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










Publication year range
1.
Am J Physiol Gastrointest Liver Physiol ; 304(2): G142-56, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23139219

ABSTRACT

Colonic chloride secretion is regulated via the neurohormonal and immune systems. Exogenous chemicals (e.g., butyrate, propionate) can affect chloride secretion. Capsaicin, the pungent ingredient of the chili peppers, exerts various effects on gastrointestinal function. Capsaicin is known to activate the transient receptor potential vanilloid type 1 (TRPV1), expressed in the mesenteric nervous system. Recent studies have also demonstrated its presence in epithelial cells but its role remains uncertain. Because capsaicin has been reported to inhibit colonic chloride secretion, we tested whether this effect of capsaicin could occur by direct action on epithelial cells. In mouse colon and model T84 human colonic epithelial cells, we found that capsaicin inhibited forskolin-dependent short-circuit current (FSK-I(sc)). Using PCR and Western blot, we demonstrated the presence of TRPV1 in colonic epithelial cells. In T84 cells, TRPV1 localized at the basolateral membrane and in vesicular compartments. In permeabilized monolayers, capsaicin activated apical chloride conductance, had no effect on basolateral potassium conductance, but induced NKCC1 internalization demonstrated by immunocytochemistry and basolateral surface biotinylation. AMG-9810, a potent inhibitor of TRPV1, did not prevent the inhibition of the FSK-I(sc) by capsaicin. Neither resiniferatoxin nor N-oleoyldopamine, two selective agonists of TRPV1, blocked the FSK-I(sc). Conversely capsaicin, resiniferatoxin, and N-oleoyldopamine raised intracellular calcium ([Ca(2+)](i)) in T84 cells and AMG-9810 blocked the rise in [Ca(2+)](i) induced by capsaicin and resiniferatoxin suggesting the presence of a functional TRPV1 channel. We conclude that capsaicin inhibits chloride secretion in part by causing NKCC1 internalization, but by a mechanism that appears to be independent of TRPV1.


Subject(s)
Capsaicin/pharmacology , Chlorides/metabolism , Colon/drug effects , Epithelial Cells/drug effects , Intestinal Mucosa/drug effects , Sodium-Potassium-Chloride Symporters/metabolism , TRPV Cation Channels/metabolism , Acrylamides/pharmacology , Animals , Biotinylation , Blotting, Western , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Calcium/metabolism , Cell Line , Colforsin/pharmacology , Colon/metabolism , Diterpenes/pharmacology , Dopamine/analogs & derivatives , Dopamine/pharmacology , Dose-Response Relationship, Drug , Electric Conductivity , Epithelial Cells/metabolism , Humans , Immunohistochemistry , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Solute Carrier Family 12, Member 2 , TRPV Cation Channels/genetics , Time Factors
2.
PLoS One ; 6(9): e23880, 2011.
Article in English | MEDLINE | ID: mdl-21949685

ABSTRACT

Secreted Protein Acidic and Rich in Cysteine (SPARC) is one of the major non-structural proteins of the extracellular matrix (ECM) in remodeling tissues. The functional significance of SPARC is emphasized by its origin in the first multicellular organisms and its high degree of evolutionary conservation. Although SPARC has been shown to act as a critical modulator of ECM remodeling with profound effects on tissue physiology and architecture, no plausible molecular mechanism of its action has been proposed. In the present study, we demonstrate that SPARC mediates the disassembly and degradation of ECM networks by functioning as a matricellular chaperone. While it has low affinity to its targets inside the cells where the Ca(2+) concentrations are low, high extracellular concentrations of Ca(2+) activate binding to multiple ECM proteins, including collagens. We demonstrated that in vitro, this leads to the inhibition of collagen I fibrillogenesis and disassembly of pre-formed collagen I fibrils by SPARC at high Ca(2+) concentrations. In cell culture, exogenous SPARC was internalized by the fibroblast cells in a time- and concentration-dependent manner. Pulse-chase assay further revealed that internalized SPARC is quickly released outside the cell, demonstrating that SPARC shuttles between the cell and ECM. Fluorescently labeled collagen I, fibronectin, vitronectin, and laminin were co-internalized with SPARC by fibroblasts, and semi-quantitative Western blot showed that SPARC mediates internalization of collagen I. Using a novel 3-dimensional model of fluorescent ECM networks pre-deposited by live fibroblasts, we demonstrated that degradation of ECM depends on the chaperone activity of SPARC. These results indicate that SPARC may represent a new class of scavenger chaperones, which mediate ECM degradation, remodeling and repair by disassembling ECM networks and shuttling ECM proteins into the cell. Further understanding of this mechanism may provide insight into the pathogenesis of matrix-associated disorders and lead to the novel treatment strategies.


Subject(s)
Extracellular Matrix Proteins/metabolism , Extracellular Matrix/metabolism , Molecular Chaperones/metabolism , Osteonectin/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Collagen/metabolism , Collagen/pharmacokinetics , Extracellular Space/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , HEK293 Cells , Humans , Intracellular Space/metabolism , Male , Mice , Mice, 129 Strain , Mice, Knockout , Microscopy, Fluorescence , NIH 3T3 Cells , Osteonectin/genetics , Osteonectin/pharmacokinetics , Protein Transport
3.
J Biol Chem ; 285(44): 34072-85, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-20732874

ABSTRACT

The basolateral Na(+)-K(+)-2Cl(-) cotransporter (NKCC1) is a key determinant of transepithelial chloride secretion and dysregulation of chloride secretion is a common feature of many diseases including secretory diarrhea. We have previously shown that activation of protein kinase C (PKC) markedly reduces transepithelial chloride secretion in human colonic T84 cells, which correlates with both functional inhibition and loss of the NKCC1 surface expression. In the present study, we defined the specific roles of PKC isoforms in regulating epithelial NKCC1 and chloride secretion utilizing adenoviral vectors that express shRNAs targeting human PKC isoforms (α, δ, ε) (shPKCs) or LacZ (shLacZ, non-targeting control). After 72 h of adenoviral transduction, protein levels of the PKC isoforms in shPKCs-T84 cells were decreased by ∼90% compared with the shLacZ-control. Activation of PKCs by phorbol 12-myristate 13-acetate (PMA) caused a redistribution of NKCC1 immunostaining from the basolateral membrane to intracellular vesicles in both shLacZ- and shPKCα-T84 cells, whereas the effect of PMA was not observed in shPKCδ- and shPKCε- cells. These results were further confirmed by basolateral surface biotinylation. Furthermore, activation of PKCs by PMA inhibited cAMP-stimulated chloride secretion in the uninfected, shLacZ- and shPKCα-T84 monolayers, but the inhibitory effect was significantly attenuated in shPKCδ- and shPKCε-T84 monolayers. In conclusion, the activated novel isoforms PKCδ or PKCε, but not the conventional isoform PKCα, inhibits transepithelial chloride secretion through inducing internalization of the basolateral surface NKCC1. Our study reveals that the novel PKC isoform-regulated NKCC1 surface expression plays an important role in the regulation of chloride secretion.


Subject(s)
Chlorides/chemistry , Cyclic AMP/metabolism , Gene Expression Regulation, Enzymologic , Protein Kinase C-delta/metabolism , Protein Kinase C-epsilon/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Adenoviridae/metabolism , Biotinylation , Endocytosis , Humans , Ions/chemistry , Protein Isoforms , Protein Transport , RNA Interference , Solute Carrier Family 12, Member 2 , Tetradecanoylphorbol Acetate/chemistry
4.
Am J Physiol Cell Physiol ; 298(1): C85-97, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19864322

ABSTRACT

In secretory epithelial cells, the basolateral Na(+)-K(+)-2Cl(-) cotransporter (NKCC1) plays a major role in salt and fluid secretion. Our laboratory has identified NKCC1 surface expression as an important regulatory mechanism for Cl(-) secretion in the colonic crypt cell line T84, a process also present in native human colonic crypts. We previously showed that activation of protein kinase C (PKC) by carbachol and phorbol 12-myristate 13-acetate (PMA) decreases NKCC1 surface expression in T84 cells. However, the specific endocytic entry pathway has not been defined. We used a Madin-Darby canine kidney (MDCK) cell line stably transfected with enhanced green fluorescent protein (EGFP)-NKCC1 to map NKCC1 entry during PMA exposure. At given times, we fixed and stained the cells with specific markers (e.g., dynamin II, clathrin heavy chain, and caveolin-1). We also used chlorpromazine, methyl-beta-cyclodextrin, amiloride, and dynasore, blockers of the clathrin, caveolin, and macropinocytosis pathways and the vesicle "pinchase" dynamin, respectively. We found that PMA caused dose- and time-dependent NKCC1 endocytosis. After 2.5 min of PMA exposure, approximately 80% of EGFP-NKCC1 endocytic vesicles colocalized with clathrin and approximately 40% colocalized with dynamin II and with the transferrin receptor, the uptake of which is also mediated by clathrin-coated vesicles. We did not observe significant colocalization of EGFP-NKCC1 endocytic vesicles with caveolin-1, a marker of the caveolae-mediated endocytic pathway. We quantified the effect of each inhibitor on PMA-induced EGFP-NKCC1 endocytosis and found that only chlorpromazine and dynasore caused significant inhibition compared with the untreated control (61% and 25%, respectively, at 2.5 min). Together, these results strongly support the conclusion that PMA-stimulated NKCC1 endocytosis is associated with a clathrin pathway.


Subject(s)
Clathrin/physiology , Endocytosis/drug effects , Sodium-Potassium-Chloride Symporters/physiology , Tetradecanoylphorbol Acetate/pharmacology , Animals , Cell Line , Dogs , Glucose/pharmacology , Immunohistochemistry , Kidney/cytology , Kidney/drug effects , Kidney/physiology , Microscopy, Fluorescence , Solute Carrier Family 12, Member 2
5.
Physiol Genomics ; 34(3): 265-76, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18577713

ABSTRACT

The reported sequences of the human and mouse Na+-driven Cl-/HCO3(-) exchangers (NDCBEs) differ greatly in their extreme cytosolic COOH termini (Ct). In human NDCBE (NDCBE-B), a 17-amino acid (aa) sequence replaces 66 aa at the equivalent position in mouse NDCBE (NDCBE-A). We performed 5'- and 3'-rapid amplification of cDNA ends (RACE) on human brain cDNA, followed by PCR of full-length cDNAs to determine whether the human SLC4A8 gene was capable of producing the mouselike Ct sequence. Our study confirmed the presence in human cDNA of mouse NDCBE-like transcripts (human NDCBE-A) and also disclosed the existence of three further novel NDCBE transcripts that we have called NDCBE-C, NDCBE-D, and NDCBE-D'. The novel NDCBE-C/D/D' transcripts initiate at a novel "exon 0" positioned approximately 35 kb upstream of the first exon of NDCBE-A/B. NDCBE-C/D/D' protein products are predicted to be truncated by 54 aa in the cytosolic NH(2) terminus (Nt) compared with NDCBE-A/B. Our data, combined with a new in silico analysis of partial transcripts reported by others in the region of the human SLC4A8 gene, increase the known extent of the SLC4A8 gene by 49 kb, to 124 kb. A functional comparison of NDCBE-A/B/C/D expressed in Xenopus oocytes demonstrates that the Nt variation does not affect the basal functional expression of NDCBE, but those with the shorter Ct have a 25-50% reduced functional expression compared with those with the longer Ct. By comparison with an artificially truncated NDCBE that contains neither 17-aa nor 66-aa Ct cassette, we determined that the functional difference is unrelated to the 66-aa cassette of NDCBE-A/C, but is instead due to an inhibitory effect of the 17-aa cassette of NDCBE-B/D.


Subject(s)
Chloride-Bicarbonate Antiporters/genetics , Sodium-Bicarbonate Symporters/genetics , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Alternative Splicing/drug effects , Amino Acid Sequence , Animals , Base Sequence , Biological Transport/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Chloride-Bicarbonate Antiporters/chemistry , Chloride-Bicarbonate Antiporters/metabolism , Chromosomes, Human , Cloning, Molecular , Exons/genetics , Humans , Hydrogen-Ion Concentration , Membrane Potentials/drug effects , Molecular Sequence Data , Mutant Proteins/metabolism , Nucleic Acid Amplification Techniques , Oocytes/cytology , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sodium/metabolism , Sodium-Bicarbonate Symporters/chemistry , Sodium-Bicarbonate Symporters/metabolism , Solubility/drug effects , Xenopus
6.
Am J Physiol Renal Physiol ; 293(1): F110-20, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17356125

ABSTRACT

The proximal tubule (PT) is major site for the reabsorption of filtered HCO(3)(-). Previous work on the rabbit PT showed that 1) increases in basolateral (BL) CO(2) concentration ([CO(2)](BL)) raise the HCO(3)(-) reabsorption rate (J(HCO(3))), and 2) the increase that luminal angiotensin II (ANG II) produces in J(HCO(3)) is greatest at 0% [CO(2)](BL) and falls to nearly zero at 20%. Here, we investigate the role of angiotensin receptors in the [CO(2)](BL) dependence of J(HCO(3)) in isolated perfused PTs. We found that, in rabbit S2 PT segments, luminal 10(-8) M saralasin (peptide antagonist of ANG II receptors), lowers baseline J(HCO(3)) (5% CO(2)) to the value normally seen at 0% in the absence of inhibitors and eliminates the J(HCO(3)) response to changes in [CO(2)](BL). However, basolateral 10(-8) M saralasin has no effect. As with saralasin, luminal 10(-8) M candesartan (AT(1) antagonist) reduces baseline J(HCO(3)) and eliminates the [CO(2)](BL) dependence of J(HCO(3)). Luminal 10(-7) M PD 123319 (AT(2) antagonist) has no effect. Finally, we compared PTs from wild-type and AT(1A)-null mice of the same genetic background. Knocking out AT(1A) modestly lowers baseline J(HCO(3)) and, like luminal saralasin or candesartan in rabbits, eliminates the J(HCO(3)) response to changes in [CO(2)](BL). Our accumulated evidence suggests that ANG II endogenous to the PT binds to the apical AT(1A) receptor and that this interaction is critical for both baseline J(HCO(3)) and its response to changes in [CO(2)](BL). Neither apical AT(2) receptors nor basolateral ANG II receptors are involved in these processes.


Subject(s)
Bicarbonates/metabolism , Carbon Dioxide/pharmacology , Kidney Tubules, Proximal/metabolism , Receptor, Angiotensin, Type 1/physiology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds , Data Interpretation, Statistical , Female , Hydrogen-Ion Concentration , In Vitro Techniques , Kidney Tubules, Proximal/drug effects , Mice , Mice, Knockout , Perfusion , Rabbits , Receptor, Angiotensin, Type 1/genetics , Saralasin/pharmacology , Signal Transduction/drug effects , Tetrazoles/pharmacology
7.
Endocrinology ; 148(2): 831-40, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17068143

ABSTRACT

Colony-stimulating factor-1 (CSF-1) promotes the survival of osteoclasts, short-lived cells that resorb bone. Although a rise in intracellular pH (pH(i)) has been linked to inhibition of apoptosis, the effect of CSF-1 on pH(i) in osteoclasts has not been reported. The present study shows that, in the absence of CO(2)/HCO(3)(-), CSF-1 causes little change in osteoclast pH(i). In contrast, exposing these cells to CSF-1 in the presence of CO(2)/HCO(3)(-) causes a rapid and sustained cellular alkalinization. The CSF-1-induced rise in pH(i) is not blocked by 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid, an inhibitor of HCO(3)(-) transporters but is abolished by removing extracellular sodium. This inhibition profile is similar to that of the electroneutral Na/HCO(3) cotransporter NBCn1. By RT-PCR, NBCn1 transcripts are present in both osteoclasts and osteoclast-like cells (OCLs), and by immunoblotting, the protein is present in OCLs. Moreover, CSF-1 promotes osteoclast survival in the presence of CO(2)/HCO(3)(-) buffer but not in its absence. Preventing the activation of NBCn1 markedly attenuates the ability of CSF-1 to 1) block activation of caspase-8 and 2) prolong osteoclast survival. Inhibiting caspase-3 or caspase-8 in OCLs prolongs osteoclast survival to the same extent as does CSF-1. This study provides the first evidence that osteoclasts express a CSF-1-regulated Na/HCO(3) cotransporter, which may play a role in cell survival.


Subject(s)
Intracellular Fluid/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Osteoclasts/physiology , Protons , Sodium-Bicarbonate Symporters/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Alkalies/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Bicarbonates/pharmacology , Carbon Dioxide/pharmacology , Caspase Inhibitors , Cell Separation , Cell Survival/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Hydrogen-Ion Concentration , Macrolides/pharmacology , Mice , Mice, Inbred Strains , Osteoclasts/metabolism , Rats , Sodium/pharmacology
8.
Am J Physiol Renal Physiol ; 291(2): F358-67, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16705143

ABSTRACT

A previous study demonstrated that proximal tubule cells regulate HCO(3)(-) reabsorption by sensing acute changes in basolateral CO(2) concentration, suggesting that there is some sort of CO(2) sensor at or near the basolateral membrane (Zhou Y, Zhao J, Bouyer P, and Boron WF Proc Natl Acad Sci USA 102: 3875-3880, 2005). Here, we hypothesized that an early element in the CO(2) signal-transduction cascade might be either a receptor tyrosine kinase (RTK) or a receptor-associated (or soluble) tyrosine kinase (sTK). In our experiments, we found, first, that basolateral 17.5 microM genistein, a broad-spectrum tyrosine kinase inhibitor, virtually eliminates the CO(2) sensitivity of HCO(3)(-) absorption rate (J(HCO(3))). Second, we found that neither basolateral 250 nM nor basolateral 2 microM PP2, a high-affinity inhibitor for the Src family that also inhibits the Bcr-Abl sTK as well as the Kit RTK, reduces the CO(2)-stimulated increase in J(HCO(3)). Third, we found that either basolateral 35 nM PD168393, a high-affinity inhibitor of RTKs in the erbB (i.e., EGF receptor) family, or basolateral 10 nM BPIQ-I, which blocks erbB RTKs by competing with ATP, eliminates the CO(2) sensitivity. In conclusion, the transduction of the CO(2) signal requires activation of a tyrosine kinase, perhaps an erbB. The possibilities include the following: 1) a TK is simply permissive for the effect of CO(2) on J(HCO(3)); 2) a CO(2) receptor activates an sTK, which would then raise J(HCO(3)); 3) a CO(2) receptor transactivates an RTK; and 4) the CO(2) receptor could itself be an RTK.


Subject(s)
Bicarbonates/metabolism , Carbon Dioxide/pharmacology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Protein-Tyrosine Kinases/physiology , Absorption/physiology , Animals , Dimethyl Sulfoxide/pharmacology , Enzyme Inhibitors/pharmacology , Female , Genistein/pharmacology , Imidazoles/pharmacology , Kidney Tubules, Proximal/chemistry , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Quinazolines/pharmacology , Rabbits , Receptor Protein-Tyrosine Kinases/analysis , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Cell Surface/analysis , Receptors, Cell Surface/physiology , Signal Transduction/physiology
9.
Am J Physiol Renal Physiol ; 290(3): F666-73, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16204407

ABSTRACT

Previous authors showed that, at low doses, both basolateral and luminal ANG II increase the proximal tubule's HCO(3)(-) reabsorption rate (J(HCO(3))). Using out-of-equilibrium CO(2)/HCO(3)(-) solutions, we demonstrated that basolateral CO(2) increases J(HCO(3)). Here, we examine interactions between ANG II and CO(2) in isolated, perfused rabbit S2 segments. We first used equilibrated 5% CO(2)/22 mM HCO(3)(-)/pH 7.40 in bath and lumen. At 10(-11) M, basolateral (BL) ANG II increased J(HCO(3)) by 41%, and luminal ANG II increased J(HCO(3)) by 35%. At 10(-9) M, basolateral ANG II decreased J(HCO(3)) by 43%, whereas luminal ANG II was without effect. Second, we varied [CO(2)](BL) from 0 to 20% at fixed [HCO(3)(-)](BL) and pH(BL). Fractional stimulation produced by BL 10(-11) M ANG II falls when [CO(2)](BL) exceeds 5%. Fractional inhibition produced by BL 10(-9) M ANG II tends to rise when [CO(2)](BL) exceeds 5%. Regarding luminal ANG II, fractional stimulation produced by 10(-11) M ANG II fell monotonically as [CO(2)](BL) rose from 0 to 20%. Fractional inhibition produced by 10(-9) M ANG II rose monotonically with increasing [CO(2)](BL). Viewed differently, ANG II at 10(-11) M tended to reduce stimulation by CO(2), and at 10(-9) M, produced an even greater reduction. In conclusion, the mutual effects of 1) ANG II on the J(HCO(3)) response to basolateral CO(2) and 2) basolateral CO(2) on the J(HCO(3)) responses to ANG II suggest that the signal-transduction pathways for ANG II and basolateral CO(2) intersect or merge.


Subject(s)
Absorption/physiology , Angiotensin II/pharmacology , Bicarbonates/metabolism , Carbon Dioxide/metabolism , Kidney Tubules, Proximal/physiology , Absorption/drug effects , Animals , Female , Kidney Tubules, Proximal/drug effects , Kinetics , Rabbits
10.
Proc Natl Acad Sci U S A ; 102(10): 3875-80, 2005 Mar 08.
Article in English | MEDLINE | ID: mdl-15728388

ABSTRACT

Respiratory acidosis, a decrease in blood pH caused by a rise in [CO(2)], rapidly triggers a compensatory response in which the kidney markedly increases its secretion of H(+) from blood to urine. However, in this and other acid-base disturbances, the equilibrium CO(2) + H(2)O HCO(3)(-) + H(+) makes it impossible to determine whether the critical parameter is [CO(2)], [HCO(3)(-)], and/or pH. Here, we used out-of-equilibrium CO(2)/HCO(3)(-) solutions to alter basolateral (BL) [HCO(3)(-)], [CO(2)], or pH, systematically and one at a time, on isolated perfused S2 rabbit proximal tubules. We found that increasing [HCO(3)(-)](BL) from 0 to 44 mM, at a fixed [CO(2)](BL) of 5% and a fixed pH(BL) of 7.40, caused HCO(3)(-) reabsorption (J(HCO(3))) to fall by half but did not significantly affect volume reabsorption (J(V)). Increasing [CO(2)](BL) from 0% to 20%, at a fixed [HCO(3)(-)](BL) of 22 mM and pH(BL) of 7.40, caused J(HCO(3)) to rise 2.5-fold but did not significantly affect J(V). Finally, increasing pH(BL) from 6.80 to 8.00, at a fixed [HCO(3)(-)](BL) of 22 mM and [CO(2)](BL) of 5%, did not affect either J(HCO(3)) or J(V). Analysis of the J(HCO(3)) and J(V) data implies that, as the tubule alters J(HCO(3)), it compensates the reabsorption of other solutes to keep J(V) approximately constant. Because the cells cannot respond acutely to pH changes, we propose that the responses of J(HCO(3)) and the reabsorption of other solutes to changes in [HCO(3)(-)](BL) or [CO(2)](BL) involve sensors for basolateral HCO(3)(-) and CO(2).


Subject(s)
Bicarbonates/metabolism , Carbon Dioxide/metabolism , Kidney Tubules, Proximal/metabolism , Absorption , Animals , Female , Hydrogen-Ion Concentration , Rabbits
11.
J Physiol ; 559(Pt 1): 85-101, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15194736

ABSTRACT

Previous reports suggest that an important characteristic of chemosensitive neurones is an unusually large change of steady-state intracellular pH in response to a change in extracellular pH (DeltapH(i)/DeltapH(o)). To determine whether such a correlation exists between neurones from the medullary raphe (a chemosensitive brain region) and hippocampus (a non-chemosensitive region), we used BCECF to monitor pH(i) in cultured neurones subjected to extracellular acid-base disturbances. In medullary raphe neurones, respiratory acidosis (5%--> 9% CO(2)) caused a rapid fall in pH(i) (DeltapH(i) approximately 0.2) with no recovery and a large DeltapH(i)/DeltapH(o) of 0.71. Hippocampal neurones had a similar response, but with a slightly lower DeltapH(i)/DeltapH(o) (0.59). We further investigated a possible link between pH(i) regulation and chemosensitivity by following the pH(i) measurements on medullary raphe neurones with an immunocytochemistry for tryptophan hydroxylase (a marker of serotonergic neurones). We found that the DeltapH(i)/DeltapH(o) of 0.69 for serotonergic neurones (which are stimulated by acidosis) was not different from either the DeltapH(i)/DeltapH(o) of 0.75 for non-serotonergic neurones (most of which are not chemosensitive), or from the DeltapH(i)/DeltapH(o) of hippocampal neurones. For both respiratory alkalosis (5%--> 3% CO(2)) and metabolic alkalosis (22 mm--> 35 mm HCO(3)(-)), DeltapH(i)/DeltapH(o) was 0.42-0.53 for all groups of neurones studied. The only notable difference between medullary raphe and hippocampal neurones was in response to metabolic acidosis (22 mm--> 14 mm HCO(3)(-)), which caused a large pH(i) decrease in approximately 80% of medullary raphe neurones (DeltapH(i)/DeltapH(o)= 0.71), but relatively little pH(i) decrease in 70% of the hippocampal neurones (DeltapH(i)/DeltapH(o)= 0.09). Our comparison of medullary raphe and hippocampal neurones indicates that, except in response to metabolic acidosis, the neurones from the chemosensitive region do not have a uniquely high DeltapH(i)/DeltapH(o). Moreover, regardless of whether neurones were cultured from the chemosensitive or the non-chemosensitive region, pH(i) did not recover during any of the acid-base stresses.


Subject(s)
Extracellular Fluid/physiology , Hippocampus/physiology , Intracellular Fluid/physiology , Medulla Oblongata/physiology , Neurons/physiology , Raphe Nuclei/physiology , Acidosis, Respiratory/chemically induced , Acidosis, Respiratory/physiopathology , Alkalosis, Respiratory/chemically induced , Alkalosis, Respiratory/physiopathology , Animals , Bicarbonates/pharmacology , Carbon Dioxide/pharmacology , Cells, Cultured , Extracellular Fluid/drug effects , Hippocampus/drug effects , Hydrogen-Ion Concentration , Intracellular Fluid/drug effects , Medulla Oblongata/drug effects , Neurons/drug effects , Raphe Nuclei/drug effects , Rats , Rats, Sprague-Dawley
12.
Am J Physiol Renal Physiol ; 285(4): F674-87, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12812914

ABSTRACT

Working with isolated perfused S2 proximal tubules, we asked whether the basolateral CO2 sensor acts, in part, by raising intracellular Ca2+ concentration ([Ca2+]i), monitored with the dye fura 2 (or fura-PE3). In paired experiments, adding 5% CO2/22 mM HCO3- (constant pH 7.40) to the bath (basolateral) solution caused [Ca2+]i to increase from 57 +/- 3 to 97 +/- 9 nM(n = 8, P < 0.002), whereas the same maneuver in the lumen had no effect. Intracellular pH (pHi), measured with the dye BCECF, fell by 0.54 +/- 0.08 (n = 14) when we added CO2/HCO3- to the lumen. In 14 tubules in which we added CO2/HCO3- to the bath, pHi fell by 0.55 +/- 0.11 in 9 with a high initial pHi, but rose by 0.28 +/- 0.07 in the other 5 with a low initial pHi. Thus it cannot be a pHi change that triggers the [Ca2+]i increase. Introducing to the bath an out-of-equilibrium (OOE) solution containing 20% CO2/no HCO3-/pH 7.40 caused [Ca2+]i to rise by 62 +/- 17 nM (n = 10), whereas an OOE solution containing 0% CO2/22 mM HCO3-/pH 7.40 caused only a trivial increase. Removing Ca2+ from the lumen and bath, or adding 10 microM nifedipine (L- and T-type Ca2+-channel blocker) or 2 microM thapsigargin [sarco-(endo) plasmic reticulum Ca2+-ATPase inhibitor] or 4 microM rotenone (mitochondrial inhibitor) to the lumen and bath, failed to reduce the CO2-induced increase in [Ca2+]i. Adding 10 mM caffeine (ryanodine-receptor agonist) had no effect on [Ca2+]i. Thus basolateral CO2, presumably via a basolateral sensor, triggers the release of Ca2+ from a nonconventional intracellular pool.


Subject(s)
Calcium/metabolism , Carbon Dioxide/pharmacology , Intracellular Membranes/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Animals , Bicarbonates/pharmacology , Caffeine/pharmacology , Calcium Channel Blockers/pharmacology , Enzyme Inhibitors/pharmacology , Female , Hydrogen-Ion Concentration , In Vitro Techniques , Nifedipine/pharmacology , Osmolar Concentration , Rabbits , Rotenone/pharmacology , Thapsigargin/pharmacology
13.
J Physiol ; 542(Pt 1): 17-29, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12096045

ABSTRACT

For almost a century it was generally assumed that the lipid phases of all biological membranes are freely permeable to gases. However, recent observations challenge this dogma. The apical membranes of epithelial cells exposed to hostile environments, such as gastric glands, have no demonstrable permeability to the gases CO2 and NH3. Additionally, the water channel protein aquaporin 1 (AQP1), expressed at high levels in erythrocytes, can increase membrane CO2 permeability when expressed in Xenopus oocytes. Similarly, nodulin-26, which is closely related to AQP1, can act as a conduit for NH3. A key question is whether aquaporins, which are abundant in virtually every tissue that transports O2 and CO2 at high levels, ever play a physiologically significant role in the transport of small volatile molecules. Preliminary data are consistent with the hypothesis that AQP1 enhances the reabsorption of HCO3- by the renal proximal tubule by increasing the CO2 permeability of the apical membrane. Other preliminary data on Xenopus oocytes heterologously expressing the electrogenic Na+-HCO3- cotransporter (NBC), AQP1 and carbonic anhydrases are consistent with the hypothesis that the macroscopic cotransport of Na+ plus two HCO3- occurs as NBC transports Na+ plus CO3(2-) and AQP1 transports CO2 and H2O. Although data - obtained on AQP1 reconstituted into liposomes or on materials from AQP1 knockout mice - appear inconsistent with the model that AQP1 mediates substantial CO2 transport in certain preparations, the existence of unstirred layers or perfusion-limited conditions may have masked the contribution of AQP1 to CO2 permeability.


Subject(s)
Aquaporins/metabolism , Gases/metabolism , Animals , Aquaporin 1 , Aquaporins/chemistry , Biological Transport/physiology , Blood Group Antigens , Carbon Dioxide/metabolism , Humans , Permeability , Water/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...