shown to regulate multiple biological functions related to the progression and metastasis of various cancer types. However, the exact mechanisms and prognostic value of S100A11 in PDAC remain unclear. Here, we found that S100A11 expression was increased in PDAC and significantly associated with worse prognosis and disease progression. Mechanistically, S100A11 knockdown suppressed the PPP by impairing nascent mRNA synthesis of TKT (transketolase). The current study also demonstrated that H3K4me3 at the -268/+77 region of the TKT promoter was required for its transcriptional activation and S100A11 promoted H3K4me3 loading to the TKT promoter by interacting with SMYD3 protein. Taking these findings together, this study provided new insights into the potential value of S100A11 for treating pancreatic cancer, suggesting that it could be a therapeutic target for PDAC patients.
Collapse
Affiliation(s)
- Xue Zeng
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, 110042, PR China
| | - Hong Guo
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, 110042, PR China
| | - Zhuang Liu
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, 110042, PR China
| | - Zilan Qin
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, 110042, PR China
| | - Yuyang Cong
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, 110042, PR China
| | - Naihan Ren
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, 110042, PR China
| | - Yuxiang Zhang
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, 110042, PR China
| | - Na Zhang
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, 110042, PR China.
| |
Collapse
|
31
|
Barzilova VD, Drury J, Rogers B, Thomas E, Ahmed F, Bradfield A, Al-Lamee H, Hapangama DK. Role of Nucleolin in Endometrial Precancerous Hyperplasia and Carcinogenesis: Ex Vivo and In Silico Study. Int J Mol Sci 2022; 23:6228. [PMID: 35682908 PMCID: PMC9181237 DOI: 10.3390/ijms23116228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/26/2022] [Accepted: 05/29/2022] [Indexed: 12/04/2022] Open
Abstract
Endometrial cancer (EC) is the most common gynaecological malignancy. Nucleolin (NCL) is involved in rDNA transcription, cell proliferation, and apoptosis, with high expression associated with worse overall survival (OS) in other adenocarcinomas. Our aims were to assess NCL gene and protein expression and explore the differential expression of NCL-associated genes (NAGs) in endometrial carcinogenesis. Endometrial samples were obtained from 157 women to include healthy, hyperplastic (EH), EC, and metastatic groups. RT-qPCR and immunohistochemistry were employed to assess NCL gene and protein levels. In silico analysis of NAGs in TCGA and GEO datasets was performed, with the prognostic value determined via Human Protein Atlas. NCL mRNA level of EC was lower than in healthy post-menopausal endometrium (p < 0.01). EH samples had lower NCL immuno-expression scores than healthy pre-menopausal (p < 0.001), benign post-menopausal (p < 0.01), and EC (p < 0.0001) samples. Metastatic lesions demonstrated higher NCL quick scores than primary tissue (p = 0.04). Higher NCL Immuno quick scores carried a worse OS in high-grade EC (p = 0.01). Interrogating Uterine Corpus Endometrial Carcinoma (TCGA-UCEC) and Uterine Carcinosarcoma (TCGA-UCS) cohorts revealed NCL to be the most highly upregulated gene in carcinosarcoma, with S100A11, LMNB2, RERG, E2F1 and CCNA2 representing key dysregulated NAGs in EC. Since NCL is implicated in transforming hyperplastic glands into cancer, with further involvement in metastasis, it is suggested to be a promising target for better-informed diagnosis, risk stratification, and management of EC.
Collapse
Affiliation(s)
- Vanya D. Barzilova
- Centre for Women’s Health Research, Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L8 7SS, UK; (V.D.B.); (J.D.); (B.R.); (E.T.); (F.A.); (H.A.-L.)
| | - Josephine Drury
- Centre for Women’s Health Research, Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L8 7SS, UK; (V.D.B.); (J.D.); (B.R.); (E.T.); (F.A.); (H.A.-L.)
| | - Bryony Rogers
- Centre for Women’s Health Research, Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L8 7SS, UK; (V.D.B.); (J.D.); (B.R.); (E.T.); (F.A.); (H.A.-L.)
| | - Emily Thomas
- Centre for Women’s Health Research, Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L8 7SS, UK; (V.D.B.); (J.D.); (B.R.); (E.T.); (F.A.); (H.A.-L.)
| | - Fareen Ahmed
- Centre for Women’s Health Research, Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L8 7SS, UK; (V.D.B.); (J.D.); (B.R.); (E.T.); (F.A.); (H.A.-L.)
| | - Alice Bradfield
- Liverpool Women’s NHS Foundation Trust, Member of Liverpool Health Partners, Liverpool L8 7SS, UK;
| | - Hannan Al-Lamee
- Centre for Women’s Health Research, Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L8 7SS, UK; (V.D.B.); (J.D.); (B.R.); (E.T.); (F.A.); (H.A.-L.)
- Liverpool Women’s NHS Foundation Trust, Member of Liverpool Health Partners, Liverpool L8 7SS, UK;
- Hewitt Centre for Reproductive Medicine, Liverpool Women’s NHS Foundation Trust, Liverpool L8 7SS, UK
| | - Dharani K. Hapangama
- Centre for Women’s Health Research, Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L8 7SS, UK; (V.D.B.); (J.D.); (B.R.); (E.T.); (F.A.); (H.A.-L.)
- Liverpool Women’s NHS Foundation Trust, Member of Liverpool Health Partners, Liverpool L8 7SS, UK;
| |
Collapse
|
32
|
Peppercorn K, Kleffmann T, Jones O, Hughes S, Tate W. Secreted Amyloid Precursor Protein Alpha, a Neuroprotective Protein in the Brain Has Widespread Effects on the Transcriptome and Proteome of Human Inducible Pluripotent Stem Cell-Derived Glutamatergic Neurons Related to Memory Mechanisms. Front Neurosci 2022; 16:858524. [PMID: 35692428 PMCID: PMC9179159 DOI: 10.3389/fnins.2022.858524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/14/2022] [Indexed: 11/18/2022] Open
Abstract
Secreted amyloid precursor protein alpha (sAPPα) processed from a parent human brain protein, APP, can modulate learning and memory. It has potential for development as a therapy preventing, delaying, or even reversing Alzheimer’s disease. In this study a comprehensive analysis to understand how it affects the transcriptome and proteome of the human neuron was undertaken. Human inducible pluripotent stem cell (iPSC)-derived glutamatergic neurons in culture were exposed to 1 nM sAPPα over a time course and changes in the transcriptome and proteome were identified with RNA sequencing and Sequential Window Acquisition of All THeoretical Fragment Ion Spectra-Mass Spectrometry (SWATH-MS), respectively. A large subset (∼30%) of differentially expressed transcripts and proteins were functionally involved with the molecular biology of learning and memory, consistent with reported links of sAPPα to memory enhancement, as well as neurogenic, neurotrophic, and neuroprotective phenotypes in previous studies. Differentially regulated proteins included those encoded in previously identified Alzheimer’s risk genes, APP processing related proteins, proteins involved in synaptogenesis, neurotransmitters, receptors, synaptic vesicle proteins, cytoskeletal proteins, proteins involved in protein and organelle trafficking, and proteins important for cell signalling, transcriptional splicing, and functions of the proteasome and lysosome. We have identified a complex set of genes affected by sAPPα, which may aid further investigation into the mechanism of how this neuroprotective protein affects memory formation and how it might be used as an Alzheimer’s disease therapy.
Collapse
Affiliation(s)
- Katie Peppercorn
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Torsten Kleffmann
- Division of Health Sciences, Research Infrastructure Centre, University of Otago, Dunedin, New Zealand
| | - Owen Jones
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Stephanie Hughes
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Warren Tate
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- *Correspondence: Warren Tate,
| |
Collapse
|
33
|
Alarm Signal S100-Related Signature Is Correlated with Tumor Microenvironment and Predicts Prognosis in Glioma. DISEASE MARKERS 2022; 2022:4968555. [PMID: 35592707 PMCID: PMC9113871 DOI: 10.1155/2022/4968555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/15/2022] [Accepted: 04/19/2022] [Indexed: 11/17/2022]
Abstract
Glioma are the most common malignant central nervous system tumor and are characterized by uncontrolled proliferation and resistance to therapy. Dysregulation of S100 proteins may augment tumor initiation, proliferation, and metastasis by modulating immune response. However, the comprehensive function and prognostic value of S100 proteins in glioma remain unclear. Here, we explored the expression profiles of 17 S100 family genes and constructed a high-efficient prediction model for glioma based on CGGA and TCGA datasets. Immune landscape analysis displayed that the distribution of immune scores, ESTIMATE scores, and stromal scores, as well as infiltrating immune cells (macrophages M0/M1/M2, T cell CD4+ naïve, Tregs, monocyte, neutrophil, and NK activated), were significant different between risk-score subgroups. Overall, we demonstrated the value of S100 protein-related signature in the prediction of glioma patients’ prognosis and determined its relationship with the tumor microenvironment (TME) in glioma.
Collapse
|
34
|
Zhu T, Zhang L, Li C, Tan X, Liu J, Huiqin Li, Fan Q, Zhang Z, Zhan M, Fu L, Luo J, Geng J, Wu Y, Zou X, Liang B. The S100 calcium binding protein A11 promotes liver fibrogenesis by targeting TGF-β signaling. J Genet Genomics 2022; 49:338-349. [PMID: 35240304 DOI: 10.1016/j.jgg.2022.02.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 02/22/2022] [Accepted: 02/22/2022] [Indexed: 12/16/2022]
Abstract
Liver fibrosis is a key transformation stage and also a reversible pathological process in various types of chronic liver diseases. However, the pathogenesis of liver fibrosis still remains elusive. Here, we report that the calcium binding protein A11 (S100A11) is consistently upregulated in the integrated data from GSE liver fibrosis and tree shrew liver proteomics. S100A11 is also experimentally activated in liver fibrosis in mouse, rat, tree shrew, and human with liver fibrosis. While overexpression of S100A11 in vivo and in vitro exacerbates liver fibrosis, the inhibition of S100A11 improves liver fibrosis. Mechanistically, S100A11 activates hepatic stellate cells (HSCs) and the fibrogenesis process via the regulation of the deacetylation of Smad3 in the TGF-β signaling pathway. S100A11 physically interacts with SIRT6, a deacetylase of Smad2/3, which may competitively inhibit the interaction between SIRT6 and Smad2/3. The subsequent release and activation of Smad2/3 promote the activation of HSCs and fibrogenesis. Additionally, a significant elevation of S100A11 in serum is observed in clinical patients. Our study uncovers S100A11 as a novel profibrogenic factor in liver fibrosis, which may represent both a potential biomarker and a promising therapy target for treating liver fibrosis and fibrosis-related liver diseases.
Collapse
Affiliation(s)
- Tingting Zhu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Linqiang Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Chengbin Li
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, China
| | - Xiaoqiong Tan
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Jing Liu
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, China
| | - Huiqin Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Qijing Fan
- School of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China
| | - Zhiguo Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Mingfeng Zhan
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, China
| | - Lin Fu
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, China
| | - Jinbo Luo
- Infectious Diseases Department and Hepatic Diseases Department, the First People's Hospital of Yunnan Province, Kunming, Yunnan 650034, China; Infectious Diseases Department and Hepatic Diseases Department, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650034, China
| | - Jiawei Geng
- Infectious Diseases Department and Hepatic Diseases Department, the First People's Hospital of Yunnan Province, Kunming, Yunnan 650034, China; Infectious Diseases Department and Hepatic Diseases Department, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650034, China.
| | - Yingjie Wu
- School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center,Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250021, China; Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Liaoning Provence Key Lab of Genome Engineered Animal Models Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Xiaoju Zou
- School of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China.
| | - Bin Liang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, China.
| |
Collapse
|