Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Am J Physiol Renal Physiol ; 327(1): F171-F183, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38779751

ABSTRACT

Sepsis-associated acute kidney injury (SA-AKI) is a key contributor to the life-threatening sequelae attributed to sepsis. Mechanistically, SA-AKI is a consequence of unabated myeloid cell activation and oxidative stress that induces tubular injury. Iron mediates inflammatory pathways directly and through regulating the expression of myeloid-derived ferritin, an iron storage protein comprising ferritin light (FtL) and ferritin heavy chain (FtH) subunits. Previous work revealed that myeloid FtH deletion leads to a compensatory increase in intracellular and circulating FtL and is associated with amelioration of SA-AKI. We designed this study to test the hypothesis that loss of myeloid FtL and subsequently, circulating FtL will exacerbate the sepsis-induced inflammatory response and worsen SA-AKI. We generated a novel myeloid-specific FtL knockout mouse (FtLLysM-/-) and induced sepsis via cecal ligation and puncture or lipopolysaccharide endotoxemia. As expected, serum ferritin levels were significantly lower in the knockout mice, suggesting that myeloid cells dominantly contribute to circulating ferritin. Interestingly, although sepsis induction led to a marked production of pro- and anti-inflammatory cytokines, there was no statistical difference between the genotypes. There was a similar loss of kidney function, as evidenced by a rise in serum creatinine and cystatin C and renal injury identified by expression of kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin. Finally, RNA sequencing revealed upregulation of pathways for cell cycle arrest and autophagy postsepsis, but no significant differences were observed between genotypes, including in key genes associated with ferroptosis, an iron-mediated form of cell death. The loss of FtL did not impact sepsis-mediated activation of NF-κB or HIF-1a signaling, key inflammatory pathways associated with dysregulated host response. Taken together, while FtL overexpression was shown to be protective against sepsis, the loss of FtL did not influence sepsis pathogenesis.NEW & NOTEWORTHY Hyperferritinemia in sepsis is often associated with a proinflammatory phenotype and poor prognosis. We previously showed the myeloid deletion of FtH results in a compensatory increase in FtL and is associated with reduced circulating cytokines and decreased rates of SA-AKI in animal sepsis models. Here, we show that myeloid deletion of FtL does not impact the severity of SA-AKI following CLP or LPS, suggesting that FtH plays the predominant role in propagating myeloid-induced proinflammatory pathways.


Subject(s)
Acute Kidney Injury , Apoferritins , Mice, Knockout , Sepsis , Animals , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Sepsis/metabolism , Sepsis/complications , Sepsis/genetics , Apoferritins/genetics , Apoferritins/metabolism , Myeloid Cells/metabolism , Disease Models, Animal , Male , Mice , Kidney/metabolism , Kidney/pathology , Mice, Inbred C57BL , Cytokines/metabolism , Inflammation Mediators/metabolism
2.
Clin Immunol ; 248: 109213, 2023 03.
Article in English | MEDLINE | ID: mdl-36566913

ABSTRACT

Ferroptosis is a druggable, iron-dependent form of cell death that is characterized by lipid peroxidation but has received little attention in lupus nephritis. Kidneys of lupus nephritis patients and mice showed increased lipid peroxidation mainly in the tubular segments and an increase in Acyl-CoA synthetase long-chain family member 4, a pro-ferroptosis enzyme. Nephritic mice had an attenuated expression of SLC7A11, a cystine importer, an impaired glutathione synthesis pathway, and low expression of glutathione peroxidase 4, a ferroptosis inhibitor. Lipidomics of nephritic kidneys confirmed ferroptosis. Using nephrotoxic serum, we induced immune complex glomerulonephritis in congenic mice and demonstrate that impaired iron sequestration within the proximal tubules exacerbates ferroptosis. Lupus nephritis patient serum rendered human proximal tubular cells susceptibility to ferroptosis which was inhibited by Liproxstatin-2, a novel ferroptosis inhibitor. Collectively, our findings identify intra-renal ferroptosis as a pathological feature and contributor to tubular injury in human and murine lupus nephritis.


Subject(s)
Ferroptosis , Kidney Diseases , Lupus Nephritis , Humans , Mice , Animals , Iron/metabolism , Kidney Glomerulus/metabolism , Epithelial Cells/metabolism
3.
Proc Natl Acad Sci U S A ; 116(12): 5681-5686, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30833408

ABSTRACT

Malaria, the disease caused by Plasmodium spp. infection, remains a major global cause of morbidity and mortality. Host protection from malaria relies on immune-driven resistance mechanisms that kill Plasmodium However, these mechanisms are not sufficient per se to avoid the development of severe forms of disease. This is accomplished instead via the establishment of disease tolerance to malaria, a defense strategy that does not target Plasmodium directly. Here we demonstrate that the establishment of disease tolerance to malaria relies on a tissue damage-control mechanism that operates specifically in renal proximal tubule epithelial cells (RPTEC). This protective response relies on the induction of heme oxygenase-1 (HMOX1; HO-1) and ferritin H chain (FTH) via a mechanism that involves the transcription-factor nuclear-factor E2-related factor-2 (NRF2). As it accumulates in plasma and urine during the blood stage of Plasmodium infection, labile heme is detoxified in RPTEC by HO-1 and FTH, preventing the development of acute kidney injury, a clinical hallmark of severe malaria.


Subject(s)
Heme/metabolism , Kidney/metabolism , Malaria/physiopathology , Animals , Apoferritins/metabolism , Cell Line , Disease Progression , Epithelial Cells/metabolism , Ferritins/metabolism , Ferritins/physiology , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/physiology , Humans , Immune Tolerance/physiology , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/physiology , Oxidoreductases , Plasmodium berghei/metabolism , Plasmodium berghei/parasitology , Up-Regulation
4.
Am J Physiol Renal Physiol ; 321(6): F675-F688, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34658261

ABSTRACT

Expansion of renal lymphatic networks, or lymphangiogenesis (LA), is well recognized during development and is now being implicated in kidney diseases. Although LA is associated with multiple pathological conditions, very little is known about its role in acute kidney injury. The purpose of this study was to evaluate the role of LA in a model of cisplatin-induced nephrotoxicity. LA is predominately regulated by vascular endothelial growth factor (VEGF)-C and VEGF-D, ligands that exert their function through their cognate receptor VEGF receptor 3 (VEGFR3). We demonstrated that use of MAZ51, a selective VEGFR3 inhibitor, caused significantly worse structural and functional kidney damage in cisplatin nephrotoxicity. Apoptotic cell death and inflammation were also increased in MAZ51-treated animals compared with vehicle-treated animals following cisplatin administration. Notably, MAZ51 caused significant upregulation of intrarenal phospho-NF-κB, phospho-JNK, and IL-6. Cisplatin nephrotoxicity is associated with vascular congestion due to endothelial dysfunction. Using three-dimensional tissue cytometry, a novel approach to explore lymphatics in the kidney, we detected significant vascular autofluorescence attributed to erythrocytes in cisplatin alone-treated animals. Interestingly, no such congestion was detected in MAZ51-treated animals. We found increased renal vascular damage in MAZ51-treated animals, whereby MAZ51 caused a modest decrease in the endothelial markers endomucin and von Willebrand factor, with a modest increase in VEGFR2. Our findings identify a protective role for de novo LA in cisplatin nephrotoxicity and provide a rationale for the development of therapeutic approaches targeting LA. Our study also suggests off-target effects of MAZ51 on the vasculature in the setting of cisplatin nephrotoxicity.NEW & NOTEWORTHY Little is known about injury-associated LA in the kidney and its role in the pathophysiology of acute kidney injury (AKI). Observed exacerbation of cisplatin-induced AKI after LA inhibition was accompanied by increased medullary damage and cell death in the kidney. LA inhibition also upregulated compensatory expression of LA regulatory proteins, including JNK and NF-κB. These data support the premise that LA is induced during AKI and lymphatic expansion is a protective mechanism in cisplatin nephrotoxicity.


Subject(s)
Indoles/toxicity , Kidney Diseases/chemically induced , Kidney/drug effects , Lymphangiogenesis/drug effects , Lymphatic Vessels/drug effects , Naphthalenes/toxicity , Protein Kinase Inhibitors/toxicity , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cisplatin , Disease Models, Animal , Inflammation Mediators/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Kidney/enzymology , Kidney/pathology , Kidney/physiopathology , Kidney Diseases/enzymology , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Lymphatic Vessels/enzymology , Lymphatic Vessels/pathology , Lymphatic Vessels/physiopathology , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphorylation , Signal Transduction , Vascular Endothelial Growth Factor Receptor-3/metabolism
5.
Acta Neuropathol ; 140(4): 549-567, 2020 10.
Article in English | MEDLINE | ID: mdl-32651669

ABSTRACT

The proinflammatory cytokine interleukin 1 (IL-1) is crucially involved in the pathogenesis of multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). Herein, we studied the role of IL-1 signaling in blood-brain barrier (BBB) endothelial cells (ECs), astrocytes and microglia for EAE development, using mice with the conditional deletion of its signaling receptor IL-1R1. We found that IL-1 signaling in microglia and astrocytes is redundant for the development of EAE, whereas the IL-1R1 deletion in BBB-ECs markedly ameliorated disease severity. IL-1 signaling in BBB-ECs upregulated the expression of the adhesion molecules Vcam-1, Icam-1 and the chemokine receptor Darc, all of which have been previously shown to promote CNS-specific inflammation. In contrast, IL-1R1 signaling suppressed the expression of the stress-responsive heme catabolizing enzyme heme oxygenase-1 (HO-1) in BBB-ECs, promoting disease progression via a mechanism associated with deregulated expression of the IL-1-responsive genes Vcam1, Icam1 and Ackr1 (Darc). Mechanistically, our data emphasize a functional crosstalk of BBB-EC IL-1 signaling and HO-1, controlling the transcription of downstream proinflammatory genes promoting the pathogenesis of autoimmune neuroinflammation.


Subject(s)
Blood-Brain Barrier/enzymology , Encephalomyelitis, Autoimmune, Experimental/immunology , Endothelial Cells/enzymology , Heme Oxygenase-1/metabolism , Inflammation/immunology , Interleukin-1/immunology , Animals , Blood-Brain Barrier/immunology , Encephalomyelitis, Autoimmune, Experimental/enzymology , Gene Expression Regulation/immunology , Mice , Mice, Inbred C57BL , Signal Transduction/immunology
7.
Lab Invest ; 99(9): 1376-1388, 2019 09.
Article in English | MEDLINE | ID: mdl-31019289

ABSTRACT

Acute kidney injury (AKI) and chronic kidney disease (CKD) are interconnected syndromes with significant attributable morbidity and mortality. The disturbing trend of increasing incidence and prevalence of these clinical disorders highlights the urgent need for better understanding of the underlying mechanisms that are involved in pathogenesis of these conditions. Lymphangiogenesis and its involvement in various inflammatory conditions is increasingly recognized while its role in AKI and CKD remains to be fully elucidated. Here, we studied lymphangiogenesis in three models of kidney injury. Our results demonstrate that the main ligands for lymphangiogenesis, VEGF-C and VEGF-D, are abundantly present in tubules at baseline conditions and the expression pattern of these ligands is significantly altered following injury. In addition, we show that both of these ligands increase in serum and urine post-injury and suggest that such increment may serve as novel urinary biomarkers of AKI as well as in progression of kidney disease. We also provide evidence that irrespective of the nature of initial insult, lymphangiogenic pathways are rapidly and robustly induced as evidenced by higher expression of lymphatic markers within the kidney.


Subject(s)
Acute Kidney Injury/metabolism , Lymphangiogenesis/physiology , Renal Insufficiency, Chronic/metabolism , Acute Kidney Injury/pathology , Animals , Disease Models, Animal , Kidney/cytology , Kidney/metabolism , Kidney/pathology , Kidney Tubules, Proximal/pathology , Male , Mice , Mice, Inbred C57BL , Renal Insufficiency, Chronic/pathology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor D/metabolism
8.
Am J Physiol Renal Physiol ; 314(5): F702-F714, 2018 05 01.
Article in English | MEDLINE | ID: mdl-28515173

ABSTRACT

Ferroptosis is an iron-dependent form of regulated nonapoptotic cell death, which contributes to damage in models of acute kidney injury (AKI). Heme oxygenase-1 (HO-1) is a cytoprotective enzyme induced in response to cellular stress, and is protective against AKI because of its antiapoptotic and anti-inflammatory properties. However, the role of HO-1 in regulating ferroptosis is unclear. The purpose of this study was to elucidate the role of HO-1 in regulating ferroptotic cell death in renal proximal tubule cells (PTCs). Immortalized PTCs obtained from HO-1+/+ and HO-1-/- mice were treated with erastin or RSL3, ferroptosis inducers, in the presence or absence of antioxidants, an iron source, or an iron chelator. Cells were assessed for changes in morphology and metabolic activity as an indicator of cell viability. Treatment of HO-1+/+ PTCs with erastin resulted in a time- and dose-dependent increase in HO-1 gene expression and protein levels compared with vehicle-treated controls. HO-1-/- cells showed increased dose-dependent erastin- or RSL3-induced cell death in comparison to HO-1+/+ PTCs. Iron supplementation with ferric ammonium citrate in erastin-treated cells decreased cell viability further in HO-1-/- PTCs compared with HO-1+/+ cells. Cotreatment with ferrostatin-1 (ferroptosis inhibitor), deferoxamine (iron chelator), or N-acetyl-l-cysteine (glutathione replenisher) significantly increased cell viability and attenuated erastin-induced ferroptosis in both HO-1+/+ and HO-1-/- PTCs. These results demonstrate an important antiferroptotic role of HO-1 in renal epithelial cells.


Subject(s)
Acute Kidney Injury/enzymology , Heme Oxygenase-1/metabolism , Kidney Tubules, Proximal/enzymology , Membrane Proteins/metabolism , Acetylcysteine/pharmacology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Animals , Antioxidants/pharmacology , Carbolines/toxicity , Cell Death , Cell Line , Cyclohexylamines/pharmacology , Deferoxamine/pharmacology , Dose-Response Relationship, Drug , Ferric Compounds/toxicity , Glutathione/metabolism , Heme Oxygenase-1/deficiency , Heme Oxygenase-1/genetics , Iron Chelating Agents/pharmacology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, Knockout , Phenylenediamines/pharmacology , Piperazines/toxicity , Quaternary Ammonium Compounds/toxicity , Signal Transduction , Time Factors
9.
Am J Physiol Renal Physiol ; 313(3): F740-F755, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28679590

ABSTRACT

Sex and age influence susceptibility to acute kidney injury (AKI), with young females exhibiting lowest incidence. In these studies, we investigated mechanisms which may underlie the sex/age-based dissimilarities. Cisplatin (Cp)-induced AKI resulted in morphological evidence of injury in all groups. A minimal rise in plasma creatinine (PCr) was seen in Young Females, whereas in Aged Females, PCr rose precipitously. Relative to Young Males, Aged Males showed significantly, but temporally, comparably elevated PCr. Notably, Aged Females showed significantly greater mortality, whereas Young Females exhibited none. Tissue KIM-1 and plasma NGAL were significantly lower in Young Females than all others. IGFBP7 levels were modestly increased in both Young groups. IGFBP7 levels in Aged Females were significantly elevated at baseline relative to Aged Males, and increased linearly through day 3, when these levels were comparable in both Aged groups. Plasma cytokine levels similarly showed a pattern of protective effects preferentially in Young Females. Expression of the drug transporter MATE2 did not explain the sex/age distinctions. Heme oxygenase-1 (HO-1) levels (~28-kDa species) showed elevation at day 1 in all groups with highest levels seen in Young Males. Exclusively in Young Females, these levels returned to baseline on day 3, suggestive of a more efficient recovery. In aggregate, we demonstrate, for the first time, a distinctive pattern of response to AKI in Young Females relative to males which appears to be significantly altered in aging. These distinctions may offer novel targets to exploit therapeutically in both females and males in the treatment of AKI.


Subject(s)
Acute Kidney Injury/prevention & control , Aging/metabolism , Kidney/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Age Factors , Aging/pathology , Animals , Autophagy , Cell Proliferation , Cisplatin , Creatinine/blood , Cytokines/blood , Disease Models, Animal , Female , Heme Oxygenase-1/metabolism , Hepatitis A Virus Cellular Receptor 1/metabolism , Insulin-Like Growth Factor Binding Proteins/metabolism , Kidney/pathology , Lipocalin-2/blood , Male , Membrane Proteins/metabolism , Methionine Adenosyltransferase/metabolism , Mice, Inbred C57BL , Sex Factors , Signal Transduction , Time Factors
10.
Hum Mol Genet ; 24(14): 4078-93, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25904107

ABSTRACT

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common known genetic cause of Parkinson's disease, and LRRK2 is also linked to Crohn's and Hansen's disease. LRRK2 is expressed in many organs in mammals but is particularly abundant in the kidney. We find that LRRK2 protein is predominantly localized to collecting duct cells in the rat kidney, with much lower expression in other kidney cells. While genetic knockout (KO) of LRRK2 expression is well-tolerated in mice and rats, a unique age-dependent pathology develops in the kidney. The cortex and medulla of LRRK2 KO rat kidneys become darkly pigmented in early adulthood, yet aged animals display no overt signs of kidney failure. Accompanying the dark pigment we find substantial macrophage infiltration in LRRK2 KO kidneys, suggesting the presence of chronic inflammation that may predispose to kidney disease. Unexpectedly, the dark kidneys of the LRRK2 KO rats are highly resistant to rhabdomyolysis-induced acute kidney injury compared with wild-type rats. Biochemical profiling of the LRRK2 KO kidneys using immunohistochemistry, proteomic and lipidomic analyses show a massive accumulation of hemoglobin and lipofuscin in renal tubules that account for the pigmentation. The proximal tubules demonstrate a corresponding up-regulation of the cytoprotective protein heme oxygenase-1 (HO-1) which is capable of mitigating acute kidney injury. The unusual kidney pathology of LRRK2 KO rats highlights several novel physiological roles for LRRK2 and provides indirect evidence for HO-1 expression as a protective mechanism in acute kidney injury in LRRK2 deficiency.


Subject(s)
Kidney Diseases/genetics , Protein Serine-Threonine Kinases/genetics , Rhabdomyolysis/genetics , Animals , Cytoprotection , Epithelial Cells/metabolism , Genetic Predisposition to Disease , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Kidney Diseases/etiology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Male , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , Proteomics , Rats , Rhabdomyolysis/complications , Up-Regulation
11.
Am J Kidney Dis ; 69(4): 531-545, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28139396

ABSTRACT

A common clinical condition, acute kidney injury (AKI) significantly influences morbidity and mortality, particularly in critically ill patients. The pathophysiology of AKI is complex and involves multiple pathways, including inflammation, autophagy, cell-cycle progression, and oxidative stress. Recent evidence suggests that a single insult to the kidney significantly enhances the propensity to develop chronic kidney disease. Therefore, the generation of effective therapies against AKI is timely. In this context, the cytoprotective effects of heme oxygenase 1 (HO-1) in animal models of AKI are well documented. HO-1 modulates oxidative stress, autophagy, and inflammation and regulates the progression of cell cycle via direct and indirect mechanisms. These beneficial effects of HO-1 induction during AKI are mediated in part by the by-products of the HO reaction (iron, carbon monoxide, and bile pigments). This review highlights recent advances in the molecular mechanisms of HO-1-mediated cytoprotection and discusses the translational potential of HO-1 induction in AKI.


Subject(s)
Acute Kidney Injury/blood , Acute Kidney Injury/therapy , Heme Oxygenase-1/blood , Accidents, Occupational , Adult , Autophagy/physiology , Cell Cycle Checkpoints/physiology , Enzyme Induction/physiology , Heme Oxygenase-1/physiology , Humans , Inflammation/blood , Inflammation/therapy , Leg Injuries/complications , Male , Oxidative Stress/physiology , Rhabdomyolysis/blood , Rhabdomyolysis/therapy , Translational Research, Biomedical
12.
Am J Physiol Renal Physiol ; 310(5): F385-94, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26672618

ABSTRACT

Heme oxygenase-1 (HO-1) is a cytoprotective enzyme that catalyzes the breakdown of heme to biliverdin, carbon monoxide, and iron. The beneficial effects of HO-1 expression are not merely due to degradation of the pro-oxidant heme but are also credited to the by-products that have potent, protective effects, including antioxidant, anti-inflammatory, and prosurvival properties. This is well reflected in the preclinical animal models of injury in both renal and nonrenal settings. However, excessive accumulation of the by-products can be deleterious and lead to mitochondrial toxicity and oxidative stress. Therefore, use of the HO system in alleviating injury merits a targeted approach. Based on the higher susceptibility of the proximal tubule segment of the nephron to injury, we generated transgenic mice using cre-lox technology to enable manipulation of HO-1 (deletion or overexpression) in a cell-specific manner. We demonstrate the validity and feasibility of these mice by breeding them with proximal tubule-specific Cre transgenic mice. Similar to previous reports using chemical modulators and global transgenic mice, we demonstrate that whereas deletion of HO-1, specifically in the proximal tubules, aggravates structural and functional damage during cisplatin nephrotoxicity, selective overexpression of HO-1 in proximal tubules is protective. At the cellular level, cleaved caspase-3 expression, a marker of apoptosis, and p38 signaling were modulated by HO-1. Use of these transgenic mice will aid in the evaluation of the effects of cell-specific HO-1 expression in response to injury and assist in the generation of targeted approaches that will enhance recovery with reduced, unwarranted adverse effects.


Subject(s)
Acute Kidney Injury/prevention & control , Cisplatin , Heme Oxygenase-1/metabolism , Kidney Tubules, Proximal/enzymology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/enzymology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Apoptosis , Caspase 3/metabolism , Disease Models, Animal , Genotype , Heme Oxygenase-1/deficiency , Heme Oxygenase-1/genetics , Kidney Tubules, Proximal/pathology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Necrosis , Phenotype , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Am J Physiol Renal Physiol ; 310(10): F1136-47, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26911846

ABSTRACT

Acute kidney injury (AKI) is one of the leading causes of in-hospital morbidity and mortality, particularly in critically ill patients. Although our understanding of AKI at the molecular level remains limited due to its complex pathophysiology, recent advances in both quantitative and spatial mass spectrometric approaches offer new opportunities to assess the significance of renal metabolomic changes in AKI models. In this study, we evaluated lipid changes in early ischemia-reperfusion (IR)-related AKI in mice by using sequential window acquisition of all theoretical spectra (SWATH)-mass spectrometry (MS) lipidomics. We found a significant increase in two abundant ether-linked phospholipids following IR at 6 h postinjury, a plasmanyl choline, phosphatidylcholine (PC) O-38:1 (O-18:0, 20:1), and a plasmalogen, phosphatidylethanolamine (PE) O-42:3 (O-20:1, 22:2). Both of these lipids correlated with the severity of AKI as measured by plasma creatinine. In addition to many more renal lipid changes associated with more severe AKI, PC O-38:1 elevations were maintained at 24 h post-IR, while renal PE O-42:3 levels decreased, as were all ether PEs detected by SWATH-MS at this later time point. To further assess the significance of this early increase in PC O-38:1, we used matrix-assisted laser desorption ionization imaging mass spectrometry (MALDI-IMS) to determine that it occurred in proximal tubules, a region of the kidney that is most prone to IR injury and also rich in the rate-limiting enzymes involved in ether-linked phospholipid biosynthesis. Use of SWATH-MS lipidomics in conjunction with MALDI-IMS for lipid localization will help in elucidating the role of lipids in the pathobiology of AKI.


Subject(s)
Acute Kidney Injury/metabolism , Lipid Metabolism , Metabolomics/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Acute Kidney Injury/etiology , Animals , Male , Mice, Inbred C57BL , Reperfusion Injury/metabolism
14.
J Am Soc Nephrol ; 26(9): 2139-51, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25677389

ABSTRACT

Renal ischemia-reperfusion injury is mediated by a complex cascade of events, including the immune response, that occur secondary to injury to renal epithelial cells. We tested the hypothesis that heme oxygenase-1 (HO-1) expression, which is protective in ischemia-reperfusion injury, regulates trafficking of myeloid-derived immune cells in the kidney. Age-matched male wild-type (HO-1(+/+)), HO-1-knockout (HO-1(-/-)), and humanized HO-1-overexpressing (HBAC) mice underwent bilateral renal ischemia for 10 minutes. Ischemia-reperfusion injury resulted in significantly worse renal structure and function and increased mortality in HO-1(-/-) mice. In addition, there were more macrophages (CD45(+) CD11b(hi)F4/80(lo)) and neutrophils (CD45(+) CD11b(hi) MHCII(-) Gr-1(hi)) in HO-1(-/-) kidneys than in sham and HO-1(+/+) control kidneys subjected to ischemia-reperfusion. However, ischemic injury resulted in a significant decrease in the intrarenal resident dendritic cell (DC; CD45(+)MHCII(+)CD11b(lo)F4/80(hi)) population in HO-1(-/-) kidneys compared with controls. Syngeneic transplant experiments utilizing green fluorescent protein-positive HO-1(+/+) or HO-1(-/-) donor kidneys and green fluorescent protein-negative HO-1(+/+) recipients confirmed increased migration of the resident DC population from HO-1(-/-) donor kidneys, compared to HO-1(+/+) donor kidneys, to the peripheral lymphoid organs. This effect on renal DC migration was corroborated in myeloid-specific HO-1(-/-) mice subjected to bilateral ischemia. These mice also displayed impaired renal recovery and increased fibrosis at day 7 after injury. These results highlight an important role for HO-1 in orchestrating the trafficking of myeloid cells in AKI, which may represent a key pathway for therapeutic intervention.


Subject(s)
Acute Kidney Injury/pathology , Cell Movement/physiology , Heme Oxygenase-1/physiology , Myeloid Cells , Acute Kidney Injury/etiology , Acute Kidney Injury/physiopathology , Animals , Cell Movement/genetics , Dendritic Cells , Fibrosis , Heme Oxygenase-1/genetics , Immunity, Innate , Interleukin-6/metabolism , Ischemia/etiology , Kidney/blood supply , Kidney/pathology , Lymph Nodes/pathology , Macrophages , Male , Mice , Mice, Knockout , Mice, Transgenic , Myeloid Cells/metabolism , Neutrophils , Reperfusion Injury/complications , Spleen/pathology , Tumor Necrosis Factor-alpha/metabolism
15.
Kidney Int ; 88(1): 95-108, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25874599

ABSTRACT

Inflammation culminating in fibrosis contributes to progressive kidney disease. Cross-talk between the tubular epithelium and interstitial cells regulates inflammation by a coordinated release of cytokines and chemokines. Here we studied the role of heme oxygenase-1 (HO-1) and the heavy subunit of ferritin (FtH) in macrophage polarization and renal inflammation. Deficiency in HO-1 was associated with increased FtH expression, accumulation of macrophages with a dysregulated polarization profile, and increased fibrosis following unilateral ureteral obstruction in mice: a model of renal inflammation and fibrosis. Macrophage polarization in vitro was predominantly dependent on FtH expression in isolated bone marrow-derived mouse monocytes. Using transgenic mice with conditional deletion of FtH in the proximal tubules (FtH(PT-/-)) or myeloid cells (FtH(LysM-/-)), we found that myeloid FtH deficiency did not affect polarization or accumulation of macrophages in the injured kidney compared with wild-type (FtH(+/+)) controls. However, tubular FtH deletion led to a marked increase in proinflammatory macrophages. Furthermore, injured kidneys from FtH(PT-/-) mice expressed significantly higher levels of inflammatory chemokines and fibrosis compared with kidneys from FtH(+/+) and FtH(LysM-/-) mice. Thus, there are differential effects of FtH in macrophages and epithelial cells, which underscore the critical role of FtH in tubular-macrophage cross-talk during kidney injury.


Subject(s)
Apoferritins/genetics , Epithelial Cells/metabolism , Heme Oxygenase-1/deficiency , Kidney/pathology , Macrophages/physiology , Myeloid Cells/metabolism , Nephritis/metabolism , Animals , Apoferritins/metabolism , Cells, Cultured , Chemokine CCL2/metabolism , Disease Models, Animal , Fibrosis , Gene Expression , Heme Oxygenase-1/genetics , Interleukin-10/metabolism , Interleukin-6/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Macrophage Activation , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nephritis/etiology , RNA, Messenger/metabolism , Ureteral Obstruction/complications
16.
Am J Pathol ; 182(4): 1239-47, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23399488

ABSTRACT

The origin of the myofibroblast in fibrotic lung disease is uncertain, and no effective medical therapy for fibrosis exists. We have previously demonstrated that transforming growth factor-ß1 (TGF-ß1) induces pleural mesothelial cell (PMC) transformation into myofibroblasts and haptotactic migration in vitro. Whether PMC differentiation and migration occurs in vivo, and whether this response can be modulated for therapeutic benefit, is unknown. Here, using mice recombinant for green fluorescent protein (GFP) driven by the Wilms tumor-1 (WT-1) promoter, we demonstrate PMC trafficking into the lung and differentiation into myofibroblasts. Carbon monoxide or the induction of heme oxygenase-1 (HO-1) inhibited the expression of myofibroblast markers, contractility, and haptotaxis in PMCs treated with TGF-ß1. Intrapleural HO-1 induction inhibited PMC migration after intratracheal fibrogenic injury. PMCs from patients with idiopathic pulmonary fibrosis (IPF) exhibited increased expression of myofibroblast markers and enhanced contractility and haptotaxis, compared with normal PMCs. Carbon monoxide reversed this IPF PMC profibrotic phenotype. WT-1-expressing cells were present within fibrotic regions of the lungs in IPF subjects, supporting a role for PMC differentiation and trafficking as contributors to the myofibroblast population in lung fibrosis. Our findings also support a potential role for pleural-based therapies to modulate pleural mesothelial activation and parenchymal fibrosis progression.


Subject(s)
Cell Differentiation , Cell Movement , Epithelial Cells/pathology , Idiopathic Pulmonary Fibrosis/pathology , Lung Injury/pathology , Pleura/pathology , Administration, Inhalation , Animals , Biomarkers/metabolism , Carbon Monoxide/metabolism , Cell Differentiation/drug effects , Cell Movement/drug effects , Enzyme Induction/drug effects , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/deficiency , Hemin/pharmacology , Humans , Idiopathic Pulmonary Fibrosis/enzymology , Imidazoles/pharmacology , Lung/pathology , Lung Injury/enzymology , Mice , Mice, Inbred C57BL , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology , Organometallic Compounds/pharmacology , Transforming Growth Factor beta1/pharmacology
17.
Skelet Muscle ; 14(1): 13, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38867250

ABSTRACT

BACKGROUND: Adult muscle-resident myogenic stem cells, satellite cells (SCs), that play non-redundant role in muscle regeneration, are intrinsically impaired in Duchenne muscular dystrophy (DMD). Previously we revealed that dystrophic SCs express low level of anti-inflammatory and anti-oxidative heme oxygenase-1 (HO-1, HMOX1). Here we assess whether targeted induction of HMOX1 affect SC function and alleviates hallmark symptoms of DMD. METHODS: We generated double-transgenic mouse model (mdx;HMOX1Pax7Ind) that allows tamoxifen (TX)-inducible HMOX1 expression in Pax7 positive cells of dystrophic muscles. Mdx;HMOX1Pax7Ind and control mdx mice were subjected to 5-day TX injections (75 mg/kg b.w.) followed by acute exercise protocol with high-speed treadmill (12 m/min, 45 min) and downhill running to worsen skeletal muscle phenotype and reveal immediate effects of HO-1 on muscle pathology and SC function. RESULTS: HMOX1 induction caused a drop in SC pool in mdx;HMOX1Pax7Ind mice (vs. mdx counterparts), while not exaggerating the effect of physical exercise. Upon physical exercise, the proliferation of SCs and activated CD34- SC subpopulation, was impaired in mdx mice, an effect that was reversed in mdx;HMOX1Pax7Ind mice, however, both in vehicle- and TX-treated animals. This corresponded to the pattern of HO-1 expression in skeletal muscles. At the tissue level, necrotic events of selective skeletal muscles of mdx mice and associated increase in circulating levels of muscle damage markers were blunted in HO-1 transgenic animals which showed also anti-inflammatory cytokine profile (vs. mdx). CONCLUSIONS: Targeted expression of HMOX1 plays protective role in DMD and alleviates dystrophic muscle pathology.


Subject(s)
Heme Oxygenase-1 , Mice, Inbred mdx , Mice, Transgenic , Muscle, Skeletal , Muscular Dystrophy, Duchenne , Satellite Cells, Skeletal Muscle , Animals , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Mice , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Male , Mice, Inbred C57BL , Physical Conditioning, Animal , Membrane Proteins
18.
Am J Physiol Renal Physiol ; 305(3): F255-64, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23720344

ABSTRACT

Mitochondria are both a source and target of the actions of reactive oxygen species and possess a complex system of inter-related antioxidants that control redox signaling and protect against oxidative stress. Interestingly, the antioxidant enzyme heme oxygenase-1 (HO-1) is not present in the mitochondria despite the fact that the organelle is the site of heme synthesis and contains multiple heme proteins. Detoxification of heme is an important protective mechanism since the reaction of heme with hydrogen peroxide generates pro-oxidant ferryl species capable of propagating oxidative stress and ultimately cell death. We therefore hypothesized that a mitochondrially localized HO-1 would be cytoprotective. To test this, we generated a mitochondria-targeted HO-1 cell line by transfecting HEK293 cells with a plasmid construct containing the manganese superoxide dismutase mitochondria leader sequence fused to HO-1 cDNA (Mito-HO-1). Nontargeted HO-1-overexpressing cells were generated by transfecting HO-1 cDNA (HO-1) or empty vector (Vector). Mitochondrial localization of HO-1 with increased HO activity in the mitochondrial fraction of Mito-HO-1 cells was observed, but a significant decrease in the expression of heme-containing proteins occurred in these cells. Both cytosolic HO-1- and Mito-HO-1-expressing cells were protected against hypoxia-dependent cell death and loss of mitochondrial membrane potential, but these effects were more pronounced with Mito-HO-1. Furthermore, decrement in production of tricarboxylic acid cycle intermediates following hypoxia was significantly mitigated in Mito-HO-1 cells. These data suggest that specific mitochondrially targeted HO-1 under acute pathological conditions may have beneficial effects, but the selective advantage of long-term expression is constrained by a negative impact on the synthesis of heme-containing mitochondrial proteins.


Subject(s)
Epithelial Cells/metabolism , Heme Oxygenase-1/metabolism , Kidney/metabolism , Mitochondria/enzymology , Aerobiosis/physiology , Blotting, Western , Cell Survival/drug effects , Citrate (si)-Synthase/metabolism , Citric Acid Cycle/physiology , Cytochromes c/metabolism , Electron Transport Complex IV/metabolism , Epithelial Cells/enzymology , HEK293 Cells , Heme Oxygenase-1/physiology , Humans , Immunohistochemistry , Kidney/cytology , Kidney/enzymology , Membrane Potential, Mitochondrial/physiology , Oxidative Stress/physiology , Plasmids/genetics , Plasmids/physiology , Voltage-Dependent Anion Channels/metabolism
19.
Lab Invest ; 93(8): 868-79, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23732814

ABSTRACT

The protective effect of heme oxygenase-1 (HO-1) expression in cardiovascular disease has been previously demonstrated using transgenic animal models in which HO-1 is constitutively overexpressed in the heart. However, the temporal requirements for protection by HO-1 induction relative to injury have not been investigated, but are essential to employ HO-1 as a therapeutic strategy in human cardiovascular disease states. Therefore, we generated mice with cardiac-specific, tamoxifen (TAM)-inducible overexpression of a human HO-1 (hHO-1) transgene (myosin heavy chain (MHC)-HO-1 mice) by breeding mice with cardiac-specific expression of a TAM-inducible Cre recombinase (MHC-Cre mice), with mice containing an hHO-1 transgene preceded by a floxed-stop signal. MHC-HO-1 mice overexpress HO-1 mRNA and the enzymatically active protein following TAM administration (40 mg/kg body weight on 2 consecutive days). In MHC-Cre controls, TAM administration leads to severe, acute cardiac toxicity, cardiomyocyte necrosis, and 80% mortality by day 3. This cardiac toxicity is accompanied by a significant increase in inflammatory cells in the heart that are predominantly neutrophils. In MHC-HO-1 mice, HO-1 overexpression ameliorates the depression of cardiac function and high mortality rate observed in MHC-Cre mice following TAM administration and attenuates cardiomyocyte necrosis and neutrophil infiltration. These results highlight that HO-1 induction is sufficient to prevent the depression of cardiac function observed in mice with TAM-inducible Cre recombinase expression by protecting the heart from necrosis and neutrophil infiltration. These findings are important because MHC-Cre mice are widely used in cardiovascular research despite the limitations imposed by Cre-induced cardiac toxicity, and also because inflammation is an important pathological component of many human cardiovascular diseases.


Subject(s)
Heart Diseases/prevention & control , Heme Oxygenase-1/metabolism , Integrases/biosynthesis , Myocardium/enzymology , Acute Disease , Animals , Antineoplastic Agents, Hormonal/pharmacology , Disease Models, Animal , Enzyme Induction , Female , Heart/drug effects , Heart/physiopathology , Heart Diseases/enzymology , Heart Diseases/mortality , Heart Diseases/pathology , Heme Oxygenase-1/genetics , Humans , Integrases/genetics , Longevity/drug effects , Male , Mice , Mice, Transgenic , Myocardium/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Necrosis/chemically induced , Neutrophils/drug effects , Neutrophils/pathology , Survival Rate , Tamoxifen/pharmacology
20.
Trans Am Clin Climatol Assoc ; 124: 111-22, 2013.
Article in English | MEDLINE | ID: mdl-23874015

ABSTRACT

Tissue injury may result as a consequence of a physical, chemical, or biological insult. Such injury recruits an adaptive response to restore homeostasis and protect against further injury. One of the most prompt protective and adaptive responses by all tissues is the robust activation of the highly inducible, anti-inflammatory, anti-oxidant, and anti-apoptotic protein, heme oxygenase-1 (HO-1). HO-1, a microsomal enzyme, catalyzes the breakdown of pro-oxidant heme, which is released from heme proteins to equimolar quantities of iron, carbon monoxide, and biliverdin. Biliverdin is converted to bilirubin by biliverdin reductase. The beneficial effects of HO-1 expression are not merely due to heme degradation but are also attributed to the cytoprotective properties of the byproducts of the reaction. Manipulation of this enzymatic system in a myriad of disease models has provided substantial evidence to support its role as a cytoprotective enzyme and is therefore an emerging therapeutic molecule.


Subject(s)
Anemia, Hemolytic/metabolism , Growth Disorders/metabolism , Heme Oxygenase-1/deficiency , Heme Oxygenase-1/metabolism , Iron Metabolism Disorders/metabolism , Animals , Gene Expression Regulation, Enzymologic , Heme Oxygenase-1/genetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL