Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Cell ; 171(3): 628-641.e26, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-29053969

ABSTRACT

Ferroptosis is a form of programmed cell death that is pathogenic to several acute and chronic diseases and executed via oxygenation of polyunsaturated phosphatidylethanolamines (PE) by 15-lipoxygenases (15-LO) that normally use free polyunsaturated fatty acids as substrates. Mechanisms of the altered 15-LO substrate specificity are enigmatic. We sought a common ferroptosis regulator for 15LO. We discovered that PEBP1, a scaffold protein inhibitor of protein kinase cascades, complexes with two 15LO isoforms, 15LO1 and 15LO2, and changes their substrate competence to generate hydroperoxy-PE. Inadequate reduction of hydroperoxy-PE due to insufficiency or dysfunction of a selenoperoxidase, GPX4, leads to ferroptosis. We demonstrated the importance of PEBP1-dependent regulatory mechanisms of ferroptotic death in airway epithelial cells in asthma, kidney epithelial cells in renal failure, and cortical and hippocampal neurons in brain trauma. As master regulators of ferroptotic cell death with profound implications for human disease, PEBP1/15LO complexes represent a new target for drug discovery.


Subject(s)
Acute Kidney Injury/pathology , Asthma/pathology , Brain Injuries, Traumatic/pathology , Cell Death , Phosphatidylethanolamine Binding Protein/metabolism , Acute Kidney Injury/metabolism , Animals , Apoptosis , Asthma/metabolism , Brain Injuries, Traumatic/metabolism , Cell Death/drug effects , Cell Line , Humans , Isoenzymes/metabolism , Lipoxygenase/chemistry , Lipoxygenase/metabolism , Mice , Models, Molecular , Oxazolidinones/pharmacology , Oxidation-Reduction , Phosphatidylethanolamine Binding Protein/chemistry
2.
J Biol Chem ; 298(5): 101836, 2022 05.
Article in English | MEDLINE | ID: mdl-35307350

ABSTRACT

Phosphate homeostasis, mediated by dietary intake, renal absorption, and bone deposition, is incompletely understood because of the uncharacterized roles of numerous implicated protein factors. Here, we identified a novel role for one such element, regulator of G protein signaling 14 (RGS14), suggested by genome-wide association studies to associate with dysregulated Pi levels. We show that human RGS14 possesses a carboxy-terminal PDZ ligand required for sodium phosphate cotransporter 2a (NPT2A) and sodium hydrogen exchanger regulatory factor-1 (NHERF1)-mediated renal Pi transport. In addition, we found using isotope uptake measurements combined with bioluminescence resonance energy transfer assays, siRNA knockdown, pull-down and overlay assays, and molecular modeling that secreted proteins parathyroid hormone (PTH) and fibroblast growth factor 23 inhibited Pi uptake by inducing dissociation of the NPT2A-NHERF1 complex. PTH failed to affect Pi transport in cells expressing RGS14, suggesting that it suppresses hormone-sensitive but not basal Pi uptake. Interestingly, RGS14 did not affect PTH-directed G protein activation or cAMP formation, implying a postreceptor site of action. Further pull-down experiments and direct binding assays indicated that NPT2A and RGS14 bind distinct PDZ domains on NHERF1. We showed that RGS14 expression in human renal proximal tubule epithelial cells blocked the effects of PTH and fibroblast growth factor 23 and stabilized the NPT2A-NHERF1 complex. In contrast, RGS14 genetic variants bearing mutations in the PDZ ligand disrupted RGS14 binding to NHERF1 and subsequent PTH-sensitive Pi transport. In conclusion, these findings identify RGS14 as a novel regulator of hormone-sensitive Pi transport. The results suggest that changes in RGS14 function or abundance may contribute to the hormone resistance and hyperphosphatemia observed in kidney diseases.


Subject(s)
Phosphoproteins/metabolism , RGS Proteins , Sodium-Hydrogen Exchangers/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , GTP-Binding Proteins/metabolism , Genome-Wide Association Study , Humans , Ligands , Parathyroid Hormone/metabolism , Phosphates/metabolism , RGS Proteins/genetics , RGS Proteins/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism
3.
J Transl Med ; 20(1): 204, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35538495

ABSTRACT

BACKGROUND: Post-cardiac surgery acute kidney injury (AKI) is associated with increased mortality. A high-protein meal enhances the renal blood flow and glomerular filtration rate (GFR) and might protect the kidneys from acute ischemic insults. Hence, we assessed the effect of a preoperative high-oral protein load on post-cardiac surgery renal function and used experimental models to elucidate mechanisms by which protein might stimulate kidney-protective effects. METHODS: The prospective "Preoperative Renal Functional Reserve Predicts Risk of AKI after Cardiac Operation" study follow-up was extended to postoperative 12 months for 109 patients. A 1:2 ratio propensity score matching method was used to identify a control group (n = 214) to comparatively evaluate the effects of a preoperative protein load and standard care. The primary endpoints were AKI development and postoperative estimated GFR (eGFR) loss at 3 and 12 months. We also assessed the secretion of tissue inhibitor of metalloproteases-2 (TIMP-2) and insulin-like growth factor-binding protein 7 (IGFBP7), biomarkers implicated in mediating kidney-protective mechanisms in human kidney tubular cells that we exposed to varying protein concentrations. RESULTS: The AKI rate did not differ between the protein loading and control groups (13.6 vs. 12.3%; p = 0.5). However, the mean eGFR loss was lower in the former after 3 months (0.1 [95% CI - 1.4, - 1.7] vs. - 3.3 [95% CI - 4.4, - 2.2] ml/min/1.73 m2) and 12 months (- 2.7 [95% CI - 4.2, - 1.2] vs - 10.2 [95% CI - 11.3, - 9.1] ml/min/1.73 m2; p < 0.001 for both). On stratification based on AKI development, the eGFR loss after 12 months was also found to be lower in the former (- 8.0 [95% CI - 14.1, - 1.9] vs. - 18.6 [95% CI - 23.3, - 14.0] ml/min/1.73 m2; p = 0.008). A dose-response analysis of the protein treatment of the primary human proximal and distal tubule epithelial cells in culture showed significantly increased IGFBP7 and TIMP-2 expression. CONCLUSIONS: A preoperative high-oral protein load did not reduce AKI development but was associated with greater renal function preservation in patients with and without AKI at 12 months post-cardiac surgery. The potential mechanisms of action by which protein loading may induce a kidney-protective response might include cell cycle inhibition of renal tubular epithelial cells. Clinical trial registration ClinicalTrials.gov: NCT03102541 (retrospectively registered on April 5, 2017) and ClinicalTrials.gov: NCT03092947 (retrospectively registered on March 28, 2017).


Subject(s)
Acute Kidney Injury , Cardiac Surgical Procedures , Acute Kidney Injury/etiology , Biomarkers , Cardiac Surgical Procedures/adverse effects , Cohort Studies , Female , Glomerular Filtration Rate , Humans , Kidney/physiology , Male , Postoperative Complications , Prospective Studies , Tissue Inhibitor of Metalloproteinase-2
4.
Am J Physiol Renal Physiol ; 321(2): F135-F148, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34151589

ABSTRACT

Cell-associated kidney injury molecule-1 (KIM-1) exerts an anti-inflammatory role following kidney injury by mediating efferocytosis and downregulating the NF-κB pathway. KIM-1 cleavage blunts its anti-inflammatory activities. We reported that mucin 1 (MUC1) is protective in a mouse model of ischemia-reperfusion injury (IRI). As both KIM-1 and MUC1 are induced in the proximal tubule (PT) during IRI and are a disintegrin and metalloprotease 17 (ADAM17) substrates, we tested the hypothesis that MUC1 protects KIM-1 activity. Muc1 knockout (KO) mice and wild-type (WT) littermates were subjected to IRI. KIM-1, MUC1, and ADAM17 levels (and signaling pathways) were assessed by immunoblot analysis. PT localization was assessed by confocal microscopy and an in situ proximity ligation assay. Findings were extended using human kidneys and urine as well as KIM-1-mediated efferocytosis assays in mouse PT cultures. In response to tubular injury in mouse and human kidneys, we observed induction and coexpression of KIM-1 and MUC1 in the PT. Compared with WT mice, Muc1 KO mice had higher urinary KIM-1 and lower kidney KIM-1. KIM-1 was apical in the PT of WT kidneys but predominately with luminal debris in Muc1 KO mice. Efferocytosis was reduced in Muc1 KO PT cultures compared with WT cultures, whereas inflammation was increased in Muc1 KO kidneys compared with WT kidneys. MUC1 was cleaved by ADAM17 in PT cultures and blocked KIM-1 shedding in Madin-Darby canine kidney cells. We conclude that KIM-1-mediated efferocytosis and thus anti-inflammatory activity during IRI is preserved in the injured kidney by MUC1 inhibition of KIM-1 shedding.NEW & NOTEWORTHY KIM-1 plays a key role in the recovery of the tubule epithelium during renal IRI by mediating efferocytosis and associated signaling that suppresses inflammation. Excessive cleavage of KIM-1 by ADAM17 provides a decoy receptor that aggravates efferocytosis and subsequent signaling. Our data from experiments in mice, patients, and cultured cells show that MUC1 is also induced during IRI and competes with KIM-1 for cleavage by ADAM17. Consequently, MUC1 protects KIM-1 anti-inflammatory activity in the damaged kidney.


Subject(s)
Hepatitis A Virus Cellular Receptor 1/metabolism , Inflammation/metabolism , Kidney Tubules, Proximal/metabolism , Kidney/blood supply , Mucin-1/metabolism , Reperfusion Injury/metabolism , ADAM17 Protein/metabolism , Animals , Cell Line , Dogs , Humans , Kidney/metabolism , Mice, Knockout , Mice, Transgenic , Mucin-1/genetics , Phagocytosis/physiology
5.
FASEB J ; 34(5): 7036-7057, 2020 05.
Article in English | MEDLINE | ID: mdl-32246808

ABSTRACT

The purpose was to determine the role of AMPK activation in the renal metabolic response to sepsis, the development of sepsis-induced acute kidney injury (AKI) and on survival. In a prospective experimental study, 167 10- to 12-week-old C57BL/6 mice underwent cecal ligation and puncture (CLP) and human proximal tubule epithelial cells (TEC; HK2) were exposed to inflammatory mix (IM), a combination of lipopolysaccharide (LPS) and high mobility group box 1 (HMGB1). Renal/TEC metabolic fitness was assessed by monitoring the expression of drivers of oxidative phosphorylation (OXPHOS), the rates of utilization of OXPHOS/glycolysis in response to metabolic stress, and mitochondrial function by measuring O2 consumption rates (OCR) and the membrane potential (Δψm ). Sepsis/IM resulted in AKI, increased mortality, and in renal AMPK activation 6-24 hours after CLP/IM. Pharmacologic activation of AMPK with 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) or metformin during sepsis improved the survival, while AMPK inhibition with Compound C increased mortality, impaired mitochondrial respiration, decreased OCR, and disrupted TEC metabolic fitness. AMPK-driven protection was associated with increased Sirt 3 expression and restoration of metabolic fitness. Renal AMPK activation in response to sepsis/IM is an adaptive mechanism that protects TEC, organs, and the host by preserving mitochondrial function and metabolic fitness likely through Sirt3 signaling.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Inflammation/metabolism , Kidney/metabolism , Sepsis/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Acute Kidney Injury/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Enzyme Activation , Epithelial Cells/metabolism , Humans , Kidney Tubules, Proximal/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidative Phosphorylation , Oxygen Consumption
6.
Am J Physiol Renal Physiol ; 312(2): F284-F296, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-28003188

ABSTRACT

We have characterized the expression and secretion of the acute kidney injury (AKI) biomarkers insulin-like growth factor binding protein 7 (IGFBP7) and tissue inhibitor of metalloproteinases-2 (TIMP-2) in human kidney epithelial cells in primary cell culture and tissue. We established cell culture model systems of primary kidney cells of proximal and distal tubule origin and observed that both proteins are indeed expressed and secreted in both tubule cell types in vitro. However, TIMP-2 is both expressed and secreted preferentially by cells of distal tubule origin, while IGFBP7 is equally expressed across tubule cell types yet preferentially secreted by cells of proximal tubule origin. In human kidney tissue, strong staining of IGFBP7 was seen in the luminal brush-border region of a subset of proximal tubule cells, and TIMP-2 stained intracellularly in distal tubules. Additionally, while some tubular colocalization of both biomarkers was identified with the injury markers kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin, both biomarkers could also be seen alone, suggesting the possibility for differential mechanistic and/or temporal profiles of regulation of these early AKI biomarkers from known markers of injury. Last, an in vitro model of ischemia-reperfusion demonstrated enhancement of secretion of both markers early after reperfusion. This work provides a rationale for further investigation of these markers for their potential role in the pathogenesis of acute kidney injury.


Subject(s)
Insulin-Like Growth Factor Binding Proteins/metabolism , Kidney Tubules, Distal/metabolism , Kidney Tubules, Proximal/metabolism , Reperfusion Injury/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Acute Kidney Injury/metabolism , Biomarkers , Hepatitis A Virus Cellular Receptor 1/metabolism , Humans , Lipocalin-2/metabolism , Organ Specificity
7.
JCI Insight ; 5(1)2020 01 16.
Article in English | MEDLINE | ID: mdl-31941842

ABSTRACT

Lithium (Li) is the mainstay pharmacotherapeutic mood stabilizer in bipolar disorder. Its efficacious use is complicated by acute and chronic renal side effects, including nephrogenic diabetes insipidus (NDI) and progression to chronic kidney disease (CKD). The nuclear factor erythroid-derived 2-related factor 2 (Nrf2) pathway senses and coordinates cellular responses to oxidative and electrophilic stress. Here, we identify that graded genetic activation of Nrf2 protects against Li-induced NDI (Li-NDI) and volume wasting via an aquaporin 2-independent mechanism. Renal Nrf2 activity is differentially expressed on functional segments of the nephron, and its activation along the distal tubule and collecting duct directly modulates ion transporter expression, mimicking paradoxical effects of diuretics in mitigating Li-NDI. In addition, Nrf2 reduces cyclooxygenase expression and vasoactive prostaglandin biosynthesis. Pharmacologic activation of Nrf2 confers protective effects, confirming this pathway as a potentially novel druggable target for the prevention of acute and chronic renal sequelae of Li therapy.


Subject(s)
Diabetes Insipidus, Nephrogenic/drug therapy , Lithium/adverse effects , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/pharmacology , Animals , Aquaporin 2/metabolism , Bipolar Disorder , Cyclooxygenase 1/metabolism , Diabetes Insipidus, Nephrogenic/chemically induced , Epithelial Cells , Humans , Kidney/metabolism , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , Prostaglandin-Endoperoxide Synthases/metabolism
8.
Mol Cancer Ther ; 6(10): 2664-74, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17938260

ABSTRACT

Human epidermal growth factor receptor-2 (HER2) and epidermal growth factor receptor (EGFR) heterodimerize to activate mitogenic signaling pathways. We have shown previously, using MCF7 subcloned cell lines with graded levels of HER2 expression, that responsiveness to trastuzumab and AG1478 (an anti-EGFR agent), varied directly with levels of HER2 expression. HER2 and EGFR up-regulate vascular endothelial growth factor (VEGF), a growth factor that promotes angiogenesis and participates in autocrine growth-stimulatory pathways that might be active in vitro. Here, we show that trastuzumab, erlotinib, and bevacizumab, individually and in combination, inhibit cell proliferation in a panel of unrelated human breast cancer cell lines, in proportion to their levels of HER2 expression. The combination of all three drugs provided a greater suppression of growth than any single drug or two-drug combination in the high HER2-expressing cell lines (P < 0.001). Combination index analysis suggested that the effects of these drugs in combination were additive. The pretreatment net level of VEGF production in each cell line was correlated with the level of HER2 expression (r = 0.883, P = 0.016). Trastuzumab and erlotinib each reduced total net VEGF production in all cell lines. Multiparameter flow cytometry studies indicated that erlotinib alone and the triple drug combination produced a prolonged but reversible blockade of cells in G1, but did not increase apoptosis substantially. These studies suggest that the effects of two and three-drug combinations of trastuzumab, erlotinib, and bevacizumab might offer potential therapeutic advantages in HER2-overexpressing breast cancers, although these effects are of low magnitude, and are likely to be transient.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , ErbB Receptors/metabolism , Receptor, ErbB-2/metabolism , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Bevacizumab , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Enzyme-Linked Immunosorbent Assay , Erlotinib Hydrochloride , Flow Cytometry , G1 Phase/drug effects , Humans , Immunoblotting , Quinazolines/administration & dosage , Trastuzumab , Tumor Cells, Cultured/drug effects , Vascular Endothelial Growth Factor A/metabolism
9.
Cytometry B Clin Cytom ; 70(1): 10-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16342079

ABSTRACT

BACKGROUND: Laser scanning Cytometry (LSC) is a versatile technology that makes it possible to perform multiple measurements on individual cells and correlate them cell by cell with other cellular features. It would be highly desirable to be able to perform reproducible, quantitative, correlated cell-based immunofluorescence studies on individual cells from human solid tumors. However, such studies can be challenging because of the presence of large numbers of cell aggregates and other confounding factors. Techniques have been developed to deal with cell aggregates in data sets collected by LSC. Experience has also been gained in addressing other key technical and methodological issues that can affect the reproducibility of such cell-based immunofluorescence measurements. METHODS AND RESULTS: We describe practical aspects of cell sample collection, cell fixation and staining, protocols for performing multiparameter immunofluorescence measurements by LSC, use of controls and reference samples, and approaches to data analysis that we have found useful in improving the accuracy and reproducibility of LSC data obtained in human tumor samples. We provide examples of the potential advantages of LSC in examining quantitative aspects of cell-based analysis. Improvements in the quality of cell-based multiparameter immunofluorescence measurements make it possible to extract useful information from relatively small numbers of cells. This, in turn, permits the performance of multiple multicolor panels on each tumor sample. With links among the different panels that are provided by overlapping measurements, it is possible to develop increasingly more extensive profiles of intracellular expression of multiple proteins in clinical samples of human solid tumors. Examples of such linked panels of measurements are provided. CONCLUSIONS: Advances in methodology can improve cell-based multiparameter immunofluorescence measurements on cell suspensions from human solid tumors by LSC for use in prognostic and predictive clinical applications.


Subject(s)
Fluorescent Antibody Technique/methods , Guidelines as Topic , Laser Scanning Cytometry/methods , Neoplasms/metabolism , Neoplasms/pathology , Tissue Fixation , Cell Aggregation , Cell Count , Cell Membrane Permeability , Cell Size , DNA/metabolism , Humans , Reference Standards , Reproducibility of Results , Research Design
10.
Cancer Res ; 63(1): 250-5, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12517805

ABSTRACT

The c-Jun NH(2)-terminal kinases (JNKs) have a role both in promoting apoptosis and tumorigenesis. The JNKs are encoded by three separate genes (JNK1, 2, and 3), which are spliced alternatively to create 10 JNK isoforms that are either M(r) 55,000 or 46,000 in size. However, the functional significance and distinct role for each splice variant remains unclear. We have noted previously that 86% of primary human glial tumors show activation of almost exclusively the M(r) 55,000 isoforms of JNK. To further study which isoforms are involved, we constructed glutathione S-transferase fusion proteins for all 10 JNK isoforms and examined kinase activity with or without the activating upstream kinase. Surprisingly, five JNK isoforms demonstrate autophosphorylation activity, and in addition, all four JNK2 isoforms (either M(r) 55,000 or 46,000) show a high basal level of substrate kinase activity in the absence of the upstream kinase, especially a M(r) 55,000 JNK2 isoform. Examination revealed autophosphorylation activity at the T-P-Y motif, which is critical for JNK activation, because a mutant lacking the dual phosphorylation sites did not show autophosphorylation or basal kinase activity. Using green fluorescence protein-JNK expression vectors, transient transfection into U87MG cells demonstrates that although the JNK1 isoforms localize predominantly to the cytoplasm, the JNK2 isoforms localize to the nucleus and are phosphorylated, confirming the constitutive activation seen in vitro. We then examined which JNK isoforms are active in glial tumors by performing two-dimensional electrophoresis. This revealed that the M(r) 55,000 isoforms of JNK2 are the principal active JNK isoforms present in tumors. Collectively, these results suggest that these constitutively active JNK isoforms play a significant role in glial tumors. Aside from epidermal growth factor receptor vIII, this is the only other kinase that has been shown to be basally active in glioma. The presence of constitutively active JNK isoforms may have implications for the design of inhibitors of the JNK pathway.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Brain/enzymology , Brain Neoplasms/enzymology , Enzyme Activation , Glioblastoma , Humans , Isoenzymes/metabolism , JNK Mitogen-Activated Protein Kinases , Phosphorylation , Recombinant Proteins/metabolism , Transfection , Tumor Cells, Cultured
11.
Oncogene ; 21(33): 5038-46, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12140754

ABSTRACT

The ERK pathway is typically associated with activation of the EGF receptor and has been shown to play a major role in promoting several tumor phenotypes. An analogous signaling module, the JNK pathway, has not been shown to be consistently activated by the EGF receptor but is instead more uniformly stimulated by cellular stresses and cytokines. The function of the JNK pathway in primary tumors is unclear as it has been implicated in both promoting apoptosis and cell growth in vitro, which may be a reflection of the cell lines chosen. Primary human brain tumors frequently show overexpression of the EGF receptor. To clarify the role of JNK in tumorigenesis, we have investigated the role of JNK in a large panel of primary human brain tumors and tumor derived cell lines. Here we present evidence that JNK has a major role in promoting tumorigenesis both in vivo and in vitro. Western blot analysis demonstrated that 86% (18 of 21) of primary brain tumors showed evidence of JNK activation but only 38% (8 of 21) showed evidence of ERK activation. Kinase assays revealed that 77% of brain tumor cell lines activated JNK in response to EGF (7 of 13) or had high levels of basal activity (3 of 13), whereas none of six normal cell lines analysed, including astrocytes, had these properties. Of several growth factors examined, EGF produced the highest level of JNK induction in tumor cell lines and the duration of activation was greater than that seen for ERK. Expression of a dominant-negative (dn) form of JNK potently inhibited EGF mediated anchorage independent growth and protection from cell death in two glial tumor cell lines. These findings demonstrate that enhanced JNK activation is frequently found in primary brain tumors and that this activation contributes to phenotypes related to transformation.


Subject(s)
Brain Neoplasms/enzymology , Mitogen-Activated Protein Kinases/metabolism , Apoptosis , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Division , Cell Survival , Cell Transformation, Neoplastic , Contact Inhibition , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Humans , JNK Mitogen-Activated Protein Kinases , Time Factors , Tumor Cells, Cultured
12.
Semin Nephrol ; 35(1): 85-95, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25795502

ABSTRACT

Acute kidney injury (AKI) occurs frequently in critically ill patients with sepsis, in whom it doubles the mortality rate and half of the survivors suffer permanent kidney damage or chronic kidney disease. Failure in the development of viable therapies has prompted studies to better elucidate the cellular and molecular etiologies of AKI, which have generated novel theories and paradigms for the mechanisms of this disease. These studies have shown multifaceted origins and elements of AKI that, in addition to/in lieu of ischemia, include the generation of damage-associated molecular patterns and pathogen-associated molecular patterns, the inflammatory response, humoral and cellular immune activation, perturbation of microvascular flow and oxidative stress, bioenergetic alterations, cell-cycle alterations, and cellular de-differentiation/re-differentiation. It is becoming clear that a major etiologic effector of all these inputs is the renal tubule epithelial cell (RTEC). This review discusses these elements and their effects on RTECs, and reviews the current hypotheses of how these effects may determine the fate of RTECs during sepsis-induced AKI.


Subject(s)
Acute Kidney Injury/physiopathology , Epithelial Cells/metabolism , Kidney/metabolism , Sepsis/physiopathology , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Epithelial Cells/physiology , Humans , Inflammation , Kidney/physiopathology , Kidney Tubules, Distal/cytology , Kidney Tubules, Proximal/cytology , Oxidative Stress , Sepsis/complications , Sepsis/metabolism
13.
Cancer Res ; 74(4): 1238-49, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24366881

ABSTRACT

The relationship between mutated proteins and the cancer stem-cell population is unclear. Glioblastoma tumors frequently express EGFRvIII, an EGF receptor (EGFR) variant that arises via gene rearrangement and amplification. However, expression of EGFRvIII is restricted despite the prevalence of the alteration. Here, we show that EGFRvIII is highly coexpressed with CD133 and that EGFRvIII(+)/CD133(+) defines the population of cancer stem cells (CSC) with the highest degree of self-renewal and tumor-initiating ability. EGFRvIII(+) cells are associated with other stem/progenitor markers, whereas markers of differentiation are found in EGFRvIII(-) cells. EGFRvIII expression is lost in standard cell culture, but its expression is maintained in tumor sphere culture, and cultured cells also retain the EGFRvIII(+)/CD133(+) coexpression, self-renewal, and tumor initiating abilities. Elimination of the EGFRvIII(+)/CD133(+) population using a bispecific antibody reduced tumorigenicity of implanted tumor cells better than any reagent directed against a single epitope. This work demonstrates that a mutated oncogene can have CSC-specific expression and be used to specifically target this population.


Subject(s)
ErbB Receptors/metabolism , Glioblastoma/therapy , Molecular Targeted Therapy/methods , Neoplastic Stem Cells/metabolism , AC133 Antigen , Animals , Antibodies, Bispecific/therapeutic use , Antigens, CD/immunology , Antineoplastic Agents , Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Separation , ErbB Receptors/immunology , Glioblastoma/metabolism , Glioblastoma/pathology , Glycoproteins/immunology , Humans , Immunoconjugates/therapeutic use , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/pathology , Peptides/immunology , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Tumor Cells, Cultured
14.
Cell Stress Chaperones ; 16(2): 181-93, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20927616

ABSTRACT

Efrapeptins (EF), a family of fungal peptides, inhibit proteasomal enzymatic activities and the in vitro and in vivo growth of HT-29 cells. They are also known inhibitors of F(1)F(0)-ATPase, a mitochondrial enzyme that functions as an Hsp90 co-chaperone. We have previously shown that treatment of cancer cells with EF results in disruption of the Hsp90:F(1)F(0)-ATPase complex and inhibition of Hsp90 chaperone activity. The present study examines the effect of EF on breast cancer growth in vitro and in vivo. As a monotherapy, EF inhibited cell proliferation in vitro with an IC(50) value ranging from 6 nM to 3.4 µM. Inhibition of Hsp90 chaperone function appeared to be the dominant mechanism of action and the factor determining cellular sensitivity to EF. In vitro inhibition of proteasome became prominent in the absence of adequate levels of Hsp90 and F(1)F(0)-ATPase as in the case of the relatively EF-resistant MDA-MB-231 cell line. In vivo, EF inhibited MCF-7 and MDA-MB-231 xenograft growth with a maximal inhibition of 60% after administration of 0.15 and 0.3 mg/kg EF, respectively. 2-Deoxyglucose (2DG), a known inhibitor of glycolysis, acted synergistically with EF in vitro and antagonistically in vivo. In vitro, the synergistic effect was attributed to a prolonged endoplasmic reticulum (ER) stress. In vivo, the antagonistic effect was ascribed to the downregulation of tumoral and/or stromal F(1)F(0)-ATPase by 2DG.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Deoxyglucose/pharmacology , Peptides/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Down-Regulation/drug effects , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Inhibitory Concentration 50
15.
Cancer Sci ; 94(1): 50-6, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12708474

ABSTRACT

The epidermal growth factor receptor (EGFR) variant type III (variously called EGFRvIII, de2-7 EGFR or deltaEGFR) has an in-frame deletion of the extracellular domain and is found in numerous types of human tumors. Since EGFRvIII has been reported to be tumor-specific and has oncogenic potential, it is being investigated as a potential therapeutic target. Because the cell-specific expression of EGFRvIII in lung has not been well documented, we examined the expression of EGFRvIII in 76 non-small cell lung cancers (NSCLCs) and 10 non-neoplastic lung tissues by immunohistochemistry using a new monoclonal antibody specific for this variant receptor. We found a higher incidence (30 of 76, 39%) of enhanced EGFRvIII expression in NSCLC than previously described. Interestingly, the presence of EGFRvIII was also observed in several normal tissue components of lung (e.g., normal bronchial epithelium). Given the high prevalence of EGFRvIII in NSCLC, a newly developed phospho-specific (activated) EGFR antibody was employed for immunohistochemical analysis that permitted visualization of activated EGFR and/or EGFRvIII in tumors. This study presents evidence, for the first time, that EGFRvIII expressed in human tumors is phosphorylated and hence activated. Our results suggest that the sustained activation of EGFRvIII is implicated in the pathogenesis of NSCLC and thus EGFRvIII is a potential therapeutic target in this challenging disease.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/metabolism , Lung Neoplasms/metabolism , Neoplasm Proteins/metabolism , 3T3 Cells , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Alternative Splicing , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Carcinoma, Large Cell/genetics , Carcinoma, Large Cell/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Differentiation , Enzyme Activation , ErbB Receptors/genetics , ErbB Receptors/immunology , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Lung/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Molecular Sequence Data , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Phosphorylation , Protein Processing, Post-Translational , Rabbits , Rats
16.
Am J Pathol ; 160(2): 441-7, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11839564

ABSTRACT

Cell surface adhesion molecules are crucial for the development and/or pathogenesis of various diseases including cancer. CD44 has received much interest as a major adhesion molecule that is involved in tumor progression. We have previously demonstrated that the ectodomain of CD44 undergoes proteolytic cleavage by membrane-associated metalloproteases in various tumor cell lines. The remaining membrane-bound CD44 cleavage product can be detected using antibodies against the cytoplasmic domain of CD44 (anti-CD44cyto antibody). However, the cleavage of CD44 in primary human tumors has not been investigated. Using Western blots with anti-CD44cyto antibody to assay human tumor tissues, we show that the CD44 cleavage product can be detected in 58% (42 of 72) of gliomas but not in normal brain. Enhanced CD44 cleavage was also found in 67% (28 of 42) of breast carcinomas, 45% (5 of 11) of non-small cell lung carcinomas, 90% (9 of 10) of colon carcinomas, and 25% (3 of 12) of ovarian carcinomas. Tumors expressing a CD44 splice variant showed a significantly higher incidence of enhanced CD44 cleavage. The wide prevalence of CD44 cleavage suggests that it plays an important role in the pathogenesis of human tumors.


Subject(s)
Brain Neoplasms/physiopathology , Glioma/physiopathology , Hyaluronan Receptors/metabolism , Neoplasms/physiopathology , Brain/metabolism , Brain Neoplasms/pathology , Breast Neoplasms/physiopathology , Colonic Neoplasms/physiopathology , Female , Glioma/pathology , Humans , Lung Neoplasms/physiopathology , Neoplasms/pathology , Ovarian Neoplasms/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL