1
|
Akagbosu CO, McCauley KE, Namasivayam S, Romero-Soto HN, O’Brien W, Bacorn M, Bohrnsen E, Schwarz B, Mistry S, Burns AS, Perez-Chaparro PJ, Chen Q, LaPoint P, Patel A, Krausfeldt LE, Subramanian P, Sellers BA, Cheung F, Apps R, Douagi I, Levy S, Nadler EP, Hourigan SK. Gut microbiome shifts in adolescents after sleeve gastrectomy with increased oral-associated taxa and pro-inflammatory potential. Gut Microbes 2025; 17:2467833. [PMID: 39971742 PMCID: PMC11845021 DOI: 10.1080/19490976.2025.2467833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/30/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025] Open
Abstract
Bariatric surgery is highly effective in achieving weight loss in children and adolescents with severe obesity, however the underlying mechanisms are incompletely understood, and gut microbiome changes are unknown. Here, we show that adolescents exhibit significant gut microbiome and metabolome shifts several months after laparoscopic vertical sleeve gastrectomy (VSG), with increased alpha diversity and notably with enrichment of oral-associated taxa. To assess causality of the microbiome/metabolome changes in phenotype, pre-VSG and post-VSG stool was transplanted into germ-free mice. Post-VSG stool was not associated with any beneficial outcomes such as adiposity reduction compared pre-VSG stool. However, post-VSG stool exhibited a potentially inflammatory phenotype with increased intestinal Th17 and decreased regulatory T cells. Concomitantly, we found elevated fecal calprotectin and an enrichment of proinflammatory pathways in a subset of adolescents post-VSG. We show that in some adolescents, microbiome changes post-VSG may have inflammatory potential, which may be of importance considering the increased incidence of inflammatory bowel disease post-VSG.
Collapse
Affiliation(s)
- Cynthia O. Akagbosu
- Department of Gastroenterology, Weill Cornell Medicine, New York, New York, USA
| | - Kathryn E. McCauley
- Bioinformatics and Computational Biosciences Branch National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sivaranjani Namasivayam
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Hector N. Romero-Soto
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Wade O’Brien
- Dartmouth Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Mickayla Bacorn
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Eric Bohrnsen
- Research Technologies Branch, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, USA
| | - Benjamin Schwarz
- Research Technologies Branch, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, USA
| | - Shreni Mistry
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew S. Burns
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - P. Juliana Perez-Chaparro
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Qing Chen
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Phoebe LaPoint
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Anal Patel
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Lauren E. Krausfeldt
- Bioinformatics and Computational Biosciences Branch National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Poorani Subramanian
- Bioinformatics and Computational Biosciences Branch National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Brian A. Sellers
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, USA
| | - Foo Cheung
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, USA
| | - Richard Apps
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, USA
| | - Iyadh Douagi
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, USA
| | - Shira Levy
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Suchitra K. Hourigan
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
2
|
Chu D, Schwartz N, Ampudia J, Guthridge J, James J, Buyon JP, Connelly S, Fung M, Ng CT, Fava A, Petri M, Mohan C, Putterman C. Soluble urine activated leukocyte cell adhesion molecule is a strong predictor of lupus nephritis. Rheumatology (Oxford) 2025; 64:2676-2687. [PMID: 39404817 PMCID: PMC12048083 DOI: 10.1093/rheumatology/keae559] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/01/2024] [Accepted: 09/22/2024] [Indexed: 05/04/2025] Open
Abstract
OBJECTIVES To evaluate urinary activated leucocyte cell adhesion molecule (ALCAM) and CD6 as predictors of LN progression or disease resolution across a 1-year study. METHODS Serum and urine samples from biopsy proven LN subjects (n = 122) were prospectively collected over the course of a year at 3- or 6-month intervals (weeks 0, 12, 26 and 52) across multiple study sites and assessed for soluble ALCAM and CD6 levels. Urine creatinine from the same urine sample was used to normalize the levels of urinary ALCAM and urinary CD6. Measured levels of serum and urine ALCAM and CD6 were then analysed against disease metrics cross-sectionally and longitudinally. RESULTS Cross-sectional analysis at baseline revealed that urinary ALCAM significantly correlated with urine protein creatinine ratio, renal SLEDAI, and the Physician Global Assessment (PGA), and negatively correlated with serum C3 and C4. Receiver operating characteristic curve analysis demonstrated that urinary ALCAM is a predictor of LN with an area under the curve (AUC) of 0.97, compared with urinary CD6 with an AUC of 0.71. Importantly, the change in urinary ALCAM over a 3-month period distinguished between non-responders and responders at week 52. CONCLUSION Urinary ALCAM is reflective of changes in LN and may be predictive of response status.
Collapse
Affiliation(s)
| | - Noa Schwartz
- Department of Medicine, Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Joel Guthridge
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Judith James
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Jill P Buyon
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, NY, USA
| | | | | | | | - Andrea Fava
- Department of Medicine, Division of Rheumatology, John Hopkins Medicine, Baltimore, MD, USA
| | - Michelle Petri
- Department of Medicine, Division of Rheumatology, John Hopkins Medicine, Baltimore, MD, USA
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Chaim Putterman
- Department of Medicine, Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Galilee Research Institute, Nahariya, Israel
| |
Collapse
|
3
|
Hiramoto K, Saito S, Hanaoka H, Kikuchi J, Fukui H, Hashiguchi A, Suzuki K, Takeuchi T, Kaneko Y. Urinary Biomarkers Associated With Pathogenic Pathways Reflecting Histologic Findings in Lupus Nephritis. Arthritis Rheumatol 2025; 77:298-310. [PMID: 39317671 PMCID: PMC11865699 DOI: 10.1002/art.43017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 07/07/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
OBJECTIVE There is a pressing need to understand the pathogenesis of histologic findings and identify the biomarkers for predicting the histologic severity in lupus nephritis (LN). This study aimed to identify the pathogenic signal pathway and elucidate urinary biomarkers for predicting the presence or severity of histologic findings in LN. METHODS Urine samples from patients with biopsy-proven active LN were screened for 1,305 proteins using an aptamer-based proteomic assay. The diversity and expansion of individual renal histologic features in LN were quantified to identify the urinary proteins associated with the histologic findings found in each score. Candidate urinary proteins were validated in a validation cohort. Immunohistochemical staining of the renal tissues was performed to clarify the localization of the candidate proteins. RESULTS Cluster analysis extracted five histologic subgroups according to their correlations with each histologic finding in LN. Protein groups that correlated with each histologic subgroup revealed a distinct pathogenesis in LN using pathway analyses. Enzyme-linked immunosorbent assay validation revealed that urinary calgranulin B (S100A9), monocyte chemotactic protein 1 (MCP-1), and insulin-like growth factor binding protein 5 (IGFBP-5) levels could specifically predict the presence and severity of active glomerular lesions, interstitial inflammation, and interstitial fibrosis, respectively. Immunohistochemical staining revealed the localization of these proteins in each lesion. CONCLUSION Renal histologic findings may reflect the different pathogeneses involved in each lesion, and estimating the urinary calgranulin B, MCP-1, and IGFBP-5 levels may be useful in predicting the presence and severity of histologic findings in LN.
Collapse
Affiliation(s)
| | | | | | - Jun Kikuchi
- Keio University School of MedicineTokyoJapan
| | | | | | | | - Tsutomu Takeuchi
- Keio University School of Medicine, Tokyo, Japan and Saitama Medical UniversitySaitamaJapan
| | - Yuko Kaneko
- Keio University School of MedicineTokyoJapan
| |
Collapse
|
4
|
Umeda M, Karino K, Satyam A, Yoshida N, Hisada R, Bhargava R, Vichos T, Kunzler AL, Igawa T, Ichinose K, Torigoe K, Nishino T, Maeda T, Owen CA, Abdi R, Kawakami A, Tsokos GC. Hypoxia Promotes the Expression of ADAM9 by Tubular Epithelial Cells, Which Enhances Transforming Growth Factor β1 Activation and Promotes Tissue Fibrosis in Patients With Lupus Nephritis. Arthritis Rheumatol 2025; 77:180-189. [PMID: 39279154 PMCID: PMC11785507 DOI: 10.1002/art.42987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 08/07/2024] [Accepted: 08/22/2024] [Indexed: 09/18/2024]
Abstract
OBJECTIVE Enhanced expression of transforming growth factor (TGF) β in the kidneys of patients with lupus nephritis (LN) can lead to progressive fibrosis, resulting in end-organ damage. ADAM9 activates TGFβ1 by cleaving the latency-associated peptide (LAP). We hypothesized that ADAM9 in the kidney may accelerate fibrogenesis by activating TGFβ1. METHODS We assessed the expression of ADAM9 in the kidneys of mice and humans who were lupus prone. In vitro experiments were conducted using tubular epithelial cells (TECs) isolated from mice and explored the mechanisms responsible for the up-regulation of ADAM9 and the subsequent activation of TGFβ1. To assess the role of ADAM9 in the development of tubular-intestinal fibrosis in individuals with LN, we generated MRL/lpr mice who were Adam9 deficient. RESULTS ADAM9 was highly expressed in tubules from MRL/lpr mice. The transcription factor hypoxia-inducible factor-1α was found to promote the transcription of ADAM9 in TECs. TECs from mice who were Adam9 deficient and exposed to the hypoxia mimetic agent dimethyloxalylglycine failed to cleave the LAP to produce bioactive TGFβ1 from latent TGFβ1. Coculture of TECs from mice who were Adam9 deficient with fibroblasts in the presence of dimethyloxalylglycine and latent TGFβ1 produced decreased amounts of type I collagen and α-smooth muscle actin (SMA) by fibroblasts. MRL/lpr mice who were Adam9 deficient showed reduced interstitial fibrosis. At the translational level, ADAM9 expression in tissues and urine of patients with LN was found to increase. CONCLUSION Hypoxia promotes the expression of ADAM9 by TECs, which is responsible for the development of interstitial fibrosis in patients with LN by enhancing the TGFβ1 activation, which promotes fibroblasts to produce collagen and α-SMA.
Collapse
Affiliation(s)
- Masataka Umeda
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA, USA
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Research Unit for Preventive and Clinical Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kohei Karino
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA, USA
| | - Abhigyan Satyam
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA, USA
| | - Nobuya Yoshida
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA, USA
| | - Ryo Hisada
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA, USA
| | - Rhea Bhargava
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA, USA
| | - Theodoros Vichos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA, USA
| | - Ana Laura Kunzler
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA, USA
| | - Takashi Igawa
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kunihiro Ichinose
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Rheumatology, Shimane University Faculty of Medicine
| | - Kenta Torigoe
- Department of Nephrology, Nagasaki University Hospital, Nagasaki, Japan
| | - Tomoya Nishino
- Department of Nephrology, Nagasaki University Hospital, Nagasaki, Japan
| | - Takahiro Maeda
- Research Unit for Preventive and Clinical Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Caroline A. Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston MA, USA
| | - Reza Abdi
- Transplantation Research Center, Nephrology Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Atsushi Kawakami
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Research Unit for Preventive and Clinical Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - George C. Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA, USA
| |
Collapse
|
5
|
Tang C, Teymur A, Wu T. Urinary Immune Complexes Reflect Renal Pathology in Lupus Nephritis. Diagnostics (Basel) 2024; 14:2787. [PMID: 39767148 PMCID: PMC11727095 DOI: 10.3390/diagnostics14242787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES Lupus nephritis (LN) is a serious complication of systemic lupus erythematosus (SLE), involving immune complex deposition in the kidneys. While renal biopsy is the diagnostic gold standard, its invasiveness limits frequent use, driving the need for non-invasive urinary biomarkers to monitor disease progression and response to treatment. This study aimed to identify and validate urinary biomarkers for LN. METHODS Data from 10 LN-related omics databases, including urine, PBMCs, and kidney tissue, were analyzed. Differentially expressed proteins (DEPs) and genes (DEGs) were identified, and candidate biomarkers were validated via ELISA in an independent cohort of 87 urine samples. RESULTS We identified 78 biomarkers, with 14 overlapping across transcriptomic categories. Novel urinary biomarkers, including SERPING1, SLPI, and CD48, were validated. Urinary CD163, VCAM1, and ALCAM levels showed significant differences between LN and healthy controls, while urinary immune complexes (ICx) demonstrated superior diagnostic performance, with urinary ALCAM-ICx and CCL21-ICx achieving the highest AUC values. CONCLUSIONS Our findings highlight the potential of urinary immune complexes and antigens as non-invasive biomarkers for LN. ALCAM, CD163, and SERPING1-ICx, in particular, were found as promising candidates for a urinary biomarker panel to aid in the diagnosis and monitoring of LN.
Collapse
Affiliation(s)
| | | | - Tianfu Wu
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (C.T.); (A.T.)
| |
Collapse
|
6
|
Fu Y, Zhang Z, He C, Jia L, Ge C, Long L, Bai Y, Zhang N, Shen L, Du Q, Zhao H. Performance of urinary C-C motif chemokine ligand 14 and the renal resistive index in predicting persistent AKI. Ren Fail 2024; 46:2438855. [PMID: 39676224 DOI: 10.1080/0886022x.2024.2438855] [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: 07/16/2024] [Revised: 11/14/2024] [Accepted: 12/01/2024] [Indexed: 12/17/2024] Open
Abstract
OBJECTIVES The purpose of this study was to evaluate the performance of urinary C-C motif chemokine ligand 14 (CCL14) and the renal resistive index (RI) in predicting persistent AKI in unselected critically ill patients. METHODS This prospective observational study was conducted in a tertiary hospital's general intensive care unit (ICU). Consecutive adults who were admitted to the ICU were enrolled, with a primary endpoint of AKI lasting 48 h or longer. Urinary CCL14 was evaluated upon inclusion, and the renal RI was determined within 12 h of ICU admission. The individual discriminative ability of urinary CCL14 and the renal RI to predict persistent AKI was evaluated by the area under the receiver operating characteristic curve (AUC). RESULTS Overall, 166 patients were included, of whom 56 had persistent AKI. Urinary CCL14 showed good ability to predict persistent AKI, with an AUC of 0.817. However, the overall performance of the renal RI was fair, with an AUC of 0.739. Forty-nine patients presented with mild AKI at inclusion, and the values of CCL14 were significantly lower than those of patients with moderate or severe AKI (0.205 [0.125-0.300] vs. 0.302 [0.157-0.501]; p = 0.034). In the subgroup analysis, although the diagnostic performance of CCL14 was excellent in patients with moderate or severe AKI, it was fair in patients with mild AKI [AUC = 0. 738; 95% confidence interval (CI) 0.593-0.853]. CONCLUSION Urinary CCL14 was an excellent predictor of persistent AKI in patients with moderate or severe AKI, but its performance was not good in patients with mild AKI. The renal RI cannot discriminate between transient and persistent AKI.
Collapse
Affiliation(s)
- You Fu
- Department of Intensive Care Unit, Hebei General Hospital, Shijiazhuang City, China
| | - Ze Zhang
- Department of Intensive Care Unit, Hebei General Hospital, Shijiazhuang City, China
| | - Cong He
- Department of Intensive Care Unit, Hebei General Hospital, Shijiazhuang City, China
| | - Lijing Jia
- Department of Intensive Care Unit, Hebei General Hospital, Shijiazhuang City, China
| | - Chen Ge
- Department of Intensive Care Unit, Hebei General Hospital, Shijiazhuang City, China
| | - Ling Long
- Department of Intensive Care Unit, Hebei General Hospital, Shijiazhuang City, China
| | - Yinxiang Bai
- Department of Intensive Care Unit, Hebei General Hospital, Shijiazhuang City, China
| | - Na Zhang
- Department of Intensive Care Unit, Hebei General Hospital, Shijiazhuang City, China
| | - Limin Shen
- Department of Intensive Care Unit, Hebei General Hospital, Shijiazhuang City, China
| | - Quansheng Du
- Department of Intensive Care Unit, Hebei General Hospital, Shijiazhuang City, China
| | - Heling Zhao
- Department of Intensive Care Unit, Hebei General Hospital, Shijiazhuang City, China
| |
Collapse
|
7
|
Vier J, Häcker G, Kirschnek S. Contribution of A1 to macrophage survival in cooperation with MCL-1 and BCL-X L in a murine cell model of myeloid differentiation. Cell Death Dis 2024; 15:677. [PMID: 39285161 PMCID: PMC11405755 DOI: 10.1038/s41419-024-07064-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/20/2024]
Abstract
Myeloid cells are the first line of defence against pathogens. Mitochondrial apoptosis signalling is a crucial regulator of myeloid cell lifespan and modulates the function of myeloid cells. The anti-apoptotic protein BCL-2-family protein BCL2A1/A1/BFL-1 is strongly upregulated in inflammation in macrophages. We analysed the contribution of A1 to apoptosis regulation in a conditional system of in vitro differentiation of murine macrophages from immortalised progenitors. We disabled the expression of A1 by targeting all murine A1 isoforms in the genome. Specific inhibitors were used to inactivate other anti-apoptotic proteins. Macrophage progenitor survival mainly depended on the anti-apoptotic proteins MCL-1, BCL-XL and A1 but not BCL-2. Deletion of A1 on its own had little effect on progenitor cell survival but was sensitised to cell death induction when BCL-XL or MCL-1 was neutralised. In progenitors, A1 was required for survival in the presence of the inflammatory stimulus LPS. Differentiated macrophages were resistant to inhibition of single anti-apoptotic proteins, but A1 was required to protect macrophages against inhibition of either BCL-XL or MCL-1; BCL-2 only had a minor role in these cells. Cell death by neutralisation of anti-apoptotic proteins completely depended on BAX with a small contribution of BAK only in progenitors in the presence of LPS. A1 and NOXA appeared to stabilise each other at the posttranscriptional level suggesting direct binding. Co-immunoprecipitation experiments showed the binding of A1 to NOXA and BIM. Interaction between A1 and Noxa may indirectly prevent neutralisation and destabilization of MCL-1. Our findings suggest a unique role for A1 as a modulator of survival in the macrophage lineage in concert with MCL-1 and BCL-XL, especially in a pro-inflammatory environment.
Collapse
Affiliation(s)
- Juliane Vier
- Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Georg Häcker
- Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Susanne Kirschnek
- Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Faculty of Medicine, Freiburg, Germany.
| |
Collapse
|
8
|
Wang S, Rao Z, Cao R, Blaes AH, Coresh J, Deo R, Dubin R, Joshu CE, Lehallier B, Lutsey PL, Pankow JS, Post WS, Rotter JI, Sedaghat S, Tang W, Thyagarajan B, Walker KA, Ganz P, Platz EA, Guan W, Prizment A. Development, characterization, and replication of proteomic aging clocks: Analysis of 2 population-based cohorts. PLoS Med 2024; 21:e1004464. [PMID: 39316596 PMCID: PMC11460707 DOI: 10.1371/journal.pmed.1004464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 10/08/2024] [Accepted: 08/22/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Biological age may be estimated by proteomic aging clocks (PACs). Previous published PACs were constructed either in smaller studies or mainly in white individuals, and they used proteomic measures from only one-time point. In this study, we created de novo PACs and compared their performance to published PACs at 2 different time points in the Atherosclerosis Risk in Communities (ARIC) study of white and black participants (around 75% white and 25% black). MEDTHODS AND FINDINGS A total of 4,712 plasma proteins were measured using SomaScan in blood samples collected in 1990 to 1992 from 11,761 midlife participants (aged 46 to 70 years) and in 2011 to 2013 from 5,183 late-life participants (aged 66 to 90 years). The de novo ARIC PACs were constructed by training them against chronological age using elastic net regression in two-thirds of healthy participants in midlife and late life and validated in the remaining one-third of healthy participants at the corresponding time point. We also computed 3 published PACs. We estimated age acceleration for each PAC as residuals after regressing each PAC on chronological age. We also calculated the change in age acceleration from midlife to late life. We examined the associations of age acceleration and change in age acceleration with mortality through 2019 from all-cause, cardiovascular disease (CVD), cancer, and lower respiratory disease (LRD) using Cox proportional hazards regression in participants (irrespective of health) after excluding the training set. The model was adjusted for chronological age, smoking, body mass index (BMI), and other confounders. We externally validated the midlife PAC using the Multi-Ethnic Study of Atherosclerosis (MESA) Exam 1 data. The ARIC PACs had a slightly stronger correlation with chronological age than published PACs in healthy participants at each time point. Associations with mortality were similar for the ARIC PACs and published PACs. For late-life and midlife age acceleration for the ARIC PACs, respectively, hazard ratios (HRs) per 1 standard deviation were 1.65 and 1.38 (both p < 0.001) for all-cause mortality, 1.37 and 1.20 (both p < 0.001) for CVD mortality, 1.21 (p = 0.028) and 1.04 (p = 0.280) for cancer mortality, and 1.68 and 1.36 (both p < 0.001) for LRD mortality. For the change in age acceleration, HRs for all-cause, CVD, and LRD mortality were comparable to the HRs for late-life age acceleration. The association between the change in age acceleration and cancer mortality was not significant. The external validation of the midlife PAC in MESA showed significant associations with mortality, as observed for midlife participants in ARIC. The main limitation is that our PACs were constructed in midlife and late-life participants. It is unknown whether these PACs could be applied to young individuals. CONCLUSIONS In this longitudinal study, we found that the ARIC PACs and published PACs were similarly associated with an increased risk of mortality. These findings suggested that PACs show promise as biomarkers of biological age. PACs may be serve as tools to predict mortality and evaluate the effect of anti-aging lifestyle and therapeutic interventions.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Zexi Rao
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Rui Cao
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Anne H. Blaes
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Josef Coresh
- Departments of Population Health and Medicine, New York University Glossman School of Medicine, New York, New York, United States of America
| | - Rajat Deo
- Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ruth Dubin
- Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Corinne E. Joshu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
| | | | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Wendy S. Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Sanaz Sedaghat
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Peter Ganz
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
| | - Weihua Guan
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Anna Prizment
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
9
|
Lopez LN, Durbin-Johnson B, Vargas CR, Ruzinski J, Goodling A, Mehrotra R, Vaisar T, Rocke DM, Afkarian M. Comparative Analysis of Protein Quantification by the SomaScan Assay versus Orthogonal Methods in Urine from People with Diabetic Kidney Disease. J Proteome Res 2024; 23:2598-2607. [PMID: 38965919 DOI: 10.1021/acs.jproteome.4c00322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
To our knowledge, calibration curves or other validations for thousands of SomaScan aptamers are not publicly available. Moreover, the abundance of urine proteins obtained from these assays is not routinely validated with orthogonal methods (OMs). We report an in-depth comparison of SomaScan readout for 23 proteins in urine samples from patients with diabetic kidney disease (n = 118) vs OMs, including liquid chromatography-targeted mass spectrometry (LC-MS), ELISA, and nephelometry. Pearson correlation between urine abundance of the 23 proteins from SomaScan 3.2 vs OMs ranged from -0.58 to 0.86, with a median (interquartile ratio, [IQR]) of 0.49 (0.18, 0.53). In multivariable linear regression, the SomaScan readout for 6 of the 23 examined proteins (26%) was most strongly associated with the OM-derived abundance of the same (target) protein. For 3 of 23 (13%), the SomaScan and OM-derived abundance of each protein were significantly associated, but the SomaScan readout was more strongly associated with OM-derived abundance of one or more "off-target" proteins. For the remaining 14 proteins (61%), the SomaScan readouts were not significantly associated with the OM-derived abundance of the targeted proteins. In 6 of the latest group, the SomaScan readout was not associated with urine abundance of any of the 23 quantified proteins. To sum, over half of the SomaScan results could not be confirmed by independent orthogonal methods.
Collapse
Affiliation(s)
- Lauren N Lopez
- Division of Nephrology, Department of Medicine, University of California, Davis, California 95616, United States
| | - Blythe Durbin-Johnson
- Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California, Davis, California 95616, United States
| | - Chenoa R Vargas
- Division of Nephrology, Department of Medicine, University of California, Davis, California 95616, United States
| | - John Ruzinski
- Kidney Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Anne Goodling
- Kidney Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Rajnish Mehrotra
- Kidney Research Institute, Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Tomas Vaisar
- Diabetes Institute, Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle,Washington98195,United States
| | - David M Rocke
- Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California, Davis, California 95616, United States
| | - Maryam Afkarian
- Division of Nephrology, Department of Medicine, University of California, Davis, California 95616, United States
| |
Collapse
|
10
|
Chen Q, Xu J, Liu L, Ye Q, Lin W, Liao Y, Gao R, Zhang X, Chen R, Xiong Y, Chen S, Ye X, Wei L. Proteomic Analysis of Idiopathic Nephrotic Syndrome Triggered by Primary Podocytopathies in Adults: Regulatory Mechanisms and Diagnostic Implications. J Proteome Res 2024; 23:2090-2099. [PMID: 38728052 DOI: 10.1021/acs.jproteome.4c00074] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Idiopathic nephrotic syndrome (NS) is a heterogeneous group of glomerular disorders which includes two major phenotypes: minimal change disease (MCD) and focal segmental glomerulosclerosis (FSGS). MCD and FSGS are classic types of primary podocytopathies. We aimed to explore the molecular mechanisms in NS triggered by primary podocytopathies and evaluate diagnostic value of the selected proteomic signatures by analyzing blood proteome profiling. Totally, we recruited 90 participants in two cohorts. The first cohort was analyzed using label-free quantitative (LFQ) proteomics to discover differential expressed proteins and identify enriched biological process in NS which were further studied in relation to clinical markers of kidney injury. The second cohort was analyzed using parallel reaction monitoring-based quantitative proteomics to verify the data of LFQ proteomics and assess the diagnostic performance of the selected proteins using receiver-operating characteristic curve analysis. Several biological processes (such as immune response, cell adhesion, and response to hypoxia) were found to be associated with kidney injury during MCD and FSGS. Moreover, three proteins (CSF1, APOC3, and LDLR) had over 90% sensitivity and specificity in detecting adult NS triggered by primary podocytopathies. The identified biological processes may play a crucial role in MCD and FSGS pathogenesis. The three blood protein markers are promising for diagnosing adult NS triggered by primary podocytopathies.
Collapse
Affiliation(s)
- Qiaoling Chen
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Institute of Clinical Immunology, Fuzhou 350001, China
| | - Jiaming Xu
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Lifang Liu
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Institute of Clinical Immunology, Fuzhou 350001, China
| | - Qiuping Ye
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Institute of Clinical Immunology, Fuzhou 350001, China
| | - Wanjun Lin
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Institute of Clinical Immunology, Fuzhou 350001, China
| | - Yonggen Liao
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Institute of Clinical Immunology, Fuzhou 350001, China
| | - Ruiyu Gao
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Xinyu Zhang
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Ruoyan Chen
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Yunfeng Xiong
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Sihui Chen
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Xiaoyi Ye
- Department of Nephrology, Mindong Hospital Affiliated to Fujian Medical University, Ningde 352000, China
| | - Lixin Wei
- Department of Nephrology, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Institute of Clinical Immunology, Fuzhou 350001, China
| |
Collapse
|
11
|
Zhang Z, Yang Z. A more detailed description of study methods could be needed: Comments on "Clinico-serological associations of urinary activated leukocyte cell adhesion molecule in systemic lupus erythematosus and lupus nephritis". Clin Rheumatol 2024; 43:2175-2176. [PMID: 38619728 DOI: 10.1007/s10067-024-06944-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/06/2024] [Accepted: 03/16/2024] [Indexed: 04/16/2024]
Affiliation(s)
- Zhencheng Zhang
- Department of Laboratory Medicine, Taizhou First People's Hospital, Huangyan Hospital of Wenzhou Medical University, 218 Hengjie Road, Taizhou, 318020, Zhejiang, China
| | - Zaixing Yang
- Department of Laboratory Medicine, Taizhou First People's Hospital, Huangyan Hospital of Wenzhou Medical University, 218 Hengjie Road, Taizhou, 318020, Zhejiang, China.
| |
Collapse
|
12
|
Li Z, Gan H, Ji K, Yang M, Pan T, Meng X, Liu T, Wang Z, Gong B, Liu K, Qi D, Fan H. Protopanaxadiol improves lupus nephritis by regulating the PTX3/MAPK/ERK1/2 pathway. J Nat Med 2024; 78:474-487. [PMID: 38431911 DOI: 10.1007/s11418-023-01777-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/25/2023] [Indexed: 03/05/2024]
Abstract
Lupus nephritis (LN) is a kidney disease that occurs after systemic lupus erythematosus (SLE) affects the kidneys. Pentraxin 3 (PTX3) is highly expressed in the serum of patients with LN. Renal PTX3 deposition is directly related to clinical symptoms such as proteinuria and inflammation. The excessive proliferation of mesangial cells (MCs) is one of the representative pathological changes in the progression of LN, which is closely related to its pathogenesis. Protopanaxadiol (PPD) is the main component of ginsenoside metabolism and has not been reported in LN. The aim of this study was to investigate the relationship between PTX3 and mesangial cell proliferation and to evaluate the potential role and mechanism of PPD in improving LN. PTX3 is highly expressed in the kidneys of LN patients and LN mice and is positively correlated with renal pathological indicators, including proteinuria and PCNA. The excessive expression of PTX3 facilitated the proliferation of MCs, facilitated the activation of the MAPK/ERK1/2 signaling pathway, and increased the expression of HIF-1α. Further studies showed that PPD can effectively inhibit the abnormal proliferation of MCs with high expression of PTX3 and significantly improve LN symptoms such as proteinuria in MRL/lpr mice. The mechanism may be related to the inhibition of the PTX3/MAPK/ERK1/2 pathway. In this study, both in vitro, in vivo, and clinical sample results show that PTX3 is involved in the regulation of MCs proliferation and the early occurrence of LN. Natural active compound PPD can improve LN by regulating the PTX3/MAPK/ERK1/2 pathway.
Collapse
Affiliation(s)
- Zhenyuan Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, 264005, Shandong, People's Republic of China
| | - Hailin Gan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, 264005, Shandong, People's Republic of China
| | - Kai Ji
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, 264005, Shandong, People's Republic of China
| | - Mingyan Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, 264005, Shandong, People's Republic of China
| | - Tao Pan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, 264005, Shandong, People's Republic of China
| | - Xiangting Meng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, 264005, Shandong, People's Republic of China
| | - Teng Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, 264005, Shandong, People's Republic of China
| | - Zhixia Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, 264005, Shandong, People's Republic of China
| | - Baifang Gong
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, 264005, Shandong, People's Republic of China
| | - Ke Liu
- Shandong Boyuan Biomedical Co., Ltd, Yantai, 264003, People's Republic of China
| | - Dong Qi
- Department of Nephrology, Yu-Huang-Ding Hospital/Qingdao University, No. 20 Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong Province, People's Republic of China.
| | - Huaying Fan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, 264005, Shandong, People's Republic of China.
| |
Collapse
|
13
|
Vanarsa K, Zhang T, Hutcheson J, Kumar SR, Nukala S, Inthavong H, Stanley B, Wu T, Mok CC, Saxena R, Mohan C. iTRAQ-based mass spectrometry screen to identify serum biomarkers in systemic lupus erythematosus. Lupus Sci Med 2024; 11:e000673. [PMID: 38782493 PMCID: PMC11116855 DOI: 10.1136/lupus-2022-000673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 09/15/2022] [Indexed: 05/25/2024]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a complex systemic autoimmune disorder with no reliable serum biomarkers currently available other than autoantibodies. METHODS In the present study, isobaric tags for relative and absolute quantitation-based mass spectrometry was used to screen the sera of patients with SLE to uncover potential disease biomarkers. RESULTS 85 common proteins were identified, with 16 being elevated (≥1.3) and 23 being decreased (≤0.7) in SLE. Of the 16 elevated proteins, serum alpha-1-microglobulin/bikunin precursor (AMBP), zinc alpha-2 glycoprotein (AZGP) and retinol-binding protein 4 (RBP4) were validated in independent cross-sectional cohorts (Cohort I, N=52; Cohort II, N=117) using an orthogonal platform, ELISA. Serum AMBP, AZGP and RBP4 were validated to be significantly elevated in both patients with inactive SLE and patients with active SLE compared with healthy controls (HCs) (p<0.05, fold change >2.5) in Cohort I. All three proteins exhibited good discriminatory power for distinguishing active SLE and inactive SLE (area under the curve=0.82-0.96), from HCs. Serum AMBP exhibited the largest fold change in active SLE (5.96) compared with HCs and correlated with renal disease activity. The elevation in serum AMBP was validated in a second cohort of patients with SLE of different ethnic origins, correlating with serum creatinine (r=0.60, p<0.001). CONCLUSION Since serum AMBP is validated to be elevated in SLE and correlated with renal disease, the clinical utility of this novel biomarker warrants further analysis in longitudinal cohorts of patients with lupus and lupus nephritis.
Collapse
Affiliation(s)
- Kamala Vanarsa
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Ting Zhang
- University of Houston, Houston, Texas, USA
- Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | | | - Sneha Ravi Kumar
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| | | | - Haleigh Inthavong
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| | | | - Tianfu Wu
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - C C Mok
- Medicine, Tuen Mun Hospital, Hong Kong
| | - Ramesh Saxena
- The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chandra Mohan
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| |
Collapse
|
14
|
Umeda M, Satyam A, Yoshida N, Kawakami A. A Disintegrin and metalloproteinase carves T cell abnormalities and pathogenesis in systemic lupus erythematosus. Clin Immunol 2024; 262:110168. [PMID: 38458301 PMCID: PMC11009040 DOI: 10.1016/j.clim.2024.110168] [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: 01/27/2024] [Revised: 02/29/2024] [Accepted: 03/03/2024] [Indexed: 03/10/2024]
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disorder impacting various organs, notably prevalent in women of reproductive age. This review explores the involvement of a disintegrin and metalloproteinases (ADAMs) in SLE pathogenesis. Despite advancements in understanding SLE through genome and transcriptome studies, the role of ADAMs in post-translational regulations remains insufficiently explored. ADAMs, transmembrane proteins with diverse functions, impact cell adhesion, migration, and inflammation by shedding cell surface proteins, growth factors, and receptors. Notably, ADAM9 is implicated in Th17 cell differentiation, which is crucial in SLE pathology. ADAM10 and ADAM17 play pivotal roles in T-cell biology, influencing immune cell development and differentiation. Elevated soluble ADAM substrates in SLE patients serve as potential biomarkers correlating with disease activity. Targeting ADAMs or their substrates offers promising therapeutic avenues for SLE management and treatment enhancement.
Collapse
Affiliation(s)
- Masataka Umeda
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | - Abhigyan Satyam
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nobuya Yoshida
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Atsushi Kawakami
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
15
|
Li Y, Tang C, Vanarsa K, Thai N, Castillo J, Lea GAB, Lee KH, Kim S, Pedroza C, Wu T, Saxena R, Mok CC, Mohan C. Proximity extension assay proteomics and renal single cell transcriptomics uncover novel urinary biomarkers for active lupus nephritis. J Autoimmun 2024; 143:103165. [PMID: 38194790 DOI: 10.1016/j.jaut.2023.103165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/11/2024]
Abstract
OBJECTIVE To identify urinary biomarkers that can distinguish active renal involvement in Lupus Nephritis (LN), a severe manifestation of systemic lupus erythematosus (SLE). METHODS Urine from 117 subjects, comprised of inactive SLE, active non-renal lupus, active LN, and healthy controls, were subjected to Proximity Extension Assay (PEA) based comprehensive proteomics followed by ELISA validation in an independent, ethnically diverse cohort. Proteomic data is also cross-referenced to renal transcriptomic data to elucidate cellular origins of biomarkers. RESULTS Systems biology analyses revealed progressive activation of cytokine signaling, chemokine activity and coagulation pathways, with worsening renal disease. In addition to validating 30 previously reported biomarkers, this study uncovers several novel candidates. Following ELISA validation in an independent cohort of different ethnicity, the six most discriminatory biomarkers for active LN were urinary ICAM-2, FABP4, FASLG, IGFBP-2, SELE and TNFSF13B/BAFF, with ROC AUC ≥80%, with most correlating strongly with clinical disease activity. Transcriptomic analyses of LN kidneys mapped the likely origin of these proteins to intra-renal myeloid cells (CXCL16, IL-1RT2, TNFSF13B/BAFF), T/NK cells (FASLG), leukocytes (ICAM2) and endothelial cells (SELE). CONCLUSION In addition to confirming the diagnostic potential of urine ALCAM, CD163, MCP1, SELL, ICAM1, VCAM1, NGAL and TWEAK for active LN, this study adds urine ICAM-2, FABP4, FASLG, IGFBP-2, SELE, and TNFSF13B/BAFF as additional markers that warrant systematic validation in larger cross-sectional and longitudinal cohorts.
Collapse
Affiliation(s)
- Yaxi Li
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Chenling Tang
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Kamala Vanarsa
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Nga Thai
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Jessica Castillo
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | | | - Kyung Hyun Lee
- Center for Clinical Research and Evidence-Based Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Soojin Kim
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Claudia Pedroza
- Center for Clinical Research and Evidence-Based Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Tianfu Wu
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Ramesh Saxena
- Department of Internal Medicine, Division of Nephrology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Chi Chiu Mok
- Department of Medicine, Tuen Mun Hospital, New Territories, Hong Kong, China
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA.
| |
Collapse
|
16
|
Fava A, Buyon J, Magder L, Hodgin J, Rosenberg A, Demeke DS, Rao DA, Arazi A, Celia AI, Putterman C, Anolik JH, Barnas J, Dall’Era M, Wofsy D, Furie R, Kamen D, Kalunian K, James JA, Guthridge J, Atta MG, Monroy Trujillo J, Fine D, Clancy R, Belmont HM, Izmirly P, Apruzzese W, Goldman D, Berthier CC, Hoover P, Hacohen N, Raychaudhuri S, Davidson A, Diamond B, Petri M. Urine proteomic signatures of histological class, activity, chronicity, and treatment response in lupus nephritis. JCI Insight 2024; 9:e172569. [PMID: 38258904 PMCID: PMC10906224 DOI: 10.1172/jci.insight.172569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024] Open
Abstract
Lupus nephritis (LN) is a pathologically heterogenous autoimmune disease linked to end-stage kidney disease and mortality. Better therapeutic strategies are needed as only 30%-40% of patients completely respond to treatment. Noninvasive biomarkers of intrarenal inflammation may guide more precise approaches. Because urine collects the byproducts of kidney inflammation, we studied the urine proteomic profiles of 225 patients with LN (573 samples) in the longitudinal Accelerating Medicines Partnership in RA/SLE cohort. Urinary biomarkers of monocyte/neutrophil degranulation (i.e., PR3, S100A8, azurocidin, catalase, cathepsins, MMP8), macrophage activation (i.e., CD163, CD206, galectin-1), wound healing/matrix degradation (i.e., nidogen-1, decorin), and IL-16 characterized the aggressive proliferative LN classes and significantly correlated with histological activity. A decline of these biomarkers after 3 months of treatment predicted the 1-year response more robustly than proteinuria, the standard of care (AUC: CD206 0.91, EGFR 0.9, CD163 0.89, proteinuria 0.8). Candidate biomarkers were validated and provide potentially treatable targets. We propose these biomarkers of intrarenal immunological activity as noninvasive tools to diagnose LN and guide treatment and as surrogate endpoints for clinical trials. These findings provide insights into the processes involved in LN activity. This data set is a public resource to generate and test hypotheses and validate biomarkers.
Collapse
Affiliation(s)
- Andrea Fava
- Division of Rheumatology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jill Buyon
- New York University School of Medicine, New York, New York, USA
| | | | - Jeff Hodgin
- University of Michigan, Ann Arbor, Michigan, USA
| | - Avi Rosenberg
- Division of Renal Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Deepak A. Rao
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Maryland, USA
| | - Arnon Arazi
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
| | - Alessandra Ida Celia
- Division of Rheumatology, Johns Hopkins University, Baltimore, Maryland, USA
- Università La Sapienza, Rome, Italy
| | - Chaim Putterman
- Albert Einstein College of Medicine, New York, New York, USA
- Azrieli Faculty of Medicine of Bar-Ilan University, Zefat, Israel
| | | | | | - Maria Dall’Era
- University of California, San Francisco, San Francisco, California, USA
| | - David Wofsy
- University of California, San Francisco, San Francisco, California, USA
| | - Richard Furie
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
| | - Diane Kamen
- Medical University of South Carolina, Charleston, South Carolina, USA
| | | | - Judith A. James
- Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Joel Guthridge
- Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Mohamed G. Atta
- Division of Nephrology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Derek Fine
- Division of Nephrology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Robert Clancy
- New York University School of Medicine, New York, New York, USA
| | | | - Peter Izmirly
- New York University School of Medicine, New York, New York, USA
| | - William Apruzzese
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Maryland, USA
| | - Daniel Goldman
- Division of Rheumatology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | | | - Soumya Raychaudhuri
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Maryland, USA
- Broad Institute, Boston, Maryland, USA
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Anne Davidson
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
| | - Betty Diamond
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
| | | | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
17
|
Guo Z, Guo Q, Li X, Gao X, Zhang L, Xu K. Urinary biomarkers associated with podocyte injury in lupus nephritis. Front Pharmacol 2024; 15:1324540. [PMID: 38313309 PMCID: PMC10834635 DOI: 10.3389/fphar.2024.1324540] [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: 11/30/2023] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
The most prevalent and devastating form of organ damage in systemic lupus erythematosus (SLE) is lupus nephritis (LN). LN is characterized by glomerular injury, inflammation, cell proliferation, and necrosis, leading to podocyte injury and tubular epithelial cell damage. Assays for urine biomarkers have demonstrated significant promise in the early detection of LN, evaluation of disease activity, and tracking of reaction to therapy. This is because they are non-invasive, allow for frequent monitoring and easy self-collection, transport and storage. Podocyte injury is believed to be a essential factor in LN. The extent and type of podocyte injury could be connected to the severity of proteinuria, making podocyte-derived cellular debris and injury-related urinary proteins potential markers for the diagnosis and monitoring of LN. This article focuses on studies examining urinary biomarkers associated with podocyte injury in LN, offering fresh perspectives on the application of biomarkers in the early detection and management of LN.
Collapse
Affiliation(s)
| | | | | | | | | | - Ke Xu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
18
|
Alduraibi FK, Tsokos GC. Lupus Nephritis Biomarkers: A Critical Review. Int J Mol Sci 2024; 25:805. [PMID: 38255879 PMCID: PMC10815779 DOI: 10.3390/ijms25020805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Lupus nephritis (LN), a major complication in individuals diagnosed with systemic lupus erythematosus, substantially increases morbidity and mortality. Despite marked improvements in the survival of patients with severe LN over the past 50 years, complete clinical remission after immunosuppressive therapy is achieved in only half of the patients. Therefore, timely detection of LN is vital for initiating prompt therapeutic interventions and improving patient outcomes. Biomarkers have emerged as valuable tools for LN detection and monitoring; however, the complex role of these biomarkers in LN pathogenesis remains unclear. Renal biopsy remains the gold standard for the identification of the histological phenotypes of LN and guides disease management. However, the molecular pathophysiology of specific renal lesions remains poorly understood. In this review, we provide a critical, up-to-date overview of the latest developments in the field of LN biomarkers.
Collapse
Affiliation(s)
- Fatima K. Alduraibi
- Department of Medicine, Division of Clinical Immunology and Rheumatology, Beth Israel Deaconess Medical Center, Harvard Teaching Hospital, Boston, MA 02215, USA
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Medicine, Division of Clinical Immunology and Rheumatology, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - George C. Tsokos
- Department of Medicine, Division of Clinical Immunology and Rheumatology, Beth Israel Deaconess Medical Center, Harvard Teaching Hospital, Boston, MA 02215, USA
| |
Collapse
|
19
|
Ding H, Shen Y, Hong SM, Xiang C, Shen N. Biomarkers for systemic lupus erythematosus - a focus on organ damage. Expert Rev Clin Immunol 2024; 20:39-58. [PMID: 37712757 DOI: 10.1080/1744666x.2023.2260098] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 08/16/2023] [Accepted: 09/13/2023] [Indexed: 09/16/2023]
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is complex autoimmune disease with heterogenous manifestations, unpredictable disease course and response to treatment. One of the critical needs in SLE management is the identification of reliable biomarkers that can aid in early diagnosis, accurate monitoring of disease activity, and assessment of treatment response. AREAS COVERED In the current review, we focus on the commonly affected organs (skin, kidney, and nervous system) in SLE to summarize the emerging biomarkers that show promise in disease diagnosis, monitoring and treatment response assessment. The subtitles within each organ domain were determined based on the most relevant and promising biomarkers for that specific organ damage. EXPERT OPINION Biomarkers have the potential to significantly benefit the management of SLE by aiding in diagnosis, disease activity monitoring, prognosis, and treatment response assessment. However, despite decades of research, none has been validated and implemented for routine clinical use. Novel biomarkers could lead to the development of precision medicine for SLE, guide personalized treatment, and improve patient outcomes. Challenges in biomarker research in SLE include defining clear and clinically relevant questions, accounting for the heterogeneity of SLE, and confirming initial findings in larger, multi-center, multi-ethnic, independent cohorts that reflect real-world clinical scenarios.
Collapse
Affiliation(s)
- Huihua Ding
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Yiwei Shen
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Soon-Min Hong
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Chunyan Xiang
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Nan Shen
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- China-Australia Centre for Personalized Immunology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Department of Rheumatology, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Center for Autoimmune Genomics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Collaborative Innovation Centre for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Yung S, Chan TM. Endothelial cell activation and glycocalyx shedding - potential as biomarkers in patients with lupus nephritis. Front Immunol 2023; 14:1251876. [PMID: 37854589 PMCID: PMC10579905 DOI: 10.3389/fimmu.2023.1251876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
Lupus nephritis (LN) is a common and severe manifestation of systemic lupus erythematosus and an important cause of acute and chronic kidney injury. Early diagnosis of LN and preventing relapses are key to preserving renal reserve. However, due to the complexity and heterogeneity of the disease, clinical management remains challenging. Kidney biopsy remains the gold standard for confirming the diagnosis of LN and subsequent assessment of kidney histopathology, but it is invasive and cannot be repeated frequently. Current clinical indicators of kidney function such as proteinuria and serum creatinine level are non-specific and do not accurately reflect histopathological changes, while anti-dsDNA antibody and C3 levels reflect immunological status but not kidney injury. Identification of novel and specific biomarkers for LN is prerequisite to improve management. Renal function deterioration is associated with changes in the endothelial glycocalyx, a delicate gel-like layer located at the interface between the endothelium and bloodstream. Inflammation induces endothelial cell activation and shedding of glycocalyx constituents into the circulation. This review discusses the potential role of soluble glycocalyx components as biomarkers of active LN, especially in patients in whom conventional serological and biochemical markers do not appear helpful.
Collapse
Affiliation(s)
- Susan Yung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Tak Mao Chan
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
21
|
Avasare R, Drexler Y, Caster DJ, Mitrofanova A, Jefferson JA. Management of Lupus Nephritis: New Treatments and Updated Guidelines. KIDNEY360 2023; 4:1503-1511. [PMID: 37528520 PMCID: PMC10617804 DOI: 10.34067/kid.0000000000000230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 07/25/2023] [Indexed: 08/03/2023]
Abstract
Management of lupus nephritis has evolved considerably over the past years. Here, we provide a comprehensive review of clinical trials that form the basis for the Kidney Disease: Improving Global Outcomes and EULAR/ERA-EDTA updated guidelines and present day trials that will change the landscape of lupus nephritis therapy in years to come. In addition, we highlight the issues related to cost of therapy, resistant disease, and downstream adverse effects of specific therapies.
Collapse
Affiliation(s)
- Rupali Avasare
- Nephrology and Hypertension, Oregon Health & Science University School of Medicine, Portland, Oregon
| | - Yelena Drexler
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Dawn J. Caster
- Division of Nephrology and Hypertension, University of Louisville School of Medicine, Louisville, Kentucky
| | - Alla Mitrofanova
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | | |
Collapse
|
22
|
Wang S, Rao Z, Cao R, Blaes AH, Coresh J, Joshu CE, Lehallier B, Lutsey PL, Pankow JS, Sedaghat S, Tang W, Thyagarajan B, Walker KA, Ganz P, Platz EA, Guan W, Prizment A. Development and Characterization of Proteomic Aging Clocks in the Atherosclerosis Risk in Communities (ARIC) Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.06.23295174. [PMID: 37732184 PMCID: PMC10508816 DOI: 10.1101/2023.09.06.23295174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Biological age may be estimated by proteomic aging clocks (PACs). Previous published PACs were constructed either in smaller studies or mainly in White individuals, and they used proteomic measures from only one-time point. In the Atherosclerosis Risk in Communities (ARIC) study of about 12,000 persons followed for 30 years (around 75% White, 25% Black), we created de novo PACs and compared their performance to published PACs at two different time points. We measured 4,712 plasma proteins by SomaScan in 11,761 midlife participants, aged 46-70 years (1990-92), and 5,183 late-life pariticpants, aged 66-90 years (2011-13). All proteins were log2-transformed to correct for skewness. We created de novo PACs by training them against chronological age using elastic net regression in two-thirds of healthy participants in midlife and late life and compared their performance to three published PACs. We estimated age acceleration (by regressing each PAC on chronological age) and its change from midlife to late life. We examined their associations with mortality from all-cause, cardiovascular disease (CVD), cancer, and lower respiratory disease (LRD) using Cox proportional hazards regression in all remaining participants irrespective of health. The model was adjusted for chronological age, smoking, body mass index (BMI), and other confounders. The ARIC PACs had a slightly stronger correlation with chronological age than published PACs in healthy participants at each time point. Associations with mortality were similar for the ARIC and published PACs. For late-life and midlife age acceleration for the ARIC PACs, respectively, hazard ratios (HRs) per one standard deviation were 1.65 and 1.38 (both p<0.001) for all-cause mortality, 1.37 and 1.20 (both p<0.001) for CVD mortality, 1.21 (p=0.03) and 1.04 (p=0.19) for cancer mortality, and 1.46 and 1.68 (both p<0.001) for LRD mortality. For the change in age acceleration, HRs for all-cause, CVD, and LRD mortality were comparable to those observed for late-life age acceleration. The association between the change in age acceleration and cancer mortality was insignificant. In this prospective study, the ARIC and published PACs were similarly associated with an increased risk of mortality and advanced testing in relation to various age-related conditions in future studies is suggested.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| | - Zexi Rao
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Rui Cao
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Anne H. Blaes
- Division of Hematology, Oncology and Transplantation, Medical School, University of Minnesota, Minneapolis, MN
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Corinne E. Joshu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Benoit Lehallier
- Alkahest Inc, San Carlos, CA, United States, Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA
| | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Sanaz Sedaghat
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute on Aging, Baltimore, MD
| | - Peter Ganz
- Division of Cardiology, Zuckerberg San Francisco General Hospital and Department of Medicine, University of California, San Francisco, CA
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Weihua Guan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Anna Prizment
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| |
Collapse
|
23
|
Shen Y, Vanarsa K, Yin Z, Zhang T, Castillo J, Dai M, Zou L, Qin L, Wang J, Guo Q, Saxena R, Petri M, Shen N, Ye Z, Mohan C, Ding H. Urine L-selectin reflects clinical and histological renal disease activity and treatment response in lupus nephritis across multi-ethnicity. Front Immunol 2023; 14:1200167. [PMID: 37720233 PMCID: PMC10500131 DOI: 10.3389/fimmu.2023.1200167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/17/2023] [Indexed: 09/19/2023] Open
Abstract
Objective There is an urgent need for novel biomarkers in lupus nephritis (LN). We report a non-invasive urinary biomarker, L-selectin, in two independent multi-ethnic cohorts. Methods uL-selectin was tested cross-sectionally in a Chinese cohort (n=255) and a US cohort (n=219) of SLE patients and controls using ELISA. A longitudinal cohort includes 20 active Chinese LN patients. Results uL-selectin was significantly increased in active LN patients compared to active non-renal SLE, inactive LN, inactive non-renal SLE, chronic kidney disease patients, and healthy controls. uL-selectin positively correlated with global and renal disease activities as well as histological activity index and chronicity index (CI). Low uL-selectin was an independent predictor for high CI. During follow-up, uL-selectin levels decreased significantly in the complete renal remission group. Conclusion uL-selectin is a novel biomarker of disease activity and renal histopathology in LN across multiple ethnicities. It also reflects treatment response in LN patients during follow up.
Collapse
Affiliation(s)
- Yiwei Shen
- Department of Rheumatology, Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kamala Vanarsa
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Zhihua Yin
- Department of Rheumatology, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Department of Rheumatology, Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Ting Zhang
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
- Division of Rheumatology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Jessica Castillo
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Min Dai
- Department of Rheumatology, Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Linghua Zou
- Department of Rheumatology, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Department of Rehabilitation, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Ling Qin
- Department of Nephrology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jieying Wang
- Clinical Research Unit, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiang Guo
- Department of Rheumatology, Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ramesh Saxena
- Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nan Shen
- Department of Rheumatology, Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Rheumatology, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Zhizhong Ye
- Department of Rheumatology, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Department of Rheumatology, Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Huihua Ding
- Department of Rheumatology, Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
24
|
Mackay S, Frazer LC, Bailey GK, Miller CM, Gong Q, Dewitt ON, Singh DK, Good M. Identification of serum biomarkers for necrotizing enterocolitis using aptamer-based proteomics. Front Pediatr 2023; 11:1184940. [PMID: 37325361 PMCID: PMC10264655 DOI: 10.3389/fped.2023.1184940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/10/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction Necrotizing enterocolitis (NEC) is a potentially fatal intestinal disease primarily affecting preterm infants. Early diagnosis of neonates with NEC is crucial to improving outcomes; however, traditional diagnostic tools remain inadequate. Biomarkers represent an opportunity to improve the speed and accuracy of diagnosis, but they are not routinely used in clinical practice. Methods In this study, we utilized an aptamer-based proteomic discovery assay to identify new serum biomarkers of NEC. We compared levels of serum proteins in neonates with and without NEC and identified ten differentially expressed serum proteins between these groups. Results We detected two proteins, C-C motif chemokine ligand 16 (CCL16) and immunoglobulin heavy constant alpha 1 and 2 heterodimer (IGHA1 IGHA2), that were significantly increased during NEC and eight that were significantly decreased. Generation of receiver operating characteristic (ROC) curves revealed that alpha-fetoprotein (AUC = 0.926), glucagon (AUC = 0.860), and IGHA1 IGHA2 (AUC = 0.826) were the proteins that best differentiated patients with and without NEC. Discussion These findings indicate that further investigation into these serum proteins as a biomarker for NEC is warranted. In the future, laboratory tests incorporating these differentially expressed proteins may improve the ability of clinicians to diagnose infants with NEC rapidly and accurately.
Collapse
Affiliation(s)
- Stephen Mackay
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, NC, United States
| | - Lauren C. Frazer
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, NC, United States
| | - Grace K. Bailey
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, NC, United States
| | - Claire M. Miller
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, NC, United States
| | - Qingqing Gong
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Olivia N. Dewitt
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Dhirendra K. Singh
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, NC, United States
| | - Misty Good
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, NC, United States
| |
Collapse
|
25
|
Mohan C, Zhang T, Putterman C. Pathogenic cellular and molecular mediators in lupus nephritis. Nat Rev Nephrol 2023:10.1038/s41581-023-00722-z. [PMID: 37225921 DOI: 10.1038/s41581-023-00722-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2023] [Indexed: 05/26/2023]
Abstract
Kidney involvement in patients with systemic lupus erythematosus - lupus nephritis (LN) - is one of the most important and common clinical manifestations of this disease and occurs in 40-60% of patients. Current treatment regimens achieve a complete kidney response in only a minority of affected individuals, and 10-15% of patients with LN develop kidney failure, with its attendant morbidity and considerable prognostic implications. Moreover, the medications most often used to treat LN - corticosteroids in combination with immunosuppressive or cytotoxic drugs - are associated with substantial side effects. Advances in proteomics, flow cytometry and RNA sequencing have led to important new insights into immune cells, molecules and mechanistic pathways that are instrumental in the pathogenesis of LN. These insights, together with a renewed focus on the study of human LN kidney tissue, suggest new therapeutic targets that are already being tested in lupus animal models and early-phase clinical trials and, as such, are hoped to eventually lead to meaningful improvements in the care of patients with systemic lupus erythematosus-associated kidney disease.
Collapse
Affiliation(s)
- Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA.
| | - Ting Zhang
- Division of Rheumatology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chaim Putterman
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
- Division of Rheumatology and Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
26
|
Vanarsa K, Castillo J, Wang L, Lee KH, Pedroza C, Lotan Y, Mohan C. Comprehensive proteomics and platform validation of urinary biomarkers for bladder cancer diagnosis and staging. BMC Med 2023; 21:133. [PMID: 37016361 PMCID: PMC10074794 DOI: 10.1186/s12916-023-02813-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 03/02/2023] [Indexed: 04/06/2023] Open
Abstract
BACKGROUND Bladder cancer (BC) is among the most common cancers diagnosed in men in the USA. The current gold standards for the diagnosis of BC are invasive or lack the sensitivity to correctly identify the disease. METHODS An aptamer-based screen analyzed the expression of 1317 proteins in BC compared to urology clinic controls. The top hits were subjected to systems biology analyses. Next, 30 urine proteins were ELISA-validated in an independent cohort of 68 subjects. Three of these proteins were next validated in an independent BC cohort of differing ethnicity. RESULTS Systems biology analysis implicated molecular functions related to the extracellular matrix, collagen, integrin, heparin, and transmembrane tyrosine kinase signaling in BC susceptibility, with HNF4A and NFKB1 emerging as key molecular regulators. STEM analysis of the dysregulated pathways implicated a functional role for the immune system, complement, and interleukins in BC disease progression. Of 21 urine proteins that discriminated BC from urology clinic controls (UC), urine D-dimer displayed the highest accuracy (0.96) and sensitivity of 97%. Furthermore, 8 urine proteins significantly discriminated MIBC from NMIBC (AUC = 0.75-0.99), with IL-8 and IgA being the best performers. Urine IgA and fibronectin exhibited the highest specificity of 80% at fixed sensitivity for identifying advanced BC. CONCLUSIONS Given the high sensitivity (97%) of urine D-dimer for BC, it may have a role in the initial diagnosis or detection of cancer recurrence. On the other hand, urine IL-8 and IgA may have the potential in identifying disease progression during patient follow-up. The use of these biomarkers for initial triage could have a significant impact as the current cystoscopy-based diagnostic and surveillance approach is costly and invasive when compared to a simple urine test.
Collapse
Affiliation(s)
- Kamala Vanarsa
- Department Biomedical Engineering, University of Houston, 3517 Cullen Blvd., Room 2027, Houston, TX, 77204-5060, USA
| | - Jessica Castillo
- Department Biomedical Engineering, University of Houston, 3517 Cullen Blvd., Room 2027, Houston, TX, 77204-5060, USA
| | - Long Wang
- Department of Urology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Kyung Hyun Lee
- Center for Clinical Research and Evidence-Based Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Claudia Pedroza
- Center for Clinical Research and Evidence-Based Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yair Lotan
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Chandra Mohan
- Department Biomedical Engineering, University of Houston, 3517 Cullen Blvd., Room 2027, Houston, TX, 77204-5060, USA.
| |
Collapse
|
27
|
Qian BS, Jia HM, Weng YB, Li XC, Chen CD, Guo FX, Han YZ, Huang LF, Zheng Y, Li WX. Analysis of urinary C-C motif chemokine ligand 14 (CCL14) and first-generation urinary biomarkers for predicting renal recovery from acute kidney injury: a prospective exploratory study. J Intensive Care 2023; 11:11. [PMID: 36941674 PMCID: PMC10026399 DOI: 10.1186/s40560-023-00659-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/07/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a frequent syndrome in the intensive care unit (ICU). AKI patients with kidney function recovery have better short-term and long-term prognoses compared with those with non-recovery. Numerous studies focus on biomarkers to distinguish them. To better understand the predictive performance of urinary biomarkers of renal recovery in patients with AKI, we evaluated C-C motif chemokine ligand 14 (CCL14) and two first-generation biomarkers (cell cycle arrest biomarkers and neutrophil gelatinase-associated lipocalin) in two ICU settings. METHODS We performed a prospective study to analyze urinary biomarkers for predicting renal recovery from AKI. Patients who developed AKI after ICU admission were enrolled and urinary biomarkers including tissue inhibitor of metalloproteinase-2 (TIMP-2), insulin-like growth factor-binding protein 7 (IGFBP7), CCL14, and neutrophil gelatinase-associated lipocalin (NGAL) were detected on the day of AKI diagnosis. The primary endpoint was non-recovery from AKI within 7 days. The individual discriminative ability of CCL14, [TIMP-2] × [IGFBP7] and NGAL to predict renal non-recovery were evaluated by the area under receiver operating characteristics curve (AUC). RESULTS Of 164 AKI patients, 64 (39.0%) failed to recover from AKI onset. CCL14 showed a fair prediction ability for renal non-recovery with an AUC of 0.71 (95% CI 0.63-0.77, p < 0.001). [TIMP-2] × [IGFBP7] showed the best prediction for renal non-recovery with an AUC of 0.78 (95% CI 0.71-0.84, p < 0.001). However, NGAL had no use in predicting non-recovery with an AUC of 0.53 (95% CI 0.45-0.60, p = 0.562). A two-parameter model (non-renal SOFA score and AKI stage) predicted renal non-recovery with an AUC of 0.77 (95% CI 0.77-0.83, p = 0.004). When [TIMP-2] × [IGFBP7] was combined with the clinical factors, the AUC was significantly improved to 0.82 (95% CI 0.74-0.87, p = 0.049). CONCLUSIONS Urinary CCL14 and [TIMP-2] × [IGFBP7] were fair predictors of renal non-recovery from AKI. Combing urinary [TIMP-2] × [IGFBP7] with a clinical model consisting of non-renal SOFA score and AKI stage enhanced the predictive power for renal non-recovery. Urinary CCL14 showed no significant advantage in predicting renal non-recovery compared to [TIMP-2] × [IGFBP7].
Collapse
Affiliation(s)
- Ben-Shu Qian
- Department of Surgical Intensive Critical Unit, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing, 100020, China
| | - Hui-Miao Jia
- Department of Surgical Intensive Critical Unit, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing, 100020, China
| | - Yi-Bing Weng
- Department of Emergent Intensive Critical Unit, Beijing Lu-He Hospital, Capital Medical University, Beijing, 101100, China
| | - Xin-Cheng Li
- Department of Surgical Intensive Critical Unit, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing, 100020, China
| | - Chao-Dong Chen
- Department of Surgical Intensive Critical Unit, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing, 100020, China
| | - Fang-Xing Guo
- Department of Surgical Intensive Critical Unit, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing, 100020, China
| | - Yu-Zhen Han
- Department of Surgical Intensive Critical Unit, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing, 100020, China
| | - Li-Feng Huang
- Department of Surgical Intensive Critical Unit, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing, 100020, China
| | - Yue Zheng
- Department of Surgical Intensive Critical Unit, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing, 100020, China.
| | - Wen-Xiong Li
- Department of Surgical Intensive Critical Unit, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
28
|
Louis Sam Titus ASC, Vanarsa K, Soomro S, Patel A, Prince J, Kugathasan S, Mohan C. Resistin, Elastase, and Lactoferrin as Potential Plasma Biomarkers of Pediatric Inflammatory Bowel Disease Based on Comprehensive Proteomic Screens. Mol Cell Proteomics 2023; 22:100487. [PMID: 36549591 PMCID: PMC9918796 DOI: 10.1016/j.mcpro.2022.100487] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 11/10/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Inflammatory bowel disease (IBD) is an immune-mediated chronic inflammation of the intestine, which can present in the form of ulcerative colitis (UC) or as Crohn's disease (CD). Biomarkers are needed for reliable diagnosis and disease monitoring in IBD, especially in pediatric patients. Plasma samples from a pediatric IBD cohort were interrogated using an aptamer-based screen of 1322 proteins. The elevated biomarkers identified using the aptamer screen were further validated by ELISA using an independent cohort of 76 pediatric plasma samples, drawn from 30 CD, 30 UC, and 16 healthy controls. Of the 1322 proteins screened in plasma from IBD patients, 129 proteins were significantly elevated when compared with healthy controls. Of these 15 proteins had a fold change greater than 2 and 28 proteins had a fold change >1.5. Neutrophil and extracellular vesicle signatures were detected among the elevated plasma biomarkers. When seven of these proteins were validated by ELISA, resistin was the only protein that was significantly higher in both UC and CD (p < 0.01), with receiver operating characteristic area under the curve value of 0.82 and 0.77, respectively, and the only protein that exhibited high sensitivity and specificity for both CD and UC. The next most discriminatory plasma proteins were elastase and lactoferrin, particularly for UC, with receiver operating characteristic area under the curve values of 0.74 and 0.69, respectively. We have identified circulating resistin, elastase, and lactoferrin as potential plasma biomarkers of IBD in pediatric patients using two independent diagnostic platforms and two independent patient cohorts.
Collapse
Affiliation(s)
| | - Kamala Vanarsa
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Sanam Soomro
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Anjali Patel
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Jarod Prince
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Subra Kugathasan
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA.
| | - Chandra Mohan
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA.
| |
Collapse
|
29
|
Faustini F, Idborg H, Fuzzi E, Larsson A, Lie WR, Pötzsch S, Okitsu SL, Svenungsson E, Gunnarsson I. Urine Galectin-3 binding protein reflects nephritis activity in systemic lupus erythematosus. Lupus 2023; 32:252-262. [PMID: 36508734 PMCID: PMC9939930 DOI: 10.1177/09612033221145534] [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] [Indexed: 12/14/2022]
Abstract
BACKGROUND Lupus nephritis (LN) is a major and severe organ involvement in systemic lupus erythematosus (SLE), whose diagnosis and treatment necessitate to perform kidney biopsy, which is an invasive procedure. Non-invasive urine biomarkers are an active area of investigation to support LN diagnosis and management. OBJECTIVE To investigate the role of urinary galectin-3 binding protein (u-Gal-3BP) as a candidate biomarker of renal disease in biopsy proven LN. PATIENTS AND METHODS Levels of u-Gal-3BP were investigated in a cross-sectional fashion by ELISA in 270 subjects: 86 LN patients, 63 active SLE patients with no kidney involvement, 73 SLE patients with inactive disease and 48 age and sex-matched population-based controls (PBC). Moreover, urine samples were analysed separately by ELISA for additional markers of kidney pathology: neutrophil gelatinase-associated lipocalin (NGAL), osteopontin (OPN), kidney injury molecule-1 (KIM-1) and galectin-3 (Gal-3). The concentrations of all studied molecules were normalized to urine creatinine levels. In 10 patients, post-treatment levels of the biomarkers were measured. RESULTS Normalized u-Gal-3BP levels were higher in LN patients compared to the other groups (p < .0001). Comparing different LN classes, u-Gal-3BP levels were higher among patients with proliferative (class III/IV) and membranous (class V) as compared to mesangial (class II) forms (p = .04). In proliferative forms, u-Gal-3BP levels correlated with the activity index in renal biopsies (r = 0.42, p = .004). Moreover, in a subset of 10 patients with repeated kidney biopsy and urine sampling before and after induction treatment, a significant decrease of u-Gal-3BP was observed (p = .03). Among the other markers, KIM-1 was also able to discriminate LN from the other groups, while NGAL, OPN and Gal-3 could not in this cohort. CONCLUSION Given its ability to discriminate LN patients from active non-renal and inactive SLE patients, the observed correlation with the activity index in renal biopsies, and its levels declining following treatment, u-Gal-3BP shows promise as a non-invasive urinary biomarker to help detecting and to monitor renal involvement in SLE patients and should be validated in larger cohorts.
Collapse
Affiliation(s)
- Francesca Faustini
- Division of Rheumatology, Department of Medicine Solna, Karolinska University Hospital, 27106Karolinska Institute, Stockholm, Sweden
| | - Helena Idborg
- Division of Rheumatology, Department of Medicine Solna, Karolinska University Hospital, 27106Karolinska Institute, Stockholm, Sweden
| | - Enrico Fuzzi
- Division of Rheumatology, Department of Medicine Solna, Karolinska University Hospital, 27106Karolinska Institute, Stockholm, Sweden.,Division of Rheumatology, Department of Medicine DIMED, Padua University Hospital, Padua, Italy
| | - Anders Larsson
- Department of Medical Sciences/Clinical Chemistry, 8097Uppsala University, Uppsala, Sweden
| | | | | | - Shinji L Okitsu
- 189697EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Elisabet Svenungsson
- Division of Rheumatology, Department of Medicine Solna, Karolinska University Hospital, 27106Karolinska Institute, Stockholm, Sweden
| | - Iva Gunnarsson
- Division of Rheumatology, Department of Medicine Solna, Karolinska University Hospital, 27106Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
30
|
Bellocchi C, Assassi S, Lyons M, Marchini M, Mohan C, Santaniello A, Beretta L. Proteomic aptamer analysis reveals serum markers that characterize preclinical systemic sclerosis (SSc) patients at risk for progression toward definite SSc. Arthritis Res Ther 2023; 25:15. [PMID: 36707842 PMCID: PMC9881382 DOI: 10.1186/s13075-023-02989-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 01/04/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND The study of molecular mechanisms characterizing disease progression may be relevant to get insights into systemic sclerosis (SSc) pathogenesis and to intercept patients at very early stage. We aimed at investigating the proteomic profile of preclinical systemic sclerosis (PreSSc) via a discovery/validation two-step approach. METHODS SOMAcan aptamer-based analysis was performed on a serum sample of 13 PreSSc (discovery cohort) according to 2001 LeRoy and Medsger criteria (characterized solely by Raynaud phenomenon plus a positive nailfold capillaroscopy and SSc-specific antibodies without any other sign of definite disease) and 8 healthy controls (HCs) age, gender, and ethnicity matched. Prospective data were available up to 4±0.6 years to determine the progression to definite SSc according to the EULAR/ACR 2013 classification criteria. In proteins with relative fluorescence units (RFU) > |1.5|-fold vs HCs values, univariate analysis was conducted via bootstrap aggregating models to determine the predicting accuracy (progression vs non-progression) of categorized baseline protein values. Gene Ontologies (GO terms) and Reactome terms of significant proteins at the adjusted 0.05 threshold were explored. Significant proteins from the discovery cohort were finally validated via ELISAs in an independent validation cohort of 50 PreSSc with clinical prospective data up to 5 years. Time-to-event analysis for interval-censored data was used to evaluate disease progression. RESULTS In the discovery cohort, 286 out of 1306 proteins analyzed via SomaScan, were differentially expressed versus HCs. Ten proteins were significantly associated with disease progression; analysis through GO and Reactome showed differentially enriched pathways involving angiogenesis, endothelial cell chemotaxis, and endothelial cell chemotaxis to fibroblast growth factor (FGF). In the validation cohort, endostatin (HR=10.23, CI95=2.2-47.59, p=0.003) was strongly associated with disease progression, as well as bFGF (HR=0.84, CI95=0.709-0.996, p=0.045) and PAF-AHβ (HR=0.372, CI95=0.171-0.809, p=0.013) CONCLUSIONS: A distinct protein profile characterized PreSSc from HCs and proteins associated with hypoxia, vasculopathy, and fibrosis regulation are linked with the progression from preclinical to definite SSc. These proteins, in particular endostatin, can be regarded both as markers of severity and molecules with pathogenetic significance as well as therapeutic targets.
Collapse
Affiliation(s)
- Chiara Bellocchi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Scleroderma Unit, Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Shervin Assassi
- Department of Internal Medicine – Rheumatology, University of Texas Health Science Center at Houston, Houston, TX USA
| | - Marka Lyons
- Department of Internal Medicine – Rheumatology, University of Texas Health Science Center at Houston, Houston, TX USA
| | - Maurizio Marchini
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Scleroderma Unit, Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX USA
| | - Alessandro Santaniello
- Scleroderma Unit, Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Beretta
- Scleroderma Unit, Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
31
|
Abstract
Uncontrolled alternative pathway activation is the primary driver of several diseases, and it contributes to the pathogenesis of many others. Consequently, diagnostic tests to monitor this arm of the complement system are increasingly important. Defects in alternative pathway regulation are strong risk factors for disease, and drugs that specifically block the alternative pathway are entering clinical use. A range of diagnostic tests have been developed to evaluate and monitor the alternative pathway, including assays to measure its function, expression of alternative pathway constituents, and activation fragments. Genetic studies have also revealed many disease-associated variants in alternative pathway genes that predict the risk of disease and prognosis. Newer imaging modalities offer the promise of non-invasively detecting and localizing pathologic complement activation. Together, these various tests help in the diagnosis of disease, provide important prognostic information, and can help guide therapy with complement inhibitory drugs.
Collapse
Affiliation(s)
- Joshua M. Thurman
- Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Veronique Fremeaux-Bacchi
- Assistance Publique-Hôpitaux de Paris, European Hospital Georges Pompidou, Department of Immunology Biology and INSERM UMRS1138, Centre de Recherche des Cordeliers, Team "Inflammation, Complement and Cancer", Paris, France
| |
Collapse
|
32
|
Lei R, Vu B, Kourentzi K, Soomro S, Danthanarayana AN, Brgoch J, Nadimpalli S, Petri M, Mohan C, Willson RC. A novel technology for home monitoring of lupus nephritis that tracks the pathogenic urine biomarker ALCAM. Front Immunol 2022; 13:1044743. [PMID: 36569940 PMCID: PMC9780296 DOI: 10.3389/fimmu.2022.1044743] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
Introduction The gold standard for diagnosis of active lupus nephritis (ALN), a kidney biopsy, is invasive with attendant morbidity and cannot be serially repeated. Urinary ALCAM (uALCAM) has shown high diagnostic accuracy for renal pathology activity in ALN patients. Methods Lateral flow assays (LFA) for assaying uALCAM were engineered using persistent luminescent nanoparticles, read by a smartphone. The stability and reproducibility of the assembled LFA strips and freeze-dried conjugated nanoparticles were verified, as was analyte specificity. Results The LFA tests for both un-normalized uALCAM (AUC=0.93) and urine normalizer (HVEM)-normalized uALCAM (AUC=0.91) exhibited excellent accuracies in distinguishing ALN from healthy controls. The accuracies for distinguishing ALN from all other lupus patients were 0.86 and 0.74, respectively. Conclusion Periodic monitoring of uALCAM using this easy-to-use LFA test by the patient at home could potentially accelerate early detection of renal involvement or disease flares in lupus patients, and hence reduce morbidity and mortality.
Collapse
Affiliation(s)
- Rongwei Lei
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Binh Vu
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, United States
| | - Katerina Kourentzi
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, United States
| | - Sanam Soomro
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | | | - Jakoah Brgoch
- Department of Chemistry, University of Houston, Houston, TX, United States
| | - Suma Nadimpalli
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Richard C. Willson
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, United States
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
- Escuela de Medicina y Ciencias de Salud, Tecnológico de Monterrey, Monterrey, NL, Mexico
| |
Collapse
|
33
|
Fan X, Zhang X, Liu LC, Zhang S, Pelger CB, Lughmani HY, Haller ST, Gunning WT, Cooper CJ, Gong R, Dworkin LD, Gupta R. Hemopexin accumulates in kidneys and worsens acute kidney injury by causing hemoglobin deposition and exacerbation of iron toxicity in proximal tubules. Kidney Int 2022; 102:1320-1330. [PMID: 36007598 DOI: 10.1016/j.kint.2022.07.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 06/29/2022] [Accepted: 07/11/2022] [Indexed: 01/12/2023]
Abstract
Hemopexin, a heme scavenging protein, accumulates in the kidneys during acute kidney injury (AKI). However, the function of this accumulated hemopexin in the kidney is unclear. In both the cisplatin-induced and the unilateral kidney ischemia-reperfusion injury models of AKI, we found accumulation of hemoglobin and hemopexin in the kidneys localized to the proximal tubules. Next, hemopexin wild-type and knockout mice were compared in both AKI models and hemopexin wild type mice had significantly worse kidney injury. Furthermore, there was increased kidney expression of kidney injury molecule-1 (a biomarker of AKI) and heme oxygenase-1 (an indicator of oxidative stress) in hemopexin wild type compared with knockout mice in both models of AKI. Next, the interaction of hemopexin and hemoglobin in vitro was investigated using cultured proximal tubular cells. Co-incubation of hemopexin with hemoglobin resulted in hemoglobin deposition and exaggerated hemoglobin-induced injury. Deferoxamine, an iron chelator, and ferrostatin-1, a ferroptosis inhibitor, inhibited this deleterious effect of hemoglobin and hemopexin in proximal tubular cells, implicating iron toxicity in the mechanism of hemopexin mediated injury. Furthermore, the protective effect of deferoxamine in cisplatin-induced AKI was apparent in hemopexin wild type, but not in hemopexin knockout mice, further implicating hemopexin as a mediator of iron toxicity in AKI. Thus, our findings demonstrate that hemopexin accumulates in the kidneys and worsens kidney injury in AKI by increasing hemoglobin deposition on proximal tubular cells to exaggerate hemoglobin-induced cell injury.
Collapse
Affiliation(s)
- Xiaoming Fan
- Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Xiaolu Zhang
- Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Lijun C Liu
- Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Shungang Zhang
- Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Cole B Pelger
- Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Haroon Y Lughmani
- Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Steven T Haller
- Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - William T Gunning
- Department of Pathology, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Christopher J Cooper
- Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Rujun Gong
- Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Lance D Dworkin
- Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Rajesh Gupta
- Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA.
| |
Collapse
|
34
|
Palazzo L, Lindblom J, Mohan C, Parodis I. Current Insights on Biomarkers in Lupus Nephritis: A Systematic Review of the Literature. J Clin Med 2022; 11:5759. [PMID: 36233628 PMCID: PMC9570701 DOI: 10.3390/jcm11195759] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/19/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
Lupus nephritis (LN) is a major cause of morbidity and mortality among patients with systemic lupus erythematosus (SLE). However, promising emerging biomarkers pave the way toward an improved management of patients with LN. We have reviewed the literature over the past decade, and we herein summarise the most relevant biomarkers for diagnosis, monitoring, and prognosis in LN. An initial systematic search of Medline was conducted to identify pertinent articles. A total of 104 studies were selected to be included in this review. Several diagnostic biomarkers, including MCP-1, TWEAK, NGAL, and uric acid, exhibited good ability to differentiate LN patients from non-renal SLE patients. Several cytokines and chemokines, including IL-10, IL-17, MCP-1, and IP-10, hold promise for assessing LN disease activity, as do cell adhesion molecules (CAMs). Angiogenesis-related and haemostasis-related proteins have also displayed potential for monitoring disease activity. Biomarkers of responses to therapy include Axl, CD163, and BAFF, whereas VCAM-1, ALCAM, and ANCAs have been reported as prognostic markers, along with traditional markers. In addition, novel renal tissue biomarkers may prove to be a useful complement to histological evaluations. The overall heterogeneity of the inclusion criteria and outcome measures across different studies, along with a lack of validation in multi-centre cohorts, call for future collaborative efforts. Nevertheless, we foresee that several biomarkers hold promise toward optimisation of the management of LN, with the use of integrated omics and panels of less invasive biomarkers paving the way towards personalised medicine.
Collapse
Affiliation(s)
- Leonardo Palazzo
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, 171 77 Stockholm, Sweden
- Medical Unit of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Julius Lindblom
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, 171 77 Stockholm, Sweden
- Medical Unit of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Chandra Mohan
- Department Biomedical Engineering, University of Houston, Houston, TX 77204, USA
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, 171 77 Stockholm, Sweden
- Medical Unit of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, 171 76 Stockholm, Sweden
- Department of Rheumatology, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
| |
Collapse
|
35
|
Soliman SA, Stanley S, Vanarsa K, Ismail F, Mok CC, Mohan C. Exploring urine:serum fractional excretion ratios as potential biomarkers for lupus nephritis. Front Immunol 2022; 13:910993. [PMID: 36091001 PMCID: PMC9449537 DOI: 10.3389/fimmu.2022.910993] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 08/01/2022] [Indexed: 11/26/2022] Open
Abstract
Objectives The goal of this exploratory study is to determine if urine:serum fractional excretion ratios can outperform the corresponding urinary biomarker proteins in identifying active renal disease in systemic lupus erythematosus (SLE). Methods Thirty-six adult SLE patients and twelve healthy controls were examined for serum and urine levels of 8 protein markers, namely ALCAM, calpastatin, hemopexin, peroxiredoxin 6 (PRDX6), platelet factor 4 (PF4), properdin, TFPI and VCAM-1, by ELISA. Fractional excretion of analyzed biomarkers was calculated after normalizing both the urine and serum biomarker levels against creatinine. A further validation cohort of fifty SLE patients was included to validate the initial findings. Results The FE ratios of all 8 proteins interrogated outperformed conventional disease activity markers such as anti-dsDNA, C3 and C4 in identifying renal disease activity. All but VCAM-1FE were superior to the corresponding urine biomarkers levels in differentiating LN activity, exhibiting positive correlation with renal SLEDAI. ALCAMFE, PF4FE and properdinFE ratios exhibited the highest accuracy (AUC>0.9) in distinguishing active LN from inactive SLE. Four of the FE ratios exhibited perfect sensitivity (calpastatin, PRDX6, PF4 and properdin), while ALCAMFE, PF4FE and properdinFE exhibited the highest specificity values for active LN. In addition, several of these novel biomarkers were associated with higher renal pathology activity indices. In the validation cohort ALCAMFE, PF4FE and properdinFE once again exhibited higher accuracy metrics, surpassing corresponding urine and serum biomarkers levels, with ALCAMFE exhibiting 95% accuracy in distinguishing active LN from inactive SLE. Conclusions With most of the tested proteins, urine:serum fractional excretion ratios outperformed corresponding urine and serum protein measurements in identifying active renal involvement in SLE. Hence, this novel class of biomarkers in SLE ought to be systemically evaluated in larger independent cohorts for their diagnostic utility in LN assessment.
Collapse
Affiliation(s)
- Samar A. Soliman
- Department of Rheumatology & Rehabilitation, Faculty of Medicine, Minia University, Minia, Egypt
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Samantha Stanley
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Kamala Vanarsa
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Faten Ismail
- Department of Rheumatology & Rehabilitation, Faculty of Medicine, Minia University, Minia, Egypt
| | - Chi Chiu Mok
- Department of Medicine, Tuen Mun Hospital, Hong Kong, Hong Kong SAR, China
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
- *Correspondence: Chandra Mohan,
| |
Collapse
|
36
|
Chen J, Luo P, Wang C, Yang C, Bai Y, He X, Zhang Q, Zhang J, Yang J, Wang S, Wang J. Integrated single-cell transcriptomics and proteomics reveal cellular-specific responses and microenvironment remodeling in aristolochic acid nephropathy. JCI Insight 2022; 7:e157360. [PMID: 35852860 PMCID: PMC9462482 DOI: 10.1172/jci.insight.157360] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 07/14/2022] [Indexed: 11/30/2022] Open
Abstract
Aristolochic acid nephropathy (AAN) is characterized by acute proximal tubule necrosis and immune cell infiltration, contributing to the global burden of chronic kidney disease and urothelial cancer. Although the proximal tubule has been defined as the primary target of aristolochic acids I (AAI), the mechanistic underpinning of gross renal deterioration caused by AAI has not been explicitly explained, prohibiting effective therapeutic intervention. To this point, we employed integrated single-cell RNA-Seq, bulk RNA-Seq, and mass spectrometry-based proteomics to analyze the mouse kidney after acute AAI exposure. Our results reveal a dramatic reduction of proximal tubule epithelial cells, associated with apoptotic and inflammatory pathways, indicating permanent damage beyond repair. We found the enriched development pathways in other nephron segments, suggesting activation of reparative programs triggered by AAI. The divergent response may be attributed to the segment-specific distribution of organic anion channels along the nephron, including OAT1 and OAT3. Moreover, we observed dramatic activation and recruitment of cytotoxic T and macrophage M1 cells, highlighting inflammation as a principal contributor to permanent renal injury. Ligand-receptor pairing revealed that critical intercellular crosstalk underpins damage-induced activation of immune cells. These results provide potentially novel insight into the AAI-induced kidney injury and point out possible pathways for future therapeutic intervention.
Collapse
Affiliation(s)
- Jiayun Chen
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Piao Luo
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chen Wang
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chuanbin Yang
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Yunmeng Bai
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Xueling He
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Zhang
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junzhe Zhang
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Yang
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Shuang Wang
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Jigang Wang
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Center for Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Southern Medical University, Dongguan, Guangdong, China
| |
Collapse
|
37
|
Yu C, Li P, Dang X, Zhang X, Mao Y, Chen X. Lupus nephritis: new progress in diagnosis and treatment. J Autoimmun 2022; 132:102871. [PMID: 35999111 DOI: 10.1016/j.jaut.2022.102871] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 02/07/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic multifactorial autoimmune disease that affects many organs, including the kidney. Lupus nephritis (LN) is a common manifestation characterized by heterogeneous clinical and histopathological findings, and often associates with poor prognosis. The diagnosis and treatment of LN is challenging, depending largely on renal biopsy, and there is no reliable non-invasive LN biomarker. Up to now, the complete remission rate of LN is only 20%∼30% after receiving six months of standard treatment, which is far from satisfactory. Moreover, adverse reactions to immunosuppressants, especially glucocorticoids, further compromise the prognosis of LN. Biological reagents targetting autoimmune responses and inflammatory pathways, bring hope to the treatment of intractable lupus. The European Renal Association-European Dialysis and Transplant Association (EULAR/ERA-EDTA) and KDIGO (Kidney Disease: Improving Global Outcomes) have been working on and launched the recommendations for the management of LN. In this review, we update our knowledge in the pathogenesis, diagnosis, and management of LN and prospect for the future potential targets in the management of LN.
Collapse
Affiliation(s)
- Chen Yu
- Department of Nephrology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Ping Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Xin Dang
- Department of Nephrology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yonghui Mao
- Department of Nephrology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China.
| |
Collapse
|
38
|
Guo X, Zhou X. Risk stratification of acute myeloid leukemia: Assessment using a novel prediction model based on ferroptosis-immune related genes. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:11821-11839. [PMID: 36653976 DOI: 10.3934/mbe.2022551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
In acute myeloid leukemia (AML), the link between ferroptosis and the immune microenvironment has profound clinical significance. The objective of this study was to investigate the role of ferroptosis-immune related genes (FIRGs) in predicting the prognosis and therapeutic sensitivity in patients with AML. Using The Cancer Genome Atlas dataset, single sample gene set enrichment analysis was performed to calculate the ferroptosis score of AML samples. To search for FIRGs, differentially expressed genes between the high- and low-ferroptosis score groups were identified and then cross-screened with immune related genes. Univariate Cox and LASSO regression analyses were performed on the FIRGs to establish a prognostic risk score model with five signature FIRGs (BMP2, CCL3, EBI3, ELANE, and S100A6). The prognostic risk score model was then used to divide the patients into high- and low-risk groups. For external validation, two Gene Expression Omnibus cohorts were employed. Overall survival was poorer in the high-risk group than in the low-risk group. The novel risk score model was an independent prognostic factor for overall survival in patients with AML. Infiltrating immune cells were also linked to high-risk scores. Treatment targeting programmed cell death protein 1 may be more effective in high-risk patients. This FIRG-based prognostic risk model may aid in optimizing prognostic risk stratification and treatment of AML.
Collapse
Affiliation(s)
- Xing Guo
- Department of Hematology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Xiaogang Zhou
- Department of Hematology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| |
Collapse
|
39
|
Mahroum N, Elsalti A, Alwani A, Seida I, Alrais M, Seida R, Esirgun SN, Abali T, Kiyak Z, Zoubi M, Shoenfeld Y. The mosaic of autoimmunity - Finally discussing in person. The 13 th international congress on autoimmunity 2022 (AUTO13) Athens. Autoimmun Rev 2022; 21:103166. [PMID: 35932955 PMCID: PMC9349027 DOI: 10.1016/j.autrev.2022.103166] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 07/31/2022] [Indexed: 11/29/2022]
Abstract
While autoimmunity is a branch of medicine linked to every single organ system via direct and indirect pathways, meeting in person to discuss autoimmunity during the 13th international congress on autoimmunity (AUTO13) with participants from all over the world had a very good reason. The mechanisms involved in autoimmune diseases are of extreme importance and in fact critical in understanding the course of diseases as well as selecting proper therapies. COVID-19 has served as a great example of how autoimmunity is deeply involved in the disease and directly correlated to severity, morbidity, and mortality. For instance, initially the term cytokine storm dominated, then COVID-19 was addressed as the new member of the hyperferritinemic syndrome, and also the use of immunosuppressants in patients with COVID-19 throughout the pandemic, all shed light on the fundamental role of autoimmunity. Unsurprisingly, SARS-CoV-2 was called the “autoimmune virus” during AUTO13. Subsequently, the correlation between autoimmunity and COVID-19 vaccines and post-COVID, all were discussed from different autoimmune aspects during the congress. In addition, updates on the mechanisms of diseases, autoantibodies, novel diagnostics and therapies in regard to autoimmune diseases such as antiphospholipid syndrome, systemic lupus erythematosus, systemic sclerosis and others, were discussed in dedicated sessions. Due to the magnificence of the topics discussed, we aimed to bring in our article hereby, the pearls of AUTO13 in terms of updates, new aspects of autoimmunity, and interesting findings. While more than 500 abstract were presented, concluding all the topics was not in reach, hence major findings were summarized.
Collapse
Affiliation(s)
- Naim Mahroum
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey.
| | - Abdulrahman Elsalti
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Abdulkarim Alwani
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Isa Seida
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Mahmoud Alrais
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ravend Seida
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Sevval Nil Esirgun
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Tunahan Abali
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Zeynep Kiyak
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Majdi Zoubi
- Department of Internal Medicine B, HaEmek Medical Center, Afula, Israel, Affiliated to Technion, Faculty of Medicine, Haifa, Israel
| | | |
Collapse
|
40
|
Soomro S, Stanley S, Lei R, Saxena R, Petri M, Mohan C. Comprehensive Urinomic Identification of Protein Alternatives to Creatinine Normalization for Diagnostic Assessment of Lupus Nephritis. Front Immunol 2022; 13:853778. [PMID: 35774777 PMCID: PMC9237323 DOI: 10.3389/fimmu.2022.853778] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/26/2022] [Indexed: 12/12/2022] Open
Abstract
Introduction The current gold standard used for urine biomarker normalization, creatinine, poses a challenge to translate to the point of care because antibodies to creatinine are difficult to develop and currently available ligands to creatinine are sub-optimal for this purpose. Hence, protein alternatives to creatinine are clearly needed. To address this need, lupus nephritis was selected as a model disease where urine protein assessment is required for diagnosis. Methods A comprehensive proteomic screen of 1129 proteins in healthy and lupus nephritis urine was executed to identify protein alternatives to creatinine for the normalization of urine biomarkers. Urinary proteins that correlated well with creatinine but did not vary with disease were further validated by ELISA in an independent cohort of lupus nephritis subjects. Results The comprehensive proteomic screen identified 14 urine proteins that correlated significantly with urine creatinine but did not differ significantly between SLE and controls. Of the top five proteins selected for ELISA validation, urine HVEM and RELT once again showed significant correlation with urine creatinine in independent cohorts. Normalizing a lupus nephritis biomarker candidate ALCAM using urinary HVEM demonstrated comparable diagnostic ability to creatinine normalization when distinguishing active lupus nephritis from inactive SLE patients. Conclusions The discovery of urine HVEM as a protein alternative to creatinine for biomarker normalization has applications in the engineering of antibody-based point of care diagnostics for monitoring lupus nephritis progression.
Collapse
Affiliation(s)
- Sanam Soomro
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Samantha Stanley
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Rongwei Lei
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Ramesh Saxena
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Michelle Petri
- Division of Rheumatology, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
- *Correspondence: Chandra Mohan,
| |
Collapse
|
41
|
Whittall-Garcia L, Goliad K, Kim M, Bonilla D, Gladman D, Urowitz M, Fortin PR, Atenafu EG, Touma Z, Wither J. Identification and Validation of a Urinary Biomarker Panel to Accurately Diagnose and Predict Response to Therapy in Lupus Nephritis. Front Immunol 2022; 13:889931. [PMID: 35711439 PMCID: PMC9196040 DOI: 10.3389/fimmu.2022.889931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/02/2022] [Indexed: 11/15/2022] Open
Abstract
Background We have previously shown that 15 urinary biomarkers (of 129 tested by Luminex), discriminate between active Lupus Nephritis (ALN) and non-LN patients. The aim of this study was to evaluate the ability of these 15 previously-identified urinary biomarkers to predict treatment responses to conventional therapy, and for the most predictive of these biomarkers to validate their utility to identify ALN patients in an independent prospectively-acquired lupus cohort. Methods Our study had a 3-stage approach. In stage 1, we used Luminex to examine whether our previously identified urinary biomarkers at the time of the renal flare ( ± 3 months) or 12 ± 3 months after treatment of biopsy-proven ALN could predict treatment responses. In stage 2, a larger prospectively-acquired cross-sectional cohort was used to further validate the utility of the most predictive urinary biomarkers (identified in stage 1) to detect ALN patients. In this 2nd stage, cut-offs with the best operating characteristics to detect ALN patients were produced for each biomarker and different combinations and/or numbers of elevated biomarkers needed to accurately identify ALN patients were analyzed. In stage 3, we aimed to further corroborate the sensitivity of the cut-offs created in stage 2 to detect ALN patients in a biopsy-proven ALN cohort who had a urine sample collection within 3 months of their biopsy. Results Twenty-one patients were included in stage 1. Twelve (57.1%), 4 (19.1%), and 5 (23.8%) patients had a complete (CR), partial (PR) and no (NR) remission at 24 ± 3 months, respectively. The percentage decrease following 12 ± 3 months of treatment for Adiponectin, MCP-1, sVCAM-1, PF4, IL-15 and vWF was significantly higher in patients with CR in comparison to those with PR/NR. In stage 2, a total of 247 SLE patients were included, of which 24 (9.7%) had ALN, 79 (31.9%) had LN in remission (RLN) and 144 (58.3%) were non-LN (NLN) patients. Based on the combinations of biomarkers with the best operating characteristics we propose “rule out” and “rule in” ALN criteria. In stage 3, 53 biopsy-proven ALN patients were included, 35 with proliferative LN and 18 with non-proliferative ALN, demonstrating that our “rule in ALN” criteria operate better in detecting active proliferative than non-proliferative classes. Conclusions Our results provide further evidence to support the role of Adiponectin, MCP-1, sVCAM-1 and PF4 in the detection of proliferative ALN cases. We further show the clinical utility of measuring multiple rather than a single biomarker and we propose novel “rule in” and “rule out” criteria for the detection of proliferative ALN with excellent operating characteristics.
Collapse
Affiliation(s)
- Laura Whittall-Garcia
- Division of Rheumatology, Department of Medicine, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Kirubel Goliad
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Michael Kim
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Dennisse Bonilla
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Dafna Gladman
- Division of Rheumatology, Department of Medicine, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Murray Urowitz
- Division of Rheumatology, Department of Medicine, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Paul R. Fortin
- Division of Rheumatology, Department of Medicine, Centre de recherche du CHU de Québec–Université Laval, Quebec City, QC, Canada
| | - Eshetu G. Atenafu
- Department of Biostatistics, University Health Network, Toronto, ON, Canada
| | - Zahi Touma
- Division of Rheumatology, Department of Medicine, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Joan Wither
- Division of Rheumatology, Department of Medicine, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- *Correspondence: Joan Wither,
| |
Collapse
|
42
|
Fava A, Rao DA, Mohan C, Zhang T, Rosenberg A, Fenaroli P, Belmont HM, Izmirly P, Clancy R, Monroy Trujillo J, Fine D, Arazi A, Berthier CC, Davidson A, James JA, Diamond B, Hacohen N, Wofsy D, Raychaudhuri S, Apruzzese W, Buyon J, Petri M. Urine Proteomics and Renal Single-Cell Transcriptomics Implicate Interleukin-16 in Lupus Nephritis. Arthritis Rheumatol 2022; 74:829-839. [PMID: 34783463 PMCID: PMC9050800 DOI: 10.1002/art.42023] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 10/13/2021] [Accepted: 11/09/2021] [Indexed: 11/10/2022]
Abstract
OBJECTIVE Current lupus nephritis (LN) treatments are effective in only 30% of patients, emphasizing the need for novel therapeutic strategies. We undertook this study to develop mechanistic hypotheses and explore novel biomarkers by analyzing the longitudinal urinary proteomic profiles in LN patients undergoing treatment. METHODS We quantified 1,000 urinary proteins in 30 patients with LN at the time of the diagnostic renal biopsy and after 3, 6, and 12 months. The proteins and molecular pathways detected in the urine proteome were then analyzed with respect to baseline clinical features and longitudinal trajectories. The intrarenal expression of candidate biomarkers was evaluated using single-cell transcriptomics of renal biopsy sections from LN patients. RESULTS Our analysis revealed multiple biologic pathways, including chemotaxis, neutrophil activation, platelet degranulation, and extracellular matrix organization, which could be noninvasively quantified and monitored in the urine. We identified 237 urinary biomarkers associated with LN, as compared to controls without systemic lupus erythematosus. Interleukin-16 (IL-16), CD163, and transforming growth factor β mirrored intrarenal nephritis activity. Response to treatment was paralleled by a reduction in urinary IL-16, a CD4 ligand with proinflammatory and chemotactic properties. Single-cell RNA sequencing independently demonstrated that IL16 is the second most expressed cytokine by most infiltrating immune cells in LN kidneys. IL-16-producing cells were found at key sites of kidney injury. CONCLUSION Urine proteomics may profoundly change the diagnosis and management of LN by noninvasively monitoring active intrarenal biologic pathways. These findings implicate IL-16 in LN pathogenesis, designating it as a potentially treatable target and biomarker.
Collapse
Affiliation(s)
- Andrea Fava
- Division of Rheumatology, Johns Hopkins University, Baltimore, MD, USA
| | - Deepak A. Rao
- Division of Rheumatology, Immunology, Allergy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Avi Rosenberg
- Division of Renal Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Paride Fenaroli
- Division of Renal Pathology, Johns Hopkins University, Baltimore, MD, USA
- Nephrology Unit, Parma University Hospital, Department of Medicine and Surgery, Parma, Italy
| | | | - Peter Izmirly
- New York University School of Medicine, New York, New York, USA
| | - Robert Clancy
- New York University School of Medicine, New York, New York, USA
| | | | - Derek Fine
- Division of Nephrology, Johns Hopkins University, Baltimore, MD, USA
| | - Arnon Arazi
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | | | | | - Judith A. James
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Betty Diamond
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | | | - David Wofsy
- University of California San Francisco, San Francisco, CA, USA
| | - Soumya Raychaudhuri
- Division of Rheumatology, Immunology, Allergy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Data Sciences, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, UK
| | - William Apruzzese
- Division of Rheumatology, Immunology, Allergy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Jill Buyon
- New York University School of Medicine, New York, New York, USA
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
43
|
Zhou X, Wang N, Zhang Y, Yu P. Expression of CCL2, FOS, and JUN May Help to Distinguish Patients With IgA Nephropathy From Healthy Controls. Front Physiol 2022; 13:840890. [PMID: 35464092 PMCID: PMC9021889 DOI: 10.3389/fphys.2022.840890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/28/2022] [Indexed: 11/26/2022] Open
Abstract
Background IgA nephropathy (IgAN), the most common type of glomerulonephritis worldwide, can only be diagnosed mainly by renal biopsy owing to lack of effective biomarkers. It is urgent to explore and identify the potential diagnostic biomarkers through assessing the gene expression profiles of patients with IgAN. Methods Two datasets were obtained from the Gene Expression Omnibus (GEO) database, including GSE115857 (55 IgAN, 7 living healthy donors) and GSE35487 (25 IgAN, 6 living healthy donors), then underwent differentially expressed genes (DEGs) and function enrichment analyses utilizing R packages. The common gene list was screened out between DEGs and immune-associated genes by Venn diagram, then performed gene-gene interaction, protein-protein interaction (PPI) and function enrichment analyses. Top three immune-associated hub genes were selected by Maximal Clique Centrality (MCC) method, then the expression and diagnostic value of these hub genes were determined. Consensus clustering algorithm was applied to conduct the unsupervised cluster analysis of the immune-associated hub gene list in IgAN. Finally, the Nephroseq V5 tool was applied to identify the expression level of CCL2, FOS, JUN in kidney diseases, as well as the correlation between CCL2, FOS, JUN expression and renal function in the patients with IgAN. Results A total of 129 DEGs were obtained through comparing IgAN with healthy controls via the GSE115857 and GSE35487 datasets. Then, we screened out 24 immune-associated IgAN DEGs. CCL2, JUN, and FOS were identified as the top three hub genes, and they were all remarkably downregulated in IgAN. More importantly, CCL2, JUN, and FOS had a high accuracy [area under the curve (AUC) reached almost 1] in predicting IgAN, which could easily distinguish between IgAN patients and healthy individuals. Three distinct subgroups of IgAN were determined based on 24 immune-associated DEGs, with significant differences in the expression of CCL2, JUN, and FOS genes. Finally, CCL2, FOS, JUN were manifested a meaningful association with proteinuria, glomerular filtration rate (GFR), and serum creatinine level. Conclusion In summary, our study comprehensively uncovers that CCL2, JUN, and FOS may function as promising biomarkers for diagnosis of IgAN.
Collapse
Affiliation(s)
- Xue Zhou
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, China
- Department of Nephrology, Tianjin Haihe Hospital, Tianjin, China
| | - Ning Wang
- Tianjin Third Central Hospital, Tianjin, China
| | - Yuefeng Zhang
- Department of Nephrology, Tianjin Haihe Hospital, Tianjin, China
| | - Pei Yu
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, China
- *Correspondence: Pei Yu,
| |
Collapse
|
44
|
Lindblom J, Mohan C, Parodis I. Diagnostic, predictive and prognostic biomarkers in systemic lupus erythematosus: current insights. Curr Opin Rheumatol 2022; 34:139-149. [PMID: 35013077 DOI: 10.1097/bor.0000000000000862] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Biomarkers for diagnosis, monitoring and prognosis still constitute an unmet need for systemic lupus erythematosus (SLE). Focusing on recent findings, this review summarises the current landscape of biomarkers in lupus. RECENT FINDINGS Urine activated leukocyte cell adhesion molecule (ALCAM) exhibited good diagnostic ability in SLE and lupus nephritis (LN) whereas cerebrospinal fluid neutrophil gelatinase-associated lipocalin (NGAL) showed promise in neuropsychiatric SLE. Urine ALCAM, CD163 and vascular cell adhesion molecule 1 (VCAM-1) may be useful in surveillance of LN. Urine monocyte chemoattractant protein 1 was found to predict treatment response in SLE, and urine CD163 and NGAL treatment response in LN. Serum complement component 3 (C3) and urinary VCAM-1 have been reported to portend long-term renal prognosis in LN. SUMMARY NGAL holds promise as a versatile biomarker in SLE whereas urine ALCAM, CD163 and VCAM-1 displayed good performance as biomarkers in LN. The overall lack of concerted corroboration of leading candidates across multiple cohorts and diverse populations leaves the current biomarker landscape in SLE in an urgent need for further survey and systematic validation.
Collapse
Affiliation(s)
- Julius Lindblom
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Chandra Mohan
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
45
|
Li M, Yao B, Jing C, Chen H, Zhang Y, Zhou N. Engineering a G-quadruplex-based logic gate platform for sensitive assay of dual biomarkers of ovarian cancer. Anal Chim Acta 2022; 1198:339559. [DOI: 10.1016/j.aca.2022.339559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/06/2022] [Accepted: 01/26/2022] [Indexed: 11/24/2022]
|
46
|
Parodis I, Tamirou F, Houssiau FA. Treat-to-Target in Lupus Nephritis. What is the Role of the Repeat Kidney Biopsy? Arch Immunol Ther Exp (Warsz) 2022; 70:8. [PMID: 35147824 PMCID: PMC8837511 DOI: 10.1007/s00005-022-00646-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 10/12/2021] [Indexed: 01/14/2023]
Abstract
Kidney involvement, termed lupus nephritis (LN), develops in 35-60% of patients with systemic lupus erythematosus, often early during the disease course. When not treated promptly and efficiently, LN may lead to rapid and severe loss of kidney function, being the reason why it is considered one of the most severe lupus manifestations. Despite improved pharmacotherapy, 5-20% of LN patients develop end-stage kidney disease within ten years from the LN diagnosis. While the principal ground of LN therapy is prevention of renal function worsening, resembling a race against nephron loss, consensual agreement upon outcome measures and clinically meaningful short- and long-term targets of LN therapy have yet to be determined. Literature points to the importance of inclusion of tissue-based approaches in the determination of those targets, and evidence accumulates regarding the importance of per-protocol repeat kidney biopsies in the evaluation of the initial phase of therapy and prediction of long-term renal prognosis. The latter leads to the hypothesis that the information gleaned from repeat biopsies may contribute to optimised therapeutic decision making, and, therefore, increased probability to attain complete renal response in the short term, and a more favourable renal prognosis within a longer prospect. The multinational project ReBioLup was recently designed to serve as a key contributor to form evidence about the role of per-protocol repeat biopsies in a randomised fashion and aspires to unify the global LN community towards improved kidney and patient survival.
Collapse
Affiliation(s)
- Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, 171 76, Stockholm, Sweden. .,Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
| | - Farah Tamirou
- Pôle de Pathologies Rhumatismales Inflammatoires et Systémiques, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.,Rheumatology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Frédéric A Houssiau
- Pôle de Pathologies Rhumatismales Inflammatoires et Systémiques, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.,Rheumatology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
47
|
Vanarsa K, Sasidharan P, Duran V, Gokaraju S, Nidhi M, Louis Sam Titus ASC, Soomro S, Stock AD, Der E, Putterman C, Greenberg B, Mok CC, Hanly JG, Mohan C. Aptamer-based screen of Neuropsychiatric Lupus cerebrospinal fluid reveals potential biomarkers that overlap with the choroid plexus transcriptome. Arthritis Rheumatol 2022; 74:1223-1234. [PMID: 35099126 DOI: 10.1002/art.42080] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 12/28/2021] [Accepted: 01/27/2022] [Indexed: 11/07/2022]
Abstract
OBJECTIVES As no gold-standard diagnostic test exists for neuropsychiatric systemic lupus erythematosus (NPSLE), we executed a broad screen of NPSLE cerebrospinal fluid (CSF) using an aptamer-based platform. METHODS CSF were obtained from NPSLE patients and subjected to proteomic assay using the aptamer-based screen. Potential biomarkers were identified and validated in independent NPSLE cohorts in comparison with other neurological diseases. RESULTS 40 proteins out of 1129 screened were elevated in NPSLE CSF. By ELISA validation, CSF Angiostatin, α2-Macroglobulin, DAN, Fibronectin, HCC-1, IgM, Lipocalin 2, M-CSF and SERPING1 were significantly elevated in a predominantly Caucasian NPSLE cohort (n=24), compared to patients with other neurological diseases (n=54), with CSF IgM (AUC=0.95) and M-CSF (AUC=0.91) being the most discriminatory. In a second, Hong Kong NPSLE cohort, CSF IgM (AUC=0.78) and Lipocalin-2 (AUC=0.85) were the most discriminatory. Several CSF proteins exhibited high diagnostic specificity for NPSLE in both cohorts. Elevated CSF C3 was associated with acute confusional state. Eleven molecules elevated in NPSLE CSF exhibited concordant elevation in the choroid plexus, suggesting shared origins. CONCLUSIONS CSF Lipocalin-2, M-CSF, IgM and complement C3 emerge as promising CSF biomarkers of NPSLE with diagnostic potential.
Collapse
Affiliation(s)
- Kamala Vanarsa
- Department Biomedical Engineering, University of Houston, Houston, TX
| | | | - Valeria Duran
- Department Biomedical Engineering, University of Houston, Houston, TX
| | - Sirisha Gokaraju
- Department Biomedical Engineering, University of Houston, Houston, TX
| | - Malavika Nidhi
- Department Biomedical Engineering, University of Houston, Houston, TX
| | | | - Sanam Soomro
- Department Biomedical Engineering, University of Houston, Houston, TX
| | | | - Evan Der
- Albert Einstein College of Medicine, Bronx, NY
| | | | | | | | - John G Hanly
- Division of Rheumatology, Queen Elizabeth II Health Sciences Center and Dalhousie University Halifax, Nova Scotia, Canada
| | - Chandra Mohan
- Department Biomedical Engineering, University of Houston, Houston, TX
| |
Collapse
|
48
|
Chalmers SA, Ayilam Ramachandran R, Garcia SJ, Der E, Herlitz L, Ampudia J, Chu D, Jordan N, Zhang T, Parodis I, Gunnarsson I, Ding H, Shen N, Petri M, Mok CC, Saxena R, Polu KR, Connelly S, Ng CT, Mohan C, Putterman C. The CD6/ALCAM pathway promotes lupus nephritis via T cell-mediated responses. J Clin Invest 2022; 132:e147334. [PMID: 34981775 PMCID: PMC8718154 DOI: 10.1172/jci147334] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 11/10/2021] [Indexed: 12/18/2022] Open
Abstract
T cells are central to the pathogenesis of lupus nephritis (LN), a common complication of systemic lupus erythematosus (SLE). CD6 and its ligand, activated leukocyte cell adhesion molecule (ALCAM), are involved in T cell activation and trafficking. Previously, we showed that soluble ALCAM is increased in urine (uALCAM) of patients with LN, suggesting that this pathway contributes to disease. To investigate, uALCAM was examined in 1038 patients with SLE and LN from 5 ethnically diverse cohorts; CD6 and ALCAM expression was assessed in LN kidney cells; and disease contribution was tested via antibody blockade of CD6 in murine models of SLE and acute glomerulonephritis. Extended cohort analysis offered resounding validation of uALCAM as a biomarker that distinguishes active renal involvement in SLE, irrespective of ethnicity. ALCAM was expressed by renal structural cells whereas CD6 expression was exclusive to T cells, with elevated numbers of CD6+ and ALCAM+ cells in patients with LN. CD6 blockade in models of spontaneous lupus and immune-complex glomerulonephritis revealed significant decreases in immune cells, inflammatory markers, and disease measures. Our data demonstrate the contribution of the CD6/ALCAM pathway to LN and SLE, supporting its use as a disease biomarker and therapeutic target.
Collapse
Affiliation(s)
- Samantha A. Chalmers
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | | | - Sayra J. Garcia
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Evan Der
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Leal Herlitz
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | - Nicole Jordan
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Ting Zhang
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institute and Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Iva Gunnarsson
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institute and Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Huihua Ding
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nan Shen
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Ramesh Saxena
- Division of Nephrology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | | | | | | | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Chaim Putterman
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Research Institute, Galilee Medical Center, Nahariya, Israel
| |
Collapse
|
49
|
Lucarelli V, Colbert D, Li S, Cumming M, Linklater W, Mitchell J, Travas-Sejdic J, Kralicek A. Selection and characterization of DNA aptamers for the rat major urinary protein 13 (MUP13) as selective biorecognition elements for sensitive detection of rat pests. Talanta 2021; 240:123073. [PMID: 35026634 DOI: 10.1016/j.talanta.2021.123073] [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: 07/01/2021] [Revised: 11/07/2021] [Accepted: 11/14/2021] [Indexed: 10/19/2022]
Abstract
Among invasive mammalian predators, rats represent a major threat, endangering ecosystem functioning worldwide. After rat-control operations, detecting their continued presence or reinvasion requires more sensitive and lower cost detection technologies. Here, we develop a new sensing paradigm by using a specific rat urine biomarker (MUP13) to unambiguously signal the presence of rats. As the first step towards a new remote surveillance technology, aptamers were selected to MUP13 using the Flu-Mag SELEX method. Six aptamer candidates were initially screened by dot blot and two of them (Apt-2.5 and Apt-1.4) exhibited high affinity and specificity. Both aptamers were further characterized by bead-based assay to confirm affinity and selectivity. The lead aptamer candidates were then applied to fluorescence anisotropy (FA) and surface plasmon resonance (SPR)-based biosensor platforms, showing dissociation constants in the nanomolar range and high specificity towards their target. The SPR biosensor had limits of detection of 13.8 and 7.5 nM for Apt-2.5 and Apt-1.4, respectively, which are more than three orders of magnitude lower than the physiological concentrations found in rat urine. Selectivity of the aptamers, when comparing with other major urinary proteins, was excellent, indicating strong efficacy in specific detection of rats. In order to validate the aptamer Apt-2.5 for use with real world samples a FA-based assay was performed on a rat urine sample. The assay showed that the aptamer could detect recombinant MUP13 spiked in filtered urine and the natural MUP13 in unfiltered urine, as a first step into translation to real world application. These are the first known assays to detect and quantify a MUP biomarker of rats.
Collapse
Affiliation(s)
- Valentina Lucarelli
- The New Zealand Institute for Plant and Food Research Limited, Private Bag 92169, Auckland, 1142, New Zealand; Polymer Biointerface Centre, School of Chemical Sciences, The University of Auckland, Auckland, 1023, New Zealand; MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington, 6140, New Zealand
| | - Damon Colbert
- The New Zealand Institute for Plant and Food Research Limited, Private Bag 92169, Auckland, 1142, New Zealand
| | - Shiwei Li
- The New Zealand Institute for Plant and Food Research Limited, Private Bag 92169, Auckland, 1142, New Zealand
| | - Mathew Cumming
- The New Zealand Institute for Plant and Food Research Limited, Private Bag 92169, Auckland, 1142, New Zealand
| | - Wayne Linklater
- Department of Environmental Studies, California State University, Sacramento, California, USA
| | - John Mitchell
- The New Zealand Institute for Plant and Food Research Limited, Private Bag 92169, Auckland, 1142, New Zealand
| | - Jadranka Travas-Sejdic
- Polymer Biointerface Centre, School of Chemical Sciences, The University of Auckland, Auckland, 1023, New Zealand; MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington, 6140, New Zealand.
| | - Andrew Kralicek
- The New Zealand Institute for Plant and Food Research Limited, Private Bag 92169, Auckland, 1142, New Zealand.
| |
Collapse
|
50
|
Li X, Huang C, Zhang X, Yang T, Zuo S, Fu C, Zhang Y, Yang C, Chen L. Discovery of bladder cancer biomarkers in paired pre- and postoperative urine samples. Transl Androl Urol 2021; 10:3402-3414. [PMID: 34532265 PMCID: PMC8421825 DOI: 10.21037/tau-21-562] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Bladder cancer (BC), a common cancer of the urinary system, has a low mortality but an extremely high recurrence rate. Patients who have undergone initial surgical treatment often undergo frequent prognostic examinations with a substantial burden of discomfort and costs. Urine samples can reflect early disease processes in the urinary system and may be an excellent source of biomarkers. METHODS In the present study, we used the liquid chromatography with tandem mass spectrometry (LC-MS/MS) to perform proteomic analysis of pre- and postoperative urine samples from patients with stage III BC to identify biomarkers of cancer prognosis. Candidate biomarkers from proteomic analysis were simultaneously validated using western blotting in an independent cohort and immunohistochemical (IHC) staining, combined with gene expression data of BC samples in The Cancer Genome Atlas (TCGA). RESULTS The comparison of pre- and postoperative urine samples from the same patients led to the discovery of several significantly differentially expressed proteins, whose functions could be closely related to the occurrence and development of BC. We confirmed a representative group of candidate biomarker molecules, such as cadherin-related family member 2 (CDHR2), heat shock protein beta-1 (HSP27), and heterogeneous nuclear ribonucleoproteins A2/B1 (HNRNPA2B1). CONCLUSIONS The candidate biomarker molecules can distinguish between pre- and postoperative urine samples, and alterations in their expression levels are significantly associated with recurrence rates in patients with BC. Therefore, these molecules may become useful biomarkers for the monitoring and prognosis of BC.
Collapse
Affiliation(s)
- Xuechao Li
- Medical School of Chinese PLA, Beijing, China
- Department of Urology, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Chuanxi Huang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China
| | - Xueli Zhang
- Department of pathology, the Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Tao Yang
- Department of Urology, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Shidong Zuo
- Department of Urology, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Chengwei Fu
- Department of Urology, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Yongjie Zhang
- Department of Urology, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Chunyuan Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China
| | - Lijun Chen
- Department of Urology, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|