1
|
Lee SJ, Kim D, Han G, Hong SP, Choi I, Cho C. Functional Characterization and Heterogeneity Analysis of Ribosomal Proteins in Mouse Preimplantation Embryos. FASEB J 2025; 39:e70662. [PMID: 40421595 DOI: 10.1096/fj.202500574rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 05/06/2025] [Accepted: 05/13/2025] [Indexed: 05/28/2025]
Abstract
Translational control is important during the mammalian preimplantation phase, when maternal RNAs and proteins are degraded and de novo synthesis of RNAs and proteins increases. Proteins are synthesized in ribosomes, which are assembled from ~82 ribosomal proteins (RPs). The function of ribosomes varies depending on the resident RPs, suggesting that ribosome heterogeneity can lead to functional specialization. Only a few studies have investigated the function of RPs during preimplantation embryonic development. Here, we performed functional analyses on six RP-encoding genes-Rpl4, Rps9, Rps11, Rpl13a, Rpl19, and Rpl39-in mouse preimplantation embryos. Knockdown (KD) of each of these RP genes, except Rpl39, affected morula-to-blastocyst transition, producing phenotypes that varied somewhat in their details. Rpl4-, Rpl13a-, and Rpl19-KD embryos showed fragmentation and strong arrest of cell proliferation, whereas Rps9- and Rps11-KD embryos showed severe fragmentation with relatively weak arrest of cell proliferation. In the case of Rpl39, single-KD embryos developed normally, but double-KD embryos with its paralog Rpl39-like (Rpl39l) inhibited normal blastocyst development. Protein misfolding signals were also activated in Rpl39-KD and Rpl39l + Rpl39 double-KD embryos, confirming a previous finding that RPL39 and RPL39L are associated with ribosome exit tunnels. Our results suggest the presence of different groups of proteins that require an RPL39-containing ribosome or RPL39/RPL39L-containing ribosome for correct folding in early embryos. Taken together, the results of the present study demonstrate that ribosomal proteins are fundamentally important for normal blastocyst formation and development, but not all ribosomal proteins contribute equally to embryonic development, providing a novel example of ribosome heterogeneity in preimplantation embryos.
Collapse
Affiliation(s)
- Seung Jae Lee
- Department of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Dayeon Kim
- Department of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Gwidong Han
- Department of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Seung Pyo Hong
- Department of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Inchul Choi
- Department of Animal and Dairy Sciences, Chungnam National University, Daejeon, Republic of Korea
| | - Chunghee Cho
- Department of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| |
Collapse
|
2
|
Das AS, Basu A, Mukhopadhyay R. Ribosomal proteins: the missing piece in the inflammation puzzle? Mol Cell Biochem 2025; 480:785-797. [PMID: 38951378 DOI: 10.1007/s11010-024-05050-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/09/2024] [Indexed: 07/03/2024]
Abstract
Ribosomal proteins (RPs) are constituents of macromolecular machinery, ribosome that translates genetic information into proteins. Besides ribosomal functions, RPs are now getting appreciated for their 'moonlighting'/extra-ribosomal functions modulating many cellular processes. Accumulating evidence suggests that a number of RPs are involved in inflammation. Though acute inflammation is a part of the innate immune response, uncontrolled inflammation is a driving factor for several chronic inflammatory diseases. An in-depth understanding of inflammation regulation has always been valued for the better management of associated diseases. Hence, this review first outlines the common livelihood of RPs and then provides a comprehensive account of five RPs that significantly contribute to the inflammation process. Finally, we discuss the possible therapeutic uses of RPs against chronic inflammatory diseases.
Collapse
Affiliation(s)
- Anindhya Sundar Das
- Department of Molecular Biology and Biotechnology, Tezpur University, Assam, 784028, India.
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, 02912, USA.
| | - Anandita Basu
- Department of Molecular Biology and Biotechnology, Tezpur University, Assam, 784028, India
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, 02903, USA
| | - Rupak Mukhopadhyay
- Department of Molecular Biology and Biotechnology, Tezpur University, Assam, 784028, India.
| |
Collapse
|
3
|
Ferhat M, Mayer J, Costa LH, Prendecki M, Tarazona AAP, Schinagl A, Kerschbaumer RJ, Tam FWK, Landlinger C, Thiele M. Targeting of oxidized Macrophage Migration Inhibitory Factor (oxMIF) with antibody ON104 attenuates the severity of glomerulonephritis. PLoS One 2024; 19:e0311837. [PMID: 39374239 PMCID: PMC11458038 DOI: 10.1371/journal.pone.0311837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/20/2024] [Indexed: 10/09/2024] Open
Abstract
The oxidized form of Macrophage Migration Inhibitory Factor (oxMIF) has been identified as the disease-related isoform of MIF, exerting pathological functions in inflamed tissue. In this study, we aimed to explore the in vivo effects of the neutralizing anti-oxMIF antibody ON104 in a rat model of crescentic glomerulonephritis (CGN), to better understand its disease modifying activities. WKY rats received a single intravenous injection of a rabbit nephrotoxic serum (NTS), targeting rat glomerular basement membrane to induce CGN. On day 4 and day 6, ON104 was given intraperitoneally (i.p.) and on day 8 urine, blood and kidney tissue were collected. ON104 substantially attenuated the severity of CGN demonstrated by reduced proteinuria, hematuria, as well as lower levels of kidney injury molecule (KIM)-1. ON104 treatment preserved the glomerular morphology and suppressed crescent formation, a hallmark of the disease. On the cellular level, oxMIF neutralization by ON104 strongly reduced the number of macrophages and neutrophils within the inflamed kidneys. In vitro, we identified human neutrophils, but not monocytes, as main producers of oxMIF among total peripheral cells. The present study demonstrates that oxMIF is a pertinent therapeutic target in a model of CGN which mechanistically resembles human immune mediated CGN. In this model, neutralization of oxMIF by ON104 leads to an improvement in both urinary abnormalities and histological pathological characteristics of the disease. ON104, thus has the potential to become a novel disease-modifying drug for the treatment of glomerulonephritis and other inflammatory kidney diseases.
Collapse
Affiliation(s)
- Maroua Ferhat
- OncoOne Research & Development GmbH, Vienna, Austria
| | - Julia Mayer
- OncoOne Research & Development GmbH, Vienna, Austria
| | - Lyndon H. Costa
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Hammersmith Hospital, Imperial College London (ICL), London, United Kingdom
| | - Maria Prendecki
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Hammersmith Hospital, Imperial College London (ICL), London, United Kingdom
| | | | | | | | - Frederick W. K. Tam
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Hammersmith Hospital, Imperial College London (ICL), London, United Kingdom
| | | | | |
Collapse
|
4
|
Huang YA, Wang X, Kim JC, Yao X, Sethi A, Strohm A, Doherty TA. PIP-seq identifies novel heterogeneous lung innate lymphocyte population activation after combustion product exposure. Sci Rep 2024; 14:20167. [PMID: 39215111 PMCID: PMC11364781 DOI: 10.1038/s41598-024-70880-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Innate lymphoid cells (ILCs) are a heterogeneous population that play diverse roles in airway inflammation after exposure to allergens and infections. However, how ILCs respond after exposure to environmental toxins is not well understood. Here we show a novel method for studying the heterogeneity of rare lung ILC populations by magnetic enrichment for lung ILCs followed by particle-templated instant partition sequencing (PIP-seq). Using this method, we were able to identify novel group 1 and group 2 ILC subsets that exist after exposure to both fungal allergen and burn pit-related constituents (BPC) that include dioxin, aromatic hydrocarbon, and particulate matter. Toxin exposure in combination with fungal allergen induced activation of specific ILC1/NK and ILC2 populations as well as promoted neutrophilic lung inflammation. Oxidative stress pathways and downregulation of specific ribosomal protein genes (Rpl41 and Rps19) implicated in anti-inflammatory responses were present after BPC exposure. Increased IFNγ expression and other pro-neutrophilic mediator transcripts were increased in BPC-stimulated lung innate lymphoid cells. Further, the addition of BPC induced Hspa8 (encodes HSC70) and aryl hydrocarbon transcription factor activity across multiple lung ILC subsets. Overall, using an airway disease model that develops after occupational and environmental exposures, we demonstrate an effective method to better understand heterogenous ILC subset activation.
Collapse
Affiliation(s)
- Yung-An Huang
- Divison of Allergy and Immunology, Department of Medicine, University of California San Diego, Biomedical Sciences Building, Room 5090, 9500 Gilman Drive, La Jolla, CA, 92093-0635, USA
- Veterans Affairs San Diego Health Care System, San Diego, CA, USA
| | - Xinyu Wang
- Divison of Allergy and Immunology, Department of Medicine, University of California San Diego, Biomedical Sciences Building, Room 5090, 9500 Gilman Drive, La Jolla, CA, 92093-0635, USA
- Veterans Affairs San Diego Health Care System, San Diego, CA, USA
| | - Jong-Chan Kim
- Divison of Allergy and Immunology, Department of Medicine, University of California San Diego, Biomedical Sciences Building, Room 5090, 9500 Gilman Drive, La Jolla, CA, 92093-0635, USA
- Veterans Affairs San Diego Health Care System, San Diego, CA, USA
| | - Xiang Yao
- Divison of Allergy and Immunology, Department of Medicine, University of California San Diego, Biomedical Sciences Building, Room 5090, 9500 Gilman Drive, La Jolla, CA, 92093-0635, USA
- Veterans Affairs San Diego Health Care System, San Diego, CA, USA
| | - Anshika Sethi
- Divison of Allergy and Immunology, Department of Medicine, University of California San Diego, Biomedical Sciences Building, Room 5090, 9500 Gilman Drive, La Jolla, CA, 92093-0635, USA
- Veterans Affairs San Diego Health Care System, San Diego, CA, USA
| | - Allyssa Strohm
- Divison of Allergy and Immunology, Department of Medicine, University of California San Diego, Biomedical Sciences Building, Room 5090, 9500 Gilman Drive, La Jolla, CA, 92093-0635, USA
- Veterans Affairs San Diego Health Care System, San Diego, CA, USA
| | - Taylor A Doherty
- Divison of Allergy and Immunology, Department of Medicine, University of California San Diego, Biomedical Sciences Building, Room 5090, 9500 Gilman Drive, La Jolla, CA, 92093-0635, USA.
- Veterans Affairs San Diego Health Care System, San Diego, CA, USA.
| |
Collapse
|
5
|
Huang YA, Wang X, Kim JC, Yao X, Sethi A, Strohm A, Doherty TA. PIP-Seq identifies novel heterogeneous lung innate lymphocyte population activation after combustion product exposure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600420. [PMID: 38979234 PMCID: PMC11230265 DOI: 10.1101/2024.06.24.600420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Innate lymphoid cells (ILCs) are a heterogeneous population that play diverse roles in airway inflammation after exposure to allergens and infections. However, how ILCs respond after exposure to environmental toxins is not well understood. Here we show a novel method for studying the heterogeneity of rare lung ILC populations by magnetic enrichment for lung ILCs followed by particle-templated instant partition sequencing (PIP-seq). Using this method, we were able to identify novel group 1 and group 2 ILC subsets that exist after exposure to both fungal allergen and burn pit-related constituents (BPC) that include dioxin, aromatic hydrocarbon, and particulate matter. Toxin exposure in combination with fungal allergen induced activation of specific ILC1/NK and ILC2 populations as well as promoted neutrophilic lung inflammation. Oxidative stress pathways and downregulation of specific ribosomal protein genes ( Rpl41 and Rps19 ) implicated in anti-inflammatory responses were present after BPC exposure. Increased IFNγ expression and other pro-neutrophilic mediator transcripts were increased in BPC-stimulated lung innate lymphoid cells. Further, the addition of BPC induced Hspa8 (encodes HSC70) and aryl hydrocarbon transcription factor activity across multiple lung ILC subsets. Overall, using an airway disease model that develops after occupational and environmental exposures, we demonstrate an effective method to better understand heterogenous ILC subset activation.
Collapse
|
6
|
Yang H, Li J, Huang XR, Bucala R, Xu A, Lan HY. Macrophage-derived macrophage migration inhibitory factor mediates renal injury in anti-glomerular basement membrane glomerulonephritis. Front Immunol 2024; 15:1361343. [PMID: 38846956 PMCID: PMC11153660 DOI: 10.3389/fimmu.2024.1361343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 04/30/2024] [Indexed: 06/09/2024] Open
Abstract
Macrophages are a rich source of macrophage migration inhibitory factor (MIF). It is well established that macrophages and MIF play a pathogenic role in anti-glomerular basement membrane crescentic glomerulonephritis (anti-GBM CGN). However, whether macrophages mediate anti-GBM CGN via MIF-dependent mechanism remains unexplored, which was investigated in this study by specifically deleting MIF from macrophages in MIFf/f-lysM-cre mice. We found that compared to anti-GBM CGN induced in MIFf/f control mice, conditional ablation of MIF in macrophages significantly suppressed anti-GBM CGN by inhibiting glomerular crescent formation and reducing serum creatinine and proteinuria while improving creatine clearance. Mechanistically, selective MIF depletion in macrophages largely inhibited renal macrophage and T cell recruitment, promoted the polarization of macrophage from M1 towards M2 via the CD74/NF-κB/p38MAPK-dependent mechanism. Unexpectedly, selective depletion of macrophage MIF also significantly promoted Treg while inhibiting Th1 and Th17 immune responses. In summary, MIF produced by macrophages plays a pathogenic role in anti-GBM CGN. Targeting macrophage-derived MIF may represent a novel and promising therapeutic approach for the treatment of immune-mediated kidney diseases.
Collapse
Affiliation(s)
- Hui Yang
- Department of Nephrology, Sun Yat‐Sen Memorial Hospital, Sun Yat‐Sen University, Guangzhou, China
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jinhong Li
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat‐sen University, SunYat‐sen University, Shenzhen, China
| | - Xiao-ru Huang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Departments of Nephrology and Pathology, Guangdong Provincial Hospital, Southern Medical University, Guangzhou, China
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Anping Xu
- Department of Nephrology, Sun Yat‐Sen Memorial Hospital, Sun Yat‐Sen University, Guangzhou, China
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Departments of Nephrology and Pathology, Guangdong Provincial Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Ahmed TA, Ahmed SM, Elkhenany H, El-Desouky MA, Magdeldin S, Osama A, Anwar AM, Mohamed IK, Abdelgawad ME, Hanna DH, El-Badri N. The cross talk between type II diabetic microenvironment and the regenerative capacities of human adipose tissue-derived pericytes: a promising cell therapy. Stem Cell Res Ther 2024; 15:36. [PMID: 38331889 PMCID: PMC10854071 DOI: 10.1186/s13287-024-03643-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/21/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Pericytes (PCs) are multipotent contractile cells that wrap around the endothelial cells (ECs) to maintain the blood vessel's functionality and integrity. The hyperglycemia associated with Type 2 diabetes mellitus (T2DM) was shown to impair the function of PCs and increase the risk of diabetes complications. In this study, we aimed to investigate the deleterious effect of the diabetic microenvironment on the regenerative capacities of human PCs. METHODS PCs isolated from human adipose tissue were cultured in the presence or absence of serum collected from diabetic patients. The functionality of PCs was analyzed after 6, 14, and 30 days. RESULTS Microscopic examination of PCs cultured in DS (DS-PCs) showed increased aggregate formation and altered surface topography with hyperbolic invaginations. Compared to PCs cultured in normal serum (NS-PCs), DS-PCs showed more fragmented mitochondria and thicker nuclear membrane. DS caused impaired angiogenic differentiation of PCs as confirmed by tube formation, decreased VEGF-A and IGF-1 gene expression, upregulated TSP1, PF4, actin-related protein 2/3 complex, and downregulated COL21A1 protein expression. These cells suffered more pronounced apoptosis and showed higher expression of Clic4, apoptosis facilitator BCl-2-like protein, serine/threonine protein phosphatase, and caspase-7 proteins. DS-PCs showed dysregulated DNA repair genes CDKN1A, SIRT1, XRCC5 TERF2, and upregulation of the pro-inflammatory genes ICAM1, IL-6, and TNF-α. Further, DS-treated cells also showed disruption in the expression of the focal adhesion and binding proteins TSP1, TGF-β, fibronectin, and PCDH7. Interestingly, DS-PCs showed resistance mechanisms upon exposure to diabetic microenvironment by maintaining the intracellular reactive oxygen species (ROS) level and upregulation of extracellular matrix (ECM) organizing proteins as vinculin, IQGAP1, and tubulin beta chain. CONCLUSION These data showed that the diabetic microenvironment exert a deleterious effect on the regenerative capacities of human adipose tissue-derived PCs, and may thus have possible implications on the vascular complications of T2DM. Nevertheless, PCs have shown remarkable protective mechanisms when initially exposed to DS and thus they could provide a promising cellular therapy for T2DM.
Collapse
Affiliation(s)
- Toka A Ahmed
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
- Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo, Egypt
| | - Sara M Ahmed
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Hoda Elkhenany
- Department of Surgery, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 22785, Egypt
| | - Mohamed A El-Desouky
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Sameh Magdeldin
- Proteomics and Metabolomics Research Program, Basic Research Department, Children's Cancer Hospital, Cairo, 57357, Egypt
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Aya Osama
- Proteomics and Metabolomics Research Program, Basic Research Department, Children's Cancer Hospital, Cairo, 57357, Egypt
| | - Ali Mostafa Anwar
- Proteomics and Metabolomics Research Program, Basic Research Department, Children's Cancer Hospital, Cairo, 57357, Egypt
| | - Ihab K Mohamed
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Mohamed Essameldin Abdelgawad
- Biochemistry and Molecular Biotechnology Division, Chemistry Department, Faculty of Science, Innovative Cellular Microenvironment Optimization Platform (ICMOP), Precision Therapy Unit, Helwan University, Cairo, Egypt
- The Egyptian Network of Bioinformatics "BioNetMasr", Cairo, Egypt
| | - Demiana H Hanna
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt.
| |
Collapse
|
8
|
Single-Cell RNA-Seq Analysis Reveals Macrophages Are Involved in the Pathogenesis of Human Sporadic Acute Type A Aortic Dissection. Biomolecules 2023; 13:biom13020399. [PMID: 36830768 PMCID: PMC9952989 DOI: 10.3390/biom13020399] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/26/2023] [Accepted: 02/14/2023] [Indexed: 02/23/2023] Open
Abstract
Macrophages play an important role in the progression of sporadic acute type A aortic dissection (ATAAD). The aim of this study was to characterize the cellular heterogeneity of macrophages in ATAAD tissues by scRNA-seq. Ascending aortic wall tissue from six ATAAD patients and three heart transplant donors was assessed by scRNA-seq and then analyzed and validated by various bioinformatic algorithms and histopathology experiments. The results revealed that the proportion of macrophages in ATAAD tissues (24.51%) was significantly higher than that in normal tissues (13.69%). Among the six macrophage subclusters, pro-inflammatory macrophages accounted for 14.96% of macrophages in the AD group and 0.18% in the normal group. Chemokine- and inflammation-related genes (CCL2, CCL20, S100A8, and S100A9) were expressed more intensively in macrophages in ATAAD tissue than in those in normal tissue. Additionally, intercellular communication analysis and transcription factor analysis indicated the activation of inflammation and degradation of the extracellular matrix in ATAAD tissue. Finally, immunohistochemistry, immunofluorescence, and Western blot experiments confirmed the overexpression of macrophage marker genes (CD68 and CD163) and matrix metalloproteinases (MMP9 and MMP2) in ATAAD tissue. Collectively, our study provides a preliminary evaluation of the role of macrophages in ATAAD, and the results could aid in the development of therapeutic options in the future.
Collapse
|
9
|
Głuchowska A, Cysewski D, Baj-Krzyworzeka M, Szatanek R, Węglarczyk K, Podszywałow-Bartnicka P, Sunderland P, Kozłowska E, Śliwińska MA, Dąbrowski M, Sikora E, Mosieniak G. Unbiased proteomic analysis of extracellular vesicles secreted by senescent human vascular smooth muscle cells reveals their ability to modulate immune cell functions. GeroScience 2022; 44:2863-2884. [PMID: 35900662 PMCID: PMC9768090 DOI: 10.1007/s11357-022-00625-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/12/2022] [Indexed: 01/07/2023] Open
Abstract
Atherosclerosis, a common age-related disease, is characterized by intense immunological activity. Atherosclerotic plaque is composed of endothelial cells, vascular smooth muscle cells (VSMCs), lipids and immune cells infiltrating from the blood. During progression of the disease, VSMCs undergo senescence within the plaque and secrete SASP (senescence-associated secretory phenotype) factors that can actively modulate plaque microenvironment. We demonstrated that senescent VSMCs secrete increased number of extracellular vesicles (senEVs). Based on unbiased proteomic analysis of VMSC-derived EVs and of the soluble fraction of SASP (sSASP), more than 900 proteins were identified in each of SASP compartments. Comparison of the composition of VMSC-derived EVs with the SASP atlas revealed several proteins, including Serpin Family F Member 1 (SERPINF1) and Thrombospondin 1 (THBS1), as commonly upregulated components of EVs secreted by senescent VSMCs and fibroblasts. Among soluble SASP factors, only Growth Differentiation Factor 15 (GDF15) was universally increased in the secretome of senescent VSMCs, fibroblasts, and epithelial cells. Bioinformatics analysis of EV proteins distinguished functionally organized protein networks involved in immune cell function regulation. Accordingly, EVs released by senescent VSMCs induced secretion of IL-17, INFγ, and IL-10 by T cells and of TNFα produced by monocytes. Moreover senEVs influenced differentiation of monocytes favoring mix M1/M2 polarization with proinflammatory characteristics. Altogether, our studies provide a complex, unbiased analysis of VSMC SASP and prove that EVs derived from senescent VSMCs influence the cytokine milieu by modulating immune cell activity. Our results strengthen the role of senescent cells as an important inducer of inflammation in atherosclerosis.
Collapse
Affiliation(s)
- Agata Głuchowska
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3 St., 02-093, Warsaw, Poland
| | - Dominik Cysewski
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
- Clinical Research Centre, Medical University of Bialystok, Białystok, Poland
| | - Monika Baj-Krzyworzeka
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Rafał Szatanek
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Kazimierz Węglarczyk
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | | | - Piotr Sunderland
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3 St., 02-093, Warsaw, Poland
| | - Ewa Kozłowska
- Department of Immunology, Institute of Functional Biology and Ecology, Faculty of Biology University of Warsaw, Warsaw, Poland
| | - Małgorzata A Śliwińska
- Laboratory of Imaging Tissue Structure and Function, Nencki Institute of Experimental Biology Polish Academy of Sciences, Warsaw, Poland
| | - Michał Dąbrowski
- Laboratory of Bioinformatics, Neurobiology Centre, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Ewa Sikora
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3 St., 02-093, Warsaw, Poland
| | - Grażyna Mosieniak
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3 St., 02-093, Warsaw, Poland.
| |
Collapse
|
10
|
Du Y, Hao H, Ma H, Liu H. Macrophage migration inhibitory factor in acute kidneyinjury. Front Physiol 2022; 13:945827. [PMID: 36117692 PMCID: PMC9478040 DOI: 10.3389/fphys.2022.945827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Acute kidney injury (AKI) is a complex clinical syndrome with multiple etiologies and pathogenesis, which lacks early biomarkers and targeted therapy. Recently, macrophage migration inhibitory factor (MIF) family protein have received increasing attention owing to its pleiotropic protein molecule character in acute kidney injury, where it performed a dual role in the pathological process. macrophage migration inhibitory factor and macrophage migration inhibitory factor-2 are released into the peripheral circulation when Acute kidney injury occurs and interact with various cellular pathways. On the one hand, macrophage migration inhibitory factor exerts a protective effect in anti-oxidation and macrophage migration inhibitory factor-2 promotes cell proliferation and ameliorates renal fibrosis. On the other hand, macrophage migration inhibitory factor aggravates renal injury as an upstream inflammation factor. Herein, we provide an overview on the biological role and possible mechanisms of macrophage migration inhibitory factor and macrophage migration inhibitory factor-2 in the process of Acute kidney injury and the clinical application prospects of macrophage migration inhibitory factor family proteins as a potential therapeutic target.
Collapse
Affiliation(s)
- Yiwei Du
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
| | - Hao Hao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
| | - Heng Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
- *Correspondence: Hongbao Liu, ; Heng Ma,
| | - Hongbao Liu
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
- *Correspondence: Hongbao Liu, ; Heng Ma,
| |
Collapse
|
11
|
Macrophage Migration Inhibitory Factor (MIF) as a Stress Molecule in Renal Inflammation. Int J Mol Sci 2022; 23:ijms23094908. [PMID: 35563296 PMCID: PMC9102975 DOI: 10.3390/ijms23094908] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 02/06/2023] Open
Abstract
Renal inflammation is an initial pathological process during progressive renal injury regardless of the initial cause. Macrophage migration inhibitory factor (MIF) is a truly proinflammatory stress mediator that is highly expressed in a variety of both inflammatory cells and intrinsic kidney cells. MIF is released from the diseased kidney immediately upon stimulation to trigger renal inflammation by activating macrophages and T cells, and promoting the production of proinflammatory cytokines, chemokines, and stress molecules via signaling pathways involving the CD74/CD44 and chemokine receptors CXCR2, CXCR4, and CXCR7 signaling. In addition, MIF can function as a stress molecule to counter-regulate the immunosuppressive effect of glucocorticoid in renal inflammation. Given the critical position of MIF in the upstream inflammatory cascade, this review focuses on the regulatory role and molecular mechanisms of MIF in kidney diseases. The therapeutic potential of targeting MIF signaling to treat kidney diseases is also discussed.
Collapse
|
12
|
Li ZH, Guo XY, Quan XY, Yang C, Liu ZJ, Su HY, An N, Liu HF. The Role of Parietal Epithelial Cells in the Pathogenesis of Podocytopathy. Front Physiol 2022; 13:832772. [PMID: 35360248 PMCID: PMC8963495 DOI: 10.3389/fphys.2022.832772] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/07/2022] [Indexed: 02/05/2023] Open
Abstract
Podocytopathy is the most common feature of glomerular disorder characterized by podocyte injury- or dysfunction-induced excessive proteinuria, which ultimately develops into glomerulosclerosis and results in persistent loss of renal function. Due to the lack of self-renewal ability of podocytes, mild podocyte depletion triggers replacement and repair processes mostly driven by stem cells or resident parietal epithelial cells (PECs). In contrast, when podocyte recovery fails, activated PECs contribute to the establishment of glomerular lesions. Increasing evidence suggests that PECs, more than just bystanders, have a crucial role in various podocytopathies, including minimal change disease, focal segmental glomerulosclerosis, membranous nephropathy, diabetic nephropathy, IgA nephropathy, and lupus podocytopathy. In this review, we attempt to dissect the diverse role of PECs in the pathogenesis of podocytopathy based on currently available information.
Collapse
|
13
|
Thankam FG, Huynh J, Fang W, Chen Y, Agrawal DK. Exosomal-ribosomal proteins-driven heterogeneity of epicardial adipose tissue derived stem cells under ischemia for cardiac regeneration. J Tissue Eng Regen Med 2022; 16:396-408. [PMID: 35142442 DOI: 10.1002/term.3289] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/19/2022] [Accepted: 01/31/2022] [Indexed: 12/22/2022]
Abstract
Extracellular ribosomal proteins secreted in exosomes elicit biological/regenerative responses; however, ribosomal proteins contained in the exosomes of ischemia-challenged epicardial adipose tissue-derived stem cells (EATDS) remain unexplored. This study focuses on the identification of ribosomal proteins in the exosomes of ischemia-challenged EATDS and their sub-populations based on the key ribosomal proteins using single-cell genomics. Exosomes were isolated from control, ischemic (ISC), and reperfused (ISC/R) EATDS harvested from hyperlipidemic microswine, and the proteins were detected using Liquid chromatography with tandem mass spectrometry (LC-MS/MS). One hundred ninety-nine proteins and 177 proteins were detected in ISC and ISC/R groups, respectively with significant fold-change compared to controls. Five ribosomal proteins, RPL10A, 40SRPS18, 40SRPS30, 60SRPL14, and 40SRPSA, were significant owing to their abundance based on LC-MS/MS data. Expression of these proteins, except RPL10A, at transcript and protein levels were lower in ISC group compared to the control. scRNAseq analysis revealed EATDS heterogeneity based on the upregulation of 40SRPSA, 40SRPL18, and 40SRPS18. Pro-inflammatory sub-populations upregulated CCL5, anti-inflammatory sub-population upregulated IL-11, proliferative sub-population upregulated cell cycle and DNA replication mediators, and non-proliferative population downregulated the cell cycle and DNA replication mediators. Overall, the functional role of extracellular ribosomal proteins in driving unique phenotypes of EATDS population offers promise for designing effective translational approaches for myocardial regeneration.
Collapse
Affiliation(s)
- Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
| | - James Huynh
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
| | - William Fang
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
| | - Yu Chen
- Molecular Instrumentation Center, University of California-Los Angeles, Los Angeles, California, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
14
|
Song S, Xiao Z, Dekker FJ, Poelarends GJ, Melgert BN. Macrophage migration inhibitory factor family proteins are multitasking cytokines in tissue injury. Cell Mol Life Sci 2022; 79:105. [PMID: 35091838 PMCID: PMC8799543 DOI: 10.1007/s00018-021-04038-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 02/06/2023]
Abstract
The family of macrophage migration inhibitory factor (MIF) proteins in humans consist of MIF, its functional homolog D-dopachrome tautomerase (D-DT, also known as MIF-2) and the relatively unknown protein named DDT-like (DDTL). MIF is a pleiotropic cytokine with multiple properties in tissue homeostasis and pathology. MIF was initially found to associate with inflammatory responses and therefore established a reputation as a pro-inflammatory cytokine. However, increasing evidence demonstrates that MIF influences many different intra- and extracellular molecular processes important for the maintenance of cellular homeostasis, such as promotion of cellular survival, antioxidant signaling, and wound repair. In contrast, studies on D-DT are scarce and on DDTL almost nonexistent and their functions remain to be further investigated as it is yet unclear how similar they are compared to MIF. Importantly, the many and sometimes opposing functions of MIF suggest that targeting MIF therapeutically should be considered carefully, taking into account timing and severity of tissue injury. In this review, we focus on the latest discoveries regarding the role of MIF family members in tissue injury, inflammation and repair, and highlight the possibilities of interventions with therapeutics targeting or mimicking MIF family proteins.
Collapse
|
15
|
Li N, Chen J, Wang P, Fan H, Hou S, Gong Y. Major signaling pathways and key mediators of macrophages in acute kidney injury (Review). Mol Med Rep 2021; 23:455. [PMID: 33880578 PMCID: PMC8072315 DOI: 10.3892/mmr.2021.12094] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/17/2021] [Indexed: 12/16/2022] Open
Abstract
Acute kidney injury (AKI) has become a global public health problem with high morbidity and mortality rates, as well as high healthcare costs. Immune cells, particularly macrophages, which regulate tissue development, destroy pathogens, control homeostasis and repair wounds, play crucial and complex roles in AKI. In various types of AKI, numerous rapidly recruited monocytes and tissue-resident macrophages act in a coordinated manner. Thus, elucidating the phenotypic and functional characteristics of macrophages in AKI is essential for identifying potential therapeutic targets. Macrophage-sensing mediators and macrophage-derived mediators participate in the major macrophage-related signaling pathways in AKI, which regulate macrophage polarization and determine disease progression. In conclusion, macrophages change their roles and regulatory mechanisms during the occurrence and development of AKI. The aim of the present review was to contribute to an improved understanding of AKI and to the identification of novel therapeutic targets for this condition.
Collapse
Affiliation(s)
- Ning Li
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, P.R. China
| | - Jiale Chen
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, P.R. China
| | - Pengtao Wang
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, P.R. China
| | - Haojun Fan
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, P.R. China
| | - Shike Hou
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, P.R. China
| | - Yanhua Gong
- Institute of Disaster Medicine, Tianjin University, Tianjin 300072, P.R. China
| |
Collapse
|
16
|
Szczęśniak P, Henke T, Fröhlich S, Plessmann U, Urlaub H, Leng L, Bucala R, Grosse R, Meinhardt A, Klug J. Extracellular MIF, but not its homologue D-DT, promotes fibroblast motility independently of its receptor complex CD74/CD44. J Cell Sci 2021; 134:jcs.217356. [PMID: 33328325 DOI: 10.1242/jcs.217356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 12/07/2020] [Indexed: 11/20/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) and its homologue D-dopachrome tautomerase (D-DT) are widely expressed pro-inflammatory cytokines with chemokine-like functions that coordinate a wide spectrum of biological activities, such as migration. Here, we biotin-tagged intracellular MIF/D-DT in vivo to identify important cytosolic interactors and found a plethora of actin cytoskeleton-associated proteins. Although the receptor complex between CD74 and CD44 (CD74/CD44) is essential for signalling transduction in fibroblasts via extracellular MIF/D-DT, our interactome data suggested direct effects. We, thus, investigated whether MIF/D-DT can modulate cell migration independently of CD74/CD44. To distinguish between receptor- and non-receptor-mediated motility, we used fibroblasts that are either deficient or that express CD74/CD44 proteins, and treated them with recombinant MIF/D-DT. Interestingly, only MIF could stimulate chemokinesis in the presence or absence of CD74/CD44. The pro-migratory effects of MIF depended on lipid raft/caveolae-mediated but not clathrin-mediated endocytosis, on its tautomerase activity and, probably, on its thiol protein oxidoreductase activity. As MIF treatment restrained actin polymerisation in vitro, our findings establish a new intracellular role for MIF/D-DT in driving cell motility through modulation of the actin cytoskeleton.
Collapse
Affiliation(s)
- Paweł Szczęśniak
- Department of Anatomy and Cell Biology, Justus Liebig University, Aulweg 123, Gießen 35392, Germany
| | - Tamara Henke
- Department of Anatomy and Cell Biology, Justus Liebig University, Aulweg 123, Gießen 35392, Germany
| | - Suada Fröhlich
- Department of Anatomy and Cell Biology, Justus Liebig University, Aulweg 123, Gießen 35392, Germany
| | - Uwe Plessmann
- Max Planck Institute for Biophysical Chemistry, Bioanalytical Mass Spectrometry Group, Am Fassberg 11, 37077 Göttingen, Germany
| | - Henning Urlaub
- Max Planck Institute for Biophysical Chemistry, Bioanalytical Mass Spectrometry Group, Am Fassberg 11, 37077 Göttingen, Germany.,Institute for Clinical Chemistry, Research Group 'Bioanalytics', University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Lin Leng
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Robert Grosse
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Medical Faculty, Albertstraße 25, 79104 Freiburg, Germany
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology, Justus Liebig University, Aulweg 123, Gießen 35392, Germany
| | - Jörg Klug
- Department of Anatomy and Cell Biology, Justus Liebig University, Aulweg 123, Gießen 35392, Germany
| |
Collapse
|
17
|
Sun W, Zhu Q, Yan L, Shao F. Mesenchymal stem cells alleviate acute kidney injury via miR-107-mediated regulation of ribosomal protein S19. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:765. [PMID: 32042781 DOI: 10.21037/atm.2019.11.89] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background The characteristics of mesenchymal stem cells (MSCs) in the repair of acute kidney injury (AKI) have been extensively studied. However, some potential molecular mechanisms remain indistinct. The aim of this study was to combine published microRNA (miRNA) transcriptional profiling with quantitative proteomic analyses to reveal specific miRNAs or genes for MSC-based therapy in AKI. Methods Transcriptome data containing significantly changed miRNAs in renal tissue from AKI mice treated with and without MSCs were downloaded. Proteomics resources were downloaded from a human proximal renal tubule cell line (HK-2) that served as a good in vitro model for AKI treated with MSCs. We connected the proteomics data with transcriptional records based on miRNA function. Differentially expressed genes (DEGs) were sorted. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis was conducted, and protein-protein interaction (PPI) chains were formed. The genes identified in the analyses were verified in a cisplatin-induced AKI rat model and in HK-2 cells exposed to cisplatin and cocultured with MSCs. Results A total of 207 specific DEGs were sorted. The ribosomal pathway was identified in pathway enrichment, and ribosomal proteins were identified from the PPI network complex. The targeting of the microRNAs, miR-107 to RPS19, was directly verified by the dual-luciferase method. miR-107 knockdown induced RPS19 expression, protected HK-2 cells from cisplatin-induced apoptosis, and promoted cell proliferation. Conclusions By analyzing comprehensive bioinformatics data, we have confirmed the DEGs and pathways in AKI treated with MSCs. Bone marrow-derived MSCs reduce miR-107 expression and increase RPS19 expression by repressing the proliferation of cisplatin-induced AKI cells and initiating apoptosis.
Collapse
Affiliation(s)
- Weinan Sun
- Department of Nephrology, People's Hospital of Zhengzhou University, He'nan Provincial People's Hospital, He'nan Provincial Key Laboratory of Kidney Disease and Immunology, Zhengzhou 450003, China
| | - Qing Zhu
- Department of Nephrology, People's Hospital of Zhengzhou University, He'nan Provincial People's Hospital, He'nan Provincial Key Laboratory of Kidney Disease and Immunology, Zhengzhou 450003, China
| | - Lei Yan
- Department of Nephrology, People's Hospital of Zhengzhou University, He'nan Provincial People's Hospital, He'nan Provincial Key Laboratory of Kidney Disease and Immunology, Zhengzhou 450003, China
| | - Fengmin Shao
- Department of Nephrology, People's Hospital of Zhengzhou University, He'nan Provincial People's Hospital, He'nan Provincial Key Laboratory of Kidney Disease and Immunology, Zhengzhou 450003, China
| |
Collapse
|
18
|
Velásquez E, Martins-de-Souza D, Velásquez I, Carneiro GRA, Schmitt A, Falkai P, Domont GB, Nogueira FCS. Quantitative Subcellular Proteomics of the Orbitofrontal Cortex of Schizophrenia Patients. J Proteome Res 2019; 18:4240-4253. [PMID: 31581776 DOI: 10.1021/acs.jproteome.9b00398] [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: 12/15/2022]
Abstract
Schizophrenia is a chronic disease characterized by the impairment of mental functions with a marked social dysfunction. A quantitative proteomic approach using iTRAQ labeling and SRM, applied to the characterization of mitochondria (MIT), crude nuclear fraction (NUC), and cytoplasm (CYT), can allow the observation of dynamic changes in cell compartments providing valuable insights concerning schizophrenia physiopathology. Mass spectrometry analyses of the orbitofrontal cortex from 12 schizophrenia patients and 8 healthy controls identified 655 protein groups in the MIT fraction, 1500 in NUC, and 1591 in CYT. We found 166 groups of proteins dysregulated among all enriched cellular fractions. Through the quantitative proteomic analysis, we detect as the main biological pathways those related to calcium and glutamate imbalance, cell signaling disruption of CREB activation, axon guidance, and proteins involved in the activation of NF-kB signaling along with the increase of complement protein C3. Based on our data analysis, we suggest the activation of NF-kB as a possible pathway that links the deregulation of glutamate, calcium, apoptosis, and the activation of the immune system in schizophrenia patients. All MS data are available in the ProteomeXchange Repository under the identifier PXD015356 and PXD014350.
Collapse
Affiliation(s)
- Erika Velásquez
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry , Federal University of Rio de Janeiro , Rio de Janeiro 21941-909 , Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry, Institute of Biology , University of Campinas (UNICAMP) , Campinas 13083-970 , Brazil.,Experimental Medicine Research Cluster (EMRC) University of Campinas , Campinas 13083-887 , SP , Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION) , Conselho Nacional de Desenvolvimento Cientı́fico e Tecnológico (CNPq) , São Paulo , Brazil
| | | | - Gabriel Reis Alves Carneiro
- Laboratory of Proteomics, LADETEC, Institute of Chemistry , Federal University of Rio de Janeiro , Rio de Janeiro 21941-598 , Brazil
| | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy , Ludwig Maximilian University of Munich (LMU) , 80539 Munich , Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy , Ludwig Maximilian University of Munich (LMU) , 80539 Munich , Germany
| | - Gilberto B Domont
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry , Federal University of Rio de Janeiro , Rio de Janeiro 21941-909 , Brazil
| | - Fabio C S Nogueira
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry , Federal University of Rio de Janeiro , Rio de Janeiro 21941-909 , Brazil.,Laboratory of Proteomics, LADETEC, Institute of Chemistry , Federal University of Rio de Janeiro , Rio de Janeiro 21941-598 , Brazil
| |
Collapse
|
19
|
Li J, Tang Y, Tang PMK, Lv J, Huang XR, Carlsson-Skwirut C, Da Costa L, Aspesi A, Fröhlich S, Szczęśniak P, Lacher P, Klug J, Meinhardt A, Fingerle-Rowson G, Gong R, Zheng Z, Xu A, Lan HY. Blocking Macrophage Migration Inhibitory Factor Protects Against Cisplatin-Induced Acute Kidney Injury in Mice. Mol Ther 2018; 26:2523-2532. [PMID: 30077612 DOI: 10.1016/j.ymthe.2018.07.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 01/08/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF) is elevated in patients with acute kidney injury (AKI) and is suggested as a potential predictor for renal replacement therapy in AKI. In this study, we found that MIF also plays a pathogenic role and is a therapeutic target for AKI. In a cisplatin-induced AKI mouse model, elevated plasma MIF correlated with increased serum creatinine and the severity of renal inflammation and tubular necrosis, whereas deletion of MIF protected the kidney from cisplatin-induced AKI by largely improving renal functional and histological injury, and suppressing renal inflammation including upregulation of cytokines such as interleukin (IL)-1β, tumor necrosis factor-alpha (TNF-α), IL-6, inducible nitric oxide synthase (iNOS), MCP-1, IL-8, and infiltration of macrophages, neutrophils, and T cells. We next developed a novel therapeutic strategy for AKI by blocking the endogenous MIF with an MIF inhibitor, ribosomal protein S19 (RPS19). Similar to the MIF-knockout mice, treatment with RPS19, but not the mutant RPS19, suppressed cisplatin-induced AKI. Mechanistically, we found that both genetic knockout and pharmacological inhibition of MIF protected against AKI by inactivating the CD74-nuclear factor κB (NF-κB) signaling. In conclusion, MIF is pathogenic in cisplatin-induced AKI. Targeting MIF with an MIF inhibitor RPS19 could be a promising therapeutic potential for AKI.
Collapse
Affiliation(s)
- Jinhong Li
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Department of Anatomical and Cellular Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Ying Tang
- Department of Nephrology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Patrick M K Tang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Department of Anatomical and Cellular Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Lv
- Department of Nephrology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiao-Ru Huang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Department of Anatomical and Cellular Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Christine Carlsson-Skwirut
- Department of Woman and Child Health, Paediatric Endocrinology Unit, Astrid Lindgren Children's Hospital, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Lydie Da Costa
- AP-HP, Service d'Hématologie Biologique, Hôpital R. Debré, Université Paris Diderot, Sorbonne Paris Cité, Paris, France; INSERM U1149, CRI, Faculté de Médecine Bichat-Claude Bernard, Laboratoire d'Excellence GR-Ex, Paris, France
| | - Anna Aspesi
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy; Interdepartmental Center for Studies on Asbestos and Other Toxic Particulates "G. Scansetti," University of Turin, Turin, Italy
| | - Suada Fröhlich
- Department of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Pawel Szczęśniak
- Department of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Philipp Lacher
- Department of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Jörg Klug
- Department of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Günter Fingerle-Rowson
- Department I of Internal Medicine, University Hospital Cologne and Center for Integrated Oncology Köln-Bonn, Cologne, Germany
| | - Rujun Gong
- Division of Kidney Diseases and Hypertension, Rhode Island Hospital, Brown University School of Medicine, Providence, RI, USA
| | - Zhihua Zheng
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Anping Xu
- Department of Nephrology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Department of Anatomical and Cellular Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
20
|
Hollmann T, Kim TK, Tirloni L, Radulović ŽM, Pinto AFM, Diedrich JK, Yates JR, da Silva Vaz I, Mulenga A. Identification and characterization of proteins in the Amblyomma americanum tick cement cone. Int J Parasitol 2017; 48:211-224. [PMID: 29258831 DOI: 10.1016/j.ijpara.2017.08.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 08/26/2017] [Indexed: 01/08/2023]
Abstract
The adaptation of hard ticks to feed for long periods is facilitated by the cement cone, which securely anchors the tick mouthparts onto host skin and protects the tick from being groomed off by the host. Thus, preventing tick cement deposition is an attractive target for the development of innovative tick control. We used LC-MS/MS sequencing to identify 160 Amblyomma americanum tick cement proteins that include glycine-rich proteins (GRP, 19%), protease inhibitors (12%), proteins of unknown function (11%), mucin (4%), detoxification, storage, and lipocalin at 1% each, and housekeeping proteins (50%). Spatiotemporal transcription analysis showing mRNA expression in multiple tick organs and transcript abundance increasing with feeding suggest that selected GRPs (n = 13) regulate multiple tick feeding functions, being classified as constitutively expressed (CE), feeding induced (FI), and up-regulated with feeding (UR). We show that transcription of CE GRPs is likely under the control of tick appetence associated factors in that mRNA abundance increased several thousand fold in 1 week old adult ticks, the time period that coincides with tick attainment of appetence. Given the high number of targets, we synthesized and injected unfed ticks with combinatorial (co) double stranded (ds)RNA and disrupted GRP mRNA in clusters according to similar transcription patterns: CE (n = 3), FI, (n = 4), and UR (n = 6) to streamline the work. Our data suggest that CE and FI GRPs are important for maintenance of the tick feeding site in that reddening and subsequent bleeding were observed around the mouthparts of CE and FI GRP co-dsRNA injected ticks during feeding. Furthermore, although not significantly different, indices for blood meal size and fecundity were apparently reduced in FI and UR ticks. We discuss our data with reference to A. americanum tick feeding physiology.
Collapse
Affiliation(s)
- Taylor Hollmann
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Tae Kwon Kim
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Lucas Tirloni
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA; Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Željko M Radulović
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Antônio F M Pinto
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil; Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA; Mass Spectrometry Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jolene K Diedrich
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA; Mass Spectrometry Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - John R Yates
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Itabajara da Silva Vaz
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Albert Mulenga
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
21
|
Yang C, Huang XR, Fung E, Liu HF, Lan HY. The Regulatory T-cell Transcription Factor Foxp3 Protects against Crescentic Glomerulonephritis. Sci Rep 2017; 7:1481. [PMID: 28469165 PMCID: PMC5431186 DOI: 10.1038/s41598-017-01515-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 03/29/2017] [Indexed: 12/31/2022] Open
Abstract
Regulatory T cells (Tregs) have been shown to play a protective role in glomerulonephritis (GN) and Foxp3 is a master transcription factor in Treg development. In this study, we examined the functional role and mechanisms of Foxp3 in a mouse model of accelerated anti-glomerular basement membrane (anti-GBM) GN induced in antigen-primed Foxp3 transgenic (Tg) mice. Compared with littermate of wildtype (WT) mice in which induced severe crescentic GN developed with progressive renal dysfunction, Foxp3 Tg mice had reduced crescent formation, urinary protein excretion, plasma creatinine and decline in creatinine clearance. The protective role of Foxp3 in crescentic GN was associated with a markedly suppressed expression of proinflammatory interleukin-1 beta (IL-1β), tumour necrosis factor-alpha (TNF-α) and monocyte chemoattractant protein 1 (MCP-1), and diminished infiltration of the kidneys by CD3+ T cells and F4/80+ macrophages. Moreover, overexpression of Foxp3 resulted in a significant increase in CD4+ Foxp3+ Tregs systemically and in the diseased kidneys, thereby blunting Th1, Th2, and Th17 responses systemically and in the kidneys. In conclusion, Foxp3 protects against kidney injury in crescentic GN through enhancement of Treg numbers and function, and suppression of Th1, Th2 and Th17 immune responses at the systemic and local tissue levels.
Collapse
Affiliation(s)
- Chen Yang
- Institute of Nephrology, Guangdong Medical University, Zhanjiang, Guangdong, China.,Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, and Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiao-Ru Huang
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, and Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Erik Fung
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, and Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Hua-Feng Liu
- Institute of Nephrology, Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, and Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
22
|
The biological function and significance of CD74 in immune diseases. Inflamm Res 2016; 66:209-216. [DOI: 10.1007/s00011-016-0995-1] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 09/30/2016] [Indexed: 12/25/2022] Open
|
23
|
Valiño-Rivas L, Baeza-Bermejillo C, Gonzalez-Lafuente L, Sanz AB, Ortiz A, Sanchez-Niño MD. CD74 in Kidney Disease. Front Immunol 2015; 6:483. [PMID: 26441987 PMCID: PMC4585214 DOI: 10.3389/fimmu.2015.00483] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 09/05/2015] [Indexed: 12/17/2022] Open
Abstract
CD74 (invariant MHC class II) regulates protein trafficking and is a receptor for macrophage migration inhibitory factor (MIF) and d-dopachrome tautomerase (d-DT/MIF-2). CD74 expression is increased in tubular cells and/or glomerular podocytes and parietal cells in human metabolic nephropathies, polycystic kidney disease, graft rejection and kidney cancer and in experimental diabetic nephropathy and glomerulonephritis. Stressors like abnormal metabolite (glucose, lyso-Gb3) levels and inflammatory cytokines increase kidney cell CD74. MIF activates CD74 to increase inflammatory cytokines in podocytes and tubular cells and proliferation in glomerular parietal epithelial cells and cyst cells. MIF overexpression promotes while MIF targeting protects from experimental glomerular injury and kidney cysts, and interference with MIF/CD74 signaling or CD74 deficiency protected from crescentic glomerulonephritis. However, CD74 may protect from interstitial kidney fibrosis. Furthermore, CD74 expression by stressed kidney cells raises questions about the kidney safety of cancer therapy strategies delivering lethal immunoconjugates to CD74-expressing cells. Thus, understanding CD74 biology in kidney cells is relevant for kidney therapeutics.
Collapse
Affiliation(s)
- Lara Valiño-Rivas
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Universidad Autónoma de Madrid , Madrid , Spain
| | - Ciro Baeza-Bermejillo
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Universidad Autónoma de Madrid , Madrid , Spain ; Red de Investigación Renal (REDINREN) , Madrid , Spain
| | - Laura Gonzalez-Lafuente
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Universidad Autónoma de Madrid , Madrid , Spain
| | - Ana Belen Sanz
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Universidad Autónoma de Madrid , Madrid , Spain ; Red de Investigación Renal (REDINREN) , Madrid , Spain
| | - Alberto Ortiz
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Universidad Autónoma de Madrid , Madrid , Spain ; Red de Investigación Renal (REDINREN) , Madrid , Spain ; School of Medicine, Universidad Autónoma de Madrid , Madrid , Spain ; Fundacion Renal Iñigo Alvarez de Toledo-IRSIN , Madrid , Spain
| | - Maria Dolores Sanchez-Niño
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Universidad Autónoma de Madrid , Madrid , Spain ; Red de Investigación Renal (REDINREN) , Madrid , Spain
| |
Collapse
|
24
|
Díaz-Martín V, Manzano-Román R, Oleaga A, Pérez-Sánchez R. New salivary anti-haemostatics containing protective epitopes from Ornithodoros moubata ticks: Assessment of their individual and combined vaccine efficacy. Vet Parasitol 2015; 212:336-49. [PMID: 26293586 DOI: 10.1016/j.vetpar.2015.08.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/31/2015] [Accepted: 08/03/2015] [Indexed: 12/28/2022]
Abstract
Ornithodoros moubata is the main vector of the pathogens causing African swine fever and human relapsing fever in Africa. The development of an efficient vaccine against this tick would facilitate its control and the prevention of the diseases it transmits to a considerable extent. Previous efforts to identify vaccine target candidates led us to the discovery of novel salivary proteins that probably act as anti-haemostatics at the host-tick interface, including a secreted phospholipase A2 (PLA2), a 7DB-like protein (7DB-like), a riboprotein 60S L10 (RP-60S), an apyrase (APY), and a new platelet aggregation inhibitor peptide, designated mougrin (MOU). In this work, the corresponding recombinant proteins were expressed in Escherichia coli and their individual vaccine efficacy was tested in rabbit vaccination trials. All of them, except the less immunogenic RP-60S, induced strong humoral responses that reduced tick feeding and survival, providing vaccine efficacies of 44.2%, 43.2% and 27.2%, 19.9% and 17.3% for PLA2, APY, MOU, RP-60S and 7DB-like, respectively. In the case of the more protective recombinant antigens (PLA2, APY and MOU), the immunodominant protective linear B-cell epitopes were identified and their combined vaccine efficacy was tested in a second vaccine trial using different adjuvants. In comparison with the best efficacy of individual antigens, the multicomponent vaccine increased vaccine efficacy by 13.6%, indicating additive protective effects rather than a synergistic effect. Tick saliva inoculated during natural tick-host contacts had a boosting effect on vaccinated animals, increasing specific antibody levels and protection.
Collapse
Affiliation(s)
- Verónica Díaz-Martín
- Parasitología Animal, Instituto de Recursos Naturales y Agrobiología de Salamanca (IRNASA, CSIC), Cordel de Merinas, 40-52, 37008 Salamanca, Spain.
| | - Raúl Manzano-Román
- Parasitología Animal, Instituto de Recursos Naturales y Agrobiología de Salamanca (IRNASA, CSIC), Cordel de Merinas, 40-52, 37008 Salamanca, Spain.
| | - Ana Oleaga
- Parasitología Animal, Instituto de Recursos Naturales y Agrobiología de Salamanca (IRNASA, CSIC), Cordel de Merinas, 40-52, 37008 Salamanca, Spain.
| | - Ricardo Pérez-Sánchez
- Parasitología Animal, Instituto de Recursos Naturales y Agrobiología de Salamanca (IRNASA, CSIC), Cordel de Merinas, 40-52, 37008 Salamanca, Spain.
| |
Collapse
|
25
|
Lewis LA, Radulović ŽM, Kim TK, Porter LM, Mulenga A. Identification of 24h Ixodes scapularis immunogenic tick saliva proteins. Ticks Tick Borne Dis 2015; 6:424-34. [PMID: 25825233 PMCID: PMC4415496 DOI: 10.1016/j.ttbdis.2015.03.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 02/19/2015] [Accepted: 03/09/2015] [Indexed: 12/13/2022]
Abstract
Ixodes scapularis is arguably the most medically important tick species in the United States. This tick transmits 5 of the 14 human tick-borne disease (TBD) agents in the USA: Borrelia burgdorferi, Anaplasma phagocytophilum, B. miyamotoi, Babesia microti, and Powassan virus disease. Except for the Powassan virus disease, I. scapularis-vectored TBD agents require more than 24h post attachment to be transmitted. This study describes identification of 24h immunogenic I. scapularis tick saliva proteins, which could provide opportunities to develop strategies to stop tick feeding before transmission of the majority of pathogens. A 24h fed female I. scapularis phage display cDNA expression library was biopanned using rabbit antibodies to 24h fed I. scapularis female tick saliva proteins, subjected to next generation sequencing, de novo assembly, and bioinformatic analyses. A total of 182 contigs were assembled, of which ∼19% (35/182) are novel and did not show identity to any known proteins in GenBank. The remaining ∼81% (147/182) of contigs were provisionally identified based on matches in GenBank including ∼18% (27/147) that matched protein sequences previously annotated as hypothetical and putative tick saliva proteins. Others include proteases and protease inhibitors (∼3%, 5/147), transporters and/or ligand binding proteins (∼6%, 9/147), immunogenic tick saliva housekeeping enzyme-like (17%, 25/147), ribosomal protein-like (∼31%, 46/147), and those classified as miscellaneous (∼24%, 35/147). Notable among the miscellaneous class include antimicrobial peptides (microplusin and ricinusin), myosin-like proteins that have been previously found in tick saliva, and heat shock tick saliva protein. Data in this study provides the foundation for in-depth analysis of I. scapularis feeding during the first 24h, before the majority of TBD agents can be transmitted.
Collapse
Affiliation(s)
- Lauren A Lewis
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, United States
| | - Željko M Radulović
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, United States
| | - Tae K Kim
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, United States
| | - Lindsay M Porter
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, United States
| | - Albert Mulenga
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, United States.
| |
Collapse
|
26
|
Zhou X, Liao WJ, Liao JM, Liao P, Lu H. Ribosomal proteins: functions beyond the ribosome. J Mol Cell Biol 2015; 7:92-104. [PMID: 25735597 DOI: 10.1093/jmcb/mjv014] [Citation(s) in RCA: 475] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 12/05/2014] [Indexed: 01/05/2023] Open
Abstract
Although ribosomal proteins are known for playing an essential role in ribosome assembly and protein translation, their ribosome-independent functions have also been greatly appreciated. Over the past decade, more than a dozen of ribosomal proteins have been found to activate the tumor suppressor p53 pathway in response to ribosomal stress. In addition, these ribosomal proteins are involved in various physiological and pathological processes. This review is composed to overview the current understanding of how ribosomal stress provokes the accumulation of ribosome-free ribosomal proteins, as well as the ribosome-independent functions of ribosomal proteins in tumorigenesis, immune signaling, and development. We also propose the potential of applying these pieces of knowledge to the development of ribosomal stress-based cancer therapeutics.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Wen-Juan Liao
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jun-Ming Liao
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Peng Liao
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Hua Lu
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
27
|
Wang W, Nag S, Zhang X, Wang MH, Wang H, Zhou J, Zhang R. Ribosomal proteins and human diseases: pathogenesis, molecular mechanisms, and therapeutic implications. Med Res Rev 2014; 35:225-85. [PMID: 25164622 DOI: 10.1002/med.21327] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ribosomes are essential components of the protein synthesis machinery. The process of ribosome biogenesis is well organized and tightly regulated. Recent studies have shown that ribosomal proteins (RPs) have extraribosomal functions that are involved in cell proliferation, differentiation, apoptosis, DNA repair, and other cellular processes. The dysfunction of RPs has been linked to the development and progression of hematological, metabolic, and cardiovascular diseases and cancer. Perturbation of ribosome biogenesis results in ribosomal stress, which triggers activation of the p53 signaling pathway through RPs-MDM2 interactions, resulting in p53-dependent cell cycle arrest and apoptosis. RPs also regulate cellular functions through p53-independent mechanisms. We herein review the recent advances in several forefronts of RP research, including the understanding of their biological features and roles in regulating cellular functions, maintaining cell homeostasis, and their involvement in the pathogenesis of human diseases. We also highlight the translational potential of this research for the identification of molecular biomarkers, and in the discovery and development of novel treatments for human diseases.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, 79106; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, 79106
| | | | | | | | | | | | | |
Collapse
|
28
|
Radulović ŽM, Kim TK, Porter LM, Sze SH, Lewis L, Mulenga A. A 24-48 h fed Amblyomma americanum tick saliva immuno-proteome. BMC Genomics 2014; 15:518. [PMID: 24962723 PMCID: PMC4099483 DOI: 10.1186/1471-2164-15-518] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 06/12/2014] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Multiple tick saliva proteins, the majority of which are unknown, confer tick resistance in repeatedly infested animals. The objective of this study was to identify the 24-48 h fed Amblyomma americanum tick saliva immuno-proteome. The 24-48 h tick-feeding phase is critical to tick parasitism as it precedes important events in tick biology, blood meal feeding and disease agent transmission. Fed male, 24 and 96 h fed female phage display cDNA expression libraries were biopanned using rabbit antibodies to 24 and 48 h fed A. americanum female tick saliva proteins. Biopanned immuno-cDNA libraries were subjected to next generation sequencing, de novo assembly, and bioinformatic analysis. RESULTS More than 800 transcripts that code for 24-48 h fed A. americanum immuno-proteins are described. Of the 895 immuno-proteins, 52% (464/895) were provisionally identified based on matches in GenBank. Of these, ~19% (86/464) show high level of identity to other tick hypothetical proteins, and the rest include putative proteases (serine, cysteine, leukotriene A-4 hydrolase, carboxypeptidases, and metalloproteases), protease inhibitors (serine and cysteine protease inhibitors, tick carboxypeptidase inhibitor), and transporters and/or ligand binding proteins (histamine binding/lipocalin, fatty acid binding, calreticulin, hemelipoprotein, IgG binding protein, ferritin, insulin-like growth factor binding proteins, and evasin). Others include enzymes (glutathione transferase, cytochrome oxidase, protein disulfide isomerase), ribosomal proteins, and those of miscellaneous functions (histamine release factor, selenoproteins, tetraspanin, defensin, heat shock proteins). CONCLUSIONS Data here demonstrate that A. americanum secretes a complex cocktail of immunogenic tick saliva proteins during the first 24-48 h of feeding. Of significance, previously validated immunogenic tick saliva proteins including AV422 protein, calreticulin, histamine release factor, histamine binding/lipocalins, selenoproteins, and paramyosin were identified in this screen, supporting the specificity of the approach in this study. While descriptive, this study opens opportunities for in-depth tick feeding physiology studies.
Collapse
Affiliation(s)
- Željko M Radulović
- />Department of Entomology, AgriLife Research, Texas A & M University, 2475 TAMU, College Station, TX77843 USA
| | - Tae K Kim
- />Department of Entomology, AgriLife Research, Texas A & M University, 2475 TAMU, College Station, TX77843 USA
| | - Lindsay M Porter
- />Department of Entomology, AgriLife Research, Texas A & M University, 2475 TAMU, College Station, TX77843 USA
| | - Sing-Hoi Sze
- />Department of Computer Sciences and Engineering, Texas A & M University, College Station, TX77843 USA
- />Department of Biochemistry & Biophysics, Texas A & M University, College Station, TX77843 USA
| | - Lauren Lewis
- />Department of Entomology, AgriLife Research, Texas A & M University, 2475 TAMU, College Station, TX77843 USA
| | - Albert Mulenga
- />Department of Entomology, AgriLife Research, Texas A & M University, 2475 TAMU, College Station, TX77843 USA
| |
Collapse
|
29
|
Acquadro E, Caron I, Tortarolo M, Bucci EM, Bendotti C, Corpillo D. Human SOD1-G93A specific distribution evidenced in murine brain of a transgenic model for amyotrophic lateral sclerosis by MALDI imaging mass spectrometry. J Proteome Res 2014; 13:1800-9. [PMID: 24579824 DOI: 10.1021/pr400942n] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive, fatal neurodegenerative disease caused by the degeneration of motor neurons. The transgenic mouse model carrying the human SOD1G93A mutant gene (hSOD1G93A mouse) represents one of the most reliable and widely used model of this pathology. In the present work, the innovative technique of matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) was applied in the study of pathological alterations at the level of small brain regions such as facial and trigeminal nuclei, which in rodents are extremely small and would be difficult to analyze with classical proteomics approaches. Comparing slices from three mice groups (transgenic hSOD1G93A, transgenic hSOD1WT, and nontransgenic, Ntg), this technique allowed us to evidence the accumulation of hSOD1G93A in the facial and trigeminal nuclei, where it generates aggregates. This phenomenon is likely to be correlated to the degeneration observed in these regions. Moreover, a statistical analysis allowed us to highlight other proteins as differentially expressed among the three mice groups analyzed. Some of them were identified by reverse-phase HPLC fractionation of extracted proteins and mass spectrometric analysis before and after trypsin digestion. In particular, the 40S ribosomal protein S19 (RPS19) was upregulated in the parenkyma and reactive glial cells in facial nuclei of hSOD1G93A mice when compared to transgenic hSOD1WT and nontransgenic ones.
Collapse
Affiliation(s)
- Elena Acquadro
- ABLE Bioscences, BioIndustry Park Silvano Fumero S.p.A., Via Ribes 5, 10010 Colleretto Giacosa, TO, Italy
| | | | | | | | | | | |
Collapse
|
30
|
Knight JRP, Willis AE, Milner J. Active regulator of SIRT1 is required for ribosome biogenesis and function. Nucleic Acids Res 2013; 41:4185-97. [PMID: 23462953 PMCID: PMC3627601 DOI: 10.1093/nar/gkt129] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Active regulator of SIRT1 (AROS) binds and upregulates SIRT1, an NAD(+)-dependent deacetylase. In addition, AROS binds RPS19, a structural ribosomal protein, which also functions in ribosome biogenesis and is implicated in multiple disease states. The significance of AROS in relation to ribosome biogenesis and function is unknown. Using human cells, we now show that AROS localizes to (i) the nucleolus and (ii) cytoplasmic ribosomes. Co-localization with nucleolar proteins was verified by confocal immunofluorescence of endogenous protein and confirmed by AROS depletion using RNAi. AROS association with cytoplasmic ribosomes was analysed by sucrose density fractionation and immunoprecipitation, revealing that AROS selectively associates with 40S ribosomal subunits and also with polysomes. RNAi-mediated depletion of AROS leads to deficient ribosome biogenesis with aberrant precursor ribosomal RNA processing, reduced 40S subunit ribosomal RNA and 40S ribosomal proteins (including RPS19). Together, this results in a reduction in 40S subunits and translating polysomes, correlating with reduced overall cellular protein synthesis. Interestingly, knockdown of AROS also results in a functionally significant increase in eIF2α phosphorylation. Overall, our results identify AROS as a factor with a role in both ribosome biogenesis and ribosomal function.
Collapse
Affiliation(s)
- John R P Knight
- Department of Biology, University of York, Wentworth Way, York, YO10 5DD, UK
| | | | | |
Collapse
|