1
|
Yang Q, Su S, Luo N, Cao G. Adenine-induced animal model of chronic kidney disease: current applications and future perspectives. Ren Fail 2024; 46:2336128. [PMID: 38575340 PMCID: PMC10997364 DOI: 10.1080/0886022x.2024.2336128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024] Open
Abstract
Chronic kidney disease (CKD) with high morbidity and mortality all over the world is characterized by decreased kidney function, a condition which can result from numerous risk factors, including diabetes, hypertension and obesity. Despite significant advances in our understanding of the pathogenesis of CKD, there are still no treatments that can effectively combat CKD, which underscores the urgent need for further study into the pathological mechanisms underlying this condition. In this regard, animal models of CKD are indispensable. This article reviews a widely used animal model of CKD, which is induced by adenine. While a physiologic dose of adenine is beneficial in terms of biological activity, a high dose of adenine is known to induce renal disease in the organism. Following a brief description of the procedure for disease induction by adenine, major mechanisms of adenine-induced CKD are then reviewed, including inflammation, oxidative stress, programmed cell death, metabolic disorders, and fibrillation. Finally, the application and future perspective of this adenine-induced CKD model as a platform for testing the efficacy of a variety of therapeutic approaches is also discussed. Given the simplicity and reproducibility of this animal model, it remains a valuable tool for studying the pathological mechanisms of CKD and identifying therapeutic targets to fight CKD.
Collapse
Affiliation(s)
- Qiao Yang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Songya Su
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Nan Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Gang Cao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
2
|
Wu Z, Hu J, Li Y, Yao X, Ouyang S, Ren K. Assessment of renal pathophysiological processes and protective effect of quercetin on contrast-induced acute kidney injury in type 1 diabetic mice using diffusion tensor imaging. Redox Rep 2024; 29:2398380. [PMID: 39284588 PMCID: PMC11407404 DOI: 10.1080/13510002.2024.2398380] [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: 09/19/2024] Open
Abstract
Purpose: To investigate the renal pathophysiological processes and protective effect of quercetin on contrast-induced acute kidney injury (CI-AKI) in mice with type 1 diabetic mellitus(DM) using diffusion tensor imaging(DTI).Methods: Mice with DM were divided into two groups. In the diabetic + contrast medium(DCA) group, the changes of the mice kidneys were monitored at 1, 24, 48, and 72 h after the injection of iodixanol(4gI/kg). The mice in the diabetic + contrast medium + quercetin(DCA + QE) group were orally given different concentrations of quercetin for seven days before injection of iodixanol. In vitro experiments, renal tubular epithelial (HK-2) cells exposed to high glucose conditions were treated with various quercetin concentrations before treatment with iodixanol(250 mgI/mL).Results: DTI-derived mean diffusivity(MD) and fractional anisotropy(FA) values can be used to evaluate CI-AKI effectively. Quercetin significantly increased the expression of Sirt 1 and reduced oxidative stress by increasing Nrf 2/HO-1/SOD1. The antiapoptotic effect of quercetin on CI-AKI was revealed by decreasing proteins level and by reducing the number of apoptosis-positive cells. In addition, flow cytometry indicated quercetin-mediated inhibition of M1 macrophage polarization in the CI-AKI.Conclusions: DTI will be an effective noninvasive tool in diagnosing CI-AKI. Quercetin attenuates CI-AKI on the basis of DM through anti-oxidative stress, apoptosis, and inflammation.
Collapse
Affiliation(s)
- Ziqian Wu
- Department of Radiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen Radiological Control Center, Xiamen, People's Republic of China
| | - Jingyi Hu
- Department of Radiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen Radiological Control Center, Xiamen, People's Republic of China
| | - Yanfei Li
- Cell Therapy Research Center, Xiamen Humanity Hospital, Xiamen, People's Republic of China
| | - Xiang Yao
- Department of Neurosurgery, Zhongshan Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Siyu Ouyang
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Ke Ren
- Department of Radiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen Radiological Control Center, Xiamen, People's Republic of China
| |
Collapse
|
3
|
Li G, Yang H, Zhang D, Zhang Y, Liu B, Wang Y, Zhou H, Xu ZX, Wang Y. The role of macrophages in fibrosis of chronic kidney disease. Biomed Pharmacother 2024; 177:117079. [PMID: 38968801 DOI: 10.1016/j.biopha.2024.117079] [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: 05/08/2024] [Revised: 06/23/2024] [Accepted: 06/29/2024] [Indexed: 07/07/2024] Open
Abstract
Macrophages are widely distributed throughout various tissues of the body, and mounting evidence suggests their involvement in regulating the tissue microenvironment, thereby influencing disease onset and progression through direct or indirect actions. In chronic kidney disease (CKD), disturbances in renal functional homeostasis lead to inflammatory cell infiltration, tubular expansion, glomerular atrophy, and subsequent renal fibrosis. Macrophages play a pivotal role in this pathological process. Therefore, understanding their role is imperative for investigating CKD progression, mitigating its advancement, and offering novel research perspectives for fibrosis treatment from an immunological standpoint. This review primarily delves into the intrinsic characteristics of macrophages, their origins, diverse subtypes, and their associations with renal fibrosis. Particular emphasis is placed on the transition between M1 and M2 phenotypes. In late-stage CKD, there is a shift from the M1 to the M2 phenotype, accompanied by an increased prevalence of M2 macrophages. This transition is governed by the activation of the TGF-β1/SMAD3 and JAK/STAT pathways, which facilitate macrophage-to-myofibroblast transition (MMT). The tyrosine kinase Src is involved in both signaling cascades. By thoroughly elucidating macrophage functions and comprehending the modes and molecular mechanisms of macrophage-fibroblast interaction in the kidney, novel, tailored therapeutic strategies for preventing or attenuating the progression of CKD can be developed.
Collapse
Affiliation(s)
- Guangtao Li
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Hongxia Yang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Dan Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Yanghe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yuxiong Wang
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Zhi-Xiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| |
Collapse
|
4
|
Cheng X, Bai X, Shang WY, Wei L, Jia JY, Yan TK, Gu QH. Profiling dendritic cells subsets in renal tissue of patients with crescentic glomerulonephritis. Int Urol Nephrol 2024:10.1007/s11255-024-04175-6. [PMID: 39069601 DOI: 10.1007/s11255-024-04175-6] [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: 01/16/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Dendritic cells (DCs) have been speculated to be involved in the pathogenesis of glomerular diseases. However, the numbers and distribution of DC subsets in the kidneys of patients with crescentic glomerulonephritis (CrGN) have not been clearly elucidated. METHODS A total of 26 patients with biopsy-proven CrGN were enrolled. Indirect immunofluorescence staining was used to quantify DC subsets in renal specimens. Double staining of HLA with CD11C, BDCA2 and CD209 respectively was performed to detect DC subsets. The correlation between DC subsets infiltrated in the kidney and clinical and pathological parameters was investigated. RESULTS DC subsets were predominantly present in the kidney interstitium, particularly in the peri-glomerular area. The numbers of CD11C+DCs, BDCA2+DCs and CD209+DCs increased in the patients with CrGN and varied among different types of CrGN. Though significant correlation between DC subsets and the percentage of crescents had not been identified, a notable increase in the number of CD11C+DCs were observed with the chronic development of crescents. Furthermore, patients with severe tubulointerstitial injury exhibited significantly more infiltrations of CD11C+DCs, BDCA2+DCs and CD209+DCs. Moreover, the numbers of CD11C+DCs and BDCA2+DCs were found to correlate with the level of serum C3. CONCLUSIONS Patients with CrGN showed increased kidney infiltration of DC subsets, primarily localized in the renal interstitium and peri-glomerular region. The correlation between DC subsets and fibrosis of crescent and severe tubulointerstitial injury implied a potential involvement of DCs in the development of CrGN.
Collapse
Affiliation(s)
- Xi Cheng
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xue Bai
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Wen-Ya Shang
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Li Wei
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jun-Ya Jia
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Tie-Kun Yan
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Qiu-Hua Gu
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
5
|
Obi Y, Raimann JG, Kalantar-Zadeh K, Murea M. Residual Kidney Function in Hemodialysis: Its Importance and Contribution to Improved Patient Outcomes. Toxins (Basel) 2024; 16:298. [PMID: 39057938 PMCID: PMC11281084 DOI: 10.3390/toxins16070298] [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: 04/30/2024] [Revised: 05/27/2024] [Accepted: 06/11/2024] [Indexed: 07/28/2024] Open
Abstract
Individuals afflicted with advanced kidney dysfunction who require dialysis for medical management exhibit different degrees of native kidney function, called residual kidney function (RKF), ranging from nil to appreciable levels. The primary focus of this manuscript is to delve into the concept of RKF, a pivotal yet under-represented topic in nephrology. To begin, we unpack the definition and intrinsic nature of RKF. We then juxtapose the efficiency of RKF against that of hemodialysis in preserving homeostatic equilibrium and facilitating physiological functions. Given the complex interplay of RKF and overall patient health, we shed light on the extent of its influence on patient outcomes, particularly in those living with advanced kidney dysfunction and on dialysis. This manuscript subsequently presents methodologies and measures to assess RKF, concluding with the potential benefits of targeted interventions aimed at preserving RKF.
Collapse
Affiliation(s)
- Yoshitsugu Obi
- Division of Nephrology, Department of Medicine, The University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Jochen G. Raimann
- Renal Research Institute, New York, NY 10065, USA;
- Katz School of Science and Health, Yeshiva University, New York, NY 10033, USA
| | - Kamyar Kalantar-Zadeh
- Tibor Rubin Veterans Affairs Long Beach Healthcare System, Long Beach, CA 90822, USA;
- The Lundquist Institute at Harbor, UCLA Medical Center, Torrance, CA 90502, USA
- Division of Nephrology, Hypertension, and Kidney Transplantation, University of California Irvine, Orange, CA 92868, USA
| | - Mariana Murea
- Department of Internal Medicine, Section on Nephrology, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| |
Collapse
|
6
|
Yang H, Li J, Huang XR, Bucala R, Xu A, Lan HY. Macrophage-derived macrophage migration inhibitory factor mediates renal injury in anti-glomerular basement membrane glomerulonephritis. Front Immunol 2024; 15:1361343. [PMID: 38846956 PMCID: PMC11153660 DOI: 10.3389/fimmu.2024.1361343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 04/30/2024] [Indexed: 06/09/2024] Open
Abstract
Macrophages are a rich source of macrophage migration inhibitory factor (MIF). It is well established that macrophages and MIF play a pathogenic role in anti-glomerular basement membrane crescentic glomerulonephritis (anti-GBM CGN). However, whether macrophages mediate anti-GBM CGN via MIF-dependent mechanism remains unexplored, which was investigated in this study by specifically deleting MIF from macrophages in MIFf/f-lysM-cre mice. We found that compared to anti-GBM CGN induced in MIFf/f control mice, conditional ablation of MIF in macrophages significantly suppressed anti-GBM CGN by inhibiting glomerular crescent formation and reducing serum creatinine and proteinuria while improving creatine clearance. Mechanistically, selective MIF depletion in macrophages largely inhibited renal macrophage and T cell recruitment, promoted the polarization of macrophage from M1 towards M2 via the CD74/NF-κB/p38MAPK-dependent mechanism. Unexpectedly, selective depletion of macrophage MIF also significantly promoted Treg while inhibiting Th1 and Th17 immune responses. In summary, MIF produced by macrophages plays a pathogenic role in anti-GBM CGN. Targeting macrophage-derived MIF may represent a novel and promising therapeutic approach for the treatment of immune-mediated kidney diseases.
Collapse
Affiliation(s)
- Hui Yang
- Department of Nephrology, Sun Yat‐Sen Memorial Hospital, Sun Yat‐Sen University, Guangzhou, China
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jinhong Li
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat‐sen University, SunYat‐sen University, Shenzhen, China
| | - Xiao-ru Huang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Departments of Nephrology and Pathology, Guangdong Provincial Hospital, Southern Medical University, Guangzhou, China
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Anping Xu
- Department of Nephrology, Sun Yat‐Sen Memorial Hospital, Sun Yat‐Sen University, Guangzhou, China
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Departments of Nephrology and Pathology, Guangdong Provincial Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Chen L, Li L, Huang C, Cao X, Jiang Y. Dynamic mRNA network profiles in macrophages challenged with lipopolysaccharide. Am J Transl Res 2024; 16:1643-1659. [PMID: 38883351 PMCID: PMC11170596 DOI: 10.62347/kmaj3260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/18/2024] [Indexed: 06/18/2024]
Abstract
OBJECTIVES To elucidate the transcriptome of macrophages in an inflammation model induced by lipopolysaccharide (LPS), providing insight into the molecular basis of inflammation. METHODS We utilized RNA sequencing (RNA-seq) to analyze dynamic changes in gene expression in RAW264.7 macrophages treated with LPS at multiple time points. Differentially expressed genes (DEGs) were identified using the edgeR package. Short Time-series Expression Miner (STEM) and KEGG pathway enrichment analyses were conducted to determine temporal expression patterns during inflammation. RESULTS We identified 2,512 DEGs, with initial inflammatory responses occurring in two distinct phases at 1 h and 3 h. Venn diagram analysis revealed 78 consistently dysregulated genes throughout the inflammatory process. A key module of 18 dysregulated genes was identified, including Irg1, which may exert an inhibitory effect on inflammation. Further, a second metabolic shift in activated macrophages was observed at the late middle stage (12 h). Multi-omics analysis highlighted the ribosome's potential regulatory role in the inflammatory response. CONCLUSIONS This study provides a detailed view of the molecular mechanisms underlying inflammation in macrophages and reveals a dynamic genetic landscape crucial for further research. Our findings underscore the complex interaction between gene expression, metabolic shifts, and ribosomal functions in response to LPS-induced inflammation.
Collapse
Affiliation(s)
- Li Chen
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University Guangzhou 510515, Guangdong, China
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Southern Medical University Guangzhou 510630, Guangdong, China
| | - Lei Li
- Department of Neurology, Shenzhen Hospital of Southern Medical University Shenzhen 518000, Guangdong, China
| | - Chenyang Huang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University Guangzhou 510515, Guangdong, China
| | - Xusong Cao
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University Guangzhou 510515, Guangdong, China
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University Guangzhou 510515, Guangdong, China
- Department of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital, Guangdong Provincial Key Laboratory of Proteomics, Southern Medical University (Dongguan People's Hospital) Dongguan 523059, Guangdong, China
| |
Collapse
|
8
|
Ashraf MI, Mengwasser J, Reutzel-Selke A, Polenz D, Führer K, Lippert S, Tang P, Michaelis E, Catar R, Pratschke J, Witzel C, Sauer IM, Tullius SG, Kern B. Depletion of donor dendritic cells ameliorates immunogenicity of both skin and hind limb transplants. Front Immunol 2024; 15:1395945. [PMID: 38799435 PMCID: PMC11116604 DOI: 10.3389/fimmu.2024.1395945] [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: 03/04/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
Acute cellular rejection remains a significant obstacle affecting successful outcomes of organ transplantation including vascularized composite tissue allografts (VCA). Donor antigen presenting cells (APCs), particularly dendritic cells (DCs), orchestrate early alloimmune responses by activating recipient effector T cells. Employing a targeted approach, we investigated the impact of donor-derived conventional DCs (cDCs) and APCs on the immunogenicity of skin and skin-containing VCA grafts, using mouse models of skin and hind limb transplantation. By post-transplantation day 6, skin grafts demonstrated severe rejections, characterized by predominance of recipient CD4 T cells. In contrast, hind limb grafts showed moderate rejection, primarily infiltrated by CD8 T cells. Notably, the skin component exhibited heightened immunogenicity when compared to the entire VCA, evidenced by increased frequencies of pan (CD11b-CD11c+), mature (CD11b-CD11c+MHCII+) and active (CD11b-CD11c+CD40+) DCs and cDC2 subset (CD11b+CD11c+ MHCII+) in the lymphoid tissues and the blood of skin transplant recipients. While donor depletion of cDC and APC reduced frequencies, maturation and activation of DCs in all analyzed tissues of skin transplant recipients, reduction in DC activities was only observed in the spleen of hind limb recipients. Donor cDC and APC depletion did not impact all lymphocyte compartments but significantly affected CD8 T cells and activated CD4 T in lymph nodes of skin recipients. Moreover, both donor APC and cDC depletion attenuated the Th17 immune response, evident by significantly reduced Th17 (CD4+IL-17+) cells in the spleen of skin recipients and reduced levels of IL-17E and lymphotoxin-α in the serum samples of both skin and hind limb recipients. In conclusion, our findings underscore the highly immunogenic nature of skin component in VCA. The depletion of donor APCs and cDCs mitigates the immunogenicity of skin grafts while exerting minimal impact on VCA.
Collapse
Affiliation(s)
- Muhammad Imtiaz Ashraf
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Joerg Mengwasser
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Anja Reutzel-Selke
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Dietrich Polenz
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Kirsten Führer
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Steffen Lippert
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Peter Tang
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Edward Michaelis
- Department of Pathology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Rusan Catar
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Healthy, Berlin, Germany
| | - Johann Pratschke
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Christian Witzel
- Department of Plastic Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Igor M. Sauer
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Stefan G. Tullius
- Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Einstein Berlin Institute of Health Visiting Fellow, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Barbara Kern
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Department of Plastic Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin Institute of Health (BIH) Biomedical Innovation Academy, Berlin Institute of Health (BIH) Charité Clinician Scientist Program, Berlin, Germany
| |
Collapse
|
9
|
Nachiappa Ganesh R, Garcia G, Truong L. Monocytes and Macrophages in Kidney Disease and Homeostasis. Int J Mol Sci 2024; 25:3763. [PMID: 38612574 PMCID: PMC11012230 DOI: 10.3390/ijms25073763] [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: 01/26/2024] [Revised: 03/19/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
The monocyte-macrophage lineage of inflammatory cells is characterized by significant morphologic and functional plasticity. Macrophages have broad M1 and M2 phenotype subgroups with distinctive functions and dual reno-toxic and reno-protective effects. Macrophages are a major contributor to injury in immune-complex-mediated, as well as pauci-immune, glomerulonephritis. Macrophages are also implicated in tubulointerstitial and vascular disease, though there have not been many human studies. Patrolling monocytes in the intravascular compartment have been reported in auto-immune injury in the renal parenchyma, manifesting as acute kidney injury. Insights into the pathogenetic roles of macrophages in renal disease suggest potentially novel therapeutic and prognostic biomarkers and targeted therapy. This review provides a concise overview of the macrophage-induced pathogenetic mechanism as a background for the latest findings about macrophages' roles in different renal compartments and common renal diseases.
Collapse
Affiliation(s)
- Rajesh Nachiappa Ganesh
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA;
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
| | - Gabriela Garcia
- Department of Medicine, Renal Division, University of Colorado, Anschutz Medical Campus, Aurora, CO 605006, USA;
| | - Luan Truong
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA;
| |
Collapse
|
10
|
Hao XM, Liu Y, Hailaiti D, Gong Y, Zhang XD, Yue BN, Liu JP, Wu XL, Yang KZ, Wang J, Liu QG. Mechanisms of inflammation modulation by different immune cells in hypertensive nephropathy. Front Immunol 2024; 15:1333170. [PMID: 38545112 PMCID: PMC10965702 DOI: 10.3389/fimmu.2024.1333170] [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: 11/04/2023] [Accepted: 02/15/2024] [Indexed: 04/10/2024] Open
Abstract
Hypertensive nephropathy (HTN) is the second leading cause of end-stage renal disease (ESRD) and a chronic inflammatory disease. Persistent hypertension leads to lesions of intrarenal arterioles and arterioles, luminal stenosis, secondary ischemic renal parenchymal damage, and glomerulosclerosis, tubular atrophy, and interstitial fibrosis. Studying the pathogenesis of hypertensive nephropathy is a prerequisite for diagnosis and treatment. The main cause of HTN is poor long-term blood pressure control, but kidney damage is often accompanied by the occurrence of immune inflammation. Some studies have found that the activation of innate immunity, inflammation and acquired immunity is closely related to the pathogenesis of HTN, which can cause damage and dysfunction of target organs. There are more articles on the mechanism of diabetic nephropathy, while there are fewer studies related to immunity in hypertensive nephropathy. This article reviews the mechanisms by which several different immune cells and inflammatory cytokines regulate blood pressure and renal damage in HTN. It mainly focuses on immune cells, cytokines, and chemokines and inhibitors. However, further comprehensive and large-scale studies are needed to determine the role of these markers and provide effective protocols for clinical intervention and treatment.
Collapse
Affiliation(s)
- Xiao-Min Hao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | | | - Yu Gong
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Xu-Dong Zhang
- Department of Chinese Medicine, Beijing Jishuitan Hospital, Beijing, China
| | - Bing-Nan Yue
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ji-Peng Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao-Li Wu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ke-Zhen Yang
- Department of Rehabilitation Medicine, Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qing-Guo Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
11
|
Xu C, Tsihlis G, Chau K, Trinh K, Rogers NM, Julovi SM. Novel Perspectives in Chronic Kidney Disease-Specific Cardiovascular Disease. Int J Mol Sci 2024; 25:2658. [PMID: 38473905 PMCID: PMC10931927 DOI: 10.3390/ijms25052658] [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: 01/19/2024] [Revised: 02/18/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Chronic kidney disease (CKD) affects > 10% of the global adult population and significantly increases the risk of cardiovascular disease (CVD), which remains the leading cause of death in this population. The development and progression of CVD-compared to the general population-is premature and accelerated, manifesting as coronary artery disease, heart failure, arrhythmias, and sudden cardiac death. CKD and CV disease combine to cause multimorbid cardiorenal syndrome (CRS) due to contributions from shared risk factors, including systolic hypertension, diabetes mellitus, obesity, and dyslipidemia. Additional neurohormonal activation, innate immunity, and inflammation contribute to progressive cardiac and renal deterioration, reflecting the strong bidirectional interaction between these organ systems. A shared molecular pathophysiology-including inflammation, oxidative stress, senescence, and hemodynamic fluctuations characterise all types of CRS. This review highlights the evolving paradigm and recent advances in our understanding of the molecular biology of CRS, outlining the potential for disease-specific therapies and biomarker disease detection.
Collapse
Affiliation(s)
- Cuicui Xu
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; (C.X.); (K.T.)
| | - George Tsihlis
- Renal and Transplantation Medicine, Westmead Hospital, Westmead, NSW 2145, Australia;
| | - Katrina Chau
- Department of Renal Services, Blacktown Hospital, Blacktown, NSW 2148, Australia;
- Blacktown Clinical School, School of Medicine, Western Sydney University, Sydney, NSW 2148, Australia
| | - Katie Trinh
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; (C.X.); (K.T.)
- Department of Renal Services, Blacktown Hospital, Blacktown, NSW 2148, Australia;
| | - Natasha M. Rogers
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; (C.X.); (K.T.)
- Renal and Transplantation Medicine, Westmead Hospital, Westmead, NSW 2145, Australia;
- Faculty of Medicine and Health, The University of Sydney, Science Rd., Camperdown, NSW 2050, Australia
| | - Sohel M. Julovi
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; (C.X.); (K.T.)
- Faculty of Medicine and Health, The University of Sydney, Science Rd., Camperdown, NSW 2050, Australia
| |
Collapse
|
12
|
Zhu XY, Klomjit N, Pawar AS, Puranik AS, Yang ZZ, Lutgens E, Eirin A, Lerman A, Textor SC, Lerman LO. Altered immune cell phenotypes within chronically ischemic human kidneys distal to occlusive renal artery disease. Am J Physiol Renal Physiol 2024; 326:F257-F264. [PMID: 38031731 PMCID: PMC11198973 DOI: 10.1152/ajprenal.00234.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/04/2023] [Accepted: 11/22/2023] [Indexed: 12/01/2023] Open
Abstract
Renal artery stenosis (RAS) is a major cause of ischemic kidney disease, which is largely mediated by inflammation. Mapping the immune cell composition in ischemic kidneys might provide useful insight into the disease pathogenesis and uncover therapeutic targets. We used mass cytometry (CyTOF) to explore the single-cell composition in a unique data set of human kidneys nephrectomized due to chronic occlusive vascular disease (RAS, n = 3), relatively healthy donor kidneys (n = 6), and unaffected sections of kidneys with renal cell carcinoma (RCC, n = 3). Renal fibrosis and certain macrophage populations were also evaluated in renal sections. Cytobank analysis showed in RAS kidneys decreased cell populations expressing epithelial markers (CD45-/CD13+) and increased CD45+ inflammatory cells, whereas scattered tubular-progenitor-like cells (CD45-/CD133+/CD24+) increased compared with kidney donors. Macrophages switched to proinflammatory phenotypes in RAS, and the numbers of IL-10-producing dendritic cells (DC) were also lower. Compared with kidney donors, RAS kidneys had decreased overall DC populations but increased plasmacytoid DC. Furthermore, senescent active T cells (CD45+/CD28+/CD57+), aged neutrophils (CD45+/CD15+/CD24+/CD11c+), and regulatory B cells (CD45+/CD14-/CD24+/CD44+) were increased in RAS. RCC kidneys showed a distribution of cell phenotypes comparable with RAS but less pronounced, accompanied by an increase in CD34+, CD370+, CD103+, and CD11c+/CD103+ cells. Histologically, RAS kidneys showed significantly increased fibrosis and decreased CD163+/CD141+ cells. The single-cell platform CyTOF enables the detection of significant changes in renal cells, especially in subsets of immune cells in ischemic human kidneys. Endogenous pro-repair cell types in RAS warrant future study for potential immune therapy.NEW & NOTEWORTHY The single-cell platform mass cytometry (CyTOF) enables detection of significant changes in one million of renal cells, especially in subsets of immune cells in ischemic human kidneys distal to renal artery stenosis (RAS). We found that pro-repair cell types such as scattered tubular-progenitor-like cells, aged neutrophils, and regulatory B cells show a compensatory increase in RAS. Immune cell phenotype changes may reflect ongoing inflammation and impaired immune defense capability in the kidneys.
Collapse
Affiliation(s)
- Xiang-Yang Zhu
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Nattawat Klomjit
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Aditya S Pawar
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Amrutesh S Puranik
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Zhi-Zhang Yang
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, United States
| | - Esther Lutgens
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, United States
| | - Alfonso Eirin
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, United States
| | - Stephen C Textor
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Lilach O Lerman
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota, United States
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
13
|
Schrezenmeier E, Dörner T, Halleck F, Budde K. Cellular Immunobiology and Molecular Mechanisms in Alloimmunity-Pathways of Immunosuppression. Transplantation 2024; 108:148-160. [PMID: 37309030 DOI: 10.1097/tp.0000000000004646] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Current maintenance immunosuppression commonly comprises a synergistic combination of tacrolimus as calcineurin inhibitor (CNI), mycophenolic acid, and glucocorticoids. Therapy is often individualized by steroid withdrawal or addition of belatacept or inhibitors of the mechanistic target of rapamycin. This review provides a comprehensive overview of their mode of action, focusing on the cellular immune system. The main pharmacological action of CNIs is suppression of the interleukin-2 pathway that leads to inhibition of T cell activation. Mycophenolic acid inhibits the purine pathway and subsequently diminishes T and B cell proliferation but also exerts a variety of effects on almost all immune cells, including inhibition of plasma cell activity. Glucocorticoids exert complex regulation via genomic and nongenomic mechanisms, acting mainly by downregulating proinflammatory cytokine signatures and cell signaling. Belatacept is potent in inhibiting B/T cell interaction, preventing formation of antibodies; however, it lacks the potency of CNIs in preventing T cell-mediated rejections. Mechanistic target of rapamycin inhibitors have strong antiproliferative activity on all cell types interfering with multiple metabolic pathways, partly explaining poor tolerability, whereas their superior effector T cell function might explain their benefits in the case of viral infections. Over the past decades, clinical and experimental studies provided a good overview on the underlying mechanisms of immunosuppressants. However, more data are needed to delineate the interaction between innate and adaptive immunity to better achieve tolerance and control of rejection. A better and more comprehensive understanding of the mechanistic reasons for failure of immunosuppressants, including individual risk/benefit assessments, may permit improved patient stratification.
Collapse
Affiliation(s)
- Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Academy, Clinician Scientist Program Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Dörner
- Department of Rheumatology and Clinical Immunology - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
14
|
Yamashita N, Kramann R. Mechanisms of kidney fibrosis and routes towards therapy. Trends Endocrinol Metab 2024; 35:31-48. [PMID: 37775469 DOI: 10.1016/j.tem.2023.09.001] [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: 07/18/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 10/01/2023]
Abstract
Kidney fibrosis is the final common pathway of virtually all chronic kidney diseases (CKDs) and is therefore considered to be a promising therapeutic target for these conditions. However, despite great progress in recent years, no targeted antifibrotic therapies for the kidney have been approved, likely because the complex mechanisms that initiate and drive fibrosis are not yet completely understood. Recent single-cell genomic approaches have allowed novel insights into kidney fibrosis mechanisms in mouse and human, particularly the heterogeneity and differentiation processes of myofibroblasts, the role of injured epithelial cells and immune cells, and their crosstalk mechanisms. In this review we summarize the key mechanisms that drive kidney fibrosis, including recent advances in understanding the mechanisms, as well as potential routes for developing novel targeted antifibrotic therapeutics.
Collapse
Affiliation(s)
- Noriyuki Yamashita
- Department of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany; Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany; Department of Internal Medicine, Nephrology, and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
15
|
Liu M, Guo P, Zeng M, Zhang Y, Jia J, Liu Y, Chen X, Kuang H, Feng W, Zheng X. Effects and mechanisms of frehmaglutin D and rehmaionoside C improve LPS-induced acute kidney injury through the estrogen receptor-mediated TLR4 pathway in vivo and in vitro. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155218. [PMID: 37980806 DOI: 10.1016/j.phymed.2023.155218] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/30/2023] [Accepted: 11/12/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Sepsis-induced acute kidney injury (S-AKI) is an inflammatory disease with sex differences and there has no effective drugs to cure it. Frehmaglutin D (Fre D) and rehmaionoside C (Reh C) are two violetone compounds with estrogenic activity isolated from Rehmannia glutinosa. However, whether these two drugs exert protective effects on S-AKI through their estrogen-like activity are unclear. PURPOSE This study aimed to explore the effects and mechanisms of Fre D and Reh C on lipopolysaccharide (LPS)-induced S-AKI through the estrogen receptor pathway in vivo and in vitro and to explore the interaction between ER and TLR4 for the first time. METHODS The LPS-induced female BALB/c mice S-AKI mouse model was established by adding the estrogen receptor antagonist ICI182,780. Renal function, inflammation, oxidative stress, apoptosis, immune cells, and expression of key proteins of the ER-TLR4-IL-1β pathway were tested. The affinity of Fre D and Reh C for the ER was investigated by molecular docking. Then, an in vitro S-AKI model was established, and ERα/ERβ antagonists (MPP/PHTPP) were added and combined with gene overexpression techniques. The interaction between ER and TLR4 was further explored by Co-IP, GST pull-down and SPR techniques. RESULTS Fre D and Reh C ameliorated LPS-induced renal damage, inflammation in mice, regulated the immune cells, decreased ROS levels, increased ERα and ERβ protein expression, and decreased TLR4, caspase 11 and IL-1β protein expression. These effects were blocked by ICI182,780. Molecular docking results showed that Fre D and Reh C bound ERα and ERβ with similar potency. The results of in vitro suggested that Fre D and Reh C reduced the levels of inflammation, ROS and apoptosis, TLR4, caspase 11, and IL-1β protein expression and increased ERα/ERβ protein expression in cells. All of these effects were reversed by the addition of MPP/PHTPP and further enhanced after ERα/ERβ gene overexpression with no significant difference in effects. Moreover, there was an indirect or direct interaction between ER and TLR4, and the binding of ERα and ERβ to TLR4 was concentration dependent. CONCLUSION Fre D and Reh C may improve S-AKI through the ER-TLR4-IL-1β pathway and may act on both ERα and ERβ receptors. Moreover, ERα and ERβ may interact directly or indirectly with TLR4, which was studied for the first time.
Collapse
Affiliation(s)
- Meng Liu
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150000, Heilongjiang, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Pengli Guo
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Mengnan Zeng
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Yuhan Zhang
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Jufang Jia
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Yanling Liu
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Xu Chen
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Haixue Kuang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150000, Heilongjiang, China
| | - Weisheng Feng
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China.
| | - Xiaoke Zheng
- Department of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China.
| |
Collapse
|
16
|
Long H, Lichtnekert J, Andrassy J, Schraml BU, Romagnani P, Anders HJ. Macrophages and fibrosis: how resident and infiltrating mononuclear phagocytes account for organ injury, regeneration or atrophy. Front Immunol 2023; 14:1194988. [PMID: 37868987 PMCID: PMC10587486 DOI: 10.3389/fimmu.2023.1194988] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
Mononuclear phagocytes (MP), i.e., monocytes, macrophages, and dendritic cells (DCs), are essential for immune homeostasis via their capacities to clear pathogens, pathogen components, and non-infectious particles. However, tissue injury-related changes in local microenvironments activate resident and infiltrating MP towards pro-inflammatory phenotypes that contribute to inflammation by secreting additional inflammatory mediators. Efficient control of injurious factors leads to a switch of MP phenotype, which changes the microenvironment towards the resolution of inflammation. In the same way, MP endorses adaptive structural responses leading to either compensatory hypertrophy of surviving cells, tissue regeneration from local tissue progenitor cells, or tissue fibrosis and atrophy. Under certain circumstances, MP contribute to the reversal of tissue fibrosis by clearance of the extracellular matrix. Here we give an update on the tissue microenvironment-related factors that, upon tissue injury, instruct resident and infiltrating MP how to support host defense and recover tissue function and integrity. We propose that MP are not intrinsically active drivers of organ injury and dysfunction but dynamic amplifiers (and biomarkers) of specific tissue microenvironments that vary across spatial and temporal contexts. Therefore, MP receptors are frequently redundant and suboptimal targets for specific therapeutic interventions compared to molecular targets upstream in adaptive humoral or cellular stress response pathways that influence tissue milieus at a contextual level.
Collapse
Affiliation(s)
- Hao Long
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany
- Department of Urology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Julia Lichtnekert
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Joachim Andrassy
- Department of General, Visceral and Transplant Surgery, University Hospital of Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Barbara U. Schraml
- Institute for Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-University (LMU), Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Paola Romagnani
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio”, University of Firenze, Nephrology and Dialysis Unit, Meyer Children’s Hospital, Firenze, Italy
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany
| |
Collapse
|
17
|
Yoshikawa T, Oguchi A, Toriu N, Sato Y, Kobayashi T, Ogawa O, Haga H, Sakurai S, Yamamoto T, Murakawa Y, Yanagita M. Tertiary Lymphoid Tissues Are Microenvironments with Intensive Interactions between Immune Cells and Proinflammatory Parenchymal Cells in Aged Kidneys. J Am Soc Nephrol 2023; 34:1687-1708. [PMID: 37548710 PMCID: PMC10561819 DOI: 10.1681/asn.0000000000000202] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 07/10/2023] [Indexed: 08/08/2023] Open
Abstract
SIGNIFICANCE STATEMENT Ectopic lymphoid structures called tertiary lymphoid tissues (TLTs) develop in several kidney diseases and are associated with poor renal prognosis. However, the mechanisms underlying TLT expansion and their effect on renal regeneration remain unclear. The authors report that single-nucleus RNA sequencing and validation experiments demonstrate that TLTs potentially amplify inflammation in aged injured kidneys. Lymphocytes within TLTs promote proinflammatory phenotypes of the surrounding proximal tubules and fibroblasts within the TLTs via proinflammatory cytokine production. These proinflammatory parenchymal cells then interact with immune cells by chemokine or cytokine production. Such cell-cell interactions potentially increase inflammation, expand TLTs, and exacerbate kidney injury. These findings help illuminate renal TLT pathology and suggest potential therapeutic targets. BACKGROUND Ectopic lymphoid structures called tertiary lymphoid tissues (TLTs) develop in several kidney diseases and are associated with poor renal prognosis. However, the mechanisms that expand TLTs and underlie exacerbation of kidney injury remain unclear. METHODS We performed single-nucleus RNA sequencing (snRNA-seq) on aged mouse kidneys with TLTs after ischemia-reperfusion injury. The results were validated using immunostaining, in situ hybridization of murine and human kidneys, and in vitro experiments. RESULTS Using snRNA-seq, we identified proinflammatory and profibrotic Vcam1+ injured proximal tubules (PTs) with NF κ B and IFN-inducible transcription factor activation. VCAM1 + PTs were preferentially localized around TLTs and drove inflammation and fibrosis via the production of multiple chemokines or cytokines. Lymphocytes within TLTs expressed Tnf and Ifng at high levels, which synergistically upregulated VCAM1 and chemokine expression in cultured PT cells. In addition, snRNA-seq also identified proinflammatory and profibrotic fibroblasts, which resided within and outside TLTs, respectively. Proinflammatory fibroblasts exhibited STAT1 activation and various chemokine or cytokine production, including CXCL9/CXCL10 and B cell-activating factor, contributing to lymphocyte recruitment and survival. IFN γ upregulated the expression of these molecules in cultured fibroblasts in a STAT1-dependent manner, indicating potential bidirectional interactions between IFN γ -producing CXCR3 + T cells and proinflammatory fibroblasts within TLTs. The cellular and molecular components described in this study were confirmed in human kidneys with TLTs. CONCLUSIONS These findings suggest that TLTs potentially amplify inflammation by providing a microenvironment that allows intense interactions between renal parenchymal and immune cells. These interactions may serve as novel therapeutic targets in kidney diseases involving TLT formation.
Collapse
Affiliation(s)
- Takahisa Yoshikawa
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akiko Oguchi
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Naoya Toriu
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Yuki Sato
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Kobayashi
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Osamu Ogawa
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hironori Haga
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoko Sakurai
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Takuya Yamamoto
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Yasuhiro Murakawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- IFOM-ETS, Milan, Italy
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| |
Collapse
|
18
|
Ma K, Liu JF, Zheng ZR, Li HY, Hu B, Meng Y. The polarization of M2 macrophages can be adjusted to alleviate renal injury by methylprednisolone in sepsis-AKI. Arch Biochem Biophys 2023; 747:109738. [PMID: 37696383 DOI: 10.1016/j.abb.2023.109738] [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: 06/17/2023] [Revised: 08/21/2023] [Accepted: 09/03/2023] [Indexed: 09/13/2023]
Abstract
Acute kidney injury in sepsis patients has an extreme mortality rate in clinical. It obviously seems that immune cells, for example, macrophages are involved with this process. Macrophages, as highly important immune cells, play a significant role in the development of human kidney diseases. But the specific role of macrophages in this process is still unclear. Under different timeline points, we surprisingly found that macrophages had the most dynamic changes in acute kidney injury immune cells. Based on macrophages' functions, they are primarily classified into M1 macrophages (pro-inflammatory) and M2 macrophages (anti-inflammatory). The polarization of M2 macrophages is closely associated with the seriousness of sepsis-induced kidney injury, but how to modulate their polarization to alleviate sepsis-associated renal damage remains unknown. We discovered that the polarization of M2 macrophages after methylprednisolone injection can significantly alleviate acute kidney injury by reducing secreted cytokine. This study suggests that the proportion of macrophage subtypes can be regulated by methylprednisolone to alleviate acute kidney injury in sepsis to provide a new sight for a clinical to provide a promising strategy for renal injury caused.
Collapse
Affiliation(s)
- Ke Ma
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510000, China; Institute of Nephrology, Jinan University, Guangzhou, 510632, China
| | - Jin-Feng Liu
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510000, China; Institute of Nephrology, Jinan University, Guangzhou, 510632, China
| | - Zi-Run Zheng
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510000, China; Institute of Nephrology, Jinan University, Guangzhou, 510632, China
| | - Hong-Yue Li
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510000, China; Institute of Nephrology, Jinan University, Guangzhou, 510632, China
| | - Bo Hu
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510000, China; Department of Nephrology, The Fifth Affiliated Hospital of Jinan University, Heyuan, 570000, China; Institute of Nephrology, Jinan University, Guangzhou, 510632, China.
| | - Yu Meng
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510000, China; Department of Nephrology, The Fifth Affiliated Hospital of Jinan University, Heyuan, 570000, China; Institute of Nephrology, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
19
|
Aoki M, Jin ZW, Ueda K, Kamimura G, Takeda-Harada A, Murakami G, Sato M. Localization of macrophages and dendritic cells in human thoracic lymph nodes: An immunohistochemical study using surgically obtained specimens. J Anat 2023; 243:504-516. [PMID: 37024113 PMCID: PMC10439373 DOI: 10.1111/joa.13870] [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: 11/29/2022] [Revised: 03/15/2023] [Accepted: 03/15/2023] [Indexed: 04/08/2023] Open
Abstract
Both dendritic cells (DCs) and macrophages are bone marrow-derived cells that perform antigen presentation. The distribution of DCs and CD68-positive macrophages were immunohistochemically examined in 103 thoracic nodes obtained from 23 lung cancer patients (50-84 years old) without metastasis. Among three antibodies tested initially-CD209/DCsign, fascin, and CD83-DCsign was chosen as the DC marker. For comparison, 137 nodes from 12 patients with cancer metastasis were also examined histologically. In patients without metastasis, DCs were found as (1) clusters along the subcapsular sinus and in a border area between the medullary sinus and cortex (mean sectional area of multiple nodes at one site, 8.4%) and, (2) rosette-like structures in the cortex (mean number in multiple nodes at one site, 20.5). Notably, DC clusters and rosettes contained no or few macrophages and were surrounded by smooth muscle actin (SMA)-positive, endothelium-like cells. The subcapsular linear cluster corresponded to 5%-85% (mean, 34.0%) of the nodal circumferential length and was shorter in older patients (p = 0.009). DC rosettes, solitary, or communicating with a cluster, were usually connected to a paracortical lymph sinus. Few differences were found between nodes with or without metastasis, but DC cluster sometimes contained abundant macrophages in cancer metastasis patients. The subcapsular DC cluster is not known in the rodent model, in which the subcapsular sinus is filled with macrophages. This quite different, even complementary, distribution suggests no, or less, cooperation between DCs and macrophages in humans.
Collapse
Affiliation(s)
- Masaya Aoki
- Department of General Thoracic Surgery, Kagoshima University School of Medicine, Kagoshima, Japan
| | - Zhe-Wu Jin
- Department of Anatomy, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Kazuhiro Ueda
- Department of General Thoracic Surgery, Kagoshima University School of Medicine, Kagoshima, Japan
| | - Go Kamimura
- Department of General Thoracic Surgery, Kagoshima University School of Medicine, Kagoshima, Japan
| | - Aya Takeda-Harada
- Department of General Thoracic Surgery, Kagoshima University School of Medicine, Kagoshima, Japan
| | - Gen Murakami
- Department of Anatomy, Tokyo Dental College, Tokyo, Japan
| | - Masami Sato
- Department of General Thoracic Surgery, Kagoshima University School of Medicine, Kagoshima, Japan
| |
Collapse
|
20
|
Cheng AS, Li X. The Potential Biotherapeutic Targets of Contrast-Induced Acute Kidney Injury. Int J Mol Sci 2023; 24:8254. [PMID: 37175958 PMCID: PMC10178966 DOI: 10.3390/ijms24098254] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Contrast-induced acute kidney injury (CI-AKI) is manifested by an abrupt decline in kidney function as a consequence of intravascular exposure to contrast media. With the increased applicability of medical imaging and interventional procedures that utilize contrast media for clinical diagnosis, CI-AKI is becoming the leading cause of renal dysfunction. The pathophysiological mechanism associated with CI-AKI involves renal medullary hypoxia, the direct toxicity of contrast agents, oxidative stress, apoptosis, inflammation, and epigenetic regulation. To date, there is no effective therapy for CI-AKI, except for the development of strategies that could reduce the toxicity profiles of contrast media. While most of these strategies have failed, evidence has shown that the proper use of personalized hydration, contrast medium, and high-dose statins may reduce the occurrence of CI-AKI. However, adequate risk predication and attempts to develop preventive strategies can be considered as the key determinants that can help eliminate CI-AKI. Additionally, a deeper understanding of the pathophysiological mechanism of CI-AKI is crucial to uncover molecular targets for the prevention of CI-AKI. This review has taken a step further to solidify the current known molecular mechanisms of CI-AKI and elaborate the biomarkers that are used to detect early-stage CI-AKI. On this foundation, this review will analyze the molecular targets relating to apoptosis, inflammation, oxidative stress, and epigenetics, and, thus, provide a strong rationale for therapeutic intervention in the prevention of CI-AKI.
Collapse
Affiliation(s)
- Alice Shasha Cheng
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
21
|
Bhargava R, Li H, Tsokos GC. Pathogenesis of lupus nephritis: the contribution of immune and kidney resident cells. Curr Opin Rheumatol 2023; 35:107-116. [PMID: 35797522 DOI: 10.1097/bor.0000000000000887] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Lupus nephritis is associated with significant mortality and morbidity. We lack effective therapeutics and biomarkers mostly because of our limited understanding of its complex pathogenesis. We aim to present an overview of the recent advances in the field to gain a deeper understanding of the underlying cellular and molecular mechanisms involved in lupus nephritis pathogenesis. RECENT FINDINGS Recent studies have identified distinct roles for each resident kidney cell in the pathogenesis of lupus nephritis. Podocytes share many elements of innate and adaptive immune cells and they can present antigens and participate in the formation of crescents in coordination with parietal epithelial cells. Mesangial cells produce pro-inflammatory cytokines and secrete extracellular matrix contributing to glomerular fibrosis. Tubular epithelial cells modulate the milieu of the interstitium to promote T cell infiltration and formation of tertiary lymphoid organs. Modulation of specific genes in kidney resident cells can ward off the effectors of the autoimmune response including autoantibodies, cytokines and immune cells. SUMMARY The development of lupus nephritis is multifactorial involving genetic susceptibility, environmental triggers and systemic inflammation. However, the role of resident kidney cells in the development of lupus nephritis is becoming more defined and distinct. More recent studies point to the restoration of kidney resident cell function using cell targeted approaches to prevent and treat lupus nephritis.
Collapse
Affiliation(s)
- Rhea Bhargava
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard, Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
22
|
Hoeft K, Schaefer GJL, Kim H, Schumacher D, Bleckwehl T, Long Q, Klinkhammer BM, Peisker F, Koch L, Nagai J, Halder M, Ziegler S, Liehn E, Kuppe C, Kranz J, Menzel S, Costa I, Wahida A, Boor P, Schneider RK, Hayat S, Kramann R. Platelet-instructed SPP1 + macrophages drive myofibroblast activation in fibrosis in a CXCL4-dependent manner. Cell Rep 2023; 42:112131. [PMID: 36807143 PMCID: PMC9992450 DOI: 10.1016/j.celrep.2023.112131] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/11/2022] [Accepted: 01/31/2023] [Indexed: 02/19/2023] Open
Abstract
Fibrosis represents the common end stage of chronic organ injury independent of the initial insult, destroying tissue architecture and driving organ failure. Here we discover a population of profibrotic macrophages marked by expression of Spp1, Fn1, and Arg1 (termed Spp1 macrophages), which expands after organ injury. Using an unbiased approach, we identify the chemokine (C-X-C motif) ligand 4 (CXCL4) to be among the top upregulated genes during profibrotic Spp1 macrophage differentiation. In vitro and in vivo studies show that loss of Cxcl4 abrogates profibrotic Spp1 macrophage differentiation and ameliorates fibrosis after both heart and kidney injury. Moreover, we find that platelets, the most abundant source of CXCL4 in vivo, drive profibrotic Spp1 macrophage differentiation. Single nuclear RNA sequencing with ligand-receptor interaction analysis reveals that macrophages orchestrate fibroblast activation via Spp1, Fn1, and Sema3 crosstalk. Finally, we confirm that Spp1 macrophages expand in both human chronic kidney disease and heart failure.
Collapse
Affiliation(s)
- Konrad Hoeft
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany; Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Gideon J L Schaefer
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany; Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Hyojin Kim
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - David Schumacher
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany; Department of Anesthesiology, RWTH Aachen University, Aachen, Germany
| | - Tore Bleckwehl
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Qingqing Long
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | | | - Fabian Peisker
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Lars Koch
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany; Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - James Nagai
- Institute for Computational Genomics, RWTH Aachen University Hospital, Aachen, Germany; Joint Research Center for Computational Biomedicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Maurice Halder
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Susanne Ziegler
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Elisa Liehn
- Institute for Molecular Medicine, University of South Denmark, Odense, Denmark
| | - Christoph Kuppe
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany; Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Jennifer Kranz
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany; Department of Urology, RWTH Aachen University, Aachen, Germany; Department of Urology and Kidney Transplantation, Martin-Luther-University, Halle (Saale), Germany
| | - Sylvia Menzel
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Ivan Costa
- Institute for Computational Genomics, RWTH Aachen University Hospital, Aachen, Germany; Joint Research Center for Computational Biomedicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Adam Wahida
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany; Division of Gynecological Oncology, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Peter Boor
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany; Department of Pathology, RWTH Aachen University, Aachen, Germany
| | - Rebekka K Schneider
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands; Department of Cell Biology, Institute for Biomedical Technologies, RWTH Aachen University, Aachen, Germany
| | - Sikander Hayat
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Rafael Kramann
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany; Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany; Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
23
|
The role of non-coding RNA in lupus nephritis. Hum Cell 2023; 36:923-936. [PMID: 36840837 DOI: 10.1007/s13577-023-00883-w] [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: 11/19/2022] [Accepted: 02/16/2023] [Indexed: 02/26/2023]
Abstract
Systemic lupus erythematosus (SLE) is a common autoimmune disease with multiple manifestations. The renal implication, also called lupus nephritis (LN) is the most regular type of complication and results in adverse outcomes. Multiple studies revealed the importance of non-coding RNA in diseases, likewise observed in nephropathies, particularly LN. Long-non-coding RNA (lncRNA) is a group of RNA that are more than 200 nucleotides in length. And in circular RNA (circRNA), the head and tail of RNA are connected by a 3' → 5' phosphodiester bond. Both two types of non-coding RNA play important roles in LN pathogenesis through the competitive endogenous RNA (ceRNA) effect. LncRNAs and circRNAs can sponge miRNAs and consequently act on downstream signaling pathways, which are capable to influence various aspects of LN, including cell proliferation, inflammation, and oxidative stress. And lncRNAs and circRNAs have the potential to act as biomarkers to diagnose LN and distinguish whether SLE patients with LN or not. In the future, lncRNAs and circRNAs may be accessible therapeutic targets.
Collapse
|
24
|
Alduraibi FK, Sullivan KA, Chatham WW, Hsu HC, Mountz JD. Interrelation of T cell cytokines and autoantibodies in systemic lupus erythematosus: A cross-sectional study. Clin Immunol 2023; 247:109239. [PMID: 36682593 PMCID: PMC10118038 DOI: 10.1016/j.clim.2023.109239] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/11/2023] [Accepted: 01/15/2023] [Indexed: 01/21/2023]
Abstract
T-helper cytokines interferon gamma (IFNɣ), interleukin 17 (IL-17) and IL-10 impact systemic lupus erythematosus (SLE) directly and indirectly via modulation of autoAb production. We determined the separate and combined effects on clinical manifestations of SLE (N = 62). IFNɣ, IL-17 but not IL-10 were significantly elevated in patients with SLE. IFNɣ positively correlated with anti-DNA and anti-SSA. IL-17 positively correlated with anti-SSA and was significantly higher in patients with discoid rash and class V LN. IL-10 did not correlate with circulating autoantibodies but was significantly elevated in patients with LN. Patients with LN had elevated plasma levels of anti-DNA and anti-Sm/ribonuclear protein (RNP). Anti-Sm/RNP levels were decreased in patients with acute mucocutaneous manifestations, including photosensitivity and/or malar rash. The study provides critical insights into pathological mechanisms of LN, which could help guide future diagnoses and therapies.
Collapse
Affiliation(s)
- Fatima K Alduraibi
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA; Medicine Service, Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA; Department of Medicine, Division of Clinical Immunology and Rheumatology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Kathryn A Sullivan
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - W Winn Chatham
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hui-Chen Hsu
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - John D Mountz
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA; Medicine Service, Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA.
| |
Collapse
|
25
|
The Response of Macrophages in Sepsis-Induced Acute Kidney Injury. J Clin Med 2023; 12:jcm12031101. [PMID: 36769749 PMCID: PMC9917612 DOI: 10.3390/jcm12031101] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/23/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Sepsis-induced acute kidney injury (SAKI) is common in critically ill patients and often leads to poor prognosis. At present, the pathogenesis of SAKI has not been fully clarified, and there is no effective treatment. Macrophages are immune cells that play an important role in the pathogenesis of SAKI. The phenotype and role of macrophages can vary from early to later stages of SAKI. Elucidating the role of macrophages in SAKI will be beneficial to its diagnosis and treatment. This article reviews past studies describing the role of macrophages in SAKI, with the aim of identifying novel therapeutic targets.
Collapse
|
26
|
Liu T, Zhuang XX, Qin XJ, Wei LB, Gao JR. Identifying effective diagnostic biomarkers and immune infiltration features in chronic kidney disease by bioinformatics and validation. Front Pharmacol 2022; 13:1069810. [PMID: 36642989 PMCID: PMC9838551 DOI: 10.3389/fphar.2022.1069810] [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: 10/14/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022] Open
Abstract
Background: Chronic kidney disease (CKD), characterized by sustained inflammation and immune dysfunction, is highly prevalent and can eventually progress to end-stage kidney disease. However, there is still a lack of effective and reliable diagnostic markers and therapeutic targets for CKD. Methods: First, we merged data from GEO microarrays (GSE104948 and GSE116626) to identify differentially expressed genes (DEGs) in CKD and healthy patient samples. Then, we conducted GO, KEGG, HPO, and WGCNA analyses to explore potential functions of DEGs and select clinically significant modules. Moreover, STRING was used to analyse protein-protein interactions. CytoHubba and MCODE algorithms in the cytoscape plug-in were performed to screen hub genes in the network. We then determined the diagnostic significance of the obtained hub genes by ROC and two validation datasets. Meanwhile, the expression level of the biomarkers was verified by IHC. Furthermore, we examined immunological cells' relationships with hub genes. Finally, GSEA was conducted to determine the biological functions that biomarkers are significantly enriched. STITCH and AutoDock Vina were used to predict and validate drug-gene interactions. Results: A total of 657 DEGs were screened and functional analysis emphasizes their important role in inflammatory responses and immunomodulation in CKD. Through WGCNA, the interaction network, ROC curves, and validation set, four hub genes (IL10RA, CD45, CTSS, and C1QA) were identified. Furthermore, IHC of CKD patients confirmed the results above. Immune infiltration analysis indicated that CKD had a significant increase in monocytes, M0 macrophages, and M1 macrophages but a decrease in regulatory T cells, activated dendritic cells, and so on. Moreover, four hub genes were statistically correlated with them. Further analysis exhibited that IL10RA, which obtained the highest expression level in hub genes, was involved in abnormalities in various immune cells and regulated a large number of immune system responses and inflammation-related pathways. In addition, the drug-gene interaction network contained four potential therapeutic drugs targeting IL10RA, and molecular docking might make this relationship viable. Conclusion: IL10RA and its related hub molecules might play a key role in the development of CKD and could be potential biomarkers in CKD.
Collapse
Affiliation(s)
- Tao Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Xing Xing Zhuang
- Department of Pharmacy, Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Xiu Juan Qin
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Liang Bing Wei
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Jia Rong Gao
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China,*Correspondence: Jia Rong Gao,
| |
Collapse
|
27
|
McEvoy CM, Murphy JM, Zhang L, Clotet-Freixas S, Mathews JA, An J, Karimzadeh M, Pouyabahar D, Su S, Zaslaver O, Röst H, Arambewela R, Liu LY, Zhang S, Lawson KA, Finelli A, Wang B, MacParland SA, Bader GD, Konvalinka A, Crome SQ. Single-cell profiling of healthy human kidney reveals features of sex-based transcriptional programs and tissue-specific immunity. Nat Commun 2022; 13:7634. [PMID: 36496458 PMCID: PMC9741629 DOI: 10.1038/s41467-022-35297-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 11/27/2022] [Indexed: 12/13/2022] Open
Abstract
Knowledge of the transcriptional programs underpinning the functions of human kidney cell populations at homeostasis is limited. We present a single-cell perspective of healthy human kidney from 19 living donors, with equal contribution from males and females, profiling the transcriptome of 27677 cells to map human kidney at high resolution. Sex-based differences in gene expression within proximal tubular cells were observed, specifically, increased anti-oxidant metallothionein genes in females and aerobic metabolism-related genes in males. Functional differences in metabolism were confirmed in proximal tubular cells, with male cells exhibiting higher oxidative phosphorylation and higher levels of energy precursor metabolites. We identified kidney-specific lymphocyte populations with unique transcriptional profiles indicative of kidney-adapted functions. Significant heterogeneity in myeloid cells was observed, with a MRC1+LYVE1+FOLR2+C1QC+ population representing a predominant population in healthy kidney. This study provides a detailed cellular map of healthy human kidney, and explores the complexity of parenchymal and kidney-resident immune cells.
Collapse
Affiliation(s)
- Caitriona M McEvoy
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, ON, Canada
| | - Julia M Murphy
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Lin Zhang
- Department of Statistical Sciences, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Sergi Clotet-Freixas
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Jessica A Mathews
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - James An
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Mehran Karimzadeh
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Vector Institute, Toronto, ON, Canada
| | - Delaram Pouyabahar
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Shenghui Su
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Olga Zaslaver
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Hannes Röst
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Rangi Arambewela
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Lewis Y Liu
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Sally Zhang
- Division of Urology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Keith A Lawson
- Division of Urology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Antonio Finelli
- Division of Urology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Bo Wang
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Vector Institute, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Sonya A MacParland
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Gary D Bader
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Ana Konvalinka
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada.
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, ON, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| | - Sarah Q Crome
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada.
- Department of Immunology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
28
|
Fu J, Sun Z, Wang X, Zhang T, Yuan W, Salem F, Yu SMW, Zhang W, Lee K, He JC. The single-cell landscape of kidney immune cells reveals transcriptional heterogeneity in early diabetic kidney disease. Kidney Int 2022; 102:1291-1304. [PMID: 36108806 PMCID: PMC9691617 DOI: 10.1016/j.kint.2022.08.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 08/03/2022] [Accepted: 08/12/2022] [Indexed: 01/12/2023]
Abstract
The pathogenesis of diabetic kidney disease (DKD) involves multifactorial processes that converge to initiate and advance the disease. Although DKD is not typically classified as an inflammatory glomerular disease, mounting evidence supports the involvement of kidney inflammation as a key contributor in DKD pathogenesis, particularly through macrophages. However, detailed identification and corresponding phenotypic changes of macrophages in DKD remain poorly understood. To capture the gene expression changes in specific macrophage cell subsets in early DKD, we performed single-cell transcriptomic analysis of CD45-enriched kidney immune cells from type 1 diabetic OVE26 mice at two time points during the disease development. We also undertook a focused analysis of mononuclear phagocytes (macrophages and dendritic cells). Our results show increased resident and infiltrating macrophage subsets in the kidneys of mice with diabetes over time, with heightened expression of pro-inflammatory or anti-inflammatory genes in a subset-specific manner. Further analysis of macrophage polarization states in each subset in the kidneys showed changes consistent with the continuum of activation and differentiation states, with gene expression tending to shift toward undifferentiated phenotypes but with increased M1-like inflammatory phenotypes over time. By deconvolution analysis of RNAseq samples and by immunostaining of biopsies from patients with DKD, we further confirmed a differential expression of select genes in specific macrophage subsets essentially recapitulating the studies in mice. Thus, our study provides a comprehensive analysis of macrophage transcriptomic profiles in early DKD that underscores the dynamic macrophage phenotypes in disease progression.
Collapse
Affiliation(s)
- Jia Fu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Zeguo Sun
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Xuan Wang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Division of Nephrology, Department of Medicine, Shanghai First People Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Tuo Zhang
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, USA
| | - Weijie Yuan
- Division of Nephrology, Department of Medicine, Shanghai First People Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Fadi Salem
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Samuel Mon-Wei Yu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Weijia Zhang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kyung Lee
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Renal Program, James J Peters VA Medical Center at Bronx, Bronx, New York, USA.
| |
Collapse
|
29
|
Wynne BM, Samson TK, Moyer HC, van Elst HJ, Moseley AS, Hecht G, Paul O, Al-Khalili O, Gomez-Sanchez C, Ko B, Eaton DC, Hoover RS. Interleukin 6 mediated activation of the mineralocorticoid receptor in the aldosterone-sensitive distal nephron. Am J Physiol Cell Physiol 2022; 323:C1512-C1523. [PMID: 35912993 PMCID: PMC9662807 DOI: 10.1152/ajpcell.00272.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 06/28/2022] [Accepted: 07/11/2022] [Indexed: 11/22/2022]
Abstract
Hypertension is characterized by increased sodium (Na+) reabsorption along the aldosterone-sensitive distal nephron (ASDN) as well as chronic systemic inflammation. Interleukin-6 (IL-6) is thought to be a mediator of this inflammatory process. Interestingly, increased Na+ reabsorption within the ASDN does not always correlate with increases in aldosterone (Aldo), the primary hormone that modulates Na+ reabsorption via the mineralocorticoid receptor (MR). Thus, understanding how increased ASDN Na+ reabsorption may occur independent of Aldo stimulation is critical. Here, we show that IL-6 can activate the MR by activating Rac1 and stimulating the generation of reactive oxygen species (ROS) with a consequent increase in thiazide-sensitive Na+ uptake. Using an in vitro model of the distal convoluted tubule (DCT2), mDCT15 cells, we observed nuclear translocation of eGFP-tagged MR after IL-6 treatment. To confirm the activation of downstream transcription factors, mDCT15 cells were transfected with mineralocorticoid response element (MRE)-luciferase reporter constructs; then treated with vehicle, Aldo, or IL-6. Aldosterone or IL-6 treatment increased luciferase activity that was reversed with MR antagonist cotreatment, but IL-6 treatment was reversed by Rac1 inhibition or ROS reduction. In both mDCT15 and mpkCCD cells, IL-6 increased amiloride-sensitive transepithelial Na+ current. ROS and IL-6 increased 22Na+ uptake via the thiazide-sensitive sodium chloride cotransporter (NCC). These results are the first to demonstrate that IL-6 can activate the MR resulting in MRE activation and that IL-6 increases NCC-mediated Na+ reabsorption, providing evidence for an alternative mechanism for stimulating ASDN Na+ uptake during conditions where Aldo-mediated MR stimulation may not occur.
Collapse
Affiliation(s)
- Brandi M Wynne
- Department of Medicine, Nephrology, Emory University, Atlanta, Georgia
- Department of Internal Medicine, Nephrology & Hypertension, University of Utah, Salt Lake City, Utah
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
- Immunology, Inflammation and Infectious Disease Initiative, University of Utah, Salt Lake City, Utah
| | - Trinity K Samson
- Department of Medicine, Nephrology, Emory University, Atlanta, Georgia
| | - Hayley C Moyer
- Department of Medicine, Nephrology, Emory University, Atlanta, Georgia
| | - Henrieke J van Elst
- Department of Medicine, Nephrology, Emory University, Atlanta, Georgia
- Department of Physiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Auriel S Moseley
- Department of Medicine, Nephrology, Emory University, Atlanta, Georgia
| | - Gillian Hecht
- Department of Medicine, Nephrology, Emory University, Atlanta, Georgia
| | - Oishi Paul
- Department of Medicine, Nephrology, Emory University, Atlanta, Georgia
| | - Otor Al-Khalili
- Department of Medicine, Nephrology, Emory University, Atlanta, Georgia
| | - Celso Gomez-Sanchez
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Mississippi
| | - Benjamin Ko
- Department of Medicine, Nephrology, University of Chicago, Chicago, Illinois
| | - Douglas C Eaton
- Department of Medicine, Nephrology, Emory University, Atlanta, Georgia
| | - Robert S Hoover
- Department of Medicine, Nephrology, Emory University, Atlanta, Georgia
- Research Service, Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
- Section of Nephrology and Hypertension, Deming Department of Medicine, Tulane University, New Orleans, Louisiana
| |
Collapse
|
30
|
The Importance of Toll-like Receptor 9 Expression on Monocytes and Dendritic Cells in the Context of Epstein–Barr Virus Infection in the Immunopathogenesis of Primary Glomerulonephritis. Int J Mol Sci 2022; 23:ijms231911796. [PMID: 36233099 PMCID: PMC9570264 DOI: 10.3390/ijms231911796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/24/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Toll-like receptor 9 (TLR9) is activated by unmethylated cytosine-phosphate-guanosine (CpG) dinucleotides found in the genomes of pathogens such as Epstein–Barr virus (EBV). The aim of this study was to determine the role of TLR9 in the immunopathogenesis of IgA nephropathy (IgAN) and membranoproliferative glomerulonephritis (MPGN) in the context of Epstein–Barr virus (EBV) infection. For this purpose, the frequency of TLR9-positive monocytes and dendritic cells (DCs, i.e., BDCA-1; myeloid dendritic cells, and BDCA-2; plasmocytoid dendritic cells) was studied, and a quantitative analysis of the concentration of TLR9 in the serum of patients diagnosed with IgAN and MPGN was undertaken. Higher frequencies of TLR9-positive DCs and monocytes in IgAN and MPGN patients were observed as compared with the control group. Patients diagnosed with GN exhibited a higher percentage of BDCA-1+CD19− and BDCA-2+CD123+ DCs than patients in the control group. Moreover, serum TLR9 concentration was shown to be significantly correlated with EBV DNA copy number/µg DNA, IgG, IgM, serum albumin, total protein in 24-h urine collection test and the frequency of BDCA-2+CD123+ DCs in peripheral blood. Our findings confirm that TLR9 may be involved in the development of IgAN and MPGN.
Collapse
|
31
|
Li X, Zhou X, Liu X, Li X, Jiang X, Shi B, Wang S. Spermidine protects against acute kidney injury by modulating macrophage NLRP3 inflammasome activation and mitochondrial respiration in an eIF5A hypusination-related pathway. Mol Med 2022; 28:103. [PMID: 36058905 PMCID: PMC9441050 DOI: 10.1186/s10020-022-00533-1] [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: 03/25/2022] [Accepted: 08/21/2022] [Indexed: 11/26/2022] Open
Abstract
Background Acute kidney injury (AKI) is still a critical problem in clinical practice, with a heavy burden for national health system around the world. It is notable that sepsis is the predominant cause of AKI for patients in the intensive care unit and the mortality remains considerably high. The treatment for AKI relies on supportive therapies and almost no specific treatment is currently available. Spermidine is a naturally occurring polyamine with pleiotropic effects. However, the renoprotective effect of spermidine and the underlying mechanism remain elusive. Methods We employed mice sepsis-induced AKI model and explored the potential renoprotective effect of spermidine in vivo with different administration time and routes. Macrophage depleting was utilized to probe the role of macrophage. In vitro experiments were conducted to examine the effect of spermidine on macrophage cytokine secretion, NLRP3 inflammasome activation and mitochondrial respiration. Results We confirmed that spermidine improves AKI with different administration time and routes and that macrophages serves as an essential mediator in this protective effect. Meanwhile, spermidine downregulates NOD-like receptor protein 3 (NLRP3) inflammasome activation and IL-1 beta production in macrophages directly. Mechanically, spermidine enhances mitochondrial respiration capacity and maintains mitochondria function which contribute to the NLRP3 inhibition. Importantly, we showed that eukaryotic initiation factor 5A (eIF5A) hypusination plays an important role in regulating macrophage bioactivity. Conclusions Spermidine administration practically protects against sepsis-induced AKI in mice and macrophages serve as an essential mediator in this protective effect. Our study identifies spermidine as a promising pharmacologic approach to prevent AKI. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00533-1.
Collapse
Affiliation(s)
- Xianzhi Li
- Department of Urology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China.,Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, 250014, China
| | - Xiaojun Zhou
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, 250014, China
| | - Xigao Liu
- Department of Urology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Xiaoyun Li
- Department of Otolaryngology, Qingdao Eighth People's Hospital, Qingdao, 266121, China
| | - Xianzhou Jiang
- Department of Urology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Benkang Shi
- Department of Urology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Shuo Wang
- Department of Urology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
32
|
Lin L, Hu K. Annexin A2 and Kidney Diseases. Front Cell Dev Biol 2022; 10:974381. [PMID: 36120574 PMCID: PMC9478026 DOI: 10.3389/fcell.2022.974381] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/08/2022] [Indexed: 11/22/2022] Open
Abstract
Annexin A2 is a Ca2+- and phospholipid-binding protein which is widely expressed in various types of cells and tissues. As a multifunctional molecule, annexin A2 is found to be involved in diverse cell functions and processes, such as cell exocytosis, endocytosis, migration and proliferation. As a receptor of plasminogen and tissue plasminogen activator, annexin A2 promotes plasmin generation and regulates the homeostasis of blood coagulation, fibrinolysis and matrix degradation. As an antigen expressed on cell membranes, annexin A2 initiates local inflammation and damage through binding to auto-antibodies. Annexin A2 also mediates multiple signaling pathways induced by various growth factors and oxidative stress. Aberrant expression of annexin A2 has been found in numerous kidney diseases. Annexin A2 has been shown to act as a co-receptor of integrin CD11b mediating NF-kB-dependent kidney inflammation, which is further amplified through annexin A2/NF-kB-triggered macrophage M2 to M1 phenotypic change. It also modulates podocyte cytoskeleton rearrangement through Cdc42 and Rac1/2/3 Rho pathway causing proteinuria. Thus, annexin A2 is implicated in the pathogenesis and progression of various kidney diseases. In this review, we focus on the current understanding of the role of annexin A2 in kidney diseases.
Collapse
Affiliation(s)
- Ling Lin
- *Correspondence: Ling Lin, ; Kebin Hu,
| | - Kebin Hu
- *Correspondence: Ling Lin, ; Kebin Hu,
| |
Collapse
|
33
|
Corneal Dendritic Cell Dynamics Are Associated with Clinical Factors in Type 1 Diabetes. J Clin Med 2022; 11:jcm11092611. [PMID: 35566743 PMCID: PMC9101330 DOI: 10.3390/jcm11092611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 01/11/2023] Open
Abstract
Time-lapsed in vivo corneal confocal microscopy (IVCCM) has shown that corneal dendritic cells (DCs) migrate at approximately 1 µm/min in healthy humans. We have undertaken IVCCM of the whorl region to compare the density of rounded DCs, and DCs with (wDCs) and without (woDCs) dendrites and dynamics; trajectory (length travelled/time), displacement (distance from origin to endpoint/time) speeds and persistence ratio (displacement/trajectory) of woDCs in subjects with type 1 diabetes (T1D) (n = 20) and healthy controls (n = 10). Only the wDC density was higher (p = 0.02) in subjects with T1D compared to controls. There was no significant difference in cell dynamics between subjects with T1D and controls. woDC density correlated directly with HDL cholesterol (r = 0.59, p = 0.007) and inversely with triglycerides (r = −0.61, p = 0.005), whilst round-shaped cell density correlated inversely with HDL cholesterol (r = −0.54, p = 0.007). Displacement, trajectory, and persistency correlated significantly with eGFR (mL/min) (r = 0.74, p < 0.001; r = 0.48, p = 0.031; r = 0.58, p = 0.008, respectively). We show an increase in wDC density but no change in any other DC sub-type or alteration in cell dynamics in T1D. However, there were associations between DC density and lipid parameters and between DC dynamics and renal function. IVCCM provides evidence of a link between immune cell dynamics with lipid levels and renal function.
Collapse
|
34
|
Kim K, Kim YG, Jung SW, Kim YG, Lee SH, Kwon SH, Moon JY. Three-Dimensional Visualization With Tissue Clearing Uncovers Dynamic Alterations of Renal Resident Mononuclear Phagocytes After Acute Kidney Injury. Front Immunol 2022; 13:844919. [PMID: 35359999 PMCID: PMC8960144 DOI: 10.3389/fimmu.2022.844919] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/15/2022] [Indexed: 12/19/2022] Open
Abstract
Traditional histologic methods are limited in detecting dynamic changes in immune cells during acute kidney injury (AKI). Recently, optical tissue clearing combined with multiphoton microscopy (MPM) or light sheet fluorescence microscopy (LSFM) has become an emerging method for deep tissue evaluation and three-dimensional visualization. These new approaches have helped expand our understanding of tissue injury and repair processes, including tracing the changes in immune cells. We designed this study to investigate the morphological and functional alterations of renal mononuclear phagocytes (MNPs) in lipopolysaccharide (LPS)-induced AKI using renal clearing in CD11c-YFP mice. We also evaluated the effect of the NLRP3 inhibitor MCC950 to determine whether NLRP3 inhibition attenuates the activation of CD11c+ cells in an LPS-induced AKI model. Transverse sectioned whole mouse kidney imaging by LSFM showed that CD11c+ cells were mainly distributed in the cortex, especially the tubulointerstitial area. The number of CD11c+ cells was significantly more densely interspersed, particularly in periglomerular and perivascular lesions, in the saline-treated LPS-exposed kidney than in the control kidney. Deep imaging of the kidney cortex by MPM demonstrated an increased number of CD11c+ cells in the saline-treated LPS group compared with the control group. This quantitative alteration of CD11c+ cells in AKI was accompanied by morphological changes at high resolution, showing an increased number and level of dendrites. These morphological and behavioral changes in the saline-treated LPS group were accompanied by increased MHC class II and CD86 on CD11c-YFP+ cells. MCC950 attenuated the activation of CD11c+ cells after AKI and improved renal function. In conclusion, wide and deep three-dimensional visualization using MPM or LSFM combined with kidney clearing uncovers dynamic changes of renal MNPs, which are directly linked to renal function in AKI.
Collapse
Affiliation(s)
- Kipyo Kim
- Division of Nephrology and Hypertension, Department of Internal Medicine, Inha University School of Medicine, Incheon, South Korea
| | - Yun-Gyeong Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, College of Medicine, Seoul, South Korea
| | - Su Woong Jung
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, College of Medicine, Seoul, South Korea
| | - Yang Gyun Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, College of Medicine, Seoul, South Korea
| | - Sang-Ho Lee
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, College of Medicine, Seoul, South Korea
| | - Seung-Hae Kwon
- Korea Basic Science Institute, Seoul Center, Seoul, South Korea
| | - Ju-Young Moon
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, College of Medicine, Seoul, South Korea
| |
Collapse
|
35
|
He Y, Deng B, Liu S, Luo S, Ning Y, Pan X, Wan R, Chen Y, Zhang Z, Jiang J, Xu H, Xia M, Li J. Myeloid
Piezo1
Deletion Protects Renal Fibrosis by Restraining Macrophage Infiltration and Activation. Hypertension 2022; 79:918-931. [DOI: 10.1161/hypertensionaha.121.18750] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background:
Macrophages play important roles in renal fibrosis, partially by sensing mechanical forces, including shear stress and increased stiffness. The mechanically activated cationic channel Piezo1 drives vascular formation and blood pressure regulation to inflammatory responses, or cancer, but its role in macrophages in fibrotic kidney is elusive. Here, we hypothesized that Piezo1 in macrophages may have functions in renal fibrosis.
Methods:
We established a genetically engineered mouse model with Piezo1 specific knockout in myeloid cells and challenged with unilateral ureteric obstruction operation and folic acid treatment to induce the renal fibrosis, aiming to investigate the function of the mechanical-sensitive protein Piezo1 in macrophages in renal fibrosis and its underlying mechanisms.
Results:
Myeloid
Piezo1
was indispensable for renal fibrosis generation.
Piezo1
gene deletion in the myeloid lineage was protective in mice with renal fibrosis. Further analyses revealed that macrophage accumulation in the injured kidney depended on the Piezo1-regulated C-C motif chemokine ligand 2, C-C motif chemokine receptor 2 pathway, and Notch signaling cascade. Moreover,
Piezo1
deletion restrained macrophage inflammation and consequently suppressed kidney fibrosis and epithelial-mesenchymal transition. In vitro assays showed that
Piezo1
deficiency blocked lipopolysaccharide and Piezo1 activation-induced inflammatory responses in bone marrow–derived macrophages. Mechanistically, Piezo1 regulated inflammation through the Ca
2+
-dependent intracellular cysteine protease, as the pharmacological inhibition of calpain blocked the proinflammatory role of Piezo1.
Conclusions:
This study characterized the important function of Piezo1 in renal fibrosis. Targeting the Piezo1 channels by genetic or pharmacological manipulations may be a promising strategy for the treatment of renal fibrosis.
Collapse
Affiliation(s)
- Yu He
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
| | - Bo Deng
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
| | - Silin Liu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
| | - Shangfei Luo
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
| | - Yile Ning
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
| | - Xianmei Pan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
| | - Rentao Wan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
| | - Yuan Chen
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
| | - Ziyan Zhang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
| | - Jintao Jiang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
| | - Honglin Xu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
| | - Mingfeng Xia
- Medical Research Center, Shandong University of Chinese Medicine, Jinan, China (M.X.)
| | - Jing Li
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine. (Y.H., B.D., S. Liu, S. Luo, Y.N., X.P., R.W., Y.C., Z.Z., J.J., H.X., J.L.)
- Faculty of Biological Sciences, University of Leeds, United Kingdom (J.L.)
| |
Collapse
|
36
|
Abstract
Macrophages have emerged at the forefront of research in immunology and transplantation because of recent advances in basic science. New findings have illuminated macrophage populations not identified previously, expanded upon traditional macrophage phenotypes, and overhauled macrophage ontogeny. These advances have major implications for the field of transplant immunology. Macrophages are known to prime adaptive immune responses, perpetuate T-cell-mediated rejection and antibody-mediated rejection, and promote allograft fibrosis. In this review, macrophage phenotypes and their role in allograft injury of solid organ transplants will be discussed with an emphasis on kidney transplantation. Additionally, consideration will be given to the prospect of manipulating macrophage phenotypes as cell-based therapy. Innate immunity and macrophages represent important players in allograft injury and a promising target to improve transplant outcomes.
Collapse
Affiliation(s)
- Sarah E. Panzer
- Department of Medicine, Division of Nephrology, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
37
|
Kitching AR, Hickey MJ. Immune cell behaviour and dynamics in the kidney - insights from in vivo imaging. Nat Rev Nephrol 2022; 18:22-37. [PMID: 34556836 DOI: 10.1038/s41581-021-00481-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2021] [Indexed: 02/08/2023]
Abstract
The actions of immune cells within the kidney are of fundamental importance in kidney homeostasis and disease. In disease settings such as acute kidney injury, anti-neutrophil cytoplasmic antibody-associated vasculitis, lupus nephritis and renal transplant rejection, immune cells resident within the kidney and those recruited from the circulation propagate inflammatory responses with deleterious effects on the kidney. As in most forms of inflammation, intravital imaging - particularly two-photon microscopy - has been critical to our understanding of immune cell responses in the renal microvasculature and interstitium, enabling visualization of immune cell dynamics over time rather than statically. These studies have demonstrated differences in the recruitment and function of these cells from those in more conventional vascular beds, and provided a wealth of information on the actions of blood-borne immune cells such as neutrophils, monocytes and T cells, as well as kidney-resident mononuclear phagocytes, in a range of diseases affecting different kidney compartments. In particular, in vivo imaging has furthered our understanding of leukocyte function within the glomerulus in acute glomerulonephritis, and in the tubulointerstitium and interstitial microvasculature during acute kidney injury and following transplantation, revealing mechanisms of immune surveillance, antigen presentation and inflammation in the kidney.
Collapse
Affiliation(s)
- A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia. .,Departments of Nephrology and Paediatric Nephrology, Monash Medical Centre, Clayton, Victoria, Australia.
| | - Michael J Hickey
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| |
Collapse
|
38
|
Mikulin JA, Bates BL, Wilson TJ. A simplified method for separating renal MPCs using SLAMF9. Cytometry A 2021; 99:1209-1217. [PMID: 34092043 PMCID: PMC9930532 DOI: 10.1002/cyto.a.24469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/01/2021] [Accepted: 05/31/2021] [Indexed: 12/23/2022]
Abstract
Mononuclear phagocytes comprise an array of tissue-resident and monocyte-derived cells with important roles in tissue homeostasis and resistance to infection. Their diverse phenotypes make functional characterization within tissues challenging, because multiple surface markers are typically required for subset identification and isolation by cell sorting methods. Analysis of SLAMF9 expression within renal mononuclear phagocyte populations by multi-parametric flow cytometry indicates that SLAMF9 is a specific marker for identification of kidney-resident CD45+ CD11c+ MHC-II+ cells corresponding to prominent tissue-resident MPC populations derived from dendritic cell progenitors in adult mice. High SLAMF9 expression was sufficient to identify and sort these cells from disaggregated tissue using a user-operated cell sorter. The population can be further subdivided according to expression of CD11b and CD14 to identify IRF8high cDC1 cells and cleanly separate the CD11bhigh F4/80low and CD11bint F4/80high CD11c+ MPC subsets. Therefore, SLAMF9 expression allows for the identification and sorting of kidney-resident CD11b+ CD11c+ CD64+ F4/80+ CX3 CR1+ MHC-II+ MPCs without the need for complex antibody panels or reporter mice, simplifying isolation of these cells for study ex vivo.
Collapse
|
39
|
Li J, Thomson AW, Rogers NM. Myeloid and Mesenchymal Stem Cell Therapies for Solid Organ Transplant Tolerance. Transplantation 2021; 105:e303-e321. [PMID: 33756544 PMCID: PMC8455706 DOI: 10.1097/tp.0000000000003765] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Transplantation is now performed globally as a routine procedure. However, the increased demand for donor organs and consequent expansion of donor criteria has created an imperative to maximize the quality of these gains. The goal is to balance preservation of allograft function against patient quality-of-life, despite exposure to long-term immunosuppression. Elimination of immunosuppressive therapy to avoid drug toxicity, with concurrent acceptance of the allograft-so-called operational tolerance-has proven elusive. The lack of recent advances in immunomodulatory drug development, together with advances in immunotherapy in oncology, has prompted interest in cell-based therapies to control the alloimmune response. Extensive experimental work in animals has characterized regulatory immune cell populations that can induce and maintain tolerance, demonstrating that their adoptive transfer can promote donor-specific tolerance. An extension of this large body of work has resulted in protocols for manufacture, as well as early-phase safety and feasibility trials for many regulatory cell types. Despite the excitement generated by early clinical trials in autoimmune diseases and organ transplantation, there is as yet no clinically validated, approved regulatory cell therapy for transplantation. In this review, we summarize recent advances in this field, with a focus on myeloid and mesenchymal cell therapies, including current understanding of the mechanisms of action of regulatory immune cells, and clinical trials in organ transplantation using these cells as therapeutics.
Collapse
Affiliation(s)
- Jennifer Li
- Center of Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, Australia
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Angus W Thomson
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Natasha M Rogers
- Center of Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, Australia
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
40
|
Abstract
Elderly individuals with chronic disorders tend to develop inflammaging, a condition associated with elevated levels of blood inflammatory markers, and increased susceptibility to chronic disease progression. Native and adaptive immunity are both involved in immune system senescence, kidney fibrosis and aging. The innate immune system is characterized by a limited number of receptors, constantly challenged by self and non-self stimuli. Circulating and kidney resident myeloid and lymphoid cells are all equipped with pattern recognition receptors (PRRs). Recent reports on PRRs show kidney overexpression of toll-like receptors (TLRs) in inflammaging autoimmune renal diseases, vasculitis, acute kidney injury and kidney transplant rejection. TLR upregulation leads to proinflammatory cytokine induction, fibrosis, and chronic kidney disease progression. TLR2 blockade in a murine model of renal ischemia reperfusion injury prevented the escape of natural killer cells and neutrophils by inflammaging kidney injury. Tumor necrosis factor-α blockade in endothelial cells with senescence-associated secretory phenotype significantly reduced interleukin-6 release. These findings should encourage experimental and translational clinical trials aimed at modulating renal inflammaging by native immunity blockade.
Collapse
|
41
|
Robertson H, Li J, Kim HJ, Rhodes JW, Harman AN, Patrick E, Rogers NM. Transcriptomic Analysis Identifies A Tolerogenic Dendritic Cell Signature. Front Immunol 2021; 12:733231. [PMID: 34745103 PMCID: PMC8564488 DOI: 10.3389/fimmu.2021.733231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/30/2021] [Indexed: 12/23/2022] Open
Abstract
Dendritic cells (DC) are central to regulating innate and adaptive immune responses. Strategies that modify DC function provide new therapeutic opportunities in autoimmune diseases and transplantation. Current pharmacological approaches can alter DC phenotype to induce tolerogenic DC (tolDC), a maturation-resistant DC subset capable of directing a regulatory immune response that are being explored in current clinical trials. The classical phenotypic characterization of tolDC is limited to cell-surface marker expression and anti-inflammatory cytokine production, although these are not specific. TolDC may be better defined using gene signatures, but there is no consensus definition regarding genotypic markers. We address this shortcoming by analyzing available transcriptomic data to yield an independent set of differentially expressed genes that characterize human tolDC. We validate this transcriptomic signature and also explore gene differences according to the method of tolDC generation. As well as establishing a novel characterization of tolDC, we interrogated its translational utility in vivo, demonstrating this geneset was enriched in the liver, a known tolerogenic organ. Our gene signature will potentially provide greater understanding regarding transcriptional regulators of tolerance and allow researchers to standardize identification of tolDC used for cellular therapy in clinical trials.
Collapse
Affiliation(s)
- Harry Robertson
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Jennifer Li
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Hani Jieun Kim
- Computational Systems Biology Group, Children's Medical Research Institute, Westmead, NSW, Australia.,School of Mathematics and Statistics, University of Sydney, Camperdown, NSW, Australia
| | - Jake W Rhodes
- Centre for Virus Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Andrew N Harman
- Centre for Virus Research, Westmead Institute for Medical Research, Westmead, NSW, Australia.,The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health Sydney, Sydney, NSW, Australia
| | - Ellis Patrick
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia.,School of Mathematics and Statistics, University of Sydney, Camperdown, NSW, Australia.,Centre for Virus Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Natasha M Rogers
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia.,Renal and Transplantation Medicine, Westmead Hospital, Westmead, NSW, Australia.,Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
42
|
Yang F, Chen J, Huang XR, Yiu WH, Yu X, Tang SCW, Lan HY. Regulatory role and mechanisms of myeloid TLR4 in anti-GBM glomerulonephritis. Cell Mol Life Sci 2021; 78:6721-6734. [PMID: 34568976 PMCID: PMC8558180 DOI: 10.1007/s00018-021-03936-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/05/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022]
Abstract
Myeloid cells and TLR4 play a critical role in acute kidney injury. This study investigated the regulatory role and mechanisms of myeloid TLR4 in experimental anti-glomerular basement membrane (GBM) glomerulonephritis (GN). Anti-GBM GN was induced in tlr4flox/flox and tlr4flox/flox−lysM−cre mice by intravenous injection of the sheep anti-mouse GBM antibody. Compared to control mice, conditional disruption of tlr4 from myeloid cells, largely macrophages (> 85%), suppressed glomerular crescent formation and attenuated progressive renal injury by lowering serum creatinine and 24-h urine protein excretion while improving creatinine clearance. Mechanistically, deletion of myeloid tlr4 markedly inhibited renal infiltration of macrophages and T cells and resulted in a shift of infiltrating macrophages from F4/80+iNOS+ M1 to F4/80+CD206+ M2 phenotype and inhibited the upregulation of renal proinflammatory cytokines IL-1β and MCP-1. Importantly, deletion of myeloid tlr4 suppressed T cell-mediated immune injury by shifting Th1 (CD4+IFNγ+) and Th17 (CD4+IL-17a+) to Treg (CD4+CD25+FoxP3+) immune responses. Transcriptome analysis also revealed that disrupted myeloid TLR4 largely downregulated genes involving immune and cytokine-related pathways. Thus, myeloid TLR4 plays a pivotal role in anti-GBM GN by immunological switching from M1 to M2 and from Th1/Th17 to Treg and targeting myeloid TLR4 may be a novel therapeutic strategy for immune-mediated kidney diseases.
Collapse
Affiliation(s)
- Fuye Yang
- Department of Nephrology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People's Republic of China.,Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, People's Republic of China
| | - Jiaoyi Chen
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, People's Republic of China
| | - Xiao Ru Huang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, People's Republic of China.,Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, 510080, People's Republic of China
| | - Wai Han Yiu
- Division of Nephrology, Department of Medicine, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Xueqing Yu
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, 510080, People's Republic of China
| | - Sydney C W Tang
- Division of Nephrology, Department of Medicine, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Hui Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, People's Republic of China. .,The CUHK-Guangdong Provincial People's Hospital Joint Research Laboratory on Immunological and Genetic Kidney Diseases, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China.
| |
Collapse
|
43
|
Xu L. The Role of Myeloid Cells in Acute Kidney Injury and Kidney Repair. KIDNEY360 2021; 2:1852-1864. [PMID: 35372990 PMCID: PMC8785849 DOI: 10.34067/kid.0000672021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 09/17/2021] [Indexed: 02/04/2023]
Abstract
AKI remains highly prevalent, yet no optimal therapy is available to prevent it or promote recovery after initial insult. Experimental studies have demonstrated that both innate and adaptive immune responses play a central role during AKI. In response to injury, myeloid cells are first recruited and activated on the basis of specific signals from the damaged microenvironment. The subsequent recruitment and activation state of the immune cells depends on the stage of injury and recovery, reflecting a dynamic and diverse spectrum of immunophenotypes. In this review, we highlight our current understanding of the mechanisms by which myeloid cells contribute to injury, repair, and fibrosis after AKI.
Collapse
Affiliation(s)
- Leyuan Xu
- Department of Internal Medicine, Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
44
|
Wang X, Chen J, Xu J, Xie J, Harris DCH, Zheng G. The Role of Macrophages in Kidney Fibrosis. Front Physiol 2021; 12:705838. [PMID: 34421643 PMCID: PMC8378534 DOI: 10.3389/fphys.2021.705838] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/07/2021] [Indexed: 12/27/2022] Open
Abstract
The phenotypic heterogeneity and functional diversity of macrophages confer on them complexed roles in the development and progression of kidney diseases. After kidney injury, bone marrow-derived monocytes are rapidly recruited to the glomerulus and tubulointerstitium. They are activated and differentiated on site into pro-inflammatory M1 macrophages, which initiate Th1-type adaptive immune responses and damage normal tissues. In contrast, anti-inflammatory M2 macrophages induce Th2-type immune responses, secrete large amounts of TGF-β and anti-inflammatory cytokines, transform into αSMA+ myofibroblasts in injured kidney, inhibit immune responses, and promote wound healing and tissue fibrosis. Previous studies on the role of macrophages in kidney fibrosis were mainly focused on inflammation-associated injury and injury repair. Apart from macrophage-secreted profibrotic cytokines, such as TGF-β, evidence for a direct contribution of macrophages to kidney fibrosis is lacking. However, under inflammatory conditions, Wnt ligands are derived mainly from macrophages and Wnt signaling is central in the network of multiple profibrotic pathways. Largely underinvestigated are the direct contribution of macrophages to profibrotic signaling pathways, macrophage phenotypic heterogeneity and functional diversity in relation to kidney fibrosis, and on their cross-talk with other cells in profibrotic signaling networks that cause fibrosis. Here we aim to provide an overview on the roles of macrophage phenotypic and functional diversity in their contribution to pro-fibrotic signaling pathways, and on the therapeutic potential of targeting macrophages for the treatment of kidney fibrosis.
Collapse
Affiliation(s)
- Xiaoling Wang
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China
- Clinical Laboratory, Shanxi Academy of Traditional Chinese Medicine, Taiyuan, China
| | - Jianwei Chen
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Jun Xu
- Department of General Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Jun Xie
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China
| | - David C. H. Harris
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Guoping Zheng
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
45
|
Role of Dendritic Cell in Diabetic Nephropathy. Int J Mol Sci 2021; 22:ijms22147554. [PMID: 34299173 PMCID: PMC8308035 DOI: 10.3390/ijms22147554] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 11/18/2022] Open
Abstract
Diabetic nephropathy (DN) is one of the most significant microvascular complications in diabetic patients. DN is the leading cause of end-stage renal disease, accounting for approximately 50% of incident cases. The current treatment options, such as optimal control of hyperglycemia and elevated blood pressure, are insufficient to prevent its progression. DN has been considered as a nonimmune, metabolic, or hemodynamic glomerular disease initiated by hyperglycemia. However, recent studies suggest that DN is an inflammatory disease, and immune cells related with innate and adaptive immunity, such as macrophage and T cells, might be involved in its development and progression. Although it has been revealed that kidney dendritic cells (DCs) accumulation in the renal tissue of human and animal models of DN require activated T cells in the kidney disease, little is known about the function of DCs in DN. In this review, we describe kidney DCs and their subsets, and the role in the pathogenesis of DN. We also suggest how to improve the kidney outcomes by modulating kidney DCs optimally in the patients with DN.
Collapse
|
46
|
Nordlohne J, Hulsmann I, Schwafertz S, Zgrajek J, Grundmann M, von Vietinghoff S, Eitner F, Becker MS. A flow cytometry approach reveals heterogeneity in conventional subsets of murine renal mononuclear phagocytes. Sci Rep 2021; 11:13251. [PMID: 34168267 PMCID: PMC8225656 DOI: 10.1038/s41598-021-92784-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 06/14/2021] [Indexed: 01/07/2023] Open
Abstract
Mononuclear phagocytes (MNPs) participate in inflammation and repair after kidney injury, reflecting their complex nature. Dissection into refined functional subunits has been challenging and would benefit understanding of renal pathologies. Flow cytometric approaches are limited to classifications of either different MNP subsets or functional state. We sought to combine these two dimensions in one protocol that considers functional heterogeneity in each MNP subset. We identified five distinct renal MNP subsets based on a previously described strategy. In vitro polarization of bone marrow-derived macrophages (BMDM) into M1- and M2-like cells suggested functional distinction of CD86 + MHCII + CD206- and CD206 + cells. Combination of both distinction methods identified CD86 + MHCII + CD206- and CD206 + cells in all five MNP subsets, revealing their heterologous nature. Our approach revealed that MNP composition and their functional segmentation varied between different mouse models of kidney injury and, moreover, was dynamically regulated in a time-dependent manner. CD206 + cells from three analyzed MNP subsets had a higher ex vivo phagocytic capacity than CD86 + MHCII + CD206- counterparts, indicating functional uniqueness of each subset. In conclusion, our novel flow cytometric approach refines insights into renal MNP heterogeneity and therefore could benefit mechanistic understanding of renal pathology.
Collapse
Affiliation(s)
- Johannes Nordlohne
- Cardiovascular Research, Research and Development, Pharmaceuticals, Kidney Diseases, Bayer AG, Building 0500, 214, 42113, Wuppertal, Germany
| | - Ilona Hulsmann
- Cardiovascular Research, Research and Development, Pharmaceuticals, Kidney Diseases, Bayer AG, Building 0500, 214, 42113, Wuppertal, Germany
| | - Svenja Schwafertz
- Cardiovascular Research, Research and Development, Pharmaceuticals, Kidney Diseases, Bayer AG, Building 0500, 214, 42113, Wuppertal, Germany
| | - Jasmin Zgrajek
- Cardiovascular Research, Research and Development, Pharmaceuticals, Kidney Diseases, Bayer AG, Building 0500, 214, 42113, Wuppertal, Germany
| | - Manuel Grundmann
- Cardiovascular Research, Research and Development, Pharmaceuticals, Kidney Diseases, Bayer AG, Building 0500, 214, 42113, Wuppertal, Germany
| | - Sibylle von Vietinghoff
- Nephrology Section, Medical Clinic 1, University Hospital Bonn, Rheinische Friedrich-Wilhelms University, Bonn, Germany
| | - Frank Eitner
- Cardiovascular Research, Research and Development, Pharmaceuticals, Kidney Diseases, Bayer AG, Building 0500, 214, 42113, Wuppertal, Germany
| | - Michael S Becker
- Cardiovascular Research, Research and Development, Pharmaceuticals, Kidney Diseases, Bayer AG, Building 0500, 214, 42113, Wuppertal, Germany.
| |
Collapse
|
47
|
Han Q, Wang X, Ding X, He J, Cai G, Zhu H. Immunomodulatory Effects of Mesenchymal Stem Cells on Drug-Induced Acute Kidney Injury. Front Immunol 2021; 12:683003. [PMID: 34149721 PMCID: PMC8213363 DOI: 10.3389/fimmu.2021.683003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/10/2021] [Indexed: 12/29/2022] Open
Abstract
Drug-induced nephrotoxicity is an important and increasing cause of acute kidney injury (AKI), which accounts for approximately 20% of hospitalized patients. Previous reviews studies on immunity and AKI focused mainly on ischemia-reperfusion (IR), whereas no systematic review addressing drug-induced AKI and its related immune mechanisms is available. Recent studies have provided a deeper understanding on the mechanisms of drug-induced AKI, among which acute tubular interstitial injury induced by the breakdown of innate immunity was reported to play an important role. Emerging research on mesenchymal stem cell (MSC) therapy has revealed its potential as treatment for drug-induced AKI. MSCs can inhibit kidney damage by regulating the innate immune balance, promoting kidney repair, and preventing kidney fibrosis. However, it is important to note that there are various sources of MSCs, which impacts on the immunomodulatory ability of the cells. This review aims to address the immune pathogenesis of drug-induced AKI versus that of IR-induced AKI, and to explore the immunomodulatory effects and therapeutic potential of MSCs for drug-induced AKI.
Collapse
Affiliation(s)
- Qiuxia Han
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (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, China.,School of Medicine, Nankai University, Tianjin, China
| | - Xiaochen Wang
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (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, China
| | - Xiaonan Ding
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (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, China
| | - Jun He
- Department of Genetics, Changsha Hospital for Maternal and Child Health Care, Hunan, China
| | - Guangyan Cai
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (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, China.,School of Medicine, Nankai University, Tianjin, China
| | - Hanyu Zhu
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (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, China
| |
Collapse
|
48
|
Rausch M, Blanc L, De Souza Silva O, Dormond O, Griffioen AW, Nowak-Sliwinska P. Characterization of Renal Cell Carcinoma Heterotypic 3D Co-Cultures with Immune Cell Subsets. Cancers (Basel) 2021; 13:2551. [PMID: 34067456 PMCID: PMC8197009 DOI: 10.3390/cancers13112551] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/12/2022] Open
Abstract
Two-dimensional cell culture-based platforms are easy and reproducible, however, they do not resemble the heterotypic cell-cell interactions or the complex tumor microenvironment. These parameters influence the treatment response and the cancer cell fate. Platforms to study the efficacy of anti-cancer treatments and their impact on the tumor microenvironment are currently being developed. In this study, we established robust, reproducible, and easy-to-use short-term spheroid cultures to mimic clear cell renal cell carcinoma (ccRCC). These 3D co-cultures included human endothelial cells, fibroblasts, immune cell subsets, and ccRCC cell lines, both parental and sunitinib-resistant. During spheroid formation, cells induce the production and secretion of the extracellular matrix. We monitored immune cell infiltration, surface protein expression, and the response to a treatment showing that the immune cells infiltrated the spheroid co-cultures within 6 h. Treatment with an optimized drug combination or the small molecule-based targeted drug sunitinib increased immune cell infiltration significantly. Assessing the therapeutic potential of this drug combination in this platform, we revealed that the expression of PD-L1 increased in 3D co-cultures. The cost- and time-effective establishment of our 3D co-culture model and its application as a pre-clinical drug screening platform can facilitate the treatment validation and clinical translation.
Collapse
Affiliation(s)
- Magdalena Rausch
- School of Pharmaceutical Sciences, Faculty of Science, University of Geneva, 1211 Geneva, Switzerland; (M.R.); (L.B.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- Translational Research Center in Oncohaematology, 1211 Geneva, Switzerland
| | - Léa Blanc
- School of Pharmaceutical Sciences, Faculty of Science, University of Geneva, 1211 Geneva, Switzerland; (M.R.); (L.B.)
| | - Olga De Souza Silva
- Department of Visceral Surgery, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (O.D.S.S.); (O.D.)
| | - Olivier Dormond
- Department of Visceral Surgery, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (O.D.S.S.); (O.D.)
| | - Arjan W. Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Patrycja Nowak-Sliwinska
- School of Pharmaceutical Sciences, Faculty of Science, University of Geneva, 1211 Geneva, Switzerland; (M.R.); (L.B.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- Translational Research Center in Oncohaematology, 1211 Geneva, Switzerland
| |
Collapse
|
49
|
Macrophages and Stem Cells-Two to Tango for Tissue Repair? Biomolecules 2021; 11:biom11050697. [PMID: 34066618 PMCID: PMC8148606 DOI: 10.3390/biom11050697] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/26/2021] [Accepted: 05/04/2021] [Indexed: 12/25/2022] Open
Abstract
Macrophages (MCs) are present in all tissues, not only supporting homeostasis, but also playing an important role in organogenesis, post-injury regeneration, and diseases. They are a heterogeneous cell population due to their origin, tissue specificity, and polarization in response to aggression factors, depending on environmental cues. Thus, as pro-inflammatory M1 phagocytic MCs, they contribute to tissue damage and even fibrosis, but the anti-inflammatory M2 phenotype participates in repairing processes and wound healing through a molecular interplay with most cells in adult stem cell niches. In this review, we emphasize MC phenotypic heterogeneity in health and disease, highlighting their systemic and systematic contribution to tissue homeostasis and repair. Unraveling the intervention of both resident and migrated MCs on the behavior of stem cells and the regulation of the stem cell niche is crucial for opening new perspectives for novel therapeutic strategies in different diseases.
Collapse
|
50
|
Hsiao CC, Hou YS, Liu YH, Ko JY, Lee CT. Combined Melatonin and Extracorporeal Shock Wave Therapy Enhances Podocyte Protection and Ameliorates Kidney Function in a Diabetic Nephropathy Rat Model. Antioxidants (Basel) 2021; 10:antiox10050733. [PMID: 34066452 PMCID: PMC8148201 DOI: 10.3390/antiox10050733] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/30/2021] [Accepted: 05/02/2021] [Indexed: 01/18/2023] Open
Abstract
(1) Background: Diabetic nephropathy (DN) is common complication of diabetes. Current therapy for DN does not include promotion of podocyte protection. Therefore, we investigated the therapeutic effect of melatonin (Mel) combined extracorporeal shock wave (SW) therapy on a DN rat model. (2) Methods: The DN rats were treated with Mel (5 mg/kg) twice a week for 6 weeks and SW treatment once a week (0.13 mJ/mm2) for 6 weeks. We assessed urine microalbumin, albumin to creatinine ratio (ACR), glomerular hypertrophy, glomerular fibrosis, podocyte markers (Wilm’s tumor protein-1, synaptopodin and nephrin), cell proliferation, cell survival, cell apoptosis, renal inflammation and renal oxidative stress. (3) Results: The Mel combined SW therapy regimen significantly reduced urine microalbumin excretion (3.3 ± 0.5 mg/dL, p < 0.001), ACR (65.2 ± 8.3 mg/g, p < 0.001), glomerular hypertrophy (3.1 ± 0.1 × 106 μm3, p < 0.01) and glomerular fibrosis (0.9 ± 0.4 relative mRNA fold, p < 0.05). Moreover, the Mel combined SW therapy regimen significantly increased podocyte number (44.1 ± 5.0% area of synaptopodin, p < 0.001) in the Mel combined SW group. This is likely primarily because Mel combined with SW therapy significantly reduced renal inflammation (753 ± 46 pg/mg, p < 0.01), renal oxidative stress (0.6 ± 0.04 relative density, p < 0.05), and apoptosis (0.3 ± 0.03 relative density, p < 0.001), and also significantly increased cell proliferation (2.0 ± 0.2% area proliferating cell nuclear antigen (PCNA), p < 0.01), cell survival, and nephrin level (4.2 ± 0.4 ng/mL, p < 0.001). (4) Conclusions: Mel combined SW therapy enhances podocyte protection and ameliorates kidney function in a DN rat model. Mel combined SW therapy may serve as a novel noninvasive and effective treatment of DN.
Collapse
Affiliation(s)
- Chang-Chun Hsiao
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (C.-C.H.); (Y.-S.H.); (Y.-H.L.)
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
| | - You-Syuan Hou
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (C.-C.H.); (Y.-S.H.); (Y.-H.L.)
| | - Yu-Hsuan Liu
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (C.-C.H.); (Y.-S.H.); (Y.-H.L.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang-Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Jih-Yang Ko
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
- Department of Orthopedic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Chien-Te Lee
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang-Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Correspondence: ; Tel.: +886-7731-7123 (ext. 8306)
| |
Collapse
|