Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(16)2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37628828

ABSTRACT

Acid sphingomyelinase deficiency (ASMD) or Niemann-Pick disease type A (NPA), type B (NPB) and type A/B (NPA/B), is a rare lysosomal storage disease characterized by progressive accumulation of sphingomyelin (SM) in the liver, lungs, bone marrow and, in severe cases, neurons. A disease model was established by generating liver organoids from a NPB patient carrying the p.Arg610del variant in the SMPD1 gene. Liver organoids were characterized by transcriptomic and lipidomic analysis. We observed altered lipid homeostasis in the patient-derived organoids showing the predictable increase in sphingomyelin (SM), together with cholesterol esters (CE) and triacylglycerides (TAG), and a reduction in phosphatidylcholine (PC) and cardiolipins (CL). Analysis of lysosomal gene expression pointed to 24 downregulated genes, including SMPD1, and 26 upregulated genes that reflect the lysosomal stress typical of the disease. Altered genes revealed reduced expression of enzymes that could be involved in the accumulation in the hepatocytes of sphyngoglycolipids and glycoproteins, as well as upregulated genes coding for different glycosidases and cathepsins. Lipidic and transcriptome changes support the use of hepatic organoids as ideal models for ASMD investigation.


Subject(s)
Niemann-Pick Disease, Type A , Niemann-Pick Diseases , Humans , Niemann-Pick Disease, Type A/genetics , Sphingomyelins , Liver , Gene Expression
2.
Cancer Res ; 81(8): 2142-2156, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33593822

ABSTRACT

The extraordinary plasticity of glioma cells allows them to contribute to different cellular compartments in tumor vessels, reinforcing the vascular architecture. It was recently revealed that targeting glioma-derived pericytes, which represent a big percentage of the mural cell population in aggressive tumors, increases the permeability of the vessels and improves the efficiency of chemotherapy. However, the molecular determinants of this transdifferentiation process have not been elucidated. Here we show that mutations in EGFR stimulate the capacity of glioma cells to function as pericytes in a BMX- (bone marrow and X-linked) and SOX9-dependent manner. Subsequent activation of platelet-derived growth factor receptor beta in the vessel walls of EGFR-mutant gliomas stabilized the vasculature and facilitated the recruitment of immune cells. These changes in the tumor microenvironment conferred a growth advantage to the tumors but also rendered them sensitive to pericyte-targeting molecules such as ibrutinib or sunitinib. In the absence of EGFR mutations, high-grade gliomas were enriched in blood vessels, but showed a highly disrupted blood-brain barrier due to the decreased BMX/SOX9 activation and pericyte coverage, which led to poor oxygenation, necrosis, and hypoxia. Overall, these findings identify EGFR mutations as key regulators of the glioma-to-pericyte transdifferentiation, highlighting the intricate relationship between the tumor cells and their vascular and immune milieu. Our results lay the foundations for a vascular-dependent stratification of gliomas and suggest different therapeutic vulnerabilities determined by the genetic status of EGFR. SIGNIFICANCE: This study identifies the EGFR-related mechanisms that govern the capacity of glioma cells to transdifferentiate into pericytes, regulating the vascular and immune phenotypes of the tumors. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2142/F1.large.jpg.


Subject(s)
Brain Neoplasms/blood supply , Cell Transdifferentiation , Cellular Microenvironment , Glioma/blood supply , Mutation , Pericytes/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , Blood-Brain Barrier/metabolism , Bone Marrow , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Line, Tumor , Chromosomes, Human, X , ErbB Receptors/genetics , Glioma/immunology , Glioma/pathology , Humans , Immunity, Cellular , Isocitrate Dehydrogenase/genetics , Mice , Pericytes/drug effects , Pericytes/metabolism , Piperidines/pharmacology , Receptor, Platelet-Derived Growth Factor beta/metabolism , SOX9 Transcription Factor , Sunitinib/pharmacology , Tumor Hypoxia , Tumor Microenvironment
3.
Cancers (Basel) ; 12(1)2020 Jan 14.
Article in English | MEDLINE | ID: mdl-31947645

ABSTRACT

Despite the high frequency of EGFR and TP53 genetic alterations in gliomas, little is known about their crosstalk during tumor progression. Here, we described a mutually exclusive distribution between mutations in these two genes. We found that wild-type p53 gliomas are more aggressive than their mutant counterparts, probably because the former accumulate amplifications and/or mutations in EGFR and show a stronger activation of this receptor. In addition, we identified a series of genes associated with vesicular trafficking of EGFR in p53 wild-type gliomas. Among these genes, TMEM167A showed the strongest implication in overall survival in this group of tumors. In agreement with this observation, inhibition of TMEM167A expression impaired the subcutaneous and the intracranial growth of wild-type p53 gliomas, regardless of the presence of EGFR mutations. In the absence of p53 mutations, TMEM167A knockdown reduced the acidification of intracellular vesicles, affecting the autophagy process and impairing EGFR trafficking and signaling. This effect was mimicked by an inhibitor of the vacuolar ATPase. We propose that the increased aggressiveness of wild-type p53 gliomas might be due to the increase in growth factor signaling activity, which depends on the regulation of vesicular trafficking by TMEM167A.

4.
Oxid Med Cell Longev ; 2019: 9719730, 2019.
Article in English | MEDLINE | ID: mdl-31467641

ABSTRACT

Glioblastoma (GBM) is the most common and devastating primary brain tumor. The presence of cancer stem cells (CSCs) has been linked to their therapy resistance. Molecular and cellular components of the tumor microenvironment also play a fundamental role in the aggressiveness of these tumors. In particular, high levels of hypoxia and reactive oxygen species participate in several aspects of GBM biology. Moreover, GBM contains a large number of macrophages, which normally behave as immunosuppressive tumor-supportive cells. In fact, the presence of both, hypoxia and M2-like macrophages, correlates with malignancy and poor prognosis in gliomas. Antioxidant agents, as nutritional supplements, might have antitumor activity. Ocoxin® oral solution (OOS), in particular, has anti-inflammatory and antioxidant properties, as well as antitumor properties in several neoplasia, without known side effects. Here, we describe how OOS affects stem cell properties in certain GBMs, slowing down their tumor growth. In parallel, OOS has a direct effect on macrophage polarization in vitro and in vivo, inhibiting the protumoral features of M2 macrophages. Therefore, OOS could be a feasible candidate to be used in combination therapies during GBM treatment because it can target the highly resilient CSCs as well as their supportive immune microenvironment, without adding toxicity to conventional treatments.


Subject(s)
Ascorbic Acid/therapeutic use , Glioblastoma/drug therapy , Macrophages/metabolism , Neoplastic Stem Cells/metabolism , Plant Extracts/therapeutic use , Vitamin B 12/therapeutic use , Vitamin B 6/therapeutic use , Animals , Ascorbic Acid/pharmacology , Folic Acid , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Pantothenic Acid , Plant Extracts/pharmacology , Vitamin B 12/pharmacology , Vitamin B 6/pharmacology , Zinc Sulfate
5.
Oncotarget ; 8(6): 9767-9782, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28038459

ABSTRACT

Neogenin-1 (NEO1) is a transmembrane receptor involved in axonal guidance, angiogenesis, neuronal cell migration and cell death, during both embryonic development and adult homeostasis. It has been described as a dependence receptor, because it promotes cell death in the absence of its ligands (Netrin and Repulsive Guidance Molecule (RGM) families) and cell survival when they are present. Although NEO1 and its ligands are involved in tumor progression, their precise role in tumor cell survival and migration remain unclear. Public databases contain extensive information regarding the expression of NEO1 and its ligands Netrin-1 (NTN1) and Netrin-4 (NTN4) in primary neuroblastoma (NB) tumors. Analysis of this data revealed that patients with high expression levels of both NEO1 and NTN4 have a poor survival rate. Accordingly, our analyses in NB cell lines with different genetic backgrounds revealed that knocking-down NEO1 reduces cell migration, whereas silencing of endogenous NTN4 induced cell death. Conversely, overexpression of NEO1 resulted in higher cell migration in the presence of NTN4, and increased apoptosis in the absence of ligand. Increased apoptosis was prevented when utilizing physiological concentrations of exogenous Netrin-4. Likewise, cell death induced after NTN4 knock-down was rescued when NEO1 was transiently silenced, thus revealing an important role for NEO1 in NB cell survival. In vivo analysis, using the chicken embryo chorioallantoic membrane (CAM) model, showed that NEO1 and endogenous NTN4 are involved in tumor extravasation and metastasis. Our data collectively demonstrate that endogenous NTN4/NEO1 maintain NB growth via both pro-survival and pro-migratory molecular signaling.


Subject(s)
Cell Movement , Chorioallantoic Membrane/blood supply , Nerve Tissue Proteins/metabolism , Netrins/metabolism , Neuroblastoma/metabolism , Receptors, Cell Surface/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Survival , Chick Embryo , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Neoplasm Invasiveness , Nerve Tissue Proteins/genetics , Netrins/genetics , Neuroblastoma/genetics , Neuroblastoma/mortality , Neuroblastoma/secondary , RNA Interference , Receptors, Cell Surface/genetics , Signal Transduction , Time Factors , Transfection
6.
FEBS Lett ; 587(14): 2272-7, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23747309

ABSTRACT

Alternative forms of proinsulin mRNA with differential translational capacities and unknown significance are expressed in several developing tissues and in the adult pancreas. In the chick embryo developing heart, we observed low expression of the translationally active transcript of embryonic proinsulin (Pro1B), and predominant expression of the intron 1-unspliced variant, translationally inactive. In the embryonic mouse heart, intron 1-unspliced isoform appeared after E12.5. This tight regulation is required for normal development, since overexpression of Pro1B resulted in abnormal cardiac morphogenesis in 40% of chick embryos, and was accompanied by changes in gene expression of Amhc1and Vmhc1.


Subject(s)
Alternative Splicing , Heart/embryology , Morphogenesis , Proinsulin/genetics , RNA, Messenger/genetics , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Chick Embryo , Female , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Proinsulin/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism
7.
Cell Calcium ; 45(3): 243-50, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19100620

ABSTRACT

Mitochondrial Ca(2+) activates many processes, from mitochondrial metabolism to opening of the permeability transition pore (PTP) and apoptosis. However, there is considerable controversy regarding the free mitochondrial [Ca(2+)] ([Ca(2+)](M)) levels that can be attained during cell activation or even in mitochondrial preparations. Studies using fluorescent dyes (rhod-2 or similar), have reported that phosphate precipitation precludes [Ca(2+)](M) from increasing above 2-3 microM. Instead, using low-Ca(2+)-affinity aequorin probes, we have measured [Ca(2+)](M) values more than two orders of magnitude higher. We confirm here these values by making a direct in situ calibration of mitochondrial aequorin, and we show that a prolonged increase in [Ca(2+)](M) to levels of 0.5-1mM was actually observed at any phosphate concentration (0-10mM) during continuous perfusion of 3.5-100 microM Ca(2+)-buffers. In spite of this high and maintained (>10 min) [Ca(2+)](M), mitochondria retained functionality and the [Ca(2+)](M) drop induced by a protonophore was fully reversible. In addition, this high [Ca(2+)](M) did not induce PTP opening unless additional activators (phenyl arsine oxide, PAO) were present. PAO induced a rapid, concentration-dependent and irreversible drop in [Ca(2+)](M). In conclusion [Ca(2+)](M) levels of 0.5-1mM can be reached and maintained for prolonged periods (>10 min) in phosphate-containing medium, and massive opening of PTP requires additional pore activators.


Subject(s)
Calcium/metabolism , Mitochondria/metabolism , Aequorin/metabolism , Animals , Arsenicals/pharmacology , Buffers , Calcium/pharmacology , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cattle , Cells, Cultured , Chromaffin Cells/drug effects , Chromaffin Cells/metabolism , HeLa Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mutant Proteins/metabolism , Perfusion , Permeability/drug effects , Phosphates/pharmacology
8.
Eur J Neurosci ; 28(7): 1265-74, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18973554

ABSTRACT

The secretory granules constitute one of the less well-known compartments in terms of Ca2+ dynamics. They contain large amounts of total Ca2+, but the free intragranular [Ca2+] ([Ca2+]SG), the mechanisms for Ca2+ uptake and release from the granules and their physiological significance regarding exocytosis are still matters of debate. We used in the present work an aequorin chimera targeted to the granules to investigate [Ca2+]SG homeostasis in bovine adrenal chromaffin cells. We found that most of the intracellular aequorin chimera is present in a compartment with 50-100 microM Ca2+. Ca2+ accumulation into this compartment takes place mainly through an ATP-dependent mechanism, namely, a thapsigargin-sensitive Ca2+-ATPase. In addition, fast Ca2+ release was observed in permeabilized cells after addition of inositol 1,4,5-trisphosphate (InsP3) or caffeine, suggesting the presence of InsP3 and ryanodine receptors in the vesicular membrane. Stimulation of intact cells with the InsP3-producing agonist histamine or with caffeine also induced Ca2+ release from the vesicles, whereas acetylcholine or high-[K+] depolarization induced biphasic changes in vesicular[Ca2+], suggesting heterogeneous responses of different vesicle populations, some of them releasing and some taking up Ca2+during stimulation. In conclusion, our data show that chromaffin cell secretory granules have the machinery required for rapid uptake and release of Ca2+, and this strongly supports the hypothesis that granular Ca2+ may contribute to its own secretion.


Subject(s)
Adrenal Medulla/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Chromaffin Cells/metabolism , Secretory Vesicles/metabolism , Adenosine Triphosphate/metabolism , Adrenal Medulla/cytology , Aequorin/genetics , Aequorin/metabolism , Animals , Calcium/pharmacology , Calcium Signaling/drug effects , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/metabolism , Catecholamines/metabolism , Cattle , Cell Compartmentation/drug effects , Cell Compartmentation/physiology , Cells, Cultured , Chromaffin Cells/drug effects , Enzyme Inhibitors/pharmacology , Inositol 1,4,5-Trisphosphate/metabolism , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ryanodine Receptor Calcium Release Channel/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Secretory Vesicles/drug effects , Thapsigargin/pharmacology
9.
J Physiol ; 580(Pt 1): 39-49, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17234694

ABSTRACT

The recent availability of activators of the mitochondrial Ca(2+) uniporter allows direct testing of the influence of mitochondrial Ca(2+) uptake on the overall Ca(2+) homeostasis of the cell. We show here that activation of mitochondrial Ca(2+) uptake by 4,4',4''-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT) or kaempferol stimulates histamine-induced Ca(2+) release from the endoplasmic reticulum (ER) and that this effect is enhanced if the mitochondrial Na(+)-Ca(2+) exchanger is simultaneously inhibited with CGP37157. This suggests that both Ca(2+) uptake and release from mitochondria control the ability of local Ca(2+) microdomains to produce feedback inhibition of inositol 1,4,5-trisphosphate receptors (InsP(3)Rs). In addition, the ability of mitochondria to control Ca(2+) release from the ER allows them to modulate cytosolic Ca(2+) oscillations. In histamine stimulated HeLa cells and human fibroblasts, both PPT and kaempferol initially stimulated and later inhibited oscillations, although kaempferol usually induced a more prolonged period of stimulation. Both compounds were also able to induce the generation of Ca(2+) oscillations in previously silent fibroblasts. Our data suggest that cytosolic Ca(2+) oscillations are exquisitely sensitive to the rates of mitochondrial Ca(2+) uptake and release, which precisely control the size of the local Ca(2+) microdomains around InsP(3)Rs and thus the ability to produce feedback activation or inhibition of Ca(2+) release.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Fibroblasts/physiology , Mitochondria/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Clonazepam/analogs & derivatives , Clonazepam/pharmacology , Cytosol/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Estrogen Antagonists/pharmacology , Fibroblasts/drug effects , HeLa Cells , Histamine/pharmacology , Humans , Inositol 1,4,5-Trisphosphate Receptors/drug effects , Kaempferols/pharmacology , Mitochondria/drug effects , Stimulation, Chemical , Thiazepines/pharmacology
10.
Cell Calcium ; 40(1): 53-61, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16720043

ABSTRACT

There is increasing evidence that mitochondria play an important role in the control of cytosolic Ca2+ signaling. We show here that the main mitochondrial Ca2+-exit pathway, the mitochondrial Na+/Ca2+ exchanger, controls the pattern of cytosolic Ca2+ oscillations in non-excitable cells. In HeLa cells, the inhibitor of the mitochondrial Na+/Ca2+ exchanger CGP37157 changed the pattern of the oscillations induced by histamine from a high-frequency irregular one to a lower frequency baseline spike type, surprisingly with little changes in the average Ca2+ values of a large cell population. In human fibroblasts, CGP37157 increased the frequency of the baseline oscillations in cells having spontaneous activity and induced the generation of oscillations in cells without spontaneous activity. This effect was dose-dependent, disappeared when the inhibitor was washed out and was not mimicked by mitochondrial depolarization. CGP37157 increased mitochondrial [Ca2+] and ATP production in histamine-stimulated HeLa cells, but the effect on ATP production was only transient. CGP37157 also activated histamine-induced Ca2+ release from the endoplasmic reticulum and increased the size of the cytosolic Ca2+ peak induced by histamine in HeLa cells. Our results suggest that the mitochondrial Na+/Ca2+ exchanger directly modulates inositol 1,4,5-trisphosphate-induced Ca2+ release and in that way controls cytosolic Ca2+ oscillations.


Subject(s)
Calcium/metabolism , Cytosol/metabolism , Mitochondria/physiology , Sodium-Calcium Exchanger/physiology , HeLa Cells , Humans
11.
Br J Pharmacol ; 145(7): 862-71, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15912132

ABSTRACT

Ca(2+) uptake by mitochondria is a key element in the control of cellular Ca(2+) homeostasis and Ca(2+)-dependent phenomena. It has been known for many years that this Ca(2+) uptake is mediated by the mitochondrial Ca(2+) uniporter, a specific Ca(2+) channel of the inner mitochondrial membrane. We have shown previously that this channel is strongly activated by a series of natural phytoestrogenic flavonoids. We show here that several agonists and antagonists of estrogen receptors (ERs) also modulate the activity of the uniporter. The specific alpha-ER agonist 4,4',4''-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT) was the strongest activator, increasing the rate of mitochondrial Ca(2+) uptake in permeabilized HeLa cells by 10-fold at 2 microM. Consistently, PPT largely increased the histamine-induced mitochondrial [Ca(2+)] peak and reduced the cytosolic one. Diethylstilbestrol and 17-beta-estradiol (but not 17-alpha-estradiol) were active at pharmacological concentrations while the beta-estrogen-receptor agonist 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN) was little effective. The ER modulators tamoxifen and 4-hydroxy-tamoxifen inhibited mitochondrial Ca(2+) uptake (IC(50) 2.5+/-1.5 and 2.5+/-1.4 microM, mean+/-s.d., respectively) both in the presence and in the absence of PPT, but raloxifene and the pure estrogen antagonist ICI 182,780 produced no effect. Activation by PPT was immediate and inhibition by tamoxifen or 4-hydroxy-tamoxifen required only 5 min to reach maximum. Tamoxifen did not modify mitochondrial membrane potential and PPT induced a slow mitochondrial depolarization at higher concentrations than those required to activate mitochondrial Ca(2+) uptake. These results suggest that some kind of ER or related protein located in mitochondria controls the activity of the Ca(2+) uniporter by a nongenomic mechanism. This novel mechanism of action of estrogen agonists and antagonists can provide a new interpretation for several previously reported effects of these compounds.


Subject(s)
Calcium Channels/drug effects , Calcium/metabolism , Mitochondria/metabolism , Phenols/pharmacology , Pyrazoles/pharmacology , Receptors, Estrogen/agonists , Selective Estrogen Receptor Modulators/pharmacology , Aequorin/biosynthesis , Aequorin/genetics , Calcium/analysis , Cell Line, Tumor , Diethylstilbestrol/pharmacology , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogens, Non-Steroidal/pharmacology , Female , HeLa Cells , Humans , Luminescent Agents , Membrane Potentials/drug effects , Mitochondria/drug effects , Organometallic Compounds , Receptors, Estrogen/antagonists & inhibitors , Tamoxifen/analogs & derivatives , Tamoxifen/metabolism , Tamoxifen/pharmacology , Transfection
12.
Cell Calcium ; 37(6): 555-64, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15862346

ABSTRACT

We have used an aequorin chimera targeted to the membrane of the secretory granules to monitor the free [Ca(2+)] inside them in neurosecretory PC12 cells. More than 95% of the probe was located in a compartment with an homogeneous [Ca(2+)] around 40 microM. Cell stimulation with either ATP, caffeine or high-K(+) depolarization increased cytosolic [Ca(2+)] and decreased secretory granule [Ca(2+)] ([Ca(2+)](SG)). Inositol-(1,4,5)-trisphosphate, cyclic ADP ribose and nicotinic acid adenine dinucleotide phosphate were all ineffective to release Ca(2+) from the granules. Changes in cytosolic [Na(+)] (0-140 mM) or [Ca(2+)] (0-10 microM) did not modify either ([Ca(2+)](SG)). Instead, [Ca(2+)](SG) was highly sensitive to changes in the pH gradient between the cytosol and the granules. Both carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP) and nigericin, as well as cytosolic acidification, reversibly decreased [Ca(2+)](SG), while cytosolic alcalinization reversibly increased [Ca(2+)](SG). These results are consistent with the operation of a H(+)/Ca(2+) antiporter in the vesicular membrane. This antiporter could also mediate the effects of ATP, caffeine and high-K(+) on [Ca(2+)](SG), because all of them induced a transient cytosolic acidification. The FCCP-induced decrease in [Ca(2+)](SG) was reversible in 10-15 min even in the absence of cytosolic Ca(2+) or ATP, suggesting that most of the calcium content of the vesicles is bound to a slowly exchanging Ca(2+) buffer. This large store buffers [Ca(2+)](SG) changes in the long-term but allows highly dynamic free [Ca(2+)](SG) changes to occur in seconds or minutes.


Subject(s)
Calcium/metabolism , Catecholamines/metabolism , Neurons/metabolism , Secretory Vesicles/metabolism , Aequorin/genetics , Aequorin/metabolism , Animals , Membrane Proteins/genetics , Membrane Proteins/metabolism , PC12 Cells , Protein Transport , R-SNARE Proteins , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
13.
Biochem J ; 384(Pt 1): 19-24, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15324303

ABSTRACT

During cell activation, mitochondria play an important role in Ca2+ homoeostasis due to the presence of a fast and specific Ca2+ channel in its inner membrane, the mitochondrial Ca2+ uniporter. This channel allows mitochondria to buffer local cytosolic [Ca2+] changes and controls the intramitochondrial Ca2+ levels, thus modulating a variety of phenomena from respiratory rate to apoptosis. We have described recently that SB202190, an inhibitor of p38 MAPK (mitogen-activated protein kinase), strongly activated the uniporter. We show in the present study that a series of natural plant flavonoids, widely distributed in foods, produced also a strong stimulation of the mitochondrial Ca2+ uniporter. This effect was of the same magnitude as that induced by SB202190 (an approx. 20-fold increase in the mitochondrial Ca2+ uptake rate), developed without measurable delay and was rapidly reversible. In intact cells, the mitochondrial Ca2+ peak induced by histamine was also largely increased by the flavonoids. Stimulation of the uniporter by either flavonoids or SB202190 did not require ATP, suggesting a direct effect on the uniporter or an associated protein which is not mediated by protein phosphorylation. The most active compound, kaempferol, increased the rate of mitochondrial Ca2+ uptake by 85+/-15% (mean+/-S.E.M., n=4) and the histamine-induced mitochondrial Ca2+ peak by 139+/-19% (mean+/-S.E.M., n=5) at a concentration of 1 microM. Given that flavonoids can reach this concentration range in plasma after ingestion of flavonoid-rich food, these compounds could be modulating the uniporter under physiological conditions.


Subject(s)
Calcium-Binding Proteins/physiology , Calcium/metabolism , Flavonoids/pharmacology , Mitochondria/metabolism , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Flavonoids/chemistry , HeLa Cells/chemistry , HeLa Cells/metabolism , Humans , Ion Transport/drug effects , Ion Transport/physiology , Mitochondrial Proteins/physiology , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL
...