1
|
Lee K, Jang HR, Rabb H. Lymphocytes and innate immune cells in acute kidney injury and repair. Nat Rev Nephrol 2024; 20:789-805. [PMID: 39095505 DOI: 10.1038/s41581-024-00875-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 08/04/2024]
Abstract
Acute kidney injury (AKI) is a common and serious disease entity that affects native kidneys and allografts but for which no specific treatments exist. Complex intrarenal inflammatory processes driven by lymphocytes and innate immune cells have key roles in the development and progression of AKI. Many studies have focused on prevention of early injury in AKI. However, most patients with AKI present after injury is already established. Increasing research is therefore focusing on mechanisms of renal repair following AKI and prevention of progression from AKI to chronic kidney disease. CD4+ and CD8+ T cells, B cells and neutrophils are probably involved in the development and progression of AKI, whereas regulatory T cells, double-negative T cells and type 2 innate lymphoid cells have protective roles. Several immune cells, such as macrophages and natural killer T cells, can have both deleterious and protective effects, depending on their subtype and/or the stage of AKI. The immune system not only participates in injury and repair processes during AKI but also has a role in mediating AKI-induced distant organ dysfunction. Targeted manipulation of immune cells is a promising therapeutic strategy to improve AKI outcomes.
Collapse
Affiliation(s)
- Kyungho Lee
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Cell and Gene Therapy Institute, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Nephrology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hye Ryoun Jang
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Cell and Gene Therapy Institute, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hamid Rabb
- Nephrology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Collett JA, Flannery AH, Liu LJ, Takeuchi T, Basile DP, Neyra JA. IL-17A Levels and Progression of Kidney Disease Following Hospitalization with and without Acute Kidney Injury. KIDNEY360 2024; 5:1623-1632. [PMID: 39230981 DOI: 10.34067/kid.0000000000000559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/16/2024] [Indexed: 09/06/2024]
Abstract
Key Points
IL-17A was higher in patients with AKI versus without AKI during hospitalization and up to 1-year postdischarge.IL-17A was higher in patients with progression of kidney disease but not independently associated with subsequent progression of kidney disease.
Background
AKI is associated with increased mortality and new or progressive CKD. Inflammatory cells play an important role in acute organ injury. We previously demonstrated that serum IL-17A levels were significantly elevated in critically ill patients with AKI and independently associated with hospital mortality. We hypothesize that IL-17A levels are elevated in hospitalized patients with AKI at diagnosis, and sustained elevation after discharge is associated with subsequent CKD incidence or progression.
Methods
This was an observational convenience sampling study of hospital survivors of stage 2 or 3 AKI and controls without AKI from the Assessment, Serial Evaluation, and Subsequent Sequelae of AKI study. Patients were classified as progression or nonprogression on the basis of a composite of CKD incidence, progression, or ESKD. IL-17A levels were evaluated with S-Plex assay (Meso Scale Discovery) at 0 (during hospitalization), 3, and 12 months postdischarge and analyzed along with clinical and biomarker data up to 84 months after discharge.
Results
Among 171 AKI and 175 non-AKI participants, IL-17A levels were elevated in AKI versus non-AKI patients at 0-, 3-, and 12-month time points (P < 0.05 for all comparisons). Furthermore, IL-17A levels were elevated in the progression versus nonprogression group at the 3- and 12-month time points for outcomes occurring at 3–6 and 12–84 months, respectively (P < 0.05 for both). In adjusted multivariable models, IL-17A levels were not independently associated with progression of kidney disease. IL-17A levels were positively correlated with kidney disease and immune activation biomarkers at all time points (P < 0.001).
Conclusions
IL-17A was higher in patients with AKI versus without AKI during hospitalization and up to 1-year postdischarge. IL-17A was higher in patients with progression of kidney disease after hospitalization, but not independently associated with subsequent progression of kidney disease in fully adjusted models.
Collapse
Affiliation(s)
- Jason A Collett
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Alexander H Flannery
- Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, Kentucky
| | - Lucas J Liu
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Tomonori Takeuchi
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - David P Basile
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Javier A Neyra
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
3
|
Khoza S, George JA, Naicker P, Stoychev SH, Fabian J, Govender IS. Proteomic Analysis Identifies Dysregulated Proteins in Albuminuria: A South African Pilot Study. BIOLOGY 2024; 13:680. [PMID: 39336107 PMCID: PMC11428484 DOI: 10.3390/biology13090680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024]
Abstract
Albuminuria may precede decreases in the glomerular filtration rate (GFR) and both tests are insensitive predictors of early stages of kidney disease. Our aim was to characterise the urinary proteome in black African individuals with albuminuria and well-preserved GFR from South Africa. This case-controlled study compared the urinary proteomes of 52 normoalbuminuric (urine albumin: creatinine ratio (uACR) < 3 mg/mmol) and 56 albuminuric (uACR ≥ 3 mg/mmol) adults of black African ethnicity. Urine proteins were precipitated, reduced, alkylated, digested, and analysed using an Evosep One LC (Evosep Biosystems, Odense, Denmark) coupled to a Sciex 5600 Triple-TOF (Sciex, Framingham, MA, USA) in data-independent acquisition mode. The data were searched on SpectronautTM 15. Differentially abundant proteins (DAPs) were filtered to include those with a ≥2.25-fold change and a false discovery rate ≤ 1%. Receiver-operating characteristic curves were used to assess the discriminating abilities of proteins of interest. Pathway analysis was performed using Enrichr software. As expected, the albuminuric group had higher uACR (7.9 vs. 0.55 mg/mmol, p < 0.001). The median eGFR (mL/min/1.73 m2) showed no difference between the groups (111 vs. 114, p = 0.707). We identified 80 DAPs in the albuminuria group compared to the normoalbuminuria group, of which 59 proteins were increased while 21 proteins were decreased in abundance. We found 12 urinary proteins with an AUC > 0.8 and a p < 0.001 in the multivariate analysis. Furthermore, an 80-protein model was developed that showed a high AUC ˃ 0.907 and a predictive accuracy of 91.3% between the two groups. Pathway analysis found that the DAPs were involved in insulin growth factor (IGF) functions, innate immunity, platelet degranulation, and extracellular matrix organization. In albuminuric individuals with a well-preserved eGFR, pathways involved in preventing the release and uptake of IGF by insulin growth factor binding protein were significantly enriched. These proteins are indicative of a homeostatic imbalance in a variety of cellular processes underlying renal dysfunction and are implicated in chronic kidney disease.
Collapse
Affiliation(s)
- Siyabonga Khoza
- Department of Chemical Pathology, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Jaya A George
- Wits Diagnostic Innovation Hub, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Previn Naicker
- Future Production Chemicals, Council for Scientific and Industrial Research, Pretoria 0001, South Africa
| | - Stoyan H Stoychev
- ReSyn BioSciences, Edenvale 1610, South Africa
- Evosep Biosystems, 5230 Odense, Denmark
| | - June Fabian
- Wits Donald Gordon Medical Centre, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
- Medical Research Council/Wits University Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Ireshyn S Govender
- Future Production Chemicals, Council for Scientific and Industrial Research, Pretoria 0001, South Africa
- ReSyn BioSciences, Edenvale 1610, South Africa
| |
Collapse
|
4
|
Fuhrmann B, Jiang J, Mcleod P, Huang X, Balaji S, Arp J, Diao H, Ma S, Peng T, Haig A, Gunaratnam L, Zhang ZX, Jevnikar AM. Inhibition of NK cell cytotoxicity by tubular epithelial cell expression of Clr-b and Clr-f. CURRENT RESEARCH IN IMMUNOLOGY 2024; 5:100081. [PMID: 39113760 PMCID: PMC11303997 DOI: 10.1016/j.crimmu.2024.100081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 06/21/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
NK cells participate in ischemia reperfusion injury (IRI) and transplant rejection. Endogenous regulatory systems may exist to attenuate NK cell activation and cytotoxicity in IRI associated with kidney transplantation. A greater understanding of NK regulation will provide insights in transplant outcomes and could direct new therapeutic strategies. Kidney tubular epithelial cells (TECs) may negatively regulate NK cell activation by their surface expression of a complex family of C-type lectin-related proteins (Clrs). We have found that Clr-b and Clr-f were expressed by TECs. Clr-b was upregulated by inflammatory cytokines TNFα and IFNγ in vitro. Silencing of both Clr-b and Clr-f expression using siRNA resulted in increased NK cell killing of TECs compared to silencing of either Clr-b or Clr-f alone (p < 0.01) and when compared to control TECs (p < 0.001). NK cells treated in vitro with soluble Clr-b and Clr-f proteins reduced their capacity to kill TECs (p < 0.05). Hence, NK cell cytotoxicity can be inhibited by Clr proteins on the surface of TECs. Our study suggests a synergistic effect of Clr molecules in regulating NK cell function in renal cells and this may represent an important endogenous regulatory system to limit NK cell-mediated organ injury during inflammation.
Collapse
Affiliation(s)
- Benjamin Fuhrmann
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Jifu Jiang
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
| | - Patrick Mcleod
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
| | - Xuyan Huang
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
| | - Shilpa Balaji
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Jaqueline Arp
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
| | - Hong Diao
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
| | - Shengwu Ma
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Tianqing Peng
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Aaron Haig
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Lakshman Gunaratnam
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Multi-Organ Transplantation Program, London Health Sciences Centre, London, Ontario, Canada
- Division of Nephrology, Department of Medicine, Western University, London, Ontario, Canada
| | - Zhu-Xu Zhang
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
- Multi-Organ Transplantation Program, London Health Sciences Centre, London, Ontario, Canada
- Division of Nephrology, Department of Medicine, Western University, London, Ontario, Canada
| | - Anthony M. Jevnikar
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Multi-Organ Transplantation Program, London Health Sciences Centre, London, Ontario, Canada
- Division of Nephrology, Department of Medicine, Western University, London, Ontario, Canada
| |
Collapse
|
5
|
Palit S, Shrestha AK, Thapa S, L. Grimm S, Coarfa C, Theis F, Simon LM, Shivanna B. Leveraging Integrated RNA Sequencing to Decipher Adrenomedullin's Protective Mechanisms in Experimental Bronchopulmonary Dysplasia. Genes (Basel) 2024; 15:806. [PMID: 38927741 PMCID: PMC11202456 DOI: 10.3390/genes15060806] [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: 05/06/2024] [Revised: 06/12/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease commonly affecting premature infants, with limited therapeutic options and increased long-term consequences. Adrenomedullin (Adm), a proangiogenic peptide hormone, has been found to protect rodents against experimental BPD. This study aims to elucidate the molecular and cellular mechanisms through which Adm influences BPD pathogenesis using a lipopolysaccharide (LPS)-induced model of experimental BPD in mice. Bulk RNA sequencing of Adm-sufficient (wild-type or Adm+/+) and Adm-haplodeficient (Adm+/-) mice lungs, integrated with single-cell RNA sequencing data, revealed distinct gene expression patterns and cell type alterations associated with Adm deficiency and LPS exposure. Notably, computational integration with cell atlas data revealed that Adm-haplodeficient mouse lungs exhibited gene expression signatures characteristic of increased inflammation, natural killer (NK) cell frequency, and decreased endothelial cell and type II pneumocyte frequency. Furthermore, in silico human BPD patient data analysis supported our cell type frequency finding, highlighting elevated NK cells in BPD infants. These results underscore the protective role of Adm in experimental BPD and emphasize that it is a potential therapeutic target for BPD infants with an inflammatory phenotype.
Collapse
Affiliation(s)
- Subarna Palit
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | - Amrit Kumar Shrestha
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA (B.S.)
| | - Shyam Thapa
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA (B.S.)
| | - Sandra L. Grimm
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Fabian Theis
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Mathematics, Technical University of Munich, 85748 Garching, Germany
| | - Lukas M. Simon
- Therapeutic Innovation Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Binoy Shivanna
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA (B.S.)
| |
Collapse
|
6
|
Valdes AM, Ikram A, Taylor LA, Zheng A, Kouraki A, Kelly A, Ashraf W, Vijay A, Miller S, Nightingale J, Selby NM, Ollivere BJ. Preoperative inflammatory biomarkers reveal renal involvement in postsurgical mortality in hip fracture patients: an exploratory study. Front Immunol 2024; 15:1372079. [PMID: 38919625 PMCID: PMC11197399 DOI: 10.3389/fimmu.2024.1372079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/20/2024] [Indexed: 06/27/2024] Open
Abstract
Background Hip fractures in frail patients result in excess mortality not accounted for by age or comorbidities. The mechanisms behind the high risk of mortality remain undetermined but are hypothesized to be related to the inflammatory status of frail patients. Methods In a prospective observational exploratory cohort study of hospitalized frail hip fracture patients, 92 inflammatory markers were tested in pre-operative serum samples and markers were tested against 6-month survival post-hip fracture surgery and incidence of acute kidney injury (AKI). After correcting for multiple testing, adjustments for comorbidities and demographics were performed on the statistically significant markers. Results Of the 92 markers tested, circulating levels of fibroblast growth factor 23 (FGF-23) and interleukin-15 receptor alpha (IL15RA), both involved in renal disease, were significantly correlated with 6-month mortality (27.5% overall) after correcting for multiple testing. The incidence of postoperative AKI (25.4%) was strongly associated with 6-month mortality, odds ratio = 10.57; 95% CI [2.76-40.51], and with both markers plus estimated glomerular filtration rate (eGFR)- cystatin C (CYSC) but not eGFR-CRE. The effect of these markers on mortality was significantly mediated by their effect on postoperative AKI. Conclusion High postoperative mortality in frail hip fracture patients is highly correlated with preoperative biomarkers of renal function in this pilot study. The effect of preoperative circulating levels of FGF-23, IL15RA, and eGFR-CYSC on 6-month mortality is in part mediated by their effect on postoperative AKI. Creatinine-derived preoperative renal function measures were very poorly correlated with postoperative outcomes in this group.
Collapse
Affiliation(s)
- Ana M. Valdes
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Adeel Ikram
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Lauren A. Taylor
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Amy Zheng
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Afroditi Kouraki
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Anthony Kelly
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Waheed Ashraf
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Amrita Vijay
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Suzanne Miller
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Jessica Nightingale
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Nicholas M. Selby
- Centre for Kidney Research and Innovation, Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Benjamin J. Ollivere
- Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| |
Collapse
|
7
|
Li S, Pang W, Wang Y, Zhang Y. Cordyceps sinensis extract protects against acute kidney injury by inhibiting perforin expression in NK cells via the STING/IRF3 pathway. Aging (Albany NY) 2024; 16:5887-5904. [PMID: 38517396 PMCID: PMC11042953 DOI: 10.18632/aging.205676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 02/13/2024] [Indexed: 03/23/2024]
Abstract
Acute kidney injury (AKI) is associated with immune cell activation and inflammation. However, the putative pathogenic mechanisms of this injury have not been thoroughly investigated. Natural killer (NK) cells play an important role in immune regulation; however, whether NK cells regulate AKI remains unclear. Cordyceps sinensis (CS), a modern Chinese patented medicine preparation, has been widely used in treating patients with chronic kidney disease (CKD) owing to its anti-inflammatory effects and maintenance of immune homeostasis. Whether 2'-deoxyadenosine, a major active component in CS, can ameliorate renal AKI by regulating immunity, particularly in NK cells, has not been reported. This study is the first to demonstrate how NK cells promote AKI by releasing perforin, interferon-gamma (IFN-γ) and other inflammatory factors in vivo and in vitro. Differential gene expression between AKI and normal tissues was assessed using bioinformatic analyses. Quantitative real-time PCR, western blotting, and immunohistochemical staining were used to detect target protein mRNA and protein expression. Levels of inflammatory factors were measured using enzyme-linked immunosorbent assay. We found the high doses of the 2'-deoxyadenosine treatment significantly alleviated FA-induced renal damage in vivo, and alleviated the NK cells of renal injury by activating the STING/IRF3 pathway to inhibit perforin release in vitro. The results showed that 2'-deoxyadenosine could mitigate AKI by downregulating the activity of NK cells (by decreasing the expressions of perforin and IFN-γ) and inhibiting the stimulator of interferon genes and phosphorylated IFN regulatory factor 3. This may provide valuable evidence supporting the clinical use of CS in treating patients with AKI.
Collapse
Affiliation(s)
- Shuang Li
- General Department of Western Medicine, Yangjing Community Health Service Center, Shanghai 200135, China
| | - Wei Pang
- Department of Emergency Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yuzhu Wang
- General Department of Western Medicine, Yangjing Community Health Service Center, Shanghai 200135, China
| | - Yiting Zhang
- General Department of Traditional Chinese Medicine, Yangjing Community Health Service Center, Shanghai 200135, China
| |
Collapse
|
8
|
Laskarin G, Babarovic E, Kifer N, Bulimbasic S, Sestan M, Held M, Frkovic M, Gagro A, Coric M, Jelusic M. Involvement of M1-Activated Macrophages and Perforin/Granulysin Expressing Lymphocytes in IgA Vasculitis Nephritis. Int J Mol Sci 2024; 25:2253. [PMID: 38396930 PMCID: PMC10889255 DOI: 10.3390/ijms25042253] [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: 01/06/2024] [Revised: 02/02/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
We investigated the polarisation of CD68+ macrophages and perforin and granulysin distributions in kidney lymphocyte subsets of children with IgA vasculitis nephritis (IgAVN). Pro-inflammatory macrophage (M)1 (CD68/iNOS) or regulatory M2 (CD68/arginase-1) polarisation; spatial arrangement of macrophages and lymphocytes; and perforin and granulysin distribution in CD3+ and CD56+ cells were visulaised using double-labelled immunofluorescence. In contrast to the tubules, iNOS+ cells were more abundant than the arginase-1+ cells in the glomeruli. CD68+ macrophage numbers fluctuated in the glomeruli and were mostly labelled with iNOS. CD68+/arginase-1+ cells are abundant in the tubules. CD56+ cells, enclosed by CD68+ cells, were more abundant in the glomeruli than in the tubuli, and co-expressed NKp44. The glomerular and interstitial/intratubular CD56+ cells express perforin and granulysin, respectively. The CD3+ cells did not express perforin, while a minority expressed granulysin. Innate immunity, represented by M1 macrophages and CD56+ cells rich in perforin and granulysin, plays a pivotal role in the acute phase of IgAVN.
Collapse
Affiliation(s)
- Gordana Laskarin
- Department of Physiology, Immunology and Pathophysiology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
- Hospital for Medical Rehabilitation of Hearth and Lung Diseases and Rheumatism “Thalassotherapia-Opatija”, 51410 Opatija, Croatia
| | - Emina Babarovic
- Department of Pathology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia;
| | - Nastasia Kifer
- Division of Rheumatology and Immunology, Department of Paediatrics, School of Medicine, University Hospital Centre Zagreb, University of Zagreb, 10000 Zagreb, Croatia; (N.K.); (M.S.); (M.H.); (M.F.); (M.J.)
| | - Stela Bulimbasic
- Department of Pathology and Cytology, School of Medicine, University Hospital Centre Zagreb, University of Zagreb, 10000 Zagreb, Croatia; (S.B.); (M.C.)
| | - Mario Sestan
- Division of Rheumatology and Immunology, Department of Paediatrics, School of Medicine, University Hospital Centre Zagreb, University of Zagreb, 10000 Zagreb, Croatia; (N.K.); (M.S.); (M.H.); (M.F.); (M.J.)
| | - Martina Held
- Division of Rheumatology and Immunology, Department of Paediatrics, School of Medicine, University Hospital Centre Zagreb, University of Zagreb, 10000 Zagreb, Croatia; (N.K.); (M.S.); (M.H.); (M.F.); (M.J.)
| | - Marijan Frkovic
- Division of Rheumatology and Immunology, Department of Paediatrics, School of Medicine, University Hospital Centre Zagreb, University of Zagreb, 10000 Zagreb, Croatia; (N.K.); (M.S.); (M.H.); (M.F.); (M.J.)
| | - Alenka Gagro
- Children’s Hospital Zagreb, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia;
| | - Marijana Coric
- Department of Pathology and Cytology, School of Medicine, University Hospital Centre Zagreb, University of Zagreb, 10000 Zagreb, Croatia; (S.B.); (M.C.)
| | - Marija Jelusic
- Division of Rheumatology and Immunology, Department of Paediatrics, School of Medicine, University Hospital Centre Zagreb, University of Zagreb, 10000 Zagreb, Croatia; (N.K.); (M.S.); (M.H.); (M.F.); (M.J.)
| |
Collapse
|
9
|
Aguilar OA, Qualls AE, Gonzalez-Hinojosa MDR, Obeidalla S, Kerchberger VE, Tsao T, Singer JP, Looney MR, Raymond W, Hays SR, Golden JA, Kukreja J, Shaver CM, Ware LB, Christie J, Diamond JM, Lanier LL, Greenland JR, Calabrese DR. MICB Genomic Variant Is Associated with NKG2D-mediated Acute Lung Injury and Death. Am J Respir Crit Care Med 2024; 209:70-82. [PMID: 37878820 PMCID: PMC10870895 DOI: 10.1164/rccm.202303-0472oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023] Open
Abstract
Rationale: Acute lung injury (ALI) carries a high risk of mortality but has no established pharmacologic therapy. We previously found that experimental ALI occurs through natural killer (NK) cell NKG2D receptor activation and that the cognate human ligand, MICB, was associated with ALI after transplantation. Objectives: To investigate the association of a common missense variant, MICBG406A, with ALI. Methods: We assessed MICBG406A genotypes within two multicenter observational study cohorts at risk for ALI: primary graft dysfunction (N = 619) and acute respiratory distress syndrome (N = 1,376). Variant protein functional effects were determined in cultured and ex vivo human samples. Measurements and Main Results: Recipients of MICBG406A-homozygous allografts had an 11.1% absolute risk reduction (95% confidence interval [CI], 3.2-19.4%) for severe primary graft dysfunction after lung transplantation and reduced risk for allograft failure (hazard ratio, 0.36; 95% CI, 0.13-0.98). In participants with sepsis, we observed 39% reduced odds of moderately or severely impaired oxygenation among MICBG406A-homozygous individuals (95% CI, 0.43-0.86). BAL NK cells were less frequent and less mature in participants with MICBG406A. Expression of missense variant protein MICBD136N in cultured cells resulted in reduced surface MICB and reduced NKG2D ligation relative to wild-type MICB. Coculture of variant MICBD136N cells with NK cells resulted in less NKG2D activation and less susceptibility to NK cell killing relative to the wild-type cells. Conclusions: These data support a role for MICB signaling through the NKG2D receptor in mediating ALI, suggesting a novel therapeutic approach.
Collapse
Affiliation(s)
- Oscar A. Aguilar
- Department Microbiology and Immunology
- Parker Institute for Cancer Immunotherapy
| | | | | | | | | | | | | | | | | | | | | | - Jasleen Kukreja
- Department of Surgery, University of California San Francisco, San Francisco, California
| | | | - Lorraine B. Ware
- Department Medicine and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jason Christie
- Department Medicine and
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | | | - Lewis L. Lanier
- Department Microbiology and Immunology
- Parker Institute for Cancer Immunotherapy
| | - John R. Greenland
- Department Medicine
- San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Daniel R. Calabrese
- Department Medicine
- San Francisco Veterans Affairs Medical Center, San Francisco, California
| |
Collapse
|
10
|
Burfeind KG, Funahashi Y, Munhall AC, Eiwaz M, Hutchens MP. Natural Killer Lymphocytes Mediate Renal Fibrosis Due to Acute Cardiorenal Syndrome. KIDNEY360 2024; 5:8-21. [PMID: 38037228 PMCID: PMC10833608 DOI: 10.34067/kid.0000000000000305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023]
Abstract
Key Points Natural killer cells infiltrate the kidney after cardiac arrest and medial renal fibrosis Granzyme A is produced by natural killer cells and causes mesenchymal cell expansion and fibrosis in type 1 cardiorenal syndrome Background The AKI to CKD transition presents an opportunity for intervention to prevent CKD. Our laboratory developed a novel murine model of AKI-CKD transition and cardiac arrest/cardiopulmonary resuscitation (CA/CPR), in which all animals develop CKD at 7 weeks. The purpose of this study was to identify potential immune drivers of fibrosis after CA/CPR. Methods Cardiac arrest was induced by potassium chloride, and mice were resuscitated with chest compressions and epinephrine. The kidney immune landscape after CA/CPR was profiled using 11-color flow cytometry analysis and immunofluorescence. Immune cell-derived mediators of fibrosis were identified by analyzing data from three previously published single-cell or single-nuclear RNA sequencing studies. NRK49F fibroblasts were treated with granzyme A (GzA) in vitro , and then cell proliferation was quantified using 5-ethynyl-2′-deoxyuridine. GzA was pharmacologically inhibited both in vitro and in vivo . Results Immune cells infiltrated the kidney after CA/CPR, consisting primarily of innate immune cells, including monocytes/macrophages, neutrophils, and natural killer (NK) cells. NK cell infiltration immediately preceded mesenchymal cell expansion, which occurred starting 7 days after CA/CPR. Immune cells colocalized with mesenchymal cells, accumulating in the areas of fibrosis. Analysis of previously published single-cell or single-nuclear RNA sequencing data revealed GzA as a potential mediator of immune to mesenchymal communication. GzA administration to fibroblasts in vitro induced cell growth and proliferation. Pharmacologic blockade of GzA signaling in vivo attenuated fibrosis and improved renal function after CA/CPR. Conclusions Renal inflammation occurs during cardiorenal syndrome, which correlates with mesenchymal cell expansion. GzA, produced by NK cells, presents a novel therapeutic target to prevent the transition to CKD after AKI.
Collapse
Affiliation(s)
- Kevin G. Burfeind
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon
| | | | | | - Mahaba Eiwaz
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon
- Portland VA Medical Center, Portland, Oregon
| | - Michael P. Hutchens
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon
- Portland VA Medical Center, Portland, Oregon
| |
Collapse
|
11
|
Goto H, Kinoshita M, Oshima N. Heatstroke-induced acute kidney injury and the innate immune system. Front Med (Lausanne) 2023; 10:1250457. [PMID: 37614951 PMCID: PMC10442538 DOI: 10.3389/fmed.2023.1250457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 07/28/2023] [Indexed: 08/25/2023] Open
Abstract
Heatstroke can cause multiple organ failure and systemic inflammatory response syndrome as the body temperature rises beyond the body's ability to regulate temperature in a hot environment. Previous studies have indicated that heatstroke-induced acute kidney injury (AKI) can lead to chronic kidney disease. Therefore, there is an urgent need to elucidate the mechanism of heatstroke-induced AKI and to establish methods for its prevention and treatment. Recent reports have revealed that innate immunity, including neutrophils, macrophages, lymphocytes, and mast cells, is deeply involved in heat-induced AKI. In this review, we will discuss the roles of each immune cell in heat-induced renal injury and their potential therapeutic use.
Collapse
Affiliation(s)
- Hiroyasu Goto
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Japan
| | - Manabu Kinoshita
- Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Japan
| | - Naoki Oshima
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Japan
| |
Collapse
|
12
|
Tamura T, Cheng C, Chen W, Merriam LT, Athar H, Kim YH, Manandhar R, Amir Sheikh MD, Pinilla-Vera M, Varon J, Hou PC, Lawler PR, Oldham WM, Seethala RR, Tesfaigzi Y, Weissman AJ, Baron RM, Ichinose F, Berg KM, Bohula EA, Morrow DA, Chen X, Kim EY. Single-cell transcriptomics reveal a hyperacute cytokine and immune checkpoint axis after cardiac arrest in patients with poor neurological outcome. MED 2023; 4:432-456.e6. [PMID: 37257452 PMCID: PMC10524451 DOI: 10.1016/j.medj.2023.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 03/06/2023] [Accepted: 05/02/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND Most patients hospitalized after cardiac arrest (CA) die because of neurological injury. The systemic inflammatory response after CA is associated with neurological injury and mortality but remains poorly defined. METHODS We determine the innate immune network induced by clinical CA at single-cell resolution. FINDINGS Immune cell states diverge as early as 6 h post-CA between patients with good or poor neurological outcomes 30 days after CA. Nectin-2+ monocyte and Tim-3+ natural killer (NK) cell subpopulations are associated with poor outcomes, and interactome analysis highlights their crosstalk via cytokines and immune checkpoints. Ex vivo studies of peripheral blood cells from CA patients demonstrate that immune checkpoints are a compensatory mechanism against inflammation after CA. Interferon γ (IFNγ)/interleukin-10 (IL-10) induced Nectin-2 on monocytes; in a negative feedback loop, Nectin-2 suppresses IFNγ production by NK cells. CONCLUSIONS The initial hours after CA may represent a window for therapeutic intervention in the resolution of inflammation via immune checkpoints. FUNDING This work was supported by funding from the American Heart Association, Brigham and Women's Hospital Department of Medicine, the Evergreen Innovation Fund, and the National Institutes of Health.
Collapse
Affiliation(s)
- Tomoyoshi Tamura
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Changde Cheng
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Wenan Chen
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Louis T Merriam
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Humra Athar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Yaunghyun H Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Reshmi Manandhar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Muhammad Dawood Amir Sheikh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Mayra Pinilla-Vera
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jack Varon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Peter C Hou
- Harvard Medical School, Boston, MA 02115, USA; Division of Emergency Critical Care Medicine, Department of Emergency Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Patrick R Lawler
- Peter Munk Cardiac Centre, Toronto General Hospital, Toronto, ON M5G 2N2, Canada; McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - William M Oldham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Raghu R Seethala
- Harvard Medical School, Boston, MA 02115, USA; Division of Emergency Critical Care Medicine, Department of Emergency Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Yohannes Tesfaigzi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Alexandra J Weissman
- Department of Emergency Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Rebecca M Baron
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Fumito Ichinose
- Harvard Medical School, Boston, MA 02115, USA; Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Katherine M Berg
- Harvard Medical School, Boston, MA 02115, USA; Division of Pulmonary, Critical Care, and Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Erin A Bohula
- Harvard Medical School, Boston, MA 02115, USA; Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - David A Morrow
- Harvard Medical School, Boston, MA 02115, USA; Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Xiang Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Edy Y Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
13
|
Mak ML, Reid KT, Crome SQ. Protective and pathogenic functions of innate lymphoid cells in transplantation. Clin Exp Immunol 2023; 213:23-39. [PMID: 37119279 PMCID: PMC10324558 DOI: 10.1093/cei/uxad050] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/27/2023] [Accepted: 04/28/2023] [Indexed: 05/01/2023] Open
Abstract
Innate lymphoid cells (ILCs) are a family of lymphocytes with essential roles in tissue homeostasis and immunity. Along with other tissue-resident immune populations, distinct subsets of ILCs have important roles in either promoting or inhibiting immune tolerance in a variety of contexts, including cancer and autoimmunity. In solid organ and hematopoietic stem cell transplantation, both donor and recipient-derived ILCs could contribute to immune tolerance or rejection, yet understanding of protective or pathogenic functions are only beginning to emerge. In addition to roles in directing or regulating immune responses, ILCs interface with parenchymal cells to support tissue homeostasis and even regeneration. Whether specific ILCs are tissue-protective or enhance ischemia reperfusion injury or fibrosis is of particular interest to the field of transplantation, beyond any roles in limiting or promoting allograft rejection or graft-versus host disease. Within this review, we discuss the current understanding of ILCs functions in promoting immune tolerance and tissue repair at homeostasis and in the context of transplantation and highlight where targeting or harnessing ILCs could have applications in novel transplant therapies.
Collapse
Affiliation(s)
- Martin L Mak
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Kyle T Reid
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Sarah Q Crome
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| |
Collapse
|
14
|
Anrather J, Marks K. Highlighting a New Potential Target in Stroke: Immunoreceptor NKG2D. J Am Heart Assoc 2023:e030482. [PMID: 37301754 DOI: 10.1161/jaha.123.030482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 06/12/2023]
Affiliation(s)
- Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine New York NY USA
| | - Kimberly Marks
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine New York NY USA
| |
Collapse
|
15
|
David C, Ruck T, Rolfes L, Mencl S, Kraft P, Schuhmann MK, Schroeter CB, Jansen R, Langhauser F, Mausberg AK, Fender AC, Meuth SG, Kleinschnitz C. Impact of NKG2D Signaling on Natural Killer and T-Cell Function in Cerebral Ischemia. J Am Heart Assoc 2023:e029529. [PMID: 37301761 DOI: 10.1161/jaha.122.029529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/04/2023] [Indexed: 06/12/2023]
Abstract
Background Typically defined as a thromboinflammatory disease, ischemic stroke features early and delayed inflammatory responses, which determine the extent of ischemia-related brain damage. T and natural killer cells have been implicated in neuronal cytotoxicity and inflammation, but the precise mechanisms of immune cell-mediated stroke progression remain poorly understood. The activating immunoreceptor NKG2D is expressed on both natural killer and T cells and may be critically involved. Methods and Results An anti-NKG2D blocking antibody alleviated stroke outcome in terms of infarct volume and functional deficits, coinciding with reduced immune cell infiltration into the brain and improved survival in the animal model of cerebral ischemia. Using transgenic knockout models devoid of certain immune cell types and immunodeficient mice supplemented with different immune cell subsets, we dissected the functional contribution of NKG2D signaling by different NKG2D-expressing cells in stroke pathophysiology. The observed effect of NKG2D signaling in stroke progression was shown to be predominantly mediated by natural killer and CD8+ T cells. Transfer of T cells with monovariant T-cell receptors into immunodeficient mice with and without pharmacological blockade of NKG2D revealed activation of CD8+ T cells irrespective of antigen specificity. Detection of the NKG2D receptor and its ligands in brain samples of patients with stroke strengthens the relevance of preclinical observations in human disease. Conclusions Our findings provide a mechanistic insight into NKG2D-dependent natural killer- and T-cell-mediated effects in stroke pathophysiology.
Collapse
Affiliation(s)
- Christina David
- Department of Neurology With Center for Translational Neuro- and Behavioral Sciences (C-TNBS) University Hospital Essen, University Duisburg-Essen Essen Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty Heinrich-Heine-University Düsseldorf Germany
| | - Leoni Rolfes
- Department of Neurology, Medical Faculty Heinrich-Heine-University Düsseldorf Germany
| | - Stine Mencl
- Department of Neurology With Center for Translational Neuro- and Behavioral Sciences (C-TNBS) University Hospital Essen, University Duisburg-Essen Essen Germany
| | - Peter Kraft
- Department of Neurology Hospital Main-Spessart Lohr am Main Germany
- Department of Neurology University Hospital Würzburg Würzburg Germany
| | | | - Christina B Schroeter
- Department of Neurology, Medical Faculty Heinrich-Heine-University Düsseldorf Germany
| | - Robin Jansen
- Department of Neurology, Medical Faculty Heinrich-Heine-University Düsseldorf Germany
| | - Friederike Langhauser
- Department of Neurology With Center for Translational Neuro- and Behavioral Sciences (C-TNBS) University Hospital Essen, University Duisburg-Essen Essen Germany
| | - Anne K Mausberg
- Department of Neurology With Center for Translational Neuro- and Behavioral Sciences (C-TNBS) University Hospital Essen, University Duisburg-Essen Essen Germany
| | - Anke C Fender
- Department of Pharmacology University Hospital Essen, University of Duisburg-Essen Essen Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty Heinrich-Heine-University Düsseldorf Germany
| | - Christoph Kleinschnitz
- Department of Neurology With Center for Translational Neuro- and Behavioral Sciences (C-TNBS) University Hospital Essen, University Duisburg-Essen Essen Germany
| |
Collapse
|
16
|
Ma K, Zheng ZR, Meng Y. Natural Killer Cells, as the Rising Point in Tissues, Are Forgotten in the Kidney. Biomolecules 2023; 13:biom13050748. [PMID: 37238618 DOI: 10.3390/biom13050748] [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: 03/03/2023] [Revised: 04/13/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Natural killer (NK) cells are members of a rapidly expanding family of innate lymphoid cells (ILCs). NK cells play roles in the spleen, periphery, and in many tissues, such as the liver, uterine, lung, adipose, and so on. While the immunological functions of NK cells are well established in these organs, comparatively little is known about NK cells in the kidney. Our understanding of NK cells is rapidly rising, with more and more studies highlighting the functional significance of NK cells in different types of kidney diseases. Recent progress has been made in translating these findings to clinical diseases that occur in the kidney, with indications of subset-specific roles of NK cells in the kidney. For the development of targeted therapeutics to delay kidney disease progression, a better understanding of the NK cell with respect to the mechanisms of kidney diseases is necessary. In order to promote the targeted treatment ability of NK cells in clinical diseases, in this paper we demonstrate the roles that NK cells play in different organs, especially the functions of NK cells in the kidney.
Collapse
Affiliation(s)
- Ke Ma
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou 510000, China
| | - Zi-Run Zheng
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou 510000, 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
| |
Collapse
|
17
|
Makita K, Otsuka N, Tomaru U, Taniguchi K, Kasahara M. NKG2D Ligand Expression Induced by Oxidative Stress Mitigates Cutaneous Ischemia-Reperfusion Injury. J Histochem Cytochem 2023; 71:61-72. [PMID: 36762536 PMCID: PMC10088101 DOI: 10.1369/00221554221147582] [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: 09/19/2022] [Accepted: 12/07/2022] [Indexed: 02/11/2023] Open
Abstract
Pressure ulcers represent a crucial clinical problem, especially in hospitalized patients. Ischemia-reperfusion (I-R) is an important cause of these lesions. Natural killer (NK), invariant NK T (iNKT), and dendritic epidermal T-cells, which express the natural killer group 2, member D (NKG2D) receptor, have been reported to have physiological roles in skin tissue repair and wound healing. However, a role for NKG2D-NKG2D ligand interactions in I-R-induced skin injury has not been determined. Using a murine pressure ulcer model, we demonstrated that I-R-induced ulcers in NKG2D-deficient mice were larger than those in wild-type or T-cell receptor δ knockout mice. Histopathological evaluation revealed that accumulation of macrophages and neutrophils at the peripheral deep dermis and subcutaneous tissue of the ulcers was enhanced in NKG2D-deficient mice. Rae-1 mRNA, which encodes an NKG2D ligand, was induced, and RAE-1 protein was detected immunohistochemically in fibroblasts and inflammatory cells in the dermis after reperfusion. RAE-1 expression was also increased in primary mouse fibroblasts treated with sodium arsenite. These results suggested that NKG2D ligand expression was induced by oxidative stress after I-R injury and support a putative role for this ligand in wound repair. Furthermore, the influx of NKG2D-positive cells at I-R sites may mitigate pressure ulcers via NKG2D-NKG2D ligand interactions.
Collapse
Affiliation(s)
- Keishi Makita
- Department of Pathology, Graduate School of Medicine and
Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Department of Pathology, Sapporo City General Hospital,
Sapporo, Japan
| | - Noriyuki Otsuka
- Department of Pathology, Graduate School of Medicine and
Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Department of Surgical Pathology, Hokkaido University
Hospital, Sapporo, Japan
| | - Utano Tomaru
- Department of Pathology, Graduate School of Medicine and
Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Department of Surgical Pathology, Hokkaido University
Hospital, Sapporo, Japan
| | - Koji Taniguchi
- Department of Pathology, Graduate School of Medicine and
Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Masanori Kasahara
- Department of Pathology, Graduate School of Medicine and
Faculty of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
18
|
Immune response associated with ischemia and reperfusion injury during organ transplantation. Inflamm Res 2022; 71:1463-1476. [PMID: 36282292 PMCID: PMC9653341 DOI: 10.1007/s00011-022-01651-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 12/03/2022] Open
Abstract
Background Ischemia and reperfusion injury (IRI) is an ineluctable immune-related pathophysiological process during organ transplantation, which not only causes a shortage of donor organs, but also has long-term and short-term negative consequences on patients. Severe IRI-induced cell death leads to the release of endogenous substances, which bind specifically to receptors on immune cells to initiate an immune response. Although innate and adaptive immunity have been discovered to play essential roles in IRI in the context of organ transplantation, the pathway and precise involvement of the immune response at various stages has not yet to be elucidated. Methods We combined “IRI” and “organ transplantation” with keywords, respectively such as immune cells, danger signal molecules, macrophages, neutrophils, natural killer cells, complement cascade, T cells or B cells in PubMed and the Web of Science to search for relevant literatures. Conclusion Comprehension of the immune mechanisms involved in organ transplantation is promising for the treatment of IRI, this review summarizes the similarities and differences in both innate and adaptive immunity and advancements in the immune response associated with IRI during diverse organ transplantation.
Collapse
|
19
|
Zhao S, Liu Y, Zhou C, Chen Z, Cai Z, Han J, Xiao J, Xiao Q. Prediction model of delayed graft function based on clinical characteristics combined with serum IL-2 levels. BMC Nephrol 2022; 23:284. [PMID: 35971094 PMCID: PMC9377118 DOI: 10.1186/s12882-022-02908-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/04/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Kidney transplantation is an effective treatment for end-stage renal disease (ESRD). Delayed graft function (DGF) is a common complication after kidney transplantation and exerts substantial effects on graft function and long-term graft survival. Therefore, the construction of an effective model to predict the occurrence of DGF is particularly important. METHODS Seventy-one patients receiving their first kidney transplant at the First Affiliated Hospital of Nanchang University from October 2020 to October 2021 were enrolled in the discovery cohort. Based on clinical characteristics and serum markers, a logistic regression model was used to simulate the risk of DGF in the discovery cohort. The DGF prediction model was named the prediction system and was composed of risk factors related to DGF. Thirty-two patients receiving a kidney transplant at the First Affiliated Hospital of Nanchang University from October 2021 to February 2022 were enrolled in the validation cohort. The validation cohort was used to verify the accuracy and reliability of the prediction model. RESULTS Cold ischemia time (CIT), donor history of diabetes mellitus, donor interleukin-2 (IL-2) level and donor terminal creatinine level constitute the prediction system. In the validation test, the area under the receiver operating characteristic curve (AUC) was 0.867 for the prediction system, and good calibration of the model was confirmed in the validation cohort. CONCLUSIONS This study constructed a reliable and highly accurate prediction model that provides a practical tool for predicting DGF. Additionally, IL-2 participates in the kidney injury process and may be a potential marker of kidney injury.
Collapse
Affiliation(s)
- Shitao Zhao
- Department of Transplantation, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie Street, Nanchang, 330006, Jiangxi, China
| | - Yuan Liu
- Department of Transplantation, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie Street, Nanchang, 330006, Jiangxi, China
| | - Chen Zhou
- Department of Transplantation, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie Street, Nanchang, 330006, Jiangxi, China
| | - Zide Chen
- Department of Transplantation, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie Street, Nanchang, 330006, Jiangxi, China
| | - Zeyu Cai
- Department of Transplantation, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie Street, Nanchang, 330006, Jiangxi, China
| | - JiaLiang Han
- Department of Transplantation, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie Street, Nanchang, 330006, Jiangxi, China
| | - Jiansheng Xiao
- Department of Transplantation, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie Street, Nanchang, 330006, Jiangxi, China.
| | - Qi Xiao
- Department of Transplantation, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zhengjie Street, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
20
|
Kang J, Liggett JR, Patil D, Ranjit S, Loh K, Duttargi A, Cui Y, Oza K, Frank BS, Kwon D, Kallakury B, Robson SC, Fishbein TM, Cui W, Khan K, Kroemer A. Type 1 Innate Lymphoid Cells Are Proinflammatory Effector Cells in Ischemia-Reperfusion Injury of Steatotic Livers. Front Immunol 2022; 13:899525. [PMID: 35833123 PMCID: PMC9272906 DOI: 10.3389/fimmu.2022.899525] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
Innate lymphoid cells (ILCs), the most recently described family of lymphoid cells, play fundamental roles in tissue homeostasis through the production of key cytokine. Group 1 ILCs, comprised of conventional natural killer cells (cNKs) and type 1 ILCs (ILC1s), have been implicated in regulating immune-mediated inflammatory diseases. However, the role of ILC1s in nonalcoholic fatty liver disease (NAFLD) and ischemia-reperfusion injury (IRI) is unclear. Here, we investigated the role of ILC1 and cNK cells in a high-fat diet (HFD) murine model of partial warm IRI. We demonstrated that hepatic steatosis results in more severe IRI compared to non-steatotic livers. We further elicited that HFD-IRI mice show a significant increase in the ILC1 population, whereas the cNK population was unchanged. Since ILC1 and cNK are major sources of IFN-γ and TNF-α, we measured the level of ex vivo cytokine expression in normal diet (ND)-IRI and HFD-IRI conditions. We found that ILC1s in HFD-IRI mice produce significantly more IFN-γ and TNF-α when compared to ND-IRI. To further assess whether ILC1s are key proinflammatory effector cells in hepatic IRI of fatty livers, we studied both Rag1−/− mice, which possess cNK cells, and a substantial population of ILC1s versus the newly generated Rag1−/−Tbx21−/− double knockout (Rag1-Tbet DKO) mice, which lack type 1 ILCs, under HFD IRI conditions. Importantly, HFD Rag1-Tbet DKO mice showed significant protection from hepatic injury upon IRI when compared to Rag1−/− mice, suggesting that T-bet-expressing ILC1s play a role, at least in part, as proinflammatory effector cells in hepatic IRI under steatotic conditions.
Collapse
Affiliation(s)
- Jiman Kang
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, United States
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
| | - Jedson R. Liggett
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, United States
- Naval Medical Center Portsmouth, Portsmouth, VA, United States
| | - Digvijay Patil
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, United States
| | - Suman Ranjit
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
| | - Katrina Loh
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, United States
| | - Anju Duttargi
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States
| | - Yuki Cui
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, United States
| | - Kesha Oza
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, United States
| | - Brett S. Frank
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, United States
| | - DongHyang Kwon
- Department of Pathology, MedStar Georgetown University Hospital, Washington, DC, United States
| | - Bhaskar Kallakury
- Department of Pathology, MedStar Georgetown University Hospital, Washington, DC, United States
| | - Simon C. Robson
- Departments of Anesthesiology and Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Thomas M. Fishbein
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, United States
| | - Wanxing Cui
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, United States
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
| | - Khalid Khan
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, United States
| | - Alexander Kroemer
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, United States
- *Correspondence: Alexander Kroemer, ;
| |
Collapse
|
21
|
Krupa A, Krupa MM, Pawlak K. Indoleamine 2,3 Dioxygenase 1-The Potential Link between the Innate Immunity and the Ischemia-Reperfusion-Induced Acute Kidney Injury? Int J Mol Sci 2022; 23:6176. [PMID: 35682852 PMCID: PMC9181334 DOI: 10.3390/ijms23116176] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/30/2022] [Indexed: 12/13/2022] Open
Abstract
Ischemia-reperfusion injury (IRI) is of the most common causes of acute kidney injury (AKI); nevertheless, the mechanisms responsible for both early kidney injury and the reparative phase are not fully recognised. The inflammatory response following ischemia is characterised by the crosstalk between cells belonging to the innate immune system-dendritic cells (DCs), macrophages, neutrophils, natural killer (NK) cells, and renal tubular epithelial cells (RTECs). A tough inflammatory response can damage the renal tissue; it may also have a protective effect leading to the repair after IRI. Indoleamine 2,3 dioxygenase 1 (IDO1), the principal enzyme of the kynurenine pathway (KP), has a broad spectrum of immunological activity from stimulation to immunosuppressive activity in inflamed areas. IDO1 expression occurs in cells of the innate immunity and RTECs during IRI, resulting in local tryptophan (TRP) depletion and generation of kynurenines, and both of these mechanisms contribute to the immunosuppressive effect. Nonetheless, it is unknown if the above mechanism can play a harmful or preventive role in IRI-induced AKI. Despite the scarcity of literature in this field, the current review attempts to present a possible role of IDO1 activation in the regulation of the innate immune system in IRI-induced AKI.
Collapse
Affiliation(s)
- Anna Krupa
- Department of Internal Medicine and Metabolic Diseases, Medical University of Bialystok, M. Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland;
| | - Mikolaj M. Krupa
- Department of Monitored Pharmacotherapy, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland;
| | - Krystyna Pawlak
- Department of Monitored Pharmacotherapy, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland;
| |
Collapse
|
22
|
Li D, Zhang H, Wu X, Dai Q, Tang S, Liu Y, Yang S, Zhang W. Role of tRNA derived fragments in renal ischemia-reperfusion injury. Ren Fail 2022; 44:815-825. [PMID: 35546262 PMCID: PMC9116270 DOI: 10.1080/0886022x.2022.2072336] [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] [Indexed: 11/21/2022] Open
Abstract
Background Ischemia–reperfusion injury (IRI) is one of the major causes of acute kidney injury (AKI). tRNA derived fragments (tRFs/tiRNAs) are groups of small noncoding RNAs derived from tRNAs. To date, the role of tRFs/tiRNAs in renal IRI has not been reported. Herein, we aimed to investigate the involvement of tRFs/tiRNAs in the occurrence and development of ischemia–reperfusion-induced AKI. Methods Moderate/severe renal IRI mouse models were established by bilateral renal pedicle clamping. The tRF/tiRNA profiles of healthy controls and moderate/severe IRI-stressed kidney tissues were sequenced by Illumina NextSeq 500. Candidate differentially expressed tiRNAs were further verified by RT-qPCR. Biological analysis was also performed. Results Overall, 152 tRFs/tiRNAs were differentially expressed in the moderate ischemic injury group compared with the normal control group (FC > 2, p < 0.05), of which 47 were upregulated and 105 were downregulated; in the severe ischemic injury group, 285 tRFs/tiRNAs were differentially expressed (FC > 2, p < 0.05), of which 157 were upregulated, and 128 were downregulated. RT-qPCR determination of eight abundantly expressed tiRNAs was consistent with the sequencing results. Gene Ontology analysis for target genes of the tRFs/tiRNAs showed that the most enriched cell components, molecular functions and biological processes were Golgi apparatus, cytoplasmic vesicles, protein binding, cellular protein localization and multicellular organism development. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that these target genes were mainly involved in the natural killer cell mediated cytotoxicity pathway, citrate cycle, and regulation of actin cytoskeleton signaling pathway. Conclusion Our results indicated that tRFs/tiRNAs were involved in renal IRI. These tRFs/tiRNAs may be effective partly via regulation of renal immunity, inflammation and metabolism processes. Candidate genes, including tiRNA-Gly-GCC-003, tiRNA-Lys-CTT-003, and tiRNA-His-GTG-002, might be potential biomarkers and therapeutic targets of ischemia–reperfusion injury-induced acute kidney injury.
Collapse
Affiliation(s)
- Dan Li
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hao Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xueqin Wu
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qing Dai
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Shiqi Tang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yan Liu
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Shikun Yang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
23
|
Marshall JL, Noel T, Wang QS, Chen H, Murray E, Subramanian A, Vernon KA, Bazua-Valenti S, Liguori K, Keller K, Stickels RR, McBean B, Heneghan RM, Weins A, Macosko EZ, Chen F, Greka A. High-resolution Slide-seqV2 spatial transcriptomics enables discovery of disease-specific cell neighborhoods and pathways. iScience 2022; 25:104097. [PMID: 35372810 PMCID: PMC8971939 DOI: 10.1016/j.isci.2022.104097] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/15/2022] [Accepted: 03/11/2022] [Indexed: 12/21/2022] Open
Abstract
High-resolution spatial transcriptomics enables mapping of RNA expression directly from intact tissue sections; however, its utility for the elucidation of disease processes and therapeutically actionable pathways remains unexplored. We applied Slide-seqV2 to mouse and human kidneys, in healthy and distinct disease paradigms. First, we established the feasibility of Slide-seqV2 in tissue from nine distinct human kidneys, which revealed a cell neighborhood centered around a population of LYVE1+ macrophages. Second, in a mouse model of diabetic kidney disease, we detected changes in the cellular organization of the spatially restricted kidney filter and blood-flow-regulating apparatus. Third, in a mouse model of a toxic proteinopathy, we identified previously unknown, disease-specific cell neighborhoods centered around macrophages. In a spatially restricted subpopulation of epithelial cells, we discovered perturbations in 77 genes associated with the unfolded protein response. Our studies illustrate and experimentally validate the utility of Slide-seqV2 for the discovery of disease-specific cell neighborhoods.
Collapse
Affiliation(s)
- Jamie L. Marshall
- Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Teia Noel
- Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Qingbo S. Wang
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Program in Bioinformatics and Integrative Genomics, Harvard Medical School, Boston, MA 02115, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Statistical Genetics, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Haiqi Chen
- Program in Cell Circuits and Epigenetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Evan Murray
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ayshwarya Subramanian
- Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Katherine A. Vernon
- Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Silvana Bazua-Valenti
- Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Katie Liguori
- Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Keith Keller
- Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Robert R. Stickels
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Graduate School of Arts and Sciences, Harvard University, Cambridge, MA 02115, USA
- Division of Medical Science, Harvard University, Boston, MA 02115, USA
| | - Breanna McBean
- Broad Summer Research Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Rowan M. Heneghan
- Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Astrid Weins
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Evan Z. Macosko
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Fei Chen
- Program in Cell Circuits and Epigenetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Anna Greka
- Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
24
|
Collett JA, Ortiz-Soriano V, Li X, Flannery AH, Toto RD, Moe OW, Basile DP, Neyra JA. Serum IL-17 levels are higher in critically ill patients with AKI and associated with worse outcomes. Crit Care 2022; 26:107. [PMID: 35422004 PMCID: PMC9008961 DOI: 10.1186/s13054-022-03976-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 04/03/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Interleukin-17 (IL-17) antagonism in rats reduces the severity and progression of AKI. IL-17-producing circulating T helper-17 (TH17) cells is increased in critically ill patients with AKI indicating that this pathway is also activated in humans. We aim to compare serum IL-17A levels in critically ill patients with versus without AKI and to examine their relationship with mortality and major adverse kidney events (MAKE). METHODS Multicenter, prospective study of ICU patients with AKI stage 2 or 3 and without AKI. Samples were collected at 24-48 h after AKI diagnosis or ICU admission (in those without AKI) [timepoint 1, T1] and 5-7 days later [timepoint 2, T2]. MAKE was defined as the composite of death, dependence on kidney replacement therapy or a reduction in eGFR of ≥ 30% from baseline up to 90 days following hospital discharge. RESULTS A total of 299 patients were evaluated. Patients in the highest IL-17A tertile (versus lower tertiles) at T1 had higher acuity of illness and comorbidity scores. Patients with AKI had higher levels of IL-17A than those without AKI: T1 1918.6 fg/ml (692.0-5860.9) versus 623.1 fg/ml (331.7-1503.4), p < 0.001; T2 2167.7 fg/ml (839.9-4618.9) versus 1193.5 fg/ml (523.8-2198.7), p = 0.006. Every onefold higher serum IL-17A at T1 was independently associated with increased risk of hospital mortality (aOR 1.35, 95% CI: 1.06-1.73) and MAKE (aOR 1.26, 95% CI: 1.02-1.55). The highest tertile of IL-17A (vs. the lowest tertile) was also independently associated with higher risk of MAKE (aOR 3.03, 95% CI: 1.34-6.87). There was no effect modification of these associations by AKI status. IL-17A levels remained significantly elevated at T2 in patients that died or developed MAKE. CONCLUSIONS Serum IL-17A levels measured by the time of AKI diagnosis or ICU admission were differentially elevated in critically ill patients with AKI when compared to those without AKI and were independently associated with hospital mortality and MAKE.
Collapse
Affiliation(s)
- Jason A Collett
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Victor Ortiz-Soriano
- Division of Nephrology, Department of Internal Medicine, Bone and Mineral Metabolism, University of Kentucky Medical Center, University of Kentucky, 800 Rose St., MN668, Lexington, KY, 40536, USA
| | - Xilong Li
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alexander H Flannery
- Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, KY, USA
| | - Robert D Toto
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - David P Basile
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Javier A Neyra
- Division of Nephrology, Department of Internal Medicine, Bone and Mineral Metabolism, University of Kentucky Medical Center, University of Kentucky, 800 Rose St., MN668, Lexington, KY, 40536, USA.
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
25
|
Murphy JM, Ngai L, Mortha A, Crome SQ. Tissue-Dependent Adaptations and Functions of Innate Lymphoid Cells. Front Immunol 2022; 13:836999. [PMID: 35359972 PMCID: PMC8960279 DOI: 10.3389/fimmu.2022.836999] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/11/2022] [Indexed: 12/21/2022] Open
Abstract
Tissue-resident immune cells reside in distinct niches across organs, where they contribute to tissue homeostasis and rapidly respond to perturbations in the local microenvironment. Innate lymphoid cells (ILCs) are a family of innate immune cells that regulate immune and tissue homeostasis. Across anatomical locations throughout the body, ILCs adopt tissue-specific fates, differing from circulating ILC populations. Adaptations of ILCs to microenvironmental changes have been documented in several inflammatory contexts, including obesity, asthma, and inflammatory bowel disease. While our understanding of ILC functions within tissues have predominantly been based on mouse studies, development of advanced single cell platforms to study tissue-resident ILCs in humans and emerging patient-based data is providing new insights into this lymphocyte family. Within this review, we discuss current concepts of ILC fate and function, exploring tissue-specific functions of ILCs and their contribution to health and disease across organ systems.
Collapse
Affiliation(s)
- Julia M. Murphy
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Louis Ngai
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Arthur Mortha
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Sarah Q. Crome
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
26
|
Hematological Ratios Are Associated with Acute Kidney Injury and Mortality in Patients That Present with Suspected Infection at the Emergency Department. J Clin Med 2022; 11:jcm11041017. [PMID: 35207289 PMCID: PMC8874958 DOI: 10.3390/jcm11041017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/03/2022] [Accepted: 02/10/2022] [Indexed: 01/27/2023] Open
Abstract
The early recognition of acute kidney injury (AKI) is essential to improve outcomes and prevent complications such as chronic kidney disease, the need for renal-replacement therapy, and an increased length of hospital stay. Increasing evidence shows that inflammation plays an important role in the pathophysiology of AKI and mortality. Several inflammatory hematological ratios can be used to measure systemic inflammation. Therefore, the association between these ratios and outcomes (AKI and mortality) in patients suspected of having an infection at the emergency department was investigated. Data from the SPACE cohort were used. Cox regression was performed to investigate the association between seven hematological ratios and outcomes. A total of 1889 patients were included, of which 160 (8.5%) patients developed AKI and 102 (5.4%) died in <30 days. The Cox proportional-hazards model revealed that the neutrophil-to-lymphocyte ratio (NLR), segmented-neutrophil-to-monocyte ratio (SMR), and neutrophil-lymphocyte-platelet ratio (NLPR) are independently associated with AKI <30 days after emergency-department presentation. Additionally, the NLR, SMR and NLPR were associated with 30-day all-cause mortality. These findings are an important step forward for the early recognition of AKI. The use of these markers might enable emergency-department physicians to recognize and treat AKI in an early phase to potentially prevent complications.
Collapse
|
27
|
Uchida T, Seki S, Oda T. Infections, Reactions of Natural Killer T Cells and Natural Killer Cells, and Kidney Injury. Int J Mol Sci 2022; 23:ijms23010479. [PMID: 35008905 PMCID: PMC8745257 DOI: 10.3390/ijms23010479] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 01/21/2023] Open
Abstract
Natural killer T (NKT) cells and NK cells are representative innate immune cells that perform antitumor and antimicrobial functions. The involvement of these cells in various renal diseases, including acute kidney injury (AKI), has recently become evident. Murine NKT cells are activated and cause AKI in response to various stimuli, such as their specific ligand, cytokines, and bacterial components. Both renal vascular endothelial cell injury (via the perforin-mediated pathway) and tubular epithelial cell injury (via the tumor necrosis factor-alpha/Fas ligand pathway) are independently involved in the pathogenesis of AKI. NK cells complement the functions of NKT cells, thereby contributing to the development of infection-associated AKI. Human CD56+ T cells, which are a functional counterpart of murine NKT cells, as well as a subpopulation of CD56+ NK cells, strongly damage intrinsic renal cells in vitro upon their activation, possibly through mechanisms similar to those in mice. These cells are also thought to be involved in the acute exacerbation of pre-existing glomerulonephritis triggered by infection in humans, and their roles in sepsis-associated AKI are currently under investigation. In this review, we will provide an overview of the recent advances in the understanding of the association among infections, NKT and NK cells, and kidney injury, which is much more profound than previously considered. The important role of liver macrophages in the activation of NKT cells will also be introduced.
Collapse
Affiliation(s)
- Takahiro Uchida
- Kidney Disease Center, Department of Nephrology and Blood Purification, Tokyo Medical University Hachioji Medical Center, Tokyo 193-0998, Japan;
| | - Shuhji Seki
- Department of Immunology and Microbiology, National Defense Medical College, Saitama 359-8513, Japan;
| | - Takashi Oda
- Kidney Disease Center, Department of Nephrology and Blood Purification, Tokyo Medical University Hachioji Medical Center, Tokyo 193-0998, Japan;
- Correspondence: ; Tel.: +81-42-665-5611; Fax: +81-42-665-1796
| |
Collapse
|
28
|
Callemeyn J, Lamarthée B, Koenig A, Koshy P, Thaunat O, Naesens M. Allorecognition and the spectrum of kidney transplant rejection. Kidney Int 2021; 101:692-710. [PMID: 34915041 DOI: 10.1016/j.kint.2021.11.029] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 10/05/2021] [Accepted: 11/08/2021] [Indexed: 12/18/2022]
Abstract
Detection of mismatched human leukocyte antigens by adaptive immune cells is considered as the main cause of transplant rejection, leading to either T-cell mediated rejection or antibody-mediated rejection. This canonical view guided the successful development of immunosuppressive therapies and shaped the diagnostic Banff classification for kidney transplant rejection that is used in clinics worldwide. However, several observations have recently emerged that question this dichotomization between T-cell mediated rejection and antibody-mediated rejection, related to heterogeneity in the serology, histology, and prognosis of the rejection phenotypes. In parallel, novel insights were obtained concerning the dynamics of donor-specific anti-human leukocyte antigen antibodies, the immunogenicity of donor-recipient non-human leukocyte antigen mismatches, and the autoreactivity against self-antigens. Moreover, the potential of innate allorecognition was uncovered, as exemplified by natural killer cell-mediated microvascular inflammation through missing self, and by the emerging evidence on monocyte-driven allorecognition. In this review, we highlight the gaps in the current classification of rejection, provide an overview of the expanding insights into the mechanisms of allorecognition, and critically appraise how these could improve our understanding and clinical approach to kidney transplant rejection. We argue that consideration of the complex interplay of various allorecognition mechanisms can foster a more integrated view of kidney transplant rejection and can lead to improved risk stratification, targeted therapies, and better outcome after kidney transplantation.
Collapse
Affiliation(s)
- Jasper Callemeyn
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium; Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Baptiste Lamarthée
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium; Necker-Enfants Malades Institute, French National Institute of Health and Medical Research (INSERM) Unit 1151, Paris, France
| | - Alice Koenig
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, University Lyon, Lyon, France; Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France; Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Priyanka Koshy
- Department of Morphology and Molecular Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Olivier Thaunat
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, University Lyon, Lyon, France; Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France; Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Maarten Naesens
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium; Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
29
|
Charmetant X, Bachelet T, Déchanet-Merville J, Walzer T, Thaunat O. Innate (and Innate-like) Lymphoid Cells: Emerging Immune Subsets With Multiple Roles Along Transplant Life. Transplantation 2021; 105:e322-e336. [PMID: 33859152 DOI: 10.1097/tp.0000000000003782] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Transplant immunology is currently largely focused on conventional adaptive immunity, particularly T and B lymphocytes, which have long been considered as the only cells capable of allorecognition. In this vision, except for the initial phase of ischemia/reperfusion, during which the role of innate immune effectors is well established, the latter are largely considered as "passive" players, recruited secondarily to amplify graft destruction processes during rejection. Challenging this prevalent dogma, the recent progresses in basic immunology have unraveled the complexity of the innate immune system and identified different subsets of innate (and innate-like) lymphoid cells. As most of these cells are tissue-resident, they are overrepresented among passenger leukocytes. Beyond their role in ischemia/reperfusion, some of these subsets have been shown to be capable of allorecognition and/or of regulating alloreactive adaptive responses, suggesting that these emerging immune players are actively involved in most of the life phases of the grafts and their recipients. Drawing upon the inventory of the literature, this review synthesizes the current state of knowledge of the role of the different innate (and innate-like) lymphoid cell subsets during ischemia/reperfusion, allorecognition, and graft rejection. How these subsets also contribute to graft tolerance and the protection of chronically immunosuppressed patients against infectious and cancerous complications is also examined.
Collapse
Affiliation(s)
- Xavier Charmetant
- CIRI, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon I, Lyon, France
| | - Thomas Bachelet
- Clinique Saint-Augustin-CTMR, ELSAN, Bordeaux, France
- Department of Nephrology, Transplantation, Dialysis and Apheresis, Bordeaux University Hospital, Bordeaux, France
| | | | - Thierry Walzer
- CIRI, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon I, Lyon, France
| | - Olivier Thaunat
- CIRI, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon I, Lyon, France
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| |
Collapse
|
30
|
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
|
31
|
Krebs R, Tikkanen JM, Raissadati A, Hollmén M, Dhaygude K, Lemström KB. Inhibition of Vascular Endothelial Growth Factor Receptors 1 and 2 Attenuates Natural Killer Cell and Innate Immune Responses in an Experimental Model for Obliterative Bronchiolitis. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 192:254-269. [PMID: 34774518 DOI: 10.1016/j.ajpath.2021.10.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/30/2021] [Accepted: 10/25/2021] [Indexed: 01/02/2023]
Abstract
Obliterative bronchiolitis (OB) after lung transplantation is a nonreversible, life-threatening complication. We investigated the role of vascular endothelial growth factor receptor (VEGFR)-1 and -2 in the development of obliterative airway disease (OAD), an experimental model for OB. The nonimmunosuppressed recipients underwent transplantation with fully major histocompatibility complex mismatched heterotopic tracheal allografts and received VEGFR-1 and -2-specific monoclonal antibodies either alone or in combination or rat IgG as a control. The treatment with VEGFR-1- or -2-blocking antibody significantly decreased intragraft mRNA expression of natural killer cell activation markers early after transplantation. This was followed by reduced infiltration of CD11b+ cells and CD4+ T cells as well as down-regulated mRNA expression of proinflammatory chemokines and profibrotic growth factors. However, blocking of both VEGFR-1 and -2 was necessary to reduce luminal occlusion. Furthermore, concomitant inhibition of the calcineurin activation pathway almost totally abolished the development of OAD. This study proposes that blocking of VEGF receptors blunted natural killer cell and innate immune responses early after transplantation and attenuated the development of OAD. The results of this study suggest that further studies on the role of VEGFR-1 and -2 blocking in development of obliterative airway lesions might be rewarding.
Collapse
Affiliation(s)
- Rainer Krebs
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland.
| | - Jussi M Tikkanen
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland; Department of Cardiothoracic Surgery, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Alireza Raissadati
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland
| | - Maria Hollmén
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland
| | - Kishor Dhaygude
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland
| | - Karl B Lemström
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, Helsinki, Finland; Department of Cardiothoracic Surgery, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| |
Collapse
|
32
|
Siren EMJ, Luo HD, Tam F, Montgomery A, Enns W, Moon H, Sim L, Rey K, Guan Q, Wang JJ, Wardell CM, Monajemi M, Mojibian M, Levings MK, Zhang ZJ, Du C, Withers SG, Choy JC, Kizhakkedathu JN. Prevention of vascular-allograft rejection by protecting the endothelial glycocalyx with immunosuppressive polymers. Nat Biomed Eng 2021; 5:1202-1216. [PMID: 34373602 DOI: 10.1038/s41551-021-00777-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/30/2021] [Indexed: 02/07/2023]
Abstract
Systemic immunosuppression for the mitigation of immune rejection after organ transplantation causes adverse side effects and constrains the long-term benefits of the transplanted graft. Here we show that protecting the endothelial glycocalyx in vascular allografts via the enzymatic ligation of immunosuppressive glycopolymers under cold-storage conditions attenuates the acute and chronic rejection of the grafts after transplantation in the absence of systemic immunosuppression. In syngeneic and allogeneic mice that received kidney transplants, the steric and immunosuppressive properties of the ligated polymers largely protected the transplanted grafts from ischaemic reperfusion injury, and from immune-cell adhesion and thereby immunocytotoxicity. Polymer-mediated shielding of the endothelial glycocalyx following organ procurement should be compatible with clinical procedures for transplant preservation and perfusion, and may reduce the damage and rejection of transplanted organs after surgery.
Collapse
Affiliation(s)
- Erika M J Siren
- Centre for Blood Research, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada.,Department of Chemistry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Haiming D Luo
- Centre for Blood Research, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada.,Department of Chemistry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Franklin Tam
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Ashani Montgomery
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Winnie Enns
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Haisle Moon
- Centre for Blood Research, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Lyann Sim
- Department of Chemistry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Kevin Rey
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Qiunong Guan
- Department of Urologic Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Jiao-Jing Wang
- Comprehensive Transplant Center, Northwestern University, Chicago, IL, USA
| | - Christine M Wardell
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Surgery, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Mahdis Monajemi
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Surgery, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Majid Mojibian
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Surgery, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Megan K Levings
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Surgery, The University of British Columbia, Vancouver, British Columbia, Canada.,School of Biomedical Engineering, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Zheng J Zhang
- Comprehensive Transplant Center, Northwestern University, Chicago, IL, USA
| | - Caigan Du
- Department of Urologic Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephen G Withers
- Department of Chemistry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan C Choy
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.
| | - Jayachandran N Kizhakkedathu
- Centre for Blood Research, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada. .,Department of Chemistry, The University of British Columbia, Vancouver, British Columbia, Canada. .,Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada. .,School of Biomedical Engineering, The University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
33
|
Zografos CG, Chrysikos D, Pittaras T, Karampelias V, Chairakakis A, Galanos A, Sfiniadakis I, Felekouras E, Zografos GC, Sideris M, Papadopoulou K, Papalois AE. The Effects of Ascorbic Acid and U-74389G on Renal Ischemia-Reperfusion Injury in a Rat Model. In Vivo 2021; 34:2475-2484. [PMID: 32871775 DOI: 10.21873/invivo.12063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND/AIM U-74389G and ascorbic acid protect the cells from oxidation. This study aimed to depict their role in ischemia-reperfusion injury in a renal rat model. MATERIALS AND METHODS Sixty Wistars rats were randomized into six groups of 10 animals each. Group A Ischemia 30 min, reperfusion 60 min; Group B Ischemia 30 min, reperfusion 120 min; Group C Ischemia 30 min, ascorbic acid administration, reperfusion 60 min; Group D Ischemia 30 min, ascorbic acid administration, reperfusion 120 min; Group E Ischemia 30 min, U-74389G administration, reperfusion 60 min; Group F Ischemia 30 min, U-74389G administration, reperfusion 120 min. We then collected tissue and blood samples. RESULTS Histology and the significantly decreased malondialdehyde and tumor necrosis factor-α levels indicated that ascorbic acid was superior to U-74389G, at pre-defined time intervals. CONCLUSION Ascorbic acid and U-74389G ameliorated renal damage induced by ischemia-reperfusion injury, suggesting a therapeutic effect.
Collapse
Affiliation(s)
- Constantinos G Zografos
- Experimental, Educational and Research Center ELPEN, Athens, Greece.,1 Department of Propaedeutic Surgery, Hippokration Hospital, Athens, Greece
| | - Dimosthenis Chrysikos
- Experimental, Educational and Research Center ELPEN, Athens, Greece.,1 Department of Propaedeutic Surgery, Hippokration Hospital, Athens, Greece
| | - Theodoros Pittaras
- Experimental, Educational and Research Center ELPEN, Athens, Greece.,Hematology Laboratory - Blood Bank, National and Kapodistrian University of Athens School of Medicine, Aretaieion Hospital, Athens, Greece
| | | | | | - Antonis Galanos
- Experimental, Educational and Research Center ELPEN, Athens, Greece
| | | | - Evangelos Felekouras
- First Department of Surgery, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - George C Zografos
- 1 Department of Propaedeutic Surgery, Hippokration Hospital, Athens, Greece
| | - Michail Sideris
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | | | - Apostolos E Papalois
- Experimental, Educational and Research Center ELPEN, Athens, Greece .,School of Medicine, European University Cyprus, Nicosia, Cyprus
| |
Collapse
|
34
|
Hamada S, Dubois V, Koenig A, Thaunat O. Allograft recognition by recipient's natural killer cells: Molecular mechanisms and role in transplant rejection. HLA 2021; 98:191-199. [PMID: 34050618 DOI: 10.1111/tan.14332] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/24/2022]
Abstract
The current transplant immunology dogma defends that allograft rejection is initiated by recipient's adaptive immune system. In this prevalent model, innate immune cells in general, and natural killer (NK) cells in particular, are merely considered as downstream effectors which participate in the destruction of the graft only upon recruitment by adaptive effectors: alloreactive T cells or donor-specific antibodies (DSA). Challenging this vision, recent data demonstrated that recipients' NK cells are capable of a form of allorecognition because they can sense the absence of self HLA class I molecules on the surface of graft endothelial cells. Missing-self triggers mTORC1-dependent activation of NK cells, which in turn promote the development of graft microvascular inflammation and detrimentally impact graft survival. The fact that some patients develop chronic vascular rejection in absence of DSA or genetically-predicted missing self suggests that other molecular mechanisms could underly NK cell allorecognition. This review provides an overview of these proven and putative molecular mechanisms and discusses future research directions in this emerging field in organ transplant immunology.
Collapse
Affiliation(s)
- Sarah Hamada
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Valérie Dubois
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France.,HLA Laboratory, French National Blood Service (EFS), Décines-Charpieu, France
| | - Alice Koenig
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Olivier Thaunat
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| |
Collapse
|
35
|
Nieuwenhuijs-Moeke GJ, Bosch DJ, Leuvenink HG. Molecular Aspects of Volatile Anesthetic-Induced Organ Protection and Its Potential in Kidney Transplantation. Int J Mol Sci 2021; 22:ijms22052727. [PMID: 33800423 PMCID: PMC7962839 DOI: 10.3390/ijms22052727] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 12/16/2022] Open
Abstract
Ischemia reperfusion injury (IRI) is inevitable in kidney transplantation and negatively impacts graft and patient outcome. Reperfusion takes place in the recipient and most of the injury following ischemia and reperfusion occurs during this reperfusion phase; therefore, the intra-operative period seems an attractive window of opportunity to modulate IRI and improve short- and potentially long-term graft outcome. Commonly used volatile anesthetics such as sevoflurane and isoflurane have been shown to interfere with many of the pathophysiological processes involved in the injurious cascade of IRI. Therefore, volatile anesthetic (VA) agents might be the preferred anesthetics used during the transplantation procedure. This review highlights the molecular and cellular protective points of engagement of VA shown in in vitro studies and in vivo animal experiments, and the potential translation of these results to the clinical setting of kidney transplantation.
Collapse
Affiliation(s)
- Gertrude J. Nieuwenhuijs-Moeke
- Department of Anesthesiology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
- Correspondence: ; Tel.: +31-631623075
| | - Dirk J. Bosch
- Department of Anesthesiology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
| | - Henri G.D. Leuvenink
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
| |
Collapse
|
36
|
Calabrese DR, Aminian E, Mallavia B, Liu F, Cleary SJ, Aguilar OA, Wang P, Singer JP, Hays SR, Golden JA, Kukreja J, Dugger D, Nakamura M, Lanier LL, Looney MR, Greenland JR. Natural killer cells activated through NKG2D mediate lung ischemia-reperfusion injury. J Clin Invest 2021; 131:137047. [PMID: 33290276 PMCID: PMC7852842 DOI: 10.1172/jci137047] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022] Open
Abstract
Pulmonary ischemia-reperfusion injury (IRI) is a clinical syndrome of acute lung injury that occurs after lung transplantation or remote organ ischemia. IRI causes early mortality and has no effective therapies. While NK cells are innate lymphocytes capable of recognizing injured cells, their roles in acute lung injury are incompletely understood. Here, we demonstrated that NK cells were increased in frequency and cytotoxicity in 2 different IRI mouse models. We showed that NK cells trafficked to the lung tissue from peripheral reservoirs and were more mature within lung tissue. Acute lung ischemia-reperfusion injury was blunted in a NK cell-deficient mouse strain but restored with adoptive transfer of NK cells. Mechanistically, NK cell NKG2D receptor ligands were induced on lung endothelial and epithelial cells following IRI, and antibody-mediated NK cell depletion or NKG2D stress receptor blockade abrogated acute lung injury. In human lung tissue, NK cells were increased at sites of ischemia-reperfusion injury and activated NK cells were increased in prospectively collected human bronchoalveolar lavage in subjects with severe IRI. These data support a causal role for recipient peripheral NK cells in pulmonary IRI via NK cell NKG2D receptor ligation. Therapies targeting NK cells may hold promise in acute lung injury.
Collapse
Affiliation(s)
- Daniel R. Calabrese
- Department of Medicine, University of California, San Francisco, California
- Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Emily Aminian
- Department of Medicine, University of California, San Francisco, California
| | - Benat Mallavia
- Department of Medicine, University of California, San Francisco, California
| | - Fengchun Liu
- Department of Medicine, University of California, San Francisco, California
| | - Simon J. Cleary
- Department of Medicine, University of California, San Francisco, California
| | - Oscar A. Aguilar
- Department of Microbiology and Immunology, University of California, San Francisco, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Ping Wang
- Department of Medicine, University of California, San Francisco, California
| | - Jonathan P. Singer
- Department of Medicine, University of California, San Francisco, California
| | - Steven R. Hays
- Department of Medicine, University of California, San Francisco, California
| | - Jeffrey A. Golden
- Department of Medicine, University of California, San Francisco, California
| | - Jasleen Kukreja
- Department of Surgery, University of California, San Francisco, California
| | - Daniel Dugger
- Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Mary Nakamura
- Department of Medicine, University of California, San Francisco, California
- Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Lewis L. Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Mark R. Looney
- Department of Medicine, University of California, San Francisco, California
| | - John R. Greenland
- Department of Medicine, University of California, San Francisco, California
- Medical Service, Veterans Affairs Health Care System, San Francisco, California
| |
Collapse
|
37
|
Čepcová D, Kema IP, Sandovici M, Deelman LE, Šišková K, Klimas J, Vavrinec P, Vavrincová-Yaghi D. The protective effect of 1-methyltryptophan isomers in renal ischemia-reperfusion injury is not exclusively dependent on indolamine 2,3-dioxygenase inhibition. Biomed Pharmacother 2021; 135:111180. [PMID: 33433354 DOI: 10.1016/j.biopha.2020.111180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/14/2020] [Accepted: 12/26/2020] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Indolamine 2,3-dioxygenase (IDO), an enzyme that catalyses the metabolism of tryptophan, may play a detrimental role in ischemia-reperfusion injury (IRI). IDO can be inhibited by 1-methyl-tryptophan, which exists in a D (D-MT) or L (L-MT) isomer. These forms show different pharmacological effects besides IDO inhibition. Therefore, we sought to investigate whether these isomers can play a protective role in renal IRI, either IDO-dependent or independent. EXPERIMENTAL APPROACH We studied the effect of both isomers in a rat renal IRI model with a focus on IDO-dependent and independent effects. KEY RESULTS Both MT isomers reduced creatinine and BUN levels, with D-MT having a faster onset of action but shorter duration and L-MT a slower onset but longer duration (24 h and 48 h vs 48 h and 96 h reperfusion time). Interestingly, this effect was not exclusively dependent on IDO inhibition, but rather from decreased TLR4 signalling, mimicking changes in renal function. Additionally, L-MT increased the overall survival of rats. Moreover, both MT isomers interfered with TGF-β signalling and epithelial-mesenchymal transition. In order to study the effect of isomers in all mechanisms involved in IRI, a series of in vitro experiments was performed. The isomers affected signalling pathways in NK cells and tubular epithelial cells, as well as in dendritic cells and T cells. CONCLUSION AND IMPLICATIONS This study shows that both MT isomers have a renoprotective effect after ischemia-reperfusion injury, mostly independent of IDO inhibition, involving mutually different mechanisms. We bring novel findings in the pharmacological properties and mechanism of action of MT isomers, which could become a novel therapeutic target of renal IRI.
Collapse
Affiliation(s)
- Diana Čepcová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Slovak Republic.
| | - Ido P Kema
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Maria Sandovici
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Leo E Deelman
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Katarína Šišková
- Department of Cell and Molecular Biology of Drugs, Faculty of Pharmacy, Comenius University in Bratislava, Slovak Republic.
| | - Ján Klimas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Slovak Republic.
| | - Peter Vavrinec
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Slovak Republic.
| | - Diana Vavrincová-Yaghi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Slovak Republic.
| |
Collapse
|
38
|
Moloudizargari M, Govahi A, Fallah M, Rezvanfar MA, Asghari MH, Abdollahi M. The mechanisms of cellular crosstalk between mesenchymal stem cells and natural killer cells: Therapeutic implications. J Cell Physiol 2020; 236:2413-2429. [PMID: 32892356 DOI: 10.1002/jcp.30038] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/11/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) are mesenchymal precursors of various origins, with well-known immunomodulatory effects. Natural killer (NK) cells, the major cells of the innate immune system, are critical for the antitumor and antiviral defenses; however, in certain cases, they may be the main culprits in the pathogenesis of some NK-related conditions such as autoimmunities and hematological malignancies. On the other hand, these cells seem to be the major responders in beneficial phenomena like graft versus leukemia. Substantial data suggest that MSCs can variably affect NK cells and can be affected by these cells. Accordingly, acquiring a profound understanding of the crosstalk between MSCs and NK cells and the involved mechanisms seems to be a necessity to develop therapeutic approaches based on such interactions. Therefore, in this study, we made a thorough review of the existing literature on the interactions between MSCs and NK cells with a focus on the underlying mechanisms. The current knowledge herein suggests that MSCs possess a great potential to be used as tools for therapeutic targeting of NK cells in disease context and that preconditioning of MSCs, as well as their genetic manipulation before administration, may provide a wider variety of options in terms of eliciting more specific and desirable therapeutic outcomes. Nevertheless, our knowledge regarding the effects of MSCs on NK cells is still in its infancy, and further studies with well-defined conditions are warranted herein.
Collapse
Affiliation(s)
- Milad Moloudizargari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Govahi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marjan Fallah
- Department of Pharmacology and Toxicology, Medicinal Plant Research Centre, Faculty of Pharmacy, Islamic Azad University, Amol, Iran
| | - Mohammad A Rezvanfar
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), and Department of Toxicology and Pharmacology, School of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad H Asghari
- Department of Pharmacology and Toxicology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), and Department of Toxicology and Pharmacology, School of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
39
|
Cantoni C, Granata S, Bruschi M, Spaggiari GM, Candiano G, Zaza G. Recent Advances in the Role of Natural Killer Cells in Acute Kidney Injury. Front Immunol 2020; 11:1484. [PMID: 32903887 PMCID: PMC7438947 DOI: 10.3389/fimmu.2020.01484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/08/2020] [Indexed: 01/18/2023] Open
Abstract
Growing evidence is revealing a central role for natural killer (NK) cells, cytotoxic cells belonging to the broad family of innate lymphoid cells (ILCs), in acute and chronic forms of renal disease. NK cell effector functions include both the recognition and elimination of virus-infected and tumor cells and the capability of sensing pathogens through Toll-like receptor (TLR) engagement. Notably, they also display immune regulatory properties, exerted thanks to their ability to secrete cytokines/chemokines and to establish interactions with different innate and adaptive immune cells. Therefore, because of their multiple functions, NK cells may have a major pathogenic role in acute kidney injury (AKI), and a better understanding of the molecular mechanisms driving NK cell activation in AKI and their downstream interactions with intrinsic renal cells and infiltrating immune cells could help to identify new potential biomarkers and to select clinically valuable novel therapeutic targets. In this review, we discuss the current literature regarding the potential involvement of NK cells in AKI.
Collapse
Affiliation(s)
- Claudia Cantoni
- Laboratory of Clinical and Experimental Immunology, Integrated Department of Services and Laboratories, IRCCS Istituto Giannina Gaslini, Genoa, Italy.,Department of Experimental Medicine (DIMES) and Center of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Simona Granata
- Renal Unit, Department of Medicine, University-Hospital of Verona, Verona, Italy
| | - Maurizio Bruschi
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Grazia Maria Spaggiari
- Department of Experimental Medicine (DIMES) and Center of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Giovanni Candiano
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University-Hospital of Verona, Verona, Italy
| |
Collapse
|
40
|
Dellepiane S, Leventhal JS, Cravedi P. T Cells and Acute Kidney Injury: A Two-Way Relationship. Front Immunol 2020; 11:1546. [PMID: 32765535 PMCID: PMC7379378 DOI: 10.3389/fimmu.2020.01546] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/11/2020] [Indexed: 12/29/2022] Open
Abstract
Acute Kidney Injury (AKI) complicates up to 10% of hospital admissions substantially increasing patient morbidity and mortality. Experimental evidence supports that AKI initiation and maintenance results from immune-mediated damage. Exogenous injury sources directly damage renal cells which produce pro-inflammatory mediators recruiting immune cells and furthering kidney injury. Many AKI studies focus on activation of innate immunity; major components include complement pathways, neutrophils, and monocytes. Recently, growing evidence emphasizes T lymphocytes role in affecting AKI pathogenesis and magnitude. In particular, T helper 17 lymphocytes enhance tissue injury by recruiting neutrophils and other inflammatory cells, while regulatory T cells conversely reduce renal injury and facilitate repair. Intriguingly, evidence supports local parenchymal-T cell interactions as essential to producing T cell phenotypic changes affecting long-term kidney and patient survival. Herein, we review T cells effects on AKI and patient outcomes and discuss related new therapeutic approaches to improve outcomes of affected individuals.
Collapse
Affiliation(s)
- Sergio Dellepiane
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jeremy S Leventhal
- Division of Nephrology, White Plains Hospital, White Plains, NY, United States
| | - Paolo Cravedi
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
41
|
Callemeyn J, Lerut E, de Loor H, Arijs I, Thaunat O, Koenig A, Meas-Yedid V, Olivo-Marin JC, Halloran P, Chang J, Thorrez L, Kuypers D, Sprangers B, Van Lommel L, Schuit F, Essig M, Gwinner W, Anglicheau D, Marquet P, Naesens M. Transcriptional Changes in Kidney Allografts with Histology of Antibody-Mediated Rejection without Anti-HLA Donor-Specific Antibodies. J Am Soc Nephrol 2020; 31:2168-2183. [PMID: 32641395 DOI: 10.1681/asn.2020030306] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 04/30/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Circulating donor-specific anti-HLA antibodies (HLA-DSAs) are often absent in serum of kidney allograft recipients whose biopsy specimens demonstrate histology of antibody-mediated rejection (ABMR). It is unclear whether cases involving ABMR histology without detectable HLA-DSAs represent a distinct clinical and molecular phenotype. METHODS In this multicenter cohort study, we integrated allograft microarray analysis with extensive clinical and histologic phenotyping from 224 kidney transplant recipients between 2011 and 2017. We used the term ABMR histology for biopsy specimens that fulfill the first two Banff 2017 criteria for ABMR, irrespective of HLA-DSA status. RESULTS Of 224 biopsy specimens, 56 had ABMR histology; 26 of these (46.4%) lacked detectable serum HLA-DSAs. Biopsy specimens with ABMR histology showed overexpression of transcripts mostly related to IFNγ-induced pathways and activation of natural killer cells and endothelial cells. HLA-DSA-positive and HLA-DSA-negative biopsy specimens with ABMR histology displayed similar upregulation of pathways and enrichment of infiltrating leukocytes. Transcriptional heterogeneity observed in biopsy specimens with ABMR histology was not associated with HLA-DSA status but was caused by concomitant T cell-mediated rejection. Compared with cases lacking ABMR histology, those with ABMR histology and HLA-DSA had higher allograft failure risk (hazard ratio [HR], 7.24; 95% confidence interval [95% CI], 3.04 to 17.20) than cases without HLA-DSA (HR, 2.33; 95% CI, 0.85 to 6.33), despite the absence of transcriptional differences. CONCLUSIONS ABMR histology corresponds to a robust intragraft transcriptional signature, irrespective of HLA-DSA status. Outcome after ABMR histology is not solely determined by the histomolecular presentation but is predicted by the underlying etiologic factor. It is important to consider this heterogeneity in further research and in treatment decisions for patients with ABMR histology.
Collapse
Affiliation(s)
- Jasper Callemeyn
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium.,Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Evelyne Lerut
- Department of Morphology and Molecular Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Henriette de Loor
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Ingrid Arijs
- Department of Human Genetics, Laboratory of Translational Genetics, KU Leuven, Leuven, Belgium.,Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Olivier Thaunat
- Center for Research in Infectious Diseases, Institut National de la Santé et de la Recherche Médicale (INSERM) U1111, Claude Bernard University Lyon I, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University Lyon I, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Alice Koenig
- Center for Research in Infectious Diseases, Institut National de la Santé et de la Recherche Médicale (INSERM) U1111, Claude Bernard University Lyon I, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University Lyon I, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Vannary Meas-Yedid
- Biological Image Analysis Unit, Pasteur Institute, CNRS Unité de Recherche Associée (URA) 2582, Paris, France
| | - Jean-Christophe Olivo-Marin
- Biological Image Analysis Unit, Pasteur Institute, CNRS Unité de Recherche Associée (URA) 2582, Paris, France
| | - Philip Halloran
- Division of Nephrology and Transplant Immunology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Jessica Chang
- Division of Nephrology and Transplant Immunology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Lieven Thorrez
- Department of Development and Regeneration, KU Leuven, Kortrijk, Belgium
| | - Dirk Kuypers
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium.,Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Ben Sprangers
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute, KU Leuven, Leuven, Belgium
| | - Leentje Van Lommel
- Department of Cellular and Molecular Medicine, Gene Expression Unit, KU Leuven, Leuven, Belgium
| | - Frans Schuit
- Department of Cellular and Molecular Medicine, Gene Expression Unit, KU Leuven, Leuven, Belgium
| | - Marie Essig
- Department of Nephrology, Dialysis and Transplantation, University of Limoges, Limoges, France
| | - Wilfried Gwinner
- Department of Nephrology and Internal Medicine, Hannover Medical School, Hannover, Germany
| | - Dany Anglicheau
- Paris Descartes University, Sorbonne Paris Cité University, Paris, France.,INSERM U1151, Paris, France.,Department of Nephrology and Kidney Transplantation, Necker Hospital, Assistance Publique--Hôpitaux de Paris, Paris, France
| | - Pierre Marquet
- INSERM U1248, Limoges, France.,Department of Pharmacology and Toxicology, University Hospitals Limoges, Limoges, France
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium .,Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
42
|
Dufour L, Ferhat M, Robin A, Inal S, Favreau F, Goujon JM, Hauet T, Gombert JM, Herbelin A, Thierry A. [Ischemia-reperfusion injury after kidney transplantation]. Nephrol Ther 2020; 16:388-399. [PMID: 32571740 DOI: 10.1016/j.nephro.2020.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Ischemia-reperfusion injury is an inescapable phenomenon in kidney transplantation. It combines lesional processes of biochemical origin associated with oxydative stress and of immunological origin in connection with the recruitment and activation of innate immunity cells. Histological lesions associate acute tubular necrosis and interstitial œdema, which can progress to interstitial fibrosis. The extent of these lesions depends on donor characteristics (age, expanded criteria donor, etc.) and cold ischemia time. In the short term, ischemia-reperfusion results in delayed recovery of graft function. Cold ischemia time also impacts long-term graft survival. Preclinical models, such as murine and porcine models, have furthered understanding of the pathophysiological mechanisms of ischemia-reperfusion injury. Due to its renal anatomical proximity to humans, the porcine model is relevant to assessment of the molecules administered to a donor or recipient, and also of additives to preservation solutions. Different donor resuscitation and graft perfusion strategies can be studied. In humans, prevention of ischemia-reperfusion injury is a research subject as concerns donor conditioning, additive molecules in preservation solutions, graft reperfusion modalities and choice of the molecules administered to the recipient. Pending significant advances in research, the goal is to achieve the shortest possible cold ischemia time.
Collapse
Affiliation(s)
- Léa Dufour
- Service de néphrologie-hémodialyse-transplantation rénale, CHU de Poitiers, 2, rue de la Milétrie, 86021 Poitiers cedex, France
| | - Maroua Ferhat
- Inserm, U1082 laboratoire Irtomit, 2, rue de la Milétrie, 86021 Poitiers cedex, France
| | - Aurélie Robin
- Inserm, U1082 laboratoire Irtomit, 2, rue de la Milétrie, 86021 Poitiers cedex, France
| | - Sofiane Inal
- Inserm, U1082 laboratoire Irtomit, 2, rue de la Milétrie, 86021 Poitiers cedex, France; Service de biochimie, CHU de Poitiers, 2, rue de la Milétrie, 86021 Poitiers cedex, France
| | - Frédéric Favreau
- Inserm, U1082 laboratoire Irtomit, 2, rue de la Milétrie, 86021 Poitiers cedex, France
| | - Jean-Michel Goujon
- Service d'anatomopathologie, CHU de Poitiers, 2, rue de la Milétrie, 86021 Poitiers cedex, France
| | - Thierry Hauet
- Inserm, U1082 laboratoire Irtomit, 2, rue de la Milétrie, 86021 Poitiers cedex, France; Service de biochimie, CHU de Poitiers, 2, rue de la Milétrie, 86021 Poitiers cedex, France; Fédération hospitalo-universitaire de transplantation Survival Optimization in Organ Transplantation (Support) Tours Poitiers Limoges, CHU de Poitiers, 2, rue de la Milétrie, 86021 Poitiers cedex, France; Plateforme Infrastructures en biologie, santé et agronomie (Ibisa) Modélisation préclinique - innovation chirurgicale et technologique (Mopict), 86000 Poitiers cedex, France
| | - Jean-Marc Gombert
- Inserm, U1082 laboratoire Irtomit, 2, rue de la Milétrie, 86021 Poitiers cedex, France; Service d'immunologie, CHU de Poitiers, 2, rue de la Milétrie, 86021 Poitiers cedex, France
| | - André Herbelin
- Inserm, U1082 laboratoire Irtomit, 2, rue de la Milétrie, 86021 Poitiers cedex, France
| | - Antoine Thierry
- Service de néphrologie-hémodialyse-transplantation rénale, CHU de Poitiers, 2, rue de la Milétrie, 86021 Poitiers cedex, France; Inserm, U1082 laboratoire Irtomit, 2, rue de la Milétrie, 86021 Poitiers cedex, France; Fédération hospitalo-universitaire de transplantation Survival Optimization in Organ Transplantation (Support) Tours Poitiers Limoges, CHU de Poitiers, 2, rue de la Milétrie, 86021 Poitiers cedex, France.
| |
Collapse
|
43
|
Tang PCT, Zhang YY, Chan MKK, Lam WWY, Chung JYF, Kang W, To KF, Lan HY, Tang PMK. The Emerging Role of Innate Immunity in Chronic Kidney Diseases. Int J Mol Sci 2020; 21:ijms21114018. [PMID: 32512831 PMCID: PMC7312694 DOI: 10.3390/ijms21114018] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/28/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
Renal fibrosis is a common fate of chronic kidney diseases. Emerging studies suggest that unsolved inflammation will progressively transit into tissue fibrosis that finally results in an irreversible end-stage renal disease (ESRD). Renal inflammation recruits and activates immunocytes, which largely promotes tissue scarring of the diseased kidney. Importantly, studies have suggested a crucial role of innate immunity in the pathologic basis of kidney diseases. This review provides an update of both clinical and experimental information, focused on how innate immune signaling contributes to renal fibrogenesis. A better understanding of the underlying mechanisms may uncover a novel therapeutic strategy for ESRD.
Collapse
Affiliation(s)
- Philip Chiu-Tsun Tang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China; (P.C.-T.T.); (M.K.-K.C.); (J.Y.-F.C.); (W.W.-Y.L.); (W.K.); (K.-F.T.)
| | - Ying-Ying Zhang
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China;
| | - Max Kam-Kwan Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China; (P.C.-T.T.); (M.K.-K.C.); (J.Y.-F.C.); (W.W.-Y.L.); (W.K.); (K.-F.T.)
| | - Winson Wing-Yin Lam
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China; (P.C.-T.T.); (M.K.-K.C.); (J.Y.-F.C.); (W.W.-Y.L.); (W.K.); (K.-F.T.)
| | - Jeff Yat-Fai Chung
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China; (P.C.-T.T.); (M.K.-K.C.); (J.Y.-F.C.); (W.W.-Y.L.); (W.K.); (K.-F.T.)
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China; (P.C.-T.T.); (M.K.-K.C.); (J.Y.-F.C.); (W.W.-Y.L.); (W.K.); (K.-F.T.)
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China; (P.C.-T.T.); (M.K.-K.C.); (J.Y.-F.C.); (W.W.-Y.L.); (W.K.); (K.-F.T.)
| | - Hui-Yao Lan
- Li Ka Shing Institute of Health Sciences, and Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong 999077, China;
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China; (P.C.-T.T.); (M.K.-K.C.); (J.Y.-F.C.); (W.W.-Y.L.); (W.K.); (K.-F.T.)
- Correspondence:
| |
Collapse
|
44
|
Zhao Q, Yan T, Chopp M, Venkat P, Chen J. Brain-kidney interaction: Renal dysfunction following ischemic stroke. J Cereb Blood Flow Metab 2020; 40:246-262. [PMID: 31766979 PMCID: PMC7370616 DOI: 10.1177/0271678x19890931] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Stroke is a leading cause of mortality and morbidity, with long-term debilitating effects. Accumulating evidence from experimental studies as well as observational studies in patients suggests a cross talk between the brain and kidney after stroke. Stroke may lead to kidney dysfunction which can adversely impact patient outcome. In this review article, we discuss the epidemiology and mechanisms of brain–kidney interaction following ischemic stroke. Specifically, we discuss the role of the central autonomic network, autoregulation, inflammatory and immune responses, the role of extracellular vesicles and their cargo microRNA, in mediating brain–kidney interaction following stroke. Understanding the bidirectional nature of interaction between the brain and kidney after cerebral injury would have clinical implications for the treatment of stroke and overall patient outcome.
Collapse
Affiliation(s)
- Qiang Zhao
- Tianjin Neurological Institute, Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Yan
- Tianjin Neurological Institute, Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.,Department of Physics, Oakland University, Rochester, MI, USA
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
45
|
Güvenç M, Cellat M, Uyar A, Özkan H, Gokcek İ, İsler CT, Yakan A. Nobiletin Protects from Renal Ischemia-Reperfusion Injury in Rats by Suppressing Inflammatory Cytokines and Regulating iNOS-eNOS Expressions. Inflammation 2020; 43:336-346. [PMID: 31705353 DOI: 10.1007/s10753-019-01123-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Ischemia-reperfusion injury is an organ failure caused by hypoxia and reperfusion, which is closely associated with oxidative stress and inflammation. In this study, we investigated whether nobiletin had protective effects on inflammatory parameters, oxidative damage, iNOS-eNOS expressions, and histopathological structure of renal tissue in rats with renal ischemia-reperfusion injury. For this purpose, 24 rats were divided into 4 groups: group 1 (Control), group 2 (Ischemia-Reperfusion-IR), group 3 (Nobiletin-10 mg/kg p.o.), group 4 (Nobiletin + IR). The study was continued for 7 days. At the end of the study, urea (p < 0.05), creatine (p < 0.05), MDA (p < 0.001), TNF-alpha (p < 0.001), IL-1 beta (p < 0.05), and IL-6 (p < 0.001) levels increased in the IR group; however, a significant decrease occurred in group 4 (Nobiletin + IR) and it reached the control group levels. In the IR group, GSH (p < 0.01) levels, and GSH.Px (p < 0.01) and CAT (p < 0.05) activities decreased whereas they increased significantly in group 4 (Nobiletin + IR) and reached the same levels as the control group. In histopathological analyses, destruction and increased iNOS-eNOS expressions in the IR group showed a significant decrease in group 4 (Nobiletin + IR). As a result, the application of nobiletin has shown that it has protective effects by reducing kidney damage caused by IR injury.
Collapse
Affiliation(s)
- Mehmet Güvenç
- Mustafa Kemal University, Faculty of Veterinary Medicine, Department of Physiology, Antakya, Turkey.
| | - Mustafa Cellat
- Mustafa Kemal University, Faculty of Veterinary Medicine, Department of Physiology, Antakya, Turkey
| | - Ahmet Uyar
- Mustafa Kemal University, Faculty of Veterinary Medicine, Department of Pathology, Antakya, Turkey
| | - Hüseyin Özkan
- Mustafa Kemal University, Faculty of Veterinary Medicine, Department of Genetics, Antakya, Turkey
| | - İshak Gokcek
- Mustafa Kemal University, Faculty of Veterinary Medicine, Department of Physiology, Antakya, Turkey
| | - Cafer Tayer İsler
- Mustafa Kemal University, Faculty of Veterinary Medicine, Department Surgery, Antakya, Turkey
| | - Akın Yakan
- Mustafa Kemal University, Faculty of Veterinary Medicine, Department of Genetics, Antakya, Turkey
| |
Collapse
|
46
|
Cao Q, Wang R, Wang Y, Niu Z, Chen T, Wang C, Jin L, Huang Q, Li Q, Wang XM, Azmi F, Lee VWS, Wang YM, Zheng G, Alexander SI, Harris DCH. Regulatory innate lymphoid cells suppress innate immunity and reduce renal ischemia/reperfusion injury. Kidney Int 2020; 97:130-142. [PMID: 31685310 DOI: 10.1016/j.kint.2019.07.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 07/10/2019] [Accepted: 07/11/2019] [Indexed: 12/14/2022]
Abstract
Innate lymphoid cells are a recently recognized group of immune cells with critical roles in tissue homeostasis and inflammation. Regulatory innate lymphoid cells are a newly identified subset of innate lymphoid cells, which play a suppressive role in the innate immune response, favoring the resolution of intestinal inflammation. However, the expression and role of regulatory innate lymphoid cells in kidney has not been reported. Here, we show that regulatory innate lymphoid cells are present in both human and mouse kidney, express similar surface markers and form a similar proportion of total kidney innate lymphoid cells. Regulatory innate lymphoid cells from kidney were expanded in vitro with a combination of IL-2, IL-7 and transforming growth factor-β. These cells exhibited immunosuppressive effects on innate immune cells via secretion of IL-10 and transforming growth factor-β. Moreover, treatment with IL-2/IL-2 antibody complexes (IL-2C) promoted expansion of regulatory innate lymphoid cells in vivo, and prevent renal ischemia/reperfusion injury in Rag-/- mice that lack adaptive immune cells including Tregs. Depletion of regulatory innate lymphoid cells with anti-CD25 antibody abolished the beneficial effects of IL-2C in the Rag-/- mice. Adoptive transfer of ex vivo expanded regulatory innate lymphoid cells improved renal function and attenuated histologic damage when given before or after induction of ischemia/reperfusion injury in association with reduction of neutrophil infiltration and induction of reparative M2 macrophages in kidney. Thus, our study shows that regulatory innate lymphoid cells suppress innate renal inflammation and ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Qi Cao
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Henan, China.
| | - Ruifeng Wang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Nephrology, The Second Hospital of Anhui Medical University, Anhui, China
| | - Yiping Wang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Zhiguo Niu
- Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Henan, China
| | - Titi Chen
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Chengshi Wang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Li Jin
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Qingsong Huang
- Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Henan, China
| | - Qing Li
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Xin Maggie Wang
- Flow Cytometry Facility, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Farhana Azmi
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Vincent W S Lee
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Yuan Min Wang
- Centre for Kidney Research, Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Guoping Zheng
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Stephen I Alexander
- Centre for Kidney Research, Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - David C H Harris
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
47
|
Flüh C, Mafael V, Adamski V, Synowitz M, Held-Feindt J. Dormancy and NKG2D system in brain metastases: Analysis of immunogenicity. Int J Mol Med 2019; 45:298-314. [PMID: 31894267 PMCID: PMC6984787 DOI: 10.3892/ijmm.2019.4449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/19/2019] [Indexed: 12/18/2022] Open
Abstract
Patients with breast cancer (BC) and lung cancer (LC) are prone to developing brain metastases, which are associated with devastating prognoses. Dormant tumor cells, a population of non-apoptotic quiescent cells and immunological escape mechanisms, including the Natural Killer Group 2 member D (NKG2D) receptor-ligand system, represent potential mechanisms of tumor recurrence. To date, the immunological characteristics of dormant tumor cells concerning the NKG2D system in cerebral malignancies are mostly unknown. In the present study, an extensive characterization of dormant and NKG2D ligand (NKG2DL)+ cells in cerebral metastases was performed. The expression profiles and localization patterns of various NKG2DL and several dormancy markers were analyzed in solid human brain metastases from patients with BC and LC using immunostaining and reverse transcription-quantitative polymerase chain reaction analyses. Statistical analysis was performed using Student's t-test and Bravais-Pearson correlation analysis. Not only 'peripheral', but also 'central' dormancy markers, which had been previously described in primary brain tumors, were identified in all cerebral metastases at detectable levels at protein and mRNA levels. Notably, the majority of NKG2DL+ cells were also positive for 'central' dormancy markers, but not 'peripheral' dormancy markers in both patient groups. This cell population may represent a promising future therapeutic target.
Collapse
Affiliation(s)
- Charlotte Flüh
- Department of Neurosurgery, University Medical Center Schleswig‑Holstein, Campus Kiel, D‑24105 Kiel, Germany
| | - Victor Mafael
- Department of Neurosurgery, University Medical Center Schleswig‑Holstein, Campus Kiel, D‑24105 Kiel, Germany
| | - Vivian Adamski
- Department of Neurosurgery, University Medical Center Schleswig‑Holstein, Campus Kiel, D‑24105 Kiel, Germany
| | - Michael Synowitz
- Department of Neurosurgery, University Medical Center Schleswig‑Holstein, Campus Kiel, D‑24105 Kiel, Germany
| | - Janka Held-Feindt
- Department of Neurosurgery, University Medical Center Schleswig‑Holstein, Campus Kiel, D‑24105 Kiel, Germany
| |
Collapse
|
48
|
Ashraf MI, Sarwar A, Kühl AA, Hunger E, Sattler A, Aigner F, Regele H, Sauter M, Klingel K, Schneeberger S, Resch T, Kotsch K. Natural Killer Cells Promote Kidney Graft Rejection Independently of Cyclosporine A Therapy. Front Immunol 2019; 10:2279. [PMID: 31616441 PMCID: PMC6769038 DOI: 10.3389/fimmu.2019.02279] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 09/09/2019] [Indexed: 01/02/2023] Open
Abstract
Natural Killer (NK) cells have recently been recognized as key players in antibody-mediated chronic allograft failure, thus requiring a comprehensive understanding whether NK cells can escape conventional immunosuppressive regimens. Influence of cyclosporine A (CyA) on NK cell function was studied in a mouse model of allogeneic kidney transplantation (KTX, BALB/c to C57BL/6). Recipients were treated daily with CyA (10 mg/kg) for seven or 14 days for long term survival (day 56). Administration of CyA in recipients resulted in significantly reduced frequencies of intragraft and splenic CD8+ T cells, whereas the latter illustrated reduced IFNγ production. In contrast, intragraft and splenic NK cell frequencies remained unaffected in CyA recipients and IFNγ production and degranulation of NK cells were not reduced as compared with controls. Depletion of NK cells in combination with CyA resulted in an improvement in kidney function until day 7 and prolonged graft survival until day 56 as compared to untreated controls. Surviving animals demonstrated higher intragraft frequencies of proliferating CD4+FoxP3+Ki67+ regulatory T (TREG) cells as well as higher frequencies of CD8+CD122+ TREG. We here demonstrate that NK cell depletion combined with CyA synergistically improves graft function and prolongs graft survival, suggesting that NK cell targeting constitutes a novel approach for improving KTX outcomes.
Collapse
Affiliation(s)
| | - Attia Sarwar
- Department of General, Visceral and Vascular Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anja A Kühl
- iPath.Berlin-Immunopathology for Experimental Models, Berlin Institute of Health (BIH), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Elena Hunger
- Department of General, Visceral and Vascular Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Arne Sattler
- Department of General, Visceral and Vascular Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Felix Aigner
- Department of Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Heinz Regele
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Martina Sauter
- Department of Molecular Pathology, Tübingen University Hospital, Tübingen, Germany
| | - Karin Klingel
- Department of Molecular Pathology, Tübingen University Hospital, Tübingen, Germany
| | - Stefan Schneeberger
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Resch
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Katja Kotsch
- Department of General, Visceral and Vascular Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
49
|
Gameiro J, Lopes JA. Complete blood count in acute kidney injury prediction: a narrative review. Ann Intensive Care 2019; 9:87. [PMID: 31388845 PMCID: PMC6684666 DOI: 10.1186/s13613-019-0561-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/30/2019] [Indexed: 02/08/2023] Open
Abstract
Acute kidney injury (AKI) is a complex syndrome defined by a decrease in renal function. The incidence of AKI has raised in the past decades, and it is associated with negative impact in patient outcomes in the short and long term. Considering the impact of AKI on patient prognosis, research has focused on methods to assess patients at risk for developing AKI, diagnose subclinical AKI, and on prevention and treatment strategies, for which it is crucial an understanding of pathophysiology the of AKI. In this review, we discuss the use of easily available parameters found in a complete blood count to detect patients at risk for developing AKI, to provide an early diagnosis of AKI, and to predict associated patient outcomes.
Collapse
Affiliation(s)
- Joana Gameiro
- Division of Nephrology and Renal Transplantation, Department of Medicine, Centro Hospitalar Lisboa Norte, EPE, Av. Prof. Egas Moniz, 1649-035, Lisbon, Portugal.
| | - José António Lopes
- Division of Nephrology and Renal Transplantation, Department of Medicine, Centro Hospitalar Lisboa Norte, EPE, Av. Prof. Egas Moniz, 1649-035, Lisbon, Portugal
| |
Collapse
|
50
|
Sung B, Su Y, Jiang J, Mcleod P, Liu W, Haig A, Green DR, Zhang ZX, Jevnikar AM. Loss of receptor interacting protein kinases 3 and caspase-8 augments intrinsic apoptosis in tubular epithelial cell and promote kidney ischaemia-reperfusion injury. Nephrology (Carlton) 2019; 24:661-669. [PMID: 30175514 PMCID: PMC7706480 DOI: 10.1111/nep.13487] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Ischaemia-reperfusion injury (IRI) is associated with programmed cell death that promotes inflammation and organ dysfunction. Necroptosis is mediated by members of receptor interacting protein kinases (RIPK1/3). Inhibition of RIPK1/3 provides a pro-survival benefit in kidney IRI. Caspase-8 initiates apoptosis and contributes to IRI. We studied whether inhibiting both RIPK3 and caspase-8 would provide an additional benefit in kidney IRI. METHODS A clamp was applied to the left kidney pedicle for 45 min followed by right kidney nephrectomy. Kidney and serum from wild type, RIPK3-/- , and RIPK3-/- caspase-8-/- double knockout (DKO) mice were collected post-IRI for assessment of injury. Tubular epithelial cells (TEC) isolated from wild type, RIPK3-/- , and DKO mice were treated with interferons-γ and interleukin-1β to induce apoptotic death. RESULTS Kidney IRI of DKO mice did not show improvement over RIPK3-/- mice. We have found that DKO triggered 'intrinsic' apoptosis in TEC in response to interleukin-1β and interferons-γ. Up-regulation of the B-cell lymphoma 2 (Bcl-2)-associated death promoter, the Bcl-2-homologous antagonist killer and Bcl-2-associated X protein and enhanced activation of caspase-3 and 9 were found in DKO TEC. TEC infected with Murine cytomegalovirus that encodes multiple cell death inhibitors resist to death. CONCLUSION We show that the deletion of both RIPK3 and caspase-8 does not provide additive benefit in IRI or TEC death and may enhance injury by up-regulation of intrinsic apoptosis. This suggests blocking multiple death pathways may be required for the prevention of kidney IRI clinically.
Collapse
Affiliation(s)
- Baekjun Sung
- Matthew Mailing Centre for Translational Transplantation Studies
- Multi-Organ Transplant program, London Health Science Centre, London, Canada
- Department of Microbiology and Immunology, Western University, London, Canada
| | - Ye Su
- Matthew Mailing Centre for Translational Transplantation Studies
| | - Jifu Jiang
- Matthew Mailing Centre for Translational Transplantation Studies
- Multi-Organ Transplant program, London Health Science Centre, London, Canada
| | - Patrick Mcleod
- Matthew Mailing Centre for Translational Transplantation Studies
| | - Weihua Liu
- Department of Pathology, Western University, London, Canada
| | - Aaron Haig
- Department of Pathology, Western University, London, Canada
| | - Douglas R. Green
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Zhu-Xu Zhang
- Matthew Mailing Centre for Translational Transplantation Studies
- Multi-Organ Transplant program, London Health Science Centre, London, Canada
- Department of Pathology, Western University, London, Canada
- Division of Nephrology, Department of Medicine, Western University, London, Ontario, Canada
| | - Anthony M. Jevnikar
- Matthew Mailing Centre for Translational Transplantation Studies
- Multi-Organ Transplant program, London Health Science Centre, London, Canada
- Department of Microbiology and Immunology, Western University, London, Canada
- Division of Nephrology, Department of Medicine, Western University, London, Ontario, Canada
| |
Collapse
|