1
|
Wang B, Tan Z, She W, Wang X, Guan X, Tao Z, Guo F, Xu H, Deng Y. Characterizing Chemokine Signaling Pathways and Hub Genes in Calcium Oxalate-Induced Kidney Stone Formation: Insights from Rodent Models. Biochem Genet 2025:10.1007/s10528-025-11036-z. [PMID: 39893356 DOI: 10.1007/s10528-025-11036-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 01/12/2025] [Indexed: 02/04/2025]
Abstract
The predominant component of kidney stone is calcium oxalate monohydrate (COM), a fact widely acknowledged. Although rodent models are frequently used to induce calcium oxalate (CaOx) crystallization, further exploration of Randall's plaques (RPs) in these models is still needed. We first selected the GSE89028 and GSE75542 datasets from the Gene Expression Omnibus (GEO) database to identify commonly differentially expressed genes (co-DEGs). Based on co-DEGs, we conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses to identify significantly enriched pathways. Additionally, we performed Gene Set Enrichment Analysis (GSEA) to validate the enriched pathways. In order to identify hub genes, we established a network of protein-protein interactions (PPI). Finally, we conducted real-time PCR and Western blot to validate the findings from the bioinformatics analysis. We selected 28 co-DEGs from two datasets. The enrichment analysis using GO, KEGG, and GSEA revealed significant enrichment of chemokine-related signaling pathways. The histogram analysis showed that three chemokine factor-related genes were involved in multiple pathways. We used Cytohubba to confirm the presence of three hub genes. Subsequently, analysis of external datasets and quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot demonstrated significant upregulation of CCL2, CXCL1, and CXCL2 in HK-2 cells following CaOx treatment compared to the control group (p < 0.05). Our study demonstrated that upon stimulation by CaOx, renal tubular epithelial cells release chemokines, including CCL2, CXCL1, and CXCL2. This release of chemokines is accompanied by the activation of signaling pathways such as TNF and IL-17. These findings may provide new directions for future research on Kidney Stone Disease.
Collapse
Affiliation(s)
- Boqiang Wang
- Department of Urology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhenkun Tan
- Department of Urology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Wusheng She
- Department of Urology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xiang Wang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Xiaofeng Guan
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Zhiwei Tao
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Fuyou Guo
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Hua Xu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yaoliang Deng
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China.
| |
Collapse
|
2
|
Zhang T, Widdop RE, Ricardo SD. Transition from acute kidney injury to chronic kidney disease: mechanisms, models, and biomarkers. Am J Physiol Renal Physiol 2024; 327:F788-F805. [PMID: 39298548 DOI: 10.1152/ajprenal.00184.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/15/2024] [Accepted: 09/01/2024] [Indexed: 09/22/2024] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are increasingly recognized as interconnected conditions with overlapping pathophysiological mechanisms. This review examines the transition from AKI to CKD, focusing on the molecular mechanisms, animal models, and biomarkers essential for understanding and managing this progression. AKI often progresses to CKD due to maladaptive repair processes, persistent inflammation, and fibrosis, with both conditions sharing common pathways involving cell death, inflammation, and extracellular matrix (ECM) deposition. Current animal models, including ischemia-reperfusion injury (IRI) and nephrotoxic damage, help elucidate these mechanisms but have limitations in replicating the complexity of human disease. Emerging biomarkers such as kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and soluble tumor necrosis factor receptors (TNFRs) show promise in early detection and monitoring of disease progression. This review highlights the need for improved animal models and biomarker validation to better mimic human disease and enhance clinical translation. Advancing our understanding of the AKI-to-CKD transition through targeted therapies and refined research approaches holds the potential to significantly improve patient outcomes.
Collapse
Affiliation(s)
- Tingfang Zhang
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Robert E Widdop
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Sharon D Ricardo
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
3
|
Bondi CD, Hartman HL, Rush BM, Tan RJ. Podocyte-Specific Deletion of MCP-1 Fails to Protect against Angiotensin II- or Adriamycin-Induced Glomerular Disease. Int J Mol Sci 2024; 25:4987. [PMID: 38732210 PMCID: PMC11084322 DOI: 10.3390/ijms25094987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Investigating the role of podocytes in proteinuric disease is imperative to address the increasing global burden of chronic kidney disease (CKD). Studies strongly implicate increased levels of monocyte chemoattractant protein-1 (MCP-1/CCL2) in proteinuric CKD. Since podocytes express the receptor for MCP-1 (i.e., CCR2), we hypothesized that podocyte-specific MCP-1 production in response to stimuli could activate its receptor in an autocrine manner, leading to further podocyte injury. To test this hypothesis, we generated podocyte-specific MCP-1 knockout mice (Podo-Mcp-1fl/fl) and exposed them to proteinuric injury induced by either angiotensin II (Ang II; 1.5 mg/kg/d, osmotic minipump) or Adriamycin (Adr; 18 mg/kg, intravenous bolus). At baseline, there were no between-group differences in body weight, histology, albuminuria, and podocyte markers. After 28 days, there were no between-group differences in survival, change in body weight, albuminuria, kidney function, glomerular injury, and tubulointerstitial fibrosis. The lack of protection in the knockout mice suggests that podocyte-specific MCP-1 production is not a major contributor to either Ang II- or Adr-induced glomerular disease, implicating that another cell type is the source of pathogenic MCP-1 production in CKD.
Collapse
Affiliation(s)
- Corry D. Bondi
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 152671, USA; (H.L.H.); (B.M.R.); (R.J.T.)
| | | | | | | |
Collapse
|
4
|
Hüsing AM, Wulfmeyer VC, Gaedcke S, Fleig SV, Rong S, DeLuca D, Haller H, Schmitt R, von Vietinghoff S. Myeloid CCR2 Promotes Atherosclerosis after AKI. J Am Soc Nephrol 2022; 33:1487-1500. [PMID: 35537780 PMCID: PMC9342642 DOI: 10.1681/asn.2022010048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 04/10/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The risk of cardiovascular events rises after AKI. Leukocytes promote atherosclerotic plaque growth and instability. We established a model of enhanced remote atherosclerosis after renal ischemia-reperfusion (IR) injury and investigated the underlying inflammatory mechanisms. METHODS Atherosclerotic lesions and inflammation were investigated in native and bone marrow-transplanted LDL receptor-deficient (LDLr-/- ) mice after unilateral renal IR injury using histology, flow cytometry, and gene expression analysis. RESULTS Aortic root atherosclerotic lesions were significantly larger after renal IR injury than in controls. A gene expression screen revealed enrichment for chemokines and their cognate receptors in aortas of IR-injured mice in early atherosclerosis, and of T cell-associated genes in advanced disease. Confocal microscopy revealed increased aortic macrophage proximity to T cells. Differential aortic inflammatory gene regulation in IR-injured mice largely paralleled the pattern in the injured kidney. Single-cell analysis identified renal cell types that produced soluble mediators upregulated in the atherosclerotic aorta. The analysis revealed a marked early increase in Ccl2, which CCR2+ myeloid cells mainly expressed. CCR2 mediated myeloid cell homing to the post-ischemic kidney in a cell-individual manner. Reconstitution with Ccr2-/- bone marrow dampened renal post-ischemic inflammation, reduced aortic Ccl2 and inflammatory macrophage marker CD11c, and abrogated excess aortic atherosclerotic plaque formation after renal IR. CONCLUSIONS Our data introduce an experimental model of remote proatherogenic effects of renal IR and delineate myeloid CCR2 signaling as a mechanistic requirement. Monocytes should be considered as mobile mediators when addressing systemic vascular sequelae of kidney injury.
Collapse
Affiliation(s)
- Anne M. Hüsing
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Hannover, Germany
| | - Vera C. Wulfmeyer
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Hannover, Germany
| | - Svenja Gaedcke
- German Centre for Lung Research, Hannover Medical School, Hannover, Germany
| | - Susanne V. Fleig
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Hannover, Germany,Nephrology Section, Medical Clinic 1, University Hospital Bonn, Rheinische Friedrich Wilhelm University of Bonn, Bonn, Germany
| | - Song Rong
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Hannover, Germany
| | - David DeLuca
- German Centre for Lung Research, Hannover Medical School, Hannover, Germany
| | - Hermann Haller
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Hannover, Germany
| | - Roland Schmitt
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Hannover, Germany
| | - Sibylle von Vietinghoff
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Hannover, Germany,Nephrology Section, Medical Clinic 1, University Hospital Bonn, Rheinische Friedrich Wilhelm University of Bonn, Bonn, Germany
| |
Collapse
|
5
|
Vehicle emissions-exposure alters expression of systemic and tissue-specific components of the renin-angiotensin system and promotes outcomes associated with cardiovascular disease and obesity in wild-type C57BL/6 male mice. Toxicol Rep 2021; 8:846-862. [PMID: 33948438 PMCID: PMC8080412 DOI: 10.1016/j.toxrep.2021.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/03/2021] [Accepted: 04/09/2021] [Indexed: 12/24/2022] Open
Abstract
Vehicle emission-exposure increases systemic and adipose renin-angiotensin signaling. Emission-exposure promotes renal, vascular, and adipocyte AT1 receptor expression. Diet and emission-exposure are associated with adipocyte hypertrophy and weight gain. Emission-exposure promotes expression of adipokines and adipose inflammatory factors. High-fat diet promotes an obese adipose phenotype, exacerbated by emission-exposure.
Exposure to air pollution from traffic-generated sources is known to contribute to the etiology of inflammatory diseases, including cardiovascular disease (CVD) and obesity; however, the signaling pathways involved are still under investigation. Dysregulation of the renin-angiotensin system (RAS) can contribute to CVD and alter lipid storage and inflammation in adipose tissue. Our previous exposure studies revealed that traffic-generated emissions increase RAS signaling, further exacerbated by a high-fat diet. Thus, we investigated the hypothesis that exposure to engine emissions increases systemic and local adipocyte RAS signaling, promoting the expression of factors involved in CVD and obesity. Male C57BL/6 mice (6–8 wk old) were fed either a high-fat (HF, n = 16) or low-fat (LF, n = 16) diet, beginning 30d prior to exposures, and then exposed via inhalation to either filtered air (FA, controls) or a mixture of diesel engine + gasoline engine vehicle emissions (MVE: 100 μg PM/m3) via whole-body inhalation for 6 h/d, 7 d/wk, 30d. Endpoints were assessed via immunofluorescence and RT-qPCR. MVE-exposure promoted vascular adhesion factors (VCAM-1, ICAM-1) expression, monocyte/macrophage sequestration, and oxidative stress in the vasculature, associated with increased angiotensin II receptor type 1 (AT1) expression. In the kidney, MVE-exposure promoted the expression of renin, AT1, and AT2 receptors. In adipose tissue, both HF-diet and MVE-exposure mediated increased epididymal fat pad weight and adipocyte hypertrophy, associated with increased angiotensinogen and AT1 receptor expression; however, these outcomes were further exacerbated in the MVE + HF group. MVE-exposure also induced inflammation, monocyte chemoattractant protein (MCP)-1, and leptin, while reducing insulin receptor and glucose transporter, GLUT4, expression in adipose tissue. Our results indicate that MVE-exposure promotes systemic and local adipose RAS signaling, associated with increased expression of factors contributing to CVD and obesity, further exacerbated by HF diet consumption.
Collapse
Key Words
- ACE, angiotensin converting enzyme
- AGT, angiotensinogen
- AT1, angiotensin II receptor subtype 1
- AT2, angiotensin II receptor subtype 2
- Adipose
- Air pollution
- Ang II, angiotensin II
- CVD
- CVD, cardiovascular disease
- DHE, dihydroethidium
- FA, filtered air (controls)
- GLUT-4, glucose transporter type 4
- HF, high-fat diet
- ICAM-1, intracellular adhesion molecule-1
- IL-6, interleukin-6
- IL-β, interleukin beta
- IR, insulin receptor
- LDL, low density lipoprotein
- LF, low-fat diet
- LOX-1, lectin-like oxidized low-density lipoprotein receptor
- MCP-1, monocyte chemoattractant protein-1
- MOMA-2, anti-monocyte + macrophage antibody
- MVE, mixed gasoline and diesel vehicle emissions
- Obesity
- PM, particulate matter
- RAS, renin-angiotensin system
- ROS, reactive oxygen species
- Renin-angiotensin system
- T2D, type 2 diabetes
- TNF-α, tumor necrosis factor alpha
- VCAM-1, vascular cell adhesion molecule-1
- vWF, Von Willebrand factor
Collapse
|
6
|
Petreski T, Piko N, Ekart R, Hojs R, Bevc S. Review on Inflammation Markers in Chronic Kidney Disease. Biomedicines 2021; 9:182. [PMID: 33670423 PMCID: PMC7917900 DOI: 10.3390/biomedicines9020182] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease (CKD) is one of the major health problems of the modern age. It represents an important public health challenge with an ever-lasting rising prevalence, which reached almost 700 million by the year 2017. Therefore, it is very important to identify patients at risk for CKD development and discover risk factors that cause the progression of the disease. Several studies have tackled this conundrum in recent years, novel markers have been identified, and new insights into the pathogenesis of CKD have been gained. This review summarizes the evidence on markers of inflammation and their role in the development and progression of CKD. It will focus primarily on cytokines, chemokines, and cell adhesion molecules. Nevertheless, further large, multicenter studies are needed to establish the role of these markers and confirm possible treatment options in everyday clinical practice.
Collapse
Affiliation(s)
- Tadej Petreski
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia; (T.P.); (N.P.); (R.H.)
- Department of Internal Medicine and Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
| | - Nejc Piko
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia; (T.P.); (N.P.); (R.H.)
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia;
| | - Robert Ekart
- Department of Internal Medicine and Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia;
| | - Radovan Hojs
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia; (T.P.); (N.P.); (R.H.)
- Department of Internal Medicine and Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
| | - Sebastjan Bevc
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia; (T.P.); (N.P.); (R.H.)
- Department of Internal Medicine and Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
| |
Collapse
|
7
|
Roles Played by Biomarkers of Kidney Injury in Patients with Upper Urinary Tract Obstruction. Int J Mol Sci 2020; 21:ijms21155490. [PMID: 32752030 PMCID: PMC7432915 DOI: 10.3390/ijms21155490] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/25/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022] Open
Abstract
Partial or complete obstruction of the urinary tract is a common and challenging urological condition caused by a variety of conditions, including ureteral calculi, ureteral pelvic junction obstruction, ureteral stricture, and malignant ureteral obstruction. The condition, which may develop in patients of any age, induces tubular and interstitial injury followed by inflammatory cell infiltration and interstitial fibrosis, eventually impairing renal function. The serum creatinine level is commonly used to evaluate global renal function but is not sensitive to early changes in the glomerular filtration rate and unilateral renal damage. Biomarkers of acute kidney injury are useful for the early detection and monitoring of kidney injury induced by upper urinary tract obstruction. These markers include levels of neutrophil gelatinase-associated lipocalin (NGAL), monocyte chemotactic protein-1, kidney injury molecule 1, N-acetyl-b-D-glucosaminidase, and vanin-1 in the urine and serum NGAL and cystatin C concentrations. This review summarizes the pathophysiology of kidney injury caused by upper urinary tract obstruction, the roles played by emerging biomarkers of obstructive nephropathy, the mechanisms involved, and the clinical utility and limitations of the biomarkers.
Collapse
|
8
|
ARFIAN NUR, BUDIHARJO SANTOSA, WIBISONO DIANPRASETYO, SETYANINGSIH WIWITANANDAWAHYU, ROMI MUHAMMADMANSYUR, SAPUTRI RAMADHEALAILAAFIFAANNURWILLYA, ROFIAH EDREANAKHUSNUR, RAHMANTI TRITA, AGUSTIN MAULIDINA, SARI DWICAHYANIRATNA. Vitamin D Ameliorates Kidney Ischemia Reperfusion Injury via Reduction of Inflammation and Myofibroblast Expansion. THE KOBE JOURNAL OF MEDICAL SCIENCES 2020; 65:E138-E143. [PMID: 32201429 PMCID: PMC7447095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/04/2019] [Indexed: 06/10/2023]
Abstract
The incidence rate of Acute Kidney Injury (AKI) gets escalated each year. Kidney ischemia/reperfusion injury (IR injury) is the main cause of AKI after major cardiovascular surgery, trauma, or kidney transplantation. Reperfusion is considered essential for ischemic tissue. However, the evidence revealed that reperfusion itself has impact in cellular destruction. Vitamin D is not only known as calcium regulating hormone, but also as renoprotective agent. This study aimed to investigate the effect of vitamin D treatment on kidney IR injury in mice. Kidney IR injury was performed using 30 minutes of bilateral clamping of renal pedicles, then released in male Swiss Webster mice (3 months, 30-40 grams, n=20), which were divided into three groups: sham operation (SO) group, IR injury (IRI) group, and IR injury with 0.25 µg/ kg body weight of vitamin D treatment (IR7+VD). Mice were terminated at day 7 post operation, kidneys were harvested and used for paraffin making, immunostaining and RNA extraction. Tubular injury was quantified based on Periodic Acid-Schiff's (PAS) staining. Immunostaining was done for quantification of macrophage (CD68) and myofibroblast (α-SMA). Reverse Transcriptase PCR (RT-PCR) was done to examine Monocyte Chemoattractant Protein-1 (MCP-1) and Toll-like Receptor 4 (TLR4) mRNA expression. Kidney IR injury induced significant increase of tubular injury, which was associated with higher myofibroblast and macrophage number. Meanwhile, Vitamin D treatment significantly reduced tubular, myofibroblast and macrophage number. RTPCR revealed reduction of TLR4 and MCP-1 mRNA expressions after Vitamin D treatment (p<0.05 vs IR group). Vitamin D ameliorates kidney IR injury through reducing inflammation and myofibroblast formation.
Collapse
Affiliation(s)
- NUR ARFIAN
- Department of Anatomy, Faculty of Medicine, Public Health and Nursing Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - SANTOSA BUDIHARJO
- Department of Anatomy, Faculty of Medicine, Public Health and Nursing Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - DIAN PRASETYO WIBISONO
- Department of Anatomy, Faculty of Medicine, Public Health and Nursing Universitas Gadjah Mada, Yogyakarta, Indonesia
| | | | - MUHAMMAD MANSYUR ROMI
- Department of Anatomy, Faculty of Medicine, Public Health and Nursing Universitas Gadjah Mada, Yogyakarta, Indonesia
| | | | - EDREANA KHUSNUR ROFIAH
- Department of Anatomy, Faculty of Medicine, Public Health and Nursing Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - TRITA RAHMANTI
- Department of Anatomy, Faculty of Medicine, Public Health and Nursing Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - MAULIDINA AGUSTIN
- Department of Anatomy, Faculty of Medicine, Public Health and Nursing Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - DWI CAHYANI RATNA SARI
- Department of Anatomy, Faculty of Medicine, Public Health and Nursing Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
9
|
IL-20 in Acute Kidney Injury: Role in Pathogenesis and Potential as a Therapeutic Target. Int J Mol Sci 2020; 21:ijms21031009. [PMID: 32028746 PMCID: PMC7037658 DOI: 10.3390/ijms21031009] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/07/2020] [Accepted: 01/10/2020] [Indexed: 02/06/2023] Open
Abstract
Acute kidney injury (AKI) causes over 1 million deaths worldwide every year. AKI is now recognized as a major risk factor in the development and progression of chronic kidney disease (CKD). Diabetes is the main cause of CKD as well. Renal fibrosis and inflammation are hallmarks in kidney diseases. Various cytokines contribute to the progression of renal diseases; thus, many drugs that specifically block cytokine function are designed for disease amelioration. Numerous studies showed IL-20 functions as a pro-inflammatory mediator to regulate cytokine expression in several inflammation-mediated diseases. In this review, we will outline the effects of pro-inflammatory cytokines in the pathogenesis of AKI and CKD. We also discuss the role of IL-20 in kidney diseases and provide a potential therapeutic approach of IL-20 blockade for treating renal diseases.
Collapse
|
10
|
Tajima S, Yamamoto N, Masuda S. Clinical prospects of biomarkers for the early detection and/or prediction of organ injury associated with pharmacotherapy. Biochem Pharmacol 2019; 170:113664. [PMID: 31606409 DOI: 10.1016/j.bcp.2019.113664] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/08/2019] [Indexed: 12/31/2022]
Abstract
Several biomarkers are used to monitor organ damage caused by drug toxicity. Traditional markers of kidney function, such as serum creatinine and blood urea nitrogen are commonly used to estimate glomerular filtration rate. However, these markers have several limitations including poor specificity and sensitivity. A number of serum and urine biomarkers have recently been described to detect kidney damage caused by drugs such as cisplatin, gentamicin, vancomycin, and tacrolimus. Neutrophil gelatinase-associated lipocalin (NGAL), liver-type fatty acid-binding protein (L-FABP), kidney injury molecule-1 (KIM-1), monocyte chemotactic protein-1 (MCP-1), and cystatin C have been identified as biomarkers for early kidney damage. Hy's Law is widely used as to predict a high risk of severe drug-induced liver injury caused by drugs such as acetaminophen. Recent reports have indicated that glutamate dehydrogenase (GLDH), high-mobility group box 1 (HMGB-1), Keratin-18 (k18), MicroRNA-122 and ornithine carbamoyltransferase (OCT) are more sensitive markers of hepatotoxicity compared to the traditional markers including the blood levels of amiotransferases and total bilirubin. Additionally, the rapid development of proteomic technologies in biofluids and tissue provides a new multi-marker panel, leading to the discovery of more sensitive biomarkers. In this review, an update topics of biomarkers for the detection of kidney or liver injury associated with pharmacotherapy.
Collapse
Affiliation(s)
- Soichiro Tajima
- Department of Pharmacy, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Nanae Yamamoto
- Department of Pharmacy, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Satohiro Masuda
- Department of Pharmacy, Kyushu University Hospital, Fukuoka 812-8582, Japan; Department of Clinical Pharmacology and Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Department of Pharmacy, International University of Health and Welfare Narita Hospital, Japan; Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, International University of Health and Welfare Narita Hospital, Japan.
| |
Collapse
|
11
|
Murinello S, Usui Y, Sakimoto S, Kitano M, Aguilar E, Friedlander HM, Schricker A, Wittgrove C, Wakabayashi Y, Dorrell MI, Westenskow PD, Friedlander M. miR-30a-5p inhibition promotes interaction of Fas + endothelial cells and FasL + microglia to decrease pathological neovascularization and promote physiological angiogenesis. Glia 2019; 67:332-344. [PMID: 30484883 PMCID: PMC6349526 DOI: 10.1002/glia.23543] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 09/05/2018] [Accepted: 09/05/2018] [Indexed: 12/18/2022]
Abstract
Ischemia-induced angiogenesis contributes to various neuronal and retinal diseases, and often results in neurodegeneration and visual impairment. Current treatments involve the use of anti-VEGF agents but are not successful in all cases. In this study we determined that miR-30a-5p is another important mediator of retinal angiogenesis. Using a rodent model of ischemic retinopathy, we show that inhibiting miR-30a-5p reduces neovascularization and promotes tissue repair, through modulation of microglial and endothelial cell cross-talk. miR-30a-5p inhibition results in increased expression of the death receptor Fas and CCL2, to decrease endothelial cell survival and promote microglial migration and phagocytic function in focal regions of ischemic injury. Our data suggest that miR-30a-5p inhibition accelerates tissue repair by enhancing FasL-Fas crosstalk between microglia and endothelial cells, to promote endothelial cell apoptosis and removal of dead endothelial cells. Finally, we found that miR-30a levels were increased in the vitreous of patients with proliferative diabetic retinopathy. Our study identifies a role for miR-30a in the pathogenesis of neovascular retinal disease by modulating microglial and endothelial cell function, and suggests it may be a therapeutic target to treat ischemia-mediated conditions.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Apoptosis/drug effects
- Apoptosis/genetics
- Cell Line, Transformed
- Cell Proliferation/drug effects
- Cell Proliferation/genetics
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Disease Models, Animal
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/genetics
- Humans
- Lectins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Microglia/metabolism
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/metabolism
- Neovascularization, Physiologic/drug effects
- Neovascularization, Physiologic/physiology
- RNA Interference/physiology
- RNA, Messenger/metabolism
- fas Receptor/metabolism
Collapse
Affiliation(s)
- Salome Murinello
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Yoshihiko Usui
- Department of Ophthalmology, Tokyo Medical University, Tokyo Japan
| | - Susumu Sakimoto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- Present address: Department of Ophthalmology, Osaka Medical University, Osaka, Japan
| | - Maki Kitano
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Edith Aguilar
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - H. Maura Friedlander
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Amelia Schricker
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Carli Wittgrove
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | | | - Michael I. Dorrell
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- The Lowy Medical Research Institute, La Jolla, CA
- Department of Biology, Point Loma Nazarene University, San Diego, CA
| | - Peter D. Westenskow
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- Present address: Department of Ophthalmology, Baylor College of Medicine, Houston, TX
| | - Martin Friedlander
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- The Lowy Medical Research Institute, La Jolla, CA
| |
Collapse
|
12
|
Kashyap S, Osman M, Ferguson CM, Nath MC, Roy B, Lien KR, Nath KA, Garovic VD, Lerman LO, Grande JP. Ccl2 deficiency protects against chronic renal injury in murine renovascular hypertension. Sci Rep 2018; 8:8598. [PMID: 29872089 PMCID: PMC5988825 DOI: 10.1038/s41598-018-26870-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/22/2018] [Indexed: 01/11/2023] Open
Abstract
Inflammation plays an important role in the pathogenesis of renal and cardiovascular disease in renovascular hypertension (RVH). Ccl2 is an important mediator of inflammation, and is induced within 24 hours following surgery to establish RVH in the murine 2 kidney 1 clip model, a time prior to onset of interstitial inflammation, fibrosis, or tubular atrophy. We tested the hypothesis that Ccl2 deficiency protects the stenotic kidney (STK) from development of chronic renal damage in mice with renovascular hypertension due to renal artery stenosis (RAS). RAS surgery was performed on wild type (WT) and Ccl2 knock out (KO) mice; animals were studied for four weeks. Renal blood flow was reduced to similar extent in both WT and Ccl2 KO mice with RVH. Perfusion of the stenotic kidney was significantly reduced in Ccl2 KO mice as assessed by magnetic resonance imaging (MRI). Stenotic kidney volume in WT, but not in Ccl2 KO mice, was significantly reduced following surgery. Cortical hypoxia was observed in the stenotic kidney of Ccl2 KO mice, as assessed by blood oxygen level-dependent MRI (BOLD-MRI). Ccl2 KO mice showed less cortical atrophy than WT RAS mice. Ccl2 deficiency reduced the number of infiltrating mononuclear cells and expression of Ccl5, Ccl7, Ccl8, Ccr2 and Cd206. We conclude that Ccl2 is a critical mediator of chronic renal injury in RVH.
Collapse
Affiliation(s)
- Sonu Kashyap
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, MN, USA
| | - Mazen Osman
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Meryl C Nath
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, MN, USA
| | - Bhaskar Roy
- Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Karen R Lien
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, MN, USA
| | - Karl A Nath
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Vesna D Garovic
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Lilach O Lerman
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Joseph P Grande
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, MN, USA. .,Division of Nephrology & Hypertension, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
13
|
Morozov D, Morozova O, Pervouchine D, Severgina L, Tsyplakov A, Zakharova N, Sushentsev N, Maltseva L, Budnik I. Hypoxic renal injury in newborns with abdominal compartment syndrome (clinical and experimental study). Pediatr Res 2018; 83:520-526. [PMID: 29053704 DOI: 10.1038/pr.2017.263] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 09/29/2017] [Indexed: 12/13/2022]
Abstract
BackgroundSurgical treatment for gastroschisis and congenital diaphragmatic hernia (CDH) commonly leads to abdominal compartment syndrome (ACS) associated with hypoxic renal injury. We hypothesized that measurement of urinary and serum concentrations of vascular endothelial growth factor (VEGF), π-glutathione S-transferase (π-GST), and monocyte chemoattractant protein-1 (MCP-1) may serve for noninvasive detection of hypoxic renal injury in such patients.MethodsIntra-abdominal pressure (IAP), renal excretory function, and the biomarker levels were analyzed before, 4, and 10 days after surgery. Association between the biomarker levels and renal histology was investigated using an original model of ACS in newborn rats.ResultsFour days after surgery, IAP increased, renal excretory function decreased, and the levels of VEGF, π-GST, and MCP-1 increased, indicating renal injury. Ten days after surgery, IAP partially decreased, renal excretory function completely restored, but the biomarker levels remained elevated, suggesting the ongoing kidney injury. In the model of ACS, increase in the biomarker levels was associated with progressing kidney morphological alteration.ConclusionSurgical treatment for gastroschisis and CDH is associated with prolonged hypoxic kidney injury despite complete restoration of renal excretory function. Follow-up measurement of VEGF, π-GST, and MCP-1 levels may provide a better tool for noninvasive assessment of renal parenchyma in newborns with ACS.
Collapse
Affiliation(s)
- Dmitry Morozov
- Department of Pediatric Surgery, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Olga Morozova
- Department of Pathophysiology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Dmitri Pervouchine
- Center for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Lubov Severgina
- Department of Pathological Anatomy, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alexei Tsyplakov
- Research Institute for Fundamental and Clinical Uronephrology, Saratov State Medical University n. a. V. I. Razumovsky, Saratov, Russia
| | - Natalya Zakharova
- Research Institute for Fundamental and Clinical Uronephrology, Saratov State Medical University n. a. V. I. Razumovsky, Saratov, Russia
| | - Nikita Sushentsev
- Department of Pathophysiology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Larisa Maltseva
- Department of Pathophysiology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ivan Budnik
- Department of Pathophysiology, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
14
|
Mansour SG, Verma G, Pata RW, Martin TG, Perazella MA, Parikh CR. Kidney Injury and Repair Biomarkers in Marathon Runners. Am J Kidney Dis 2017; 70:252-261. [PMID: 28363731 PMCID: PMC5526736 DOI: 10.1053/j.ajkd.2017.01.045] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/11/2017] [Indexed: 01/05/2023]
Abstract
BACKGROUND Investigation into strenuous activity and kidney function has gained interest given increasing marathon participation. STUDY DESIGN Prospective observational study. SETTING & PARTICIPANTS Runners participating in the 2015 Hartford Marathon. PREDICTOR Completing a marathon. OUTCOMES Acute kidney injury (AKI) as defined by AKI Network (AKIN) criteria. Stage 1 AKI was defined as 1.5- to 2-fold or 0.3-mg/dL increase in serum creatinine level within 48 hours of day 0 and stage 2 was defined as a more than 2- to 3-fold increase in creatinine level. Microscopy score was defined by the number of granular casts and renal tubular epithelial cells. MEASUREMENTS Samples were collected 24 hours premarathon (day 0), immediately postmarathon (day 1), and 24 hours postmarathon (day 2). Measurements of serum creatinine, creatine kinase, and urine albumin were completed, as well as urine microscopy analysis. 6 injury urine biomarkers (IL-6, IL-8, IL-18, kidney injury molecule 1, neutrophil gelatinase-associated lipocalin, and tumor necrosis factor α) and 2 repair urine biomarkers (YKL-40 and monocyte chemoattractant protein 1) were measured. RESULTS 22 marathon runners were included. Mean age was 44 years and 41% were men. 82% of runners developed an increase in creatinine level equivalent to AKIN-defined AKI stages 1 and 2. 73% had microscopy diagnoses of tubular injury. Serum creatinine, urine albumin, and injury and repair biomarker levels peaked on day 1 and were significantly elevated compared to day 0 and day 2. Serum creatine kinase levels continued to significantly increase from day 0 to day 2. LIMITATIONS Small sample size and limited clinical data available at all time points. CONCLUSIONS Marathon runners developed AKI and urine sediment diagnostic of tubular injury. An increase in injury and repair biomarker levels suggests structural damage to renal tubules occurring after marathon. The results of our study should be validated in larger cohorts with longer follow-up of kidney function.
Collapse
Affiliation(s)
- Sherry G Mansour
- Program of Applied Translational Research, Department of Medicine, New Haven, CT; Section of Nephrology, Yale University School of Medicine, New Haven, CT
| | - Gagan Verma
- Program of Applied Translational Research, Department of Medicine, New Haven, CT
| | | | | | - Mark A Perazella
- Section of Nephrology, Yale University School of Medicine, New Haven, CT; Veterans Affairs Connecticut Healthcare System, New Haven, CT
| | - Chirag R Parikh
- Program of Applied Translational Research, Department of Medicine, New Haven, CT; Section of Nephrology, Yale University School of Medicine, New Haven, CT; Veterans Affairs Connecticut Healthcare System, New Haven, CT.
| |
Collapse
|
15
|
Mansour SG, Puthumana J, Reese PP, Hall IE, Doshi MD, Weng FL, Schröppel B, Thiessen-Philbrook H, Bimali M, Parikh CR. Associations between Deceased-Donor Urine MCP-1 and Kidney Transplant Outcomes. Kidney Int Rep 2017; 2:749-758. [PMID: 28730184 PMCID: PMC5512592 DOI: 10.1016/j.ekir.2017.03.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Introduction Existing methods to predict recipient allograft function during deceased-donor kidney procurement are imprecise. Understanding the potential renal reparative role for monocyte chemoattractant protein-1 (MCP-1), a cytokine involved in macrophage recruitment after injury, might help to predict allograft outcomes. Methods We conducted a substudy of the multicenter prospective Deceased Donor Study cohort that evaluated deceased kidney donors from 5 organ procurement organizations from May 2010 to December 2013. We measured urine MCP-1 (uMCP-1) concentrations from donor samples collected at nephrectomy to determine associations with donor acute kidney injury (AKI), recipient delayed graft function (DGF), 6-month estimated glomerular filtration rate (eGFR), and graft failure. We also assessed perfusate MCP-1 concentrations from pumped kidneys for associations with DGF and 6-month eGFR. Results AKI occurred in 111 donors (9%). The median (interquartile range) uMCP-1 concentration was higher in donors with AKI compared with donors without AKI (1.35 [0.41–3.93] ng/ml vs. 0.32 [0.11–0.80] ng/ml, P < 0.001). DGF occurred in 756 recipients (31%), but uMCP-1 was not independently associated with DGF. Higher donor uMCP-1 concentrations were independently associated with a higher 6-month eGFR in those without DGF (0.77 [0.10–1.45] ml/min per 1.73 m2 per doubling of uMCP1). However, there were no independent associations between uMCP-1 and graft failure over a median follow-up of ∼2 years. Lastly, perfusate MCP-1 concentrations significantly increased during pump perfusion but were not associated with DGF or 6-month eGFR. Discussion Donor uMCP-1 concentrations were modestly associated with higher recipient 6-month eGFR in those without DGF. However, the results suggest that donor uMCP-1 has minimal clinical utility given no associations with graft failure.
Collapse
Affiliation(s)
- S G Mansour
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, CT.,Section of Nephrology, Yale University School of Medicine, New Haven, CT
| | - J Puthumana
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, CT
| | - P P Reese
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - I E Hall
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT
| | | | - F L Weng
- Saint Barnabas Medical Center, Livingston, NJ
| | - B Schröppel
- Section of Nephrology, University Hospital, Ulm, Germany
| | - H Thiessen-Philbrook
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, CT
| | - M Bimali
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, CT
| | - C R Parikh
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, CT.,Section of Nephrology, Yale University School of Medicine, New Haven, CT.,Veterans Affairs Connecticut Healthcare System, New Haven, CT
| |
Collapse
|
16
|
CCR2 contributes to the recruitment of monocytes and leads to kidney inflammation and fibrosis development. Inflammopharmacology 2017; 26:403-411. [PMID: 28168553 DOI: 10.1007/s10787-017-0317-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 01/23/2017] [Indexed: 12/24/2022]
Abstract
Chemokines are a large family of proteins that, once associated to its receptor on leukocytes, stimulate their movement and migration from blood to tissues. Once in the tissue, immune cells trigger inflammation that, when uncontrolled, leads to fibrosis development. Among the immune cells, macrophages take a special role in fibrosis formation, since macrophage depletion reflects less collagen deposition. The majority of tissue macrophages is derived from monocytes, especially monocytes expressing the chemokine receptor CCR2. Here, we investigated the role of infiltrating CCR2+ cells in the development of fibrosis, and specifically, the dynamic of infiltration of these cells into kidneys under chronic obstructive lesion. Using liposome-encapsulated clodronate, we observed that macrophage depletion culminated in less collagen deposition and reduced chemokines milieu that were released in the damaged kidney after obstructive nephropathy. We also obstructed the kidneys of CCL3-/-, CCR2-/-, CCR4-/-, CCR5-/-, and C57BL/6 mice and we found that among all animals, CCR2-/- mice demonstrated the more robust protection, reflected by less inflammatory and Th17-related cytokines and less collagen formation. Next we evaluated the dynamic of CCR2+/rfp cell infiltration and we observed that they adhere onto the vessels at early stages of disease, culminating in increased recruitment of CCR2+/rfp cells at later stages. On the other hand, CCR2rfp/rfp animals exhibited less fibrosis formation and reduced numbers of recruited cells at later stages. We have experimentally demonstrated that inflammatory CCR2+ cells that reach the injured kidney at initial stages after tissue damage are responsible for the fibrotic pattern observed at later time points in the context of UUO.
Collapse
|
17
|
Emal D, Rampanelli E, Stroo I, Butter LM, Teske GJ, Claessen N, Stokman G, Florquin S, Leemans JC, Dessing MC. Depletion of Gut Microbiota Protects against Renal Ischemia-Reperfusion Injury. J Am Soc Nephrol 2016; 28:1450-1461. [PMID: 27927779 DOI: 10.1681/asn.2016030255] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 11/03/2016] [Indexed: 12/15/2022] Open
Abstract
An accumulating body of evidence shows that gut microbiota fulfill an important role in health and disease by modulating local and systemic immunity. The importance of the microbiome in the development of kidney disease, however, is largely unknown. To study this concept, we depleted gut microbiota with broad-spectrum antibiotics and performed renal ischemia-reperfusion (I/R) injury in mice. Depletion of the microbiota significantly attenuated renal damage, dysfunction, and remote organ injury and maintained tubular integrity after renal I/R injury. Gut flora-depleted mice expressed lower levels of F4/80 and chemokine receptors CX3CR1 and CCR2 in the F4/80+ renal resident macrophage population and bone marrow (BM) monocytes than did control mice. Additionally, compared with control BM monocytes, BM monocytes from gut flora-depleted mice had decreased migratory capacity toward CX3CL1 and CCL2 ligands. To study whether these effects were driven by depletion of the microbiota, we performed fecal transplants in antibiotic-treated mice and found that transplant of fecal material from an untreated mouse abolished the protective effect of microbiota depletion upon renal I/R injury. In conclusion, we show that depletion of gut microbiota profoundly protects against renal I/R injury by reducing maturation status of F4/80+ renal resident macrophages and BM monocytes. Therefore, dampening the inflammatory response by targeting microbiota-derived mediators might be a promising therapy against I/R injury.
Collapse
Affiliation(s)
- Diba Emal
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; and
| | - Elena Rampanelli
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; and
| | - Ingrid Stroo
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; and
| | - Loes M Butter
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; and
| | - Gwendoline J Teske
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; and
| | - Nike Claessen
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; and
| | - Geurt Stokman
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; and
| | - Sandrine Florquin
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; and.,Department of Pathology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Jaklien C Leemans
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; and
| | - Mark C Dessing
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; and
| |
Collapse
|
18
|
Iseri K, Iyoda M, Ohtaki H, Matsumoto K, Wada Y, Suzuki T, Yamamoto Y, Saito T, Hihara K, Tachibana S, Honda K, Shibata T. Therapeutic effects and mechanism of conditioned media from human mesenchymal stem cells on anti-GBM glomerulonephritis in WKY rats. Am J Physiol Renal Physiol 2016; 310:F1182-91. [DOI: 10.1152/ajprenal.00165.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 03/24/2016] [Indexed: 02/06/2023] Open
Abstract
Recent studies have demonstrated that conditioned media derived from mesenchymal stem cells (MSC-CM) have therapeutic effects in various experimental diseases. However, the therapeutic mechanism is not fully understood. In the present study, we investigated the therapeutic effects and mechanism of MSC-CM in experimental antiglomerular basement membrane glomerulonephritis. We administered either MSC-CM or vehicle from day 0 to day 10 after the induction of nephrotoxic serum nephritis in Wistar-Kyoto rats. In vitro, we analyzed the effects of MSC-CM on TNF-α-mediated cytokine production in cultured normal human mesangial cells, proximal tubular (HK-2) cells, human umbilical vein endothelial cells, and monocytes (THP-1 and peripheral blood mononuclear cells). Compared with vehicle treatment, MSC-CM treatment improved proteinuria and renal dysfunction. Histologically, MSC-CM-treated rats had reduced crescent formation and glomerular ED1+ macrophage infiltration and increased glomerular ED2+ macrophage infiltration. Increased serum monocyte chemoattractant protein (MCP)-1 levels were observed in MSC-CM-treated rats. Renal cortical mRNA expression levels of proinflammatory cytokines, such as TNF-α and IL-6, and of the T helper cell 1 cytokine interferon-γ were greatly decreased by MSC-CM treatment. In vitro, pretreatment with MSC-CM blocked TNF-α-mediated IL-8 release in normal human mesangial cells and HK-2 cells. TNF-α-mediated MCP-1 release was enhanced by pretreatment with MSC-CM in human umbilical vein endothelial cells and HK-2 cells and was strikingly enhanced in THP-1 cells. Stimulation of peripheral blood mononuclear cells with a combination of MCP-1 and IL-4 enhanced the expression of M2-associated genes compared with IL-4 alone. We demonstrated that MSC-CM had therapeutic effects in experimental antiglomerular basement membrane glomerulonephritis that were mediated through anti-inflammatory effects that were partly due to acceleration of M2 macrophage polarization, which might be mediated by MCP-1 enhancement.
Collapse
Affiliation(s)
- Ken Iseri
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Masayuki Iyoda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Hirokazu Ohtaki
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Kei Matsumoto
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Yukihiro Wada
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Taihei Suzuki
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Yasutaka Yamamoto
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Tomohiro Saito
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Kei Hihara
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Shohei Tachibana
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Kazuho Honda
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Takanori Shibata
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| |
Collapse
|
19
|
Cardiac Stem Cell Secretome Protects Cardiomyocytes from Hypoxic Injury Partly via Monocyte Chemotactic Protein-1-Dependent Mechanism. Int J Mol Sci 2016; 17:ijms17060800. [PMID: 27231894 PMCID: PMC4926334 DOI: 10.3390/ijms17060800] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 05/10/2016] [Accepted: 05/12/2016] [Indexed: 12/20/2022] Open
Abstract
Cardiac stem cells (CSCs) were known to secrete diverse paracrine factors leading to functional improvement and beneficial left ventricular remodeling via activation of the endogenous pro-survival signaling pathway. However, little is known about the paracrine factors secreted by CSCs and their roles in cardiomyocyte survival during hypoxic condition mimicking the post-myocardial infarction environment. We established Sca-1+/CD31- human telomerase reverse transcriptase-immortalized CSCs (Sca-1+/CD31- CSCs(hTERT)), evaluated their stem cell properties, and paracrine potential in cardiomyocyte survival during hypoxia-induced injury. Sca-1+/CD31- CSCs(hTERT) sustained proliferation ability even after long-term culture exceeding 100 population doublings, and represented multi-differentiation potential into cardiomyogenic, endothelial, adipogenic, and osteogenic lineages. Dominant factors secreted from Sca-1+/CD31- CSCs(hTERT) were EGF, TGF-β1, IGF-1, IGF-2, MCP-1, HGF R, and IL-6. Among these, MCP-1 was the most predominant factor in Sca-1+/CD31- CSCs(hTERT) conditioned medium (CM). Sca-1+/CD31- CSCs(hTERT) CM increased survival and reduced apoptosis of HL-1 cardiomyocytes during hypoxic injury. MCP-1 silencing in Sca-1+/CD31- CSCs(hTERT) CM resulted in a significant reduction in cardiomyocyte apoptosis. We demonstrated that Sca-1+/CD31- CSCs(hTERT) exhibited long-term proliferation capacity and multi-differentiation potential. Sca-1+/CD31- CSCs(hTERT) CM protected cardiomyocytes from hypoxic injury partly via MCP-1-dependent mechanism. Thus, they are valuable sources for in vitro and in vivo studies in the cardiovascular field.
Collapse
|