1
|
Gallo PM, Chain RW, Xu J, Whiteman LM, Palladino A, Caricchio R, Costa-Reis P, Sullivan KE, Gallucci S. EGFR-ErbB2 dual kinase inhibitor lapatinib decreases autoantibody levels and worsens renal disease in Interferon α-accelerated murine lupus. Int Immunopharmacol 2024; 140:112692. [PMID: 39079344 PMCID: PMC11456265 DOI: 10.1016/j.intimp.2024.112692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 09/01/2024]
Abstract
Glomerulonephritis remains a major cause of morbidity and mortality in systemic lupus erythematosus (SLE). We have reported that expression of HER2/ErbB2, a member of the EGFR family, is increased in kidneys of patients and mice with lupus nephritis. We therefore asked if EGFR-family inhibition could ameliorate murine lupus nephritis. We used lapatinib, an EGFR-ErbB2 dual kinase inhibitor in female lupus-prone NZBxW/F1 mice, in which lupus onset was accelerated by injecting an IFN-α-expressing adenovirus. Mice received lapatinib (75 mg/Kg) or vehicle from the beginning of the acceleration or after the mice developed severe proteinuria (>300 mg/dL). Autoantibodies, kidney disease and markers of fibrosis and wound healing were analyzed. Exposure to IFNα induced ErbB2 expression in the kidney of lupus prone mice. Lapatinib, administered before but not after renal disease onset, lowered autoantibody titers and lessened immune complex deposition in the kidney. However, lapatinib increased proteinuria, kidney fibrosis and mouse mortality. Lapatinib also inhibited an in vitro wound healing assay testing renal cells. Our results suggest that EGFR-ErbB2 dual kinase inhibitor lapatinib decreases autoimmunity but worsens renal disease in IFNα-accelerated lupus, by increasing fibrosis and inhibiting wound healing. Type I Interferons are highlighted as important regulators of HER2/ErbB2 expression in the kidney. Further studies are required to parse the beneficial aspects of EGFR inhibition on autoimmunity from its negative effects on wound healing in lupus nephritis.
Collapse
Affiliation(s)
- Paul M Gallo
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Robert W Chain
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Jun Xu
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Leah M Whiteman
- Division of Innate Immunity, Department of Medicine, UMass Chan Medical School, Worcester, MA, USA
| | - Annette Palladino
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Roberto Caricchio
- Section of Rheumatology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Patricia Costa-Reis
- Division of Allergy Immunology, Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kathleen E Sullivan
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; Division of Innate Immunity, Department of Medicine, UMass Chan Medical School, Worcester, MA, USA; Section of Rheumatology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; Division of Allergy Immunology, Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Stefania Gallucci
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; Division of Innate Immunity, Department of Medicine, UMass Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
2
|
Liu Z, Fu Y, Yan M, Zhang S, Cai J, Chen G, Dong Z. microRNAs in kidney diseases: Regulation, therapeutics, and biomarker potential. Pharmacol Ther 2024; 262:108709. [PMID: 39181246 DOI: 10.1016/j.pharmthera.2024.108709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/01/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
MicroRNAs (miRNAs) are small, non-coding RNA molecules that play a crucial role in regulating gene expression by inhibiting the translation of their specific target messenger RNAs. To date, numerous studies have demonstrated changes in the expression of miRNAs in the kidneys throughout the progression of both acute kidney injury (AKI) and chronic kidney disease (CKD) in both human patients and experimental models. The role of specific microRNAs in the pathogenesis of kidney diseases has also been demonstrated. Further studies have elucidated the regulation of these microRNAs in diseased kidneys. Besides, certain miRNAs are detected in plasma and/or urine in kidney diseases and are potential diagnostic biomarkers. In this review, we provide an overview of recent developments in our understanding of how miRNAs contribute to kidney diseases. We also explore the potential of miRNAs as both biomarkers and therapeutic targets for these conditions, and highlight future research directions.
Collapse
Affiliation(s)
- Zhiwen Liu
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China.
| | - Ying Fu
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Mingjuan Yan
- Changde Hospital, Xiangya School of Medicine, Central South University, China
| | - Subing Zhang
- Youxian People's Hospital, Youxian, Hunan 412300, China
| | - Juan Cai
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Guochun Chen
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, USA.
| |
Collapse
|
3
|
Tawengi M, Al-Dali Y, Tawengi A, Benter IF, Akhtar S. Targeting the epidermal growth factor receptor (EGFR/ErbB) for the potential treatment of renal pathologies. Front Pharmacol 2024; 15:1394997. [PMID: 39234105 PMCID: PMC11373609 DOI: 10.3389/fphar.2024.1394997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024] Open
Abstract
Epidermal growth factor receptor (EGFR), which is referred to as ErbB1/HER1, is the prototype of the EGFR family of receptor tyrosine kinases which also comprises ErbB2 (Neu, HER2), ErbB3 (HER3), and ErbB4 (HER4). EGFR, along with other ErbBs, is expressed in the kidney tubules and is physiologically involved in nephrogenesis and tissue repair, mainly following acute kidney injury. However, its sustained activation is linked to several kidney pathologies, including diabetic nephropathy, hypertensive nephropathy, glomerulonephritis, chronic kidney disease, and renal fibrosis. This review aims to provide a summary of the recent findings regarding the consequences of EGFR activation in several key renal pathologies. We also discuss the potential interplay between EGFR and the reno-protective angiotensin-(1-7) (Ang-(1-7), a heptapeptide member of the renin-angiotensin-aldosterone system that counter-regulates the actions of angiotensin II. Ang-(1-7)-mediated inhibition of EGFR transactivation might represent a potential mechanism of action for its renoprotection. Our review suggests that there is a significant body of evidence supporting the potential inhibition of EGFR/ErbB, and/or administration of Ang-(1-7), as potential novel therapeutic strategies in the treatment of renal pathologies. Thus, EGFR inhibitors such as Gefitinib and Erlinotib that have an acceptable safety profile and have been clinically used in cancer chemotherapy since their FDA approval in the early 2000s, might be considered for repurposing in the treatment of renal pathologies.
Collapse
Affiliation(s)
- Mohamed Tawengi
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Yazan Al-Dali
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Ibrahim F Benter
- Faculty of Pharmacy, Final International University, Kyrenia, Cyprus
| | - Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
4
|
Guo Q, Qiao P, Wang J, Zhao L, Guo Z, Li X, Fan X, Yu C, Zhang L. Investigating the value of urinary biomarkers in relation to lupus nephritis histopathology: present insights and future prospects. Front Pharmacol 2024; 15:1421657. [PMID: 39104393 PMCID: PMC11298450 DOI: 10.3389/fphar.2024.1421657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Lupus nephritis (LN), a leading cause of death in Systemic Lupus Erythematosus (SLE) patients, presents significant diagnostic and prognostic challenges. Although renal pathology offers critical insights regarding the diagnosis, classification, and therapy for LN, its clinical utility is constrained by the invasive nature and limited reproducibility of renal biopsies. Moreover, the continuous monitoring of renal pathological changes through repeated biopsies is impractical. Consequently, there is a growing interest in exploring urine as a non-invasive, easily accessible, and dynamic "liquid biopsy" alternative to guide clinical management. This paper examines novel urinary biomarkers from a renal pathology perspective, encompassing cellular components, cytokines, adhesion molecules, auto-antibodies, soluble leukocyte markers, light chain fragments, proteins, small-molecule peptides, metabolomics, urinary exosomes, and ribonucleic acids. We also discuss the application of combined models comprising multiple biomarkers in assessing lupus activity. These innovative biomarkers and models offer insights into LN disease activity, acute and chronic renal indices, fibrosis, thrombotic microangiopathy, podocyte injury, and other pathological changes, potentially improving the diagnosis, management, and prognosis of LN. These urinary biomarkers or combined models may serve as viable alternatives to traditional renal pathology, potentially revolutionizing the method for future LN diagnosis and observation.
Collapse
Affiliation(s)
- Qianyu Guo
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, China
| | - Pengyan Qiao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, China
| | - Juanjuan Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Stem Cell Translational Laboratory, Shanxi Bethune Hospital, Taiyuan, China
| | - Li Zhao
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Zhiying Guo
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, China
| | - Xiaochen Li
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, China
| | - Xiuying Fan
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Office of Drug Clinical Trial Institution, Taiyuan, China
| | - Chong Yu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, China
| | - Liyun Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, China
- Stem Cell Translational Laboratory, Shanxi Bethune Hospital, Taiyuan, China
- Office of Drug Clinical Trial Institution, Taiyuan, China
| |
Collapse
|
5
|
Garmaa G, Bunduc S, Kói T, Hegyi P, Csupor D, Ganbat D, Dembrovszky F, Meznerics FA, Nasirzadeh A, Barbagallo C, Kökény G. A Systematic Review and Meta-Analysis of microRNA Profiling Studies in Chronic Kidney Diseases. Noncoding RNA 2024; 10:30. [PMID: 38804362 PMCID: PMC11130806 DOI: 10.3390/ncrna10030030] [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: 04/08/2024] [Revised: 04/24/2024] [Accepted: 04/28/2024] [Indexed: 05/29/2024] Open
Abstract
Chronic kidney disease (CKD) represents an increasing health burden. Evidence suggests the importance of miRNA in diagnosing CKD, yet the reports are inconsistent. This study aimed to determine novel miRNA biomarkers and potential therapeutic targets from hypothesis-free miRNA profiling studies in human and murine CKDs. Comprehensive literature searches were conducted on five databases. Subgroup analyses of kidney diseases, sample types, disease stages, and species were conducted. A total of 38 human and 12 murine eligible studies were analyzed using Robust Rank Aggregation (RRA) and vote-counting analyses. Gene set enrichment analyses of miRNA signatures in each kidney disease were conducted using DIANA-miRPath v4.0 and MIENTURNET. As a result, top target genes, Gene Ontology terms, the interaction network between miRNA and target genes, and molecular pathways in each kidney disease were identified. According to vote-counting analysis, 145 miRNAs were dysregulated in human kidney diseases, and 32 were dysregulated in murine CKD models. By RRA, miR-26a-5p was significantly reduced in the kidney tissue of Lupus nephritis (LN), while miR-107 was decreased in LN patients' blood samples. In both species, epithelial-mesenchymal transition, Notch, mTOR signaling, apoptosis, G2/M checkpoint, and hypoxia were the most enriched pathways. These miRNA signatures and their target genes must be validated in large patient cohort studies.
Collapse
Affiliation(s)
- Gantsetseg Garmaa
- Institute of Translational Medicine, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary; (G.G.); (A.N.)
- Center for Translational Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (S.B.); (T.K.); (P.H.); (D.C.); (F.D.); (F.A.M.)
- Department of Pathology, School of Medicine, Mongolian National University of Medical Sciences, Ulan-Bator 14210, Mongolia;
| | - Stefania Bunduc
- Center for Translational Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (S.B.); (T.K.); (P.H.); (D.C.); (F.D.); (F.A.M.)
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Dionisie Lupu Street 37, 020021 Bucharest, Romania
- Fundeni Clinical Institute, Fundeni Street 258, 022328 Bucharest, Romania
- Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, Baross út 22-24, 1085 Budapest, Hungary
| | - Tamás Kói
- Center for Translational Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (S.B.); (T.K.); (P.H.); (D.C.); (F.D.); (F.A.M.)
- Department of Stochastics, Institute of Mathematics, Budapest University of Technology and Economics, Műegyetem rkp. 3, 1111 Budapest, Hungary
| | - Péter Hegyi
- Center for Translational Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (S.B.); (T.K.); (P.H.); (D.C.); (F.D.); (F.A.M.)
- Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, Baross út 22-24, 1085 Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Dezső Csupor
- Center for Translational Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (S.B.); (T.K.); (P.H.); (D.C.); (F.D.); (F.A.M.)
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
- Institute of Clinical Pharmacy, University of Szeged, Szikra utca 8, 6725 Szeged, Hungary
| | - Dariimaa Ganbat
- Department of Pathology, School of Medicine, Mongolian National University of Medical Sciences, Ulan-Bator 14210, Mongolia;
- Department of Public Health, Graduate School of Medicine, International University of Health and Welfare, Tokyo 107-840, Japan
| | - Fanni Dembrovszky
- Center for Translational Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (S.B.); (T.K.); (P.H.); (D.C.); (F.D.); (F.A.M.)
- Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, Baross út 22-24, 1085 Budapest, Hungary
| | - Fanni Adél Meznerics
- Center for Translational Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (S.B.); (T.K.); (P.H.); (D.C.); (F.D.); (F.A.M.)
- Department of Dermatology, Venereology and Dermatooncology, Semmelweis University, Mária utca 41, 1085 Budapest, Hungary
| | - Ailar Nasirzadeh
- Institute of Translational Medicine, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary; (G.G.); (A.N.)
| | - Cristina Barbagallo
- Section of Biology and Genetics “G. Sichel”, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Gábor Kökény
- Institute of Translational Medicine, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary; (G.G.); (A.N.)
- International Nephrology Research and Training Center, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary
| |
Collapse
|
6
|
Nezhad Nezhad MT, Rajabi M, Nekooeizadeh P, Sanjari S, Pourvirdi B, Heidari MM, Veradi Esfahani P, Abdoli A, Bagheri S, Tobeiha M. Systemic lupus erythematosus: From non-coding RNAs to exosomal non-coding RNAs. Pathol Res Pract 2023; 247:154508. [PMID: 37224659 DOI: 10.1016/j.prp.2023.154508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/01/2023] [Accepted: 05/05/2023] [Indexed: 05/26/2023]
Abstract
Systemic lupus erythematosus (SLE), as an immunological illness, frequently impacts young females. Both vulnerabilities to SLE and the course of the illness's clinical symptoms have been demonstrated to be affected by individual differences in non-coding RNA expression. Many non-coding RNAs (ncRNAs) are out of whack in patients with SLE. Because of the dysregulation of several ncRNAs in peripheral blood of patients suffering from SLE, these ncRNAs to be showed valuable as biomarkers for medication response, diagnosis, and activity. NcRNAs have also been demonstrated to influence immune cell activity and apoptosis. Altogether, these facts highlight the need of investigating the roles of both families of ncRNAs in the progress of SLE. Being aware of the significance of these transcripts perhaps elucidates the molecular pathogenesis of SLE and could open up promising avenues to create tailored treatments during this condition. In this review we summarized various non-coding RNAs and Exosomal non-coding RNAs in SLE.
Collapse
Affiliation(s)
| | - Mohammadreza Rajabi
- Student Research Committee، Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pegah Nekooeizadeh
- Student Research Committee، Shiraz University of Medical Sciences, Shiraz, Iran
| | - Siavash Sanjari
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran; School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Bita Pourvirdi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran; School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Mehdi Heidari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Department of Pediatric, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Pegah Veradi Esfahani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran; School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Amirhossein Abdoli
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran; School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Sahar Bagheri
- Diabetes Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Mohammad Tobeiha
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Department of Pediatric, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
7
|
Choi D, Kim J, Yang JW, Kim JH, Park S, Shin JI. Dysregulated MicroRNAs in the Pathogenesis of Systemic Lupus Erythematosus: A Comprehensive Review. Int J Biol Sci 2023; 19:2495-2514. [PMID: 37215992 PMCID: PMC10197884 DOI: 10.7150/ijbs.74315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 12/11/2022] [Indexed: 05/24/2023] Open
Abstract
Systemic lupus erythematosus is a chronic autoimmune disease of which clinical presentation is vastly heterogeneous, ranging from mild skin rashes to severe renal diseases. Treatment goal of this illness is to minimize disease activity and prevent further organ damage. In recent years, much research has been done on the epigenetic aspects of SLE pathogenesis, for among the various factors known to contribute to the pathogenic process, epigenetic factors, especially microRNAs, bear the most therapeutic potential that can be altered unlike congenital genetic factors. This article reviews and updates what has been discovered so far about the pathogenesis of lupus, while focusing on the dysregulation of microRNAs in lupus patients in comparison to healthy controls along with the potentially pathogenic roles of the microRNAs commonly reported to be either upregulated or downregulated. Furthermore, this review includes microRNAs of which results are controversial, suggesting possible explanations for such discrepancies and directions for future research. Moreover, we aimed to emphasize the point that had been overlooked so far in studies regarding microRNA expression levels; that is, which specimen was used to assess the dysregulation of microRNAs. To our surprise, a vast number of studies have not considered this factor and have analyzed the potential role of microRNAs in general. Despite extensive investigations done on microRNA levels, their significance and potential role remain a mystery, which calls for further studies on this particular subject in regard of which specimen is used for assessment.
Collapse
Affiliation(s)
- Daeun Choi
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jimin Kim
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Won Yang
- Department of Nephrology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Ji Hong Kim
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seoyeon Park
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
8
|
Roointan A, Gholaminejad A, Shojaie B, Hudkins KL, Gheisari Y. Candidate MicroRNA Biomarkers in Lupus Nephritis: A Meta-analysis of Profiling Studies in Kidney, Blood and Urine Samples. Mol Diagn Ther 2023; 27:141-158. [PMID: 36520403 DOI: 10.1007/s40291-022-00627-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2022] [Indexed: 12/16/2022]
Abstract
CONTEXT Lupus nephritis (LN) is a kidney disease caused by systemic lupus erythematosus in which kidneys are attacked by the immune system. So far, various investigations have reported altered miRNA expression profiles in LN patients and different miRNAs have been introduced as biomarkers and/or therapeutic targets in LN. The aim of this study was to introduce a consensus panel of potential miRNA biomarkers by performing a meta-analysis of miRNA profiles in the LN patients. MATERIALS AND METHODS A comprehensive literature review approach was performed to find LN-related miRNA expression profiles in renal tissues, blood, and urine samples. After selecting the eligible studies and performing the data extraction, meta-analysis was done based on the vote-counting rank strategy as well as meta-analysis of p-values. The meta-miRNAs and their related genes were subjected to functional enrichment analyses and network construction. RESULTS The results of the meta-analysis of 41 studies were three lists of consensus miRNAs with altered expression profiles in the various tissue samples of LN patients (meta-analysis of p-values < 0.05). Of the 13 studies on kidney tissue, the meta-miRNAs were let-7a, miR-198, let-7e, miR-145, and miR-26a. In addition, meta-miRNAs of miR-199a, miR-21, miR-423, miR-1260b, miR-589, miR-150, miR-155, miR-146a, and miR-183 from 21 studies on blood samples, and miR-146a, miR-204, miR-30c, miR-3201, and miR-1273e from 11 studies on urine samples can be considered as non-invasive biomarker panels for LN. Functional enrichment analysis on the meta-miRNA lists confirmed the involvement of their target genes in nephropathy-related signaling pathways. CONCLUSION Using a meta-analytical approach, our study proposes three meta-miRNA panels that could be the target of further research to assess their potential as therapeutic targets/biomarkers in LN disease.
Collapse
Affiliation(s)
- Amir Roointan
- Faculty of Medicine, Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Hezar jarib St, Isfahan, 81746-73461, Iran
| | - Alieh Gholaminejad
- Faculty of Medicine, Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Hezar jarib St, Isfahan, 81746-73461, Iran.
| | - Behrokh Shojaie
- Faculty of Medicine, Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Hezar jarib St, Isfahan, 81746-73461, Iran
| | - Kelly L Hudkins
- Department of Pathology, School of Medicine, University of Washington, Seattle, USA
| | - Yousof Gheisari
- Faculty of Medicine, Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Hezar jarib St, Isfahan, 81746-73461, Iran
| |
Collapse
|
9
|
Exploring the Molecular Mechanism of Zhi Bai Di Huang Wan in the Treatment of Systemic Lupus Erythematosus Based on Network Pharmacology and Molecular Docking Techniques. Processes (Basel) 2022. [DOI: 10.3390/pr10101914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective: To investigate the molecular mechanism and simulated validation of Zhi Bai Di Huang Pill (ZBDHP) for the treatment of systemic lupus erythematosus (SLE) using network pharmacology and molecular docking techniques. Methods: The active ingredients of ZBDHP were obtained through the TCMSP database and the Canonical SMILES of the active ingredients were queried through Pubchem. The targets of the active ingredients were predicted in the SwissTarget database based on the SMILES. The SLE-related disease targets were obtained through the GeneCards, OMIM and DisGenets databases, and the intersection targets of ZBDHP and SLE were obtained using the Venny 2.1.0 online platform. Intersection targets build a visual protein interaction network (PPI) through the STRING database, and the core targets were identified by network topology analysis. GO analysis and KEGG pathway enrichment analysis of the intersecting targets were performed using the DAVID database. Finally, the molecular docking of the first four active ingredients and the first four core target genes were verified by Pubchem, the PDB database and CB-Dock online molecular docking technology. Results: ZBDHP screened 91 potential active ingredients and 816 potential targets. Among them, 141 genes were intersected by ZBDHP and SLE. The network topology analysis showed that the main active ingredients were Hydroxygenkwanin, Alisol B, asperglaucide, Cerevisterol, etc., and the key target genes were TNF, AKT1, EGFR, STAT3, etc. GO and KEGG enrichment analysis showed that common targets interfere with biological processes or molecular functions such as signal transduction protein phosphorylation, inflammatory response, transmembrane receptor protein tyrosine kinase activity, etc., through multiple signaling pathways, such as pathways in cancer, Kaposi sarcoma-associated herpesvirus infection, the PI3K-Akt signaling pathway, lipid and atherosclerosis, hepatitis B, etc. Molecular docking results showed that the active components of ZBDHP have good binding activity to the core targets of SLE. Conclusions: This study reveals that the ZBDHP treatment of SLE is a complex mechanistic process with multi-components, multi-targets and multi-pathways, and it may play a therapeutic role in SLE by inhibiting the production, proliferation and apoptosis of inflammatory factors. In conclusion, the present study provides a theoretical basis for further research on ZBDHP.
Collapse
|
10
|
Zeng H, Chen S, Lu X, Yan Z. Investigating the molecular mechanism of iguratimod act on SLE using network pharmacology and molecular docking analysis. FRONTIERS IN BIOINFORMATICS 2022; 2:932114. [PMID: 36304300 PMCID: PMC9580962 DOI: 10.3389/fbinf.2022.932114] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/04/2022] [Indexed: 11/21/2022] Open
Abstract
Objective: Iguratimod (IGU) is a novel small disease-modifying compound widely used in Asia for the treatment of rheumatic diseases. IGU is a methane sulfonanilide. We applied network pharmacology to investigate the pharmacological mechanisms of IGU act on SLE. Methods: We used PharmMapper, UniProt, and OMIM databases to screen the potential targets of IGU, and the SLE-related disease targets were predicted. Hub target genes among the intersections of the potential targets (IGU) and related genes (SLE) were validated using the PPI network generated by the String database. GO and KEGG enrichment analyses were carried out using the David online platform. Finally, the molecular docking of hub targets and their corresponding compounds were completed through AutoDock Vina and PyMOL software for visualization. Result: A total of 292 potential targets of IGU, 6501 related disease targets of SLE, and 114 cross targets were screened from the aforementioned database. Network topology analysis identified 10 hub targets, such as CASP3, AKT1, EGFR, MMP9, and IGF1. GO enrichment analysis mainly focuses on the negative regulation of the apoptotic process and signal transduction. KEGG enrichment analysis illustrated that the PI3K-AKT signaling pathway, MAPK signaling pathway, and FoxO signaling pathway might play a significant role in the pharmacological mechanisms of IGU act on SLE. Molecular docking confirmed that the IGU ligand had strong binding activity to the hub targets. Conclusion: This study based on network pharmacology and molecular docking validation preliminarily revealed the protein targets affected by IGU acting on SLE through, and explored potential therapeutic mechanism role of IGU in SLE treatment by multi pathways.
Collapse
Affiliation(s)
- Huiqiong Zeng
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- *Correspondence: Huiqiong Zeng,
| | - Shuai Chen
- Department of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Xiaoping Lu
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Zhenbo Yan
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| |
Collapse
|
11
|
Tabouni M, Ali A, Aljaberi N, Alblooshi H. Lupus nephritis: A focus on the United Arab Emirates and the potential role of genetics. Lupus 2022; 31:1415-1422. [PMID: 36017600 DOI: 10.1177/09612033221122982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lupus nephritis (LN) is a severe manifestation of systemic lupus erythematosus (SLE), characterized by chronic and progressive inflammation of the kidneys. As with many other autoimmune diseases, LN is a multifactorial disease caused by genetic and environmental factors. Globally, LN can affect around 60% of SLE patients, and it was observed to be less frequent and severe in Caucasian patients compared to other ethnic groups, including Arabs. Data on LN in the United Arab Emirates (UAE) are scattered and scarce in literature. Nevertheless, LN is common, occurring in around 43%-55% of SLE patients in the UAE. Anecdotally, the demographics and clinical features of SLE in the UAE have been distinct. However, the paucity of supporting literature makes it difficult to draw meaningful conclusions. Over the past two decades, there have been improvements in understanding the pathogenesis of LN; however, many cellular and molecular mechanisms which are implicated in the disease development and progression remain ambiguous. Investigating the clinical, pathological, and genetic characteristics of LN in different cohorts of patients is of importance for a better understanding of its pathogenesis, and thus improving its outcome. As a result, we acknowledge the need for large-scale epidemiological, clinical, and genetic investigation of LN cohorts in the UAE and surrounding regions.
Collapse
Affiliation(s)
- Mohammed Tabouni
- Department of Genetics and Genomics, College of Medicine and Health Sciences, 62776United Arab Emirates University, Al Ain, United Arab Emirates
| | - Amanat Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, 62776United Arab Emirates University, Al Ain, United Arab Emirates
| | - Najla Aljaberi
- Department of Pediatrics, College of Medicine and Health Sciences, 62776United Arab Emirates University, Al Ain, United Arab Emirates
| | - Hiba Alblooshi
- Department of Genetics and Genomics, College of Medicine and Health Sciences, 62776United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
12
|
Mahtal N, Lenoir O, Tinel C, Anglicheau D, Tharaux PL. MicroRNAs in kidney injury and disease. Nat Rev Nephrol 2022; 18:643-662. [PMID: 35974169 DOI: 10.1038/s41581-022-00608-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2022] [Indexed: 11/09/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression by degrading or repressing the translation of their target messenger RNAs. As miRNAs are critical regulators of cellular homeostasis, their dysregulation is a crucial component of cell and organ injury. A substantial body of evidence indicates that miRNAs are involved in the pathophysiology of acute kidney injury (AKI), chronic kidney disease and allograft damage. Different subsets of miRNAs are dysregulated during AKI, chronic kidney disease and allograft rejection, which could reflect differences in the physiopathology of these conditions. miRNAs that have been investigated in AKI include miR-21, which has an anti-apoptotic role, and miR-214 and miR-668, which regulate mitochondrial dynamics. Various miRNAs are downregulated in diabetic kidney disease, including the miR-30 family and miR-146a, which protect against inflammation and fibrosis. Other miRNAs such as miR-193 and miR-92a induce podocyte dedifferentiation in glomerulonephritis. In transplantation, miRNAs have been implicated in allograft rejection and injury. Further work is needed to identify and validate miRNAs as biomarkers of graft function and of kidney disease development and progression. Use of combinations of miRNAs together with other molecular markers could potentially improve diagnostic or predictive power and facilitate clinical translation. In addition, targeting specific miRNAs at different stages of disease could be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Nassim Mahtal
- Paris Cardiovascular Research Center - PARCC, Inserm, Université Paris Cité, Paris, France
| | - Olivia Lenoir
- Paris Cardiovascular Research Center - PARCC, Inserm, Université Paris Cité, Paris, France.
| | - Claire Tinel
- Service de Néphrologie et Transplantation Adulte, Hôpital Necker-Enfants Malades, Université Paris Cité, Assistance Publique-Hôpitaux de Paris, Paris, France.,Institut Necker-Enfants Malades, Inserm, Université Paris Cité, Paris, France
| | - Dany Anglicheau
- Service de Néphrologie et Transplantation Adulte, Hôpital Necker-Enfants Malades, Université Paris Cité, Assistance Publique-Hôpitaux de Paris, Paris, France.,Institut Necker-Enfants Malades, Inserm, Université Paris Cité, Paris, France
| | - Pierre-Louis Tharaux
- Paris Cardiovascular Research Center - PARCC, Inserm, Université Paris Cité, Paris, France.
| |
Collapse
|
13
|
Costa-Reis P, Maurer K, Petri MA, Levy Erez D, Zhao X, Faig W, Burnham J, O'Neil K, Klein-Gitelman MS, von Scheven E, Schanberg LE, Sullivan KE. Urinary HER2, TWEAK and VCAM-1 levels are associated with new-onset proteinuria in paediatric lupus nephritis. Lupus Sci Med 2022; 9:9/1/e000719. [PMID: 35918102 PMCID: PMC9351344 DOI: 10.1136/lupus-2022-000719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/27/2022] [Indexed: 01/13/2023]
Abstract
Objective Lupus nephritis is a key driver of morbidity and mortality in SLE. Detecting active nephritis on a background of pre-existing renal damage is difficult, leading to potential undertreatment and accumulating injury. An unmet need is a biomarker that distinguishes active lupus nephritis, particularly important in paediatrics where minimising invasive procedures is desirable. Methods This was a multicentre, prospective study of 113 paediatric patients with biopsy-proven lupus nephritis. Clinical data and urine were obtained every 3–4 months and patients averaged 2 years on study with seven time points. Urine was analysed for human epidermal growth factor receptor 2 (HER2), tumour necrosis factor-like weak inducer of apoptosis and vascular cell adhesion molecule-1 (VCAM-1) by ELISA. We defined active disease as either a rise in serum creatinine ≥0.3 mg/dL from baseline or a rise in renal Systemic Lupus Erythematosus Disease Activity Index score from the previous visit. These markers were also studied in patients with acute kidney injury, juvenile idiopathic arthritis (JIA), amplified pain syndrome and healthy controls. Results The rate of active disease was 56% over an average of 2 years of follow-up. HER2 and VCAM-1 were significantly elevated at time points with active disease defined by increased serum creatinine compared with time points with inactive disease or patients who never flared. All three biomarkers were associated with new-onset proteinuria and VCAM-1 was elevated at time points preceding new-onset proteinuria. These biomarkers were not increased in acute kidney injury or JIA. Conclusion All three biomarkers were associated with new onset proteinuria and increased VCAM-1 may predict impending proteinuria. These biomarkers provide potential non-invasive measures for monitoring that may be more sensitive to impending flare than conventional measures.
Collapse
Affiliation(s)
| | - Kelly Maurer
- Division of Allergy Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Michelle A Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniella Levy Erez
- Department of Nephrology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Xue Zhao
- Division of Allergy Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Walter Faig
- Biostatistics and Data Management Core, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jon Burnham
- Division of Rheumatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kathleen O'Neil
- Department of Rheumatology, Riley Hospital for Children, Indianapolis, Indiana, USA
| | - Marisa S Klein-Gitelman
- Department of Pediatrics, Ann and Robert H Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | | | - Laura Eve Schanberg
- Department of Pediatrics, Duke Children's Hospital and Health Center, Durham, North Carolina, USA
| | - Kathleen E Sullivan
- Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Barratt J, Pawluczyk I, Selvaskandan H. Clinical application of microRNAs in glomerular diseases. Nephrol Dial Transplant 2022; 38:1375-1384. [PMID: 35906877 DOI: 10.1093/ndt/gfac230] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
RNA interference (RNAi) occurs in all organisms and modulates most, if not all, biological pathways. It is the process by which non-coding RNAs, including microRNAs (miRs), regulate gene transcription and post-transcriptional processing of messenger RNA (mRNA). A single miR can modulate several genes within a cell, and several miRs can regulate expression of the same gene, adding tiers of complexity to regulation of gene expression. MicroRNAs and other RNAi approaches have been successfully used in vitro and in vivo to selectively manipulate gene transcription, making them pivotal agents for basic science research and candidates for targeted therapeutics. This review will focus on miRs and their potential as biomarkers and novel therapeutics for glomerular disease.
Collapse
Affiliation(s)
- Jonathan Barratt
- University of Leicester, Department of Cardiovascular Sciences, Leicester, UK
| | - Izabella Pawluczyk
- University of Leicester, Department of Cardiovascular Sciences, Leicester, UK
| | - Haresh Selvaskandan
- University of Leicester, Department of Cardiovascular Sciences, Leicester, UK
| |
Collapse
|
15
|
Mei X, Jin H, Zhao M, Lu Q. Association of Immune-Related Genetic and Epigenetic Alterations with Lupus Nephritis. KIDNEY DISEASES (BASEL, SWITZERLAND) 2022; 8:286-296. [PMID: 36157263 PMCID: PMC9386430 DOI: 10.1159/000524937] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 05/02/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The familial clustering phenomenon together with environmental influences indicates the presence of a genetic and epigenetic predisposition to systematic lupus erythematosus (SLE). Interestingly, regarding lupus nephritis (LN), the worst complication of SLE, mortality, and morbidity were not consistent with SLE in relation to sexuality and ethnicity. SUMMARY Genetic and epigenetic alterations in LN include genes and noncoding RNAs that are involved in antigen-presenting, complements, immune cell infiltration, interferon pathways, and so on. Once genetic or epigenetic change occurs alone or simultaneously, they will promote the formation of immune complexes with autoantibodies that target various autoantigens, which results in inflammatory cytokines and autoreactive immune cells colonizing renal tissues and contributing to LN. KEY MESSAGES Making additional checks for immunopathology-related heredity and epigenetic factors may lead to a more holistic perspective of LN.
Collapse
Affiliation(s)
- Xiaole Mei
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immunological Dermatology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
| | - Hui Jin
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
| | - Ming Zhao
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immunological Dermatology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
| |
Collapse
|
16
|
Lindblom J, Mohan C, Parodis I. Biomarkers in Neuropsychiatric Systemic Lupus Erythematosus: A Systematic Literature Review of the Last Decade. Brain Sci 2022; 12:192. [PMID: 35203955 PMCID: PMC8869794 DOI: 10.3390/brainsci12020192] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/23/2022] [Accepted: 01/26/2022] [Indexed: 11/17/2022] Open
Abstract
Nervous system involvement in patients with SLE, termed neuropsychiatric SLE (NPSLE), constitutes a diagnostic challenge, and its management is still poorly optimised. This review summarises recent insights over the past decade in laboratory biomarkers of diagnosis, monitoring, and prognosis of NPSLE. An initial systematic search in the Medline and Web of Science was conducted to guide the selection of articles. Emerging diagnostic biomarkers in NPSLE that displayed satisfactory ability to discriminate between NPSLE and controls include serum interleukin (IL)-6, microRNA (miR)-23a, miR-155, and cerebrospinal fluid (CSF) α-Klotho. CSF lipocalin-2, macrophage colony-stimulating factor (M-CSF), and immunoglobulin (Ig)M also displayed such ability in two ethnically diverse cohorts. Serum interferon (IFN)-α and neuron specific enolase (NSE) were recently reported to moderately correlate with disease activity in patients with active NPSLE. CSF IL-8, IL-13, and granulocyte colony-stimulating factor (G-CSF) exhibited excellent sensitivity, yet poorer specificity, as predictors of response to therapy in patients with NPSLE. The overall lack of validation studies across multiple and diverse cohorts necessitates further and well-concerted investigations. Nevertheless, we propound CSF lipocalin 2 among molecules that hold promise as reliable diagnostic biomarkers in NPSLE.
Collapse
Affiliation(s)
- Julius Lindblom
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institute and Karolinska University Hospital, 17176 Stockholm, Sweden;
| | - Chandra Mohan
- Department Biomedical Engineering, University of Houston, Houston, TX 77204, USA;
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institute and Karolinska University Hospital, 17176 Stockholm, Sweden;
- Department of Rheumatology, Faculty of Medicine and Health, Örebro University, 70182 Örebro, Sweden
| |
Collapse
|
17
|
Su CT, See DHW, Huang JW. Lipid-Based Nanocarriers in Renal RNA Therapy. Biomedicines 2022; 10:283. [PMID: 35203492 PMCID: PMC8869454 DOI: 10.3390/biomedicines10020283] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
Kidney disease is a multifactorial problem, with a growing prevalence and an increasing global burden. With the latest worldwide data suggesting that chronic kidney disease (CKD) is the 12th leading cause of death, it is no surprise that CKD remains a public health problem that requires urgent attention. Multiple factors contribute to kidney disease, each with its own pathophysiology and pathogenesis. Furthermore, microRNAs (miRNAs) have been linked to several types of kidney diseases. As dysregulation of miRNAs is often seen in some diseases, there is potential in the exploitation of this for therapeutic applications. In addition, uptake of interference RNA has been shown to be rapid in kidneys making them a good candidate for RNA therapy. The latest advancements in RNA therapy and lipid-based nanocarriers have enhanced the effectiveness and efficiency of RNA-related drugs, thereby making RNA therapy a viable treatment option for renal disease. This is especially useful for renal diseases, for which a suitable treatment is not yet available. Moreover, the high adaptability of RNA therapy combined with the low risk of lipid-based nanocarriers make for an attractive treatment choice. Currently, there are only a small number of RNA-based drugs related to renal parenchymal disease, most of which are in different stages of clinical trials. We propose the use of miRNAs or short interfering RNAs coupled with a lipid-based nanocarrier as a delivery vehicle for managing renal disease.
Collapse
Affiliation(s)
- Chi-Ting Su
- Department of Medicine, National Taiwan University Cancer Centre, Taipei 10672, Taiwan; (C.-T.S.); (D.H.W.S.)
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Douliu 640, Taiwan
| | - Daniel H. W. See
- Department of Medicine, National Taiwan University Cancer Centre, Taipei 10672, Taiwan; (C.-T.S.); (D.H.W.S.)
| | - Jenq-Wen Huang
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Douliu 640, Taiwan
| |
Collapse
|
18
|
Renal Tissue miRNA Expression Profiles in ANCA-Associated Vasculitis—A Comparative Analysis. Int J Mol Sci 2021; 23:ijms23010105. [PMID: 35008531 PMCID: PMC8745125 DOI: 10.3390/ijms23010105] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/14/2021] [Accepted: 12/20/2021] [Indexed: 11/17/2022] Open
Abstract
Anti-neutrophil cytoplasm antibody (ANCA)-associated vasculitis (AAV) comprises autoimmune disease entities that cause target organ damage due to relapsing-remitting small vessel necrotizing vasculitis, and which affects various vascular beds. The pathogenesis of AAV is incompletely understood, which translates to considerable disease- and treatment-related morbidity and mortality. Recent advances have implicated microRNAs (miRNAs) in AAV; however, their accurate characterization in renal tissue is lacking. The goal of this study was to identify the intrarenal miRNA expression profile in AAV relative to healthy, non-inflammatory and inflammatory controls to identify candidate-specific miRNAs. Formalin-fixed, paraffin-embedded renal biopsy tissue samples from 85 patients were obtained. Comprehensive miRNA expression profiles were performed using panels with 752 miRNAs and revealed 17 miRNA that differentiated AAV from both controls. Identified miRNAs were annotated to characterize their involvement in pathways and to define their targets. A considerable subset of differentially expressed miRNAs was related to macrophage and lymphocyte polarization and cytokines previously deemed important in AAV pathogenesis, lending credence to the obtained results. Interestingly, several members of the miR-30 family were detected. However, a validation study of these differentially expressed miRNAs in an independent, larger sample cohort is needed to establish their potential diagnostic utility.
Collapse
|
19
|
Fava A, Rao DA. Cellular and molecular heterogeneity in systemic lupus erythematosus. Semin Immunol 2021; 58:101653. [PMID: 36184357 DOI: 10.1016/j.smim.2022.101653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/15/2021] [Accepted: 11/15/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Andrea Fava
- Division of Rheumatology, Johns Hopkins University, Baltimore, MD, USA.
| | - Deepak A Rao
- Division of Rheumatology, Inflammation, Immunity, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
So BYF, Yap DYH, Chan TM. MicroRNAs in Lupus Nephritis-Role in Disease Pathogenesis and Clinical Applications. Int J Mol Sci 2021; 22:10737. [PMID: 34639078 PMCID: PMC8509214 DOI: 10.3390/ijms221910737] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRs) are non-coding small RNAs that act as epigenetic modulators to regulate the protein levels of target mRNAs without modifying the genetic sequences. The role of miRs in the pathogenesis of lupus nephritis (LN) is increasingly recognized and highly complex. Altered levels of different miRs are observed in the blood, urine and kidney tissues of murine LN models and LN patients. Accumulating evidence suggests that these miRs can modulate immune cells and various key inflammatory pathways, and their perturbations contribute to the aberrant immune response in LN. The dysregulation of miRs in different resident renal cells and urinary exosomes can also lead to abnormal renal cell proliferation, inflammation and kidney fibrosis in LN. While miRs may hold promise in various clinical applications in LN patients, there are still many potential limitations and safety concerns for their use. Further studies are worthwhile to examine the clinical utility of miRs in the diagnosis, disease activity monitoring, prognostication and treatment of LN.
Collapse
Affiliation(s)
| | - Desmond Y. H. Yap
- Division of Nephrology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong; (B.Y.F.S.); (T.M.C.)
| | | |
Collapse
|
21
|
Panfoli I, Granata S, Candiano G, Verlato A, Lombardi G, Bruschi M, Zaza G. Analysis of urinary exosomes applications for rare kidney disorders. Expert Rev Proteomics 2021; 17:735-749. [PMID: 33395324 DOI: 10.1080/14789450.2020.1866993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Exosomes are nanovesicles that play important functions in a variety of physiological and pathological conditions. They are powerful cell-to-cell communication tool thanks to the protein, mRNA, miRNA, and lipid cargoes they carry. They are also emerging as valuable diagnostic and prognostic biomarker sources. Urinary exosomes carry information from all the cells of the urinary tract, downstream of the podocyte. Rare kidney diseases are a subset of an inherited diseases whose genetic diagnosis can be unclear, and presentation can vary due to genetic, epigenetic, and environmental factors. Areas covered: In this review, we focus on a group of rare and often neglected kidney diseases, for which we have sufficient available literature data on urinary exosomes. The analysis of their content can help to comprehend pathological mechanisms and to identify biomarkers for diagnosis, prognosis, and therapeutic targets. Expert opinion: The foreseeable large-scale application of system biology approach to the profiling of exosomal proteins as a source of renal disease biomarkers will be also useful to stratify patients with rare kidney diseases whose penetrance, phenotypic presentation, and age of onset vary sensibly. This can ameliorate the clinical management.
Collapse
Affiliation(s)
- Isabella Panfoli
- Department of Pharmacy-DIFAR, University of Genoa , Genoa, Italy
| | - Simona Granata
- Renal Unit, Department of Medicine, University-Hospital of Verona , Verona, Italy
| | - Giovanni Candiano
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini , Genoa, Italy
| | - Alberto Verlato
- Renal Unit, Department of Medicine, University-Hospital of Verona , Verona, Italy
| | - Gianmarco Lombardi
- Renal Unit, Department of Medicine, University-Hospital of Verona , Verona, Italy
| | - Maurizio Bruschi
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini , Genoa, Italy
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University-Hospital of Verona , Verona, Italy
| |
Collapse
|
22
|
Tsai CY, Shen CY, Liu CW, Hsieh SC, Liao HT, Li KJ, Lu CS, Lee HT, Lin CS, Wu CH, Kuo YM, Yu CL. Aberrant Non-Coding RNA Expression in Patients with Systemic Lupus Erythematosus: Consequences for Immune Dysfunctions and Tissue Damage. Biomolecules 2020; 10:biom10121641. [PMID: 33291347 PMCID: PMC7762297 DOI: 10.3390/biom10121641] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex systemic autoimmune disease with heterogeneous clinical manifestations. A diverse innate and adaptive immune dysregulation is involved in the immunopathogenesis of SLE. The dysregulation of immune-related cells may derive from the intricate interactions among genetic, epigenetic, environmental, and immunological factors. Of these contributing factors, non-coding RNAs (ncRNAs), including microRNAs (miRNAs, miRs), and long non-coding RNAs (lncRNAs) play critical roles in the post-transcriptional mRNA expression of cytokines, chemokines, and growth factors, which are essential for immune modulation. In the present review, we emphasize the roles of ncRNA expression in the immune-related cells and cell-free plasma, urine, and tissues contributing to the immunopathogenesis and tissue damage in SLE. In addition, the circular RNAs (circRNA) and their post-translational regulation of protein synthesis in SLE are also briefly described. We wish these critical reviews would be useful in the search for biomarkers/biosignatures and novel therapeutic strategies for SLE patients in the future.
Collapse
MESH Headings
- Adaptive Immunity/genetics
- Autoimmunity/genetics
- Chemokines/genetics
- Chemokines/immunology
- Dendritic Cells/immunology
- Dendritic Cells/pathology
- Gene Expression Regulation
- Humans
- Immunity, Innate/genetics
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/immunology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Lupus Erythematosus, Systemic/blood
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/pathology
- MicroRNAs/genetics
- MicroRNAs/immunology
- Neutrophils/immunology
- Neutrophils/pathology
- RNA, Circular/genetics
- RNA, Circular/immunology
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/immunology
- RNA, Messenger/genetics
- RNA, Messenger/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
Collapse
Affiliation(s)
- Chang-Youh Tsai
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei 11217, Taiwan; (C.-W.L.); (H.-T.L.)
- Correspondence: (C.-Y.T.); (C.-L.Y.)
| | - Chieh-Yu Shen
- Division of Rheumatology, Immunology, & Allergy, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (S.-C.H.); (K.-J.L.); (C.-S.L.); (C.-H.W.); (Y.-M.K.)
- Institute of Clinical Medicine, National Taiwan University School of Medicine, Taipei 10002, Taiwan
| | - Chih-Wei Liu
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei 11217, Taiwan; (C.-W.L.); (H.-T.L.)
| | - Song-Chou Hsieh
- Division of Rheumatology, Immunology, & Allergy, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (S.-C.H.); (K.-J.L.); (C.-S.L.); (C.-H.W.); (Y.-M.K.)
| | - Hsien-Tzung Liao
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei 11217, Taiwan; (C.-W.L.); (H.-T.L.)
| | - Ko-Jen Li
- Division of Rheumatology, Immunology, & Allergy, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (S.-C.H.); (K.-J.L.); (C.-S.L.); (C.-H.W.); (Y.-M.K.)
| | - Cheng-Shiun Lu
- Division of Rheumatology, Immunology, & Allergy, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (S.-C.H.); (K.-J.L.); (C.-S.L.); (C.-H.W.); (Y.-M.K.)
| | - Hui-Ting Lee
- Mackay Memorial Hospital and Mackay College of Medicine, Taipei 10449, Taiwan;
| | - Cheng-Sung Lin
- Department of Thoracic Surgery, Ministry of Health and Welfare Taipei Hospital, New Taipei City 24213, Taiwan;
| | - Cheng-Han Wu
- Division of Rheumatology, Immunology, & Allergy, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (S.-C.H.); (K.-J.L.); (C.-S.L.); (C.-H.W.); (Y.-M.K.)
| | - Yu-Min Kuo
- Division of Rheumatology, Immunology, & Allergy, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (S.-C.H.); (K.-J.L.); (C.-S.L.); (C.-H.W.); (Y.-M.K.)
| | - Chia-Li Yu
- Division of Rheumatology, Immunology, & Allergy, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (C.-Y.S.); (S.-C.H.); (K.-J.L.); (C.-S.L.); (C.-H.W.); (Y.-M.K.)
- Correspondence: (C.-Y.T.); (C.-L.Y.)
| |
Collapse
|
23
|
Mir-30b-5p Promotes Proliferation, Migration, and Invasion of Breast Cancer Cells via Targeting ASPP2. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7907269. [PMID: 32420372 PMCID: PMC7210518 DOI: 10.1155/2020/7907269] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 01/06/2020] [Accepted: 01/16/2020] [Indexed: 12/18/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtypes of breast cancer, which has few effective targeted therapies. Various sources of evidence confirm that microRNAs (miRNAs) contribute to the progression and metastasis of human breast cancer. However, the molecular mechanisms underlying the changes in miRNAs expression and the regulation of miRNAs functions have not been well clarified. In this study, we found that the expression of miR-30b-5p was upregulated in breast cancer tissues and breast cancer cell lines, compared to paracancer tissues and normal breast cell lines. Moreover, induced overexpression of miR-30b-5p promoted the MDA-MB-231 and HCC 1937 cell growth, migration, and invasion and reduced the cellular apoptosis. Further studies confirmed that miR-30b-5p could directly target ASPP2 and then activate the AKT signaling pathway. Our results suggested that miR-30b-5p could act as a tumor promoter in TNBC. The newly identified miR-30b-5p/ASPP2/AKT axis represents a novel therapeutic strategy for treating TNBC.
Collapse
|
24
|
Lopez-Pedrera C, Barbarroja N, Patiño-Trives AM, Luque-Tévar M, Torres-Granados C, Aguirre-Zamorano MA, Collantes-Estevez E, Pérez-Sánchez C. Role of microRNAs in the Development of Cardiovascular Disease in Systemic Autoimmune Disorders. Int J Mol Sci 2020; 21:E2012. [PMID: 32188016 PMCID: PMC7139533 DOI: 10.3390/ijms21062012] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/11/2022] Open
Abstract
Rheumatoid Arthritis (RA), Systemic lupus erythematosus (SLE) and antiphospholipid syndrome (APS) are the systemic autoimmune diseases (SADs) most associated with an increased risk of developing cardiovascular (CV) events. Cardiovascular disease (CVD) in SADs results from a complex interaction between traditional CV-risk factors, immune deregulation and disease activity. Oxidative stress, dyslipidemia, endothelial dysfunction, inflammatory/prothrombotic mediators (cytokines/chemokines, adipokines, proteases, adhesion-receptors, NETosis-derived-products, and intracellular-signaling molecules) have been implicated in these vascular pathologies. Genetic and genomic analyses further allowed the identification of signatures explaining the pro-atherothrombotic profiles in RA, SLE and APS. However, gene modulation has left significant gaps in our understanding of CV co-morbidities in SADs. MicroRNAs (miRNAs) are emerging as key post-transcriptional regulators of a suite of signaling pathways and pathophysiological effects. Abnormalities in high number of miRNA and their associated functions have been described in several SADs, suggesting their involvement in the development of atherosclerosis and thrombosis in the setting of RA, SLE and APS. This review focusses on recent insights into the potential role of miRNAs both, as clinical biomarkers of atherosclerosis and thrombosis in SADs, and as therapeutic targets in the regulation of the most influential processes that govern those disorders, highlighting the potential diagnostic and therapeutic properties of miRNAs in the management of CVD.
Collapse
|
25
|
Erez DL, Denburg MR, Afolayan S, Jodele S, Wallace G, Davies SM, Seif AE, Bunin N, Laskin BL, Sullivan KE. Acute Kidney Injury in Children after Hematopoietic Cell Transplantation Is Associated with Elevated Urine CXCL10 and CXCL9. Biol Blood Marrow Transplant 2020; 26:1266-1272. [PMID: 32165324 DOI: 10.1016/j.bbmt.2020.02.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/23/2020] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
Acute kidney injury (AKI) is nearly universally associated with worse outcomes, especially among children after hematopoietic stem cell transplant (HCT). Our objective was to examine urinary immune biomarkers of AKI after HCT to provide insights into novel mechanisms of kidney injury in this population. Studying patients undergoing allogeneic HCT provides a unique opportunity to examine immune markers of AKI because the risk of AKI is high and the immune system newly develops after transplant. Children (>2 years old) and young adults undergoing their first allogeneic HCT and enrolled in a prospective, observational cohort study at 2 large children's hospitals had urine collected pre-HCT and monthly for the first 4 months after HCT. Urine samples at each monthly time point were assayed for 8 immune-related biomarkers. AKI was defined as a 1.5-fold increase in the monthly serum creatinine value, which was recorded ±1 day from when the research urine sample was obtained, as compared with the pre-HCT baseline. Generalized estimating equation regression analysis evaluated the association between the monthly repeated measures (urinary biomarkers and AKI). A total of 176 patients were included from 2 pediatric centers. Thirty-six patients from 1 center were analyzed as a discovery cohort and the remaining 140 patients from the second center were analyzed as a validation cohort. AKI rates were 18% to 35% depending on the monthly time point after HCT. Urine CXCL10 and CXCL9 concentrations were significantly higher among children who developed AKI compared with children who did not (P < .01) in both cohorts. In order to gain a better understanding of the cellular source for these biomarkers in the urine, we also analyzed in vitro expression of CXCL10 and CXCL9 in kidney cell lines after stimulation with interferon-γ and interferon-α. HEK293-epithelial kidney cells demonstrated interferon-induced expression of CXCL10 and CXCL9, suggesting a potential mechanism driving the key finding. CXCL10 and CXCL9 are associated with AKI after HCT and are therefore promising biomarkers to guide improved diagnostic and treatment strategies for AKI in this high-risk population.
Collapse
Affiliation(s)
- Daniella Levy Erez
- Division of Nephrology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Michelle R Denburg
- Division of Nephrology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Simisola Afolayan
- Division of Nephrology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sonata Jodele
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Gregory Wallace
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stella M Davies
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Alix E Seif
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Nancy Bunin
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Benjamin L Laskin
- Division of Nephrology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kathleen E Sullivan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
26
|
Zhang L, Wu H, Zhao M, Lu Q. Identifying the differentially expressed microRNAs in autoimmunity: A systemic review and meta-analysis. Autoimmunity 2020; 53:122-136. [DOI: 10.1080/08916934.2019.1710135] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Lian Zhang
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Central South University, Changsha, China
| | - Haijing Wu
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Central South University, Changsha, China
| | - Ming Zhao
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Central South University, Changsha, China
| | - Qianjin Lu
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Central South University, Changsha, China
| |
Collapse
|
27
|
De Angelis MT, Santamaria G, Parrotta EI, Scalise S, Lo Conte M, Gasparini S, Ferlazzo E, Aguglia U, Ciampi C, Sgura A, Cuda G. Establishment and characterization of induced pluripotent stem cells (iPSCs) from central nervous system lupus erythematosus. J Cell Mol Med 2019; 23:7382-7394. [PMID: 31536674 PMCID: PMC6815917 DOI: 10.1111/jcmm.14598] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/02/2019] [Accepted: 08/06/2019] [Indexed: 12/16/2022] Open
Abstract
Involvement of the central nervous system (CNS) is an uncommon feature in systemic lupus erythematosus (SLE), making diagnosis rather difficult and challenging due to the poor specificity of neuropathic symptoms and neurological symptoms. In this work, we used human‐induced pluripotent stem cells (hiPSCs) derived from CNS‐SLE patient, with the aim to dissect the molecular insights underlying the disease by gene expression analysis and modulation of implicated pathways. CNS‐SLE‐derived hiPSCs allowed us to provide evidence of Erk and Akt pathways involvement and to identify a novel cohort of potential biomarkers, namely CHCHD2, IDO1, S100A10, EPHA4 and LEFTY1, never reported so far. We further extended the study analysing a panel of oxidative stress‐related miRNAs and demonstrated, under normal or stress conditions, a strong dysregulation of several miRNAs in CNS‐SLE‐derived compared to control hiPSCs. In conclusion, we provide evidence that iPSCs reprogrammed from CNS‐SLE patient are a powerful useful tool to investigate the molecular mechanisms underlying the disease and to eventually develop innovative therapeutic approaches.
Collapse
Affiliation(s)
- Maria Teresa De Angelis
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Gianluca Santamaria
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Elvira Immacolata Parrotta
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Stefania Scalise
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Michela Lo Conte
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Sara Gasparini
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Edoardo Ferlazzo
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy.,Regional Epilepsy Centre, Great Metropolitan Hospital, Reggio Calabria, Italy
| | - Umberto Aguglia
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy.,Regional Epilepsy Centre, Great Metropolitan Hospital, Reggio Calabria, Italy
| | - Clara Ciampi
- Department of Science, University of Rome " Roma Tre", Rome, Italy
| | - Antonella Sgura
- Department of Science, University of Rome " Roma Tre", Rome, Italy
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| |
Collapse
|
28
|
Li X, Pan X, Fu X, Yang Y, Chen J, Lin W. MicroRNA-26a: An Emerging Regulator of Renal Biology and Disease. Kidney Blood Press Res 2019; 44:287-297. [PMID: 31163420 DOI: 10.1159/000499646] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
MicroRNAs (miRNAs) are short, single-stranded, noncoding RNAs that modulate many key biological processes by simultaneously suppressing multiple target genes. Among them, miR-26a, a conserved miRNA among vertebrates, is highly expressed in various tissues. Accumulating evidence demonstrates that miR-26a plays pivotal roles in cellular differentiation, cell growth, apoptosis, and metastasis, thereby participating in the initiation and development of various human diseases, such as metabolic disease and cancer. More recently, miR-26a was found as a versatile regulator of renal biology and disease. miR-26a is intensively involved in the maintenance of podocyte homeostasis and the actin cytoskeleton. It is also able to modulate the homeostasis and function of mesangial cells. In addition, miR-26a affects the expansion of regulatory T cells in the context of ischemia-reperfusion injury and autoimmune diabetes and thus protects the renal system from immune attack. These available data strongly suggest that renal miR-26a possesses critical pathological functions and represents a potential target for renal disease therapies. This review summarizes current knowledge of miR-26a in renal biology and disease, laying the foundation for exploring its previously unknown functions and mechanisms in the renal system.
Collapse
Affiliation(s)
- Xiaoyan Li
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Pan
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianghui Fu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Yang
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianghua Chen
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weiqiang Lin
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, .,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China,
| |
Collapse
|
29
|
Role of MicroRNAs in Renal Parenchymal Diseases-A New Dimension. Int J Mol Sci 2018; 19:ijms19061797. [PMID: 29914215 PMCID: PMC6032378 DOI: 10.3390/ijms19061797] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 06/07/2018] [Accepted: 06/08/2018] [Indexed: 11/18/2022] Open
Abstract
Since their discovery in 1993, numerous microRNAs (miRNAs) have been identified in humans and other eukaryotic organisms, and their role as key regulators of gene expression is still being elucidated. It is now known that miRNAs not only play a central role in the processes that ensure normal development and physiology, but they are often dysregulated in various diseases. In this review, we present an overview of the role of miRNAs in normal renal development and physiology, in maladaptive renal repair after injury, and in the pathogenesis of renal parenchymal diseases. In addition, we describe methods used for their detection and their potential as therapeutic targets. Continued research on renal miRNAs will undoubtedly improve our understanding of diseases affecting the kidneys and may also lead to new therapeutic agents.
Collapse
|
30
|
Kim Y, Shim SC. Wolves Trapped in the NETs–The Pathogenesis of Lupus Nephritis. JOURNAL OF RHEUMATIC DISEASES 2018. [DOI: 10.4078/jrd.2018.25.2.81] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Young Kim
- Division of Internal Medicine, Daejeon Veterans Hospital, Daejeon, Korea
| | - Seung Cheol Shim
- Division of Rheumatology, Department of Internal Medicine, Daejeon Rheumatoid and Degenerative Arthritis Center, Chungnam National University Hospital, Chungnam National University College of Medicine, Daejeon, Korea
| |
Collapse
|
31
|
Tangtanatakul P, Thammasate B, Jacquet A, Reantragoon R, Pisitkun T, Avihingsanon Y, Leelahavanichkul A, Hirankarn N. Transcriptomic profiling in human mesangial cells using patient-derived lupus autoantibodies identified miR-10a as a potential regulator of IL8. Sci Rep 2017; 7:14517. [PMID: 29109423 PMCID: PMC5673966 DOI: 10.1038/s41598-017-15160-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 10/23/2017] [Indexed: 11/09/2022] Open
Abstract
Autoantibody-mediated inflammation directed at resident kidney cells mediates lupus nephritis (LN) pathogenesis. This study investigated the role of miRNA in human mesangial cells (HMCs) stimulated with auto anti-dsDNA immunoglobulin (Ig)G antibodies. HMCs were treated with antibodies purified from active LN patients or non-specific IgG controls in the presence of normal serum. Aberrant miRNA was screened using high throughput sequencing. Anti-dsDNA IgG up-regulated 103 miRNAs and down-regulated 30 miRNAs. The miRNAs regulated genes in the cell cycle, catabolic processes, regulation of transcription and apoptosis signalling. miR-10a was highly abundant in HMCs but was specifically downregulated upon anti-dsDNA IgG induction. Interestingly, the expression of miR-10a in kidney biopsies from class III and IV LN patients (n = 26) was downregulated compared with cadaveric donor kidneys (n = 6). Functional studies highlighted the downstream regulator of miR-10a in the chemokine signalling and cell proliferation or apoptosis pathways. Luciferase assay confirmed for the first time that IL8 was a direct target of miR-10a in HMCs. In conclusion, anti-dsDNA IgG Ab down-regulated miR-10a expression in HMCs resulting in the induction of various target genes involved in HMC proliferation and chemokine expression.
Collapse
Affiliation(s)
- Pattarin Tangtanatakul
- Medical Microbiology Interdisciplinary Program, Graduate School, Chulalongkorn University, Bangkok, 10330, Thailand.,Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Boonyakiat Thammasate
- Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Alain Jacquet
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Rangsima Reantragoon
- Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Trairak Pisitkun
- Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.,Chulalongkorn University Systems Biology (CUSB), Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Yingyos Avihingsanon
- Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.,Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Asada Leelahavanichkul
- Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Nattiya Hirankarn
- Center of Excellence in Immunology and Immune-mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
32
|
Wang H, Wang J, Xia Y. Defective Suppressor of Cytokine Signaling 1 Signaling Contributes to the Pathogenesis of Systemic Lupus Erythematosus. Front Immunol 2017; 8:1292. [PMID: 29085365 PMCID: PMC5650678 DOI: 10.3389/fimmu.2017.01292] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 09/26/2017] [Indexed: 12/19/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease involving injuries in multiple organs and systems. Exaggerated inflammatory responses are characterized as end-organ damage in patients with SLE. Although the explicit pathogenesis of SLE remains unclear, increasing evidence suggests that dysregulation of cytokine signals contributes to the progression of SLE through the Janus kinase/signal transducer and activator of transcription (STAT) signaling pathway. Activated STAT proteins translocate to the cell nucleus and induce transcription of target genes, which regulate downstream cytokine production and inflammatory cell infiltration. The suppressor of cytokine signaling 1 (SOCS1) is considered as a classical inhibitor of cytokine signaling. Recent studies have demonstrated that SOCS1 expression is decreased in patients with SLE and in murine lupus models, and this negatively correlates with the magnitude of inflammation. Dysregulation of SOCS1 signals participates in various pathological processes of SLE such as hematologic abnormalities and autoantibody generation. Lupus nephritis is one of the most serious complications of SLE, and it correlates with suppressed SOCS1 signals in renal tissues. Moreover, SOCS1 insufficiency affects the function of several other organs, including skin, central nervous system, liver, and lungs. Therefore, SOCS1 aberrancy contributes to the development of both systemic and local inflammation in SLE patients. In this review, we discuss recent studies regarding the roles of SOCS1 in the pathogenesis of SLE and its therapeutic implications.
Collapse
Affiliation(s)
- Huixia Wang
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jiaxing Wang
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
33
|
Ma TKW, McAdoo SP, Tam FWK. Targeting the tyrosine kinase signalling pathways for treatment of immune-mediated glomerulonephritis: from bench to bedside and beyond. Nephrol Dial Transplant 2017; 32:i129-i138. [PMID: 28391340 PMCID: PMC5410974 DOI: 10.1093/ndt/gfw336] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 08/18/2016] [Indexed: 12/25/2022] Open
Abstract
Glomerulonephritis (GN) affects patients of all ages and is an important cause of morbidity and mortality. Non-selective immunosuppressive drugs have been used in immune-mediated GN but often result in systemic side effects and occasionally fatal infective complications. There is increasing evidence from both preclinical and clinical studies that abnormal activation of receptor and non-receptor tyrosine kinase signalling pathways are implicated in the pathogenesis of immune-mediated GN. Activation of spleen tyrosine kinase (SYK), Bruton's tyrosine kinase (BTK), platelet-derived growth factor receptor (PDGFR), epidermal growth factor receptor (EGFR) and discoidin domain receptor 1 (DDR1) have been demonstrated in anti-GBM disease. SYK is implicated in the pathogenesis of ANCA-associated GN. SYK, BTK, PDGFR, EFGR, DDR1 and Janus kinase are implicated in the pathogenesis of lupus nephritis. A representative animal model of IgA nephropathy (IgAN) is lacking. Based on the results from in vitro and human renal biopsy study results, a phase II clinical trial is ongoing to evaluate the efficacy and safety of fostamatinib (an oral SYK inhibitor) in high-risk IgAN patient. Various tyrosine kinase inhibitors (TKIs) have been approved for cancer treatment. Clinical trials of TKIs in GN may be justified given their long-term safety data. In this review we will discuss the current unmet medical needs in GN treatment and research as well as the current stage of development of TKIs in GN treatment and propose an accelerated translational research approach to investigate whether selective inhibition of tyrosine kinase provides a safer and more efficacious option for GN treatment.
Collapse
Affiliation(s)
- Terry King-Wing Ma
- Renal and Vascular Inflammation Section, Department of Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK.,Carol and Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Stephen P McAdoo
- Renal and Vascular Inflammation Section, Department of Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK
| | - Frederick Wai Keung Tam
- Renal and Vascular Inflammation Section, Department of Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK
| |
Collapse
|
34
|
Wang WJ, Cai GY, Chen XM. Cellular senescence, senescence-associated secretory phenotype, and chronic kidney disease. Oncotarget 2017; 8:64520-64533. [PMID: 28969091 PMCID: PMC5610023 DOI: 10.18632/oncotarget.17327] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/24/2017] [Indexed: 12/19/2022] Open
Abstract
Chronic kidney disease (CKD) is increasingly being accepted as a type of renal ageing. The kidney undergoes age-related alterations in both structure and function. To date, a comprehensive analysis of cellular senescence and senescence-associated secretory phenotype (SASP) in CKD is lacking. Hence, this review mainly discusses the relationship between the two phenomena to show the striking similarities between SASP and CKD-associated secretory phenotype (CASP). It has been reported that replicative senescence, stress-induced premature ageing, and epigenetic abnormalities participate in the occurrence and development of CKD. Genomic damage and external environmental stimuli cause increased levels of oxidative stress and a chronic inflammatory state as a result of irreversible cell cycle arrest and low doses of SASP. Similar to SASP, CASP factors activate tissue repair by multiple mechanisms. Once tissue repair fails, the accumulated SASP or CASP species aggravate DNA damage response (DDR) and cause the senescent cells to secrete more SASP factors, accelerating the process of cellular ageing and eventually leading to various ageing-related changes. It is concluded that cellular senescence and SASP participate in the pathological process of CKD, and correspondingly CKD accelerated the progression of cell senescence and the secretion of SASP. These results will facilitate the integration of these mechanisms into the care and management of CKD and other age-related diseases.
Collapse
Affiliation(s)
- Wen-Juan Wang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing 100853, China
- Department of Nephrology, Beijing Changping Hospital, Beijing 102200, China
| | - Guang-Yan Cai
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing 100853, China
| | - Xiang-Mei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing 100853, China
| |
Collapse
|
35
|
Novel insights of microRNAs in the development of systemic lupus erythematosus. Curr Opin Rheumatol 2017; 29:450-457. [DOI: 10.1097/bor.0000000000000420] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
|
37
|
Wang Z, Chang C, Peng M, Lu Q. Translating epigenetics into clinic: focus on lupus. Clin Epigenetics 2017; 9:78. [PMID: 28785369 PMCID: PMC5541721 DOI: 10.1186/s13148-017-0378-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 07/26/2017] [Indexed: 01/17/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic relapsing–remitting autoimmune disease with highly heterogeneous phenotypes. Biomarkers with high sensitivity and specificity are useful for early diagnosis as well as monitoring disease activity and long-term complications. Epigenetics potentially provide novel biomarkers in autoimmune diseases. These may include DNA methylation changes in relevant lupus-prone genes or histone modifications and microRNAs to upregulate and downregulate relevant gene expression. The timing and nature of epigenetic modification provide such changes. In lupus, DNA methylation alterations in cytokine genes, such as IFN-related gene and retrovirus gene, have been found to offer biomarkers for lupus diagnosis. Histone modifications such as histone methylation and acetylation lead to transcriptional alterations of several genes such as PTPN22, LRP1B, and TNFSF70. There are varieties of microRNAs applied as lupus biomarkers, including DNMT1-related microRNAs, renal function-associated microRNAs, microRNAs involved in the immune system, and microRNAs for phenotype classification. Thus, we conclude a wide range of promising roles of epigenetic biomarkers aiding in the diagnosing and monitoring of lupus diseases and the risk of organ damage.
Collapse
Affiliation(s)
- Zijun Wang
- Department of Dermatology, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, Hunan 410011 China
| | - Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA USA
| | - Mou Peng
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Qianjin Lu
- Department of Dermatology, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, Hunan 410011 China
| |
Collapse
|
38
|
Tsokos GC, Lo MS, Costa Reis P, Sullivan KE. New insights into the immunopathogenesis of systemic lupus erythematosus. Nat Rev Rheumatol 2017; 12:716-730. [PMID: 27872476 DOI: 10.1038/nrrheum.2016.186] [Citation(s) in RCA: 783] [Impact Index Per Article: 111.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The aetiology of systemic lupus erythematosus (SLE) is multifactorial, and includes contributions from the environment, stochastic factors, and genetic susceptibility. Great gains have been made in understanding SLE through the use of genetic variant identification, mouse models, gene expression studies, and epigenetic analyses. Collectively, these studies support the concept that defective clearance of immune complexes and biological waste (such as apoptotic cells), neutrophil extracellular traps, nucleic acid sensing, lymphocyte signalling, and interferon production pathways are all central to loss of tolerance and tissue damage. Increased understanding of the pathogenesis of SLE is driving a renewed interest in targeted therapy, and researchers are now on the verge of developing targeted immunotherapy directed at treating either specific organ system involvement or specific subsets of patients with SLE. Accordingly, this Review places these insights within the context of our current understanding of the pathogenesis of SLE and highlights pathways that are ripe for therapeutic targeting.
Collapse
Affiliation(s)
- George C Tsokos
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, 110 Francis Street, Boston, Massachusetts 02215, USA
| | - Mindy S Lo
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
| | - Patricia Costa Reis
- Department of Pediatrics, Lisbon Medical School, Lisbon University, Santa Maria Hospital, Avenida Professor Egas Moniz, 1649-035 Lisbon, Portugal
| | - Kathleen E Sullivan
- Division of Allergy and Immunology, The Children's Hospital of Philadelphia, The University of Pennsylvania Perelman School of Medicine, 3615 Civic Center Boulevard, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
39
|
Trionfini P, Benigni A. MicroRNAs as Master Regulators of Glomerular Function in Health and Disease. J Am Soc Nephrol 2017; 28:1686-1696. [PMID: 28232619 DOI: 10.1681/asn.2016101117] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are important regulators of gene expression, and the dysregulation of miRNAs is a common feature of several diseases. More miRNAs are identified almost daily, revealing the complexity of these transcripts in eukaryotic cellular networks. The study of renal miRNAs, using genetically modified mice or by perturbing endogenous miRNA levels, has revealed the important biologic roles miRNAs have in the major cell lineages that compose the glomerulus. Here, we provide an overview of miRNA biogenesis and function in regulating key genes and cellular pathways in glomerular cells during development and homeostasis. Moreover, we focus on the emerging mechanisms through which miRNAs contribute to different diseases affecting the glomerulus, such as FSGS, IgA nephropathy, lupus nephritis, and diabetic nephropathy. In-depth knowledge of miRNA-based gene regulation has made it possible to unravel pathomechanisms, enabling the design of new therapeutic strategies for glomerular diseases for which available therapies are not fully efficacious.
Collapse
Affiliation(s)
- Piera Trionfini
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| | - Ariela Benigni
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| |
Collapse
|
40
|
Ichii O, Ohta H, Horino T, Nakamura T, Hosotani M, Mizoguchi T, Morishita K, Nakamura K, Hoshino Y, Takagi S, Sasaki N, Takiguchi M, Sato R, Oyamada K, Kon Y. Urinary exosome-derived microRNAs reflecting the changes of renal function and histopathology in dogs. Sci Rep 2017; 7:40340. [PMID: 28074869 PMCID: PMC5225487 DOI: 10.1038/srep40340] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/05/2016] [Indexed: 12/20/2022] Open
Abstract
MicroRNAs act as post-transcriptional regulators, and urinary exosome (UExo)-derived microRNAs may be used as biomarkers. Herein, we screened for UExo-derived microRNAs reflecting kidney disease (KD) status in dogs. Examined dogs were divided into healthy kidney control (HC) and KD groups according to renal dysfunction. We confirmed the appearance of UExo having irregular globe-shapes in a dog by immunoblot detection of the exosome markers, TSG101 and CD9. Based on our previous data using KD model mice and the data obtained herein by next generation sequencing of UExo-derived microRNAs in dogs, miR-26a, miR-146a, miR-486, miR-21a, and miR-10a/b were selected as candidate microRNAs. In particular, UExo-derived miR-26a and miR-10a/b were significantly decreased in KD dogs, and miR-26a levels negatively correlated with deteriorated renal function compared to the other miRNAs. UExo-derived miR-21a levels corrected or not to that of internal control microRNAs in UExo, miR-26a and miR-191, significantly increased with renal dysfunction. In kidney tissues, the decrease of miR-26a and miR-10a/b in the glomerulus and miR-10b in the tubulointerstitium negatively correlated with deteriorated renal function and histopathology. Increased miR-21a in the tubulointerstitium rather than in the glomerulus correlated with deteriorated renal histopathology. In conclusion, microRNAs reflecting the changes in renal function and histopathology in dogs were identified in this study.
Collapse
Affiliation(s)
- Osamu Ichii
- Laboratory of Anatomy, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Hiroshi Ohta
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Taro Horino
- Department of Endocrinology, Metabolism and Nephrology, Kochi University School of Medicine, Kohasu, Oko-cho, Nankoku 783-8505, Japan
| | - Teppei Nakamura
- Laboratory of Anatomy, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan.,Section of Biological Safety Research, Chitose Laboratory, Japan Food Research Laboratories, Bunkyo 2-3, Chitose 066-0052, Japan
| | - Marina Hosotani
- Laboratory of Anatomy, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Tatsuya Mizoguchi
- Laboratory of Anatomy, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Keitaro Morishita
- Veterinary Teaching Hospital, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Kensuke Nakamura
- Veterinary Teaching Hospital, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Yuki Hoshino
- Veterinary Teaching Hospital, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Satoshi Takagi
- Veterinary Teaching Hospital, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Noboru Sasaki
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Mitsuyoshi Takiguchi
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Ryo Sato
- Matsubara Animal Hospital, Taijo 1-174-1, Matsubara 580-0044, Japan
| | - Kazuhisa Oyamada
- Matsubara Animal Hospital, Taijo 1-174-1, Matsubara 580-0044, Japan
| | - Yasuhiro Kon
- Laboratory of Anatomy, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| |
Collapse
|
41
|
Yao F, Sun L, Fang W, Wang H, Yao D, Cui R, Xu J, Wang L, Wang X. Hsa‑miR‑371‑5p inhibits human mesangial cell proliferation and promotes apoptosis in lupus nephritis by directly targeting hypoxia‑inducible factor 1α. Mol Med Rep 2016; 14:5693-5698. [PMID: 27878241 DOI: 10.3892/mmr.2016.5939] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 09/16/2016] [Indexed: 11/06/2022] Open
Abstract
MicroRNAs (miRNAs/miR) have emerged as a novel class of gene expression modulators in kidney disease. Lupus nephritis (LN) is the predominant cause of morbidity and mortality in patients with systemic lupus erythematosus (SLE). Hsa‑miR‑371‑5p has previously been reported to be dysregulated in LN using a miRNA microarray analysis. The present study aimed to determine the function and molecular mechanisms of hsa‑miR‑371‑5p in human mesangial cells of LN. Quantitative polymerase chain reaction (qPCR) was used to detect hsa‑miR‑371‑5p expression in LN tissues. Furthermore, the MTT assay and flow cytometric analyses were performed to analyze the effects of hsa‑miR‑371‑5p on mesangial cell proliferation and apoptosis. Bioinformatics analysis, luciferase reporter assay, qPCR and western blotting were also conducted to predict and confirm the target gene of hsa‑miR‑371‑5p in mesangial cells. The results demonstrated that hsa‑miR‑371‑5p expression was markedly downregulated in LN renal tissues compared with in normal kidney tissues. Restoration of hsa‑miR‑371‑5p expression using synthetic hsa‑miR‑371‑5p mimics was able to significantly inhibit mesangial cell proliferation and induce apoptosis. In addition, mechanistic exploration demonstrated that hypoxia‑inducible factor 1α (HIF‑1α) was a direct target gene of hsa‑miR‑371‑5p in mesangial cells. In conclusion, these results suggested that hsa‑miR‑371‑5p is downregulated in LN, and overexpression of hsa‑miR‑371‑5p may inhibit mesangial cell proliferation and promote apoptosis by directly targeting HIF‑1α.
Collapse
Affiliation(s)
- Feifei Yao
- Department of Nephrology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Liqiu Sun
- Department of Nephrology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Wei Fang
- Department of Nephrology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Huamin Wang
- Department of Nephrology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Dongsheng Yao
- Department of Nephrology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Rui Cui
- Department of Nephrology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Jia Xu
- Department of Nephrology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Li Wang
- Department of Nephrology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xiumei Wang
- Department of Nephrology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
42
|
Grammer AC, Ryals MM, Heuer SE, Robl RD, Madamanchi S, Davis LS, Lauwerys B, Catalina MD, Lipsky PE. Drug repositioning in SLE: crowd-sourcing, literature-mining and Big Data analysis. Lupus 2016; 25:1150-70. [DOI: 10.1177/0961203316657437] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Lupus patients are in need of modern drugs to treat specific manifestations of their disease effectively and safely. In the past half century, only one new treatment has been approved by the US Food and Drug Administration (FDA) for systemic lupus erythematosus (SLE). In 2014–2015, the FDA approved 71 new drugs, only one of which targeted a rheumatic disease and none of which was approved for use in SLE. Repositioning/repurposing drugs approved for other diseases using multiple approaches is one possible means to find new treatment options for lupus patients. “Big Data” analysis approaches this challenge from an unbiased standpoint whereas literature mining and crowd sourcing for candidates assessed by the CoLTs (Combined Lupus Treatment Scoring) system provide a hypothesis-based approach to rank potential therapeutic candidates for possible clinical application. Both approaches mitigate risk since the candidates assessed have largely been extensively tested in clinical trials for other indications. The usefulness of a multi-pronged approach to drug repositioning in lupus is highlighted by orthogonal confirmation of hypothesis-based drug repositioning predictions by “Big Data” analysis of differentially expressed genes from lupus patient samples. The goal is to identify novel therapies that have the potential to affect disease processes specifically. Involvement of SLE patients and the scientists that study this disease in thinking about new drugs that may be effective in lupus though crowd-sourcing sites such as LRxL-STAT ( www.linkedin.com/in/lrxlstat ) is important in stimulating the momentum needed to test these novel drug targets for efficacy in lupus rapidly in small, proof-of-concept trials conducted by LuCIN, the Lupus Clinical Investigators Network ( www.linkedin.com/in/lucinstat ).
Collapse
Affiliation(s)
- A C Grammer
- AMPEL BioSolutions and RILITE Foundation, University of Virginia Research Park, Charlottesville, VA, USA
| | - M M Ryals
- AMPEL BioSolutions and RILITE Foundation, University of Virginia Research Park, Charlottesville, VA, USA
| | - S E Heuer
- AMPEL BioSolutions and RILITE Foundation, University of Virginia Research Park, Charlottesville, VA, USA
| | - R D Robl
- AMPEL BioSolutions and RILITE Foundation, University of Virginia Research Park, Charlottesville, VA, USA
| | - S Madamanchi
- AMPEL BioSolutions and RILITE Foundation, University of Virginia Research Park, Charlottesville, VA, USA
| | - L S Davis
- Department of Internal Medicine, UTSW Medical Center at Dallas, Dallas, TX, USA
| | - B Lauwerys
- Université Catholique de Louvain, Brussels, Belgium
| | - M D Catalina
- AMPEL BioSolutions and RILITE Foundation, University of Virginia Research Park, Charlottesville, VA, USA
| | - P E Lipsky
- AMPEL BioSolutions and RILITE Foundation, University of Virginia Research Park, Charlottesville, VA, USA
| |
Collapse
|
43
|
Naqvi AR, Zhong S, Dang H, Fordham JB, Nares S, Khan A. Expression Profiling of LPS Responsive miRNA in Primary Human Macrophages. ACTA ACUST UNITED AC 2016; 8:136-143. [PMID: 27307950 PMCID: PMC4905593 DOI: 10.4172/1948-5948.1000276] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
microRNAs (miRNAs) have emerged as important regulators of the innate and adaptive immune response. The purpose of the present study was to interrogate miRNA profiles of primary human macrophages challenged with bacterial lipopolysaccharide (LPS) with focus on expression kinetics. We employed Nanostring platform to precisely characterize the changes in miRNA expression following different doses and durations of LPS exposure. Differentially expressed miRNAs were identified in response to LPS challenge with convergent and divergent expression profiles. Pathway analysis of LPS-responsive miRNAs revealed regulation of biological processes linked to key cell signaling (including PIK3-Akt, MAP kinase, ErbB) and pathogen response pathways. Our data provide a comprehensive miRNA profiling of human primary macrophages treated with LPS. These results show that bacterial Toll like receptor (TLR) ligands can temporally modulate macrophage miRNA expression.
Collapse
Affiliation(s)
- Afsar Raza Naqvi
- Department of Periodontics, College of Dentistry, University of Illinois at Chicago, USA
| | - Sheng Zhong
- Department of Endodontics, Center for Pain Research and Innovation, School of Dentistry, University of North Carolina, USA
| | - Hong Dang
- Cystic Fibrosis/Pulmonary Research and Treatment Center, Marisco Lung Institute, University of North Carolina, USA
| | - Jezrom B Fordham
- Department of Periodontics, College of Dentistry, University of Illinois at Chicago, USA
| | - Salvador Nares
- Department of Periodontics, College of Dentistry, University of Illinois at Chicago, USA
| | - Asma Khan
- Department of Endodontics, Center for Pain Research and Innovation, School of Dentistry, University of North Carolina, USA
| |
Collapse
|