1
|
Akita K, Maurer MS, Tower-Rader A, Fifer MA, Shimada YJ. Comprehensive Proteomics Profiling Identifies Circulating Biomarkers to Distinguish Hypertrophic Cardiomyopathy From Other Cardiomyopathies With Left Ventricular Hypertrophy. Circ Heart Fail 2025; 18:e012434. [PMID: 39523983 PMCID: PMC11753946 DOI: 10.1161/circheartfailure.124.012434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Distinguishing hypertrophic cardiomyopathy (HCM) from other cardiomyopathies with left ventricular hypertrophy (LVH), such as hypertensive LVH, transthyretin amyloid cardiomyopathy, and aortic stenosis, is sometimes challenging. Using plasma proteomics profiling, we aimed to identify circulating biomarkers and dysregulated signaling pathways specific to HCM. METHODS In this multicenter case-control study, plasma proteomics profiling was performed in cases with HCM and controls with hypertensive LVH, transthyretin amyloid cardiomyopathy, and aortic stenosis. Two-thirds of patients enrolled earlier in each disease group were defined as the training set and the remaining one-third as the test set. Protein concentrations in HCM were compared with those in hypertensive LVH (comparison 1), transthyretin amyloid cardiomyopathy (comparison 2), and aortic stenosis (comparison 3). Candidate proteins that meet the following 2 criteria were selected: (1) higher abundance in HCM throughout all 3 comparisons or lower abundance in HCM throughout all 3 comparisons with univariable P<0.05 and |log2(fold change)| >0.5 in both the training and test sets and (2) independently associated with HCM with multivariable P<0.05 after adjusting for clinical parameters significantly different between HCM and controls. Using the selected candidate proteins, a logistic regression model to distinguish HCM from controls was developed in the training set and applied to the test set. Finally, pathway analysis was performed in each comparison using proteins with different abundance. RESULTS Overall, 4979 proteins in 1415 patients (HCM, n=879; hypertensive LVH, n=331; transthyretin amyloid cardiomyopathy, n=169; aortic stenosis, n=36) were analyzed. Of those, 5 proteins were selected as candidate proteins. The logistic regression model with these 5 proteins had an area under the receiver operating characteristic curve of 0.86 (95% CI, 0.82-0.89) in the test set. The MAPK (mitogen-activated protein kinase) and HIF-1 (hypoxia-inducible factor 1) pathways were dysregulated in HCM throughout the 3 comparisons. CONCLUSIONS This study identified circulating biomarkers that distinguish HCM from other cardiomyopathies with LVH independently from confounders and revealed signaling pathways associated with HCM.
Collapse
Affiliation(s)
- Keitaro Akita
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Mathew S. Maurer
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Albree Tower-Rader
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael A. Fifer
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yuichi J. Shimada
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
2
|
Zhang H, Vandesompele J, Braeckmans K, De Smedt SC, Remaut K. Nucleic acid degradation as barrier to gene delivery: a guide to understand and overcome nuclease activity. Chem Soc Rev 2024; 53:317-360. [PMID: 38073448 DOI: 10.1039/d3cs00194f] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Gene therapy is on its way to revolutionize the treatment of both inherited and acquired diseases, by transferring nucleic acids to correct a disease-causing gene in the target cells of patients. In the fight against infectious diseases, mRNA-based therapeutics have proven to be a viable strategy in the recent Covid-19 pandemic. Although a growing number of gene therapies have been approved, the success rate is limited when compared to the large number of preclinical and clinical trials that have been/are being performed. In this review, we highlight some of the hurdles which gene therapies encounter after administration into the human body, with a focus on nucleic acid degradation by nucleases that are extremely abundant in mammalian organs, biological fluids as well as in subcellular compartments. We overview the available strategies to reduce the biodegradation of gene therapeutics after administration, including chemical modifications of the nucleic acids, encapsulation into vectors and co-administration with nuclease inhibitors and discuss which strategies are applied for clinically approved nucleic acid therapeutics. In the final part, we discuss the currently available methods and techniques to qualify and quantify the integrity of nucleic acids, with their own strengths and limitations.
Collapse
Affiliation(s)
- Heyang Zhang
- Laboratory for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium.
- Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Jo Vandesompele
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium.
- Centre for Nano- and Biophotonics, Ghent University, 9000 Ghent, Belgium
| | - Stefaan C De Smedt
- Laboratory for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Centre for Nano- and Biophotonics, Ghent University, 9000 Ghent, Belgium
| | - Katrien Remaut
- Laboratory for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
3
|
Xie J, Mao H. Functional Insight into hTRIR. Curr Mol Med 2024; 24:1445-1449. [PMID: 37867262 DOI: 10.2174/0115665240260310231016112946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 10/24/2023]
Abstract
The uncharacterized C19orf43 was discovered to be associated with hTR maturation. Our previous work indicated that C19orf43 cleaves distinct RNA types but not DNA. We then named it hTR-interacting RNase (hTRIR) (Uniprot: Q9BQ61). hTRIR works in a broad range of temperatures and pH without any divalent cations needed. hTRIR cleaves RNA at all four nucleotide sites but preferentially at purines. In addition, hTRIR digested both ends of methylated small RNA, which suggested that it was a putative ribonuclease. Later, we designed more nucleotides that methylated small RNA to determine whether it was an exo- and/or endoribonuclease. Unlike RNase A, hTRIR could digest both ends of methylated RNA oligos 5R5, which suggested it was potentially an endoribonuclease.
Collapse
Affiliation(s)
- Jumin Xie
- Biochemistry and Molecular Biology, Hubei Polytechnic University, Huangshi, Hubei 435003, P.R. China
| | - Hui Mao
- Department of Dermatology, Huangshi Central Hospital, Huangshi, Hubei, 435000, P.R. China
| |
Collapse
|
4
|
Deo R, Dubin RF, Ren Y, Murthy AC, Wang J, Zheng H, Zheng Z, Feldman H, Shou H, Coresh J, Grams M, Surapaneni AL, Bhat Z, Cohen JB, Rahman M, He J, Saraf SL, Go AS, Kimmel PL, Vasan RS, Segal MR, Li H, Ganz P. Proteomic cardiovascular risk assessment in chronic kidney disease. Eur Heart J 2023; 44:2095-2110. [PMID: 37014015 PMCID: PMC10281556 DOI: 10.1093/eurheartj/ehad115] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 01/21/2023] [Accepted: 02/16/2023] [Indexed: 04/05/2023] Open
Abstract
AIMS Chronic kidney disease (CKD) is widely prevalent and independently increases cardiovascular risk. Cardiovascular risk prediction tools derived in the general population perform poorly in CKD. Through large-scale proteomics discovery, this study aimed to create more accurate cardiovascular risk models. METHODS AND RESULTS Elastic net regression was used to derive a proteomic risk model for incident cardiovascular risk in 2182 participants from the Chronic Renal Insufficiency Cohort. The model was then validated in 485 participants from the Atherosclerosis Risk in Communities cohort. All participants had CKD and no history of cardiovascular disease at study baseline when ∼5000 proteins were measured. The proteomic risk model, which consisted of 32 proteins, was superior to both the 2013 ACC/AHA Pooled Cohort Equation and a modified Pooled Cohort Equation that included estimated glomerular filtrate rate. The Chronic Renal Insufficiency Cohort internal validation set demonstrated annualized receiver operating characteristic area under the curve values from 1 to 10 years ranging between 0.84 and 0.89 for the protein and 0.70 and 0.73 for the clinical models. Similar findings were observed in the Atherosclerosis Risk in Communities validation cohort. For nearly half of the individual proteins independently associated with cardiovascular risk, Mendelian randomization suggested a causal link to cardiovascular events or risk factors. Pathway analyses revealed enrichment of proteins involved in immunologic function, vascular and neuronal development, and hepatic fibrosis. CONCLUSION In two sizeable populations with CKD, a proteomic risk model for incident cardiovascular disease surpassed clinical risk models recommended in clinical practice, even after including estimated glomerular filtration rate. New biological insights may prioritize the development of therapeutic strategies for cardiovascular risk reduction in the CKD population.
Collapse
Affiliation(s)
- Rajat Deo
- Division of Cardiovascular Medicine, Electrophysiology Section, Perelman School of Medicine at the University of Pennsylvania, One Convention Avenue, Level 2 / City Side, Philadelphia, PA 19104, USA
| | - Ruth F Dubin
- Division of Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Yue Ren
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Ashwin C Murthy
- Division of Cardiovascular Medicine, Electrophysiology Section, Perelman School of Medicine at the University of Pennsylvania, One Convention Avenue, Level 2 / City Side, Philadelphia, PA 19104, USA
| | - Jianqiao Wang
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Haotian Zheng
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Zihe Zheng
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Harold Feldman
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Haochang Shou
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Josef Coresh
- Department of Epidemiology; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins University, 2024 E. Monument Street, Room 2-635, Suite 2-600, Baltimore, MD 21287, USA
| | - Morgan Grams
- Department of Epidemiology; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins University, 2024 E. Monument Street, Room 2-635, Suite 2-600, Baltimore, MD 21287, USA
| | - Aditya L Surapaneni
- Department of Epidemiology; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA
| | - Zeenat Bhat
- Division of Nephrology, University of Michigan, 5100 Brehm Tower, 1000 Wall Street, Ann Arbor, MI 48105, USA
| | - Jordana B Cohen
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
- Renal, Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, 831 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Mahboob Rahman
- Department of Medicine, Case Western Reserve University School of Medicine, 11100 Euclid Avenue, Wearn Bldg. 3 Floor. Rm 352, Cleveland, OH 44106, USA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, SL 18, New Orleans, LA 70112, USA
| | - Santosh L Saraf
- Division of Hematology and Oncology, University of Illinois at Chicago, 1740 West Taylor Street, Chicago, IL 60612, USA
| | - Alan S Go
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA
- Departments of Epidemiology, Biostatistics and Medicine, University of California at San Francisco, San Francisco, CA, USA
| | - Paul L Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Ramachandran S Vasan
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Section of Cardiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Mark R Segal
- Department of Epidemiology and Biostatistics, University of California, 550 16th Street, 2nd Floor, Box #0560, San Francisco, CA 94143, USA
| | - Hongzhe Li
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 215 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104, USA
| | - Peter Ganz
- Division of Cardiology, Zuckerberg San Francisco General Hospital and Department of Medicine, University of California, San Francisco, 1001 Potrero Avenue, 5G1, San Francisco, CA 94110, USA
| |
Collapse
|
5
|
Laakmann K, Eckersberg JM, Hapke M, Wiegand M, Bierwagen J, Beinborn I, Preußer C, Pogge von Strandmann E, Heimerl T, Schmeck B, Jung AL. Bacterial extracellular vesicles repress the vascular protective factor RNase1 in human lung endothelial cells. Cell Commun Signal 2023; 21:111. [PMID: 37189117 DOI: 10.1186/s12964-023-01131-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Sepsis is one of the leading causes of death worldwide and characterized by blood stream infections associated with a dysregulated host response and endothelial cell (EC) dysfunction. Ribonuclease 1 (RNase1) acts as a protective factor of vascular homeostasis and is known to be repressed by massive and persistent inflammation, associated to the development of vascular pathologies. Bacterial extracellular vesicles (bEVs) are released upon infection and may interact with ECs to mediate EC barrier dysfunction. Here, we investigated the impact of bEVs of sepsis-related pathogens on human EC RNase1 regulation. METHODS bEVs from sepsis-associated bacteria were isolated via ultrafiltration and size exclusion chromatography and used for stimulation of human lung microvascular ECs combined with and without signaling pathway inhibitor treatments. RESULTS bEVs from Escherichia coli, Klebsiella pneumoniae and Salmonella enterica serovar Typhimurium significantly reduced RNase1 mRNA and protein expression and activated ECs, while TLR2-inducing bEVs from Streptococcus pneumoniae did not. These effects were mediated via LPS-dependent TLR4 signaling cascades as they could be blocked by Polymyxin B. Additionally, LPS-free ClearColi™ had no impact on RNase1. Further characterization of TLR4 downstream pathways involving NF-кB and p38, as well as JAK1/STAT1 signaling, revealed that RNase1 mRNA regulation is mediated via a p38-dependent mechanism. CONCLUSION Blood stream bEVs from gram-negative, sepsis-associated bacteria reduce the vascular protective factor RNase1, opening new avenues for therapeutical intervention of EC dysfunction via promotion of RNase1 integrity. Video Abstract.
Collapse
Affiliation(s)
- Katrin Laakmann
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Jorina Mona Eckersberg
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Moritz Hapke
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Marie Wiegand
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Jeff Bierwagen
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Isabell Beinborn
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Christian Preußer
- Institute for Tumor Immunology and Core Facility - Extracellular Vesicles, Philipps-University Marburg, Marburg, Germany
| | - Elke Pogge von Strandmann
- Institute for Tumor Immunology and Core Facility - Extracellular Vesicles, Philipps-University Marburg, Marburg, Germany
| | - Thomas Heimerl
- Center for Synthetic Microbiology (SYNMIKRO), Philipps-University Marburg, Marburg, Germany
| | - Bernd Schmeck
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
- Center for Synthetic Microbiology (SYNMIKRO), Philipps-University Marburg, Marburg, Germany
- Core Facility Flow Cytometry - Bacterial Vesicles, Philipps-University Marburg, Marburg, Germany
- Department of Pulmonary and Critical Care Medicine, Philipps-University Marburg, Marburg, Germany
- Member of the German Center for Infectious Disease Research (DZIF), Marburg, Germany
| | - Anna Lena Jung
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany.
- Core Facility Flow Cytometry - Bacterial Vesicles, Philipps-University Marburg, Marburg, Germany.
| |
Collapse
|
6
|
Functions and cellular signaling by ribosomal extracellular RNA (rexRNA): Facts and hypotheses on a non-typical DAMP. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119408. [PMID: 36503009 DOI: 10.1016/j.bbamcr.2022.119408] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/07/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022]
Abstract
Upon microbial infections with the subsequent host response of innate immunity, a variety of fragmented RNA- and DNA-based "Pathogen-associated molecular patterns" (PAMPs) are recognized mainly by endosomal or cytoplasmic host cell "Pattern recognition receptors" (PRRs), particularly "Toll-like receptors" (TLRs). Concomitantly, various self-extracellular RNA species (exRNAs) are present in extracellular body fluids where they contribute to diverse physiological and homeostatic processes. In principle, such exRNAs, including the most abundant one, ribosomal exRNA (rexRNA), are designated as "Danger-associated molecular patterns" (DAMPs) and are prevented by e.g. natural modifications from uncontrolled signaling via TLRs to avoid hyper-inflammatory responses or autoimmunity. Upon cellular stress or tissue damage/necrosis, the levels and composition of released self-exRNA species, either in free form, in complex with proteins or in association with extracellular vesicles (EVs), can change considerably. Among the self-exRNAs, rexRNA is considered as a non-typical DAMP, since it may induce inflammatory responses by cell membrane receptors, both in the absence or presence of PAMPs. Yet, its mode of receptor activation to mount inflammatory responses remains obscure. RexRNA also serves as a universal damaging factor in cardiovascular and other diseases independent of PRRs. In general, RNase1 provides a profound antagonist in these pathologies and in rexRNA-mediated inflammatory cell responses. Based on the extrapolation of the here described aspects of rexRNA-biology, further activities of this molecular entity are hypothesized that may stimulate additional research in this area.
Collapse
|
7
|
Wang R, Xu J, Tang Y, Wang Y, Zhao J, Ding L, Peng Y, Zhang Z. Transcriptome-wide analysis reveals the coregulation of RNA-binding proteins and alternative splicing genes in the development of atherosclerosis. Sci Rep 2023; 13:1764. [PMID: 36720950 PMCID: PMC9889815 DOI: 10.1038/s41598-022-26556-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 12/16/2022] [Indexed: 02/02/2023] Open
Abstract
RNA-binding proteins (RBPs) are involved in the regulation of RNA splicing, stability, and localization. How RBPs control the development of atherosclerosis, is not fully understood. To explore the relevant RNA-binding proteins (RBPs) and alternative splicing events (ASEs) in atherosclerosis. We made a comprehensive work to integrate analyses of differentially expressed genes, including differential RBPs, and variable splicing characteristics related to different stages of atherosclerosis in dataset GSE104140. A total of 3712 differentially expressed genes (DEGs) were identified, including 2921 upregulated genes and 791 downregulated genes. Further analysis screened out 54 RBP genes, and 434 AS genes overlapped DEGs. We selected high expression ten RBP genes (SAMHD1, DDX60 L, TLR7, RBM47, MYEF2, RNASE6, PARP12, APOBEC3G, SMAD9, and RNASE1) for co-expression analysis. Meanwhile, we found seven regulated alternative splicing genes (RASGs) (ABI1, FXR1, CHID1, PLEC, PRKACB, BNIP2, PPP3CB) that could be regulated by RBPs. The co-expression network was used to further elucidate the regulatory and interaction relationship between RBPs and AS genes. Apoptotic process and innate immune response, revealed by the functional enrichment analysis of RASGs regulated by RBPs were closely related to atherosclerosis. In addition, 26 of the 344 alternative splicing genes regulated by the above 10 RBPs were transcription factors (TFs), We selected high expression nine TFs (TFDP1, RBBP7, STAT2, CREB5, ERG, ELF1, HMGN3, BCLAF1, and ZEB2) for co-expression analysis. The target genes of these TFs were mainly enriched in inflammatory and immune response pathways that were associated with atherosclerosis. indicating that AS abnormalities of these TFs may have a function in atherosclerosis. Furthermore, the expression of differentially expressed RBPs and the alternative splicing events of AS genes was validated by qRT-PCR in umbilical vein endothelial cells (HUVEC). The results showed that RBM47 were remarkedly difference in HUVEC treated with ox-LDL and the splicing ratio of AS in BCLAF1which is regulated by RBM47 significantly changed. In conclusion, the differentially expressed RBPs identified in our analysis may play important roles in the development of atherosclerosis by regulating the AS of these TF genes.
Collapse
Affiliation(s)
- Runqing Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.,Gansu Key Laboratory of Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Gansu Clinical Medical Research Center for Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Jin Xu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.,Gansu Key Laboratory of Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Gansu Clinical Medical Research Center for Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yuning Tang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.,Gansu Key Laboratory of Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Gansu Clinical Medical Research Center for Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Yongxiang Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.,Gansu Key Laboratory of Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Gansu Clinical Medical Research Center for Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Heart Center, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Jing Zhao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.,Gansu Key Laboratory of Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Gansu Clinical Medical Research Center for Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Heart Center, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Liqiong Ding
- Gansu Key Laboratory of Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Gansu Clinical Medical Research Center for Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Heart Center, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yu Peng
- Gansu Key Laboratory of Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Gansu Clinical Medical Research Center for Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,Heart Center, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Zheng Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China. .,Gansu Key Laboratory of Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, Gansu, China. .,Gansu Clinical Medical Research Center for Cardiovascular Diseases, The First Hospital of Lanzhou University, Lanzhou, Gansu, China. .,Heart Center, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.
| |
Collapse
|
8
|
Evaluating and mitigating clinical samples matrix effects on TX-TL cell-free performance. Sci Rep 2022; 12:13785. [PMID: 35962056 PMCID: PMC9374283 DOI: 10.1038/s41598-022-17583-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/27/2022] [Indexed: 12/03/2022] Open
Abstract
Cell-free biosensors are promising tools for medical diagnostics, yet their performance can be affected by matrix effects arising from the sample itself or from external components. Here we systematically evaluate the performance and robustness of cell-free systems in serum, plasma, urine, and saliva using two reporter systems, sfGFP and luciferase. In all cases, clinical samples have a strong inhibitory effect. Of the different inhibitors, only RNase inhibitor mitigated matrix effects. However, we found that the recovery potential of RNase inhibitor was partially muted by interference from glycerol contained in the commercial buffer. We solved this issue by designing a strain producing an RNase inhibitor protein requiring no additional step in extract preparation. Furthermore, our new extract yielded higher reporter levels than previous conditions and tempered interpatient variability associated with matrix effects. This systematic evaluation and improvements of cell-free system robustness unified across many types of clinical samples is a significant step towards developing cell-free diagnostics for a wide range of conditions.
Collapse
|
9
|
Iksanova AM, Arzumanian VG, Konanykhina SY, Samoylikov PV. Antimicrobial peptides and proteins in human biological fluids. MICROBIOLOGY INDEPENDENT RESEARCH JOURNAL 2022. [DOI: 10.18527/2500-2236-2022-9-1-37-55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Antimicrobial peptides and proteins (AMPs) are endogenous compounds that have a direct antimicrobial effect on bacteria (e. g., by disrupting bacterial membranes), as well as on fungi and viruses. AMPs are the main component of the innate immunity of living organisms and are produced by both epithelial cells (skin cells, cells of respiratory tract, intestine, urinary and genital tracts) and cells of the immune system and are secreted into secretory fluids. AMPs can also act as chemoattractants for immunocompetent cells (neutrophils, monocytes, T lymphocytes, dendritic cells) in the inflammation site and affect the antigen presenting cells by modulating adaptive T cell immune responses. The representatives of the main 15 AMP classes, that we describe in this review, are the most studied group of the large pool of these compounds. We discuss their localization, expression, and concentration in various biofluids of humans under normal and pathological conditions.
Collapse
|
10
|
Ribonuclease-1 treatment after traumatic brain injury preserves blood-brain barrier integrity and delays secondary brain damage in mice. Sci Rep 2022; 12:5731. [PMID: 35388024 PMCID: PMC8986812 DOI: 10.1038/s41598-022-09326-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/22/2022] [Indexed: 11/08/2022] Open
Abstract
Traumatic brain injury (TBI) involves primary mechanical damage and delayed secondary damage caused by vascular dysfunction and neuroinflammation. Intracellular components released into the parenchyma and systemic circulation, termed danger-associated molecular patterns (DAMPs), are major drivers of vascular dysfunction and neuroinflammation. These DAMPs include cell-free RNAs (cfRNAs), which damage the blood-brain barrier (BBB), thereby promoting edema, procoagulatory processes, and infiltration of inflammatory cells. We tested the hypothesis that intraperitoneal injection of Ribonuclease-1 (RNase1, two doses of 20, 60, or 180 µg/kg) at 30 min and 12 h after controlled-cortical-impact (CCI) can reduce secondary lesion expansion compared to vehicle treatment 24 h and 120 h post-CCI. The lowest total dose (40 µg/kg) was most effective at reducing lesion volume (- 31% RNase 40 µg/kg vs. vehicle), brain water accumulation (- 5.5%), and loss of BBB integrity (- 21.6%) at 24 h post-CCI. RNase1 also reduced perilesional leukocyte recruitment (- 53.3%) and microglial activation (- 18.3%) at 120 h post-CCI, but there was no difference in lesion volume at this time and no functional benefit. Treatment with RNase1 in the early phase following TBI stabilizes the BBB and impedes leukocyte immigration, thereby suppressing neuroinflammation. RNase1-treatment may be a novel approach to delay brain injury to extend the window for treatment opportunities after TBI.
Collapse
|
11
|
Garnett ER, Raines RT. Emerging biological functions of ribonuclease 1 and angiogenin. Crit Rev Biochem Mol Biol 2021; 57:244-260. [PMID: 34886717 DOI: 10.1080/10409238.2021.2004577] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Pancreatic-type ribonucleases (ptRNases) are a large family of vertebrate-specific secretory endoribonucleases. These enzymes catalyze the degradation of many RNA substrates and thereby mediate a variety of biological functions. Though the homology of ptRNases has informed biochemical characterization and evolutionary analyses, the understanding of their biological roles is incomplete. Here, we review the functions of two ptRNases: RNase 1 and angiogenin. RNase 1, which is an abundant ptRNase with high catalytic activity, has newly discovered roles in inflammation and blood coagulation. Angiogenin, which promotes neovascularization, is now known to play roles in the progression of cancer and amyotrophic lateral sclerosis, as well as in the cellular stress response. Ongoing work is illuminating the biology of these and other ptRNases.
Collapse
Affiliation(s)
- Emily R Garnett
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ronald T Raines
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
12
|
Nucera F, Lo Bello F, Shen SS, Ruggeri P, Coppolino I, Di Stefano A, Stellato C, Casolaro V, Hansbro PM, Adcock IM, Caramori G. Role of Atypical Chemokines and Chemokine Receptors Pathways in the Pathogenesis of COPD. Curr Med Chem 2021; 28:2577-2653. [PMID: 32819230 DOI: 10.2174/0929867327999200819145327] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/11/2020] [Accepted: 06/18/2020] [Indexed: 11/22/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) represents a heightened inflammatory response in the lung generally resulting from tobacco smoking-induced recruitment and activation of inflammatory cells and/or activation of lower airway structural cells. Several mediators can modulate activation and recruitment of these cells, particularly those belonging to the chemokines (conventional and atypical) family. There is emerging evidence for complex roles of atypical chemokines and their receptors (such as high mobility group box 1 (HMGB1), antimicrobial peptides, receptor for advanced glycosylation end products (RAGE) or toll-like receptors (TLRs)) in the pathogenesis of COPD, both in the stable disease and during exacerbations. Modulators of these pathways represent potential novel therapies for COPD and many are now in preclinical development. Inhibition of only a single atypical chemokine or receptor may not block inflammatory processes because there is redundancy in this network. However, there are many animal studies that encourage studies for modulating the atypical chemokine network in COPD. Thus, few pharmaceutical companies maintain a significant interest in developing agents that target these molecules as potential antiinflammatory drugs. Antibody-based (biological) and small molecule drug (SMD)-based therapies targeting atypical chemokines and/or their receptors are mostly at the preclinical stage and their progression to clinical trials is eagerly awaited. These agents will most likely enhance our knowledge about the role of atypical chemokines in COPD pathophysiology and thereby improve COPD management.
Collapse
Affiliation(s)
- Francesco Nucera
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Federica Lo Bello
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Sj S Shen
- Faculty of Science, Centre for Inflammation, Centenary Institute, University of Technology, Ultimo, Sydney, Australia
| | - Paolo Ruggeri
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Irene Coppolino
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Antonino Di Stefano
- Division of Pneumology, Cyto- Immunopathology Laboratory of the Cardio-Respiratory System, Clinical Scientific Institutes Maugeri IRCCS, Veruno, Italy
| | - Cristiana Stellato
- Department of Medicine, Surgery and Dentistry, Salerno Medical School, University of Salerno, Salerno, Italy
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry, Salerno Medical School, University of Salerno, Salerno, Italy
| | - Phil M Hansbro
- Faculty of Science, Centre for Inflammation, Centenary Institute, University of Technology, Ultimo, Sydney, Australia
| | - Ian M Adcock
- Airway Disease Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Gaetano Caramori
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| |
Collapse
|
13
|
Human ribonuclease 1 serves as a secretory ligand of ephrin A4 receptor and induces breast tumor initiation. Nat Commun 2021; 12:2788. [PMID: 33986289 PMCID: PMC8119676 DOI: 10.1038/s41467-021-23075-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 04/09/2021] [Indexed: 12/12/2022] Open
Abstract
Human ribonuclease 1 (hRNase 1) is critical to extracellular RNA clearance and innate immunity to achieve homeostasis and host defense; however, whether it plays a role in cancer remains elusive. Here, we demonstrate that hRNase 1, independently of its ribonucleolytic activity, enriches the stem-like cell population and enhances the tumor-initiating ability of breast cancer cells. Specifically, secretory hRNase 1 binds to and activates the tyrosine kinase receptor ephrin A4 (EphA4) signaling to promote breast tumor initiation in an autocrine/paracrine manner, which is distinct from the classical EphA4-ephrin juxtacrine signaling through contact-dependent cell-cell communication. In addition, analysis of human breast tumor tissue microarrays reveals a positive correlation between hRNase 1, EphA4 activation, and stem cell marker CD133. Notably, high hRNase 1 level in plasma samples is positively associated with EphA4 activation in tumor tissues from breast cancer patients, highlighting the pathological relevance of the hRNase 1-EphA4 axis in breast cancer. The discovery of hRNase 1 as a secretory ligand of EphA4 that enhances breast cancer stemness suggests a potential treatment strategy by inactivating the hRNase 1-EphA4 axis.
Collapse
|
14
|
Preissner KT, Fischer S, Deindl E. Extracellular RNA as a Versatile DAMP and Alarm Signal That Influences Leukocyte Recruitment in Inflammation and Infection. Front Cell Dev Biol 2020; 8:619221. [PMID: 33392206 PMCID: PMC7775424 DOI: 10.3389/fcell.2020.619221] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Upon vascular injury, tissue damage, ischemia, or microbial infection, intracellular material such as nucleic acids and histones is liberated and comes into contact with the vessel wall and circulating blood cells. Such "Danger-associated molecular patterns" (DAMPs) may thus have an enduring influence on the inflammatory defense process that involves leukocyte recruitment and wound healing reactions. While different species of extracellular RNA (exRNA), including microRNAs and long non-coding RNAs, have been implicated to influence inflammatory processes at different levels, recent in vitro and in vivo work has demonstrated a major impact of ribosomal exRNA as a prominent DAMP on various steps of leukocyte recruitment within the innate immune response. This includes the induction of vascular hyper-permeability and vasogenic edema by exRNA via the activation of the "vascular endothelial growth factor" (VEGF) receptor-2 system, as well as the recruitment of leukocytes to the inflamed endothelium, the M1-type polarization of inflammatory macrophages, or the role of exRNA as a pro-thrombotic cofactor to promote thrombosis. Beyond sterile inflammation, exRNA also augments the docking of bacteria to host cells and the subsequent microbial invasion. Moreover, upon vessel occlusion and ischemia, the shear stress-induced release of exRNA initiates arteriogenesis (i.e., formation of natural vessel bypasses) in a multistep process that resembles leukocyte recruitment. Although exRNA can be counteracted for by natural circulating RNase1, under the conditions mentioned, only the administration of exogenous, thermostable, non-toxic RNase1 provides an effective and safe therapeutic regimen for treating the damaging activities of exRNA. It remains to be investigated whether exRNA may also influence viral infections (including COVID-19), e.g., by supporting the interaction of host cells with viral particles and their subsequent invasion. In fact, as a consequence of the viral infection cycle, massive amounts of exRNA are liberated, which can provoke further tissue damage and enhance virus dissemination. Whether the application of RNase1 in this scenario may help to limit the extent of viral infections like COVID-19 and impact on leukocyte recruitment and emigration steps in immune defense in order to limit the extent of associated cardiovascular diseases remains to be studied.
Collapse
Affiliation(s)
- Klaus T. Preissner
- Department of Biochemistry, Medical School, Justus Liebig University Giessen, Giessen, Germany
- Kerckhoff-Heart-Research-Institute, Department of Cardiology, Medical School, Justus Liebig University Giessen, Giessen, Germany
| | - Silvia Fischer
- Department of Biochemistry, Medical School, Justus Liebig University Giessen, Giessen, Germany
| | - Elisabeth Deindl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, LMU Munich, Munich, Germany
| |
Collapse
|
15
|
Lasch M, Kumaraswami K, Nasiscionyte S, Kircher S, van den Heuvel D, Meister S, Ishikawa-Ankerhold H, Deindl E. RNase A Treatment Interferes With Leukocyte Recruitment, Neutrophil Extracellular Trap Formation, and Angiogenesis in Ischemic Muscle Tissue. Front Physiol 2020; 11:576736. [PMID: 33240100 PMCID: PMC7677187 DOI: 10.3389/fphys.2020.576736] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/16/2020] [Indexed: 01/13/2023] Open
Abstract
Background: RNase A (the bovine equivalent to human RNase 1) and RNase 5 (angiogenin) are two closely related ribonucleases. RNase 5 is described as a powerful angiogenic factor. Whether RNase A shares the same angiogenic characteristic, or interferes with vessel growth as demonstrated for arteriogenesis, has never been investigated and is the topic of this present study. Methods and Results: To investigate whether RNase A shows a pro‐ or anti-angiogenic effect, we employed a murine hindlimb model, in which femoral artery ligation (FAL) results in arteriogenesis in the upper leg, and, due to provoked ischemia, in angiogenesis in the lower leg. C57BL/6J male mice underwent unilateral FAL, whereas the contralateral leg was sham operated. Two and seven days after the surgery and intravenous injection of RNase A (50 μg/kg dissolved in saline) or saline (control), the gastrocnemius muscles of mice were isolated from the lower legs for (immuno-) histological analyses. Hematoxylin and Eosin staining evidenced that RNase A treatment resulted in a higher degree of ischemic tissue damage. This was, however, associated with reduced angiogenesis, as evidenced by a reduced capillary/muscle fiber ratio. Moreover, RNase A treatment was associated with a significant reduction in leukocyte infiltration as shown by CD45+ (pan-leukocyte marker), Ly6G+ or MPO+ (neutrophils), MPO+/CitH3+ [neutrophil extracellular traps (NETs)], and CD68+ (macrophages) staining. CD68/MRC1 double staining revealed that RNase A treated mice showed a reduced percentage of M1-like polarized (CD68+/MRC1−) macrophages whereas the percentage of M2-like polarized (CD68+/MRC1+) macrophages was increased. Conclusion: In contrast to RNase 5, RNase A interferes with angiogenesis, which is linked to reduced leukocyte infiltration and NET formation.
Collapse
Affiliation(s)
- Manuel Lasch
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.,Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Konda Kumaraswami
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.,Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Simona Nasiscionyte
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Susanna Kircher
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.,Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Dominic van den Heuvel
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sarah Meister
- Department of Obstetrics and Gynaecology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Hellen Ishikawa-Ankerhold
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Internal Medicine I, Faculty of Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Elisabeth Deindl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.,Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
16
|
Bedenbender K, Beinborn I, Vollmeister E, Schmeck B. p38 and Casein Kinase 2 Mediate Ribonuclease 1 Repression in Inflamed Human Endothelial Cells via Promoter Remodeling Through Nucleosome Remodeling and Deacetylase Complex. Front Cell Dev Biol 2020; 8:563604. [PMID: 33178683 PMCID: PMC7593526 DOI: 10.3389/fcell.2020.563604] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/28/2020] [Indexed: 11/13/2022] Open
Abstract
Vascular pathologies, such as thrombosis or atherosclerosis, are leading causes of death worldwide and are strongly associated with the dysfunction of vascular endothelial cells. In this context, the extracellular endonuclease Ribonuclease 1 (RNase1) acts as an essential protective factor in regulation and maintenance of vascular homeostasis. However, long-term inflammation causes strong repression of RNase1 expression, thereby promoting endothelial cell dysfunction. This inflammation-mediated downregulation of RNase1 in human endothelial cells is facilitated via histone deacetylase (HDAC) 2, although the underlying molecular mechanisms are still unknown. Here, we report that inhibition of c-Jun N-terminal kinase by small chemical compounds in primary human endothelial cells decreased physiological RNase1 mRNA abundance, while p38 kinase inhibition restored repressed RNase1 expression upon proinflammatory stimulation with tumor necrosis factor alpha (TNF-α) and poly I:C. Moreover, blocking of the p38 kinase- and HDAC2-associated kinase casein kinase 2 (CK2) by inhibitor as well as small interfering RNA (siRNA)-knockdown restored RNase1 expression upon inflammation of human endothelial cells. Further downstream, siRNA-knockdown of chromodomain helicase DNA binding protein (CHD) 3 and 4 of the nucleosome remodeling and deacetylase (NuRD) complex restored RNase1 repression in TNF-α treated endothelial cells implicating its role in the HDAC2-containing repressor complex involved in RNase1 repression. Finally, chromatin immunoprecipitation in primary human endothelial cells confirmed recruitment of the CHD4-containing NuRD complex and subsequent promoter remodeling via histone deacetylation at the RNASE1 promoter in a p38-dependent manner upon human endothelial cell inflammation. Altogether, our results suggest that endothelial RNase1 repression in chronic vascular inflammation is regulated by a p38 kinase-, CK2-, and NuRD complex-dependent pathway resulting in complex recruitment to the RNASE1 promoter and subsequent promoter remodeling.
Collapse
Affiliation(s)
- Katrin Bedenbender
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Marburg, Germany
| | - Isabell Beinborn
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Marburg, Germany
| | - Evelyn Vollmeister
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Marburg, Germany
| | - Bernd Schmeck
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Marburg, Germany.,Department of Pulmonary and Critical Care Medicine, Department of Medicine, University Medical Center Giessen and Marburg, Philipps-University Marburg, Marburg, Germany.,Member of the German Center for Lung Research, Member of the German Center for Infectious Disease Research, Marburg, Germany.,Center for Synthetic Microbiology, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
17
|
Bedenbender K, Schmeck BT. Endothelial Ribonuclease 1 in Cardiovascular and Systemic Inflammation. Front Cell Dev Biol 2020; 8:576491. [PMID: 33015070 PMCID: PMC7500176 DOI: 10.3389/fcell.2020.576491] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022] Open
Abstract
The vascular endothelial cell layer forms the inner lining of all blood vessels to maintain proper functioning of the vascular system. However, dysfunction of the endothelium depicts a major issue in context of vascular pathologies, such as atherosclerosis or thrombosis that cause several million deaths per year worldwide. In recent years, the endothelial extracellular endonuclease Ribonuclease 1 (RNase1) was described as a key player in regulation of vascular homeostasis by protecting endothelial cells from detrimental effects of the damage-associated molecular pattern extracellular RNA upon acute inflammation. Despite this protective function, massive dysregulation of RNase1 was observed during prolonged endothelial cell inflammation resulting in progression of several vascular diseases. For the first time, this review article outlines the current knowledge on endothelial RNase1 and its role in function and dysfunction of the endothelium, thereby focusing on the intensive research from recent years: Uncovering the underlying mechanisms of RNase1 function and regulation in response to acute as well as long-term inflammation, the role of RNase1 in context of vascular, inflammatory and infectious diseases and the potential to develop novel therapeutic options to treat these pathologies against the background of RNase1 function in endothelial cells.
Collapse
Affiliation(s)
- Katrin Bedenbender
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Marburg, Germany
| | - Bernd T. Schmeck
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Marburg, Germany
- Department of Pulmonary and Critical Care Medicine, Department of Medicine, University Medical Center Giessen and Marburg, Philipps-University Marburg, Marburg, Germany
- Member of the German Center for Lung Research, Member of the German Center for Infectious Disease Research, Marburg, Germany
- Center for Synthetic Microbiology, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
18
|
Rangel-Muñoz N, Suarez-Arnedo A, Anguita R, Prats-Ejarque G, Osma JF, Muñoz-Camargo C, Boix E, Cruz JC, Salazar VA. Magnetite Nanoparticles Functionalized with RNases against Intracellular Infection of Pseudomonas aeruginosa. Pharmaceutics 2020; 12:E631. [PMID: 32640506 PMCID: PMC7408537 DOI: 10.3390/pharmaceutics12070631] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 12/15/2022] Open
Abstract
Current treatments against bacterial infections have severe limitations, mainly due to the emergence of resistance to conventional antibiotics. In the specific case of Pseudomonas aeruginosa strains, they have shown a number of resistance mechanisms to counter most antibiotics. Human secretory RNases from the RNase A superfamily are proteins involved in a wide variety of biological functions, including antimicrobial activity. The objective of this work was to explore the intracellular antimicrobial action of an RNase 3/1 hybrid protein that combines RNase 1 high catalytic and RNase 3 bactericidal activities. To achieve this, we immobilized the RNase 3/1 hybrid on Polyetheramine (PEA)-modified magnetite nanoparticles (MNPs). The obtained nanobioconjugates were tested in macrophage-derived THP-1 cells infected with Pseudomonas aeruginosa PAO1. The obtained results show high antimicrobial activity of the functionalized hybrid protein (MNP-RNase 3/1) against the intracellular growth of P. aeruginosa of the functionalized hybrid protein. Moreover, the immobilization of RNase 3/1 enhances its antimicrobial and cell-penetrating activities without generating any significant cell damage. Considering the observed antibacterial activity, the immobilization of the RNase A superfamily and derived proteins represents an innovative approach for the development of new strategies using nanoparticles to deliver antimicrobials that counteract P. aeruginosa intracellular infection.
Collapse
Affiliation(s)
- Nathaly Rangel-Muñoz
- Department of Biomedical Engineering, Universidad de los Andes, Cra. 1E No. 19a-40, Bogotá 111711, Colombia; (N.R.-M.); (A.S.-A.); (C.M.-C.)
| | - Alejandra Suarez-Arnedo
- Department of Biomedical Engineering, Universidad de los Andes, Cra. 1E No. 19a-40, Bogotá 111711, Colombia; (N.R.-M.); (A.S.-A.); (C.M.-C.)
| | - Raúl Anguita
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (R.A.); (G.P.-E.)
| | - Guillem Prats-Ejarque
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (R.A.); (G.P.-E.)
| | - Johann F. Osma
- Department of Electrical and Electronics Engineering, Universidad de los Andes, Cra. 1E No. 19a-40, Bogotá 111711, Colombia;
| | - Carolina Muñoz-Camargo
- Department of Biomedical Engineering, Universidad de los Andes, Cra. 1E No. 19a-40, Bogotá 111711, Colombia; (N.R.-M.); (A.S.-A.); (C.M.-C.)
| | - Ester Boix
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (R.A.); (G.P.-E.)
| | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Cra. 1E No. 19a-40, Bogotá 111711, Colombia; (N.R.-M.); (A.S.-A.); (C.M.-C.)
| | - Vivian A. Salazar
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (R.A.); (G.P.-E.)
- Department of Electrical and Electronics Engineering, Universidad de los Andes, Cra. 1E No. 19a-40, Bogotá 111711, Colombia;
| |
Collapse
|
19
|
Kosgey JC, Jia L, Nyamao RM, Zhao Y, Xue T, Yang J, Fang Y, Zhang F. RNase 1, 2, 5 & 8 role in innate immunity: Strain specific antimicrobial activity. Int J Biol Macromol 2020; 160:1042-1049. [PMID: 32504708 DOI: 10.1016/j.ijbiomac.2020.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/31/2020] [Accepted: 06/01/2020] [Indexed: 01/26/2023]
Abstract
The increase in microbial resistance to conventional antimicrobial agents is driving research for the discovery of new antibiotics and antifungal agents. The greatest challenge in this endeavor is to find antimicrobial agents with broad antimicrobial activity and low toxicity. Antimicrobial peptides, for example, RNases, are one of the promising areas. The production of RNases increases during infection, but their role is still being explored. Whereas the enzymatic activity of RNases is well documented, their physiological function is still being investigated. This study aimed to evaluate the antimicrobial activity of RNase 1, 2, 5, and 8 against E. coli strains, S. aureus, Streptococcus thermophilus, P. aeruginosa, Candida albicans, and Candida glabrata. The results demonstrated that RNases have a strain-specific antimicrobial activity. RNase 1 had the highest antimicrobial activity compared to other RNases. All the microorganisms screened had varying levels of susceptibility to RNases, except P. aeruginosa and E. coli DR115. RNase 1 showed dose-dependent activity against C. albicans. The RNase killed Candida albicans by lowering the mitochondrial membrane potential but did not damage the cell membrane. We concluded that strain-specific antimicrobial activity is one of the physiological roles of RNases.
Collapse
Affiliation(s)
- Janet Cheruiyot Kosgey
- School of Biological and Life Sciences, The Technical University of Kenya, 52428-00200, Kenya; Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin 150086, China
| | - Lina Jia
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin 150086, China
| | - Rose Magoma Nyamao
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin 150086, China; School of Medicine, Kenyatta University, 43844, 00100, Kenya
| | - Yi Zhao
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin 150086, China
| | - Teng Xue
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin 150086, China
| | - Jianxun Yang
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin 150086, China; Department of Dermatology, The 2nd Hospital of Harbin Medical University, Harbin 150086, China
| | - Yong Fang
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin 150086, China
| | - Fengmin Zhang
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin 150086, China.
| |
Collapse
|
20
|
Zhang H, Rombouts K, Raes L, Xiong R, De Smedt SC, Braeckmans K, Remaut K. Fluorescence-Based Quantification of Messenger RNA and Plasmid DNA Decay Kinetics in Extracellular Biological Fluids and Cell Extracts. ACTA ACUST UNITED AC 2020; 4:e2000057. [PMID: 32402121 DOI: 10.1002/adbi.202000057] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 02/26/2020] [Indexed: 12/22/2022]
Abstract
Extracellular and intracellular degradation of nucleic acids remains an issue in non-viral gene therapy. Understanding biodegradation is critical for the rational design of gene therapeutics in order to maintain stability and functionality at the target site. However, there are only limited methods available that allow determining the stability of genetic materials in biological environments. In this context, the decay kinetics of fluorescently labeled plasmid DNA (pDNA) and messenger RNA (mRNA) in undiluted biological samples (i.e., human serum, human ascites, bovine vitreous) and cell extracts is studied using fluorescence correlation spectroscopy (FCS) and single particle tracking (SPT). It is demonstrated that FCS is suitable to follow mRNA degradation, while SPT is better suited to investigate pDNA integrity. The half-life of mRNA and pDNA is ≈1-2 min and 1-4 h in biological samples, respectively. The resistance against biodegradation drastically improves by complexation with lipid-based carriers. Taken together, FCS and SPT are able to quantify the integrity of mRNA and pDNA, respectively, as a function of time, both in the extracellular biological fluids and cell extracts. This in turn allows to focus on the important but less understood issue of nucleic acids degradation in more detail and to rationally optimize gene delivery system as therapeutics.
Collapse
Affiliation(s)
- Heyang Zhang
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Koen Rombouts
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Laurens Raes
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Ranhua Xiong
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Katrien Remaut
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| |
Collapse
|
21
|
Kluever AK, Braumandl A, Fischer S, Preissner KT, Deindl E. The Extraordinary Role of Extracellular RNA in Arteriogenesis, the Growth of Collateral Arteries. Int J Mol Sci 2019; 20:ijms20246177. [PMID: 31817879 PMCID: PMC6940760 DOI: 10.3390/ijms20246177] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/05/2019] [Accepted: 12/06/2019] [Indexed: 01/13/2023] Open
Abstract
Arteriogenesis is an intricate process in which increased shear stress in pre-existing arteriolar collaterals induces blood vessel expansion, mediated via endothelial cell activation, leukocyte recruitment and subsequent endothelial and smooth muscle cell proliferation. Extracellular RNA (eRNA), released from stressed cells or damaged tissue under pathological conditions, has recently been discovered to be liberated from endothelial cells in response to increased shear stress and to promote collateral growth. Until now, eRNA has been shown to enhance coagulation and inflammation by inducing cytokine release, leukocyte recruitment, and endothelial permeability, the latter being mediated by vascular endothelial growth factor (VEGF) signaling. In the context of arteriogenesis, however, eRNA has emerged as a transmitter of shear stress into endothelial activation, mediating the sterile inflammatory process essential for collateral remodeling, whereby the stimulatory effects of eRNA on the VEGF signaling axis seem to be pivotal. In addition, eRNA might influence subsequent steps of the arteriogenesis cascade as well. This article provides a comprehensive overview of the beneficial effects of eRNA during arteriogenesis, laying the foundation for further exploration of the connection between the damaging and non-damaging effects of eRNA in the context of cardiovascular occlusive diseases and of sterile inflammation.
Collapse
Affiliation(s)
- Anna-Kristina Kluever
- Walter-Brendel-Center of Experimental Medicine, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (A.-K.K.); (A.B.)
| | - Anna Braumandl
- Walter-Brendel-Center of Experimental Medicine, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (A.-K.K.); (A.B.)
| | - Silvia Fischer
- Institute of Biochemistry, Medical School, Justus-Liebig-University, 35392 Giessen, Germany; (S.F.); (K.T.P.)
| | - Klaus T. Preissner
- Institute of Biochemistry, Medical School, Justus-Liebig-University, 35392 Giessen, Germany; (S.F.); (K.T.P.)
| | - Elisabeth Deindl
- Walter-Brendel-Center of Experimental Medicine, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (A.-K.K.); (A.B.)
- Correspondence: ; Tel.: +49-89-2180-76504
| |
Collapse
|
22
|
Garnett ER, Lomax JE, Mohammed BM, Gailani D, Sheehan JP, Raines RT. Phenotype of ribonuclease 1 deficiency in mice. RNA (NEW YORK, N.Y.) 2019; 25:921-934. [PMID: 31053653 PMCID: PMC6633200 DOI: 10.1261/rna.070433.119] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 04/27/2019] [Indexed: 05/06/2023]
Abstract
Biological roles for extracellular RNA (eRNA) have become apparent. For example, eRNA can induce contact activation in blood via activation of the plasma proteases factor XII (FXII) and factor XI (FXI). We sought to reveal the biological role of the secretory enzyme ribonuclease 1 (RNase 1) in an organismal context by generating and analyzing RNase 1 knockout (Rnase1-/-) mice. We found that these mice are viable, healthy, and fertile, though larger than Rnase1+/+ mice. Rnase1-/- plasma contains more RNA than does the plasma of Rnase1+/+ mice. Moreover, the plasma of Rnase1-/- mice clots more rapidly than does wild-type plasma. This phenotype appeared to be due to increased levels of the active form of FXII (FXIIa) in the plasma of Rnase1-/- mice compared to Rnase1+/+ mice, and is consistent with the known effects of eRNA on FXII activation. The apparent activity of FXI in the plasma of Rnase1-/- mice was 1000-fold higher when measured in an assay triggered by a low concentration of tissue factor than in assays based on recalcification, consistent with eRNA enhancing FXI activation by thrombin. These findings suggest that one of the physiological functions of RNase 1 is to degrade eRNA in blood plasma. Loss of this function facilitates FXII and FXI activation, which could have effects on inflammation and blood coagulation. We anticipate that Rnase1-/- mice will be a useful tool for evaluating other hypotheses about the functions of RNase 1 and of eRNA in vivo.
Collapse
Affiliation(s)
- Emily R Garnett
- Graduate Program in Molecular and Cellular Pharmacology, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Jo E Lomax
- Graduate Program Molecular and Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Bassem M Mohammed
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - David Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - John P Sheehan
- Department of Medicine/Hematology-Oncology, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Ronald T Raines
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| |
Collapse
|
23
|
Bedenbender K, Scheller N, Fischer S, Leiting S, Preissner KT, Schmeck BT, Vollmeister E. Inflammation-mediated deacetylation of the ribonuclease 1 promoter via histone deacetylase 2 in endothelial cells. FASEB J 2019; 33:9017-9029. [PMID: 31039328 DOI: 10.1096/fj.201900451r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ribonuclease 1 (RNase1) is a circulating extracellular endonuclease that regulates the vascular homeostasis of extracellular RNA and acts as a vessel- and tissue-protective enzyme. Upon long-term inflammation, high amounts of proinflammatory cytokines affect endothelial cell (EC) function by down-regulation of RNase1. Here, we investigated the transcriptional regulation of RNase1 upon inflammation in HUVECs. TNF-α or IL-1β stimulation reduced the expression of RNase1 relative to the acetylation state of histone 3 at lysine 27 and histone 4 of the RNASE1 promoter. Inhibition of histone deacetylase (HDAC) 1, 2, and 3 by the specific class I HDAC inhibitor MS275 abolished the TNF-α- or IL-1β-mediated effect on the mRNA and chromatin levels of RNase1. Moreover, chromatin immunoprecipitation kinetics revealed that HDAC2 accumulates at the RNASE1 promoter upon TNF-α stimulation, indicating an essential role for HDAC2 in regulating RNase1 expression. Thus, proinflammatory stimulation induced recruitment of HDAC2 to attenuate histone acetylation at the RNASE1 promoter site. Consequently, treatment with HDAC inhibitors may provide a new therapeutic strategy to stabilize vascular homeostasis in the context of inflammation by preventing RNase1 down-regulation in ECs.-Bedenbender, K., Scheller, N., Fischer, S., Leiting, S., Preissner, K. T., Schmeck, B. T., Vollmeister, E. Inflammation-mediated deacetylation of the ribonuclease 1 promoter via histone deacetylase 2 in endothelial cells.
Collapse
Affiliation(s)
- Katrin Bedenbender
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Nicoletta Scheller
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Silvia Fischer
- Department of Biochemistry, Faculty of Medicine, Justus-Liebig-University, Giessen, Germany
| | - Silke Leiting
- Department of Biochemistry, Faculty of Medicine, Justus-Liebig-University, Giessen, Germany
| | - Klaus T Preissner
- Department of Biochemistry, Faculty of Medicine, Justus-Liebig-University, Giessen, Germany
| | - Bernd T Schmeck
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany.,Department of Pulmonary and Critical Care Medicine, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Evelyn Vollmeister
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| |
Collapse
|
24
|
Lee HH, Wang YN, Hung MC. Functional roles of the human ribonuclease A superfamily in RNA metabolism and membrane receptor biology. Mol Aspects Med 2019; 70:106-116. [PMID: 30902663 DOI: 10.1016/j.mam.2019.03.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 03/17/2019] [Indexed: 02/08/2023]
Abstract
The human ribonuclease A (hRNase A) superfamily is comprised of 13 members of secretory RNases, most of which are recognized as catabolic enzymes for their ribonucleolytic activity to degrade ribonucleic acids (RNAs) in the extracellular space, where they play a role in innate host defense and physiological homeostasis. Interestingly, human RNases 9-13, which belong to a non-canonical subgroup of the hRNase A superfamily, are ribonucleolytic activity-deficient proteins with unclear biological functions. Moreover, accumulating evidence indicates that secretory RNases, such as human RNase 5, can be internalized into cells facilitated by membrane receptors like the epidermal growth factor receptor to regulate intracellular RNA species, in particular non-coding RNAs, and signaling pathways by either a ribonucleolytic activity-dependent or -independent manner. In this review, we summarize the classical role of hRNase A superfamily in the metabolism of extracellular and intracellular RNAs and update its non-classical function as a cognate ligand of membrane receptors. We further discuss the biological significance and translational potential of using secretory RNases as predictive biomarkers or therapeutic agents in certain human diseases and the pathological settings for future investigations.
Collapse
Affiliation(s)
- Heng-Huan Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Unit 108, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Ying-Nai Wang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Unit 108, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Unit 108, 1515 Holcombe Boulevard, Houston, TX, 77030, USA; Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung, 404, Taiwan; Department of Biotechnology, Asia University, Taichung 413, Taiwan.
| |
Collapse
|
25
|
Circulating mediators of remote ischemic preconditioning: search for the missing link between non-lethal ischemia and cardioprotection. Oncotarget 2019; 10:216-244. [PMID: 30719216 PMCID: PMC6349428 DOI: 10.18632/oncotarget.26537] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 12/10/2018] [Indexed: 12/11/2022] Open
Abstract
Acute myocardial infarction (AMI) is one of the leading causes of mortality and morbidity worldwide. There has been an extensive search for cardioprotective therapies to reduce myocardial ischemia-reperfusion (I/R) injury. Remote ischemic preconditioning (RIPC) is a phenomenon that relies on the body's endogenous protective modalities against I/R injury. In RIPC, non-lethal brief I/R of one organ or tissue confers protection against subsequent lethal I/R injury in an organ remote to the briefly ischemic organ or tissue. Initially it was believed to be limited to direct myocardial protection, however it soon became apparent that RIPC applied to other organs such as kidney, liver, intestine, skeletal muscle can reduce myocardial infarct size. Intriguing discoveries have been made in extending the concept of RIPC to other organs than the heart. Over the years, the underlying mechanisms of RIPC have been widely sought and discussed. The involvement of blood-borne factors as mediators of RIPC has been suggested by a number of research groups. The main purpose of this review article is to summarize the possible circulating mediators of RIPC, and recent studies to establish the clinical efficacy of these mediators in cardioprotection from lethal I/R injury.
Collapse
|
26
|
Lu L, Li J, Moussaoui M, Boix E. Immune Modulation by Human Secreted RNases at the Extracellular Space. Front Immunol 2018; 9:1012. [PMID: 29867984 PMCID: PMC5964141 DOI: 10.3389/fimmu.2018.01012] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/23/2018] [Indexed: 12/23/2022] Open
Abstract
The ribonuclease A superfamily is a vertebrate-specific family of proteins that encompasses eight functional members in humans. The proteins are secreted by diverse innate immune cells, from blood cells to epithelial cells and their levels in our body fluids correlate with infection and inflammation processes. Recent studies ascribe a prominent role to secretory RNases in the extracellular space. Extracellular RNases endowed with immuno-modulatory and antimicrobial properties can participate in a wide variety of host defense tasks, from performing cellular housekeeping to maintaining body fluid sterility. Their expression and secretion are induced in response to a variety of injury stimuli. The secreted proteins can target damaged cells and facilitate their removal from the focus of infection or inflammation. Following tissue damage, RNases can participate in clearing RNA from cellular debris or work as signaling molecules to regulate the host response and contribute to tissue remodeling and repair. We provide here an overall perspective on the current knowledge of human RNases’ biological properties and their role in health and disease. The review also includes a brief description of other vertebrate family members and unrelated extracellular RNases that share common mechanisms of action. A better knowledge of RNase mechanism of actions and an understanding of their physiological roles should facilitate the development of novel therapeutics.
Collapse
Affiliation(s)
- Lu Lu
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Jiarui Li
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Mohammed Moussaoui
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Ester Boix
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| |
Collapse
|
27
|
Salehi ASM, Yang SO, Earl CC, Shakalli Tang MJ, Porter Hunt J, Smith MT, Wood DW, Bundy BC. Biosensing estrogenic endocrine disruptors in human blood and urine: A RAPID cell-free protein synthesis approach. Toxicol Appl Pharmacol 2018; 345:19-25. [PMID: 29499249 DOI: 10.1016/j.taap.2018.02.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 01/26/2018] [Accepted: 02/23/2018] [Indexed: 11/30/2022]
Abstract
Many diseases and disorders are linked to exposure to endocrine disrupting chemicals (EDCs) that mimic the function of natural estrogen hormones. Here we present a Rapid Adaptable Portable In-vitro Detection biosensor platform (RAPID) for detecting chemicals that interact with the human estrogen receptor β (hERβ). This biosensor consists of an allosteric fusion protein, which is expressed using cell-free protein synthesis technology and is directly assayed by a colorimetric response. The resultant biosensor successfully detected known EDCs of hERβ (BPA, E2, and DPN) at similar or better detection range than an analogous cell-based biosensor, but in a fraction of time. We also engineered cell-free protein synthesis reactions with RNAse inhibitors to increase production yields in the presence of human blood and urine. The RAPID biosensor successfully detects EDCs in these human samples in the presence of RNAse inhibitors. Engineered cell-free protein synthesis facilitates the use of protein biosensors in complex sample matrices without cumbersome protein purification.
Collapse
Affiliation(s)
- Amin S M Salehi
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| | - Seung Ook Yang
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| | - Conner C Earl
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| | - Miriam J Shakalli Tang
- Department of Chemical and Biomolecular Engineering, Ohio State University, Columbus, OH, USA
| | - J Porter Hunt
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| | - Mark T Smith
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| | - David W Wood
- Department of Chemical and Biomolecular Engineering, Ohio State University, Columbus, OH, USA.
| | - Bradley C Bundy
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA.
| |
Collapse
|
28
|
Ohashi A, Murata A, Cho Y, Ichinose S, Sakamaki Y, Nishio M, Hoshi O, Fischer S, Preissner KT, Koyama T. The expression and localization of RNase and RNase inhibitor in blood cells and vascular endothelial cells in homeostasis of the vascular system. PLoS One 2017; 12:e0174237. [PMID: 28329009 PMCID: PMC5362223 DOI: 10.1371/journal.pone.0174237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/06/2017] [Indexed: 02/02/2023] Open
Abstract
RNA may be released from vascular cells including endothelial cells in the event of injury and in vascular disease. Extracellular RNAs have been recognized as novel procoagulant and permeability-increasing factors. Extracellular RNA may function as inflammatory host alarm signals that serve to amplify the defense mechanism, but it may provide important links to thrombus formation. Extracellular RNA is degraded by RNase. We propose that RNase and its inhibitor RNase inhibitor (RI) act as modulators of homeostasis in the vasculature to control the functions of extracellular RNA. We aimed to investigate the expression and localization of RNase 1 and RI in cells that contact blood, such as platelets, mononuclear cells, polymorphonuclear cells, and red blood cells. RNase 1 and RI expression and localization in blood cells were compared with those in the human umbilical vein endothelial cell line, EAhy926. Additionally, we further investigated the effect of thrombin on the expression of RNase 1 and RI in platelets. We used an RNase activity assay, reverse transcription-polymerase chain reaction, western blot, immunocytochemistry, transmission electron microscopy, and immunoelectron microscopy (pre- and post-embedding methods). RNase activity in the supernatant from EAhy926 cells was 50 times than in blood cells (after 60 min). RNase 1 mRNA and protein expression in EAhy926 cells was highest among the cells examined. However, RI mRNA and protein expression was similar in most cell types examined. Furthermore, we observed that RNase 1 and von Willebrand factor were partially colocalized in EAhy926 cells and platelets. In conclusion, we propose that high RNase activity is ordinarily released from endothelial cells to support anticoagulation in the vasculature. On the other hand, platelets and leukocytes within thrombi at sites of vascular injury show very low RNase activity, which may support hemostatic thrombus formation. However, activated platelets and leukocytes may accelerate pathologic thrombus formation.
Collapse
Affiliation(s)
- Ayaka Ohashi
- Laboratory Molecular Genetics of Hematology, Field of Applied Laboratory Science, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Aya Murata
- Laboratory Molecular Genetics of Hematology, Field of Applied Laboratory Science, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuichiro Cho
- Anatomy and Physiological Science, Field of Applied Laboratory Science, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shizuko Ichinose
- Instrumental Analysis Research Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuriko Sakamaki
- Instrumental Analysis Research Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Miwako Nishio
- Laboratory Molecular Genetics of Hematology, Field of Applied Laboratory Science, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Osamu Hoshi
- Anatomy and Physiological Science, Field of Applied Laboratory Science, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Silvia Fischer
- Institute for Biochemistry, Medical Faculty, Justus-Liebig-Universität, Giessen, Germany
| | - Klaus T. Preissner
- Institute for Biochemistry, Medical Faculty, Justus-Liebig-Universität, Giessen, Germany
| | - Takatoshi Koyama
- Laboratory Molecular Genetics of Hematology, Field of Applied Laboratory Science, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- * E-mail:
| |
Collapse
|
29
|
Bornert O, Peking P, Bremer J, Koller U, van den Akker PC, Aartsma-Rus A, Pasmooij AMG, Murauer EM, Nyström A. RNA-based therapies for genodermatoses. Exp Dermatol 2017; 26:3-10. [PMID: 27376675 PMCID: PMC5593095 DOI: 10.1111/exd.13141] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2016] [Indexed: 12/14/2022]
Abstract
Genetic disorders affecting the skin, genodermatoses, constitute a large and heterogeneous group of diseases, for which treatment is generally limited to management of symptoms. RNA-based therapies are emerging as a powerful tool to treat genodermatoses. In this review, we discuss in detail RNA splicing modulation by antisense oligonucleotides and RNA trans-splicing, transcript replacement and genome editing by in vitro-transcribed mRNAs, and gene knockdown by small interfering RNA and antisense oligonucleotides. We present the current state of these therapeutic approaches and critically discuss their opportunities, limitations and the challenges that remain to be solved. The aim of this review was to set the stage for the development of new and better therapies to improve the lives of patients and families affected by a genodermatosis.
Collapse
Affiliation(s)
- Olivier Bornert
- Department of Dermatology, Medical Center – University of
Freiburg, Freiburg, Germany
| | - Patricia Peking
- EB House Austria, Research Program for Molecular Therapy of
Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus
Medical University, Salzburg, Austria
| | - Jeroen Bremer
- Department of Dermatology, University Medical Center Groningen,
University of Groningen, Groningen, The Netherlands
| | - Ulrich Koller
- EB House Austria, Research Program for Molecular Therapy of
Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus
Medical University, Salzburg, Austria
| | - Peter C. van den Akker
- Department of Dermatology, University Medical Center Groningen,
University of Groningen, Groningen, The Netherlands
- Department of Genetics, University Medical Center Groningen,
University of Groningen, Groningen, The Netherlands
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center,
Leiden, The Netherlands
| | - Anna M. G. Pasmooij
- Department of Dermatology, University Medical Center Groningen,
University of Groningen, Groningen, The Netherlands
| | - Eva M. Murauer
- EB House Austria, Research Program for Molecular Therapy of
Genodermatoses, Department of Dermatology, University Hospital of the Paracelsus
Medical University, Salzburg, Austria
| | - Alexander Nyström
- Department of Dermatology, Medical Center – University of
Freiburg, Freiburg, Germany
| |
Collapse
|
30
|
Thomas SP, Kim E, Kim JS, Raines RT. Knockout of the Ribonuclease Inhibitor Gene Leaves Human Cells Vulnerable to Secretory Ribonucleases. Biochemistry 2016; 55:6359-6362. [PMID: 27806571 DOI: 10.1021/acs.biochem.6b01003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Ribonuclease inhibitor (RNH1) is a cytosolic protein that binds with femtomolar affinity to human ribonuclease 1 (RNase 1) and homologous secretory ribonucleases. RNH1 contains 32 cysteine residues and has been implicated as an antioxidant. Here, we use CRISPR-Cas9 to knock out RNH1 in HeLa cells. We find that cellular RNH1 affords marked protection from the lethal ribonucleolytic activity of RNase 1 but not from oxidants. We conclude that RNH1 protects cytosolic RNA from invading ribonucleases.
Collapse
Affiliation(s)
- Sydney P Thomas
- Graduate Program in Cellular & Molecular Biology, University of Wisconsin-Madison , 1525 Linden Drive, Madison, Wisconsin 53706, United States
| | - Eunji Kim
- Center for Genome Engineering, Institute for Basic Science , Seoul 08826, Republic of Korea
| | - Jin-Soo Kim
- Center for Genome Engineering, Institute for Basic Science , Seoul 08826, Republic of Korea
| | - Ronald T Raines
- Department of Biochemistry, University of Wisconsin-Madison , 433 Babcock Drive, Madison, Wisconsin 53706, United States.,Department of Chemistry, University of Wisconsin-Madison , 1101 University Avenue, Madison, Wisconsin 53706, United States
| |
Collapse
|
31
|
Koczera P, Martin L, Marx G, Schuerholz T. The Ribonuclease A Superfamily in Humans: Canonical RNases as the Buttress of Innate Immunity. Int J Mol Sci 2016; 17:ijms17081278. [PMID: 27527162 PMCID: PMC5000675 DOI: 10.3390/ijms17081278] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/29/2016] [Accepted: 08/01/2016] [Indexed: 12/18/2022] Open
Abstract
In humans, the ribonuclease A (RNase A) superfamily contains eight different members that have RNase activities, and all of these members are encoded on chromosome 14. The proteins are secreted by a large variety of different tissues and cells; however, a comprehensive understanding of these proteins’ physiological roles is lacking. Different biological effects can be attributed to each protein, including antiviral, antibacterial and antifungal activities as well as cytotoxic effects against host cells and parasites. Different immunomodulatory effects have also been demonstrated. This review summarizes the available data on the human RNase A superfamily and illustrates the significant role of the eight canonical RNases in inflammation and the host defence system against infections.
Collapse
Affiliation(s)
- Patrick Koczera
- Department of Intensive Care and Intermediate Care, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen 52074, Germany.
- Department for Experimental Molecular Imaging, University Hospital RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen 52074, Germany.
| | - Lukas Martin
- Department of Intensive Care and Intermediate Care, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen 52074, Germany.
| | - Gernot Marx
- Department of Intensive Care and Intermediate Care, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen 52074, Germany.
| | - Tobias Schuerholz
- Department of Intensive Care and Intermediate Care, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen 52074, Germany.
| |
Collapse
|
32
|
Kleinert E, Langenmayer MC, Reichart B, Kindermann J, Griemert B, Blutke A, Troidl K, Mayr T, Grantzow T, Noyan F, Abicht JM, Fischer S, Preissner KT, Wanke R, Deindl E, Guethoff S. Ribonuclease (RNase) Prolongs Survival of Grafts in Experimental Heart Transplantation. J Am Heart Assoc 2016; 5:e003429. [PMID: 27121849 PMCID: PMC4889206 DOI: 10.1161/jaha.116.003429] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 03/03/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cell damage, tissue and vascular injury are associated with the exposure and release of intracellular components such as RNA, which promote inflammatory reactions and thrombosis. Based on the counteracting anti-inflammatory and cardioprotective functions of ribonuclease A (RNase A) in this context, its role in an experimental model of heart transplantation in rats was studied. METHODS AND RESULTS Inbred BN/OrlRj rat cardiac allografts were heterotopically transplanted into inbred LEW/OrlRj rats. Recipients were intravenously treated every other day with saline or bovine pancreatic RNase A (50 μg/kg). Toxic side effects were not found (macroscopically and histologically). Heart tissue flow cytometry and quantitative morphological analyses of explanted hearts at postoperative day 1 or postoperative day 4 showed reduced leukocyte infiltration, edema, and thrombus formation in RNase A-treated rats. In allogeneic mixed lymphocyte reactions, RNase A decreased the proliferation of effector T cells. RNase A treatment of rats resulted in prolonged median graft survival up to 10.5 days (interquartile range 1.8) compared to 6.5 days (interquartile range 1.0) in saline treatment (P=0.001). Treatment of rats with a new generated (recombinant) human pancreatic RNase 1 prolonged median graft survival similarly, unlike treatment with (recombinant) inactive human RNase 1 (each 50 μg/kg IV every other day, 11.0 days, interquartile range 0.3, versus 8.0 days, interquartile range 0.5, P=0.007). CONCLUSIONS Upon heart transplantation, RNase administration appears to present a promising and safe drug to counteract ischemia/reperfusion injury and graft rejection. Furthermore, RNase treatment may be considered in situations of critical reperfusion after percutaneous coronary interventions or in cardiac surgery using the heart-lung machine.
Collapse
Affiliation(s)
- Eike Kleinert
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Germany
| | - Martin C Langenmayer
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Germany Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-Universität München, Germany
| | - Bruno Reichart
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Germany
| | - Jana Kindermann
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Germany Department of Cardiac Surgery, Ludwig-Maximilians-Universität München, Germany
| | - Barbara Griemert
- Institute of Biochemistry, Medical School, Justus-Liebig-Universität, Giessen, Germany
| | - Andreas Blutke
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Germany
| | - Kerstin Troidl
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany Department of Vascular and Endovascular Surgery, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Tanja Mayr
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Germany Department of Anaesthesiology, Ludwig-Maximilians-Universität München, Germany
| | - Tobias Grantzow
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Germany
| | - Fatih Noyan
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Jan-Michael Abicht
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Germany Department of Anaesthesiology, Ludwig-Maximilians-Universität München, Germany
| | - Silvia Fischer
- Institute of Biochemistry, Medical School, Justus-Liebig-Universität, Giessen, Germany
| | - Klaus T Preissner
- Institute of Biochemistry, Medical School, Justus-Liebig-Universität, Giessen, Germany
| | - Ruediger Wanke
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Germany
| | - Elisabeth Deindl
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Germany
| | - Sonja Guethoff
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München, Germany Department of Cardiac Surgery, Ludwig-Maximilians-Universität München, Germany
| |
Collapse
|
33
|
Miyata S, Kurachi M, Okano Y, Sakurai N, Kobayashi A, Harada K, Yamagata H, Matsuo K, Takahashi K, Narita K, Fukuda M, Ishizaki Y, Mikuni M. Blood Transcriptomic Markers in Patients with Late-Onset Major Depressive Disorder. PLoS One 2016; 11:e0150262. [PMID: 26926397 PMCID: PMC4771207 DOI: 10.1371/journal.pone.0150262] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 02/11/2016] [Indexed: 01/09/2023] Open
Abstract
We investigated transcriptomic markers of late-onset major depressive disorder (LOD; onset age of first depressive episode ≥ 50 years) from the genes expressed in blood cells and identified state-dependent transcriptomic markers in these patients. We assessed the genes expressed in blood cells by microarray and found that the expression levels of 3,066 probes were state-dependently changed in the blood cells of patients with LOD. To select potential candidates from those probes, we assessed the genes expressed in the blood of an animal model of depression, ovariectomized female mice exposed to chronic ultra-mild stress, by microarray and cross-matched the differentially expressed genes between the patients and the model mice. We identified 14 differentially expressed genes that were similarly changed in both patients and the model mice. By assessing statistical significance using real-time quantitative PCR (RT-qPCR), the following 4 genes were selected as candidates: cell death-inducing DFFA-like effector c (CIDEC), ribonuclease 1 (RNASE1), solute carrier family 36 member-1 (SLC36A1), and serine/threonine/tyrosine interacting-like 1 (STYXL1). The discriminating ability of these 4 candidate genes was evaluated in an independent cohort that was validated. Among them, CIDEC showed the greatest discriminant validity (sensitivity 91.3% and specificity 87.5%). Thus, these 4 biomarkers should be helpful for properly diagnosing LOD.
Collapse
Affiliation(s)
- Shigeo Miyata
- Departments of Psychiatry and Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
- * E-mail:
| | - Masashi Kurachi
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yoshiko Okano
- Departments of Psychiatry and Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Noriko Sakurai
- Departments of Psychiatry and Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Ayumi Kobayashi
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Kenichiro Harada
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Hirotaka Yamagata
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Koji Matsuo
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Keisuke Takahashi
- Departments of Psychiatry and Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Kosuke Narita
- Departments of Psychiatry and Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Masato Fukuda
- Departments of Psychiatry and Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yasuki Ishizaki
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Masahiko Mikuni
- Departments of Psychiatry and Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| |
Collapse
|
34
|
Martin L, Koczera P, Simons N, Zechendorf E, Hoeger J, Marx G, Schuerholz T. The Human Host Defense Ribonucleases 1, 3 and 7 Are Elevated in Patients with Sepsis after Major Surgery--A Pilot Study. Int J Mol Sci 2016; 17:294. [PMID: 26927088 PMCID: PMC4813158 DOI: 10.3390/ijms17030294] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 02/04/2016] [Accepted: 02/16/2016] [Indexed: 01/21/2023] Open
Abstract
Sepsis is the most common cause of death in intensive care units and associated with widespread activation of host innate immunity responses. Ribonucleases (RNases) are important components of the innate immune system, however the role of RNases in sepsis has not been investigated. We evaluated serum levels of RNase 1, 3 and 7 in 20 surgical sepsis patients (Sepsis), nine surgical patients (Surgery) and 10 healthy controls (Healthy). RNase 1 and 3 were elevated in Sepsis compared to Surgery (2.2- and 3.1-fold, respectively; both p < 0.0001) or compared to Healthy (3.0- and 15.5-fold, respectively; both p < 0.0001). RNase 1 showed a high predictive value for the development of more than two organ failures (AUC 0.82, p = 0.01). Patients with renal dysfunction revealed higher RNase 1 levels than without renal dysfunction (p = 0.03). RNase 1 and 3 were higher in respiratory failure than without respiratory failure (p < 0.0001 and p = 0.02, respectively). RNase 7 was not detected in Healthy patients and only in two patients of Surgery, however RNase 7 was detected in 10 of 20 Sepsis patients. RNase 7 was higher in renal or metabolic failure than without failure (p = 0.04 and p = 0.02, respectively). In conclusion, RNase 1, 3 and 7 are secreted into serum under conditions with tissue injury, such as major surgery or sepsis. Thus, RNases might serve as laboratory parameters to diagnose and monitor organ failure in sepsis.
Collapse
Affiliation(s)
- Lukas Martin
- Department of Intensive Care and Intermediate Care, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Pauwelsstrasse 30, Aachen 52074, Germany.
| | - Patrick Koczera
- Department of Intensive Care and Intermediate Care, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Pauwelsstrasse 30, Aachen 52074, Germany.
| | - Nadine Simons
- Department of Intensive Care and Intermediate Care, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Pauwelsstrasse 30, Aachen 52074, Germany.
| | - Elisabeth Zechendorf
- Department of Intensive Care and Intermediate Care, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Pauwelsstrasse 30, Aachen 52074, Germany.
| | - Janine Hoeger
- Department of Intensive Care and Intermediate Care, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Pauwelsstrasse 30, Aachen 52074, Germany.
| | - Gernot Marx
- Department of Intensive Care and Intermediate Care, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Pauwelsstrasse 30, Aachen 52074, Germany.
| | - Tobias Schuerholz
- Department of Intensive Care and Intermediate Care, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Pauwelsstrasse 30, Aachen 52074, Germany.
| |
Collapse
|
35
|
Cabrera-Fuentes HA, Niemann B, Grieshaber P, Wollbrueck M, Gehron J, Preissner KT, Böning A. RNase1 as a potential mediator of remote ischaemic preconditioning for cardioprotection†. Eur J Cardiothorac Surg 2015; 48:732-7; discussion 737. [PMID: 25564211 DOI: 10.1093/ejcts/ezu519] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 11/26/2014] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES Remote ischaemic preconditioning (RIPC) is a non-invasive and virtually cost-free strategy for protecting the heart against acute ischaemia-reperfusion injury (IRI). We have recently shown that the inhibition of extracellular RNA (eRNA) using non-toxic RNase1 protected the heart against acute IRI, reduced myocardial infarct (MI) size and preserved left ventricular systolic function in rodent animal MI models. Based on this previous work in animals, the role of the eRNA/RNase1 system in cardiac RIPC in humans should be defined. METHODS Fourteen patients underwent cardiac surgery without RIPC; from each patient, six separate 5 ml blood specimens from radial artery and two blood specimens from coronary sinus at different time points during heart surgery were taken. Six healthy donors received RIPC (4 × 5 min upper limb ischaemia); blood parameters were quantified before and after RIPC. Twelve patients underwent cardiac surgery of which 6 received RIPC, whereas the remaining 6 were exposed to sham procedure. Circulating eRNA was quantified in plasma from arterial and coronary sinus blood obtained from patients undergoing cardiac by standard procedures. Tumour necrosis factor-α (TNF-α) production by heart tissue was assessed by enzyme-linked immuno-sorbent assay; RNase activity was quantified by an enzymatic assay. RESULTS Before surgery, eRNA levels were similar in both groups (14 ± 6 vs 13 ± 5 ng/ml; P = 0.9967). In patients without RIPC, arterial eRNA levels rose during surgery (87 ± 12 ng/ml) and peaked after (127 ± 11 ng/ml) aortic declamping; accordingly, eRNA levels in coronary sinus blood were significantly higher (206 ± 32 ng/ml; P = 0.0129) than that in radial artery. Moreover, significant elevation of TNF-α (36 ± 6 ng/ml; P = 0.0059) particularly in coronary sinus blood after opening of the aortic clamping was observed. Interestingly, applying a RIPC protocol significantly increased levels of plasma endogenous vascular RNase1 by >7-fold, and the levels of arterial (31 ± 7 ng/ml; P = 0.0024) and coronary sinus (37 ± 9 ng/ml; P < 0.0001) circulating eRNA, as well as circulating TNF-α (20 ± 4 ng/ml; P = 0.0050) levels were significantly reduced. CONCLUSIONS Upon RIPC, the level of cardioprotective RNase1 increased, while the concentration of damaging eRNA and TNF-α decreased. The present findings imply a significant contribution of the RIPC-dependent (endothelial) RNase1 for improving the outcome of cardiac surgery. However, the exact mechanism of RNase1-induced cardioprotection still remains to be explored.
Collapse
Affiliation(s)
- Hector A Cabrera-Fuentes
- Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany Department of Microbiology, Kazan Federal University, Kazan, Russian Federation
| | - Bernd Niemann
- Department of Cardiovascular Surgery, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Philippe Grieshaber
- Department of Cardiovascular Surgery, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Matthias Wollbrueck
- Department of Anaesthesia and Intensive Care Medicine, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Johannes Gehron
- Department of Cardiovascular Surgery, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Klaus T Preissner
- Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany Department of Microbiology, Kazan Federal University, Kazan, Russian Federation
| | - Andreas Böning
- Department of Cardiovascular Surgery, Medical School, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
36
|
Eller CH, Lomax JE, Raines RT. Bovine brain ribonuclease is the functional homolog of human ribonuclease 1. J Biol Chem 2014; 289:25996-26006. [PMID: 25078100 DOI: 10.1074/jbc.m114.566166] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mounting evidence suggests that human pancreatic ribonuclease (RNase 1) plays important roles in vivo, ranging from regulating blood clotting and inflammation to directly counteracting tumorigenic cells. Understanding these putative roles has been pursued with continual comparisons of human RNase 1 to bovine RNase A, an enzyme that appears to function primarily in the ruminant gut. Our results imply a different physiology for human RNase 1. We demonstrate distinct functional differences between human RNase 1 and bovine RNase A. Moreover, we characterize another RNase 1 homolog, bovine brain ribonuclease, and find pronounced similarities between that enzyme and human RNase 1. We report that human RNase 1 and bovine brain ribonuclease share high catalytic activity against double-stranded RNA substrates, a rare quality among ribonucleases. Both human RNase 1 and bovine brain RNase are readily endocytosed by mammalian cells, aided by tight interactions with cell surface glycans. Finally, we show that both human RNase 1 and bovine brain RNase are secreted from endothelial cells in a regulated manner, implying a potential role in vascular homeostasis. Our results suggest that brain ribonuclease, not RNase A, is the true bovine homolog of human RNase 1, and provide fundamental insight into the ancestral roles and functional adaptations of RNase 1 in mammals.
Collapse
Affiliation(s)
- Chelcie H Eller
- Departments of Biochemistry and University of Wisconsin, Madison, Wisconsin 53706
| | - Jo E Lomax
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin, Madison, Wisconsin 53706
| | - Ronald T Raines
- Departments of Biochemistry and University of Wisconsin, Madison, Wisconsin 53706; Departments of Chemistry, and University of Wisconsin, Madison, Wisconsin 53706.
| |
Collapse
|
37
|
Impact of extracellular RNA on endothelial barrier function. Cell Tissue Res 2014; 355:635-45. [DOI: 10.1007/s00441-014-1850-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 02/13/2014] [Indexed: 11/25/2022]
|
38
|
Gansler J, Preissner KT, Fischer S. Influence of proinflammatory stimuli on the expression of vascular ribonuclease 1 in endothelial cells. FASEB J 2013; 28:752-60. [PMID: 24174426 DOI: 10.1096/fj.13-238600] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Extracellular RNA (eRNA) released under injury or pathological conditions has been identified as a yet unrecognized vascular alarm signal to induce procoagulant, permeability-promoting, and proinflammatory activities. eRNA-induced functions were largely prevented by administration of RNase1 as a natural blood vessel-protective antagonist of eRNA. The aim of this study was to investigate the inflammatory regulation of endothelial cell RNase1, which is partly stored in Weibel-Palade bodies of these cells. Long-term treatment of human umbilical vein endothelial cells (HUVECs) with inflammatory agents like tumor necrosis factor α (TNF-α) or interleukin 1β (IL-1β), but not with eRNA, significantly decreased the release (34 ± 5%; 34 ± 7% of control) as well as the cellular expression (19.5 ± 5%; 33 ± 8% of control) of RNase1. Down-regulation of RNase1 by TNF-α stimulation or RNase1 siRNA knockdown increased the permeability of HUVEC monolayers, demonstrated by dearrangement of VE-cadherins at cell-cell borders. Mechanistically, cytokine-induced decrease of RNase1 expression did not involve the nuclear factor κ B (NFκB) signaling pathway but epigenic modifications. Since inhibition of histone deacetylases resulted in recovery of RNase1 expression and secretion after cytokine treatment, an acetylation-dependent process of RNase1 regulation is proposed. These results indicate that cytokine-mediated down-regulation of RNase1 in endothelial cells may aggravate eRNA-induced inflammatory activities and thereby disturbs the vascular homeostasis of the extracellular RNA/RNase system.
Collapse
Affiliation(s)
- Julia Gansler
- 1Institute for Biochemistry, Medical School, Justus-Liebig-University, Friedrichstrasse 24, 35392 Giessen, Germany.
| | | | | |
Collapse
|
39
|
Fischer S, Gesierich S, Griemert B, Schänzer A, Acker T, Augustin HG, Olsson AK, Preissner KT. Extracellular RNA liberates tumor necrosis factor-α to promote tumor cell trafficking and progression. Cancer Res 2013; 73:5080-9. [PMID: 23774209 DOI: 10.1158/0008-5472.can-12-4657] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Extracellular RNA (eRNA) released from injured cells promotes tissue permeability, thrombosis, and inflammation in vitro and in vivo, and RNase1 pretreatment can reduce all these effects. In this study, we investigated the role of the eRNA/RNase1 system in tumor progression and metastasis. Under quiescent and stimulatory conditions, tumor cells released much higher levels of endogenous extracellular RNA (eRNA) than nontumor cells. In glioblastomas, eRNA was detected at higher levels in tumors than nontumor tissue. eRNA induced tumor cells to adhere to and migrate through human cerebral microvascular endothelial cells (HCMEC/D3), in a manner requiring activation of VEGF signaling. In addition, eRNA liberated TNF-α from macrophages in a manner requiring activation of the TNF-α-converting enzyme TACE. Accordingly, supernatants derived from eRNA-treated macrophages enhanced tumor cell adhesion to HCMEC/D3. TNF-α release evoked by eRNA relied upon signaling activation of mitogen-activated protein kinases and the NF-κB pathway. In subcutaneous xenograft models of human cancer, administration of RNase1 but not DNase decreased tumor volume and weight. Taken together, these results suggest that eRNA released from tumor cells has the capacity to promote tumor cell invasion through endothelial barriers by both direct and indirect mechanisms, including through a mechanism involving TNF-α release from tumor-infiltrating monocytes/macrophages. Our findings establish a crucial role for eRNA in driving tumor progression, and they suggest applications for extracellular RNase1 as an antiinvasive regimen for cancer treatment.
Collapse
Affiliation(s)
- Silvia Fischer
- Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Fischer S, Preissner KT. Extracellular nucleic acids as novel alarm signals in the vascular system. Mediators of defence and disease. Hamostaseologie 2013; 33:37-42. [PMID: 23328880 DOI: 10.5482/hamo-13-01-0001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 01/08/2013] [Indexed: 01/13/2023] Open
Abstract
Upon vascular injury or tissue damage, the exposed intracellular material such as nucleic acids, histones and other macromolecules may come into contact with vessel wall cells and circulating blood cells and may thus, have an enduring influence on wound healing and body defence processes. This short review summarizes recent work related to extracellular DNA and RNA and their role as prominent alarm signals and inducers of different defence reactions related to innate immunity and thrombus formation. Of particular importance are DNA-histone complexes (nucleosome material) that, having been expelled during stimulation of the neutrophils, not only trap and eliminate bacteria but also promote thrombus formation in the arterial and venous system. Consequently therefore, the administration of DNase exhibits strong antithrombotic functions. Similarly, extracellular RNA provokes activation of the contact phase system of blood coagulation and, by interacting with specific proteins and cytokines, it promotes vascular permeability and oedema formation. The development of RNA-mediated thrombosis, vasogenic oedema or proinflammatory responses are counteracted by the administration of RNase1 in several pathogenetic animal models. Thus, extracellular nucleic acids appear not only to function as host alarm signals that serve to amplify the defence response, but they also provide important links to thrombus formation as part of the innate immune system.
Collapse
Affiliation(s)
- S Fischer
- Department of Biochemistry, Medical School, Justus-Liebig-Universität, Friedrichstr. 24, 35392 Giessen, Germany
| | | |
Collapse
|
41
|
Fischer S, Grantzow T, Pagel JI, Tschernatsch M, Sperandio M, Preissner KT, Deindl E. Extracellular RNA promotes leukocyte recruitment in the vascular system by mobilising proinflammatory cytokines. Thromb Haemost 2012; 108:730-41. [PMID: 22836360 DOI: 10.1160/th12-03-0186] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 07/16/2012] [Indexed: 12/31/2022]
Abstract
Extracellular RNA (eRNA), released from cells under conditions of injury or vascular disease, acts as potent prothrombotic factor and promotes vascular hyperpermeability related to oedema formation in vivo. In this study, we aimed to investigate the mechanism by which eRNA triggers inflammatory processes, particularly associated with different steps of leukocyte recruitment. Using intravital microscopy of murine cremaster muscle venules, eRNA (but not DNA) significantly induced leukocyte adhesion and transmigration in vivo, which was comparable in its effects to the function of tumour-necrosis-factor-α (TNF-α). In vitro, eRNA promoted adhesion and transmigration of monocytic cells on and across endothelial cell monolayers. eRNA-induced monocyte adhesion in vitro was mediated by activation of the vascular endothelial growth factor (VEGF)/VEGF-receptor-2 system and was abolished by neutralising antibodies against intercellular adhesion molecule-1 or the β2-integrin Mac-1. Additionally, eRNA induced the release of TNF-α from monocytic cells in a time- and concentration-dependent manner, which involved activation of TNF-α-converting enzyme (TACE) as well as the nuclear factor κB signalling machinery. In vivo, inhibiton of TACE significantly reduced eRNA-induced leukocyte adhesion. Our findings present evidence that eRNA in connection with tissue/vascular damage provokes a potent inflammatory response by inducing leukocyte recruitment and by mobilising proinflammatory cytokines from monocytes.
Collapse
Affiliation(s)
- Silvia Fischer
- Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany.
| | | | | | | | | | | | | |
Collapse
|
42
|
Ellis GA, McGrath NA, Palte MJ, Raines RT. Ribonuclease-Activated Cancer Prodrug. ACS Med Chem Lett 2012; 3:268-272. [PMID: 22611478 DOI: 10.1021/ml2002554] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Cancer chemotherapeutic agents often have a narrow therapeutic index that challenges the maintenance of a safe and effective dose. Consistent plasma concentrations of a drug can be obtained by using a timed-release prodrug strategy. We reasoned that a ribonucleoside 3'-phosphate could serve as a pro-moiety that also increases the hydrophilicity of a cancer chemotherapeutic agent. Herein, we report an efficient route for the synthesis of the prodrug uridine 3'-(4-hydroxytamoxifen phosphate) (UpHT). UpHT demonstrates timed-released activation kinetics with a half-life of approximately 4 h at the approximate plasma concentration of human pancreatic ribonuclease (RNase 1). MCF-7 breast cancer cells treated with UpHT showed decreased proliferation upon co-incubation with RNase 1, consistent with the release of the active drug-4-hydroxytamoxifen. These data demonstrate the utility of a human plasma enzyme as a useful activator of a prodrug.
Collapse
Affiliation(s)
- Gregory A. Ellis
- Department of Biochemistry, ‡Department of Chemistry, and §Medical Scientist Training Program and Molecular & Cellular Pharmacology Graduate Training Program, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Nicholas A. McGrath
- Department of Biochemistry, ‡Department of Chemistry, and §Medical Scientist Training Program and Molecular & Cellular Pharmacology Graduate Training Program, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Michael J. Palte
- Department of Biochemistry, ‡Department of Chemistry, and §Medical Scientist Training Program and Molecular & Cellular Pharmacology Graduate Training Program, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Ronald T. Raines
- Department of Biochemistry, ‡Department of Chemistry, and §Medical Scientist Training Program and Molecular & Cellular Pharmacology Graduate Training Program, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
43
|
The eight human "canonical" ribonucleases: molecular diversity, catalytic properties, and special biological actions of the enzyme proteins. FEBS Lett 2010; 584:2194-200. [PMID: 20388512 DOI: 10.1016/j.febslet.2010.04.018] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 04/07/2010] [Accepted: 04/07/2010] [Indexed: 01/25/2023]
Abstract
Human ribonucleases (RNases) are members of a large superfamily of rapidly evolving homologous proteins. Upon completion of the human genome, eight catalytically active RNases (numbered 1-8) were identified. These structurally distinct RNases, characterized by their various catalytic differences on different RNA substrates, constitute a gene family that appears to be the sole vertebrate-specific enzyme family. Apart from digestion of dietary RNA, a wide variety of biological actions, including neurotoxicity, angiogenesis, immunosuppressivity, and anti-pathogen activity, have been recently reported for almost all members of the family. Recent evolutionary studies suggest that RNases started off in vertebrates as host defence or angiogenic proteins.
Collapse
|
44
|
Kövér KE, Bruix M, Santoro J, Batta G, Laurents DV, Rico M. The solution structure and dynamics of human pancreatic ribonuclease determined by NMR spectroscopy provide insight into its remarkable biological activities and inhibition. J Mol Biol 2008; 379:953-65. [PMID: 18495155 DOI: 10.1016/j.jmb.2008.04.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2008] [Revised: 04/08/2008] [Accepted: 04/14/2008] [Indexed: 02/05/2023]
Abstract
Human pancreatic ribonuclease (RNase 1) is expressed in many tissues; has several important enzymatic and biological activities, including efficient cleavage of single-stranded RNA, double-stranded RNA and double-stranded RNA-DNA hybrids, digestion of dietary RNA, regulation of vascular homeostasis, inactivation of the HIV, activation of immature dendritic cells and induction of cytokine production; and furthermore shows potential as an anti-tumor agent. The solution structure and dynamics of uncomplexed, wild-type RNase 1 have been determined by NMR spectroscopy methods to better understand these activities. The family of 20 structures determined on the basis of 6115 unambiguous nuclear Overhauser enhancements is well resolved (pairwise backbone RMSD=1.07 A) and has the classic RNase A type of tertiary structure. Important structural differences compared with previously determined crystal structures of RNase 1 variants or inhibitor-bound complexes are observed in the conformation of loop regions and side chains implicated in the enzymatic as well as biological activities and binding to the cytoplasmic RNase inhibitor. Multiple side chain conformations observed for key surface residues are proposed to be crucial for membrane binding as well as translocation and efficient RNA hydrolysis. (15)N-(1)H relaxation measurements interpreted with the standard and our extended Lipari-Szabo formalism reveal rigid regions and identify more dynamic loop regions. Some of the most dynamic areas are key for binding to the cytoplasmic RNase inhibitor. This finding and the important differences observed between the structure in solution and that bound to the inhibitor are indications that RNase 1 to inhibitor binding can be better described by the "induced fit" model rather than the rigid "lock-into-key" mechanism. Translational diffusion measurements reveal that RNase 1 is predominantly dimeric above 1 mM concentration; the possible implications of this dimeric state for the remarkable biological properties of RNase 1 are discussed.
Collapse
Affiliation(s)
- K E Kövér
- Department of Chemistry, University of Debrecen, 4010 Debrecen, Hungary
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
Immunotherapy, based on mAbs specifically directed against cancer cells, is considered a precious strategy in the fight against cancer because of its selectivity and lack of multidrug resistant effects. However, there are obstacles to the complete success of current immunotherapy such as immune responses to nonhuman or even humanized antibodies and the large size of the antibodies, which hinders their diffusion into bulky tumors. Fully human, small immunoagents, capable of inhibiting tumor growth may overcome these problems and provide safe, highly selective and effective antitumor drugs. An attractive target for immunotherapy is ErbB2, a transmembrane tyrosine kinase receptor, overexpressed on tumor cells of different origin, with a key role in the development of malignancy. An anti-ErbB2 humanized monoclonal (Herceptin) is currently used with success for breast cancer therapy; however, it can engender cardiotoxicity and a high proportion of breast cancer patients are resistant to Herceptin treatment. Anti-ErbB2 immunoagents of human origin, with potentially no or very low immunogenicity have been engineered to assemble 'compact', i.e. reduced size, antibodies, one consisting of a human single-chain antibody fragment (scFv) fused to a human RNase to construct an immunoRNase and the other made up of two human scFv molecules fused to the Fc region of a human IgG1. By choosing a human antibody fragment as the immune moiety and a human RNase as the effector moiety, an immunoRNase would be both nonimmunogenic and nontoxic, as it becomes toxic only when the scFv promotes its internalization by target cells. The alternative strategy of compact antibodies was aimed at producing therapeutic agents with an increased half-life, prolonged tumor retention and the ability to recruit host effector functions. Moreover, the bivalency of compact antibodies can be exploited to construct bispecific antibodies, as well as for other therapeutic applications.
Collapse
Affiliation(s)
- Claudia De Lorenzo
- Dipartimento di Biologia Strutturale e Funzionale, Università di Napoli Federico II, Naples, Italy.
| | | |
Collapse
|
46
|
Development of Genotyping Methods for Single Nucleotide Polymorphism in the Human Pancreatic Ribonuclease Gene (RNASE1) and Their Application to Population Studies. Biochem Genet 2008; 46:145-53. [DOI: 10.1007/s10528-007-9138-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Accepted: 06/30/2007] [Indexed: 10/22/2022]
|
47
|
Rodríguez M, Moussaoui M, Benito A, Cuchillo CM, Nogués MV, Vilanova M. Human pancreatic ribonuclease presents higher endonucleolytic activity than ribonuclease A. Arch Biochem Biophys 2008; 471:191-7. [PMID: 18191631 DOI: 10.1016/j.abb.2007.12.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Revised: 12/14/2007] [Accepted: 12/15/2007] [Indexed: 11/17/2022]
Abstract
Analyzing the pattern of oligonucleotide formation induced by HP-RNase cleavage shows that the enzyme does not act randomly and follows a more endonucleolytic pattern when compared to RNase A. The enzyme prefers the binding and cleavage of longer substrate molecules, especially when the phosphodiester bond that is broken is 8-11 nucleotides away from at least one of the ends of the substrate molecule. This more endonucleolytic pattern is more appropriate for an enzyme with a regulatory role. Deleting two positive charges on the N-terminus (Arg4 and Lys6) modifies this pattern of external/internal phosphodiester bond cleavage preference, and produces a more exonucleolytic enzyme. These residues may reinforce the strength of a non-catalytic secondary phosphate binding (p2) or, alternatively, constitute a new non-catalytic phosphate binding subsite (p3).
Collapse
Affiliation(s)
- M Rodríguez
- Laboratori d'Enginyeria de Proteïnes, Departament de Biologia, Facultat de Ciències, Universitat de Girona, Campus de Montilivi s/n, 17071 Girona, Spain
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
DNA and RNA can be used to construct artificial nanodevices with strong potential for future biomedical applications. DNA nanodevices can function as biosensors, which detect and report the presence of proteins and naturally occurring nucleic acids, such as mRNA or microRNAs. Complex sensors can be realized by supporting DNA devices with DNA-based information processing. Artificial DNA-based reaction networks can be created that amplify molecular signals or evaluate logical functions to report the simultaneous presence of several disease-related molecules. Other applications for DNA nanodevices are found in controlled release and drug delivery. DNA can be used to build nanocontainers for drugs or switchable hydrogels, which can trap and release compounds. For in vivo applications of DNA nanodevices, techniques for efficient packaging and delivery have been developed and the first examples of intracellular RNA-based nanodevices have already been demonstrated.
Collapse
Affiliation(s)
- Friedrich C Simmel
- Technical University Munich, Physics Department E14, James-Franck-Straße D-85748 Garching, Germany
| |
Collapse
|
49
|
Fischer S, Gerriets T, Wessels C, Walberer M, Kostin S, Stolz E, Zheleva K, Hocke A, Hippenstiel S, Preissner KT. Extracellular RNA mediates endothelial-cell permeability via vascular endothelial growth factor. Blood 2007; 110:2457-65. [PMID: 17576819 DOI: 10.1182/blood-2006-08-040691] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Cell injury leads to exposure of intracellular material and is associated with increased permeability of vessels in the vicinity of the damage. Here, we demonstrate that natural extracellular RNA as well as artificial RNA (poly-I:C), or single-stranded RNA but not DNA, significantly increased the permeability across brain microvascular endothelial cells in vitro and in vivo. RNA-induced hyperpermeability of tight monolayers of endothelial cells correlated with disintegration of tight junctions and was mediated through vascular endothelial growth factor (VEGF), reminiscent of heparin's activities. Antisense oligonucleotides against VEGF-receptor 2 (VEGF-R2) prevented the permeability-inducing activity of extracellular RNA and heparin completely. Hence, these polyanionic substances can lead to mobilization/stabilization of VEGF with the subsequent activation of VEGF-R2. In accordance with these functional data, strong binding of VEGF as well as other growth factors to RNA was demonstrable. In in vivo rat models of FeCl(3)-induced sinus sagittal is superior thrombosis and stroke/brain edema, pretreatment of animals with RNase (but not DNase) resulted in a significant reduction of vessel occlusion, infarct volume, and prevention of brain edema formation. Together, these results identify extracellular RNA as a novel natural permeability factor, upstream of VEGF, whereas counteracting RNase treatment may serve as new vessel-protective modality.
Collapse
Affiliation(s)
- Silvia Fischer
- Department of Biochemistry Medical School, Justus-Liebig-University, Giessen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Shibamiya A, Muhl L, Tannert-Otto S, Preissner K, Kanse S. Nucleic acids potentiate Factor VII-activating protease (FSAP)-mediated cleavage of platelet-derived growth factor-BB and inhibition of vascular smooth muscle cell proliferation. Biochem J 2007; 404:45-50. [PMID: 17300216 PMCID: PMC1868837 DOI: 10.1042/bj20070166] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
FSAP (Factor VII-activating protease) can cleave and inactivate PDGF-BB (platelet-derived growth factor-BB) and thereby inhibits VSMC (vascular smooth-muscle cell) proliferation. The auto-activation of FSAP is facilitated by negatively charged polyanions such as heparin, dextransulfate or extracellular ribonucleic acids. Since auto-activation is essential for the anti-proliferative function of FSAP, the influence of nucleic acids as cofactors for the FSAP-mediated inhibition of PDGF-BB was investigated. Natural or artificial RNA was an effective cofactor for FSAP mediated PDGF-BB degradation, whereas the effect of DNA was weak. RNA-induced cleavage of PDGF-BB was inhibited by serine protease inhibitors. The pattern of PDGF-BB cleavage was identical with either heparin or RNA as a cofactor. One of the cleavage sites in PDGF-BB was at the positions 160-162 (R160KK162), which is an important region for receptor binding and activation. In VSMCs, PDGF-BB-stimulated DNA synthesis was inhibited by FSAP in the presence of RNA. RNA was more effective than DNA and the cofactor activity of RNA was neutralized after pretreatment with RNase. FSAP binding to RNA protected the nucleic acid from degradation by RNase. These data are relevant to situations where extracellular nucleic acids released from necrotic or apoptotic cells could activate local FSAP, leading to inhibition of PDGF-BB.
Collapse
Affiliation(s)
- Aya Shibamiya
- *Institute for Biochemistry, Medical Faculty, Justus-Liebig-University, 35392 Giessen, Germany
- †Graduate School of Health Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Lars Muhl
- *Institute for Biochemistry, Medical Faculty, Justus-Liebig-University, 35392 Giessen, Germany
| | - Susanne Tannert-Otto
- *Institute for Biochemistry, Medical Faculty, Justus-Liebig-University, 35392 Giessen, Germany
| | - Klaus T. Preissner
- *Institute for Biochemistry, Medical Faculty, Justus-Liebig-University, 35392 Giessen, Germany
| | - Sandip M. Kanse
- *Institute for Biochemistry, Medical Faculty, Justus-Liebig-University, 35392 Giessen, Germany
- To whom correspondence should be addressed (email )
| |
Collapse
|