Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 160
Filter
Add more filters

Publication year range
1.
Cell ; 185(26): 4887-4903.e17, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36563662

ABSTRACT

Our bodies turn over billions of cells daily via apoptosis and are in turn cleared by phagocytes via the process of "efferocytosis." Defects in efferocytosis are now linked to various inflammatory diseases. Here, we designed a strategy to boost efferocytosis, denoted "chimeric receptor for efferocytosis" (CHEF). We fused a specific signaling domain within the cytoplasmic adapter protein ELMO1 to the extracellular phosphatidylserine recognition domains of the efferocytic receptors BAI1 or TIM4, generating BELMO and TELMO, respectively. CHEF-expressing phagocytes display a striking increase in efferocytosis. In mouse models of inflammation, BELMO expression attenuates colitis, hepatotoxicity, and nephrotoxicity. In mechanistic studies, BELMO increases ER-resident enzymes and chaperones to overcome protein-folding-associated toxicity, which was further validated in a model of ER-stress-induced renal ischemia-reperfusion injury. Finally, TELMO introduction after onset of kidney injury significantly reduced fibrosis. Collectively, these data advance a concept of chimeric efferocytic receptors to boost efferocytosis and dampen inflammation.


Subject(s)
Macrophages , Phagocytosis , Animals , Mice , Macrophages/metabolism , Inflammation/metabolism , Phagocytes/metabolism , Carrier Proteins/metabolism , Apoptosis , Adaptor Proteins, Signal Transducing/metabolism
2.
Circ Res ; 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38957990

ABSTRACT

BACKGROUND: PANX1 (pannexin 1), a ubiquitously expressed ATP release membrane channel, has been shown to play a role in inflammation, blood pressure regulation, and myocardial infarction. However, the possible role of PANX1 in cardiomyocytes in the progression of heart failure has not yet been investigated. METHOD: We generated a novel mouse line with constitutive deletion of PANX1 in cardiomyocytes (Panx1MyHC6). RESULTS: PANX1 deletion in cardiomyocytes had no effect on unstressed heart function but increased the glycolytic metabolism and resulting glycolytic ATP production, with a concurrent decrease in oxidative phosphorylation, both in vivo and in vitro. In vitro, treatment of H9c2 cardiomyocytes with isoproterenol led to PANX1-dependent release of ATP and Yo-Pro-1 uptake, as assessed by pharmacological blockade with spironolactone and siRNA-mediated knockdown of PANX1. To investigate nonischemic heart failure and the preceding cardiac hypertrophy, we administered isoproterenol, and we demonstrated that Panx1MyHC6 mice were protected from systolic and diastolic left ventricle volume increases as a result of cardiomyocyte hypertrophy. Moreover, we found that Panx1MyHC6 mice showed decreased isoproterenol-induced recruitment of immune cells (CD45+), particularly neutrophils (CD11b+, Ly6g+), to the myocardium. CONCLUSIONS: Together, these data demonstrate that PANX1 deficiency in cardiomyocytes increases glycolytic metabolism and protects against cardiac hypertrophy in nonischemic heart failure at least in part by reducing immune cell recruitment. Our study implies PANX1 channel inhibition as a therapeutic approach to ameliorate cardiac dysfunction in patients with heart failure.

3.
Am J Physiol Renal Physiol ; 326(2): F167-F177, 2024 02 01.
Article in English | MEDLINE | ID: mdl-37969103

ABSTRACT

This study aimed to investigate the role of bone marrow stromal cell antigen-1 (Bst1; also known as CD157) in acute kidney injury (AKI). Bst1 is a cell surface molecule with various enzymatic activities and downstream intracellular signaling pathways that modulate the immune response. Previous research has linked Bst1 to diseases such as ovarian cancer, Parkinson's disease, and rheumatoid arthritis. We used bilateral ischemia-reperfusion injury (IRI) as an AKI model and created bone marrow chimeric mice to evaluate the role of Bst1 in bone marrow-derived cells. We also used flow cytometry to identify Bst1/CD157 expression in hematopoietic cells and evaluate immune cell dynamics in the kidney. The findings showed that Bst1-deficient (Bst1-/-) mice were protected against renal bilateral IRI. Bone marrow chimera experiments revealed that Bst1 expression on hematopoietic cells, but not parenchymal cells, induced renal IRI. Bst1 was mainly found in B cells and neutrophils by flow cytometry of the spleen and bone marrow. In vitro, migration of neutrophils from Bst1-/- mice was suppressed, and adoptive transfer of neutrophils from wild-type Bst1+/+ mice abolished the renal protective effect in Bst1 knockout mice. In conclusion, the study demonstrated that Bst1-/- mice are protected against renal IRI and that Bst1 expression in neutrophils plays a crucial role in inducing renal IRI. These findings suggest that targeting Bst1 in neutrophils could be a potential therapeutic strategy for AKI.NEW & NOTEWORTHY Acute kidney injury (AKI), a serious disease for which there is no effective Federal Drug Administration-approved treatment, is associated with high mortality rates. Bone marrow stromal cell antigen-1 (Bst1) is a cell surface molecule that can cause kidney fibrosis, but its role in AKI is largely unknown. Our study showed that Bst1-/- mice revealed a protective effect against renal bilateral ischemia-reperfusion injury (IRI). Adoptive transfer studies confirmed that Bst1 expression in hematopoietic cells, especially neutrophils, contributed to renal bilateral IRI.


Subject(s)
Acute Kidney Injury , Mesenchymal Stem Cells , Reperfusion Injury , Mice , Animals , Acute Kidney Injury/genetics , Acute Kidney Injury/prevention & control , Kidney/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/prevention & control , Neutrophils/metabolism , Mice, Knockout , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL
4.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Article in English | MEDLINE | ID: mdl-33737395

ABSTRACT

Acute kidney injury is highly prevalent and associated with high morbidity and mortality, and there are no approved drugs for its prevention and treatment. Vagus nerve stimulation (VNS) alleviates inflammatory diseases including kidney disease; however, neural circuits involved in VNS-induced tissue protection remain poorly understood. The vagus nerve, a heterogeneous group of neural fibers, innervates numerous organs. VNS broadly stimulates these fibers without specificity. We used optogenetics to selectively stimulate vagus efferent or afferent fibers. Anterograde efferent fiber stimulation or anterograde (centripetal) sensory afferent fiber stimulation both conferred kidney protection from ischemia-reperfusion injury. We identified the C1 neurons-sympathetic nervous system-splenic nerve-spleen-kidney axis as the downstream pathway of vagus afferent fiber stimulation. Our study provides a map of the neural circuits important for kidney protection induced by VNS, which is critical for the safe and effective clinical application of VNS for protection from acute kidney injury.


Subject(s)
Acute Kidney Injury/etiology , Disease Susceptibility , Neuroimmunomodulation , Spleen/immunology , Spleen/innervation , Vagus Nerve Stimulation , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Mice , Neurons , Sympathetic Nervous System/physiology
5.
Kidney Int ; 100(3): 613-620, 2021 09.
Article in English | MEDLINE | ID: mdl-34224760

ABSTRACT

Microcirculatory changes and oxidative stress have long been associated with acute kidney injury. Despite substantial progress made by two-photon microscopy of microvascular responses to acute kidney injury in rodent models, little is known about the underlying changes in blood oxygen delivery and tissue oxygen metabolism. To fill this gap, we developed a label-free kidney imaging technique based on photoacoustic microscopy, which enables simultaneous quantification of hemoglobin concentration, oxygen saturation of hemoglobin, and blood flow in peritubular capillaries in vivo. Based on these microvascular parameters, microregional oxygen metabolism was quantified. We demonstrated the utility of this technique by studying kidney hemodynamic and oxygen-metabolic responses to acute kidney injury in mice subject to lipopolysaccharide-induced sepsis. Dynamic photoacoustic microscopy of the peritubular capillary function and tissue oxygen metabolism revealed that sepsis induced an acute and significant reduction in peritubular capillary oxygen saturation of hemoglobin, concomitant with a marked reduction in kidney ATP levels and contrasted with nominal changes in peritubular capillary flow and plasma creatinine. Thus, our technique opens new opportunities to study microvascular and metabolic dysfunction in acute and chronic kidney diseases.


Subject(s)
Capillaries , Microscopy , Animals , Kidney , Mice , Microcirculation , Oxygen
6.
Am J Kidney Dis ; 77(1): 122-131, 2021 01.
Article in English | MEDLINE | ID: mdl-33077315

ABSTRACT

Recent advances in glomerular biology have expanded our understanding of glomerular diseases, leading to more precise therapeutic options. Since the discovery of the autoantigen phospholipase A2 receptor in primary membranous nephropathy 10 years ago, the serologic evaluation of glomerular diseases has become more detailed and nuanced for nephrologists. In addition to phospholipase A2 receptor antibodies, circulating autoantibodies now include thrombospondin type 1 domain-containing 7A and most recently, neural epidermal growth factor-like 1 protein for membranous nephropathy. Additionally, discoveries in C3 glomerulopathy and fibrillary glomerulonephritis are poised to improve the diagnostic approach to these disorders by using novel biomarkers to complement traditional histologic patterns on kidney biopsy. Although kidney biopsies are considered the gold standard in profiling glomerular diseases, validated novel glomerular biomarkers contribute substantially to the diagnostic and therapeutic approaches through their ability to improve sensitivity, permit dynamic longitudinal monitoring of disease activity, and capture genetic heterogeneity. We describe the value of specific biomarkers in selected glomerular diseases, with the major focus on their clinical applicability.


Subject(s)
Biological Factors/blood , Glomerulonephritis , Biomarkers/blood , Glomerulonephritis/classification , Glomerulonephritis/diagnosis , Glomerulonephritis/immunology , Glomerulonephritis/metabolism , Humans , Reproducibility of Results
7.
Am J Physiol Renal Physiol ; 319(1): F125-F138, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32508112

ABSTRACT

Acute kidney injury (AKI) is an important clinical disorder with high prevalence, serious consequences, and limited therapeutic options. Modulation of neuroimmune interaction by nonpharmacological methods is emerging as a novel strategy for treating inflammatory diseases, including AKI. Recently, pulsed ultrasound (US) treatment was shown to protect from AKI by stimulating the cholinergic anti-inflammatory pathway. Because of the relatively simple, portable, and noninvasive nature of US procedures, US stimulation may be a valuable therapeutic option for treating inflammatory conditions. This review discusses potential impacts of US bioeffects on the nervous system and how this may generate feedback onto the immune system. We also discuss recent evidence supporting the use of US as a means to treat AKI and other inflammatory diseases.


Subject(s)
Acute Kidney Injury/therapy , Neuroimmunomodulation , Ultrasonography, Interventional/methods , Humans , Inflammation/therapy
8.
Am J Physiol Renal Physiol ; 319(4): F603-F611, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32830538

ABSTRACT

The acyl-CoA synthetase medium-chain family member 2 (Acsm2) gene was first identified and cloned by our group as a kidney-specific "KS" gene. However, its expression pattern and function remain to be clarified. In the present study, we found that the Acsm2 gene was expressed specifically and at a high level in normal adult kidneys. Expression of Acsm2 in kidneys followed a maturational pattern: it was low in newborn mice and increased with kidney development and maturation. In situ hybridization and immunohistochemistry revealed that Acsm2 was expressed specifically in proximal tubular cells of adult kidneys. Data from the Encyclopedia of DNA Elements database revealed that the Acsm2 gene locus in the mouse has specific histone modifications related to the active transcription of the gene exclusively in kidney cells. Following acute kidney injury, partial unilateral ureteral obstruction, and chronic kidney diseases, expression of Acsm2 in the proximal tubules was significantly decreased. In human samples, the expression pattern of ACSM2A, a homolog of mouse Acsm2, was similar to that in mice, and its expression decreased with several types of renal injuries. These results indicate that the expression of Acsm2 parallels the structural and functional maturation of proximal tubular cells. Downregulation of its expression in several models of kidney disease suggests that Acms2 may serve as a novel marker of proximal tubular injury and/or dysfunction.


Subject(s)
Coenzyme A Ligases/metabolism , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , Mitochondrial Proteins/metabolism , Acute Kidney Injury/enzymology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Coenzyme A Ligases/genetics , Disease Models, Animal , Epithelial Cells/pathology , Fibrosis , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Humans , Integrin beta1/genetics , Integrin beta1/metabolism , Kidney Tubules, Proximal/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/genetics , Renal Insufficiency, Chronic/enzymology , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Renin/genetics , Renin/metabolism , Reperfusion Injury/enzymology , Reperfusion Injury/genetics , Reperfusion Injury/pathology
9.
Kidney Int ; 97(3): 466-476, 2020 03.
Article in English | MEDLINE | ID: mdl-32001065

ABSTRACT

Under physiological states, the nervous system and the kidneys communicate with each other to maintain normal body homeostasis. However, pathological states disrupt this interaction as seen in hypertension, and kidney damage can cause impaired renorenal reflex and sodium handling. In acute kidney injury (AKI) and chronic kidney disease (CKD), damaged kidneys can have a detrimental effect on the central nervous system. CKD is an independent risk factor for cerebrovascular disease and cognitive impairment, and many factors, including retention of uremic toxins and phosphate, have been proposed as CKD-specific factors responsible for structural and functional cerebral changes in patients with CKD. However, more studies are needed to determine the precise pathogenesis. Epidemiological studies have shown that AKI is associated with a subsequent risk for developing stroke and dementia. However, recent animal studies have shown that the renal nerve contributes to kidney inflammation and fibrosis, whereas activation of the cholinergic anti-inflammatory pathway, which involves the vagus nerve, the splenic nerve, and immune cells in the spleen, has a significant renoprotective effect. Therefore, elucidating mechanisms of communication between the nervous system and the kidney enables us not only to develop new strategies to ameliorate neurological conditions associated with kidney disease but also to design safe and effective clinical interventions for kidney disease, using the neural and neuroimmune control of kidney injury and disease.


Subject(s)
Acute Kidney Injury , Renal Insufficiency, Chronic , Reperfusion Injury , Acute Kidney Injury/pathology , Animals , Fibrosis , Humans , Kidney/pathology , Nervous System , Renal Insufficiency, Chronic/pathology , Reperfusion Injury/pathology
10.
Am J Physiol Renal Physiol ; 317(3): F658-F669, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31364375

ABSTRACT

Progressive tubulointerstitial fibrosis may occur after acute kidney injury due to persistent inflammation. Purinergic signaling by 5'-ectonucleotidase, CD73, an enzyme that converts AMP to adenosine on the extracellular surface, can suppress inflammation. The role of CD73 in progressive kidney fibrosis has not been elucidated. We evaluated the effect of deletion of CD73 from kidney perivascular cells (including pericytes and/or fibroblasts of the Foxd1+ lineage) on fibrosis. Perivascular cell expression of CD73 was necessary to suppress inflammation and prevent kidney fibrosis in Foxd1CreCD73fl/fl mice evaluated 14 days after unilateral ischemia-reperfusion injury or folic acid treatment (250 mg/kg). Kidneys of Foxd1CreCD73fl/fl mice had greater collagen deposition, expression of proinflammatory markers (including various macrophage markers), and platelet-derived growth factor recepetor-ß immunoreactivity than CD73fl/fl mice. Kidney dysfunction and fibrosis were rescued by administration of soluble CD73 or by macrophage deletion. Isolated CD73-/- kidney pericytes displayed an activated phenotype (increased proliferation and α-smooth muscle actin mRNA expression) compared with wild-type controls. In conclusion, CD73 in perivascular cells may act to suppress myofibroblast transformation and influence macrophages to promote a wound healing response. These results suggest that the purinergic signaling pathway in the kidney interstitial microenvironment orchestrates perivascular cells and macrophages to suppress inflammation and prevent progressive fibrosis.


Subject(s)
5'-Nucleotidase/metabolism , Cellular Microenvironment , Fibroblasts/metabolism , Kidney/metabolism , Macrophages/metabolism , Nephritis, Interstitial/metabolism , Pericytes/metabolism , Reperfusion Injury/metabolism , 5'-Nucleotidase/deficiency , 5'-Nucleotidase/genetics , Actins/metabolism , Animals , Cell Proliferation , Cells, Cultured , Collagen/metabolism , Disease Models, Animal , Fibroblasts/pathology , Fibrosis , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Inflammation Mediators/metabolism , Kidney/immunology , Kidney/pathology , Macrophages/pathology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Nephritis, Interstitial/genetics , Nephritis, Interstitial/immunology , Nephritis, Interstitial/pathology , Pericytes/pathology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Signal Transduction , Wound Healing
11.
Kidney Int ; 95(3): 563-576, 2019 03.
Article in English | MEDLINE | ID: mdl-30670317

ABSTRACT

The cholinergic anti-inflammatory pathway (CAP) links the nervous and immune systems and modulates innate and adaptive immunity. Activation of the CAP by vagus nerve stimulation exerts protective effects in a wide variety of clinical disorders including rheumatoid arthritis and Crohn's disease, and in murine models of acute kidney injury including ischemia/reperfusion injury (IRI). The canonical CAP pathway involves activation of splenic alpha7-nicotinic acetylcholine receptor (α7nAChR)-positive macrophages by splenic ß2-adrenergic receptor-positive CD4+ T cells. Here we demonstrate that ultrasound or vagus nerve stimulation also activated α7nAChR-positive peritoneal macrophages, and that adoptive transfer of these activated peritoneal macrophages reduced IRI in recipient mice. The protective effect required α7nAChR, and did not occur in splenectomized mice or in mice lacking T and B cells, suggesting a bidirectional interaction between α7nAChR-positive peritoneal macrophages and other immune cells including ß2-adrenergic receptor-positive CD4+ T cells. We also found that expression of hairy and enhancer of split-1 (Hes1), a basic helix-loop-helix DNA-binding protein, is induced in peritoneal macrophages by ultrasound or vagus nerve stimulation. Adoptive transfer of Hes1-overexpressing peritoneal macrophages reduced kidney IRI. Our data suggest that Hes1 is downstream of α7nAChR and is important to fully activate the CAP. Taken together, these results suggest that peritoneal macrophages play a previously unrecognized role in mediating the protective effect of CAP activation in kidney injury, and that Hes1 is a new candidate pharmacological target to activate the CAP.


Subject(s)
Acute Kidney Injury/immunology , Macrophages, Peritoneal/immunology , Reperfusion Injury/immunology , Transcription Factor HES-1/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/therapy , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/radiation effects , CD4-Positive T-Lymphocytes/transplantation , Disease Models, Animal , Gene Knockdown Techniques , Humans , Macrophage Activation , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/transplantation , Male , Mice , Neuroimmunomodulation/radiation effects , RAW 264.7 Cells , Reperfusion Injury/pathology , Reperfusion Injury/therapy , Transcription Factor HES-1/genetics , Transcription Factor HES-1/immunology , Ultrasonic Therapy , Up-Regulation/radiation effects , Vagus Nerve Stimulation , alpha7 Nicotinic Acetylcholine Receptor/immunology
12.
J Immunol ; 198(7): 2589-2601, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28219886

ABSTRACT

Glomerular damage mediated by glomerulus-infiltrating myeloid-derived cells is a key pathogenic event in lupus nephritis (LN), but the process is poorly understood. Confocal microscopy of kidney sections and flow cytometry analysis of glomerular cells from magnetic bead-purified glomeruli have identified glomerulus-infiltrating leukocyte populations in NZM2328 (NZM) lupus-prone mice with spontaneous chronic glomerulonephritis (GN) and anti-glomerular basement membrane-induced nephritis. The occurrence of a major glomerulus-infiltrating CD11b+F4/80-I-A- macrophage population exhibiting the markers programmed death ligand-1 (PD-L1), Mac-2, and macrophage mannose receptor (CD206) and producing Klf4, Il10, Retnla, Tnf, and Il6 mRNA, which are known to be expressed by alternatively activated (M2b) macrophages, correlated with proteinuria status. In NZM mice with spontaneous LN, glomerular macrophage infiltration is predominant. CD11b+F4/80-I-A- intraglomerular macrophages and polymorphonuclear neutrophils (PMN) are important in inducing GN, as anti-CD11b and -ICAM-1 mAb inhibited both proteinuria and macrophage and PMN infiltration. The predominant and high expression of PD-L1 by CD11b+F4/80-I-A- glomerular macrophages in kidneys of mice with GN and the inhibition of proteinuria by anti-PD-L1 mAb supported the pathogenic role of these macrophages but not the PD-L1- PMN in GN development and in inducing podocyte damage. In NZM mice with spontaneous chronic GN and severe proteinuria, few glomerulus-infiltrating PMN were found, leaving macrophages and, to a less extent, dendritic cells as the major infiltrating leukocytes. Taken together, these data support the important pathogenic effect of CD11b+F4/80-I-A- M2b-like glomerulus-infiltrating macrophages in LN and reinforce macrophages as a promising target for GN treatment.


Subject(s)
Kidney Glomerulus/immunology , Lupus Nephritis/immunology , Macrophages/immunology , Animals , B7-H1 Antigen/immunology , Bone Marrow Cells/immunology , Cell Separation , Disease Models, Animal , Female , Flow Cytometry , Kidney Glomerulus/pathology , Kruppel-Like Factor 4 , Lupus Nephritis/pathology , Macrophage-1 Antigen/immunology , Macrophages/pathology , Mice , Mice, Mutant Strains , Microscopy, Confocal , Real-Time Polymerase Chain Reaction
13.
J Am Soc Nephrol ; 29(1): 194-206, 2018 01.
Article in English | MEDLINE | ID: mdl-29084809

ABSTRACT

The proximal tubule epithelium relies on mitochondrial function for energy, rendering the kidney highly susceptible to ischemic AKI. Dynamin-related protein 1 (DRP1), a mediator of mitochondrial fission, regulates mitochondrial function; however, the cell-specific and temporal role of DRP1 in AKI in vivo is unknown. Using genetic murine models, we found that proximal tubule-specific deletion of Drp1 prevented the renal ischemia-reperfusion-induced kidney injury, inflammation, and programmed cell death observed in wild-type mice and promoted epithelial recovery, which associated with activation of the renoprotective ß-hydroxybutyrate signaling pathway. Loss of DRP1 preserved mitochondrial structure and reduced oxidative stress in injured kidneys. Lastly, proximal tubule deletion of DRP1 after ischemia-reperfusion injury attenuated progressive kidney injury and fibrosis. These results implicate DRP1 and mitochondrial dynamics as an important mediator of AKI and progression to fibrosis and suggest that DRP1 may serve as a therapeutic target for AKI.


Subject(s)
3-Hydroxybutyric Acid/metabolism , Acute Kidney Injury/genetics , Dynamins/genetics , Kidney Tubules, Proximal/pathology , Mitochondria/metabolism , Renal Insufficiency, Chronic/genetics , Acute Kidney Injury/etiology , Animals , Apoptosis/genetics , Disease Progression , Dynamins/antagonists & inhibitors , Fibrosis , Male , Mice , Mice, Knockout , Mitochondria/ultrastructure , Mitochondrial Dynamics/genetics , Nephritis/etiology , Nephritis/genetics , Oxidative Stress/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Reperfusion Injury/complications , Signal Transduction
14.
J Am Soc Nephrol ; 29(7): 1887-1899, 2018 07.
Article in English | MEDLINE | ID: mdl-29866797

ABSTRACT

Background Pannexin1 (Panx1), an ATP release channel, is present in most mammalian tissues, but the role of Panx1 in health and disease is not fully understood. Panx1 may serve to modulate AKI; ATP is a precursor to adenosine and may function to block inflammation, or ATP may act as a danger-associated molecular pattern and initiate inflammation.Methods We used pharmacologic and genetic approaches to evaluate the effect of Panx1 on kidney ischemia-reperfusion injury (IRI), a mouse model of AKI.Results Pharmacologic inhibition of gap junctions, including Panx1, by administration of carbenoxolone protected mice from IRI. Furthermore, global deletion of Panx1 preserved kidney function and morphology and diminished the expression of proinflammatory molecules after IRI. Analysis of bone marrow chimeric mice revealed that Panx1 expressed on parenchymal cells is necessary for ischemic injury, and both proximal tubule and vascular endothelial Panx1 tissue-specific knockout mice were protected from IRI. In vitro, Panx1-deficient proximal tubule cells released less and retained more ATP under hypoxic stress.Conclusions Panx1 is involved in regulating ATP release from hypoxic cells, and reducing this ATP release may protect kidneys from AKI.


Subject(s)
Acute Kidney Injury/metabolism , Connexins/antagonists & inhibitors , Connexins/genetics , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Acute Kidney Injury/etiology , Acute Kidney Injury/prevention & control , Adenosine Triphosphate/metabolism , Animals , Anti-Ulcer Agents/pharmacology , Bone Marrow Cells/metabolism , Carbenoxolone/pharmacology , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Endothelium, Vascular , Epithelial Cells/metabolism , Intercellular Adhesion Molecule-1/metabolism , Male , Mice , Mice, Knockout , RNA, Messenger/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control
15.
Kidney Blood Press Res ; 43(2): 555-567, 2018.
Article in English | MEDLINE | ID: mdl-29642060

ABSTRACT

BACKGROUND/AIMS: The metabolic syndrome (MetS), as assessed using dichotomous criteria, is associated with increased risk of future chronic kidney disease (CKD), though this relationship is unclear among African Americans, who have lower risk for MetS but higher risk for CKD. METHODS: We performed logistic regression using a sex- and race-specific MetS-severity z-score to assess risk of incident CKD among 2,627 African-American participants of the Jackson Heart Study, assessed at baseline and 8 years later. Based on quartile of baseline MetS severity, we further assessed prevalence of being in the lowest quartile of baseline GFR, the lowest quartile of relative GFR at follow-up, microalbuminuria and incident CKD. RESULTS: Higher MetS-severity was associated with higher prevalence of GFR in the lowest quartile at baseline among males and females. Among African-American females but not males, higher baseline MetS-severity was associated with a higher prevalence of baseline elevations in microabuminuria (p<0.01), steep decline in GFR (p<0.001) and a higher incidence of CKD (p<0.0001). Women in increasing quartiles of baseline MetS-severity exhibited a linear trend toward higher odds of future CKD (p<0.05), with those in the 4th quartile of MetS-severity (compared to the 1st) having an odds ratio of 2.47 (95% confidence interval 1.13, 5.37); no such relationship was seen among men (p value for trend 0.49). CONCLUSION: MetS-severity exhibited sex-based interactions regarding risk for future GFR deterioration and CKD, with increasing risk in women but not men. These data may have implications for triggering CKD screening among African-American women with higher degrees of MetS-severity.


Subject(s)
Glomerular Filtration Rate , Metabolic Syndrome/diagnosis , Renal Insufficiency, Chronic/etiology , Severity of Illness Index , Adult , Black or African American , Aged , Albuminuria , Female , Follow-Up Studies , Humans , Incidence , Logistic Models , Male , Metabolic Syndrome/complications , Metabolic Syndrome/ethnology , Metabolic Syndrome/physiopathology , Middle Aged , Racial Groups , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/ethnology , Risk Factors , Sex Factors , United States/epidemiology
16.
J Am Soc Nephrol ; 28(9): 2681-2693, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28539382

ABSTRACT

CD4+Foxp3+ regulatory T cells (Tregs) protect the kidney during AKI. We previously found that IL-2, which is critical for Treg homeostasis, upregulates the IL-33 receptor (ST2) on CD4+ T cells, thus we hypothesized that IL-2 and IL-33 cooperate to enhance Treg function. We found that a major subset of Tregs in mice express ST2, and coinjection of IL-2 and IL-33 increased the number of Tregs in lymphoid organs and protected mice from ischemia-reperfusion injury (IRI) more efficiently than either cytokine alone. Accordingly, we generated a novel hybrid cytokine (IL233) bearing the activities of IL-2 and IL-33 for efficient targeting to Tregs. IL233 treatment increased the number of Tregs in blood and spleen and prevented IRI more efficiently than a mixture of IL-2 and IL-33. Injection of IL233 also increased the numbers of Tregs in renal compartments. Moreover, IL233-treated mice had fewer splenic Tregs and more Tregs in kidneys after IRI. In vitro, splenic Tregs from IL233-treated mice suppressed CD4+ T cell proliferation better than Tregs from saline-treated controls. IL233 treatment also improved the ability of isolated Tregs to inhibit IRI in adoptive transfer experiments and protected mice from cisplatin- and doxorubicin-induced nephrotoxic injury. Finally, treatment with IL233 increased the proportion of ST2-bearing innate lymphoid cells (ILC2) in blood and kidneys, and adoptive transfer of ILC2 also protected mice from IRI. Thus, the novel IL233 hybrid cytokine, which utilizes the cooperation of IL-2 and IL-33 to enhance Treg- and ILC2-mediated protection from AKI, bears strong therapeutic potential.


Subject(s)
Acute Kidney Injury/immunology , Acute Kidney Injury/prevention & control , Interleukin-2/pharmacology , Interleukin-33/pharmacology , Recombinant Fusion Proteins/pharmacology , Reperfusion Injury/immunology , Reperfusion Injury/prevention & control , T-Lymphocytes, Regulatory/drug effects , Acute Kidney Injury/chemically induced , Animals , CD4 Lymphocyte Count , Cell Proliferation , Cells, Cultured , Cisplatin/adverse effects , Coculture Techniques , Doxorubicin/adverse effects , Interleukin-1 Receptor-Like 1 Protein/blood , Interleukin-2/therapeutic use , Interleukin-33/therapeutic use , Kidney/immunology , Male , Mice , Recombinant Fusion Proteins/therapeutic use , Spleen/immunology
17.
J Am Soc Nephrol ; 28(3): 888-902, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27628903

ABSTRACT

CD73-derived adenosine plays an anti-inflammatory role in various organs. However, its role in renal ischemia-reperfusion injury (IRI) is controversial. We targeted CD73 mutant mice to determine the function of CD73 expressed by various renal cell types under mild IRI conditions. Mice with CD73 deletion in proximal tubules exhibited exacerbated IRI, comparable with that of CD73-/- mice compared with WT mice. Mice with CD73 deletions in other cell types, including cortical type 1 fibroblast-like cells, mesangial cells, macrophages, and dendritic cells, showed small or no increases in injury above control mice when subjected to threshold levels of ischemia. Results from adoptive transfer experiments between WT and CD73-/- mice and pharmacologic studies modulating enzymatic activity of CD73 and extracellular adenosine levels supported a critical role of adenosine generated by proximal tubule CD73 expression in abrogating IRI. Renal adenosine levels were lower before and after ischemia in CD73-deficient mice. However, reduction in total acid-extractable renal adenosine levels was inadequate to explain the marked difference in kidney injury in these CD73-deficient mice. Furthermore, CD73 inhibition and enzyme replacement studies showed no change in total kidney adenosine levels in treated mice compared with vehicle-treated controls. Protection from IRI in neutrophil-depleted WT recipients was sustained by repopulation with bone marrow neutrophils from WT mice but not by those lacking adenosine 2a receptors (from Adora2a-/- mice). These data support the thesis that local adenosine generated by cells at the injury site is critical for protection from IRI through bone marrow-derived adenosine 2a receptors.


Subject(s)
5'-Nucleotidase/physiology , Kidney/blood supply , Reperfusion Injury/etiology , Animals , Cells, Cultured , Kidney Tubules, Proximal , Male , Mice , Mice, Inbred C57BL
18.
J Am Soc Nephrol ; 28(4): 1145-1161, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27799486

ABSTRACT

Maladaptive repair after AKI may lead to progressive fibrosis and decline in kidney function. Sphingosine 1-phosphate has an important role in kidney injury and pleiotropic effects in fibrosis. We investigated the involvement of sphingosine kinase 1 and 2 (SphK1 and SphK2), which phosphorylate sphingosine to produce sphingosine 1-phosphate, in kidney fibrosis induced by folic acid (FA) or unilateral ischemia-reperfusion injury. Analysis of Masson trichrome staining and fibrotic marker protein and mRNA expression 14 days after AKI revealed that wild-type (WT) and Sphk1-/- mice exhibited more kidney fibrosis than Sphk2-/- mice. Furthermore, kidneys of FA-treated WT and Sphk1-/- mice had greater immune cell infiltration and expression of fibrotic and inflammatory markers than kidneys of FA-treated Sphk2-/- mice. In contrast, kidneys of Sphk2-/- mice exhibited greater expression of Ifng and IFN-γ-responsive genes (Cxcl9 and Cxcl10) than kidneys of WT or Sphk1-/- mice did at this time point. Splenic T cells from untreated Sphk2-/- mice were hyperproliferative and produced more IFN-γ than did those of WT or Sphk1-/- mice. IFN-γ blocking antibody administered to Sphk2-/- mice or deletion of Ifng (Sphk2-/-Ifng-/- mice) blocked the protective effect of SphK2 deficiency in fibrosis. Moreover, adoptive transfer of Sphk2-/- (but not Sphk2-/-Ifng-/- ) CD4 T cells into WT mice blocked FA-induced fibrosis. Finally, a selective SphK2 inhibitor blocked FA-induced kidney fibrosis in WT mice. These studies demonstrate that SphK2 inhibition may serve as a novel therapeutic approach for attenuating kidney fibrosis.


Subject(s)
Interferon-gamma/physiology , Kidney Diseases/enzymology , Kidney/enzymology , Kidney/pathology , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Animals , Fibrosis/enzymology , Fibrosis/etiology , Fibrosis/prevention & control , Kidney Diseases/prevention & control , Mice , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors
19.
J Immunol ; 195(11): 5215-26, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26519533

ABSTRACT

We have previously shown that polyclonal natural IgM protects mice from renal ischemia/reperfusion injury (IRI) by inhibiting the reperfusion inflammatory response. We hypothesized that a potential mechanism involved IgM modulation of dendritic cells (DC), as we observed high IgM binding to splenic DC. To test this hypothesis, we pretreated bone marrow-derived DC (BMDC) with polyclonal murine or human IgM prior to LPS activation and demonstrated that 0.5 × 10(6) IgM/LPS-pretreated BMDC, when injected into wild-type C57BL/6 mice 24 h before renal ischemia, protect mice from developing renal IRI. We show that this switching of LPS-activated BMDC to a regulatory phenotype requires modulation of BMDC function that is mediated by IgM binding to nonapoptotic BMDC receptors. Regulatory BMDC require IL-10 and programmed death 1 as well as downregulation of CD40 and p65 NF-κB phosphorylation to protect in renal IRI. Blocking the programmed death ligand 1 binding site just before i.v. injection of IgM/LPS-pretreated BMDC or using IL-10 knockout BMDC fails to induce protection. Similarly, IgM/LPS-pretreated BMDC are rendered nonprotective by increasing CD40 expression and phosphorylation of p65 NF-κB. How IgM/LPS regulatory BMDC suppress in vivo ischemia-induced innate inflammation remains to be determined. However, we show that suppression is dependent on other in vivo regulatory mechanisms in the host, that is, CD25(+) T cells, B cells, IL-10, and circulating IgM. There was no increase in Foxp3(+) regulatory T cells in the spleen either before or after renal IRI. Collectively, these findings show that natural IgM anti-leukocyte Abs can switch BMDC to a regulatory phenotype despite the presence of LPS that ordinarily induces BMDC maturation.


Subject(s)
Bone Marrow Cells/immunology , Dendritic Cells/immunology , Immunoglobulin M/immunology , Inflammation/immunology , Kidney/blood supply , Reperfusion Injury/prevention & control , Animals , B-Lymphocytes/immunology , CD40 Antigens/metabolism , Cells, Cultured , Interleukin-10/immunology , Lipopolysaccharides/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Programmed Cell Death 1 Receptor/immunology , Reperfusion Injury/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Transcription Factor RelA/metabolism
20.
Blood Purif ; 43(1-3): 68-77, 2017.
Article in English | MEDLINE | ID: mdl-27923227

ABSTRACT

As advances in Critical Care Medicine continue, critically ill patients are surviving despite the severity of their illness. The incidence of acute kidney injury (AKI) has increased, and its impact on clinical outcomes as well as medical expenditures has been established. The role, indications and technological advancements of renal replacement therapy (RRT) have evolved, allowing more effective therapies with less complications. With these changes, Critical Care Nephrology has become an established specialty, and ongoing collaborations between critical care physicians and nephrologist have improved education of multi-disciplinary team members and patient care in the ICU. Multidisciplinary programs to support these changes have been stablished in some hospitals to maximize the delivery of care, while other programs have continue to struggle in their ability to acquire the necessary resources to maximize outcomes, educate their staff, and develop quality initiatives to evaluate and drive improvements. Clearly, the role of the nephrologist in the ICU has evolved, and varies widely among institutions. This special article will provide insights that will hopefully optimize the role of the nephrologist as the leader of the acute care nephrology program, as clinician for critically ill patients, and as teacher for all members of the health care team.


Subject(s)
Intensive Care Units/organization & administration , Nephrologists/organization & administration , Nephrology/organization & administration , Practice Guidelines as Topic/standards , Acute Kidney Injury/therapy , Critical Care/methods , Critical Care/standards , Humans , Interprofessional Relations
SELECTION OF CITATIONS
SEARCH DETAIL