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










Publication year range
2.
J Am Soc Nephrol ; 34(10): 1629-1646, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37545036

ABSTRACT

SIGNIFICANCE STATEMENT: Activation of the type 1 IL-1 receptor (IL-1R1) triggers a critical innate immune signaling cascade that contributes to the pathogenesis of AKI. However, blockade of IL-1 signaling in AKI has not consistently demonstrated kidney protection. The current murine experiments show that IL-1R1 activation in the proximal tubule exacerbates toxin-induced AKI and cell death through local suppression of apolipoprotein M. By contrast, IL-1R1 activation in endothelial cells ameliorates AKI by restoring VEGFA-dependent endothelial cell viability. Using this information, future delivery strategies can maximize the protective effects of blocking IL-1R1 while mitigating unwanted actions of IL-1R1 manipulation. BACKGROUND: Activation of the type 1 IL-1 receptor (IL-1R1) triggers a critical innate immune signaling cascade that contributes to the pathogenesis of AKI. IL-1R1 is expressed on some myeloid cell populations and on multiple kidney cell lineages, including tubular and endothelial cells. Pharmacological inhibition of the IL-1R1 does not consistently protect the kidney from injury, suggesting there may be complex, cell-specific effects of IL-1R1 stimulation in AKI. METHODS: To examine expression of IL-1 and IL-1R1 in intrinsic renal versus infiltrating immune cell populations during AKI, we analyzed single-cell RNA sequencing (scRNA-seq) data from kidney tissues of humans with AKI and mice with acute aristolochic acid exposure. We then investigated cell-specific contributions of renal IL-1R1 signaling to AKI using scRNA-seq, RNA microarray, and pharmacological interventions in mice with IL-1R1 deletion restricted to the proximal tubule or endothelium. RESULTS: scRNA-seq analyses demonstrated robust IL-1 expression in myeloid cell populations and low-level IL-1R1 expression in kidney parenchymal cells during toxin-induced AKI. Our genetic studies showed that IL-1R1 activation in the proximal tubule exacerbated toxin-induced AKI and cell death through local suppression of apolipoprotein M. By contrast, IL-1R1 activation in endothelial cells ameliorated aristolochic acid-induced AKI by restoring VEGFA-dependent endothelial cell viability and density. CONCLUSIONS: These data highlight opposing cell-specific effects of IL-1 receptor signaling on AKI after toxin exposure. Disrupting pathways activated by IL-1R1 in the tubule, while preserving those triggered by IL-1R1 activation on endothelial cells, may afford renoprotection exceeding that of global IL-1R1 inhibition while mitigating unwanted actions of IL-1R1 blockade.


Subject(s)
Acute Kidney Injury , Receptors, Interleukin-1 , Humans , Mice , Animals , Receptors, Interleukin-1/genetics , Apolipoproteins M , Endothelial Cells/metabolism , Acute Kidney Injury/pathology , Mice, Knockout , Interleukin-1 , Endothelium/metabolism , Mice, Inbred C57BL
3.
Kidney Int ; 103(3): 514-528, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36334787

ABSTRACT

The most common cause of acute kidney injury (AKI) in critically ill patients is sepsis. Kidney macrophages consist of both F4/80hi and CD11bhi cells. The role of macrophage subpopulations in septic AKI pathogenesis remains unclear. As F4/80hi macrophages are reported to contribute to immunomodulation following injury, we hypothesized that selective depletion of F4/80hi macrophages would worsen septic AKI. F4/80hi macrophages were depleted via diphtheria toxin injection in CD11cCre(+)/CX3CR1dtr/wt (F4/80 MKO mice) compared to CD11cCre(-)/CX3CR1dtr/wt (F4/80 MWT) mice. F4/80 MWT and F4/80 MKO mice were subjected to sham or cecal ligation and puncture to induce sepsis. Compared to F4/80 MWT mice, F4/80 MKO mice displayed worsened septic AKI at 24 hours as measured by serum creatinine and histologic injury scoring. Kidneys from F4/80 MKO mice elaborated higher kidney interleukin-6 levels. Mechanistically, single cell RNA sequencing identified a macrophage-endothelial cell immunoregulatory axis that underlies interleukin-6 expression. F4/80hi macrophages expressed interleukin-1 receptor antagonist and limited interleukin-6 expression in endothelial cells. In turn, anti-interleukin-6 therapy ameliorated septic AKI in F4/80 MKO mice. Thus, F4/80hi macrophages express interleukin-1 receptor antagonist and constrain interleukin-6 generation from endothelial cells to limit septic AKI, representing a targetable cellular crosstalk in septic AKI. These findings are particularly relevant owing to the efficacy of anti-interleukin-6 therapies during COVID-19 infection, a disease associated with high rates of AKI and endothelial dysfunction.


Subject(s)
Acute Kidney Injury , COVID-19 , Sepsis , Mice , Animals , Endothelial Cells/pathology , COVID-19/complications , Acute Kidney Injury/pathology , Kidney/pathology , Macrophages/metabolism , Interleukin-6/metabolism , Sepsis/complications , Receptors, Interleukin-1/metabolism , Mice, Inbred C57BL
4.
Hypertension ; 79(6): 1227-1236, 2022 06.
Article in English | MEDLINE | ID: mdl-35430875

ABSTRACT

BACKGROUND: Type 1 angiotensin (AT1) receptors are expressed on immune cells, and we previously found that bone marrow-derived AT1 receptors protect against Ang (angiotensin) II-induced hypertension. CD11c is expressed on myeloid cells derived from the bone marrow, including dendritic cells (DCs) that activate T lymphocytes. Here, we examined the role of AT1 receptors on CD11c+ cells in hypertension pathogenesis. METHODS: Mice lacking the dominant murine AT1 receptor isoform, AT1a, on CD11c+ cells (dendritic cell [DC] AT1aR knockout [KO]) and wild-type (WT) littermates were subjected to Ang II-induced hypertension. Blood pressures were measured by radiotelemetry. RESULTS: DC AT1aR KO mice had exaggerated hypertensive responses to chronic Ang II infusion with enhanced renal accumulation of effector memory T cells and CD40+ DCs. CCL5 (C-C motif chemokine ligand 5) recruits T cells into injured tissues, and CCR7 (C-C motif chemokine receptor 7) facilitates DC and T cell interactions in the kidney lymph node to allow T cell activation. DCs from the hypertensive DC AT1aR KO kidneys expressed higher levels of CCL5 and CCR7. mRNA expressions for CCR7 and tumor necrosis factor-α were increased in CD4+ T cells from the renal lymph nodes of DC AT1aR KO mice. During the second week of Ang II infusion when blood pressures between groups diverged, DC AT1aR KO mice excreted less sodium than WTs. Expressions for epithelial sodium channel subunits were increased in DC AT1aR KO kidneys. CONCLUSIONS: Following activation of the renin angiotensin system, AT1aR stimulation on DCs suppresses renal DC maturation and T cell activation with consequent protection from sodium retention and blood pressure elevation.


Subject(s)
Hypertension , Receptor, Angiotensin, Type 1 , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Dendritic Cells/metabolism , Hypertension/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Receptors, CCR7/metabolism , Sodium/metabolism , T-Lymphocytes/metabolism
5.
Am J Physiol Renal Physiol ; 322(2): F164-F174, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34894725

ABSTRACT

Interleukin (IL)-1 receptor type 1 (IL-1R1) activation triggers a proinflammatory signaling cascade that can exacerbate kidney injury. However, the functions of podocyte IL-1R1 in glomerular disease remain unclear. To study the role of IL-1R1 signaling in podocytes, we selectively ablated podocyte IL-1R1 in mice (PKO mice). We then subjected PKO mice and wild-type controls to two glomerular injury models: nephrotoxic serum (NTS)- and adriamycin-induced nephropathy. Surprisingly, we found that IL-1R1 activation in podocytes limited albuminuria and podocyte injury during NTS- and adriamycin-induced nephropathy. Moreover, deletion of IL-1R1 in podocytes drove podocyte apoptosis and glomerular injury through diminishing Akt activation. Activation of Akt signaling abrogated the differences in albuminuria and podocyte injury between wild-type and PKO mice during NTS. Thus, IL-1R1 signaling in podocytes limits susceptibility to glomerular injury via an Akt-dependent signaling pathway. These data identify an unexpected protective role for IL-1R1 signaling in podocytes in the pathogenesis of glomerular disease.NEW & NOTEWORTHY The present study establishes that activation of the receptor for interleukin-1 limits susceptibility to damage to the kidney glomerulus in preclinical mouse models by stimulating Akt signaling cascades inside the podocyte.


Subject(s)
Glomerulonephritis/metabolism , Podocytes/metabolism , Proteinuria/metabolism , Receptors, Interleukin-1 Type I/metabolism , Animals , Apoptosis/drug effects , Cell Line , Disease Models, Animal , Doxorubicin , Glomerulonephritis/chemically induced , Glomerulonephritis/pathology , Glomerulonephritis/prevention & control , Humans , Interleukin-1beta/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice, 129 Strain , Mice, Knockout , Podocytes/drug effects , Podocytes/pathology , Proteinuria/chemically induced , Proteinuria/pathology , Proteinuria/prevention & control , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Interleukin-1 Type I/agonists , Receptors, Interleukin-1 Type I/genetics , Signal Transduction
6.
JCI Insight ; 6(15)2021 08 09.
Article in English | MEDLINE | ID: mdl-34369383

ABSTRACT

The transcription factor Twist1 regulates several processes that could impact kidney disease progression, including epithelial cell differentiation and inflammatory cytokine induction. Podocytes are specialized epithelia that exhibit features of immune cells and could therefore mediate unique effects of Twist1 on glomerular disease. To study Twist1 functions in podocytes during proteinuric kidney disease, we employed a conditional mutant mouse in which Twist1 was selectively ablated in podocytes (Twist1-PKO). Deletion of Twist1 in podocytes augmented proteinuria, podocyte injury, and foot process effacement in glomerular injury models. Twist1 in podocytes constrained renal accumulation of monocytes/macrophages and glomerular expression of CCL2 and the macrophage cytokine TNF-α after injury. Deletion of TNF-α selectively from podocytes had no impact on the progression of proteinuric nephropathy. By contrast, the inhibition of CCL2 abrogated the exaggeration in proteinuria and podocyte injury accruing from podocyte Twist1 deletion. Collectively, Twist1 in podocytes mitigated urine albumin excretion and podocyte injury in proteinuric kidney diseases by limiting CCL2 induction that drove monocyte/macrophage infiltration into injured glomeruli. Myeloid cells, rather than podocytes, further promoted podocyte injury and glomerular disease by secreting TNF-α. These data highlight the capacity of Twist1 in the podocyte to mitigate glomerular injury by curtailing the local myeloid immune response.


Subject(s)
Chemokine CCL2/metabolism , Myeloid Cells/immunology , Podocytes/metabolism , Renal Insufficiency, Chronic , Tumor Necrosis Factor-alpha/metabolism , Twist-Related Protein 1/metabolism , Animals , Cell Differentiation , Gene Silencing , Immunity/immunology , Kidney Glomerulus/immunology , Kidney Glomerulus/injuries , Kidney Glomerulus/metabolism , Macrophages , Mice , Proteinuria/metabolism , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology
7.
Front Physiol ; 12: 683098, 2021.
Article in English | MEDLINE | ID: mdl-34276404

ABSTRACT

Background: Acute kidney injury (AKI) is one of the most common organ failures following surgery. We have developed a tripeptide mimetic (ANXA1sp) of the parent annexin A1 molecule that shows promise as an organ protectant limiting cellular stress; however, its potential as a kidney protective agent remains unexplored, and its mechanism of action is poorly understood. Our hypothesis was that ANXA1sp would limit kidney injury following surgical ischemic kidney injury. Methods: In a blinded fashion, wildtype mice were assigned to receive vehicle control or ANXA1sp one hour prior to and one hour after kidney vascular clamping. Our primary outcomes were markers of kidney injury and function as measured by serum creatinine and histologic injury scoring of kidney tissue sections. Immunofluorescence microscopy, real-time PCR, and Western blot were used to assess cell death, oxidative stress, and mitochondrial biomarkers. An in vitro model of oxygen-glucose deprivation in immortalized kidney tubule cells was used. Results: ANXA1sp given prior to and after ischemic kidney injury abrogated ischemic kidney injury. ANXA1sp limited cell death both in vivo and in vitro and abrogated oxidative stress following ischemia. ANXA1sp significantly increased the expression of markers associated with protective mitophagy and limited the expression of markers associated with detrimental mitochondrial fission. ANXA1sp upregulated the expression of the mitochondrial protectant sirtuin-3 (SIRT3) in the mitochondria of kidney tubular cells. Silencing of SIRT3 reversed ANXA1sp-mediated protection against hypoxic cell death. Conclusions: ANXA1sp limits kidney injury, upregulates SIRT3, and preserves mitochondrial integrity following ischemic kidney injury. ANXA1sp holds considerable promise as a perioperative kidney protectant prior to ischemia inducing surgery and kidney transplantation.

8.
Kidney360 ; 2(5): 784-794, 2021 05 27.
Article in English | MEDLINE | ID: mdl-35373065

ABSTRACT

Background: Twist1 is a basic helix-loop-helix domain-containing transcription factor that participates in diverse cellular functions, including epithelial-mesenchymal transition and the cellular immune response. Although Twist1 plays critical roles in the initiation and progression of kidney diseases, the effects of Twist1 in the T lymphocyte on the progression of renal fibrosis require elucidation. Methods: 129/SvEv mice with a floxed allele for the gene encoding Twist1 or TNFα were bred with CD4-Cre mice to yield CD4-Cre+ Twist1flox/flox (Twist1-TKO) or CD4-Cre+ TNFflox/flox (TNF-TKO) mice with robust, but selective, deletion of Twist1 or TNFα mRNA in T cells, respectively. Twist1 TKO, TNF TKO, and WT controls underwent UUO with assessment of kidney fibrosis and T-cell phenotype at 14 days. Results: Compared with WT controls, obstructed kidneys from Twist1 TKO mice had attenuated extracellular matrix deposition. Despite this diminished fibrosis, Twist1 TKO obstructed kidneys contained more CD8+ T cells than in WTs. These intrarenal CD8+ T cells exhibited greater activation and higher levels of TNFα expression than those from WT obstructed kidneys. Further, we found that selective deletion of TNFα from T cells exaggerated renal scar formation and injury after UUO, highlighting the capacity of T-cell TNF to constrain fibrosis in the kidney. Conclusions: Twist1 in T cells promotes kidney fibrogenesis, in part, by curtailing the renal accumulation of TNF-elaborating T cells.


Subject(s)
Kidney Diseases , Twist-Related Protein 1/metabolism , Ureteral Obstruction , Animals , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Fibrosis , Kidney/metabolism , Kidney Diseases/metabolism , Mice , Mice, Knockout , Ureteral Obstruction/complications
9.
Kidney Dis (Basel) ; 6(4): 247-257, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32903940

ABSTRACT

BACKGROUND: Twist1 is a basic helix-loop-helix domain containing transcription factor that regulates cell differentiation, migration, proliferation, survival, and inflammatory responses by transcriptionally regulating a wide range of downstream target genes. Its homologous protein, Twist2, shares many structural and functional similarities with Twist1. SUMMARY: Accumulating evidence from both preclinical and clinical studies suggests that Twist1 is a pivotal regulator of several forms of renal disease. Twist1 is persistently activated following renal insults, particularly in chronic kidney diseases, and contributes to the renal inflammatory responses, tubular cell transformation programs, and possibly fibroblast activation, all of which are involved in the initiation and progression of kidney diseases. KEY MESSAGE: This review will specifically focus on Twist1 and outline our understanding of its functions in kidney disorders along with the introduction of Twist2 where pertinent. The thorough knowledge of Twist1's actions in the pathogenesis of kidney diseases should facilitate the development of novel therapeutics for kidney injury.

10.
Hypertension ; 75(3): 869-876, 2020 03.
Article in English | MEDLINE | ID: mdl-31983306

ABSTRACT

Activated T lymphocytes that infiltrate blood pressure control organs make a critical contribution to the pathogenesis of hypertension. Dendritic cells act as potent antigen-presenting cells to stimulate prohypertensive T cells. However, the mechanisms that facilitate the recruitment of prohypertensive T cells and dendritic cells into the kidney's draining lymph node during hypertension require elucidation. As CCR7 (C-C motif chemokine receptor type 7) directs the homing of lymphocytes and dendritic cells into lymph nodes, we posited that dendritic cell-mediated T lymphocyte stimulation in the renal lymph node is CCR7 dependent and required for a full hypertensive response. We found that CCR7-deficient (CCR7 KO) mice had a blunted hypertensive response in our model of chronic Ang II (angiotensin II) infusion. Ang II-infused CCR7 KO animals had exaggerated accumulation of CD8+ T cells in the kidney but reduced numbers of CD4+ and CD8+ T cells in the kidney's draining lymph node. To understand whether CCR7-dependent homing of T lymphocytes or dendritic cells into the lymph node regulates the hypertensive response, we injected CCR7 KO or wild-type T cells or dendritic cells into CCR7 KO recipients, neither of which restored the full hypertensive response to Ang II infusion. However, adoptive transfer of wild-type but not CCR7 KO T lymphocytes into RAG1 (recombination-activating gene 1)-deficient mice that lack a lymphocyte niche restored full blood pressure elevation during Ang II infusion. Thus, CCR7-dependent interactions between T lymphocytes and dendritic cells are essential for T lymphocyte stimulation and hypertension accruing from inappropriate activation of the renin-angiotensin system.


Subject(s)
Chemotaxis, Leukocyte/physiology , Hypertension/immunology , Receptors, CCR7/physiology , T-Lymphocyte Subsets/immunology , Adaptive Immunity , Adoptive Transfer , Angiotensin II/toxicity , Animals , Dendritic Cells/transplantation , Genes, RAG-1 , Hypertension/physiopathology , Kidney/immunology , Kidney/physiopathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nephrectomy , Receptors, CCR7/deficiency , Receptors, CCR7/genetics
11.
12.
Am J Physiol Renal Physiol ; 318(1): F107-F116, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31736350

ABSTRACT

Nephrotoxic serum nephritis (NTN) models immune-mediated human glomerulonephritis and culminates in kidney inflammation and fibrosis, a process regulated by T lymphocytes. TNF-α is a key proinflammatory cytokine that contributes to diverse forms of renal injury. Therefore, we posited that TNF-α from T lymphocytes may contribute to NTN pathogenesis. Here, mice with T cell-specific deletion of TNF-α (TNF TKO) and wild-type (WT) control mice were subjected to the NTN model. At 14 days after NTN, kidney injury and fibrosis were increased in kidneys from TNF TKO mice compared with WT mice. PD1+CD4+ T cell numbers and mRNA levels of IL-17A were elevated in NTN kidneys of TNF TKO mice, suggesting that augmented local T helper 17 lymphocyte responses in the TNF TKO kidney may exaggerate renal injury and fibrosis. In turn, we found increased accumulation of neutrophils in TNF TKO kidneys during NTN. We conclude that TNF-α production in T lymphocytes mitigates NTN-induced kidney injury and fibrosis by inhibiting renal T helper 17 lymphocyte responses and infiltration of neutrophils.


Subject(s)
Fibrosis/metabolism , Glomerulonephritis/metabolism , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Disease Models, Animal , Fibrosis/genetics , Fibrosis/pathology , Glomerulonephritis/genetics , Glomerulonephritis/pathology , Interleukin-17/genetics , Interleukin-17/metabolism , Kidney/metabolism , Kidney/pathology , Mice , Mice, Knockout , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/genetics
13.
Kidney Int ; 97(1): 119-129, 2020 01.
Article in English | MEDLINE | ID: mdl-31685313

ABSTRACT

Tubulointerstitial disease in the kidney culminates in renal fibrosis that portents organ failure. Twist1, a basic helix-loop-helix protein 38 transcription factor, regulates several essential biological functions, but inappropriate Twist1 activity in the kidney epithelium can trigger kidney fibrogenesis and chronic kidney disease. By contrast, Twist1 in circulating myeloid cells may constrain inflammatory injury by attenuating cytokine generation. To dissect the effects of Twist1 in kidney tubular versus immune cells on renal inflammation following toxin-induced renal injury, we subjected mice with selective deletion of Twist1 in renal epithelial cells or macrophages to aristolochic acid-induced chronic kidney disease. Ablation of Twist1 in the distal nephron attenuated kidney damage, interstitial fibrosis, and renal inflammation after aristolochic acid exposure. However, macrophage-specific deletion of Twist1 did not impact the development of aristolochic acid-induced nephropathy. In vitro studies confirmed that Twist1 in renal tubular cells underpins their susceptibility to apoptosis and propensity to generate pro-fibrotic mediators in response to aristolochic acid. Moreover, co-culture studies revealed that Twist1 in renal epithelia augmented the recruitment and activation of pro-inflammatory CD64+ macrophages. Thus, Twist1 in the distal nephron rather than in infiltrating macrophages propagates chronic inflammation and fibrogenesis during aristolochic acid-induced nephropathy.


Subject(s)
Kidney Tubules, Distal/pathology , Macrophages/immunology , Nephritis, Interstitial/immunology , Renal Insufficiency, Chronic/immunology , Twist-Related Protein 1/metabolism , Animals , Apoptosis/drug effects , Apoptosis/immunology , Aristolochic Acids/toxicity , Coculture Techniques , Disease Models, Animal , Epithelial Cells , Female , Fibrosis , Gene Knockdown Techniques , Hepatitis A Virus Cellular Receptor 1/metabolism , Humans , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/immunology , Kidney Tubules, Distal/metabolism , Lipocalin-2/metabolism , Macrophages/metabolism , Mice , Mice, Transgenic , Nephritis, Interstitial/chemically induced , Nephritis, Interstitial/pathology , Primary Cell Culture , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/pathology , Twist-Related Protein 1/genetics , Up-Regulation/drug effects , Up-Regulation/immunology
14.
Hypertension ; 75(1): 131-138, 2020 01.
Article in English | MEDLINE | ID: mdl-31786985

ABSTRACT

FLT3L (Fms-like tyrosine kinase 3 ligand) stimulates the development of classical dendritic cells (DCs). Here we tested the hypothesis that classical DCs drive blood pressure elevation by promoting renal fluid retention. FLT3L-deficient (FLT3L-/-) mice that lack classical DCs in the kidney had mean arterial pressures similar to wild-types (WTs) at baseline but had blunted hypertensive responses during 4 weeks of chronic Ang II (angiotensin II) infusion. In FLT3L-/- mice, the proportions of effector memory T cells in the kidney were similar to those in WTs at baseline. However, after Ang II infusion, proportions of effector memory T cells were dramatically lower in the FLT3L-/- kidneys versus WTs, indicating that classical DCs augment the renal accumulation of effector T cells after renin-angiotensin system activation. Consistent with their lower blood pressures, the Ang II-infused FLT3L-/- mice had attenuated cardiac hypertrophy and lower renal mRNA expression for pro-hypertensive cytokines. Moreover, the Ang II-infused FLT3L-/- mice had lower urinary excretion of the oxidative stress marker 8-isoprostane and lower renal mRNA levels of nicotinamide adenine dinucleotide phosphate oxidase 2. In an intraperitoneal saline challenge test at day 7 of Ang II, FLT3L-/- mice excreted higher proportions of the injected volume and sodium than WTs. Consistent with this enhanced diuresis, mRNA expressions for the sodium chloride cotransporter and all 3 subunits of the epithelial sodium channel were diminished by >40% in FLT3L-/- kidneys compared with the WTs. Thus, classical FLT3L-dependent DCs promote renal T-cell activation with consequent oxidative stress, fluid retention, and blood pressure elevation.


Subject(s)
Dendritic Cells/metabolism , Hypertension/metabolism , Kidney/metabolism , Oxidative Stress/physiology , T-Lymphocytes/metabolism , Angiotensin II , Animals , Disease Models, Animal , Hypertension/chemically induced , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout
15.
Circ Res ; 125(12): 1055-1066, 2019 12 06.
Article in English | MEDLINE | ID: mdl-31630621

ABSTRACT

RATIONALE: The ubiquitin-editing protein A20 in dendritic cells (DCs) suppresses NF-κB (nuclear factor-κB) signaling and constrains DC-mediated T-cell stimulation, but the role of A20 in modulating the hypertensive response requires elucidation. OBJECTIVE: Here, we tested the hypothesis that A20 in CD11c-expressing myeloid cells mitigates Ang II (angiotensin II)-induced hypertension by limiting renal T-cell activation. METHODS AND RESULTS: Mice with heterozygous deletion of A20 in CD11c-expressing myeloid cells (DC ACT[Cd11c-Cre+A20flox/wt]) have spontaneous DC activation but have normal baseline blood pressures. In response to low-dose chronic Ang II infusion, DC ACT mice compared with WT (wild type) controls had an exaggerated hypertensive response and augmented proportions of CD62LloCD44hi effector memory T lymphocytes in the kidney lymph node. After 10 days of Ang II, DC ACT kidneys had increased numbers of memory effector CD8+, but not CD4+ T cells, compared with WTs. Moreover, the expressions of TNF-α (tumor necrosis factor-α) and IFN-γ (interferon-γ) were upregulated in the DC ACT renal CD8+ T cells but not CD4+ T cells. Saline challenge testing revealed enhanced renal fluid retention in the DC ACT mice. DC ACT kidneys showed augmented protein expression of γ-epithelial sodium channel and NHE3 (sodium-hydrogen antiporter 3). DC ACT mice also had greater reductions in renal blood flow following acute injections with Ang II and enhanced oxidant stress in the vasculature as evidenced by higher circulating levels of malondialdehyde compared with WT controls. To directly test whether enhanced T-cell activation in the DC ACT cohort was responsible for their exaggerated hypertensive response, we chronically infused Ang II into lymphocyte-deficient DC ACT Rag1 (recombination activating protein 1)-deficient (Rag1-/-) mice and WT (Cd11c-Cre-A20flox/wt) Rag1-/- controls. The difference in blood pressure elevation accruing from DC activation was abrogated on the Rag1-/- strain. CONCLUSIONS: Following stimulation of the renin-angiotensin system, A20 suppresses DC activation and thereby mitigates T-cell-dependent blood pressure elevation.


Subject(s)
Dendritic Cells/metabolism , Hypertension/metabolism , Kidney/metabolism , Myeloid Cells/metabolism , T-Lymphocytes/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/deficiency , Animals , Cells, Cultured , Dendritic Cells/immunology , Hypertension/immunology , Hypertension/prevention & control , Kidney/cytology , Kidney/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor alpha-Induced Protein 3/immunology
16.
Kidney Int ; 96(6): 1308-1319, 2019 12.
Article in English | MEDLINE | ID: mdl-31585741

ABSTRACT

Wnt/ß-catenin signaling is essential in the pathogenesis of renal fibrosis. We previously reported inhibition of the Wnt O-acyl transferase porcupine, required for Wnt secretion, dramatically attenuates kidney fibrosis in the murine unilateral ureteral obstruction model. Here, we investigated the tissue-specific contributions of porcupine to renal fibrosis and inflammation in ureteral obstruction using mice with porcupine deletion restricted to the kidney tubular epithelium or infiltrating myeloid cells. Obstruction of the ureter induced the renal mRNA expression of porcupine and downstream targets, ß-catenin, T-cell factor, and lymphoid enhancer factor in wild type mice. Renal tubular specific deficiency of porcupine reduced the expression of collagen I and other fibrosis markers in the obstructed kidney. Moreover, kidneys from obstructed mice with tubule-specific porcupine deficiency had reduced macrophage accumulation with attenuated expression of myeloid cytokine and chemokine mRNA. In co-culture with activated macrophages, renal tubular cells from tubular-specific porcupine knockout mice had blunted induction of fibrosis mediators compared with wild type renal tubular cells. In contrast, macrophages from macrophage-specific porcupine deficient mice in co-culture with wild type renal tubular cells had markedly enhanced expression of pro-fibrotic cytokines compared to wild type macrophages. Consequently, porcupine deletion specifically within macrophages augmented renal scar formation following ureteral obstruction. Thus, our experiments suggest a benefit of interrupting Wnt secretion specifically within the kidney epithelium while preserving Wnt O-acylation in infiltrating myeloid cells during renal fibrogenesis.


Subject(s)
Acyltransferases/metabolism , Membrane Proteins/metabolism , Nephrosclerosis/metabolism , Wnt Signaling Pathway , Animals , Chemokines/metabolism , Female , Fibrosis , Kidney Tubules/metabolism , Kidney Tubules/pathology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Myeloid Cells/metabolism , Nephrosclerosis/etiology , Ureteral Obstruction
17.
J Am Soc Nephrol ; 30(10): 1925-1938, 2019 10.
Article in English | MEDLINE | ID: mdl-31337692

ABSTRACT

BACKGROUND: Polarized macrophage populations can orchestrate both inflammation of the kidney and tissue repair during CKD. Proinflammatory M1 macrophages initiate kidney injury, but mechanisms through which persistent M1-dependent kidney damage culminates in fibrosis require elucidation. Krüppel-like factor 4 (KLF4), a zinc-finger transcription factor that suppresses inflammatory signals, is an essential regulator of macrophage polarization in adipose tissues, but the effect of myeloid KLF4 on CKD progression is unknown. METHODS: We used conditional mutant mice lacking KLF4 or TNFα (KLF4's downstream effector) selectively in myeloid cells to investigate macrophage KLF4's role in modulating CKD progression in two models of CKD that feature robust macrophage accumulation, nephrotoxic serum nephritis, and unilateral ureteral obstruction. RESULTS: In these murine CKD models, KLF4 deficiency in macrophages infiltrating the kidney augmented their M1 polarization and exacerbated glomerular matrix deposition and tubular epithelial damage. During the induced injury in these models, macrophage-specific KLF4 deletion also exacerbated kidney fibrosis, with increased levels of collagen 1 and α-smooth muscle actin in the injured kidney. CD11b+Ly6Chi myeloid cells isolated from injured kidneys expressed higher levels of TNFα mRNA versus wild-type controls. In turn, mice bearing macrophage-specific deletion of TNFα exhibited decreased glomerular and tubular damage and attenuated kidney fibrosis in the models. Moreover, treatment with the TNF receptor-1 inhibitor R-7050 during nephrotoxic serum nephritis reduced damage, fibrosis, and necroptosis in wild-type mice and mice with KLF4-deficient macrophages, and abrogated the differences between the two groups in these parameters. CONCLUSIONS: These data indicate that macrophage KLF4 ameliorates CKD by mitigating TNF-dependent injury and fibrosis.


Subject(s)
Kidney Diseases/etiology , Kidney/pathology , Kruppel-Like Transcription Factors/physiology , Macrophages/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Fibrosis/etiology , Kruppel-Like Factor 4 , Male , Mice , Tumor Necrosis Factor-alpha/antagonists & inhibitors
18.
J Am Soc Nephrol ; 30(9): 1674-1685, 2019 09.
Article in English | MEDLINE | ID: mdl-31315922

ABSTRACT

BACKGROUND: Following an acute insult, macrophages regulate renal fibrogenesis through the release of various factors that either encourage the synthesis of extracellular matrix synthesis or the degradation of matrix via endocytosis, proteolysis, or both. However, the roles of infiltrating versus resident myeloid cells in these opposing processes require elucidation. The transcription factor Twist1 controls diverse essential cellular functions through induction of several downstream targets, including matrix metalloproteinases (MMPs). In macrophages, Twist1 can influence patterns of cytokine generation, but the role of macrophage Twist1 in renal fibrogenesis remains undefined. METHODS: To study Twist1 functions in different macrophage subsets during kidney scar formation, we used two conditional mutant mouse models in which Twist1 was selectively ablated either in infiltrating, inflammatory macrophages or in resident tissue macrophages. We assessed fibrosis-related parameters, matrix metallopeptidase 13 (MMP13, or collagen 3, which catalyzes collagen degradation), inflammatory cytokines, and other factors in these Twist1-deficient mice compared with wild-type controls after subjecting the animals to unilateral ureteral obstruction. We also treated wild-type and Twist1-deficient mice with an MMP13 inhibitor after unilateral ureteral obstruction. RESULTS: Twist1 in infiltrating inflammatory macrophages but not in resident macrophages limited kidney fibrosis after ureteral obstruction by driving extracellular matrix degradation. Moreover, deletion of Twist1 in infiltrating macrophages attenuated the expression of MMP13 in CD11b+Ly6Clo myeloid cells. Inhibition of MMP13 abrogated the protection from renal fibrosis afforded by macrophage Twist1. CONCLUSIONS: Twist1 in infiltrating myeloid cells mitigates interstitial matrix accumulation in the injured kidney by promoting MMP13 production, which drives extracellular matrix degradation. These data highlight the complex cell-specific actions of Twist1 in the pathogenesis of kidney fibrosis.


Subject(s)
Extracellular Matrix/metabolism , Kidney Diseases/metabolism , Kidney/metabolism , Kidney/pathology , Macrophages/metabolism , Matrix Metalloproteinase 13/metabolism , Twist-Related Protein 1/metabolism , Actins/metabolism , Animals , Benzofurans/pharmacology , CX3C Chemokine Receptor 1/metabolism , Collagen Type I/metabolism , Disease Models, Animal , Fibrosis , Gene Expression , Hydroxyproline/metabolism , Kidney Diseases/etiology , Kidney Diseases/pathology , Macrophages, Peritoneal/metabolism , Male , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase Inhibitors/pharmacology , Mice , Morpholines/pharmacology , Myeloid Cells/enzymology , Twist-Related Protein 1/genetics , Ureteral Obstruction/complications
19.
Cell Signal ; 62: 109331, 2019 10.
Article in English | MEDLINE | ID: mdl-31154001

ABSTRACT

Upon occurrence of kidney injury, tubular cells arrested in G2/M stage may promote interstitial fibroblast activation and kidney fibrosis through producing large amounts of pro-fibrotic cytokines. MTORC1 signaling is essential for controlling cell growth, however, the role and mechanisms for mTORC1 in regulating tubular cell cycle progression during kidney fibrosis are not clear. Here we reported that p-S6 abundance was increased at 15 min, reached peak at 1 h and declined from 3 h to 24 h, while the abundance of p-4E-BP1 and p-Histone H3 was increased from 15 min to 24 h in tubular epithelial cells at the similar pattern after serum stimulation. The phosphorylation of 4E-BP1 was prohibited in NRK-52E cells by the transfection of 4E-BP1 plasmid with four phospho-sites mutation (4E-BP1A4). 4E-BP1A4 transfection led to less G2/M cell arrest as well as the production of pro-fibrotic cytokine and extracellular matrix in NRK-52E cells. In addition, aristolochic acid (AA)-induced tubular cell G2/M arrest induced by treatment was also largely attenuated in NRK-52E cells transfected with 4E-BP1A4. In mouse kidneys with UUO nephropathy, p-4E-BP1 abundance was markedly elevated in the mitotic tubular cells. Therefore, these data indicates that suppressing 4E-BP1 phosphorylation may inhibit tubular cell G2/M-arrest and kidney fibrosis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Cycle Proteins/genetics , Fibrosis/genetics , Histones/genetics , Kidney/metabolism , Animals , Apoptosis/drug effects , Aristolochic Acids/pharmacology , Cell Cycle/drug effects , Cell Division/genetics , Cell Line, Tumor , Epithelial Cells/drug effects , Fibrosis/pathology , Gene Expression Regulation, Developmental/drug effects , Humans , Kidney/pathology , Kidney Tubules/drug effects , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mitosis/genetics , Phosphorylation/drug effects , Signal Transduction/drug effects
20.
Am J Physiol Renal Physiol ; 317(2): F221-F228, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31141402

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is associated with increased chemokines, cytokines, and growth factors in the diseased kidney. We found that both isoforms of IL-1, IL-1α and IL-1ß, were upregulated in ADPKD tissues. Here, we used a unique murine ADPKD model with selective deletion of polycystin-1 (pkd1) in the kidney (KPKD1) to study the role of IL-1 signaling in ADPKD progression. In KPKD mice, genetic deletion of the IL-1 receptor [IL-1 receptor (IL-1R) knockout (KO)] prolongs survival and attenuates cyst volume. Compared with IL-1R wild-type KPKD1 kidneys, IL-1R KO KPKD1 kidneys have upregulated TNF-α gene expression, with consequent elevations in markers for TNF-dependent regulated necrosis. We further observed that regulated necrosis was increased in ADPKD tissues from both humans and mice. To confirm that enhanced necroptosis is protective in ADPKD, we treated KPKD1 mice with an inhibitor of regulated necrosis (Nec-1). Regulated necrosis suppression augments kidney weights, suggesting that regulated necrosis is required to limit kidney growth in ADPKD. Thus, IL-1R activation drives ADPKD progression by paradoxically limiting regulated necrosis.


Subject(s)
Kidney/metabolism , Polycystic Kidney, Autosomal Dominant/metabolism , Receptors, Interleukin-1 Type I/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Humans , Interleukin-1/genetics , Interleukin-1/metabolism , Kidney/pathology , Mice, Knockout , Necroptosis , Necrosis , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/pathology , Receptors, Interleukin-1 Type I/deficiency , Receptors, Interleukin-1 Type I/genetics , Signal Transduction , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL