Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
JHEP Rep ; 5(10): 100837, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37691688

ABSTRACT

Background and Aims: Although type 2 innate lymphoid cells (ILC2s) were originally found to be liver-resident lymphocytes, the role and importance of ILC2 in liver injury remains poorly understood. In the current study, we sought to determine whether ILC2 is an important regulator of hepatic ischaemia/reperfusion injury (IRI). Methods: ILC2-deficient mice (ICOS-T or NSG) and genetically modified ILC2s were used to investigate the role of ILC2s in murine hepatic IRI. Interactions between ILC2s and eosinophils or macrophages were studied in coculture. The role of human ILC2s was assessed in an immunocompromised mouse model of hepatic IRI. Results: Administration of IL-33 prevented hepatic IRI in association with reduction of neutrophil infiltration and inflammatory mediators in the liver. IL-33-treated mice had elevated numbers of ILC2s, eosinophils, and regulatory T cells. Eosinophils, but not regulatory T cells, were required for IL-33-mediated hepatoprotection in IRI mice. Depletion of ILC2s substantially abolished the protective effect of IL-33 in hepatic IRI, indicating that ILC2s play critical roles in IL-33-mediated liver protection. Adoptive transfer of ex vivo-expanded ILC2s improved liver function and attenuated histologic damage in mice subjected to IRI. Mechanistic studies combining genetic and adoptive transfer approaches identified a protective role of ILC2s through promoting IL-13-dependent induction of anti-inflammatory macrophages and IL-5-dependent elevation of eosinophils in IRI. Furthermore, in vivo expansion of human ILC2s by IL-33 or transfer of ex vivo-expanded human ILC2s ameliorated hepatic IRI in an immunocompromised mouse model of hepatic IRI. Conclusions: This study provides insight into the mechanisms of ILC2-mediated liver protection that could serve as therapeutic targets to treat acute liver injury. Impact and Implications: We report that type 2 innate lymphoid cells (ILC2s) are important regulators in a mouse model of liver ischaemia/reperfusion injury (IRI). Through manipulation of macrophage and eosinophil phenotypes, ILC2s mitigate liver inflammation and injury during liver IRI. We propose that ILC2s have the potential to serve as a therapeutic tool for protecting against acute liver injury and lay the foundation for translation of ILC2 therapy to human liver disease.

2.
Kidney Int Rep ; 6(3): 727-736, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33732987

ABSTRACT

INTRODUCTION: Individuals aged ≥65 years are increasingly prevalent on the waitlist for kidney transplantation, yet evidence on recipient and donor factors that define optimal outcomes in elderly patients after kidney transplantation is scarce. METHODS: We used multivariable Cox regression modeling to determine the factors associated with all-cause death, death with a functioning graft, and overall and death-censored graft survival, using data from the Australia and New Zealand Dialysis and Transplant (ANZDATA) registry. RESULTS: A total of 802 kidney transplant recipients aged ≥65 years underwent their first transplantation between June 2006 and December 2016. Median age at transplantation was 68 years (interquartile range = 66-69 years). The 1-year and 5-year overall patient and graft survivals (95% confidence interval [CI]) were 95.1 (93.5-96.7) and 79.0 (75.1-82.9), and 92.9 (91.1-94.7) and 75.4 (71.3-79.5), respectively. Factors associated with higher risks of all-cause death included prevalent coronary artery disease (adjusted hazard ratio [95% confidence interval] = 1.47 [1.03-2.11]), cerebrovascular disease (1.99 [1.26-3.16]), increasing graft ischemic time (1.06 per hour [1.03-1.09]), donor age (1.02 per year [1.01-1.03]), delayed graft function (1.64 [1.13-2.39]), and peritoneal dialysis pretransplantation (1.71 [1.17-2.51]). CONCLUSION: Prevalent vascular disease and peritoneal dialysis as a pretransplantation dialysis modality are risk factors associated with poorer outcomes in transplant recipients aged ≥65 years. Careful selection and evaluation of potential candidates may improve graft and patient outcomes in older patients.

4.
Kidney Int ; 97(1): 130-142, 2020 01.
Article in English | MEDLINE | ID: mdl-31685310

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-ß. These cells exhibited immunosuppressive effects on innate immune cells via secretion of IL-10 and transforming growth factor-ß. 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.


Subject(s)
Immunity, Innate , Kidney/cytology , Lymphocyte Subsets/immunology , Nephritis/immunology , Reperfusion Injury/complications , Adoptive Transfer , Animals , Cell Separation , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Flow Cytometry , Homeodomain Proteins/genetics , Humans , Interleukin-10/metabolism , Interleukin-2/antagonists & inhibitors , Interleukin-2/metabolism , Kidney/blood supply , Kidney/immunology , Kidney/pathology , Lymphocyte Subsets/metabolism , Lymphocyte Subsets/transplantation , Macrophages/immunology , Male , Mice , Mice, Knockout , Nephritis/pathology , Primary Cell Culture , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Transforming Growth Factor beta/metabolism
5.
Nephrol Dial Transplant ; 34(11): 1853-1863, 2019 11 01.
Article in English | MEDLINE | ID: mdl-30590794

ABSTRACT

BACKGROUND: Chronic kidney disease (CKD) is a global public health problem, which lacks effective treatment. Previously, we have shown that CD103+ dendritic cells (DCs) are pathogenic in adriamycin nephropathy (AN), a model of human focal segmental glomerulosclerosis (FSGS). Fms-like tyrosine kinase 3 (Flt3) is a receptor that is expressed with high specificity on tissue resident CD103+ DCs. METHODS: To test the effect on CD103+ DCs and kidney injury of inhibition of Flt3, we used a selective Flt3 inhibitor (AC220) to treat mice with AN. RESULTS: Human CD141+ DCs, homologous to murine CD103+ DCs, were significantly increased in patients with FSGS. The number of kidney CD103+ DCs, but not CD103- DCs or plasmacytoid DCs, was significantly decreased in AN mice after AC220 administration. Treatment with AC220 significantly improved kidney function and reduced kidney injury and fibrosis in AN mice. AC220-treated AN mice had decreased levels of inflammatory cytokines and chemokines, tumor necrosis factor-α, interleukin (IL)-1ß, IL-6, CCL2 and CCL5 and reduced kidney infiltration of CD4 T cells and CD8 T cells. The protective effect of AC220 was associated with its suppression of CD103+ DCs-mediated CD8 T cell proliferation and activation in AN mice. CONCLUSION: Flt3 inhibitor AC220 effectively reduced kidney injury in AN mice, suggesting that this inhibitor might be a useful pharmaceutical agent to treat CKD.


Subject(s)
Antigens, CD/metabolism , Benzothiazoles/pharmacology , Dendritic Cells/immunology , Integrin alpha Chains/metabolism , Kidney/drug effects , Lymphocyte Activation/immunology , Phenylurea Compounds/pharmacology , Renal Insufficiency, Chronic/prevention & control , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Cytokines/metabolism , Dendritic Cells/drug effects , Humans , Kidney/immunology , Kidney/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology
6.
PLoS One ; 13(4): e0195657, 2018.
Article in English | MEDLINE | ID: mdl-29641559

ABSTRACT

Studies disrupting the chemokine pathway CX3CL1 (fractalkine)/ CX3CR1 have shown decreased atherosclerosis in animal models but the techniques used to interrupt the pathway have not been easily translatable into human trials. DNA vaccination potentially overcomes the translational difficulties. We evaluated the effect of a DNA vaccine, targeted to CX3CR1, on atherosclerosis in a murine model and examined possible mechanisms of action. DNA vaccination against CX3CR1, enhanced by dendritic cell targeting using DEC-205 single chain variable region fragment (scFv), was performed in 8 week old ApoE-/- mice, fed a normal chow diet. High levels of anti-CX3CR1 antibodies were induced in vaccinated mice. There were no apparent adverse reactions to the vaccine. Arterial vessels of 34 week old mice were examined histologically for atherosclerotic plaque size, macrophage infiltration, smooth muscle cell infiltration and lipid deposition. Vaccinated mice had significantly reduced atherosclerotic plaque in the brachiocephalic artery. There was less macrophage infiltration but no significant change to the macrophage phenotype in the plaques. There was less lipid deposition in the lesions, but there was no effect on smooth muscle cell migration. Targeted DNA vaccination to CX3CR1 was well tolerated, induced a strong immune response and resulted in attenuated atherosclerotic lesions with reduced macrophage infiltration. DNA vaccination against chemokine pathways potentially offers a potential therapeutic option for the treatment of atherosclerosis.


Subject(s)
Antigens, CD/immunology , Atherosclerosis/immunology , Atherosclerosis/prevention & control , CX3C Chemokine Receptor 1/immunology , Dendritic Cells/immunology , Lectins, C-Type/immunology , Minor Histocompatibility Antigens/immunology , Receptors, Cell Surface/immunology , Vaccines, DNA/immunology , Animals , Atherosclerosis/blood , Atherosclerosis/pathology , Cholesterol/blood , Cytokines/blood , Lipid Metabolism/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/pathology , Vaccination
7.
Am J Physiol Renal Physiol ; 314(4): F561-F571, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29357438

ABSTRACT

Cell therapy using macrophages requires large amounts of cells, which are difficult to collect from patients. Patients undergoing peritoneal dialysis (PD) discard huge numbers of peritoneal macrophages in dialysate daily. Macrophages can be modulated to become regulatory macrophages, which have shown great promise as a therapeutic strategy in experimental kidney disease and human kidney transplantation. This study aimed to examine the potential of using peritoneal macrophages (PMs) from peritoneal dialysate to treat kidney disease. Monocytes/macrophages accounted for >40% of total peritoneal leukocytes in both patients and mice undergoing PD. PMs from patients and mice undergoing PD were more mature than peripheral monocytes/macrophages, as shown by low expression of C-C motif chemokine receptor 2 (CCR2) and morphological changes during in vitro culture. PMs from patients and mice undergoing PD displayed normal macrophage function and could be modulated into a regulatory (M2) phenotype. In vivo, adoptive transfer of peritoneal M2 macrophages derived from PD mice effectively protected against kidney injury in mice with adriamycin nephropathy (AN). Importantly, the transfused peritoneal M2 macrophages maintained their M2 phenotype in kidney of AN mice. In conclusion, PMs derived from patients and mice undergoing PD exhibited conventional macrophage features. Peritoneal M2 macrophages derived from PD mice are able to reduce kidney injury in AN, suggesting that peritoneal macrophages from patients undergoing PD may have the potential for clinical therapeutic application.


Subject(s)
Adoptive Transfer , Dialysis Solutions , Doxorubicin , Kidney Diseases/prevention & control , Kidney , Macrophages, Peritoneal/transplantation , Peritoneal Dialysis , Animals , Cell Plasticity , Cell Separation/methods , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Humans , Inflammation Mediators/metabolism , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Kidney Diseases/chemically induced , Kidney Diseases/immunology , Kidney Diseases/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Mice, Inbred BALB C , Phenotype , Time Factors
8.
J Am Soc Nephrol ; 29(3): 961-976, 2018 03.
Article in English | MEDLINE | ID: mdl-29295873

ABSTRACT

The IL-33-type 2 innate lymphoid cell (ILC2) axis has an important role in tissue homeostasis, inflammation, and wound healing. However, the relative importance of this innate immune pathway for immunotherapy against inflammation and tissue damage remains unclear. Here, we show that treatment with recombinant mouse IL-33 prevented renal structural and functional injury and reduced mortality in mice subjected to ischemia-reperfusion injury (IRI). Compared with control-treated IRI mice, IL-33-treated IRI mice had increased levels of IL-4 and IL-13 in serum and kidney and more ILC2, regulatory T cells (Tregs), and anti-inflammatory (M2) macrophages. Depletion of ILC2, but not Tregs, substantially abolished the protective effect of IL-33 on renal IRI. Adoptive transfer of ex vivo-expanded ILC2 prevented renal injury in mice subjected to IRI. This protective effect associated with induction of M2 macrophages in kidney and required ILC2 production of amphiregulin. Treatment of mice with IL-33 or ILC2 after IRI was also renoprotective. Furthermore, in a humanized mouse model of renal IRI, treatment with human IL-33 or transfer of ex vivo-expanded human ILC2 ameliorated renal IRI. This study has uncovered a major protective role of the IL-33-ILC2 axis in renal IRI that could be potentiated as a therapeutic strategy.


Subject(s)
Interleukin-33/therapeutic use , Kidney Diseases/prevention & control , Lymphocytes/immunology , Lymphocytes/metabolism , Reperfusion Injury/prevention & control , Amphiregulin/metabolism , Animals , Female , Humans , Immunity, Innate , Interleukin-13/metabolism , Interleukin-4/metabolism , Kidney Diseases/immunology , Kidney Diseases/pathology , Lymphocyte Count , Macrophages/immunology , Male , Mice , Recombinant Proteins/therapeutic use , Reperfusion Injury/immunology , Reperfusion Injury/pathology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology
9.
Nephrol Dial Transplant ; 32(5): 781-791, 2017 May 01.
Article in English | MEDLINE | ID: mdl-27566305

ABSTRACT

BACKGROUND: Endothelial cells are known to contribute to kidney fibrosis via endothelial-mesenchymal transition (EndoMT). Matrix metalloproteinase 9 (MMP-9) is known to be profibrotic. However, whether MMP-9 contributes to kidney fibrosis via EndoMT is unknown. METHODS: Primary mouse renal peritubular endothelial cells (MRPECs) were isolated and treated by recombinant human transforming growth factor beta 1 (rhTGF-ß1) with or without MMP-9 inhibitor or by recombinant human MMP-9 (rhMMP-9) alone. Kidney fibrosis was induced by unilateral ureteral obstruction (UUO) in MMP-9 knockout (KO) and wide-type (WT) control mice. The effects of MMP-9 on EndoMT of MRPECs and kidney fibrosis were examined. RESULTS: We showed that MRPECs underwent EndoMT after rhTGF-ß1 treatment or in UUO kidney as evidenced by decreased expression of endothelial markers, vascular endothelial cadherin (VE-cadherin) and CD31, and increased levels of mesenchymal markers, α-smooth muscle actin (α-SMA) and vimentin. The expression of fibrosis markers was also up-regulated significantly after rhTGF-ß1 treatment in MRPECs. The EndoMT and fibrosis markers were significantly less in rhTGF-ß1-treated MMP-9 KO MRPECs, whereas MMP-9 alone was sufficient to induce EndoMT in MRPECs. UUO kidney of MMP-9 KO mice showed significantly less interstitial fibrosis and EndoMT in MRPECs. Notch signaling shown by Notch intracellular domain (NICD) was increased, while Notch-1 was decreased in rhTGF-ß1-treated MRPECs of MMP-9 WT but not MMP-9 KO mice. Inhibition of MMP-9 or Notch signaling prevented rhTGF-ß1- or rhMMP-9-induced α-SMA and NICD upregulation in MRPECs. UUO kidney of MMP-9 KO mice had less staining of Notch signaling transcription factor Hey-1 in VE-cadherin-positive MRPECs than WT controls. CONCLUSIONS: Our results demonstrate that MMP-9-dependent Notch signaling plays an important role in kidney fibrosis through EndoMT of MRPECs.


Subject(s)
Endothelium/pathology , Fibrosis/pathology , Kidney Diseases/pathology , Matrix Metalloproteinase 9/metabolism , Mesoderm/pathology , Receptors, Notch/metabolism , Animals , Endothelium/metabolism , Fibrosis/metabolism , Humans , Kidney Diseases/metabolism , Male , Mesoderm/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Signal Transduction , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
10.
Am J Pathol ; 186(7): 1847-1860, 2016 07.
Article in English | MEDLINE | ID: mdl-27182643

ABSTRACT

Loss of E-cadherin marks a defect in epithelial integrity and polarity during tissue injury and fibrosis. Whether loss of E-cadherin plays a causal role in fibrosis is uncertain. α3ß1 Integrin has been identified to complex with E-cadherin in cell-cell adhesion, but little is known about the details of their cross talk. Herein, E-cadherin gene (Cdh1) was selectively deleted from proximal tubules of murine kidney by Sglt2Cre. Ablation of E-cadherin up-regulated α3ß1 integrin at cell-cell adhesion. E-cadherin-deficient proximal tubular epithelial cell displayed enhanced transforming growth factor-ß1-induced α-smooth muscle actin (α-SMA) and vimentin expression, which was suppressed by siRNA silencing of α3 integrin, but not ß1 integrin. Up-regulation of transforming growth factor-ß1-induced α-SMA was mediated by an α3 integrin-dependent increase in integrin-linked kinase (ILK). Src phosphorylation of ß-catenin and consequent p-ß-catenin-Y654/p-Smad2 transcriptional complex underlies the transcriptional up-regulation of ILK. Kidney fibrosis after unilateral ureteric obstruction or ischemia reperfusion was increased in proximal tubule E-cadherin-deficient mice in comparison to that of E-cadherin intact control mice. The exacerbation of fibrosis was explained by the α3 integrin-dependent increase of ILK, ß-catenin nuclear translocation, and α-SMA/proximal tubular-specific Cre double positive staining in proximal tubular epithelial cell. These studies delineate a nonconventional integrin/ILK signaling by α3 integrin-dependent Src/p-ß-catenin-Y654/p-Smad2-mediated up-regulation of ILK through which loss of E-cadherin leads to kidney fibrosis.


Subject(s)
Cadherins/deficiency , Integrin alpha3beta1/metabolism , Kidney Diseases/pathology , Protein Serine-Threonine Kinases/metabolism , Animals , Blotting, Western , Cell Adhesion , Chromatin Immunoprecipitation , Disease Models, Animal , Fibrosis/metabolism , Fibrosis/pathology , Immunohistochemistry , Immunoprecipitation , Kidney Diseases/metabolism , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology
11.
BMC Cell Biol ; 17(1): 21, 2016 04 29.
Article in English | MEDLINE | ID: mdl-27130612

ABSTRACT

BACKGROUND: Endothelial-mesenchymal transition (EndoMT) is a major source of myofibroblast formation in kidney fibrosis. Our previous study showed a profibrotic role for matrix metalloproteinase 9 (MMP-9) in kidney fibrosis via induction of epithelial-mesenchymal transition (EMT). Inhibition of MMP-9 activity reduced kidney fibrosis in murine unilateral ureteral obstruction. This study investigated whether MMP-9 also plays a role in EndoMT in human glomerular endothelial cells. RESULTS: TGF-ß1 (10 or 20 ng/ml) induced EndoMT in HKGECs as shown by morphological changes. In addition, VE-cadherin and CD31 were significantly downregulated, whereas α-SMA, vimentin, and N-cadherin were upregulated. RT-PCR revealed that Snail, a known inducer of EMT, was upregulated. The MMP inhibitor GM6001 abrogated TGF-ß1-induced EndoMT. Zymography indicated that MMP-9 was also upregulated in TGF-ß1-treated HKGECs. Recombinant MMP-9 (2 µg/ml) induced EndoMT in HKGECs via Notch signaling, as evidenced by increased formation of the Notch intracellular domain (NICD) and decreased Notch 1. Inhibition of MMP-9 activity by its inhibitor showed a dose-dependent response in preventing TGF-ß1-induced α-SMA and NICD in HKGECs, whereas inhibition of Notch signaling by γ-secretase inhibitor (GSI) blocked rMMP-9-induced EndoMT. CONCLUSIONS: Taken together, our results demonstrate that MMP-9 plays an important role in TGF-ß1-induced EndoMT via upregulation of Notch signaling in HKGECs.


Subject(s)
Endothelial Cells/metabolism , Kidney Glomerulus/cytology , Matrix Metalloproteinase 9/metabolism , Mesoderm/cytology , Receptors, Notch/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Dipeptides/pharmacology , Endothelial Cells/drug effects , Humans , Matrix Metalloproteinase Inhibitors/pharmacology , Mesoderm/drug effects , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology
12.
J Am Soc Nephrol ; 27(5): 1344-60, 2016 05.
Article in English | MEDLINE | ID: mdl-26376858

ABSTRACT

CD103(+) dendritic cells (DCs) in nonlymphoid organs exhibit two main functions: maintaining tolerance by induction of regulatory T cells and protecting against tissue infection through cross-presentation of foreign antigens to CD8(+) T cells. However, the role of CD103(+) DCs in kidney disease is unknown. In this study, we show that CD103(+) DCs are one of four subpopulations of renal mononuclear phagocytes in normal kidneys. CD103(+) DCs expressed DC-specific surface markers, transcription factors, and growth factor receptors and were found in the kidney cortex but not in the medulla. The number of kidney CD103(+) DCs was significantly higher in mice with adriamycin nephropathy (AN) than in normal mice, and depletion of CD103(+) DCs attenuated kidney injury in AN mice. In vitro, kidney CD103(+) DCs preferentially primed CD8(+) T cells and did not directly induce tubular epithelial cell apoptosis. Adoptive transfer of CD8(+) T cells significantly exacerbated kidney injury in AN SCID mice, whereas depletion of CD103(+) DCs in these mice impaired activation and proliferation of transfused CD8(+) T cells and prevented the exacerbation of kidney injury associated with this transfusion. In conclusion, kidney CD103(+) DCs display a pathogenic role in murine CKD via activation of CD8(+) T cells.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Doxorubicin/adverse effects , Integrin alpha Chains/immunology , Kidney Diseases/chemically induced , Kidney Diseases/immunology , Animals , Mice
13.
Curr Protoc Immunol ; 109: 15.29.1-15.29.6, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25845564

ABSTRACT

Human membranous nephritis is a major cause of end-stage kidney disease. Active Heymann nephritis (HN) is an auto-immune model of membranous nephritis induced in Lewis rats by immunization with a crude renal tubular antigen (Fx1A) or megalin (gp330). The pathogenesis of HN is through the binding of anti-Fx1A autoantibodies to the auto-antigen expressed on glomerular epithelial cells, resulting in severe glomerular injury and proteinuria. The pathological features of HN include immune deposits in glomeruli and infiltration of glomeruli and the tubulointerstitium by macrophages and T cells. This unit describes the method of the preparation of Fx1A and the induction of HN in Lewis rats by immunization with Fx1A.


Subject(s)
Glomerulonephritis, Membranous/etiology , Glomerulonephritis, Membranous/pathology , Animals , Biopsy , Disease Models, Animal , Heymann Nephritis Antigenic Complex/administration & dosage , Heymann Nephritis Antigenic Complex/immunology , Male , Rats , Rats, Inbred Lew
14.
Curr Protoc Immunol ; 108: 15.28.1-15.28.6, 2015 Feb 02.
Article in English | MEDLINE | ID: mdl-25640989

ABSTRACT

Chronic proteinuric renal injury is a major cause of end stage renal disease. Adriamycin nephropathy (AN) is a murine model of chronic proteinuric renal disease whereby chemical injury is followed by immune and structural changes that mimic human disease. This unit describes the method of AN induced by a single injection of adriamycin (ADR) in BALB/c mice. After the initial toxic injury, an immune-mediated chronic proteinuric renal disease that resembles human focal segmental glomerulosclerosis develops. The clinic pathological features of AN are nephrotic syndrome, focal glomerulosclerosis, tubular injury, and interstitial compartment expansion with mononuclear cell infiltrates that are composed largely of macrophages and T cells.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Doxorubicin/adverse effects , Kidney Diseases/chemically induced , Kidney Diseases/pathology , Animals , Antibiotics, Antineoplastic/administration & dosage , Biopsy , Disease Models, Animal , Doxorubicin/administration & dosage , Kidney/metabolism , Kidney/pathology , Kidney/ultrastructure , Kidney Diseases/metabolism , Kidney Function Tests , Male , Mice
15.
J Am Soc Nephrol ; 26(9): 2199-211, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25556172

ABSTRACT

IL-25 is an important immune regulator that can promote Th2 immune response-dependent immunity, inflammation, and tissue repair in asthma, intestinal infection, and autoimmune diseases. In this study, we examined the effects of IL-25 in renal ischemic/reperfusion injury (IRI). Treating IRI mice with IL-25 significantly improved renal function and reduced renal injury. Furthermore, IL-25 treatment increased the levels of IL-4, IL-5, and IL-13 in serum and kidney and promoted induction of alternatively activated (M2) macrophages in kidney. Notably, IL-25 treatment also increased the frequency of type 2 innate lymphoid cells (ILC2s) and multipotent progenitor type 2 (MPP(type2)) cells in kidney. IL-25-responsive ILC2 and MPP(type2) cells produced greater amounts of Th2 cytokines that associated with the induction of M2 macrophages and suppression of classically activated (M1) macrophages in vitro. Finally, adoptive transfer of ILC2s or MPP(type2) cells not only reduced renal functional and histologic injury in IRI mice but also induced M2 macrophages in kidney. In conclusion, our data identify a mechanism whereby IL-25-elicited ILC2 and MPP(type2) cells regulate macrophage phenotype in kidney and prevent renal IRI.


Subject(s)
Acute Kidney Injury/immunology , Acute Kidney Injury/prevention & control , Immunologic Factors/therapeutic use , Interleukin-17/therapeutic use , Lymphocytes/drug effects , Multipotent Stem Cells/drug effects , Reperfusion Injury/immunology , Reperfusion Injury/prevention & control , Acute Kidney Injury/etiology , Adoptive Transfer , Animals , Cell Survival , Cells, Cultured , Immunity, Innate/drug effects , Immunity, Innate/immunology , Immunologic Factors/pharmacology , Interleukin-13/metabolism , Interleukin-17/pharmacology , Interleukin-4/metabolism , Interleukin-5/metabolism , Kidney/blood supply , Kidney/metabolism , Lymphocytes/cytology , Macrophages/drug effects , Male , Mice , Mice, Inbred BALB C , Multipotent Stem Cells/cytology , Reperfusion Injury/complications , Th2 Cells/immunology
16.
J Am Soc Nephrol ; 26(2): 349-63, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25012165

ABSTRACT

Conventional markers of macrophages (Mфs) and dendritic cells (DCs) lack specificity and often overlap, leading to confusion and controversy regarding the precise function of these cells in kidney and other diseases. This study aimed to identify the phenotype and function of renal mononuclear phagocytes (rMPs) expressing key markers of both Mфs and DCs. F4/80(+)CD11c(+) cells accounted for 45% of total rMPs in normal kidneys and in those from mice with Adriamycin nephropathy (AN). Despite expression of the DC marker CD11c, these double-positive rMPs displayed the features of Mфs, including Mф-like morphology, high expression of CD68, CD204, and CD206, and high phagocytic ability but low antigen-presenting ability. F4/80(+)CD11c(+) cells were found in the cortex but not in the medulla of the kidney. In AN, F4/80(+)CD11c(+) cells displayed an M1 Mф phenotype with high expression of inflammatory mediators and costimulatory factors. Adoptive transfer of F4/80(+)CD11c(+) cells separated from diseased kidney aggravated renal injury in AN mice. Furthermore, adoptive transfer of common progenitors revealed that kidney F4/80(+)CD11c(+) cells were derived predominantly from monocytes, but not from pre-DCs. In conclusion, renal F4/80(+)CD11c(+) cells are a major subset of rMPs and display Mф-like phenotypic and functional characteristics in health and in AN.


Subject(s)
Antigens, Differentiation/metabolism , CD11 Antigens/metabolism , Kidney Diseases/pathology , Kidney/pathology , Macrophages/pathology , Phagocytes/pathology , Phenotype , Adoptive Transfer , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Biomarkers/metabolism , Disease Models, Animal , Doxorubicin/adverse effects , In Vitro Techniques , Kidney/physiology , Kidney Diseases/chemically induced , Kidney Diseases/physiopathology , Lectins, C-Type/metabolism , Macrophages/immunology , Macrophages/physiology , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phagocytes/immunology , Phagocytes/physiology , Receptors, Cell Surface/metabolism , Scavenger Receptors, Class A/metabolism
17.
Kidney Int ; 85(4): 794-806, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24048378

ABSTRACT

Alternatively activated macrophages (M2) regulate immune responses and ex vivo polarized splenic M2 are able to ameliorate renal injury including models of renal disease, such as adriamycin nephropathy. Whether M2 derived from other organs have similar protective efficacy is unknown. Here, we report adoptively transferred bone marrow M2 macrophages did not improve renal function or reduce renal injury in adriamycin nephropathy, whereas splenic M2 macrophages were protective. Bone marrow and splenic M2 macrophages showed similar regulatory phenotypes and suppressive functions in vitro. Within the inflamed kidney, suppressive phenotypes in bone marrow but not in splenic M2 macrophages, were dramatically reduced. Loss of the suppressive phenotype in bone marrow M2 was related to strong proliferation of bone marrow M2. Bone marrow M2 proliferation in vivo correlated with M-CSF expression by tubular cells in the inflamed kidney. Inhibition of M-CSF in vitro limited bone marrow M2 proliferation and prevented switch of phenotype. Proliferating cells derived from transfused bone marrow M2 were inflammatory rather than regulatory in their phenotype and function. Thus bone marrow in contrast to splenic M2 macrophages do not protect against renal structural and functional injury in murine adriamycin nephropathy. The failed renoprotection of bone marrow M2 is due to the switch of transfused M2 macrophages from a regulatory to an inflammatory phenotype.


Subject(s)
Adoptive Transfer , Kidney Diseases/therapy , Macrophages/transplantation , Spleen/immunology , Animals , Bone Marrow Cells/cytology , Cell Proliferation , Doxorubicin , Kidney Diseases/chemically induced , Kidney Tubules/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/cytology , Macrophages/physiology , Male , Mice, Inbred BALB C , Phenotype
18.
Curr Hypertens Rev ; 9(1): 76-84, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23971646

ABSTRACT

The role of innate and adaptive immunity in the pathogenesis of hypertension has received increasing recognition over the past few years. This review will focus on recent developments in this emerging area. In particular, the role of macrophages and lymphocytes will be highlighted.


Subject(s)
Hypertension/etiology , Immune System/physiology , Humans , Lymphocytes/physiology , Macrophages/physiology
19.
Kidney Int ; 84(4): 745-55, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23636175

ABSTRACT

Two types of alternatively activated macrophages, M(2a) induced by IL-4/IL-13 and M(2c) by IL-10/TGF-ß, exhibit anti-inflammatory functions in vitro and protect against renal injury in vivo. Since their relative therapeutic efficacy is unclear, we compared the effects of these two macrophage subsets in murine adriamycin nephrosis. Both subsets significantly reduced renal inflammation and renal injury; however, M(2c) macrophages more effectively reduced glomerulosclerosis, tubular atrophy, interstitial expansion, and proteinuria than M(2a) macrophages. The M(2c) macrophages were also more effective than M(2a) in reduction of macrophage and CD4(+) T-cell infiltration in kidney. Moreover, nephrotic mice treated with M(2c) had a greater reduction in renal fibrosis than those treated with M(2a). M(2c) but not M(2a) macrophages induced regulatory T cells (Tregs) from CD4(+)CD25(-) T cells in vitro, and increased Treg numbers in local draining lymph nodes of nephrotic mice. To determine whether the greater protection with M(2c) was due to their capability to induce Tregs, the Tregs were depleted by PC61 antibody in nephrotic mice treated with M(2a) or M(2c). Treg depletion diminished the superior effects of M(2c) compared to M(2a) in protection against renal injury, inflammatory infiltrates, and renal fibrosis. Thus, M(2c) are more potent than M(2a) macrophages in protecting against renal injury due to their ability to induce Tregs.


Subject(s)
Adoptive Transfer/methods , Cell- and Tissue-Based Therapy/methods , Macrophages/classification , Macrophages/physiology , Renal Insufficiency, Chronic/therapy , Animals , Cell Differentiation/physiology , Cells, Cultured , Disease Management , Disease Models, Animal , Doxorubicin/adverse effects , In Vitro Techniques , Macrophages/cytology , Male , Mice , Mice, Inbred BALB C , Nephrosis/chemically induced , Nephrosis/physiopathology , Nephrosis/prevention & control , Phenotype , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/physiopathology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/physiology
20.
Lab Invest ; 93(4): 434-49, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23358111

ABSTRACT

A pro-fibrotic role of matrix metalloproteinase-9 (MMP-9) in tubular cell epithelial-mesenchymal transition (EMT) is well established in renal fibrosis; however studies from our group and others have demonstrated some previously unrecognized complexity of MMP-9 that has been overlooked in renal fibrosis. Therefore, the aim of this study was to determine the expression pattern, origin and the exact mechanism underlying the contribution of MMP-9 to unilateral ureteral obstruction (UUO), a well-established model of renal fibrosis via MMP-9 inhibition. Renal MMP-9 expression in BALB/c mice with UUO was examined on day 1, 3, 5, 7, 9, 11 and 14. To inhibit MMP-9 activity, MMP-2/9 inhibitor or MMP-9-neutralizing antibody was administered daily for 4 consecutive days from day 0-3, 6-9 or 10-13 and tissues harvested at day 14. In UUO, there was a bi-phasic early- and late-stage upregulation of MMP-9 activity. Interestingly, tubular epithelial cells (TECs) were the predominant source of MMP-9 during early stage, whereas TECs, macrophages and myofibroblasts produced MMP-9 during late-stage UUO. Early- and late-stage inhibition of MMP-9 in UUO mice significantly reduced tubular cell EMT and renal fibrosis. Moreover, MMP-9 inhibition caused a significant reduction in MMP-9-cleaved osteopontin and macrophage infiltration in UUO kidney. Our in vitro study showed MMP-9-cleaved osteopontin enhanced macrophage transwell migration and MMP-9 of both primary TEC and macrophage induced tubular cell EMT. In summary, our result suggests that MMP-9 of both TEC and macrophage origin may directly or indirectly contribute to the pathogenesis of renal fibrosis via osteopontin cleavage, which, in turn further recruit macrophage and induce tubular cell EMT. Our study also highlights the time dependency of its expression and the potential of stage-specific inhibition strategy against renal fibrosis.


Subject(s)
Kidney Diseases/immunology , Kidney/pathology , Matrix Metalloproteinase 9/metabolism , Osteopontin/metabolism , Ureteral Obstruction/metabolism , Animals , Cell Movement , Cells, Cultured , Epithelial Cells/physiology , Epithelial-Mesenchymal Transition , Fibrosis , Kidney/metabolism , Kidney Diseases/metabolism , Kidney Diseases/pathology , Macrophages/physiology , Mice , Mice, Inbred BALB C , Snail Family Transcription Factors , Transcription Factors/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...