Journal articles on the topic 'V-ATPase proton pump'

To see the other types of publications on this topic, follow the link: V-ATPase proton pump.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'V-ATPase proton pump.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Nolta, K. V., H. Padh, and T. L. Steck. "An immunocytochemical analysis of the vacuolar proton pump in Dictyostelium discoideum." Journal of Cell Science 105, no. 3 (July 1, 1993): 849–59. http://dx.doi.org/10.1242/jcs.105.3.849.

Full text
Abstract:
Antisera were generated in rabbits against the vacuolar proton pump (V-H(+)-ATPase) purified from Dictyostelium discoideum. The antisera inhibited V-H(+)-ATPase but not F1-ATPase activity and immunoprecipitated and immunoblotted only the polypeptide subunits of the V-H(+)-ATPase from cell homogenates. Immunocytochemical analysis of intact cells and subcellular fractions showed that the predominant immunoreactive organelles were clusters of empty, irregular vacuoles of various sizes and shapes, which corresponded to the acidosomes. The cytoplasmic surfaces of lysosomes, phagosomes and the tubular spongiome of the contractile vacuole also bore the pump antigen. The lumina of multivesicular bodies were often stained intensely; the internalized antigen may have been derived from acidosomes by autophagy. Antibodies against V-H(+)-ATPases from plant and animal cells cross-reacted with the proton pumps of Dictyostelium. Antisera directed against the V-H(+)-ATPase of Dictyostelium decorated a profusion of small vacuoles scattered throughout the cytoplasm of hepatocytes, epithelial cells, macrophages and fibroblasts. The pattern paralleled that of the endocytic and acidic spaces; there was no clear indication of discrete acidosomes in these mammalian cells. We conclude that the V-H(+)-ATPase in Dictyostelium is distributed among diverse endomembrane organelles and is immunologically cross-reactive with the proton pumps on endocytic vacuoles in mammalian cells.
APA, Harvard, Vancouver, ISO, and other styles
2

Drory, Omri, and Nathan Nelson. "The Emerging Structure of Vacuolar ATPases." Physiology 21, no. 5 (October 2006): 317–25. http://dx.doi.org/10.1152/physiol.00017.2006.

Full text
Abstract:
Bioenergetics and physiology of primary pumps have been revitalized by new insights into the mechanism of energizing biomembranes. Structural information is becoming available, and the three-dimensional structure of F-ATPase is being resolved. The growing understanding of the fundamental mechanism of energy coupling may revolutionize our view of biological processes. The F- and V-ATPases (vacuolar-type ATPase) exhibit a common mechanical design in which nucleotide-binding on the catalytic sector, through a cycle of conformation changes, drives the transmembrane passage of protons by turning a membrane-embedded rotor. This motor can run in forward or reverse directions, hydrolyzing ATP as it pumps protons uphill or creating ATP as protons flow downhill. In contrast to F-ATPases, whose primary function in eukaryotic cells is to form ATP at the expense of the proton-motive force (pmf), V-ATPases function exclusively as an ATP-dependent proton pump. The pmf generated by V-ATPases in organelles and membranes of eukaryotic cells is utilized as a driving force for numerous secondary transport processes. V- and F-ATPases have similar structure and mechanism of action, and several of their subunits evolved from common ancestors. Electron microscopy studies of V-ATPase revealed its general structure at low resolution. Recently, several structures of V-ATPase subunits, solved by X-ray crystallography with atomic resolution, were published. This, together with electron microscopy low-resolution maps of the whole complex, and biochemistry cross-linking experiments, allows construction of a structural model for a part of the complex that may be used as a working hypothesis for future research.
APA, Harvard, Vancouver, ISO, and other styles
3

Kabała, Katarzyna, and Małgorzata Janicka. "Structural and Functional Diversity of Two ATP-Driven Plant Proton Pumps." International Journal of Molecular Sciences 24, no. 5 (February 24, 2023): 4512. http://dx.doi.org/10.3390/ijms24054512.

Full text
Abstract:
Two ATP-dependent proton pumps function in plant cells. Plasma membrane H+-ATPase (PM H+-ATPase) transfers protons from the cytoplasm to the apoplast, while vacuolar H+-ATPase (V-ATPase), located in tonoplasts and other endomembranes, is responsible for proton pumping into the organelle lumen. Both enzymes belong to two different families of proteins and, therefore, differ significantly in their structure and mechanism of action. The plasma membrane H+-ATPase is a member of the P-ATPases that undergo conformational changes, associated with two distinct E1 and E2 states, and autophosphorylation during the catalytic cycle. The vacuolar H+-ATPase represents rotary enzymes functioning as a molecular motor. The plant V-ATPase consists of thirteen different subunits organized into two subcomplexes, the peripheral V1 and the membrane-embedded V0, in which the stator and rotor parts have been distinguished. In contrast, the plant plasma membrane proton pump is a functional single polypeptide chain. However, when the enzyme is active, it transforms into a large twelve-protein complex of six H+-ATPase molecules and six 14-3-3 proteins. Despite these differences, both proton pumps can be regulated by the same mechanisms (such as reversible phosphorylation) and, in some processes, such as cytosolic pH regulation, may act in a coordinated way.
APA, Harvard, Vancouver, ISO, and other styles
4

Pérez-Sayáns, M., JM Suárez-Peñaranda, F. Barros-Angueira, PG Diz, JM Gándara-Rey, and A. García-García. "An update in the structure, function, and regulation of V-ATPases: the role of the C subunit." Brazilian Journal of Biology 72, no. 1 (February 2012): 189–98. http://dx.doi.org/10.1590/s1519-69842012000100023.

Full text
Abstract:
Vacuolar ATPases (V-ATPases) are present in specialized proton secretory cells in which they pump protons across the membranes of various intracellular organelles and across the plasma membrane. The proton transport mechanism is electrogenic and establishes an acidic pH and a positive transmembrane potential in these intracellular and extracellular compartments. V-ATPases have been found to be practically identical in terms of the composition of their subunits in all eukaryotic cells. They have two distinct structures: a peripheral catalytic sector (V1) and a hydrophobic membrane sector (V0) responsible for driving protons. V-ATPase activity is regulated by three different mechanisms, which control pump density, association/dissociation of the V1 and V0 domains, and secretory activity. The C subunit is a 40-kDa protein located in the V1 domain of V-ATPase. The protein is encoded by the ATP6V1C gene and is located at position 22 of the long arm of chromosome 8 (8q22.3). The C subunit has very important functions in terms of controlling the regulation of the reversible dissociation of V-ATPases.
APA, Harvard, Vancouver, ISO, and other styles
5

Holliday, L. Shannon. "Vacuolar H+-ATPase: An Essential Multitasking Enzyme in Physiology and Pathophysiology." New Journal of Science 2014 (January 23, 2014): 1–21. http://dx.doi.org/10.1155/2014/675430.

Full text
Abstract:
Vacuolar H+-ATPases (V-ATPases) are large multisubunit proton pumps that are required for housekeeping acidification of membrane-bound compartments in eukaryotic cells. Mammalian V-ATPases are composed of 13 different subunits. Their housekeeping functions include acidifying endosomes, lysosomes, phagosomes, compartments for uncoupling receptors and ligands, autophagosomes, and elements of the Golgi apparatus. Specialized cells, including osteoclasts, intercalated cells in the kidney and pancreatic beta cells, contain both the housekeeping V-ATPases and an additional subset of V-ATPases, which plays a cell type specific role. The specialized V-ATPases are typically marked by the inclusion of cell type specific isoforms of one or more of the subunits. Three human diseases caused by mutations of isoforms of subunits have been identified. Cancer cells utilize V-ATPases in unusual ways; characterization of V-ATPases may lead to new therapeutic modalities for the treatment of cancer. Two accessory proteins to the V-ATPase have been identified that regulate the proton pump. One is the (pro)renin receptor and data is emerging that indicates that V-ATPase may be intimately linked to renin/angiotensin signaling both systemically and locally. In summary, V-ATPases play vital housekeeping roles in eukaryotic cells. Specialized versions of the pump are required by specific organ systems and are involved in diseases.
APA, Harvard, Vancouver, ISO, and other styles
6

Schep, Daniel G., Jianhua Zhao, and John L. Rubinstein. "Models for the a subunits of the Thermus thermophilus V/A-ATPase and Saccharomyces cerevisiae V-ATPase enzymes by cryo-EM and evolutionary covariance." Proceedings of the National Academy of Sciences 113, no. 12 (March 7, 2016): 3245–50. http://dx.doi.org/10.1073/pnas.1521990113.

Full text
Abstract:
Rotary ATPases couple ATP synthesis or hydrolysis to proton translocation across a membrane. However, understanding proton translocation has been hampered by a lack of structural information for the membrane-embedded a subunit. The V/A-ATPase from the eubacterium Thermus thermophilus is similar in structure to the eukaryotic V-ATPase but has a simpler subunit composition and functions in vivo to synthesize ATP rather than pump protons. We determined the T. thermophilus V/A-ATPase structure by cryo-EM at 6.4 Å resolution. Evolutionary covariance analysis allowed tracing of the a subunit sequence within the map, providing a complete model of the rotary ATPase. Comparing the membrane-embedded regions of the T. thermophilus V/A-ATPase and eukaryotic V-ATPase from Saccharomyces cerevisiae allowed identification of the α-helices that belong to the a subunit and revealed the existence of previously unknown subunits in the eukaryotic enzyme. Subsequent evolutionary covariance analysis enabled construction of a model of the a subunit in the S. cerevisae V-ATPase that explains numerous biochemical studies of that enzyme. Comparing the two a subunit structures determined here with a structure of the distantly related a subunit from the bovine F-type ATP synthase revealed a conserved pattern of residues, suggesting a common mechanism for proton transport in all rotary ATPases.
APA, Harvard, Vancouver, ISO, and other styles
7

Taiz, L. "THE PLANT VACUOLE." Journal of Experimental Biology 172, no. 1 (November 1, 1992): 113–22. http://dx.doi.org/10.1242/jeb.172.1.113.

Full text
Abstract:
Plant cells are unique in containing large acidic vacuoles which occupy most of the cell volume. The vacuolar H+-ATPase (V-ATPase) is the enzyme responsible for acidifying the central vacuole, although it is also present on Golgi and coated vesicles. Many secondary transport processes are driven by the proton-motive force generated by the V-ATPase, including reactions required for osmoregulation, homeostasis, storage, plant defense and many other functions. However, a second proton pump, the V-PPase, serves as a potential back-up system and may, in addition, pump potassium. The plant V-ATPase is structurally similar to other eukaryotic V-ATPases and its subunits appear to be encoded by small multigene families. These multigene families may play important roles in the regulation of gene expression and in the sorting of V-ATPase isoforms to different organelles.
APA, Harvard, Vancouver, ISO, and other styles
8

Jansen, Eric J. R., Theo G. M. Hafmans, and Gerard J. M. Martens. "V-ATPase-Mediated Granular Acidification Is Regulated by the V-ATPase Accessory Subunit Ac45 in POMC-Producing Cells." Molecular Biology of the Cell 21, no. 19 (October 2010): 3330–39. http://dx.doi.org/10.1091/mbc.e10-04-0274.

Full text
Abstract:
The vacuolar (H+)-ATPase (V-ATPase) is an important proton pump, and multiple critical cell-biological processes depend on the proton gradient provided by the pump. Yet, the mechanism underlying the control of the V-ATPase is still elusive but has been hypothesized to involve an accessory subunit of the pump. Here we studied as a candidate V-ATPase regulator the neuroendocrine V-ATPase accessory subunit Ac45. We transgenically manipulated the expression levels of the Ac45 protein specifically in Xenopus intermediate pituitary melanotrope cells and analyzed in detail the functioning of the transgenic cells. We found in the transgenic melanotrope cells the following: i) significantly increased granular acidification; ii) reduced sensitivity for a V-ATPase-specific inhibitor; iii) enhanced early processing of proopiomelanocortin (POMC) by prohormone convertase PC1; iv) reduced, neutral pH–dependent cleavage of the PC2 chaperone 7B2; v) reduced 7B2-proPC2 dissociation and consequently reduced proPC2 maturation; vi) decreased levels of mature PC2 and consequently reduced late POMC processing. Together, our results show that the V-ATPase accessory subunit Ac45 represents the first regulator of the proton pump and controls V-ATPase-mediated granular acidification that is necessary for efficient prohormone processing.
APA, Harvard, Vancouver, ISO, and other styles
9

FINBOW, Malcolm E., and Michael A. HARRISON. "The vacuolar H+-ATPase: a universal proton pump of eukaryotes." Biochemical Journal 324, no. 3 (June 15, 1997): 697–712. http://dx.doi.org/10.1042/bj3240697.

Full text
Abstract:
The vacuolar H+-ATPase (V-ATPase) is a universal component of eukaryotic organisms. It is present in the membranes of many organelles, where its proton-pumping action creates the low intra-vacuolar pH found, for example, in lysosomes. In addition, there are a number of differentiated cell types that have V-ATPases on their surface that contribute to the physiological functions of these cells. The V-ATPase is a multi-subunit enzyme composed of a membrane sector and a cytosolic catalytic sector. It is related to the familiar FoF1 ATP synthase (F-ATPase), having the same basic architectural construction, and many of the subunits from the two display identity with one another. All the core subunits of the V-ATPase have now been identified and much is known about the assembly, regulation and pharmacology of the enzyme. Recent genetic analysis has shown the V-ATPase to be a vital component of higher eukaryotes. At least one of the subunits, i.e. subunit c (ductin), may have multifunctional roles in membrane transport, providing a possible pathway of communication between cells. The structure of the membrane sector is known in some detail, and it is possible to begin to suggest how proton pumping is coupled to ATP hydrolysis.
APA, Harvard, Vancouver, ISO, and other styles
10

Pérez-Castiñeira, José R., Agustín Hernández, Rocío Drake, and Aurelio Serrano. "A plant proton-pumping inorganic pyrophosphatase functionally complements the vacuolar ATPase transport activity and confers bafilomycin resistance in yeast." Biochemical Journal 437, no. 2 (June 28, 2011): 269–78. http://dx.doi.org/10.1042/bj20110447.

Full text
Abstract:
V-ATPases (vacuolar H+-ATPases) are a specific class of multi-subunit pumps that play an essential role in the generation of proton gradients across eukaryotic endomembranes. Another simpler proton pump that co-localizes with the V-ATPase occurs in plants and many protists: the single-subunit H+-PPase [H+-translocating PPase (inorganic pyrophosphatase)]. Little is known about the relative contribution of these two proteins to the acidification of intracellular compartments. In the present study, we show that the expression of a chimaeric derivative of the Arabidopsis thaliana H+-PPase AVP1, which is preferentially targeted to internal membranes of yeast, alleviates the phenotypes associated with V-ATPase deficiency. Phenotypic complementation was achieved both with a yeast strain with its V-ATPase specifically inhibited by bafilomycin A1 and with a vma1-null mutant lacking a catalytic V-ATPase subunit. Cell staining with vital fluorescent dyes showed that AVP1 recovered vacuole acidification and normalized the endocytic pathway of the vma mutant. Biochemical and immunochemical studies further demonstrated that a significant fraction of heterologous H+-PPase is located at the vacuolar membrane. These results raise the question of the occurrence of distinct proton pumps in certain single-membrane organelles, such as plant vacuoles, by proving yeast V-ATPase activity dispensability and the capability of H+-PPase to generate, by itself, physiologically suitable internal pH gradients. Also, they suggest new ways of engineering macrolide drug tolerance and outline an experimental system for testing alternative roles for fungal and animal V-ATPases, other than the mere acidification of subcellular organelles.
APA, Harvard, Vancouver, ISO, and other styles
11

Chatterjee, D., M. Chakraborty, M. Leit, L. Neff, S. Jamsa-Kellokumpu, R. Fuchs, M. Bartkiewicz, N. Hernando, and R. Baron. "The osteoclast proton pump differs in its pharmacology and catalytic subunits from other vacuolar H(+)-ATPases." Journal of Experimental Biology 172, no. 1 (November 1, 1992): 193–204. http://dx.doi.org/10.1242/jeb.172.1.193.

Full text
Abstract:
Osteoclasts are multinucleated cells derived from the mononuclear phagocyte system in the hematopoietic bone marrow. Their function is to resorb bone during skeletal growth and remodeling. They perform this function by acidifying an enclosed extracellular space, the bone resorbing compartment. Analysis of proton transport by inside-out vesicles derived from highly purified chicken osteoclast membranes has revealed the presence of a novel type of multisubunit vacuolar-like H(+)-ATPase. Unlike H(+)-ATPases derived from any other cell type or organelle, proton transport and ATPase activity in osteoclast vesicles are sensitive to two classes of inhibitors, namely V-ATPase inhibitors [N-ethyl-maleimide (NEM) and bafilomycin A1] and vanadate (IC50 100 mumol l-1), an inhibitor previously found to affect only P-ATPases. The osteoclast V-ATPase morphologically resembles vacuolar proton pumps and contains several vacuolar-like subunits (115 x 10(3), 39 x 10(3) and 16 x 10(3)M(r)), demonstrated by Western blot analysis. Subunits A and B of the catalytic domain of the enzyme, however, differ from that of other V-ATPases. In osteoclasts, subunit A has an M(r) of 63 x 10(3) instead of 67 x 10(3)-70 x 10(3); in contrast, monocytes, macrophages and kidney microsomes, which contain a vanadate-insensitive H(+)-ATPase, express the classical subunit A (70 x 10(3)M(r)). Moreover, two types of 57 x 10(3)-60 x 10(3)M(r) B subunits are also found: they are differentially recognized by antibodies and one is expressed predominantly in osteoclasts and the other in bone marrow cells and in kidney microsomes. Preliminary cloning data have indicated that the B subunit expressed in osteoclasts may be similar to the brain isoform. The osteoclast proton pump may, therefore, constitute a novel class of V-ATPase, with a unique pharmacology and specific isoforms of two subunits in the catalytic portion of the enzyme.
APA, Harvard, Vancouver, ISO, and other styles
12

Wieczorek, H. "The insect V-ATPase, a plasma membrane proton pump energizing secondary active transport: molecular analysis of electrogenic potassium transport in the tobacco hornworm midgut." Journal of Experimental Biology 172, no. 1 (November 1, 1992): 335–43. http://dx.doi.org/10.1242/jeb.172.1.335.

Full text
Abstract:
Goblet cell apical membranes in the larval midgut of Manduca sexta are the site of active and electrogenic K+ secretion. They possess a vacuolar-type ATPase which, in its immunopurified form, consists of at least nine polypeptides. cDNAs for the A and B subunits screened by monoclonal antibodies to the A subunit of the Manduca V-ATPase or by hybridisation with a cDNA probe for a plant V-ATPase B subunit have been cloned and sequenced. There is a high degree of identity to the sequences of the respective subunits of other V-ATPases. The M. sexta plasma membrane V-ATPase is an electrogenic proton pump which energizes, by the electrical component of the proton-motive force, electrogenic K+/nH+ antiport, resulting in net electrogenic K+ secretion. Since the midgut lacks a Na+/K(+)-ATPase, all solute fluxes in this epithelium seem to be energized by the V-ATPase. Thus, the midgut provides an alternative to the classical concept of animal plasma membrane energization by the Na(+)-motive force generated by the Na+/K(+)-ATPase.
APA, Harvard, Vancouver, ISO, and other styles
13

Sze, H., JM Ward, S. Lai, and I. Perera. "VACUOLAR-TYPE H+-TRANSLOCATING ATPases IN PLANT ENDOMEMBRANES: SUBUNIT ORGANIZATION AND MULTIGENE FAMILIES." Journal of Experimental Biology 172, no. 1 (November 1, 1992): 123–35. http://dx.doi.org/10.1242/jeb.172.1.123.

Full text
Abstract:
Acidification of endomembrane compartments by the vacuolar-type H+-translocating ATPase (V-ATPase) is vital to the growth and development of plants. The V-ATPase purified from oat roots is a large complex of 650x10(3 )Mr that contains 10 different subunits of 70, 60, 44, 42, 36, 32, 29, 16, 13 and 12x10(3 )Mr. This set of ten polypeptides is sufficient to couple ATP hydrolysis to proton pumping after reconstitution of the ATPase into liposomes. Unlike some animal V-ATPases, the purified and reconstituted V-ATPase from oat is directly stimulated by Cl-. The peripheral complex of the ATPase includes the nucleotide-binding subunits of 70 and 60x10(3 )Mr and polypeptides of 44, 42, 36 and 29x10(3 )Mr. Six copies of the 16x10(3 )Mr proteolipid together with three other polypeptides are thought to make up the integral sector that forms the H+-conducting pathway. Release of the peripheral complex from the native membrane completely inactivates the pump; however, the peripheral subunits can be reassembled with the membrane sector to form a functional H+ pump. Comparison of V-ATPases from several plants indicates considerable variations in subunit composition. Hence, several forms of the V-ATPase may exist among, and probably within, plant species. At least four distinct cDNAs encode the 16x10(3 )Mr proteolipid subunit in oat. Multiple genes could encode different subtypes of the H+ pump that are regulated by the developmental stage and physiological function specific to the cell or tissue type.
APA, Harvard, Vancouver, ISO, and other styles
14

Abe, Michiko, Mayu Saito, Ayana Tsukahara, Shuka Shiokawa, Kazuma Ueno, Hiroki Shimamura, Makoto Nagano, Junko Y. Toshima, and Jiro Toshima. "Functional complementation reveals that 9 of the 13 human V-ATPase subunits can functionally substitute for their yeast orthologs." Journal of Biological Chemistry 294, no. 20 (April 5, 2019): 8273–85. http://dx.doi.org/10.1074/jbc.ra118.006192.

Full text
Abstract:
Vacuolar-type H+-ATPase (V-ATPase) is a highly conserved proton pump responsible for acidification of intracellular organelles and potential drug target. It is a multisubunit complex comprising a cytoplasmic V1 domain responsible for ATP hydrolysis and a membrane-embedded Vo domain that contributes to proton translocation across the membrane. Saccharomyces cerevisiae V-ATPase is composed of 14 subunits, deletion of any one of which results in well-defined growth defects. As the structure of V-ATPase and the function of each subunit have been well-characterized in yeast, this organism has been recognized as a preferred model for studies of V-ATPases. In this study, to assess the functional relatedness of the yeast and human V-ATPase subunits, we investigated whether human V-ATPase subunits can complement calcium- or pH-sensitive growth, acidification of the vacuolar lumen, assembly of the V-ATPase complex, and protein sorting in yeast mutants lacking the equivalent yeast genes. These assessments revealed that 9 of the 13 human V-ATPase subunits can partially or fully complement the function of the corresponding yeast subunits. Importantly, sequence similarity was not necessarily correlated with functional complementation. We also found that besides all Vo domain subunits, the V1 F subunit is required for proper assembly of the Vo domain at the endoplasmic reticulum. Furthermore, the human H subunit fully restored the level of vacuolar acidification, but only partially rescued calcium-sensitive growth, suggesting a specific role of the H subunit in V-ATPase activity. These findings provide important insights into functional homologies between yeast and human V-ATPases.
APA, Harvard, Vancouver, ISO, and other styles
15

Sarkar, Juni, Xin Wen, Emil J. Simanian, and Michael L. Paine. "V-type ATPase proton pump expression during enamel formation." Matrix Biology 52-54 (May 2016): 234–45. http://dx.doi.org/10.1016/j.matbio.2015.11.004.

Full text
APA, Harvard, Vancouver, ISO, and other styles
16

Klein, U. "THE INSECT V-ATPase, A PLASMA MEMBRANE PROTON PUMP ENERGIZING SECONDARY ACTIVE TRANSPORT: IMMUNOLOGICAL EVIDENCE FOR THE OCCURRENCE OF A V-ATPase IN INSECT ION-TRANSPORTING EPITHELIA." Journal of Experimental Biology 172, no. 1 (November 1, 1992): 345–54. http://dx.doi.org/10.1242/jeb.172.1.345.

Full text
Abstract:
Active electrogenic K+ transport in insects serves as the energy source for secretion or absorption in gastrointestinal epithelia or for the receptor current in sensory epithelia. In the larval midgut of the tobacco hornworm Manduca sexta, a vacuolar-type proton pump (V-ATPase) and a K+/nH+ antiport represent the functional elements of the potassium pump. Several immunological findings support the hypothesis that active K+ transport in other insect epithelia may also be energized by a V-ATPase. In immunoblots, crude homogenates of sensilla-rich antennae and Malpighian tubules of M. sexta cross reacted with an immune serum directed to the purified plasma membrane V-ATPase from the midgut; the M. sexta midgut V-ATPase cross reacted with polyclonal antibodies to endomembrane V-ATPases from xenic origin. In immunocytochemical investigations of larvae of M. sexta and adults of Antheraea pernyi, monoclonal antibodies to defined subunits of the purified midgut V-ATPase or polyclonal antibodies to xenic endomembrane V-ATPase labelled the sites of active K+ transport: the goblet cell apical membrane in the midgut, the brush border of Malpighian tubules and the apical projections of the auxiliary cells in antennal sensilla. The functional mechanism of a primary H+-pumping V-ATPase and a secondary H+-dependent K+ transport postulated for K+-transporting insect epithelia may be further applicable to active Na+ or Cl- transport and would provide a unifying concept for all ouabain-insensitive electrogenic ion transport in insects. The findings from the midgut investigations, however, are the first instance in which a V-ATPase provides an alternative to the Na+/K+-ATPase in energizing secondary active transport in animal plasma membranes.
APA, Harvard, Vancouver, ISO, and other styles
17

Feng, Shengmei, Lianfu Deng, Wei Chen, Jianzhong Shao, Guoliang Xu, and Yi-Ping Li. "Atp6v1c1 is an essential component of the osteoclast proton pump and in F-actin ring formation in osteoclasts." Biochemical Journal 417, no. 1 (December 12, 2008): 195–203. http://dx.doi.org/10.1042/bj20081073.

Full text
Abstract:
Bone resorption relies on the extracellular acidification function of V-ATPase (vacuolar-type proton-translocating ATPase) proton pump(s) present in the plasma membrane of osteoclasts. The exact configuration of the osteoclast-specific ruffled border V-ATPases remains largely unknown. In the present study, we found that the V-ATPase subunit Atp6v1c1 (C1) is highly expressed in osteoclasts, whereas subunits Atp6v1c2a (C2a) and Atp6v1c2b (C2b) are not. The expression level of C1 is highly induced by RANKL [receptor activator for NF-κB (nuclear factor κB) ligand] during osteoclast differentiation; C1 interacts with Atp6v0a3 (a3) and is mainly localized on the ruffled border of activated osteoclasts. The results of the present study show for the first time that C1-silencing by lentivirus-mediated RNA interference severely impaired osteoclast acidification activity and bone resorption, whereas cell differentiation did not appear to be affected, which is similar to a3 silencing. The F-actin (filamentous actin) ring formation was severely defected in C1-depleted osteoclasts but not in a3-depleted and a3−/− osteoclasts. C1 co-localized with microtubules in the plasma membrane and its vicinity in mature osteoclasts. In addition, C1 co-localized with F-actin in the cytoplasm; however, the co-localization chiefly shifted to the cell periphery of mature osteoclasts. The present study demonstrates that Atp6v1c1 is an essential component of the osteoclast proton pump at the osteoclast ruffled border and that it may regulate F-actin ring formation in osteoclast activation.
APA, Harvard, Vancouver, ISO, and other styles
18

Liu, Tongyao, Christian Mirschberger, Lilian Chooback, Quyen Arana, Zeno Dal Sacco, Harry MacWilliams, and Margaret Clarke. "Altered expression of the 100 kDa subunit of the Dictyosteliumvacuolar proton pump impairs enzyme assembly, endocytic function and cytosolic pH regulation." Journal of Cell Science 115, no. 9 (May 1, 2002): 1907–18. http://dx.doi.org/10.1242/jcs.115.9.1907.

Full text
Abstract:
The vacuolar proton pump (V-ATPase) appears to be essential for viability of Dictyostelium cells. To investigate the function of VatM, the 100 kDa transmembrane V-ATPase subunit, we altered its level. By means of homologous recombination, the promoter for the chromosomal vatM gene was replaced with the promoter for the act6 gene, yielding the mutant strain VatMpr. The act6 promoter is much more active in cells growing axenically than on bacteria. Thus, transformants were selected under axenic growth conditions, then shifted to bacteria to determine the consequences of reduced vatM expression. When VatMpr cells were grown on bacteria,the level of the 100 kDa V-ATPase subunit dropped, cell growth slowed, and the A subunit, a component of the peripheral catalytic domain of the V-ATPase,became mislocalized. These defects were complemented by transformation of the mutant cells with a plasmid expressing vatM under the control of its own promoter. Although the principal locus of vacuolar proton pumps in Dictyostelium is membranes of the contractile vacuole system, mutant cells did not manifest osmoregulatory defects. However, bacterially grown VatMpr cells did exhibit substantially reduced rates of phagocytosis and a prolonged endosomal transit time. In addition, mutant cells manifested alterations in the dynamic regulation of cytosolic pH that are characteristic of normal cells grown in acid media, which suggested that the V-ATPase also plays a role in cytosolic pH regulation.
APA, Harvard, Vancouver, ISO, and other styles
19

Ehrenfeld, J., and U. Klein. "The key role of the H+ V-ATPase in acid-base balance and Na+ transport processes in frog skin." Journal of Experimental Biology 200, no. 2 (January 1, 1997): 247–56. http://dx.doi.org/10.1242/jeb.200.2.247.

Full text
Abstract:
Frogs are faced with various osmoregulatory problems, such as compensation of salt and water loss or metabolic acidification. Being exposed both to air and to pond water of low salinity in their natural habitat, the epithelium of the frog skin serves as one of the major organs for body fluid homeostasis. For years, the frog skin has been the guiding model for ion transport processes in animal cells energized by a Na(+)-motive force. Meanwhile, however, it was demonstrated that under natural conditions Na+ uptake is electrically coupled to active H+ secretion, mediated by an electrogenic H+ pump. A proton-motive force generated at the apical membrane of the mitochondria-rich cells (MR cells) energizes Na+ entry via apical Na+ channels. The basolateral Na+/K+ P-ATPase then pumps Na+ out of the cell into the body fluid. Thus, there are two pumps functioning in series, both involved in transepithelial Na+ transport. Our recent investigations provided conclusive evidence that the H+ pump of the frog skin is an H+ V-ATPase. In transport studies, Na+ absorption and H+ secretion were blocked by micromolar concentrations of bafilomycin A1 or concanamycin A, two highly specific inhibitors of H(+)-V-ATPases. Using immunofluorescence microscopy, H(+)-V-ATPase-like immunoreactivity was found in MR cells in the region of their apical membrane foldings and intracellularly in the apical portion of the cell at so far unidentified locations. Besides the definition of its molecular nature, these results also confirmed the localization of the H+ pump in the apical membrane of the MR cells. These cells were already candidates for H(+)-V-ATPase localization mostly from correlations between their morphological features and their epithelial H+ secretion capacity. So far, there is evidence for only one type of MR cell serving both H+ and HCO3- secretion through an apical Cl-/HCO3- antiporter. H(+)-V-ATPase-mediated H+ secretion and thus Na+ absorption can be modulated by complementary mechanisms. Changes in intracellular H+ concentration linked to the animal's acid-base status will directly influence H+ V-ATPase activity. Acute acidification increases H+ current, probably as a result of the insertion of H(+)-V-ATPase-bearing vesicles by exocytotic processes, while alkalization causes the reverse effects. Chronic metabolic acidosis induces an increase in MR cell number in response to hormonal signals.
APA, Harvard, Vancouver, ISO, and other styles
20

Peters, Lee Zeev, Rotem Hazan, Michal Breker, Maya Schuldiner, and Shay Ben-Aroya. "Formation and dissociation of proteasome storage granules are regulated by cytosolic pH." Journal of Cell Biology 201, no. 5 (May 20, 2013): 663–71. http://dx.doi.org/10.1083/jcb.201211146.

Full text
Abstract:
The 26S proteasome is the major protein degradation machinery of the cell and is regulated at many levels. One mode of regulation involves accumulation of proteasomes in proteasome storage granules (PSGs) upon glucose depletion. Using a systematic robotic screening approach in yeast, we identify trans-acting proteins that regulate the accumulation of proteasomes in PSGs. Our dataset was enriched for subunits of the vacuolar adenosine triphosphatase (V-ATPase) complex, a proton pump required for vacuole acidification. We show that the impaired ability of V-ATPase mutants to properly govern intracellular pH affects the kinetics of PSG formation. We further show that formation of other protein aggregates upon carbon depletion also is triggered in mutants with impaired activity of the plasma membrane proton pump and the V-ATPase complex. We thus identify cytosolic pH as a specific cellular signal involved both in the glucose sensing that mediates PSG formation and in a more general mechanism for signaling carbon source exhaustion.
APA, Harvard, Vancouver, ISO, and other styles
21

Malikova, Marina, Jun Shi, and Konstantin V. Kandror. "V-type ATPase is involved in biogenesis of GLUT4 vesicles." American Journal of Physiology-Endocrinology and Metabolism 287, no. 3 (September 2004): E547—E552. http://dx.doi.org/10.1152/ajpendo.00571.2003.

Full text
Abstract:
Proton pumps participate in several aspects of endocytic protein trafficking. However, their involvement specifically in the GLUT4 pathway has been a matter of great controversy. Here, we report that incubation of 3T3-L1 adipocytes with specific inhibitors of V-type ATPase, concanamycin A and bafilomycin A1, inhibits insulin-regulated glucose transport and results in accumulation of GLUT4 in heavy, rapidly sedimenting intracellular membranes. Correspondingly, the amount of small responsive GLUT4 vesicles in concanamycin A- and bafilomycin A1-treated cells is decreased. We conclude that these drugs block translocation of GLUT4 in adipose cells by inhibiting formation of small insulin-responsive vesicles on donor intracellular membranes. At the same time, proton pump inhibitors do not affect insulin-dependent translocation of preexisting vesicles or GLUT4 sorting in recycling endosomes. On the contrary, wortmannin acutely inhibits insulin-dependent translocation of the preexisting vesicles but has no effect on vesicle formation.
APA, Harvard, Vancouver, ISO, and other styles
22

Bowman, E. J., and B. J. Bowman. "Cellular role of the V-ATPase in Neurospora crassa: analysis of mutants resistant to concanamycin or lacking the catalytic subunit A." Journal of Experimental Biology 203, no. 1 (January 1, 2000): 97–106. http://dx.doi.org/10.1242/jeb.203.1.97.

Full text
Abstract:
Vacuolar ATPases (V-ATPases) are large complex enzymes that are structural and mechanistic relatives of F(1)F(o)-ATPases. They hydrolyze ATP and pump protons across membranes to hyperpolarize membranes and, often, to acidify cellular compartments. The proton gradients generated are used to drive the movement of various compounds across membranes. V-ATPases are found in membranes of archaebacteria and some eubacteria, in various components of the endomembrane system of all eukaryotes and in the plasma membranes of many specialized eukaryotic cells. They have been implicated in a wide variety of cellular processes and are associated with several diseases. Bafilomycin and concanamycin, specific inhibitors of V-ATPases, have been instrumental in implicating the V-ATPase in many of these roles. To understand further the mechanism of inhibition by these antibiotics and the physiological role of the enzyme in the cell, we have isolated mutants of the filamentous fungus Neurospora crassa that are resistant to concanamycin. Concanamycin has a dramatic effect on hyphal morphology at acid pH and is lethal at basic pH. In the resistant mutants, the cells can germinate and grow, although abnormally, in basic medium. Thus far, none of the mutants we have characterized is mutated in a gene encoding a subunit of the V-ATPase. Instead, the largest class of mutants is mutated in the gene encoding the plasma-membrane H(+)-ATPase. Mutations in at least four uncharacterized genes can also confer resistance. Inactivation of the V-ATPase by disruption of vma-1, which encodes the catalytic subunit (A) of the enzyme, causes a much more severe phenotype than inhibition by concanamycin. A strain lacking vma-1 is seriously impaired in rate of growth, differentiation and capacity to produce viable spores. It is also completely resistant to concanamycin, indicating that the inhibitory effects of concanamycin in vivo are due to inhibition of the V-ATPase. How the multiplicity of ATPases within a cell is regulated and how their activity is integrated with other metabolic reactions is poorly understood. Mutant analysis should help unravel this puzzle.
APA, Harvard, Vancouver, ISO, and other styles
23

Graner, Michael. "V-ATPase expression in gliomas—Not your grandparents' proton pump." EBioMedicine 41 (March 2019): 13–14. http://dx.doi.org/10.1016/j.ebiom.2019.02.044.

Full text
APA, Harvard, Vancouver, ISO, and other styles
24

Liégeois, Samuel, Alexandre Benedetto, Jean-Marie Garnier, Yannick Schwab, and Michel Labouesse. "The V0-ATPase mediates apical secretion of exosomes containing Hedgehog-related proteins in Caenorhabditis elegans." Journal of Cell Biology 173, no. 6 (June 19, 2006): 949–61. http://dx.doi.org/10.1083/jcb.200511072.

Full text
Abstract:
Polarized intracellular trafficking in epithelia is critical in development, immunity, and physiology to deliver morphogens, defensins, or ion pumps to the appropriate membrane domain. The mechanisms that control apical trafficking remain poorly defined. Using Caenorhabditis elegans, we characterize a novel apical secretion pathway involving multivesicularbodies and the release of exosomes at the apical plasma membrane. By means of two different genetic approaches, we show that the membrane-bound V0 sector of the vacuolar H+-ATPase (V-ATPase) acts in this pathway, independent of its contribution to the V-ATPase proton pump activity. Specifically, we identified mutations in the V0 “a” subunit VHA-5 that affect either the V0-specific function or the V0+V1 function of the V-ATPase. These mutations allowed us to establish that the V0 sector mediates secretion of Hedgehog-related proteins. Our data raise the possibility that the V0 sector mediates exosome and morphogen release in mammals.
APA, Harvard, Vancouver, ISO, and other styles
25

Manolson, M. F., D. Proteau, and E. W. Jones. "Evidence for a conserved 95-120 kDa subunit associated with and essential for activity of V-ATPases." Journal of Experimental Biology 172, no. 1 (November 1, 1992): 105–12. http://dx.doi.org/10.1242/jeb.172.1.105.

Full text
Abstract:
Vacuoles purified from Saccharomyces cerevisiae bearing the vph1-1 mutation had no detectable bafilomycin-sensitive ATPase activity or ATP-dependent proton pumping. Furthermore, the vacuolar H(+)-ATPase (V-ATPase) nucleotide binding subunits were no longer associated with vacuolar membranes yet were present at wild-type levels in yeast whole-cell extracts. The VPH1 gene was cloned by screening a lambda gt11 expression library with antibodies directed against a 95 kDa vacuolar integral membrane protein and independently cloned by complementation of the vph1-1 mutation. Deletion disruption of the VPH1 gene revealed that the VPH1 gene is required for vacuolar H(+)-ATPase assembly and vacuolar acidification but is not essential for cell viability or for targeting and maturation of vacuolar proteases. VPH1 encodes a predicted polypeptide of 840 amino acid residues (95.6 kDa) with putative membrane-spanning regions. Cell fractionation and immunodetection demonstrate that Vph1p is a vacuolar integral membrane protein that co-purifies with V-ATPase activity. Vph1p has 42% identity to the 116 kDa polypeptide of the rat clathrin-coated vesicles/synaptic vesicle proton pump, 42% identity to the TJ6 mouse immune suppressor factor, 42% identity to the Caenorhabditis elegans proton pump homologue and 54% identity to the predicted polypeptide encoded by the yeast gene STV1 (Similar To VPH1, identified as an open reading frame next to the BUB2 gene.
APA, Harvard, Vancouver, ISO, and other styles
26

Baars, Tonie L., Sebastian Petri, Christopher Peters, and Andreas Mayer. "Role of the V-ATPase in Regulation of the Vacuolar Fission–Fusion Equilibrium." Molecular Biology of the Cell 18, no. 10 (October 2007): 3873–82. http://dx.doi.org/10.1091/mbc.e07-03-0205.

Full text
Abstract:
Like numerous other eukaryotic organelles, the vacuole of the yeast Saccharomyces cerevisiae undergoes coordinated cycles of membrane fission and fusion in the course of the cell cycle and in adaptation to environmental conditions. Organelle fission and fusion processes must be balanced to ensure organelle integrity. Coordination of vacuole fission and fusion depends on the interactions of vacuolar SNARE proteins and the dynamin-like GTPase Vps1p. Here, we identify a novel factor that impinges on the fusion–fission equilibrium: the vacuolar H+-ATPase (V-ATPase) performs two distinct roles in vacuole fission and fusion. Fusion requires the physical presence of the membrane sector of the vacuolar H+-ATPase sector, but not its pump activity. Vacuole fission, in contrast, depends on proton translocation by the V-ATPase. Eliminating proton pumping by the V-ATPase either pharmacologically or by conditional or constitutive V-ATPase mutations blocked salt-induced vacuole fragmentation in vivo. In living cells, fission defects are epistatic to fusion defects. Therefore, mutants lacking the V-ATPase display large single vacuoles instead of multiple smaller vacuoles, the phenotype that is generally seen in mutants having defects only in vacuolar fusion. Its dual involvement in vacuole fission and fusion suggests the V-ATPase as a potential regulator of vacuolar morphology and membrane dynamics.
APA, Harvard, Vancouver, ISO, and other styles
27

Kitay, Alice Miriam, Marie-Therese Schneebacher, Anne Schmitt, Katharina Heschl, Sascha Kopic, Tariq Alfadda, Abrar Alsaihati, Alexander Link, and John Peter Geibel. "Modulations in extracellular calcium lead to H+-ATPase-dependent acid secretion: a clarification of PPI failure." American Journal of Physiology-Gastrointestinal and Liver Physiology 315, no. 1 (July 1, 2018): G36—G42. http://dx.doi.org/10.1152/ajpgi.00132.2017.

Full text
Abstract:
The H+,K+-ATPase was identified as the primary proton secretory pathway in the gastric parietal cell and is the pharmacological target of agents suppressing acid secretion. Recently, we identified a second acid secretory protein expressed in the parietal cell, the vacuolar H+-ATPase (V-type ATPase). The aim of the present study was to further characterize H+-ATPase activation by modulations in extracellular calcium via the calcium sensing receptor (CaSR). Isolated gastric glands were loaded with the pH indicator dye BCECF-AM [2′,7′-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein acetoxymethyl ester] to measure intracellular pH. Experiments were conducted in the absence of sodium and potassium to monitor H+-ATPase-specific transport activity. CaSR was activated with the calcimimetic R568 (400 nM) and/or by modulations in extracellular Ca2+. Elevation in calcium concentrations increased proton extrusion from the gastric parietal cell. Allosteric modification of the CaSR via R568 and calcium increased vacuolar H+-ATPase activity significantly (ΔpH/minlowCa2+(0.1mM) = 0.001 ± 0.001, ΔpH/minnormalCa2+(1.0mM) = 0.033 ± 0.004, ΔpH/minhighCa2+(5.0mM) = 0.051 ± 0.005). Carbachol significantly suppressed calcium-induced gastric acid secretion via the H+-ATPase under sodium- and potassium-free conditions. We conclude that the V-type H+-ATPase is tightly linked to CaSR activation. We observed that proton pump inhibitor (PPI) exposure does not modulate H+-ATPase activity. This elevated blood calcium activation of the H+-ATPase could provide an explanation for recurrent reflux symptoms while taking a PPI therapy. NEW & NOTEWORTHY This study emphasizes the role of the H+-ATPase in acid secretion. We further demonstrate the modification of this proton excretion pathway by extracellular calcium and the activation of the calcium sensing receptor CaSR. The novelty of this paper is based on the modulation of the H+-ATPase via both extracellular Ca (activation) and the classical secretagogues histamine and carbachol (inactivation). Both activation and inactivation of this proton pump are independent of PPI modulation.
APA, Harvard, Vancouver, ISO, and other styles
28

Lepier, A., M. Azuma, W. R. Harvey, and H. Wieczorek. "K+/H+ antiport in the tobacco hornworm midgut: the K(+)-transporting component of the K+ pump." Journal of Experimental Biology 196, no. 1 (November 1, 1994): 361–73. http://dx.doi.org/10.1242/jeb.196.1.361.

Full text
Abstract:
The midgut of the tobacco hornworm secretes K+ across the apical plasma membrane of its goblet cells. This secondary K+ transport results from K+/H+ antiport energized by the proton-motive force generated by a primary, H(+)-transporting plasma membrane V-ATPase. Thus, the lepidopteran midgut constitutes a well-established example of the emerging concept that the proton-motive force is an alternative to the classical sodium-motive force for the energization of animal plasma membranes. K+/H+ antiport in the tobacco hornworm midgut is electrophoretic, exchanging 2H+ for 1K+. Under physiological conditions, it is energized by the voltage component of the proton-motive force. The strong coupling of electrophoretic K+/2H+ antiport with the electrogenic V-ATPase provides, in principle, the minimal device for the alkalization of the midgut lumen to pH values higher than 11. K+/H+ antiport is insensitive to bafilomycin A1, but is inhibited by amiloride or Concanavalin A. Lectin staining of blots after SDS-PAGE revealed several glycosylated polypeptides in the goblet cell apical membrane which are not part of the V-ATPase and thus are candidates for the antiporter protein. Current efforts are focused on the isolation of the K+/H+ antiporter.
APA, Harvard, Vancouver, ISO, and other styles
29

Rane, Hallie S., Stella M. Bernardo, Summer R. Hayek, Jessica L. Binder, Karlett J. Parra, and Samuel A. Lee. "The Contribution of Candida albicans Vacuolar ATPase Subunit V1B, Encoded byVMA2, to Stress Response, Autophagy, and Virulence Is Independent of Environmental pH." Eukaryotic Cell 13, no. 9 (July 18, 2014): 1207–21. http://dx.doi.org/10.1128/ec.00135-14.

Full text
Abstract:
ABSTRACTCandida albicansvacuoles are central to many critical biological processes, including filamentation andin vivovirulence. The V-ATPase proton pump is a multisubunit complex responsible for organellar acidification and is essential for vacuolar biogenesis and function. To study the function of the V1B subunit ofC. albicansV-ATPase, we constructed a tetracycline-regulatableVMA2mutant, tetR-VMA2. Inhibition ofVMA2expression resulted in the inability to grow at alkaline pH and altered resistance to calcium, cold temperature, antifungal drugs, and growth on nonfermentable carbon sources. Furthermore, V-ATPase was unable to fully assemble at the vacuolar membrane and was impaired in proton transport and ATPase-specific activity.VMA2repression led to vacuolar alkalinization in addition to abnormal vacuolar morphology and biogenesis. Key virulence-related traits, including filamentation and secretion of degradative enzymes, were markedly inhibited. These results are consistent with previous studies ofC. albicansV-ATPase; however, differential contributions of the V-ATPase Voand V1subunits to filamentation and secretion are observed. We also make the novel observation that inhibition ofC. albicansV-ATPase results in increased susceptibility to osmotic stress. Notably, V-ATPase inhibition under conditions of nitrogen starvation results in defects in autophagy. Lastly, we show the first evidence that V-ATPase contributes to virulence in an acidicin vivosystem by demonstrating that the tetR-VMA2mutant is avirulent in aCaenorhabditis elegansinfection model. This study illustrates the fundamental requirement of V-ATPase for numerous key virulence-related traits inC. albicansand demonstrates that the contribution of V-ATPase to virulence is independent of host pH.
APA, Harvard, Vancouver, ISO, and other styles
30

Santos-Pereira, Cátia, Lígia R. Rodrigues, and Manuela Côrte-Real. "Plasmalemmal V-ATPase as a Potential Biomarker for Lactoferrin-Based Anticancer Therapy." Biomolecules 12, no. 1 (January 12, 2022): 119. http://dx.doi.org/10.3390/biom12010119.

Full text
Abstract:
Lactoferrin (Lf) is a milk-derived protein with well-recognized potential as a therapeutic agent against a wide variety of cancers. This natural protein exhibits health-promoting effects and has several interesting features, including its selectivity towards cancer cells, good tolerability in humans, worldwide availability, and holding a generally recognized as safe (GRAS) status. To prompt the rational clinical application of this promising anticancer compound, previous works aimed to unveil the molecular mechanisms underlying its selective anticancer activity, where plasmalemmal V-ATPase was identified as an Lf target in cancer cells. V-ATPase is a proton pump critical for cellular homeostasis that migrates to the plasma membrane of highly metastatic cancer cells contributing to the acidity of the tumor microenvironment. Cancer cells were found to be susceptible to Lf only when this proton pump is present at the plasma membrane. Plasmalemmal V-ATPase can thus be an excellent biomarker for driving treatment decisions and forecasting clinical outcomes of Lf-based anticancer strategies. Future research endeavors should thus seek to validate this biomarker by thorough preclinical and clinical studies, as well as to develop effective methods for its detection under clinical settings.
APA, Harvard, Vancouver, ISO, and other styles
31

Yadav, Jyoti, Sabina Muend, Yongqiang Zhang, and Rajini Rao. "A Phenomics Approach in Yeast Links Proton and Calcium Pump Function in the Golgi." Molecular Biology of the Cell 18, no. 4 (April 2007): 1480–89. http://dx.doi.org/10.1091/mbc.e06-11-1049.

Full text
Abstract:
The Golgi-localized Ca2+- and Mn2+-transporting ATPase Pmr1 is important for secretory pathway functions. Yeast mutants lacking Pmr1 show growth sensitivity to multiple drugs (amiodarone, wortmannin, sulfometuron methyl, and tunicamycin) and ions (Mn2+ and Ca2+). To find components that function within the same or parallel cellular pathways as Pmr1, we identified genes that shared multiple pmr1 phenotypes. These genes were enriched in functional categories of cellular transport and interaction with cellular environment, and predominantly localize to the endomembrane system. The vacuolar-type H+-transporting ATPase (V-ATPase), rather than other Ca2+ transporters, was found to most closely phenocopy pmr1Δ, including a shared sensitivity to Zn2+ and calcofluor white. However, we show that pmr1Δ mutants maintain normal vacuolar and prevacuolar pH and that the two transporters do not directly influence each other's activity. Together with a synthetic fitness defect of pmr1ΔvmaΔ double mutants, this suggests that Pmr1 and V-ATPase work in parallel toward a common function. Overlaying data sets of growth sensitivities with functional screens (carboxypeptidase secretion and Alcian Blue binding) revealed a common set of genes relating to Golgi function. We conclude that overlapping phenotypes with Pmr1 reveal Golgi-localized functions of the V-ATPase and emphasize the importance of calcium and proton transport in secretory/prevacuolar traffic.
APA, Harvard, Vancouver, ISO, and other styles
32

Muench, Stephen P., John Trinick, and Michael A. Harrison. "Structural divergence of the rotary ATPases." Quarterly Reviews of Biophysics 44, no. 3 (March 22, 2011): 311–56. http://dx.doi.org/10.1017/s0033583510000338.

Full text
Abstract:
AbstractThe rotary ATPase family of membrane protein complexes may have only three members, but each one plays a fundamental role in biological energy conversion. The F1Fo-ATPase (F-ATPase) couples ATP synthesis to the electrochemical membrane potential in bacteria, mitochondria and chloroplasts, while the vacuolar H+-ATPase (V-ATPase) operates as an ATP-driven proton pump in eukaryotic membranes. In different species of archaea and bacteria, the A1Ao-ATPase (A-ATPase) can function as either an ATP synthase or an ion pump. All three of these multi-subunit complexes are rotary molecular motors, sharing a fundamentally similar mechanism in which rotational movement drives the energy conversion process. By analogy to macroscopic systems, individual subunits can be assigned to rotor, axle or stator functions. Recently, three-dimensional reconstructions from electron microscopy and single particle image processing have led to a significant step forward in understanding of the overall architecture of all three forms of these complexes and have allowed the organisation of subunits within the rotor and stator parts of the motors to be more clearly mapped out. This review describes the emerging consensus regarding the organisation of the rotor and stator components of V-, A- and F-ATPases, examining core similarities that point to a common evolutionary origin, and highlighting key differences. In particular, it discusses how newly revealed variation in the complexity of the inter-domain connections may impact on the mechanics and regulation of these molecular machines.
APA, Harvard, Vancouver, ISO, and other styles
33

Jansen, Jos C., David Wolthuis, Monique Van Scherpenzeel, Vlad Ratziu, Joost P. H. Drenth, and Dirk J. Lefeber. "NAFLD Phenotype in Patients With V-ATPase Proton Pump Assembly Defects." Cellular and Molecular Gastroenterology and Hepatology 5, no. 3 (2018): 415–17. http://dx.doi.org/10.1016/j.jcmgh.2017.12.011.

Full text
APA, Harvard, Vancouver, ISO, and other styles
34

Fenwick, J. C., S. E. Wendelaar Bonga, and G. Flik. "In vivo bafilomycin-sensitive Na(+) uptake in young freshwater fish." Journal of Experimental Biology 202, no. 24 (December 15, 1999): 3659–66. http://dx.doi.org/10.1242/jeb.202.24.3659.

Full text
Abstract:
In vivo treatment with external bafilomycin A(1), a selective inhibitor of V-ATPase H(+) pumps, reduced whole-body Na(+) influx by up to 90 % in young tilapia and 70 % in young carp. The inhibition was rapidly reversible, with whole-body Na(+) influx rebounding to 280 % of pre-treatment values within 20 min of removal from the bafilomycin. This rebound effect is consistent with the prior accumulation of protons during the period when the cells were exposed to bafilomycin. Bafilomycin also inhibited Cl(−) uptake, an effect that was still apparent 30 min after the removal of bafilomycin. These data provide circumstantial evidence for previous suggestions that Na(+) uptake in freshwater fish is associated with a proton-motive force created by a proton pump and indirect evidence for the major significance of this mechanism in the branchial uptake of Na(+) by freshwater fish.
APA, Harvard, Vancouver, ISO, and other styles
35

Ichihara, Atsuhiro, and Kenichiro Kinouchi. "Current knowledge of (pro)renin receptor as an accessory protein of vacuolar H+-ATPase." Journal of the Renin-Angiotensin-Aldosterone System 12, no. 4 (December 2011): 638–40. http://dx.doi.org/10.1177/1470320311429227.

Full text
Abstract:
The (pro)renin receptor (P)RR is a receptor for renin and prorenin, not only allowing local production of angiotensin I from angiotensinogen, but also inducing intracellular signaling. Intriguingly, (P)RR is also called ATP6AP2 because a (P)RR fragment was demonstrated to be associated with vacuolar-type H+-ATPase (V-ATPase), which is of importance for the maintenance of intracellular pH. Recent studies implicate that deletion of (P)RR results in the dysfunction of V-ATPase, suggesting that the (P)RR is essential for its role as a proton pump. Furthermore, the novel function of (P)RR as an adaptor protein between the Wnt receptor complex and the V-ATPase was discovered. Thus, (P)RR is a multi-functional molecule with functions beyond renin and prorenin. This review focuses on the current knowledge and issues of (P)RR and V-ATPase.
APA, Harvard, Vancouver, ISO, and other styles
36

Perry, S. F., M. L. Beyers, and D. A. Johnson. "Cloning and molecular characterisation of the trout (Oncorhynchus mykiss) vacuolar H(+)-ATPase B subunit." Journal of Experimental Biology 203, no. 3 (February 1, 2000): 459–70. http://dx.doi.org/10.1242/jeb.203.3.459.

Full text
Abstract:
The current model of transepithelial ion movements in the gill of freshwater fish incorporates an apically oriented vacuolar H(+)-ATPase (H(+)V-ATPase; proton pump) that is believed to facilitate both acid excretion and Na(+) uptake. To substantiate this model, we have cloned and sequenced a cDNA encoding the B subunit of the rainbow trout (Oncorhynchus mykiss) H(+)V-ATPase. The cloning of the B subunit enabled an examination by northern analysis of its tissue distribution and expression during external hypercapnia. Degenerate oligonucleotide primers to the B subunit of the H(+)V-ATPase were designed and used in a semi-nested polymerase chain reaction (PCR) to amplify an 810 base pair (bp) product from a trout gill/kidney cDNA library. This PCR product was cloned and sequenced and then used to screen the same cDNA library. The assembled 2262 bp cDNA included an open reading frame coding for a deduced protein of 502 amino acid residues. A BLAST search of the GenBank nucleotide database revealed numerous matches to other vertebrate and invertebrate H(+)V-ATPase B subunits. Protein alignment demonstrated that the trout H(+)V-ATPase B subunit is more than 85 % identical and more than 90 % similar to those in other vertebrate species. An initial analysis of H(+)V-ATPase mRNA tissue distribution revealed significant expression in blood. Although a comparison of perfused tissues (blood removed) with non-perfused tissues demonstrated no obvious contribution of the blood to total tissue H(+)-ATPase mRNA levels, all subsequent experiments were performed using perfused tissues. Levels of H(+)V-ATPase mRNA expression were high in the gill, kidney (anterior or posterior), intestine, heart and spleen, but lower in liver and white muscle. Exposure of the fish to 12 h of external hypercapnia (water P(CO2)=7. 5 mmHg; 1 kPa) was associated with a transient increase (at 2 h) in the levels of H(+)V-ATPase B subunit mRNA in gill and kidney; liver mRNA levels were unaffected. These results are consistent with the hypothesis of an apically localised plasma membrane H(+)V-ATPase in the freshwater trout gill and that the expression of this proton pump is increased during periods of acidosis, at least in part because of an increased steady-state level of H(+)V-ATPase mRNA.
APA, Harvard, Vancouver, ISO, and other styles
37

Marsh, Kenneth, Pedro Gonzalez, and Ed Echeverría. "PPi Formation by Reversal of the Tonoplast-bound H+-pyrophosphatase from `Valencia' Orange Juice Cells." Journal of the American Society for Horticultural Science 125, no. 4 (July 2000): 420–24. http://dx.doi.org/10.21273/jashs.125.4.420.

Full text
Abstract:
Tonoplast vesicles isolated from juice cells of mature `Valencia' oranges [Citrus sinensis (L.) Osbeck] showed similar tonoplast-bound vacuolar ATPase (V-ATPase) and inorganic pyrophosphatase (V-PPiase) activity as measured by product formation. Both proton pumps were able to generate a similar pH gradient, although steady-state was reached faster with ATP as substrate. When a ΔpH of 3 units was imposed (vesicle lumen pH of 4.5 and incubation medium of 7.5), tonoplast-bound PPiase was not able to significantly amplify the existing ΔpH. Although not able to function as a H+ pump, V-PPiase effectively synthesized PPi in the presence of inorganic phosphate (Pi). Formation of PPi by V-PPiase was enhanced by ATP but inhibited by NaF, gramicidin, and by antibodies raised against V-PPiase from mung bean [Vigna radiata (L.) R. Wilcz. (Syn. Phaseolus aureus Roxb.)]. Immunological analysis demonstrated an increase in V-PPiase protein with fruit maturity. Data indicate that under in vivo conditions, the V-PPiase of mature orange juice cells acts as a source of inorganic pyrophosphate (PPi) but not as a H+ pump. We propose that synthesis of PPi provides a mechanism for recovery of stored energy in the form of the pH gradient across the vacuole during later stages of development and postharvest storage.
APA, Harvard, Vancouver, ISO, and other styles
38

Coen, Katrijn, Ronald S. Flannagan, Szilvia Baron, Luciene R. Carraro-Lacroix, Dong Wang, Wendy Vermeire, Christine Michiels, et al. "Lysosomal calcium homeostasis defects, not proton pump defects, cause endo-lysosomal dysfunction in PSEN-deficient cells." Journal of Cell Biology 198, no. 1 (July 2, 2012): 23–35. http://dx.doi.org/10.1083/jcb.201201076.

Full text
Abstract:
Presenilin (PSEN) deficiency is accompanied by accumulation of endosomes and autophagosomes, likely caused by impaired endo-lysosomal fusion. Recently, Lee et al. (2010. Cell. doi: http://dx.doi.org/10.1016/j.cell.2010.05.008) attributed this phenomenon to PSEN1 enabling the transport of mature V0a1 subunits of the vacuolar ATPase (V-ATPase) to lysosomes. In their view, PSEN1 mediates the N-glycosylation of V0a1 in the endoplasmic reticulum (ER); consequently, PSEN deficiency prevents V0a1 glycosylation, compromising the delivery of unglycosylated V0a1 to lysosomes, ultimately impairing V-ATPase function and lysosomal acidification. We show here that N-glycosylation is not a prerequisite for proper targeting and function of this V-ATPase subunit both in vitro and in vivo in Drosophila melanogaster. We conclude that endo-lysosomal dysfunction in PSEN−/− cells is not a consequence of failed N-glycosylation of V0a1, or compromised lysosomal acidification. Instead, lysosomal calcium storage/release is significantly altered in PSEN−/− cells and neurons, thus providing an alternative hypothesis that accounts for the impaired lysosomal fusion capacity and accumulation of endomembranes that accompanies PSEN deficiency.
APA, Harvard, Vancouver, ISO, and other styles
39

Breton, Sylvie, Ndona N. Nsumu, Thierry Galli, Ivan Sabolic, Peter J. S. Smith, and Dennis Brown. "Tetanus toxin-mediated cleavage of cellubrevin inhibits proton secretion in the male reproductive tract." American Journal of Physiology-Renal Physiology 278, no. 5 (May 1, 2000): F717—F725. http://dx.doi.org/10.1152/ajprenal.2000.278.5.f717.

Full text
Abstract:
Our laboratory has previously shown that the vacuolar H+-ATPase, located in a subpopulation of specialized cells establishes a luminal acidic environment in the epididymis and proximal part of the vas deferens (Breton S, Smith PJS, Lui B, and Brown D. Nat Med 2: 470–472, 1996). Low luminal pH is critical for sperm maturation and maintenance of sperm in a quiescent state during storage in these organs. In the present study we examined the regulation of proton secretion in the epididymis and vas deferens. In vivo microtubule disruption by colchicine induced an almost complete loss of H+-ATPase apical polarity. Endocytotic vesicles, visualized by Texas red-dextran internalization, contain H+-ATPase, indicating active endocytosis of the pump. Cellubrevin, an analog of the vesicle soluble N-ethyl malemide-sensitive factor attachment protein (SNAP) receptor (v-SNARE) synaptobrevin, is highly enriched in H+-ATPase-rich cells of the epididymis and vas deferens, and tetanus toxin treatment markedly inhibited bafilomycin-sensitive proton secretion by 64.3 ± 9.0% in the proximal vas deferens. Western blotting showed effective cleavage of cellubrevin by tetanus toxin in intact vas deferens, demonstrating that the toxin gained access to cellubrevin. These results suggest that H+-ATPase is actively endocytosed and exocytosed in proton-secreting cells of the epididymis and vas deferens and that net proton secretion requires the participation of the v-SNARE cellubrevin.
APA, Harvard, Vancouver, ISO, and other styles
40

Futai, M., T. Oka, G. Sun-Wada, Y. Moriyama, H. Kanazawa, and Y. Wada. "Luminal acidification of diverse organelles by V-ATPase in animal cells." Journal of Experimental Biology 203, no. 1 (January 1, 2000): 107–16. http://dx.doi.org/10.1242/jeb.203.1.107.

Full text
Abstract:
Eukaryotic cells contain organelles bounded by a single membrane in the cytoplasm. These organelles have differentiated to carry out various functions in the pathways of endocytosis and exocytosis. Their lumina are acidic, with pH ranging from 4.5 to 6.5. This article describes recent studies on these animal cell organelles focusing on (1) the primary proton pump (vacuolar-type H(+)-ATPase) and (2) the functions of the organelle luminal acidity. We also discuss similarities and differences between vacuolar-type H(+)-ATPase and F-type ATPase. Our own studies and interests are emphasized.
APA, Harvard, Vancouver, ISO, and other styles
41

Wadsworth, S. J., A. R. Spitzer, and A. Chander. "Ionic regulation of proton chemical (pH) and electrical gradients in lung lamellar bodies." American Journal of Physiology-Lung Cellular and Molecular Physiology 273, no. 2 (August 1, 1997): L427—L436. http://dx.doi.org/10.1152/ajplung.1997.273.2.l427.

Full text
Abstract:
This study investigated the pH (chemical) and electrical gradients in lamellar bodies, the acidic surfactant-secreting organelles of lung epithelial type II cells, by following the uptake of a weak fluorescent base, quinacrine, and a membrane potential-sensitive dye, bis-(3-phenyl-5-oxoisoxazol-4-yl)pentamethine oxonol (oxonol V). In isolated lung lamellar bodies, the ATP-dependent uptake of both agents could be inhibited by bafilomycin A1, a reportedly specific inhibitor of vacuolar-type H(+)-ATPase (V-ATPase) and could be dissipated by a protonophore, carbonyl cyanide p-(trifluoromethoxy)phenylhydrazone, suggesting that the V-ATPase generates an electropositive interior. A closely linked uptake of Cl- neutralizes the positive electrical potential and increases the proton pump activity. The uptake of quinacrine, but not oxonol V, was decreased by Na+. This effect of Na+ could be prevented by dimethylamiloride, suggesting the presence of electroneutral Na+/H+ exchanger in lamellar body membranes. The initial rates of quinacrine and oxonol V uptake were increased by bumetanide, but only in the presence of Na+, K+, and Cl-, suggesting that the lamellar bodies also contain an outwardly directed electroneutral Na(+)-K(+)-2Cl- cotransporter. Thus three ion transporters, H(+)-translocating V-ATPase, Na+/H+ exchanger, and Na(+)-K(+)-2Cl- cotransporter, appear to determine the chemical and electrical gradients across the lamellar body membrane.
APA, Harvard, Vancouver, ISO, and other styles
42

Matsumura, Shinya, Takeshi Ishikawa, Juichiro Yoshida, Ryuichi Morita, Tomoki Sakakida, Yuki Endo, Toshifumi Doi, et al. "Proton Pump Inhibitors Enhance the Antitumor Effect of Chemotherapy for Esophageal Squamous Cell Carcinoma." Cancers 14, no. 10 (May 12, 2022): 2395. http://dx.doi.org/10.3390/cancers14102395.

Full text
Abstract:
Background: Vacuolar ATPase (V-ATPase) is involved in cancer development. The use of proton pump inhibitors (PPIs) as V-ATPase inhibitors has been reported to enhance the effectiveness of chemotherapy in certain cancers. This study aimed to evaluate the effect of PPIs on chemotherapy for esophageal cancer. Methods: To investigate the effects of PPIs on esophageal cancer cells, human KYSE50 and 70 cells were plated and 3 PPIs (lansoprazole, esomeprazole, vonoprazan) were added at various concentrations with 5-Fluorouracil (5-FU) to the corresponding cells for a cell viability assay. To investigate the effects of PPI treatment on patients undergoing 5-FU-based therapy in the clinical setting, we retrospectively analyzed the clinical outcomes and chemotherapy-related adverse events in 40 esophageal cancer patients who received 5-FU chemotherapy in our hospital between May 2013 and April 2017. Results: In the viability assays, all PPIs significantly enhanced the cytotoxic effect of 5-FU on the two esophageal cancer cell lines. In the clinical study, PPI-treated patients showed better overall survival (OS) than patients managed without PPI treatment. A multivariate analysis revealed that PPI treatment was independently associated with OS (p = 0.009, HR, 0.33; 95% CI, 0.12–0.76). Conclusions: PPI treatment may safely enhance chemosensitivity in esophageal cancer patients.
APA, Harvard, Vancouver, ISO, and other styles
43

Collins, Michael P., Laura A. Stransky, and Michael Forgac. "AKT Ser/Thr kinase increases V-ATPase–dependent lysosomal acidification in response to amino acid starvation in mammalian cells." Journal of Biological Chemistry 295, no. 28 (May 14, 2020): 9433–44. http://dx.doi.org/10.1074/jbc.ra120.013223.

Full text
Abstract:
The vacuolar H+-ATPase (V-ATPase) is an ATP-dependent proton pump that is essential for cellular homeostasis. V-ATPase activity is controlled by the regulated assembly of the enzyme from its component V1 and V0 domains. We previously reported that amino acid starvation rapidly increases V-ATPase assembly and activity in mammalian lysosomes, but the signaling pathways controlling this effect are unknown. In testing inhibitors of pathways important for controlling cellular metabolism, we found here that the cAMP-dependent protein kinase (PKA) inhibitor H89 increases lysosomal V-ATPase activity and blocks any further change upon starvation. The AMP-activated protein kinase (AMPK) inhibitor dorsomorphin decreased lysosomal V-ATPase activity and also blocked any increase upon starvation. However, CRISPR-mediated gene editing revealed that PKA and AMPK are not required for the starvation-dependent increase in lysosomal V-ATPase activity, indicating that H89 and dorsomorphin modify V-ATPase activity through other cellular targets. We next found that the AKT Ser/Thr kinase (AKT) inhibitor MK2206 blocks the starvation-dependent increase in lysosomal V-ATPase activity without altering basal activity. Expression of AKT1 or AKT3, but not AKT2, was required for increased lysosomal V-ATPase activity in response to amino acid starvation in mouse fibroblasts. Finally, HEK293T cells expressing only AKT1 responded normally to starvation, whereas cells expressing only AKT2 displayed a significantly reduced increase in V-ATPase activity and assembly upon starvation. These results show that AKT is required for controlling the rapid response of lysosomal V-ATPase activity to changes in amino acid availability and that this response depends on specific AKT isoforms.
APA, Harvard, Vancouver, ISO, and other styles
44

Williamson, W. Ryan, Dong Wang, Adam S. Haberman, and P. Robin Hiesinger. "A dual function of V0-ATPase a1 provides an endolysosomal degradation mechanism in Drosophila melanogaster photoreceptors." Journal of Cell Biology 189, no. 5 (May 31, 2010): 885–99. http://dx.doi.org/10.1083/jcb.201003062.

Full text
Abstract:
The vesicular adenosine triphosphatase (v-ATPase) is a proton pump that acidifies intracellular compartments. In addition, mutations in components of the membrane-bound v-ATPase V0 sector cause acidification-independent defects in yeast, worm, fly, zebrafish, and mouse. In this study, we present a dual function for the neuron-specific V0 subunit a1 orthologue v100 in Drosophila melanogaster. A v100 mutant that selectively disrupts proton translocation rescues a previously characterized synaptic vesicle fusion defect and vesicle fusion with early endosomes. Correspondingly, V100 selectively interacts with syntaxins on the respective target membranes, and neither synaptic vesicles nor early endosomes require v100 for their acidification. In contrast, V100 is required for acidification once endosomes mature into degradative compartments. As a consequence of the complete loss of this neuronal degradation mechanism, photoreceptors undergo slow neurodegeneration, whereas selective rescue of the acidification-independent function accelerates cell death by increasing accumulations in degradation-incompetent compartments. We propose that V100 exerts a temporally integrated dual function that increases neuronal degradative capacity.
APA, Harvard, Vancouver, ISO, and other styles
45

Sullivan, Gary V., Steve F. Perry, and James N. Fryer. "Localization of mRNA for the proton pump (H+-ATPase) and exchanger in the rainbow trout gill." Canadian Journal of Zoology 74, no. 11 (November 1, 1996): 2095–103. http://dx.doi.org/10.1139/z96-238.

Full text
Abstract:
In situ hybridization was performed on sections of rainbow trout (Oncorhynchus mykiss) gill tissue using oligonucleotide probes complementary to the mRNA of the 31-kilodalton subunit of the bovine renal V-type H+-ATPase or rat kidney Band 3 anion exchanger ([Formula: see text] exchanger). This was conducted in conjunction with measurements of whole-body net acid fluxes and blood acid–base status during imposed conditions of respiratory acidosis (external hypercapnia) or metabolic alkalosis (NaHCO3 infusion). A positive hybridization signal for the H+-ATPase mRNA was localized predominantly in lamellar epithelial cells and was less apparent in cells associated with the filament or interlamellar regions. The H+-ATPase hybridization signal was enhanced during hypercapnic acidosis concurrently with a marked increase in whole-body net acid excretion. A positive hybridization signal for the [Formula: see text] exchanger mRNA was observed in epithelial cells on both the filament and lamella. During metabolic alkalosis induced by intra-arterial infusion of NaHCO3, there was a marked increase in the [Formula: see text] exchanger mRNA hybridization signal in cells on both the filament and lamella that occurred concurrently with a decrease in net acid excretion. The results of this study support the existence of a V-type H+-ATPase and a [Formula: see text] exchanger in rainbow trout gill epithelial cells and demonstrate that alterations in gene expression for the pump–exchanger may be a significant mechanism underlying the altered rates of net acid equivalent excretion during acid – base disturbances.
APA, Harvard, Vancouver, ISO, and other styles
46

SCOTT, David A., and Roberto DOCAMPO. "Two types of H+-ATPase are involved in the acidification of internal compartments in Trypanosoma cruzi." Biochemical Journal 331, no. 2 (April 15, 1998): 583–89. http://dx.doi.org/10.1042/bj3310583.

Full text
Abstract:
ATP-driven acidification of internal compartments of Trypanosoma cruziepimastigotes was assayed spectrophotometrically with Acridine Orange and cells permeabilized with filipin. H+-ATPase activity was not inhibited fully by either 500 nM concanamycin A or 500 µM orthovanadate, but a combination of 5 nM concanamycin A and 25 µM vanadate completely inhibited activity, suggesting the operation of separate V-type (concanamycin-sensitive) and P-type (vanadate-sensitive) H+-ATPase activities in the permeabilized cells. This was supported by different kinetics of Acridine Orange uptake seen in the presence of the different inhibitors, and by different optimal protein (cell) concentrations for the two apparent activities. The use of different buffers further distinguished the ATPases. The V-H+-ATPase activity was stimulated by K+ and inhibited by a lack of anions or the replacement of Cl- with gluconate. The P-type H+-ATPase activity was not affected by a lack of Cl- or K+ but was substantially inhibited in a largely anion-free buffer. This inhibition could be annulled by the addition of the K+ ionophore valinomycin, which probably acted via the establishment of a countercurrent efflux of K+ from the compartment containing the P-type H+-ATPase and the relief of the potential difference generated by the electrogenic proton pump. Valinomycin showed some stimulation of P-type activity in all buffers tested, but its effects on V-H+-ATPase activity were at best transient except in a K+-free buffer, which suggested that the V-H+-ATPase was located in an organelle with relatively low [K+] that was different from that which accommodated the P-type activity. On the basis of acidity and K+ content, these organelles might correspond, in part at least, to the acidocalcisomes (V-H+-ATPase activity) and the reservosomes (P-type activity) previously identified in these cells. Both activities could also be found in the human-infective forms of the parasite, amastigotes and trypomastigotes, but the P-type activity was relatively weak in these cells types, which is correlated with a lack of reservosomes in these forms.
APA, Harvard, Vancouver, ISO, and other styles
47

Collaco, Anne M., Robert L. Jakab, Nadia E. Hoekstra, Kisha A. Mitchell, Amos Brooks, and Nadia A. Ameen. "Regulated traffic of anion transporters in mammalian Brunner's glands: a role for water and fluid transport." American Journal of Physiology-Gastrointestinal and Liver Physiology 305, no. 3 (August 1, 2013): G258—G275. http://dx.doi.org/10.1152/ajpgi.00485.2012.

Full text
Abstract:
The Brunner's glands of the proximal duodenum exert barrier functions through secretion of glycoproteins and antimicrobial peptides. However, ion transporter localization, function, and regulation in the glands are less clear. Mapping the subcellular distribution of transporters is an important step toward elucidating trafficking mechanisms of fluid transport in the gland. The present study examined 1) changes in the distribution of intestinal anion transporters and the aquaporin 5 (AQP5) water channel in rat Brunner's glands following second messenger activation and 2) anion transporter distribution in Brunner's glands from healthy and disease-affected human tissues. Cystic fibrosis transmembrane conductance regulator (CFTR), AQP5, sodium-potassium-coupled chloride cotransporter 1 (NKCC1), sodium-bicarbonate cotransporter (NBCe1), and the proton pump vacuolar ATPase (V-ATPase) were localized to distinct membrane domains and in endosomes at steady state. Carbachol and cAMP redistributed CFTR to the apical membrane. cAMP-dependent recruitment of CFTR to the apical membrane was accompanied by recruitment of AQP5 that was reversed by a PKA inhibitor. cAMP also induced apical trafficking of V-ATPase and redistribution of NKCC1 and NBCe1 to the basolateral membranes. The steady-state distribution of AQP5, CFTR, NBCe1, NKCC1, and V-ATPase in human Brunner's glands from healthy controls, cystic fibrosis, and celiac disease resembled that of rat; however, the distribution profiles were markedly attenuated in the disease-affected duodenum. These data support functional transport of chloride, bicarbonate, water, and protons by second messenger-regulated traffic in mammalian Brunner's glands under physiological and pathophysiological conditions.
APA, Harvard, Vancouver, ISO, and other styles
48

Xie, Y., M. B. Coukell, and Z. Gombos. "Antisense RNA inhibition of the putative vacuolar H(+)-ATPase proteolipid of Dictyostelium reduces intracellular Ca2+ transport and cell viability." Journal of Cell Science 109, no. 2 (February 1, 1996): 489–97. http://dx.doi.org/10.1242/jcs.109.2.489.

Full text
Abstract:
Transport of Ca2+ via a P-type pump into the contractile vacuole of Dictyostelium discoideum appears to be facilitated by vacuolar proton (V-H+) ATPase activity. To investigate the involvement of the V-H(+)-ATPase in this process using molecular techniques, we cloned a cDNA (vatP) encoding the putative proteolipid subunit of this enzyme. The deduced protein product of this cDNA is composed of 196 amino acids with a calculated M(r) of 20,148 and the primary structure exhibits high amino acid sequence identity with V-H(+)-ATPase proteolipids from other organisms. vatP is a single-copy gene and it produces one approximately 900 nt transcript at relatively constant levels during growth and development. Attempts to disrupt the endogenous gene using vatP cDNA were unsuccessful. But, expression of vatP antisense RNA reduced the levels of vatP message and V-H(+)-ATPase activity by 50% or more. These antisense strains grew and developed slowly, especially under acidic conditions, and the cells seemed to have difficulty forming acidic vesicles. During prolonged cultivation, all of the antisense strains either reverted to a wild-type phenotype or died. Thus in Dictyostelium, unlike yeast, the V-H(+)-ATPase seems to be indispensable for cell viability. When different antisense strains were analyzed for Ca2+ uptake by the contractile vacuole, they all accumulated less Ca2+ than control transformants. These results are consistent with earlier pharmacological studies which suggested that the V-H(+)-ATPase functions in intracellular Ca2+ transport in this organism.
APA, Harvard, Vancouver, ISO, and other styles
49

Tyerman, Stephen D., Mary Beilby, John Whittington, Unggul Juswono, Ian Newman, and Sergey Shabala. "Oscillations in proton transport revealed from simultaneous measurements of net current and net proton fluxes from isolated root protoplasts: MIFE meets patch-clamp." Functional Plant Biology 28, no. 7 (2001): 591. http://dx.doi.org/10.1071/pp01030.

Full text
Abstract:
Proton fluxes were measured non-invasively on patch-clamped protoplasts isolated from wheat roots using an external H + electrode to measure the electrochemical gradient in the external solution. Under voltage clamp in the whole-cell configuration, the H + fluxes across the plasma membrane could be measured as a function of voltage and time and correlated with the simultaneous measurements of membrane current. Protoplasts could exist in three states based on the current–voltage (I–V) curves and the flux–V curves. In the pump-state where the membrane voltage (Vm) was more negative than the electrochemical equilibrium potential for potassium (E K ), a net efflux of H + occurred that was voltage-dependent such that the efflux increased as Vm was clamped more positive. In the K-state, where Vm was close to E K , similar flux–V curves were observed. In the depolarised state where Vm was greater than E K the proton flux was characterised by a net influx of H + (H + -influx state) that reversed direction at more positive values of Vm. The inhibitory effect of DCCD and stimulatory effect of fusicoccin were used to correlate current and H + flux through the H + -ATPase for which there was reasonably good agreement within the limits of the flux measurements. Some protoplasts were kept in the whole-cell configuration for up to 3 h revealing slow sustained oscillations (period about 40 min) in H + flux that were in phase with oscillations in free-running Vm. These oscillations were also observed under voltage clamp, with membrane current in phase with H + flux, but which became damped out after a few cycles. The oscillations encompassed the pump-state, K + -state and H + -influx-state. The H +- flux–V curves and I–V curves were used to model the electrical characteristics of the plasma membrane with H + -ATPase, inward and outward K + rectifiers, a linear conductance, and a passive H + influx possibly through gated proton channels.
APA, Harvard, Vancouver, ISO, and other styles
50

Allman, Erik, David Johnson, and Keith Nehrke. "Loss of the apical V-ATPase a-subunit VHA-6 prevents acidification of the intestinal lumen during a rhythmic behavior in C. elegans." American Journal of Physiology-Cell Physiology 297, no. 5 (November 2009): C1071—C1081. http://dx.doi.org/10.1152/ajpcell.00284.2009.

Full text
Abstract:
In Caenorhabditis elegans, oscillations of intestinal pH contribute to the rhythmic defecation behavior, but the acid-base transport mechanisms that facilitate proton movement are not well understood. Here, we demonstrate that VHA-6, an intestine-specific a-subunit of the H+-K+-ATPase complex (V-ATPase), resides in the apical membrane of the intestinal epithelial cells and is required for luminal acidification. Disruption of the vha-6 gene led to early developmental arrest; the arrest phenotype could be complemented by expression of a fluorescently labeled vha-6 transgene. To study the contribution of vha-6 to pH homeostasis in larval worms, we used a partial reduction of function through postembryonic single-generation RNA interference. We demonstrate that the inability to fully acidify the intestinal lumen coincides with a defect in pH recovery of the intestinal epithelial cells, suggesting that VHA-6 is essential for proton pumping following defecation. Moreover, intestinal dipeptide accumulation and fat storage are compromised by the loss of VHA-6, suggesting that luminal acidification promotes nutrient uptake in worms, as well as in mammals. Since acidified intracellular vesicles and autofluorescent storage granules are indistinguishable between the vha-6 mutant and controls, it is likely that the nutrient-restricted phenotype is due to a loss of plasma membrane V-ATPase activity specifically. These data establish a simple genetic model for proton pump-driven acidification. Since defecation occurs at 45-s intervals in worms, this model represents an opportunity to study acute regulation of V-ATPase activity on a short time scale and may be useful in the study of alternative treatments for acid-peptic disorders.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography