Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 22(11): 1803-11, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25857265

ABSTRACT

The receptor-interacting protein-associated ICH-1/CED-3 homologous protein with a death domain (RAIDD/CRADD) functions as a dual adaptor and is a constituent of different multi-protein complexes implicated in the regulation of inflammation and cell death. Within the PIDDosome complex, RAIDD connects the cell death-related protease, Caspase-2, with the p53-induced protein with a death domain 1 (PIDD1). As such, RAIDD has been implicated in DNA-damage-induced apoptosis as well as in tumorigenesis. As loss of Caspase-2 leads to an acceleration of tumor onset in the Eµ-Myc mouse lymphoma model, whereas loss of Pidd1 actually delays onset of this disease, we set out to interrogate the role of Raidd in cancer in more detail. Our data obtained analyzing Eµ-Myc/Raidd(-/-) mice indicate that Raidd is unable to protect from c-Myc-driven lymphomagenesis. Similarly, we failed to observe a modulatory effect of Raidd deficiency on DNA-damage-driven cancer. The role of Caspase-2 as a tumor suppressor and that of Pidd1 as a tumor promoter can therefore be uncoupled from their ability to interact with the Raidd scaffold, pointing toward the existence of alternative signaling modules engaging these two proteins in this context.


Subject(s)
CRADD Signaling Adaptor Protein/metabolism , Caspase 2/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , CRADD Signaling Adaptor Protein/genetics , Caspase 2/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Survival/genetics , Cell Survival/radiation effects , Cells, Cultured , DNA Damage/genetics , DNA Damage/radiation effects , Death Domain Receptor Signaling Adaptor Proteins/genetics , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Mice , Proto-Oncogene Proteins c-myc/genetics
2.
Lung Cancer ; 83(3): 408-10, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24412619

ABSTRACT

We report the case of a female never-smoking patient with an epidermal growth factor receptor (EGFR) mutation positive advanced non-small cell lung cancer (NSCLC) who received multiple lines of treatment. When she evolved clinical resistance to first generation EGFR tyrosine kinase inhibitors (TKI), she was treated with a fifth-line combination therapy with cetuximab and vinorelbine. This combination was highly active with a treatment response lasting for 9 months supporting the hypothesis that EGFR monoclonal antibodies in combination with chemotherapy may play a role in reversing EGFR-TKI resistance in EGFR mutation-positive NSCLC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Carcinoma, Non-Small-Cell Lung/genetics , Cetuximab , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Erlotinib Hydrochloride , Female , Gefitinib , Humans , Lung Neoplasms/genetics , Mutation/genetics , Neoplasm Staging , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Quinazolines/administration & dosage , Quinazolines/adverse effects , Remission Induction , Vinblastine/administration & dosage , Vinblastine/adverse effects , Vinblastine/analogs & derivatives , Vinorelbine
3.
Osteoporos Int ; 25(3): 1151-61, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23974861

ABSTRACT

SUMMARY: In the present study, we evaluated the potential for aminobisphosphonates to enhance the development of bone-forming osteoblasts from progenitor cells isolated from aged female osteoporotic patients. The aminobisphosphonates tested significantly enhanced osteoblast formation and thus lend further insights into their possible mode of action in the treatment of osteoporosis. INTRODUCTION: The primary aim of this study was to evaluate the influence of aminobisphosphonates on the osteogenesis of human bone marrow stromal cells (hBMSCs) and mineralization of differentiating bone-forming cells isolated from osteoporotic patients. METHODS: The influence of aminobisphosphonate treatment on hBMSC osteogenesis was assessed by the quantitative measurement of alkaline phosphatase (ALP) activity, in addition to quantitative reverse transcription polymerase chain reaction and Western blot analysis of known osteogenic markers. Mineralized matrix formation by hBMSC-derived osteoblasts was visualized and quantified using Alizarin red staining. RESULTS: hBMSC cultures treated with osteogenic medium supplemented with zoledronate demonstrated a significant increase in Alizarin red staining after 3 weeks as compared to cells cultured in osteogenic medium alone. Similarly, cultures of differentiating hBMSCs isolated from patients receiving alendronate treatment also demonstrated an increased propensity for mineralization, even in the absence of further in vitro stimulation by zoledronate. The stimulatory effects of aminobisphosphonate treatment on hBMSC-derived osteoblast-mediated mineralization were independent of any alterations in ALP activity, although significant decreases in the expression levels of osteopontin (SPP1) were evident in hBMSCs following exposure to aminobisphosphonates. Further analysis including Western blotting and loss-of-function studies revealed osteopontin as having a negative influence on the mineralization of differentiating osteoporotic bone-forming cells. CONCLUSIONS: The results presented here demonstrate for the first time that aminobisphosphonate treatment of osteoporotic hBMSCs enhances their capacity for osteoblast formation and subsequent mineral deposition, thus supporting the concept of aminobisphosphonates as having an osteoanabolic effect in osteoporosis.


Subject(s)
Alendronate/pharmacology , Bone Density Conservation Agents/pharmacology , Diphosphonates/pharmacology , Imidazoles/pharmacology , Mesenchymal Stem Cells/drug effects , Osteoporosis, Postmenopausal/pathology , Aged , Aged, 80 and over , Alendronate/therapeutic use , Alkaline Phosphatase/metabolism , Bone Density/drug effects , Bone Density Conservation Agents/therapeutic use , Cell Differentiation/drug effects , Cells, Cultured , Female , Humans , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/pathology , Osteogenesis/drug effects , Osteopontin/physiology , Osteoporosis, Postmenopausal/drug therapy , Osteoporosis, Postmenopausal/physiopathology , Zoledronic Acid
4.
Cell Death Dis ; 4: e942, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24309929

ABSTRACT

Much effort has been put in the discovery of ways to selectively kill p53-deficient tumor cells and targeting cell cycle checkpoint pathways has revealed promising candidates. Studies in zebrafish and human cell lines suggested that the DNA damage response kinase, checkpoint kinase 1 (Chk1), not only regulates onset of mitosis but also cell death in response to DNA damage in the absence of p53. This effect reportedly relies on ataxia telangiectasia mutated (ATM)-dependent and PIDDosome-mediated activation of Caspase-2. However, we show that genetic ablation of PIDDosome components in mice does not affect cell death in response to γ-irradiation. Furthermore, Chk1 inhibition largely failed to sensitize normal and malignant cells from p53(-/-) mice toward DNA damaging agents, and p53 status did not affect the death-inducing activity of DNA damage after Chk1 inhibition in human cancer cells. These observations argue against cross-species conservation of a Chk1-controlled cell survival pathway demanding further investigation of the molecular machinery responsible for cell death elicited by forced mitotic entry in the presence of DNA damage in different cell types and model organisms.


Subject(s)
Caspase 2/metabolism , DNA Damage/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Caspase 2/genetics , Cell Cycle/genetics , Cell Cycle/physiology , DNA Damage/genetics , Death Domain Receptor Signaling Adaptor Proteins/genetics , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Immunoblotting , Mice , Mice, Inbred C57BL , Mitosis/genetics , Mitosis/physiology , Tumor Suppressor Protein p53/genetics
5.
Cell Death Differ ; 20(4): 546-57, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23238565

ABSTRACT

Activation of NF-κB (nuclear factor of kappa light chain gene enhancer in B cells) in response to DNA damage is considered to contribute to repair of genetic lesions, increased cell survival and cytokine release. The molecular mechanisms orchestrating this cytoplasmic event involve core components of the nuclear DNA damage response machinery, including ATM-kinase (ataxia telangiectasia mutated kinase) and PARP-1 (poly (ADP-ribose) polymerase 1). The physiological consequences of defective NF-κB activation in this context, however, remain poorly investigated. Here we report on the role of the 'p53-induced protein with a death domain', PIDD, which appears rate limiting in this process, as is PARP-1. Despite impaired NF-κB activation, DNA damage did not increase cell death or reduce clonal survival of various cell types lacking PIDD, such as mouse embryonic fibroblasts or stem and progenitor cells of the hematopoietic system. Furthermore, lymphomagenesis induced by γ-irradiation (IR) was unaffected by deficiency for PIDD or PARP-1, indicating that loss of DNA damage-triggered NF-κB signalling does not affect IR-driven tumorigenesis. However, loss of either gene compromised cytokine release after acute IR injury. Hence, we propose that NF-κB's most notable function after DNA damage in primary cells is related to the release of cytokines, thereby contributing to sterile inflammation.


Subject(s)
Cytokines/metabolism , DNA Damage , Death Domain Receptor Signaling Adaptor Proteins/metabolism , NF-kappa B/metabolism , Animals , Apoptosis/radiation effects , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Survival/drug effects , Cell Transformation, Neoplastic/radiation effects , Cells, Cultured , DNA Damage/radiation effects , DNA-Binding Proteins/metabolism , Death Domain Receptor Signaling Adaptor Proteins/genetics , Granulocyte Colony-Stimulating Factor/pharmacology , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Radiation, Ionizing , Signal Transduction , Transcription Factor RelA/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/metabolism
6.
Cell Death Differ ; 19(10): 1722-32, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22595758

ABSTRACT

The PIDDosome, a multiprotein complex constituted of the 'p53-induced protein with a death domain (PIDD), 'receptor-interacting protein (RIP)-associated ICH-1/CED-3 homologous protein with a death domain' (RAIDD) and pro-Caspase-2 has been defined as an activating platform for this apoptosis-related protease. PIDD has been implicated in p53-mediated cell death in response to DNA damage but also in DNA repair and nuclear factor kappa-light-chain enhancer (NF-κB) activation upon genotoxic stress, together with RIP-1 kinase and Nemo/IKKγ. As all these cellular responses are critical for tumor suppression and deregulated expression of individual PIDDosome components has been noted in human cancer, we investigated their role in oncogenesis induced by DNA damage or oncogenic stress in gene-ablated mice. We observed that Pidd or Caspase-2 failed to suppress lymphoma formation triggered by γ-irradiation or 3-methylcholanthrene-driven fibrosarcoma development. In contrast, Caspase-2 showed tumor suppressive capacity in response to aberrant c-Myc expression, which did not rely on PIDD, the BH3-only protein Bid (BH3 interacting domain death agonist) or the death receptor ligand Trail (TNF-related apoptosis-inducing ligand), but associated with reduced rates of p53 loss and increased extranodal dissemination of tumor cells. In contrast, Pidd deficiency associated with abnormal M-phase progression and delayed disease onset, indicating that both proteins are differentially engaged upon oncogenic stress triggered by c-Myc, leading to opposing effects on tumor-free survival.


Subject(s)
CRADD Signaling Adaptor Protein/metabolism , Caspase 2/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Animals , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/metabolism , CRADD Signaling Adaptor Protein/antagonists & inhibitors , CRADD Signaling Adaptor Protein/genetics , Caspase 2/deficiency , Caspase 2/genetics , Cell Line , DNA Damage , Death Domain Receptor Signaling Adaptor Proteins/antagonists & inhibitors , Death Domain Receptor Signaling Adaptor Proteins/genetics , GTPase-Activating Proteins/metabolism , Gamma Rays , HCT116 Cells , Humans , I-kappa B Kinase/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Methylcholanthrene/pharmacology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Suppressor Protein p53/metabolism
7.
Oncogene ; 31(45): 4733-9, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-22266869

ABSTRACT

P53-induced protein with a death domain (PIDD) has been described as primary p53 target gene, induced upon DNA damage. More than 10 years after its discovery, its physiological role in the DNA damage response remains enigmatic, as it seems to be able to execute life-death decisions in vitro, yet genetic ablation in mice failed to reveal an obvious phenotype. Nonetheless, evidence is accumulating that it contributes to the fine-tuning of the DNA-damage response by orchestrating critical processes such as caspase activation or nuclear factor κB translocation and can also exert additional nuclear functions, for example, the modulation of translesion synthesis. In this review, we aim to integrate these observations and propose possible unexplored functions of PIDD.


Subject(s)
Death Domain Receptor Signaling Adaptor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Alternative Splicing , Animals , Apoptosis , Caspase 2/metabolism , Cell Survival , DNA Repair , Death Domain Receptor Signaling Adaptor Proteins/genetics , Drug Resistance, Neoplasm/genetics , Enzyme Activation , Humans , NF-kappa B/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Protein Binding , Protein Isoforms/genetics , Proteolysis , Tumor Suppressor Protein p53/genetics
8.
Cell Death Differ ; 18(6): 1036-45, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21415862

ABSTRACT

PIDD has been implicated in survival and apoptotic pathways in response to DNA damage, and a role for PIDD was recently identified in non-homologous end-joining (NHEJ) repair induced by γ-irradiation. Here, we present an interaction of PIDD with PCNA, first identified in a proteomics screen. PCNA has essential functions in DNA replication and repair following UV irradiation. Translesion synthesis (TLS) is a process that prevents UV irradiation-induced replication blockage and is characterized by PCNA monoubiquitination and interaction with the TLS polymerase eta (polη). Both of these processes are inhibited by p21. We report that PIDD modulates p21-PCNA dissociation, and promotes PCNA monoubiquitination and interaction with polη in response to UV irradiation. Furthermore, PIDD deficiency leads to a defect in TLS that is associated, both in vitro and in vivo, with cellular sensitization to UV-induced apoptosis. Thus, PIDD performs key functions upon UV irradiation, including TLS, NHEJ, NF-κB activation and cell death.


Subject(s)
Carrier Proteins/metabolism , DNA Damage/radiation effects , DNA Repair/radiation effects , DNA Replication/radiation effects , DNA/biosynthesis , Ultraviolet Rays , Apoptosis/genetics , Apoptosis/radiation effects , Carrier Proteins/genetics , Cell Line , DNA/genetics , DNA Damage/genetics , DNA Repair/genetics , DNA Replication/genetics , Death Domain Receptor Signaling Adaptor Proteins , Gamma Rays , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitination/genetics , Ubiquitination/radiation effects
9.
Oncogene ; 28(35): 3093-6, 2009 Sep 03.
Article in English | MEDLINE | ID: mdl-19581929

ABSTRACT

Despite the early discovery of caspase-2, its physiological function has long remained an enigma. A number of recent publications now suggest not just one, but multiple functions, including roles in apoptosis, DNA repair and tumor suppression. How can one enzyme have so many talents? Considering the diversity of interaction partners and the specific mode of pro-apoptotic action proposed in these studies, caspase-2 might in fact represent a primordial protease serving numerous pathways, established before the advent of a more elaborate functionally diversified caspases system.


Subject(s)
Caspase 2/physiology , Animals , Apoptosis/physiology , Caspase 2/deficiency , Cell Transformation, Neoplastic/metabolism , DNA Repair/physiology , Enzyme Activation , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Mice, Transgenic , Models, Biological
10.
Cell Death Differ ; 16(2): 195-207, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19023332

ABSTRACT

Proteolysis of cellular substrates by caspases (cysteine-dependent aspartate-specific proteases) is one of the hallmarks of apoptotic cell death. Although the activation of apoptotic caspases is considered a 'late-stage' event in apoptosis signaling, past the commitment stage, one caspase family member, caspase-2, splits the cell death community into half - those searching for evidence of an apical initiator function of this molecule and those considering it as an amplifier of the apoptotic caspase cascade, at best, if relevant for apoptosis at all. This review screens past and present biochemical as well as genetic evidence for caspase-2 function in cell death signaling and beyond.


Subject(s)
Caspase 2/metabolism , Apoptosis , CRADD Signaling Adaptor Protein/metabolism , Caspase 2/genetics , Cell Death , DNA Damage , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Cell Death Differ ; 14(9): 1561-75, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17627286

ABSTRACT

A number of elegant studies exploring the consequences of expression of various mutant forms of p53 in mice have been published over the last years. The results and conclusions drawn from these studies often contradict results previously obtained in biochemical assays and cell biology studies, questioning their relevance for p53 function in vivo. Owing to the multitude of post-translational modifications imposed on p53, however, the in vivo validation of their relevance for proper protein function and tumour suppression is constantly lagging behind new biochemical discoveries. Nevertheless, mouse genetics presents again its enormous power. Despite being relatively slow and tedious, it has become indispensable for researchers to sort out the wheat from the chaff in an endless sea of publications on p53.


Subject(s)
Apoptosis , Protein Processing, Post-Translational , Tumor Suppressor Protein p53/metabolism , Ubiquitin/metabolism , Acetylation , Animals , Gene Expression Regulation, Neoplastic , Genes, p53 , Humans , Methylation , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Structure-Activity Relationship , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
13.
Physiol Biochem Zool ; 74(3): 413-9, 2001.
Article in English | MEDLINE | ID: mdl-11331514

ABSTRACT

The importance of glycolysis, as an ATP-producing and substrate-providing pathway, was studied in anoxia-tolerant (goldfish) and anoxia-intolerant (trout) hepatocytes. Inhibition of glycolysis with iodoacetic acid (IAA) left aerobic ATP production largely unaffected in hepatocytes from both species but caused a significant decrease of ATP contents in the goldfish cells. Ouabain-sensitive oxygen consumption (osVo2), an estimate of mitochondrial ATP production coupled to ATP consumption by the Na(+) pump, was significantly reduced in IAA-treated goldfish hepatocytes, whereas it was unaltered in trout hepatocytes. Partial reduction of mitochondrial respiration, achieved by titration with cyanide (CN), strongly stimulated glycolytic flux but did not affect ATP contents of hepatocytes from both species. Under these conditions, osVo2 became undetectable. Rb(+)-uptake rates, providing a direct estimate of Na(+)-pump activity, were in good agreement with estimates derived from osVo2 in IAA-treated cells, showing a decrease in goldfish and no change in trout. However, they indicated persistent Na(+)-pump activity despite the lack of osVo2 in CN-treated cells. Overall, these data indicate that in goldfish hepatocytes Na(+)-pump activity is more dependent on glycolytic ATP production as compared to trout hepatocytes. Protein synthesis of goldfish hepatocytes was inhibited in IAA- and CN-treated cells, possibly reflecting the hierarchical organization of energy metabolism. In trout hepatocytes, protein synthesis could be sustained at control levels, given that energetic substrate provision was not limited.


Subject(s)
Glycolysis/physiology , Goldfish/physiology , Hepatocytes/metabolism , Mitochondria, Liver/metabolism , Oxygen Consumption/physiology , Trout/physiology , Acclimatization , Adenosine Triphosphate/metabolism , Animals , Hypoxia , Kinetics , Lactates/metabolism , Rubidium/metabolism , Species Specificity , Temperature
14.
Gen Comp Endocrinol ; 121(2): 205-13, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11178886

ABSTRACT

The effect of epinephrine on various aspects of cellular metabolism was studied in hepatocytes from the goldfish Carassius auratus. Epinephrine increased cytosolic free calcium ([Ca2+](i)) from a baseline value of 108 +/- 22 nM to a peak value of 577 +/- 127 nM in suspensions of hepatocytes. Responses of single cells ranged from a single spike (66% of hepatocytes) to variable oscillatory patterns (34%). The increase in [Ca(2+)](i) was independent of the presence of extracellular Ca2+ and was prevented by the alpha-adrenergic antagonist phentolamine. Cellular glucose release induced by epinephrine (1.7- to 3.2-fold) was significantly reduced in Ca2+-depleted cells and in the presence of phentolamine, providing evidence for the co-occurrence of alpha-adrenoceptors and a Ca2+-independent, presumably beta-adrenergic, system in these cells. Furthermore, epinephrine stimulated oxygen consumption in a Ca2+-dependent manner, which was not due to stimulated Na(+) pump activity. An increased rate of acid secretion of 50%, evoked by epinephrine, appears to be mediated by enhanced Na(+)/H(+) exchange but did not result in intracellular alkalization.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Epinephrine/pharmacology , Goldfish/physiology , Hepatocytes/drug effects , Receptors, Adrenergic, alpha/drug effects , Acids/metabolism , Animals , Calcium/metabolism , Glucose/metabolism , Hydrogen-Ion Concentration , Lactic Acid/metabolism , Oxygen Consumption/drug effects
15.
J Exp Biol ; 204(Pt 22): 3943-51, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11807112

ABSTRACT

Mechanisms of intracellular pH (pHi) regulation were investigated in anoxia-tolerant hepatocytes from goldfish Carassius auratus, and compared to the situation in the anoxia-intolerant hepatocytes from trout Oncorhynchus mykiss. Under normoxic conditions, the pHi of goldfish hepatocytes was regulated by a Na(+)/H(+) exchanger and a Na(+)-independent Cl(-)/HCO(3)(-) exchanger, the latter being activated only after acidification of the cells. Mechanisms of acid secretion appear to be fuelled, at least in part, by lactate formation under fully aerobic conditions, as inhibition of glycolysis caused a drastic reduction of steady state proton release. In trout hepatocytes both a Na(+)/H(+) exchanger and a Cl(-)/HCO(3)(-) exchanger were found to be tonically active, as described previously. During chemical anoxia a constant pHi was maintained in goldfish hepatocytes, whereas it was reversibly reduced by 0.3 units in the trout cells. Under these conditions a reversible increase in the rate of acid secretion was induced in the cells from both species. In the goldfish cells this was based on a SITS-sensitive transporter, possibly involving export of lactate, with no contribution from Na(+)/H(+) exchange. By contrast, in hepatocytes from trout, CN-induced acid secretion was dominated by the activity of the Na(+)/H(+) exchanger. Brief exposure to extracellular acidosis had no dramatic effects on the energetics of hepatocytes from either species.


Subject(s)
Cell Hypoxia , Goldfish/metabolism , Hepatocytes/metabolism , Oncorhynchus mykiss/metabolism , Adenosine Triphosphate/metabolism , Animals , Chloride-Bicarbonate Antiporters/metabolism , Homeostasis , Hydrogen-Ion Concentration , Kinetics , Lactic Acid/metabolism , Propionates/pharmacology , Sodium Cyanide/pharmacology , Sodium-Hydrogen Exchangers/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...