Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1251452, 2023.
Article in English | MEDLINE | ID: mdl-38022500

ABSTRACT

Necroptosis, a pathway of regulated necrosis, involves recruitment and activation of RIPK1, RIPK3 and MLKL, leading to cell membrane rupture, cell death and release of intracellular contents causing further injury and inflammation. Necroptosis is believed to play an important role in the pathogenesis of kidney ischemia-reperfusion injury (IRI). However, the dynamics of necroptosis in kidney IRI is poorly understood, in part due to difficulties in detecting phosphorylated MLKL (pMLKL), the executioner of the necroptosis pathway. Here, we investigated the temporal and spatial activation of necroptosis in a mouse model of unilateral warm kidney IRI, using a robust method to stain pMLKL. We identified the period 3-12 hrs after reperfusion as a critical phase for the activation of necroptosis in proximal tubular cells. After 12 hrs, the predominant pattern of pMLKL staining shifted from cytoplasmic to membrane, indicating progression to the terminal phase of necroptotic cell death. Mlkl-ko mice exhibited reduced kidney inflammation at 12 hrs and lower serum creatinine and tubular injury at 24 hrs compared to wild-type littermates. Interestingly, we observed increased apoptosis in the injured kidneys of Mlkl-ko mice, suggesting a relationship between necroptosis and apoptosis in kidney IRI. Together, our findings confirm the role of necroptosis and necroinflammation in kidney IRI, and identify the first 3 hrs following reperfusion as a potential window for targeted treatments.


Subject(s)
Necroptosis , Reperfusion Injury , Animals , Mice , Kidney/pathology , Necrosis/pathology , Inflammation/metabolism , Reperfusion Injury/metabolism
2.
Sci Rep ; 11(1): 21873, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34750424

ABSTRACT

The complement system is a potent mediator of ischemia-reperfusion injury (IRI), which detrimentally affects the function and survival of transplanted kidneys. Human complement receptor 1 (HuCR1) is an integral membrane protein that inhibits complement activation by blocking the convertases that activate C3 and C5. We have previously reported that CSL040, a truncated form of recombinant soluble HuCR1 (sHuCR1), has enhanced complement inhibitory activity and improved pharmacokinetic properties compared to the parent molecule. Here, we compared the capacity of CSL040 and full-length sHuCR1 to suppress complement-mediated organ damage in a mouse model of warm renal IRI. Mice were treated with two doses of CSL040 or sHuCR1, given 1 h prior to 22 min unilateral renal ischemia and again 3 h later. 24 h after reperfusion, mice treated with CSL040 were protected against warm renal IRI in a dose-dependent manner, with the highest dose of 60 mg/kg significantly reducing renal dysfunction, tubular injury, complement activation, endothelial damage, and leukocyte infiltration. In contrast, treatment with sHuCR1 at a molar equivalent dose to 60 mg/kg CSL040 did not confer significant protection. Our results identify CSL040 as a promising therapeutic candidate to attenuate renal IRI and demonstrate its superior efficacy over full-length sHuCR1 in vivo.


Subject(s)
Kidney/injuries , Receptors, Complement 3b/administration & dosage , Reperfusion Injury/prevention & control , Animals , Complement Activation/drug effects , Disease Models, Animal , Humans , Kidney/drug effects , Kidney/immunology , Kidney Transplantation/adverse effects , Male , Mice , Mice, Inbred C57BL , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Receptors, Complement 3b/chemistry , Reperfusion Injury/etiology , Reperfusion Injury/immunology , Solubility
3.
J Immunol ; 205(5): 1433-1440, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32839213

ABSTRACT

Ischemia-reperfusion injury (IRI) is a complex inflammatory process that detrimentally affects the function of transplanted organs. Neutrophils are important contributors to the pathogenesis of renal IRI. Signaling by G-CSF, a regulator of neutrophil development, trafficking, and function, plays a key role in several neutrophil-associated inflammatory disease models. In this study, we investigated whether targeting neutrophils with a neutralizing mAb to G-CSFR would reduce inflammation and protect against injury in a mouse model of warm renal IRI. Mice were treated with anti-G-CSFR 24 h prior to 22-min unilateral renal ischemia. Renal function and histology, complement activation, and expression of kidney injury markers, and inflammatory mediators were assessed 24 h after reperfusion. Treatment with anti-G-CSFR protected against renal IRI in a dose-dependent manner, significantly reducing serum creatinine and urea, tubular injury, neutrophil and macrophage infiltration, and complement activation (plasma C5a) and deposition (tissue C9). Renal expression of several proinflammatory genes (CXCL1/KC, CXCL2/MIP-2, MCP-1/CCL2, CXCR2, IL-6, ICAM-1, P-selectin, and C5aR) was suppressed by anti-G-CSFR, as was the level of circulating P-selectin and ICAM-1. Neutrophils in anti-G-CSFR-treated mice displayed lower levels of the chemokine receptor CXCR2, consistent with a reduced ability to traffic to inflammatory sites. Furthermore, whole transcriptome analysis using RNA sequencing showed that gene expression changes in IRI kidneys after anti-G-CSFR treatment were indistinguishable from sham-operated kidneys without IRI. Hence, anti-G-CSFR treatment prevented the development of IRI in the kidneys. Our results suggest G-CSFR blockade as a promising therapeutic approach to attenuate renal IRI.


Subject(s)
Kidney Diseases/drug therapy , Protective Agents/pharmacology , Receptors, Granulocyte Colony-Stimulating Factor/antagonists & inhibitors , Reperfusion Injury/drug therapy , Animals , Chemokines/metabolism , Complement Activation/drug effects , Creatinine/blood , Disease Models, Animal , Gene Expression/drug effects , Inflammation/blood , Inflammation/drug therapy , Inflammation/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney Diseases/blood , Kidney Diseases/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/metabolism , Reperfusion Injury/blood , Reperfusion Injury/metabolism , Urea/blood
4.
Transplant Direct ; 5(4): e341, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30993186

ABSTRACT

BACKGROUND: Complement activation plays an important role in the pathogenesis of renal ischemia-reperfusion (IR) injury (IRI), but whether this involves damage to the vasculoprotective endothelial glycocalyx is not clear. We investigated the impact of complement activation on glycocalyx integrity and renal dysfunction in a mouse model of renal IRI. METHODS: Right nephrectomized male C57BL/6 mice were subjected to 22 minutes left renal ischemia and sacrificed 24 hours after reperfusion to analyze renal function, complement activation, glycocalyx damage, endothelial cell activation, inflammation, and infiltration of neutrophils and macrophages. RESULTS: Ischemia-reperfusion induced severe renal injury, manifested by significantly increased serum creatinine and urea, complement activation and deposition, loss of glycocalyx, endothelial activation, inflammation, and innate cell infiltration. Treatment with the anti-C5 antibody BB5.1 protected against IRI as indicated by significantly lower serum creatinine (P = 0.04) and urea (P = 0.003), tissue C3b/c and C9 deposition (both P = 0.004), plasma C3b (P = 0.001) and C5a (P = 0.006), endothelial vascular cell adhesion molecule-1 expression (P = 0.003), glycocalyx shedding (tissue heparan sulfate [P = 0.001], plasma syndecan-1 [P = 0.007], and hyaluronan [P = 0.02]), inflammation (high mobility group box-1 [P = 0.0003]), and tissue neutrophil (P = 0.0009) and macrophage (P = 0.004) infiltration. CONCLUSIONS: Together, our data confirm that the terminal pathway of complement activation plays a key role in renal IRI and demonstrate that the mechanism of injury involves shedding of the glycocalyx.

5.
Nutrients ; 10(9)2018 Sep 12.
Article in English | MEDLINE | ID: mdl-30213104

ABSTRACT

The contribution of cows' milk containing beta-casein protein A1 variant to the development of type 1 diabetes (T1D) has been controversial for decades. Despite epidemiological data demonstrating a relationship between A1 beta-casein consumption and T1D incidence, direct evidence is limited. We demonstrate that early life exposure to A1 beta-casein through the diet can modify progression to diabetes in non-obese diabetic (NOD) mice, with the effect apparent in later generations. Adult NOD mice from the F0 generation and all subsequent generations (F1 to F4) were fed either A1 or A2 beta-casein supplemented diets. Diabetes incidence in F0⁻F2 generations was similar in both cohorts of mice. However, diabetes incidence doubled in the F3 generation NOD mice fed an A1 beta-casein supplemented diet. In F4 NOD mice, subclinical insulitis and altered glucose handling was evident as early as 10 weeks of age in A1 fed mice only. A significant decrease in the proportion of non-conventional regulatory T cell subset defined as CD4⁺CD25-FoxP3⁺ was evident in the F4 generation of A1 fed mice. This feeding intervention study demonstrates that dietary A1 beta-casein may affect glucose homeostasis and T1D progression, although this effect takes generations to manifest.


Subject(s)
Caseins/toxicity , Diabetes Mellitus, Type 1/etiology , Dietary Supplements/toxicity , Animal Feed , Animal Nutritional Physiological Phenomena , Animals , Biomarkers/blood , Blood Glucose/metabolism , Caseins/administration & dosage , Cells, Cultured , Coculture Techniques , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Female , Gastrointestinal Microbiome , Gene-Environment Interaction , Genetic Predisposition to Disease , Male , Mice, Inbred NOD , Risk Factors , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
6.
J Immunol ; 198(12): 4837-4845, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28500075

ABSTRACT

Deficiency in the membrane-bound complement regulators CD55 and CD59 exacerbates renal ischemia-reperfusion injury (IRI) in mouse models, but the effect of increasing CD55 and CD59 activity has not been examined. In this study, we investigated the impact of overexpression of human (h) CD55 ± hCD59 or treatment with soluble rhCD55 in a mouse model of renal IRI. Unilaterally nephrectomised mice were subjected to 18 (mild IRI) or 22 min (moderate IRI) warm renal ischemia, and analyzed 24 h after reperfusion for renal function (serum creatinine and urea), complement deposition (C3b/c and C9), and infiltration of neutrophils and macrophages. Transgenic mice expressing hCD55 alone were protected against mild renal IRI, with reduced creatinine and urea levels compared with wild type littermates. However, the renal function of the hCD55 mice was not preserved in the moderate IRI model, despite a reduction in C3b/c and C9 deposition and innate cell infiltration. Mice expressing both hCD55 and hCD59, on the other hand, were protected in the moderate IRI model, with significant reductions in all parameters measured. Wild type mice treated with rhCD55 immediately after reperfusion were also protected in the moderate IRI model. Thus, manipulation of CD55 activity to increase inhibition of the C3 and C5 convertases is protective against renal IRI, and the additional expression of hCD59, which regulates the terminal complement pathway, provides further protection. Therefore, anti-complement therapy using complement regulatory proteins may provide a potential clinical option for preventing tissue and organ damage in renal IRI.


Subject(s)
CD55 Antigens/genetics , CD55 Antigens/therapeutic use , CD59 Antigens/genetics , Kidney Diseases/therapy , Reperfusion Injury/therapy , Animals , CD55 Antigens/immunology , Complement Activation , Creatinine/blood , Humans , Kidney Diseases/immunology , Kidney Diseases/physiopathology , Macrophages/immunology , Mice , Mice, Transgenic , Neutrophils/immunology , Reperfusion Injury/immunology , Reperfusion Injury/physiopathology , Urea/blood
7.
Nephrology (Carlton) ; 22(7): 505-512, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27517975

ABSTRACT

AIM: Regulatory T cells (Treg) are important in mediating immune tolerance and outcomes of allotransplantation. CD4+ CD25+ CD39+ co-expression identifies memory Treg; CD4+ CD25- CD39+ memory T effectors. We sought to determine CD4+ CD25+/- CD39+ expression from the peripheral blood of patients with end stage renal failure, following transplantation and during episodes of acute cellular rejection. METHODS: CD4+ T cells were isolated from peripheral blood leucocytes and analysed for CD25 and CD39 expression by flow cytometry. Treg suppressive function was measured by suppression of autologous effector T-cell proliferation by Treg in co-culture. RESULTS: CD4+ CD25+/- CD39+ T-cell subsets were tracked longitudinally in the peripheral blood of 17 patients following renal transplantation. Patients with acute T-cell-mediated rejection diagnosed on biopsy had reduced CD4+ CD25+ CD39+ mTreg (P < 0.05) and CD4+ CD25- CD39+ mTeff (P < 0.01) cells compared with non-rejecting patients. CD4+ CD25+ CD39+ mTreg (P < 0.05) and CD4+ CD25- CD39+ mTeff (P = 0.057) were reduced in long-term transplant patients (>1 year) compared with non-immunosuppressed controls. Interestingly, remaining CD4+ CD25+ CD39+ mTreg in the stable transplant patients displayed more potent suppressive capacity compared with non-immunosuppressed controls (83.2% ± 3.1% vs 45.7% ± 8.0%, nTeff:Treg ratio 8:1, P < 0.01). CONCLUSION: CD4+ CD25+ CD39+ mTreg and CD4+ CD25- CD39+ mTeff in peripheral blood can be tracked in renal transplant patients. Acute cellular rejection was accompanied by reduced mTreg and mTeff. Determining changes in these T-cell subsets may help to identify patients with, or at high risk of, renal allograft rejection.


Subject(s)
Antigens, CD/blood , Apyrase/blood , Graft Rejection/immunology , Interleukin-2 Receptor alpha Subunit/blood , Kidney Failure, Chronic/surgery , Kidney Transplantation/adverse effects , T-Lymphocytes, Regulatory/immunology , Acute Disease , Adult , Aged , Allografts , Biomarkers/blood , Case-Control Studies , Cell Proliferation , Cells, Cultured , Coculture Techniques , Female , Graft Rejection/blood , Graft Rejection/diagnosis , Graft Rejection/drug therapy , Graft Survival , Humans , Immunosuppressive Agents/therapeutic use , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/immunology , Lymphocyte Activation , Male , Middle Aged , Phenotype , Prospective Studies , Risk Factors , T-Lymphocytes, Regulatory/drug effects , Time Factors , Transplantation Tolerance , Treatment Outcome
8.
Nephrology (Carlton) ; 21(5): 397-403, 2016 May.
Article in English | MEDLINE | ID: mdl-26369693

ABSTRACT

AIM: The phospholipase A2 receptor (PLA2R) is the major target antigen in idiopathic membranous nephropathy (iMN). The aim of this prospective study was to determine the prevalence of anti-PLA2R in iMN in an Australian cohort. METHODS: Serum anti-PLA2R was measured using two techniques, an enzyme-linked immunosorbent assay (ELISA) and a cell-based indirect immunofluorescence test. Kidney biopsies were also examined for the presence of PLA2R using a polyclonal antibody. A group of 21 patients with iMN were compared with a group of 19 patients with secondary MN and other glomerular diseases. RESULTS: Seventeen of 21 patients with iMN were positive for anti-PLA2R on both ELISA and indirect immunofluorescence test, and 14 of these patients also had positive staining for PLA2R in the biopsy (tissue was unavailable in two patients). Three patients with iMN had positive staining in the biopsy only, and one patient was negative in both the serum and the biopsy. None of the patients with secondary MN or other glomerular diseases had anti-PLA2R antibodies or PLA2R in the biopsy. There was wide inter-individual variation in titre on ELISA, but serial levels within an individual patient enabled monitoring of disease, and a fall in titre correlated with clinical remission. CONCLUSIONS: This is the first Australian series to examine the incidence of anti-PLA2R in iMN. It is also unique in examining sera by two separate techniques in conjunction with tissue localization of PLA2R. We have shown 100% specificity of both serum anti-PLA2R and glomerular PLA2R in iMN, with a sensitivity of 81.0%. When serum testing is combined with tissue localization of PLA2R, the sensitivity increases to 95.2%.


Subject(s)
Autoantibodies/blood , Glomerulonephritis, Membranous/immunology , Kidney/immunology , Receptors, Phospholipase A2/immunology , Biomarkers/blood , Biopsy , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Glomerulonephritis, Membranous/blood , Glomerulonephritis, Membranous/diagnosis , Humans , Kidney/pathology , Predictive Value of Tests , Prospective Studies , Reproducibility of Results , Time Factors , Victoria
9.
Am J Physiol Renal Physiol ; 305(5): F679-90, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23785098

ABSTRACT

Salt reabsorption is the major energy-requiring process in the kidney, and AMP-activated protein kinase (AMPK) is an important regulator of cellular metabolism. Mice with targeted deletion of the ß1-subunit of AMPK (AMPK-ß1(-/-) mice) had significantly increased urinary Na(+) excretion on a normal salt diet. This was associated with reduced expression of the ß-subunit of the epithelial Na(+) channel (ENaC) and increased subapical tubular expression of kidney-specific Na(+)-K(+)-2Cl(-) cotransporter 2 (NKCC2) in the medullary thick ascending limb of Henle. AMPK-ß1(-/-) mice fed a salt-deficient diet were able to conserve Na(+), but renin secretion increased 180% compared with control mice. Cyclooxygenase-2 mRNA also increased in the kidney cortex, indicating greater signaling through the macula densa tubular salt-sensing pathway. To determine whether the increase in renin secretion was due to a change in regulation of fatty acid metabolism by AMPK, mice with a mutation of the inhibitory AMPK phosphosite in acetyl-CoA carboxylase 1 [ACC1-knockin (KI)(S79A) mice] were examined. ACC1-KI(S79A) mice on a normal salt diet had no increase in salt loss or renin secretion, and expression of NKCC2, Na(+)-Cl(-) cotransporter, and ENaC-ß were similar to those in control mice. When mice were placed on a salt-deficient diet, however, renin secretion and cortical expression of cyclooxygenase-2 mRNA increased significantly in ACC1-KI(S79A) mice compared with control mice. In summary, our data suggest that renin synthesis and secretion are regulated by AMPK and coupled to metabolism by phosphorylation of ACC1.


Subject(s)
AMP-Activated Protein Kinases/genetics , Acetyl-CoA Carboxylase/metabolism , Renin/blood , AMP-Activated Protein Kinases/deficiency , Acetyl-CoA Carboxylase/genetics , Animals , Epithelial Sodium Channels/biosynthesis , Mice , Phosphorylation , Renin/biosynthesis , Sodium/urine , Sodium Chloride, Dietary/administration & dosage
10.
Nephrology (Carlton) ; 18(5): 351-5, 2013 May.
Article in English | MEDLINE | ID: mdl-23600369

ABSTRACT

CD39 (NTPDase1), a critical immune and vascular ecto-nucleotidase, hydrolyses pro-inflammatory and pro-thrombotic nucleotides (adenosine-5'-triphosphate (ATP) and adenosine diphosphate) to adenosine. In humans, CD39 is the dominant ecto-nucleotidase in placental trophoblastic tissues and modulates ATP-dependent trophoblastic functions. CD39 is an integral component of regulatory T cells (Treg), which are central to immunological tolerance and maintenance of normal pregnancy. We examined the impact of CD39 overexpression in a mouse model of preeclampsia. Matings were performed between virginal BALB/c female (wild-type (WT) or CD39 transgenic (CD39TG)) and C57BL/6 male mice. On days 10 and 12 of pregnancy BALB/c Th1-polarized cells were injected. Systolic blood pressure (SBP) was measured throughout pregnancy. Mice were sacrificed at day 15 of pregnancy. Following transfer of Th1-polarized cells, SBP of pregnant WT mice increased (118 ± 3 mmHg to 142 ± 5 mmHg). Although ultrastructural changes were evident in the kidney this was not accompanied by significant proteinuria. SBP remained unchanged (115 ± 2 mmHg to 114 ± 3 mmHg) in pregnant CD39TG mice without evidence of renal lesions. We conclude that gestational hypertension can be induced in mice following transfer of maternally derived Th1-polarized cells and that overexpression of CD39 is protective in this model.


Subject(s)
Antigens, CD/physiology , Apyrase/physiology , Pre-Eclampsia/prevention & control , Animals , Cell Polarity , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pregnancy , Systole , Th1 Cells/physiology
11.
Diabetes ; 62(6): 2026-35, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23364452

ABSTRACT

Islet allograft survival limits the long-term success of islet transplantation as a potential curative therapy for type 1 diabetes. A number of factors compromise islet survival, including recurrent diabetes. We investigated whether CD39, an ectonucleotidase that promotes the generation of extracellular adenosine, would mitigate diabetes in the T cell-mediated multiple low-dose streptozotocin (MLDS) model. Mice null for CD39 (CD39KO), wild-type mice (WT), and mice overexpressing CD39 (CD39TG) were subjected to MLDS. Adoptive transfer experiments were performed to delineate the efficacy of tissue-restricted overexpression of CD39. The role of adenosine signaling was examined using mutant mice and pharmacological inhibition. The susceptibility to MLDS-induced diabetes was influenced by the level of expression of CD39. CD39KO mice developed diabetes more rapidly and with higher frequency than WT mice. In contrast, CD39TG mice were protected. CD39 overexpression conferred protection through the activation of adenosine 2A receptor and adenosine 2B receptor. Adoptive transfer experiments indicated that tissue-restricted overexpression of CD39 conferred robust protection, suggesting that this may be a useful strategy to protect islet grafts from T cell-mediated injury.


Subject(s)
Antigens, CD/metabolism , Apyrase/metabolism , Diabetes Mellitus, Experimental/metabolism , Animals , Antigens, CD/genetics , Apyrase/genetics , Diabetes Mellitus, Experimental/genetics , Flow Cytometry , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Receptors, Purinergic P1/genetics , Receptors, Purinergic P1/metabolism
12.
J Biomed Biotechnol ; 2012: 320495, 2012.
Article in English | MEDLINE | ID: mdl-23118504

ABSTRACT

Diabetes mellitus encompasses two distinct disease processes: autoimmune Type 1 (T1D) and nonimmune Type 2 (T2D) diabetes. Despite the disparate aetiologies, the disease phenotype of hyperglycemia and the associated complications are similar. In this paper, we discuss the role of the CD39-adenosinergic axis in the pathogenesis of both T1D and T2D, with particular emphasis on the role of CD39 and CD73.


Subject(s)
Adenosine/metabolism , Antigens, CD/metabolism , Apyrase/metabolism , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Signal Transduction , Animals , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/pathology , Humans , Pancreas/enzymology , Pancreas/pathology
13.
Eur J Immunol ; 42(9): 2441-51, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22684996

ABSTRACT

CD39 is an ecto-enzyme that degrades extracellular nucleotides, such as ATP, and is highly expressed on by the vasculature and circulating cells including Foxp3+ regulatory T (Treg) cells. To study the role of purinergic regulation in renal disease, we used the adriamycin nephropathy (AN) mouse model of chronic renal injury, using human CD39-transgenic (hCD39Tg) and wild-type (WT) BALB/c mice. Effects of CD39 expression by Treg cells were assessed in AN by adoptive transfer of CD4(+) CD25(+) and CD4(+) CD25(-) T cells isolated from hCD39Tg and WT mice. hCD39Tg mice were protected from renal injury in AN with decreased urinary protein and serum creatinine, and significantly less renal injury compared with WT mice. While WT CD25(+) and hCD39Tg CD25(-) T cells conferred some protection against AN, hCD39Tg CD25(+) Treg cells offered greater protection. In vitro studies showed direct pro-apoptotic effects of ATP on renal tubular cells. In conclusion, hCD39 expressed by circulating leukocytes and intrinsic renal cells limits innate AN injury. Specifically, CD39 expression by Treg cells contributes to its protective role in renal injury. These findings suggest that extracellular nucleotides mediate AN kidney injury and that CD39, expressed by Treg cells and other cells, is protective in this model.


Subject(s)
Antigens, CD/immunology , Antigens, CD/metabolism , Apyrase/immunology , Apyrase/metabolism , Kidney Diseases/immunology , T-Lymphocytes, Regulatory/immunology , Adenosine Triphosphate/immunology , Adenosine Triphosphate/metabolism , Animals , Apoptosis/immunology , Creatinine/immunology , Creatinine/metabolism , Disease Models, Animal , Doxorubicin , Humans , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Kidney Tubules/immunology , Kidney Tubules/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism
14.
J Immunol ; 174(10): 6250-6, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15879123

ABSTRACT

Factor H-related protein 5 (FHR-5) is a recently discovered member of the factor H (fH)-related protein family. FHR proteins are structurally similar to the complement regulator fH, but their biological functions remain poorly defined. FHR-5 is synthesized in the liver and consists of 9 short consensus repeats (SCRs), which display various degrees of homology to those of fH and the other FHR proteins. FHR-5 colocalizes with complement deposits in vivo and binds C3b in vitro, suggesting a role in complement regulation or localization. The current study examined whether rFHR-5 exhibits properties similar to those of fH, including heparin binding, CRP binding, cofactor activity for the factor I-mediated degradation of C3b and decay acceleration of the C3 convertase. rFHR-5 bound heparin-BSA and heparin-agarose and a defined series of truncations expressed in Pichia pastoris localized the heparin-binding region to within SCRs 5-7. rFHR-5 bound CRP, and this binding was also localized to SCRs 5-7. FHR-5 inhibited alternative pathway C3 convertase activity in a fluid phase assay; however, dissociation of the convertase was not observed in a solid phase assay. rFHR-5 displayed factor I-dependent cofactor activity for C3b cleavage, although it was apparently less effective than fH. In addition, we demonstrate association of FHR-5 with high density lipid lipoprotein complexes in human plasma. These results demonstrate that FHR-5 shares properties of heparin and CRP binding and lipoprotein association with one or more of the other FHRs but is unique among this family of proteins in possessing independent complement-regulatory activity.


Subject(s)
Blood Proteins/physiology , C-Reactive Protein/metabolism , Complement C3-C5 Convertases/antagonists & inhibitors , Complement Factor H/physiology , Complement Inactivator Proteins/physiology , Heparin/metabolism , Lipoproteins, HDL/metabolism , Blood Proteins/biosynthesis , Blood Proteins/genetics , Blood Proteins/metabolism , Complement C3-C5 Convertases/metabolism , Complement C3b/metabolism , Complement Factor H/metabolism , Complement Inactivator Proteins/biosynthesis , Complement Inactivator Proteins/genetics , Complement Inactivator Proteins/metabolism , Complement System Proteins , Consensus Sequence , Fibrinogen/physiology , Humans , Hydrolysis , Lipoproteins, HDL/blood , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Pichia/genetics , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Repetitive Sequences, Amino Acid
15.
Genetica ; 114(2): 157-61, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12041828

ABSTRACT

The factor H family of genes has been localised to human chromosome 1q32. This region encodes various proteins involved in complement regulation and is known as the regulators of complement activation (RCA) gene cluster. The factor H genes encode seven known plasma proteins. Using fluorescence in situ hybridisation (FISH), radiation hybrid (RH) mapping and BLAST alignment analysis, we have established that the factor H-related 5 (FHR-5) gene is closely linked to the other factor H gene family members. Analysis of the genomic sequence indicates that the FHR-5 gene is situated between FHR-2 and the non-complement protein factor XIIIb (Fl3B). Like all members of the factor H family. transcription of FHR-5 is in the telomeric direction. Furthermore, the short consensus repeats (SCRs) of FHR-5 are encoded by individual exons and splicing is of type 1. These data allow the generation of a more complete map of the factor H gene family.


Subject(s)
Blood Proteins/genetics , Chromosomes, Human, Pair 1 , Base Sequence , Complement System Proteins , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Radiation Hybrid Mapping , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...