Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Biomolecules ; 10(1)2020 01 07.
Article in English | MEDLINE | ID: mdl-31936109

ABSTRACT

Autophagy is a dynamic process by which intracellular damaged macromolecules and organelles are degraded and recycled for the synthesis of new cellular components. Basal autophagy in the kidney acts as a quality control system and is vital for cellular metabolic and organelle homeostasis. Under pathological conditions, autophagy facilitates cellular adaptation; however, activation of autophagy in response to renal injury may be insufficient to provide protection, especially under dysregulated conditions. Kidney-specific deletion of Atg genes in mice has consistently demonstrated worsened acute kidney injury (AKI) outcomes supporting the notion of a pro-survival role of autophagy. Recent studies have also begun to unfold the role of autophagy in progressive renal disease and subsequent fibrosis. Autophagy also influences tubular cell death in renal injury. In this review, we reported the current understanding of autophagy regulation and its role in the pathogenesis of renal injury. In particular, the classic mammalian target of rapamycin (mTOR)-dependent signaling pathway and other mTOR-independent alternative signaling pathways of autophagy regulation were described. Finally, we summarized the impact of autophagy activation on different forms of cell death, including apoptosis and regulated necrosis, associated with the pathophysiology of renal injury. Understanding the regulatory mechanisms of autophagy would identify important targets for therapeutic approaches.


Subject(s)
Acute Kidney Injury/pathology , Autophagy/physiology , Kidney Diseases/physiopathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/therapy , Animals , Apoptosis , Fibrosis , Homeostasis , Humans , Kidney/pathology , Signal Transduction
2.
Int J Mol Sci ; 20(15)2019 Aug 03.
Article in English | MEDLINE | ID: mdl-31382550

ABSTRACT

Reactive oxygen species (ROS) are highly reactive signaling molecules that maintain redox homeostasis in mammalian cells. Dysregulation of redox homeostasis under pathological conditions results in excessive generation of ROS, culminating in oxidative stress and the associated oxidative damage of cellular components. ROS and oxidative stress play a vital role in the pathogenesis of acute kidney injury and chronic kidney disease, and it is well documented that increased oxidative stress in patients enhances the progression of renal diseases. Oxidative stress activates autophagy, which facilitates cellular adaptation and diminishes oxidative damage by degrading and recycling intracellular oxidized and damaged macromolecules and dysfunctional organelles. In this review, we report the current understanding of the molecular regulation of autophagy in response to oxidative stress in general and in the pathogenesis of kidney diseases. We summarize how the molecular interactions between ROS and autophagy involve ROS-mediated activation of autophagy and autophagy-mediated reduction of oxidative stress. In particular, we describe how ROS impact various signaling pathways of autophagy, including mTORC1-ULK1, AMPK-mTORC1-ULK1, and Keap1-Nrf2-p62, as well as selective autophagy including mitophagy and pexophagy. Precise elucidation of the molecular mechanisms of interactions between ROS and autophagy in the pathogenesis of renal diseases may identify novel targets for development of drugs for preventing renal injury.


Subject(s)
Acute Kidney Injury/genetics , Autophagy/genetics , Oxidative Stress/genetics , Renal Insufficiency, Chronic/genetics , AMP-Activated Protein Kinase Kinases , Acute Kidney Injury/pathology , Autophagy-Related Protein-1 Homolog/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Kelch-Like ECH-Associated Protein 1/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , NF-E2-Related Factor 2/genetics , Protein Kinases/genetics , RNA-Binding Proteins/genetics , Reactive Oxygen Species/metabolism , Renal Insufficiency, Chronic/pathology
3.
Cytokine ; 114: 18-25, 2019 02.
Article in English | MEDLINE | ID: mdl-30580156

ABSTRACT

Meprin metalloendopeptidases, comprising α and ß isoforms, are widely expressed in mammalian cells and organs including kidney, intestines, lungs, skin, and bladder, and in a variety of immune cells and cancer cells. Meprins proteolytically process many inflammatory mediators, including cytokines, chemokines, and other bioactive proteins and peptides that control the function of immune cells. The knowledge of meprin-mediated processing of inflammatory mediators and other target substrates provides a pathophysiologic link for the involvement of meprins in the pathogenesis of many inflammatory disorders. Meprins are now known to play important roles in inflammatory diseases including acute kidney injury, sepsis, urinary tract infections, bladder inflammation, and inflammatory bowel disease. The proteolysis of epithelial and endothelial barriers including cell junctional proteins by meprins promotes leukocyte influx into areas of tissue damage to result in inflammation. Meprins degrade extracellular matrix proteins; this ability of meprins is implicated in the cell migration of leukocytes and the invasion of tumor cells that express meprins. Proteolytic processing and maturation of procollagens provides evidence that meprins are involved in collagen maturation and deposition in the fibrotic processes involved in the formation of keloids and hypertrophic scars and lung fibrosis. This review highlights recent progress in understanding the role of meprins in inflammatory disorders in both human and mouse models.


Subject(s)
Inflammation/metabolism , Inflammation/pathology , Metalloproteases/metabolism , Amino Acid Sequence , Animals , Cytokines/metabolism , Extracellular Matrix Proteins/metabolism , Humans , Metalloproteases/chemistry , Proteolysis
4.
PLoS One ; 11(12): e0165576, 2016.
Article in English | MEDLINE | ID: mdl-27973558

ABSTRACT

Patients with chronic kidney disease (CKD) have high risk of cardiovascular complications. Plasma levels of carbamylated proteins produced by urea-derived isocyanate or thiocyanate are elevated in CKD patients and that they are significant predictors of cardiovascular events and all-cause mortality. Carbamylated LDL (cLDL) has pro-atherogenic properties and is known to affect major biological processes relevant to atherosclerosis including endothelial cell injury. The underlying mechanisms of cLDL-induced endothelial cell injury are not well understood. Although autophagy has been implicated in atherosclerosis, cLDL-mediated induction of autophagy and its role in endothelial cell injury is unknown. Our studies demonstrate that human coronary artery endothelial cells (HCAECs) respond to cLDL by specific induction of key autophagy proteins including LC3-I, beclin-1, Atg5, formation of lipid-conjugated LC3-II protein, and formation of punctate dots of autophagosome-associated LC3-II. We demonstrated that autophagy induction is an immediate response to cLDL and occurred in a dose and time-dependent manner. Inhibition of cLDL-induced autophagy by a specific siRNA to LC3 as well as by an autophagy inhibitor provided protection from cLDL-induced cell death and DNA fragmentation. Our studies demonstrate that autophagy plays an important role in cLDL-mediated endothelial cell injury and may provide one of the underlying mechanisms for the pathogenesis of cLDL-induced atherosclerosis in CKD patients.


Subject(s)
Autophagy/drug effects , Endothelial Cells/drug effects , Lipoproteins, LDL/pharmacology , Renal Insufficiency, Chronic/physiopathology , Adenine/analogs & derivatives , Adenine/pharmacology , Atherosclerosis , Autophagosomes , Autophagy-Related Protein 5/metabolism , Beclin-1/metabolism , Cell Death , Cells, Cultured , Coronary Vessels/cytology , Cytosol/metabolism , DNA Fragmentation , Endothelial Cells/cytology , Humans , L-Lactate Dehydrogenase/metabolism , Lipids/chemistry , Microcirculation , Microscopy, Fluorescence , Microtubule-Associated Proteins/metabolism , RNA, Small Interfering/metabolism
5.
Biol Chem ; 397(11): 1135-1146, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27279059

ABSTRACT

Kallikrein-related peptidase 7 (KLK7) is a serine protease encoded within the kallikrein gene cluster located on human chromosome region 19q13.3-13.4. KLK7 is overexpressed in human pancreatic ductal adenocarcinomas (PDACs), but not in normal pancreas. Examination of KLK7 mRNA levels in pancreatic cancer cell lines revealed that it is readily detected in MIA PaCa-2 and PK-1 cells, but not in Panc-1 cells. Treatment of Panc-1 cells with the histone deacetylase (HDAC) inhibitor trichostatin A (TSA) significantly induced KLK7 mRNA expression. Similarly, KLK7 is highly expressed in cervical cancer cells, but its expression in the human cervical cancer cell line HeLa is only detected following TSA treatment. Promoter deletion analysis revealed that the proximal -238 promoter region, containing a putative Sp1-binding site, was sufficient for TSA activation of luciferase reporter activity, which was abrogated by the disruption of the Sp1-binding sequence. Consistent with the notion that TSA induced KLK7 expression via Sp1, co-expression of Sp1 with the KLK7-promoter/luciferase construct produced a significant increase in reporter activity. Chromatin immunoprecipitation (ChIP) analysis revealed enriched Sp1 occupancy on the KLK7 promoter following TSA treatment. Similarly, ChIP analysis showed the histone active mark, H3K4Me3, in the KLK7 promoter region was significantly increased after exposure to TSA.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/genetics , Kallikreins/genetics , Pancreatic Neoplasms/pathology , Uterine Cervical Neoplasms/pathology , Cell Line, Tumor , Epigenesis, Genetic/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Pancreatic Neoplasms/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sp1 Transcription Factor/genetics , Transcription, Genetic , Uterine Cervical Neoplasms/genetics
6.
Kidney Int ; 89(4): 779-91, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26924060

ABSTRACT

Autophagy is a conserved multistep pathway that degrades and recycles damaged organelles and macromolecules to maintain intracellular homeostasis. The autophagy pathway is upregulated under stress conditions including cell starvation, hypoxia, nutrient and growth-factor deprivation, endoplasmic reticulum stress, and oxidant injury, most of which are involved in the pathogenesis of acute kidney injury (AKI). Recent studies demonstrate that basal autophagy in the kidney is vital for the normal homeostasis of the proximal tubules. Deletion of key autophagy proteins impaired renal function and increased p62 levels and oxidative stress. In models of AKI, autophagy deletion in proximal tubules worsened tubular injury and renal function, highlighting that autophagy is renoprotective in models of AKI. In addition to nonselective sequestration of autophagic cargo, autophagy can facilitate selective degradation of damaged organelles, particularly mitochondrial degradation through the process of mitophagy. Damaged mitochondria accumulate in autophagy-deficient kidneys of mice subjected to ischemia-reperfusion injury, but the precise mechanisms of regulation of mitophagy in AKI are not yet elucidated. Recent progress in identifying the interplay of autophagy, apoptosis, and regulated necrosis has revived interest in examining shared pathways/molecules in this crosstalk during the pathogenesis of AKI. Autophagy and its associated pathways pose potentially unique targets for therapeutic interventions in AKI.


Subject(s)
Acute Kidney Injury/etiology , Autophagy , Animals , Antineoplastic Agents/adverse effects , Apoptosis , Caspases/metabolism , Cisplatin/adverse effects , Humans , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/antagonists & inhibitors , Sepsis/complications , TOR Serine-Threonine Kinases/antagonists & inhibitors
7.
Biochem Biophys Rep ; 8: 146-150, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28955950

ABSTRACT

Monocyte chemotactic protein 1 (CCL2/MCP-1) is a small chemokine involved in the recruitment and trafficking of mononuclear immune cells to inflammation sites. Our studies demonstrate that the metalloendopeptidases meprin A (purified from kidney cortex), recombinant meprin α, and recombinant meprin ß can all process CCL2/MCP-1. The cleavage sites were determined by amino acid sequencing and mass spectrometry analysis of the generated products, and the biological activity of the products was evaluated by chemotactic migration assay using THP-1 cells. The cleavage sites generated by the meprin isoforms revealed that meprin A and meprin α cleaved the N-terminal domain of mouse CCL2/MCP-1 at the Asn6 and Ala7 bond, resulting in significant reduction in the chemotactic activity of the cleaved CCL2/MCP-1. Meprin ß was unable to cleave the N-terminus of mouse CCL2/MCP-1 but cleaved the C-terminal region between Ser74 and Glu75. Human CCL2/MCP-1 that lacks the murine C-terminal region was also cleaved by meprin α at the N-terminus resulting in significant loss of CCL2/MCP-1 biological activity, whereas meprin ß did not affect the biological activity. These studies suggest that meprin α and meprin ß may play important roles in regulating the CCL2/MCP-1 chemokine activity during inflammation.

8.
PLoS One ; 10(10): e0140025, 2015.
Article in English | MEDLINE | ID: mdl-26444017

ABSTRACT

We examined whether endoplasmic reticulum (ER) stress-induced autophagy provides cytoprotection from renal tubular epithelial cell injury due to oxidants and chemical hypoxia in vitro, as well as from ischemia-reperfusion (IR) injury in vivo. We demonstrate that the ER stress inducer tunicamycin triggers an unfolded protein response, upregulates ER chaperone Grp78, and activates the autophagy pathway in renal tubular epithelial cells in culture. Inhibition of ER stress-induced autophagy accelerated caspase-3 activation and cell death suggesting a pro-survival role of ER stress-induced autophagy. Compared to wild-type cells, autophagy-deficient MEFs subjected to ER stress had enhanced caspase-3 activation and cell death, a finding that further supports the cytoprotective role of ER stress-induced autophagy. Induction of autophagy by ER stress markedly afforded cytoprotection from oxidants H2O2 and tert-Butyl hydroperoxide and from chemical hypoxia induced by antimycin A. In contrast, inhibition of ER stress-induced autophagy or autophagy-deficient cells markedly enhanced cell death in response to oxidant injury and chemical hypoxia. In mouse kidney, similarly to renal epithelial cells in culture, tunicamycin triggered ER stress, markedly upregulated Grp78, and activated autophagy without impairing the autophagic flux. In addition, ER stress-induced autophagy markedly ameliorated renal IR injury as evident from significant improvement in renal function and histology. Inhibition of autophagy by chloroquine markedly increased renal IR injury. These studies highlight beneficial impact of ER stress-induced autophagy in renal ischemia-reperfusion injury both in vitro and in vivo.


Subject(s)
Autophagy/drug effects , Hypoxia/drug therapy , Kidney/drug effects , Protective Agents/therapeutic use , Renal Insufficiency/drug therapy , Reperfusion Injury/drug therapy , Tunicamycin/therapeutic use , Adenosine Triphosphate/metabolism , Animals , Caspases/metabolism , Cell Line , Cytoprotection/drug effects , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Heat-Shock Proteins/metabolism , Hypoxia/complications , Hypoxia/metabolism , Hypoxia/pathology , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Oxidants/adverse effects , Oxidative Stress/drug effects , Renal Insufficiency/complications , Renal Insufficiency/metabolism , Renal Insufficiency/pathology , Reperfusion Injury/complications , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Unfolded Protein Response/drug effects
9.
PLoS One ; 10(9): e0139226, 2015.
Article in English | MEDLINE | ID: mdl-26414017

ABSTRACT

Cardiovascular disease is the largest cause of morbidity and mortality among patients with chronic kidney disease (CKD) and end-stage kidney disease, with nearly half of all deaths attributed to cardiovascular disease. Hydroxychloroquine (HCQ), an anti-inflammatory drug, has been shown to have multiple pleiotropic actions relevant to atherosclerosis. We conducted a proof-of-efficacy study to evaluate the effects of hydroxychloroquine in an animal model of atherosclerosis in ApoE knockout mice with and without chronic kidney disease. Forty male, 6-week-old mice were divided into four groups in a 2 x 2 design: sham placebo group; sham treatment group; CKD placebo group; and CKD treatment group. CKD was induced by a two-step surgical procedure. All mice received a high-fat diet through the study duration and were sacrificed after 16 weeks of therapy. Mice were monitored with ante-mortem ultrasonic echography (AUE) for atherosclerosis and vascular stiffness and with post-mortem histology studies for atherosclerosis. Therapy with HCQ significantly reduced the severity of atherosclerosis in CKD mice and sham treated mice. HCQ reduced the area of aortic atherosclerosis on en face examination by approximately 60% in HCQ treated groups compared to the non-treated groups. Additionally, therapy with HCQ resulted in significant reduction in vascular endothelial dysfunction with improvement in vascular elasticity and flow patterns and better-preserved vascular wall thickness across multiple vascular beds. More importantly, we found that presence of CKD had no mitigating effect on HCQ's anti-atherosclerotic and vasculoprotective effects. These beneficial effects were not due to any significant effect of HCQ on inflammation, renal function, or lipid profile at the end of 16 weeks of therapy. This study, which demonstrates structural and functional protection against atherosclerosis by HCQ, provides a rationale to evaluate its use in CKD patients. Further studies are needed to define the exact mechanisms through which HCQ confers these benefits.


Subject(s)
Atherosclerosis/drug therapy , Atherosclerosis/physiopathology , Hydroxychloroquine/therapeutic use , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/drug therapy , Vascular Stiffness , Animals , Aorta/pathology , Aorta/physiopathology , Atherosclerosis/blood , Atherosclerosis/complications , Bilirubin/blood , Blood Glucose/metabolism , Elasticity , Hydroxychloroquine/pharmacology , Inflammation/pathology , Male , Mice, Inbred C57BL , Postmortem Changes , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/physiopathology , Urea/blood , Vascular Stiffness/drug effects
10.
Toxicol Lett ; 236(2): 110-6, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-25957482

ABSTRACT

Meprins are oligomeric metalloproteinases that are abundantly expressed in the brush-border membranes of renal proximal tubules. During acute kidney injury (AKI) induced by cisplatin or ischemia-reperfusion, membrane-bound meprins are shed and their localization is altered from the apical membranes toward the basolateral surface of the proximal tubules. Meprins are capable of cleaving basement membrane proteins in vitro, however, it is not known whether meprins are able to degrade extracellular matrix proteins under pathophysiological conditions in vivo. The present study demonstrates that a basement membrane protein, nidogen-1, is cleaved and excreted in the urine of mice subjected to cisplatin-induced nephrotoxicity, a model of AKI. Cleaved nidogen-1 was not detected in the urine of untreated mice, but during the progression of cisplatin nephrotoxicity, the excretion of cleaved nidogen-1 increased in a time-dependent manner. The meprin inhibitor actinonin markedly prevented urinary excretion of the cleaved nidogen-1. In addition, meprin ß-deficient mice, but not meprin α-deficient mice, subjected to cisplatin nephrotoxicity significantly suppressed excretion of cleaved nidogen-1, further suggesting that meprin ß is involved in the cleavage of nidogen-1. These studies provide strong evidence for a pathophysiological link between meprin ß and urinary excretion of cleaved nidogen-1 during cisplatin-induced AKI.


Subject(s)
Acute Kidney Injury/chemically induced , Basement Membrane/metabolism , Cisplatin/toxicity , Membrane Glycoproteins/metabolism , Metalloendopeptidases/metabolism , Acute Kidney Injury/metabolism , Animals , Antineoplastic Agents/toxicity , Gene Expression Regulation/drug effects , Genotype , Hydroxamic Acids , Male , Metalloendopeptidases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Transport
11.
J Biol Chem ; 289(19): 13308-22, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24662289

ABSTRACT

Meprin A, composed of α and ß subunits, is a membrane-bound metalloproteinase in renal proximal tubules. Meprin A plays an important role in tubular epithelial cell injury during acute kidney injury (AKI). The present study demonstrated that during ischemia-reperfusion-induced AKI, meprin A was shed from proximal tubule membranes, as evident from its redistribution toward the basolateral side, proteolytic processing in the membranes, and excretion in the urine. To identify the proteolytic enzyme responsible for shedding of meprin A, we generated stable HEK cell lines expressing meprin ß alone and both meprin α and meprin ß for the expression of meprin A. Phorbol 12-myristate 13-acetate and ionomycin stimulated ectodomain shedding of meprin ß and meprin A. Among the inhibitors of various proteases, the broad spectrum inhibitor of the ADAM family of proteases, tumor necrosis factor-α protease inhibitor (TAPI-1), was most effective in preventing constitutive, phorbol 12-myristate 13-acetate-, and ionomycin-stimulated shedding of meprin ß and meprin A in the medium of both transfectants. The use of differential inhibitors for ADAM10 and ADAM17 indicated that ADAM10 inhibition is sufficient to block shedding. In agreement with these results, small interfering RNA to ADAM10 but not to ADAM9 or ADAM17 inhibited meprin ß and meprin A shedding. Furthermore, overexpression of ADAM10 resulted in enhanced shedding of meprin ß from both transfectants. Our studies demonstrate that ADAM10 is the major ADAM metalloproteinase responsible for the constitutive and stimulated shedding of meprin ß and meprin A. These studies further suggest that inhibiting ADAM 10 activity could be of therapeutic benefit in AKI.


Subject(s)
ADAM Proteins/metabolism , Acute Kidney Injury/enzymology , Amyloid Precursor Protein Secretases/metabolism , Cell Membrane/enzymology , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , ADAM Proteins/genetics , ADAM10 Protein , ADAM17 Protein , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Amyloid Precursor Protein Secretases/genetics , Animals , Calcium Ionophores/pharmacology , Carcinogens/pharmacology , Cell Membrane/genetics , Cell Membrane/pathology , HEK293 Cells , Humans , Ionomycin/pharmacology , Male , Membrane Proteins/genetics , Metalloendopeptidases/genetics , Mice , Tetradecanoylphorbol Acetate/pharmacology
12.
Methods Mol Biol ; 1133: 141-54, 2014.
Article in English | MEDLINE | ID: mdl-24567100

ABSTRACT

Members of the caspase family of proteases are evolutionarily conserved cysteine proteases that play a crucial role as the central executioners of the apoptotic pathway. Since the discovery of caspases, many methods have been developed to detect their activation and are widely used in basic and clinical studies. In a mouse tissue, caspase activation can be monitored by cleavage of caspase-specific synthetic substrates and by detecting cleaved caspase by western blot analysis of the tissue extract. In tissue sections, active caspase can be detected by immunostaining using specific antibodies to the active caspase. In addition, among the myriads of caspase-specific substrates known so far, cleaved fragments produced by caspases from the substrates such as PARP, lamin A, and cytokeratin-18 can be monitored in tissue sections by immunostaining as well as western blots of tissue extracts. In general, more than one method should be used to ascertain detection of activation of caspases in a mouse tissue.


Subject(s)
Apoptosis/genetics , Caspases/isolation & purification , Molecular Biology/methods , Animals , Caspases/genetics , Mice
13.
J Am Soc Nephrol ; 25(5): 877-83, 2014 May.
Article in English | MEDLINE | ID: mdl-24480828

ABSTRACT

Treating or preventing AKI requires treating or preventing a rise in serum creatinine as well as the immediate and remote clinical consequences associated with AKI. Because a substantial number of patients with AKI progress to ESRD, identifying patients likely to progress and halting progression are important goals for treating AKI. Many therapies for AKI are being developed, including RenalGuard Therapy, which aims to maintain high urine output; α-melanocyte-stimulating hormone, with anti-inflammatory and antiapoptotic activities; alkaline phosphatase, which detoxifies proinflammatory substances; novel, small interfering RNA, directed at p53 activation; THR-184, a peptide agonist of bone morphogenetic proteins; removal of catalytic iron, important in free-radical formation; and cell-based therapies, including mesenchymal stem cells in vivo and renal cell therapy in situ. In this review, we explore what treatment of AKI really means, discuss the emerging therapies, and examine the windows of opportunity for treating AKI. Finally, we provide suggestions for accelerating the pathways toward preventing and treating AKI, such as establishing an AKI network, implementing models of catalytic philanthropy, and directing a small percentage of the Medicare ESRD budget for developing therapies to prevent and treat AKI and halt progression of CKD.


Subject(s)
Acute Kidney Injury/therapy , Nephrology/trends , Humans
15.
Am J Physiol Renal Physiol ; 304(9): F1150-8, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23427141

ABSTRACT

Meprin A, composed of α- and ß-subunits, is a membrane-associated neutral metalloendoprotease that belongs to the astacin family of zinc endopeptidases. It was first discovered as an azocasein and benzoyl-l-tyrosyl-p-aminobenzoic acid hydrolase in the brush-border membranes of proximal tubules and intestines. Meprin isoforms are now found to be widely distributed in various organs (kidney, intestines, leukocytes, skin, bladder, and a variety of cancer cells) and are capable of hydrolyzing and processing a large number of substrates, including extracellular matrix proteins, cytokines, adherens junction proteins, hormones, bioactive peptides, and cell surface proteins. The ability of meprin A to cleave various substrates sheds new light on the functional properties of this enzyme, including matrix remodeling, inflammation, and cell-cell and cell-matrix processes. Following ischemia-reperfusion (IR)- and cisplatin-induced acute kidney injury (AKI), meprin A is redistributed toward the basolateral plasma membrane, and the cleaved form of meprin A is excreted in the urine. These studies suggest that altered localization and shedding of meprin A in places other than the apical membranes may be deleterious in vivo in acute tubular injury. These studies also provide new insight into the importance of a sheddase involved in the release of membrane-associated meprin A under pathological conditions. Meprin A is injurious to the kidney during AKI, as meprin A-knockout mice and meprin inhibition provide protective roles and improve renal function. Meprin A, therefore, plays an important role in AKI and potentially is a unique target for therapeutic intervention during AKI.


Subject(s)
Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Metalloendopeptidases/physiology , Animals , Cell Membrane/pathology , Cell Membrane/physiology , Disease Models, Animal , Epithelial Cells/pathology , Epithelial Cells/physiology , Humans , Leukocytes/pathology , Leukocytes/physiology , Metalloendopeptidases/genetics , Mice , Mice, Knockout , Rats
16.
Kidney Int ; 82(12): 1250-3, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23203020

ABSTRACT

Autophagy is upregulated during ischemia-reperfusion (IR)-induced and cisplatin-induced acute kidney injury (AKI). Proximal tubule-specific Atg7 knockout mice exhibited increased renal injury compared with wild-type mice following cisplatin- and IR-induced AKI. Inhibition of autophagy by chloroquine aggravated AKI, whereas upregulation of autophagy by rapamycin recovered lost renal function and histology, further indicating a protective role of autophagy in AKI. These findings reported by Jiang et al. will provide stimulus to further examine the role and mechanism of the enhancement of autophagy in AKI.


Subject(s)
Acute Kidney Injury/prevention & control , Autophagy , Kidney Tubules, Proximal/pathology , Reperfusion Injury/prevention & control , Animals
18.
Am J Physiol Renal Physiol ; 303(8): F1239-50, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22896037

ABSTRACT

Cisplatin injury to renal tubular epithelial cells (RTEC) is accompanied by autophagy and caspase activation. However, autophagy gradually decreases during the course of cisplatin injury. The role of autophagy and the mechanism of its decrease during cisplatin injury are not well understood. This study demonstrated that autophagy proteins beclin-1, Atg5, and Atg12 were cleaved and degraded during the course of cisplatin injury in RTEC and the kidney. zVAD-fmk, a widely used pancaspase inhibitor, blocked cleavage of autophagy proteins suggesting that zVAD-fmk would promote the autophagy pathway. Unexpectedly, zVAD-fmk blocked clearance of the autophagosomal cargo, indicating lysosomal dysfunction. zVAD-fmk markedly inhibited cisplatin-induced lysosomal cathepsin B and calpain activities and therefore impaired autophagic flux. In a mouse model of cisplatin nephrotoxicity, zVAD-fmk impaired autophagic flux by blocking autophagosomal clearance as revealed by accumulation of key autophagic substrates p62 and LC3-II. Furthermore, zVAD-fmk worsened cisplatin-induced renal dysfunction. Chloroquine, a lysomotropic agent that is known to impair autophagic flux, also exacerbated cisplatin-induced decline in renal function. These findings demonstrate that impaired autophagic flux induced by zVAD-fmk or a lysomotropic agent worsened renal function in cisplatin acute kidney injury (AKI) and support a protective role of autophagy in AKI. These studies also highlight that the widely used antiapoptotic agent zVAD-fmk may be contraindicated as a therapeutic agent for preserving renal function in AKI.


Subject(s)
Amino Acid Chloromethyl Ketones/pharmacology , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Caspase Inhibitors/pharmacology , Cisplatin/pharmacology , Kidney/drug effects , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 12 , Autophagy-Related Protein 5 , Beclin-1 , Calpain/metabolism , Cathepsin B/metabolism , Cells, Cultured , Kidney/metabolism , Lysosomes/drug effects , Microtubule-Associated Proteins/metabolism , Proteins/metabolism
19.
Biochim Biophys Acta ; 1813(8): 1525-31, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21616098

ABSTRACT

The gelatinases, matrix metalloproteinase (MMP)-9 and -2, are produced as latent, inactive enzymes that can be proteolytically activated by a number of proteases. In many normal and pathological conditions, where the expression of MMPs is deregulated, changes in the expression of other proteases have also been reported. Human kallikrein-related peptidase 7 (KLK7), a chymotryptic-like serine protease, is overexpressed in many different types of neoplastic conditions, which have also been shown to express high levels of both MMP-9 and -2. Since the activation of MMPs by KLK7 has never been examined, we sought to determine whether KLK7 can activate these MMPs. To test this hypothesis KLK7 was incubated with the recombinant MMPs and the products of the reaction were analyzed for their activity. Incubation of proMMP-9 with KLK7 resulted in the production of a novel truncated, active MMP-9 lacking the C-terminal hemopexin domains. In contrast, KLK7 degraded, but did not activate, proMMP-2. The novel activation of proMMP-9 by KLK7 was further confirmed using conditioned medium prepared from an MMP-9-expressing cell line, MDA-MMP-9. Our results clearly establish that KLK7 activates proMMP-9 to produce a novel truncated, active MMP-9 product not generated by other proteases. These findings suggest that KLK7 may play an important role in the activation of MMP-9 in tumors that express high levels of both these proteases and the resulting truncated MMP may possess altered substrate specificities compared with full-length MMP-9 activated by other proteases.


Subject(s)
Kallikreins/metabolism , Matrix Metalloproteinase 9/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Enzyme Activation , Enzyme Precursors/chemistry , Enzyme Precursors/metabolism , Female , Gelatinases/chemistry , Gelatinases/metabolism , Hemopexin/chemistry , Hemopexin/metabolism , Humans , Matrix Metalloproteinase 2/chemistry , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/chemistry , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Structure, Tertiary , Substrate Specificity
20.
Shock ; 35(2): 141-7, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20577148

ABSTRACT

Sepsis-induced acute kidney injury occurs in 20% to 50% of septic patients and nearly doubles the mortality rate of sepsis. Because treatment in the septic patient is usually begun only after the onset of symptoms, therapy that is effective even when delayed would have the greatest impact on patient survival. The metalloproteinase meprin A, an oligomeric complex made of α- and ß-subunits, is highly expressed at the brush-border membranes of the kidney and capable of degrading numerous substrates including extracellular matrix proteins and cytokines. The goal of the present study was to compare the therapeutic potential of actinonin, an inhibitor of meprin A, when administered before and after the onset of sepsis. Mice were treated with actinonin at 30 min before or 7 h after induction of sepsis by cecal ligation and puncture (CLP). Intravital videomicroscopy was used to image renal peritubular capillary perfusion and reactive nitrogen species. Actinonin treatment 30 min before CLP reduced IL-1ß levels and prevented the fall in renal capillary perfusion at 7 and 18 h. Actinonin also prevented the fall in renal capillary perfusion even when administered at 7 h after CLP. In addition, even late administration of actinonin preserved renal morphology and lowered blood urea nitrogen and serum creatinine concentrations. These data suggest that agents such as actinonin should be evaluated further as possible therapeutic agents because targeting both the early systemic and later organ-damaging effects of sepsis should have the highest likelihood of success.


Subject(s)
Anti-Bacterial Agents/pharmacology , Metalloendopeptidases/antagonists & inhibitors , Microcirculation/drug effects , Renal Circulation/drug effects , Sepsis/physiopathology , Animals , Hydroxamic Acids/pharmacology , Interleukin-1beta , Kidney/blood supply , Kidney/physiopathology , Metalloendopeptidases/metabolism , Mice , Sepsis/drug therapy , Sepsis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...