Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 294(19): 7878-7891, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30923126

ABSTRACT

The Ca2+/Mn2+ transport ATPases 1a and 2 (SPCA1a/2) are closely related to the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) and are implicated in breast cancer and Hailey-Hailey skin disease. Here, we purified the human SPCA1a/2 isoforms from a yeast recombinant expression system and compared their biochemical properties after reconstitution. We observed that the purified SPCA1a displays a lower Ca2+ affinity and slightly lower Mn2+ affinity than SPCA2. Remarkably, the turnover rates of SPCA1a in the presence of Mn2+ and SPCA2 incubated with Ca2+ and Mn2+ were comparable, whereas the turnover rate of SPCA1a in Ca2+ was 2-fold higher. Moreover, we noted an unusual biphasic activation curve for the SPCA1a ATPase and autophosphorylation activity, not observed with SPCA2. We also found that the biphasic pattern and low apparent ion affinity of SPCA1a critically depends on ATP concentration. We further show that the specific properties of SPCA1a at least partially depend on an N-terminal EF-hand-like motif, which is present only in the SPCA1a isoform and absent in SPCA2. This motif binds Ca2+, and its mutation lowered the Ca2+ turnover rate relative to that of Mn2+, increased substrate affinity, and reduced the level of biphasic activation of SPCA1a. A biochemical analysis indicated that Ca2+ binding to the N-terminal EF-hand-like motif promotes the activity of SPCA1a by facilitating autophosphorylation. We propose that this regulation may be physiologically relevant in cells with a high Ca2+ load, such as mammary gland cells during lactation, or in cells with a low ATP content, such as keratinocytes.


Subject(s)
Calcium-Transporting ATPases/chemistry , Calcium/chemistry , Amino Acid Motifs , Calcium/metabolism , Calcium-Transporting ATPases/genetics , Calcium-Transporting ATPases/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Phosphorylation/genetics , Protein Domains
2.
J Biol Chem ; 292(17): 6938-6951, 2017 04 28.
Article in English | MEDLINE | ID: mdl-28264934

ABSTRACT

The Golgi/secretory pathway Ca2+/Mn2+-transport ATPase (SPCA1a) is implicated in breast cancer and Hailey-Hailey disease. Here, we purified recombinant human SPCA1a from Saccharomyces cerevisiae and measured Ca2+-dependent ATPase activity following reconstitution in proteoliposomes. The purified SPCA1a displays a higher apparent Ca2+ affinity and a lower maximal turnover rate than the purified sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA1a). The lipids cholesteryl hemisuccinate, linoleamide/oleamide, and phosphatidylethanolamine inhibit and phosphatidic acid and sphingomyelin enhance SPCA1a activity. Moreover, SPCA1a is blocked by micromolar concentrations of the commonly used SERCA1a inhibitors thapsigargin (Tg), cyclopiazonic acid, and 2,5-di-tert-butylhydroquinone. Because tissue-specific targeting of SERCA2b by Tg analogues is considered for prostate cancer therapy, the inhibition of SPCA1a by Tg might represent an off-target risk. We assessed the structure-activity relationship (SAR) of Tg for SPCA1a by in silico modeling, site-directed mutagenesis, and measuring the potency of a series of Tg analogues. These indicate that Tg and the analogues are bound via the Tg scaffold but with lower affinity to the same homologous cavity as on the membrane surface of SERCA1a. The lower Tg affinity may depend on a more flexible binding cavity in SPCA1a, with low contributions of the Tg O-3, O-8, and O-10 chains to the binding energy. Conversely, the protein interaction of the Tg O-2 side chain with SPCA1a appears comparable with that of SERCA1a. These differences define a SAR of Tg for SPCA1a distinct from that of SERCA1a, indicating that Tg analogues with a higher specificity for SPCA1a can probably be developed.


Subject(s)
Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Thapsigargin/chemistry , Animals , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Calcium/chemistry , Cholesterol/chemistry , Drug Design , Female , Humans , Hydroquinones/chemistry , Indoles/chemistry , Linoleic Acids/chemistry , Liposomes/chemistry , Male , Mutagenesis, Site-Directed , Oleic Acids/chemistry , Phosphatidic Acids/chemistry , Prostatic Neoplasms/drug therapy , Protein Binding , Protein Conformation , Rabbits , Recombinant Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Sphingomyelins/chemistry , Structure-Activity Relationship
3.
Tissue Cell ; 49(2 Pt A): 141-149, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27692665

ABSTRACT

Dysregulation of the Golgi/Secretory Pathway Ca2+ transport ATPase SPCA2 is implicated in breast cancer. During lactation and in luminal breast cancer types, SPCA2 interacts with the plasma membrane Ca2+ channel Orai1, promoting constitutive Ca2+ influx, which is termed store independent Ca2+ entry (SICE). The mechanism of SPCA2/Orai1 interaction depends on the N- and C-termini of SPCA2. These extensions may play a dual role in activating not only Orai1, but also Ca2+ transport into the Golgi/secretory pathway, which we tested by investigating the impact of various SPCA2 N- and/or C-terminal truncations on SICE and Ca2+ transport activity of SPCA2. C-terminal truncations impair SICE and SPCA2 activity, but also affect targeting, whereas N-terminal truncations affect targeting and inactivate SPCA2, but remarkably, SICE activation remains unaffected. Importantly, overexpression of SPCA2 increases the Ca2+ content of non-ER stores, which depends on Orai1 and SPCA2 activity. Thus, Orai1-mediated Ca2+-influx and SPCA2-mediated Ca2+ uptake activity into the Golgi/secretory pathway might be coupled possibly in a microdomain. This channel/pump complex may efficiently transfer Ca2+ into the secretory pathway, which might play a role in SPCA2-expressing secretory cells, such as mammary gland during lactation.


Subject(s)
Breast Neoplasms/genetics , Calcium-Transporting ATPases/genetics , Golgi Apparatus/genetics , ORAI1 Protein/genetics , Animals , Breast Neoplasms/pathology , Calcium/metabolism , Calcium-Transporting ATPases/metabolism , Endoplasmic Reticulum/genetics , Female , Gene Expression Regulation , Golgi Apparatus/metabolism , Humans , Lactation/genetics , ORAI1 Protein/metabolism , Secretory Pathway/genetics , Signal Transduction/genetics
4.
Proc Natl Acad Sci U S A ; 112(29): 9040-5, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26134396

ABSTRACT

ATP13A2 is a lysosomal P-type transport ATPase that has been implicated in Kufor-Rakeb syndrome and Parkinson's disease (PD), providing protection against α-synuclein, Mn(2+), and Zn(2+) toxicity in various model systems. So far, the molecular function and regulation of ATP13A2 remains undetermined. Here, we demonstrate that ATP13A2 contains a unique N-terminal hydrophobic extension that lies on the cytosolic membrane surface of the lysosome, where it interacts with the lysosomal signaling lipids phosphatidic acid (PA) and phosphatidylinositol(3,5)bisphosphate [PI(3,5)P2]. We further demonstrate that ATP13A2 accumulates in an inactive autophosphorylated state and that PA and PI(3,5)P2 stimulate the autophosphorylation of ATP13A2. In a cellular model of PD, only catalytically active ATP13A2 offers cellular protection against rotenone-induced mitochondrial stress, which relies on the availability of PA and PI(3,5)P2. Thus, the N-terminal binding of PA and PI(3,5)P2 emerges as a key to unlock the activity of ATP13A2, which may offer a therapeutic strategy to activate ATP13A2 and thereby reduce α-synuclein toxicity or mitochondrial stress in PD or related disorders.


Subject(s)
Lipids/chemistry , Parkinson Disease/metabolism , Proton-Translocating ATPases/metabolism , Amino Acid Sequence , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cytosol/metabolism , Endosomes/drug effects , Endosomes/metabolism , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Manganese/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Phosphatidic Acids/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/genetics , Structural Homology, Protein , Zinc/pharmacology
5.
Cold Spring Harb Protoc ; 2014(8): 865-75, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-25086012

ABSTRACT

We provide a detailed procedure to determine the Ca(2+)-dependent ATPase activity in COS or HEK293 cells overexpressing a Ca(2+) pump. The ATPase activity is determined by the Baginsky method, which allows measurement of the steady-state production of inorganic phosphate (Pi). We have adapted this widely applied method into a sensitive, fast, and semi-high-throughput protocol suitable for use in a 96-well plate format.


Subject(s)
Calcium-Transporting ATPases/analysis , High-Throughput Screening Assays , Microsomes/enzymology , Microsomes/metabolism , Animals , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Phosphates/metabolism , Sensitivity and Specificity , Time Factors
6.
Cold Spring Harb Protoc ; 2014(8): 876-86, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-25086013

ABSTRACT

The apparent Ca(2+) affinity of the isoforms of the sarco/endoplasmic reticulum Ca(2+) ATPase SERCA2 is controlled primarily by two proteins, phospholamban (PLB) and sarcolipin (SLN). The rate of ATP-driven Ca(2+) uptake into sarcoplasmic reticulum (SR)-derived vesicles can be monitored by a technique in which the net uptake of (45)Ca(2+) in the form of an intravesicular calcium oxalate precipitate is recorded. Here, we present details of a modification of such a protocol for determining the apparent Ca(2+) affinity of the Ca(2+) pump, and its control by various regulators, in crude homogenates of mouse heart.


Subject(s)
Calcium/metabolism , Myocardium/enzymology , Myocardium/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Complex Mixtures/metabolism , Mice
7.
Cold Spring Harb Protoc ; 2014(8): 876-86, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-25086021

ABSTRACT

Sarco-/endoplasmic reticulum (SR/ER) Ca(2+) pumps (SERCAs) build up vital Ca(2+) gradients across the intracellular SR/ER membrane, helping to control cell function, proliferation, growth, differentiation, and death. We describe two techniques to measure the SERCA activity either in mammalian culture cells overexpressing SERCAs or in muscle tissue containing high levels of endogenous SERCAs. As Ca(2+) transport is tightly coupled to ATP hydrolysis, it is possible to determine the rate of Ca(2+)-dependent ATP hydrolysis and use it as a measure for SERCA activity or, in a second approach, to quantify ATP-stimulated uptake of radioactive (45)Ca(2+). Here, we first provide an overview of the mechanism of Ca(2+)-transport ATPases and show how this can be taken advantage of in protocols for measuring Ca(2+) pump activity.


Subject(s)
Calcium/metabolism , Cations, Divalent/metabolism , Myocardium/enzymology , Myocardium/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/analysis , Adenosine Triphosphate/metabolism , Animals , Biological Transport , Calcium Radioisotopes/metabolism , Cells, Cultured , Gene Expression , Humans , Hydrolysis , Isotope Labeling , Mammals
8.
J Biol Chem ; 287(47): 39460-9, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23024360

ABSTRACT

The molecular mechanism underlying the characteristic high apparent Ca(2+) affinity of SERCA2b relative to SERCA1a and SERCA2a isoforms was studied. The C-terminal tail of SERCA2b consists of an 11th transmembrane helix (TM11) with an associated 11-amino acid luminal extension (LE). The effects of each of these parts and their interactions with the SERCA environment were examined by transient kinetic analysis of the partial reaction steps in the Ca(2+) transport cycle in mutant and chimeric Ca(2+)-ATPase constructs. Manipulations to the LE of SERCA2b markedly increased the rate of Ca(2+) dissociation from Ca(2)E1. Addition of the SERCA2b tail to SERCA1a slowed Ca(2+) dissociation, but only when the luminal L7/8 loop of SERCA1 was simultaneously replaced with that of SERCA2, thus suggesting that the LE interacts with L7/8 in Ca(2)E1. The interaction of LE with L7/8 is also important for the low rate of the Ca(2)E1P → E2P conformational transition. These findings can be rationalized in terms of stabilization of the Ca(2)E1 and Ca(2)E1P forms by docking of the LE near L7/8. By contrast, low rates of E2P dephosphorylation and E2 → E1 transition in SERCA2b depend critically on TM11, particularly in a SERCA2 environment, but do not at all depend on the LE or L7/8. This indicates that interaction of TM11 with SERCA2-specific sequence element(s) elsewhere in the structure is critical in the Ca(2+)-free E2/E2P states. Collectively these properties ensure a higher Ca(2+) affinity of SERCA2b relative to other SERCA isoforms, not only on the cytosolic side, but also on the luminal side.


Subject(s)
Calcium/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , COS Cells , Chlorocebus aethiops , Humans , Ion Transport/physiology , Isoenzymes/genetics , Isoenzymes/metabolism , Protein Binding/physiology , Protein Structure, Secondary , Protein Structure, Tertiary , Rabbits , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics
9.
J Neurochem ; 123(5): 824-36, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22845487

ABSTRACT

Excess Mn(2+) in humans causes a neurological disorder known as manganism, which shares symptoms with Parkinson's disease. However, the cellular mechanisms underlying Mn(2+) -neurotoxicity and the involvement of Mn(2+) -transporters in cellular homeostasis and repair are poorly understood and require further investigation. In this work, we have analyzed the effect of Mn(2+) on neurons and glia from mice in primary cultures. Mn(2+) overload compromised survival of both cell types, specifically affecting cellular integrity and Golgi organization, where the secretory pathway Ca(2+) /Mn(2+) -ATPase is localized. This ATP-driven Mn(2+) transporter might take part in Mn(2+) accumulation/detoxification at low loads of Mn(2+) , but its ATPase activity is inhibited at high concentration of Mn(2+) . Glial cells appear to be significantly more resistant to this toxicity than neurons and their presence in cocultures provided some protection to neurons against degeneration induced by Mn(2+) . Interestingly, the Mn(2+) toxicity was partially reversed upon Mn(2+) removal by wash out or by the addition of EDTA as a chelating agent, in particular in glial cells. These studies provide data on Mn(2+) neurotoxicity and may contribute to explore new therapeutic approaches for reducing Mn(2+) poisoning.


Subject(s)
Calcium-Transporting ATPases/metabolism , Golgi Apparatus/pathology , Manganese/toxicity , Neuroglia/pathology , Neurons/pathology , Animals , Apoptosis , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Immunohistochemistry , Manganese/metabolism , Mice , Neuroglia/metabolism , Neurons/metabolism , Neurotoxins/toxicity , Reverse Transcriptase Polymerase Chain Reaction , Secretory Pathway/drug effects
10.
J Cell Biol ; 198(1): 23-35, 2012 Jul 09.
Article in English | MEDLINE | ID: mdl-22753898

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.


Subject(s)
Calcium/metabolism , Drosophila Proteins/metabolism , Homeostasis/physiology , Lysosomes/metabolism , Presenilin-1/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Biological Transport , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster/enzymology , Fibroblasts/metabolism , Glycosylation , Hippocampus/metabolism , Humans , Mice , Mice, Knockout , Neurons/metabolism , Vacuolar Proton-Translocating ATPases/genetics
11.
J Biol Chem ; 287(24): 19876-85, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22528494

ABSTRACT

The housekeeping sarco(endo)plasmic reticulum Ca(2+) ATPase SERCA2b transports Ca(2+) across the endoplasmic reticulum membrane maintaining a vital Ca(2+) gradient. Compared with the muscle-specific isoforms SERCA2a and SERCA1a, SERCA2b houses an 11th transmembrane segment (TM11) and a short luminal extension (LE) at its C terminus (2b-tail). The 2b-tail imposes a 2-fold higher apparent Ca(2+) affinity and lower V(max). Previously, we assumed that LE is the sole functional region of the 2b-tail and that TM11 is a passive element providing an additional membrane passage. However, here we show that peptides corresponding to the TM11 region specifically modulate the activity of the homologous SERCA1a in co-reconstituted proteoliposomes and mimic the 2b-tail effect (i.e. lower V(max) and higher Ca(2+) affinity). Using truncated 2b-tail variants we document that TM11 regulates SERCA1a independently from LE, confirming that TM11 is a second, previously unrecognized functional region of the 2b-tail. A phylogenetic analysis further indicates that TM11 is the oldest and most conserved feature of the 2b-tail, found in the SERCA pump of all Bilateria, whereas LE is only present in Nematoda and vertebrates. Considering remarkable similarities with the Na(+),K(+)-ATPase α-ß interaction, we now propose a model for interaction of TM11 with TM7 and TM10 in the anchoring subdomain of the Ca(2+) pump. This model involves a TM11-induced helix bending of TM7. In conclusion, more than just a passive structural feature, TM11 acts as a genuine regulator of Ca(2+) transport through interaction with the pump.


Subject(s)
Endoplasmic Reticulum/enzymology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/genetics , Humans , Nematoda/enzymology , Nematoda/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Rabbits , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sodium-Potassium-Exchanging ATPase/genetics
12.
Am J Physiol Heart Circ Physiol ; 302(12): H2574-82, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22505640

ABSTRACT

Cardiomyocytes from failing hearts exhibit reduced levels of the sarcoplasmic reticulum (SR) Ca(2+)-ATPase (SERCA) and/or increased activity of the endogenous SERCA inhibitor phospholamban. The resulting reduction in the Ca(2+) affinity of SERCA impairs SR Ca(2+) cycling in this condition. We have previously investigated the physiological impact of increasing the Ca(2+) affinity of SERCA by substituting SERCA2a with the higher affinity SERCA2b pump. When phospholamban was also ablated, these double knockouts (DKO) exhibited a dramatic reduction in total SERCA levels, severe hypertrophy, and diastolic dysfunction. We presently examined the role of cardiomyocyte Ca(2+) homeostasis in both functional and structural remodeling in these hearts. Despite the low SERCA levels in DKO, we observed near-normal Ca(2+) homeostasis with rapid Ca(2+) reuptake even at high Ca(2+) loads and stimulation frequencies. Well-preserved global Ca(2+) homeostasis in DKO was paradoxically associated with marked activation of the Ca(2+)-dependent nuclear factor of activated T-cell-calcineurin pathway known to trigger hypertrophy. No activation of the MAP kinase signaling pathway was detected. These findings suggest that local changes in Ca(2+) homeostasis may play an important signaling role in DKO, perhaps due to reduced microdomain Ca(2+) buffering by SERCA2b. Furthermore, alterations in global Ca(2+) homeostasis can also not explain impaired in vivo diastolic function in DKO. Taken together, our results suggest that normalizing global cardiomyocyte Ca(2+) homeostasis does not necessarily protect against hypertrophy and heart failure development and that excessively increasing SERCA Ca(2+) affinity may be detrimental.


Subject(s)
Calcium Signaling/physiology , Calcium-Binding Proteins/genetics , Calcium/metabolism , Myocardium/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Animals , Calcium-Binding Proteins/metabolism , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/physiopathology , Homeostasis/physiology , Mice , Mice, Knockout , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
13.
Respir Res ; 13: 27, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22452949

ABSTRACT

BACKGROUND: Chronic thromboembolic pulmonary hypertension (CTEPH) is associated with proximal pulmonary artery obstruction and vascular remodeling. We hypothesized that pulmonary arterial smooth muscle (PASMC) and endothelial cells (PAEC) may actively contribute to remodeling of the proximal pulmonary vascular wall in CTEPH. Our present objective was to characterize PASMC and PAEC from large arteries of CTEPH patients and investigate their potential involvement in vascular remodeling. METHODS: Primary cultures of proximal PAEC and PASMC from patients with CTEPH, with non-thromboembolic pulmonary hypertension (PH) and lung donors have been established. PAEC and PASMC have been characterized by immunofluorescence using specific markers. Expression of smooth muscle specific markers within the pulmonary vascular wall has been studied by immunofluorescence and Western blotting. Mitogenic activity and migratory capacity of PASMC and PAEC have been investigated in vitro. RESULTS: PAEC express CD31 on their surface, von Willebrand factor in Weibel-Palade bodies and take up acetylated LDL. PASMC express various differentiation markers including α-smooth muscle actin (α-SMA), desmin and smooth muscle myosin heavy chain (SMMHC). In vascular tissue from CTEPH and non-thromboembolic PH patients, expression of α-SMA and desmin is down-regulated compared to lung donors; desmin expression is also down-regulated in vascular tissue from CTEPH compared to non-thromboembolic PH patients. A low proportion of α-SMA positive cells express desmin and SMMHC in the neointima of proximal pulmonary arteries from CTEPH patients. Serum-induced mitogenic activity of PAEC and PASMC, as well as migratory capacity of PASMC, were increased in CTEPH only. CONCLUSIONS: Modified proliferative and/or migratory responses of PASMC and PAEC in vitro, associated to a proliferative phenotype of PASMC suggest that PASMC and PAEC could contribute to proximal vascular remodeling in CTEPH.


Subject(s)
Cell Movement , Cell Proliferation , Endothelium, Vascular/pathology , Hypertension, Pulmonary/pathology , Muscle, Smooth, Vascular/pathology , Pulmonary Artery/pathology , Pulmonary Embolism/pathology , Actins/metabolism , Adult , Aged , Cells, Cultured , Chronic Disease , Desmin/metabolism , Endothelium, Vascular/metabolism , Female , Humans , Male , Middle Aged , Muscle, Smooth, Vascular/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pulmonary Artery/cytology , Smooth Muscle Myosins/metabolism , von Willebrand Factor/metabolism
14.
Biochem Soc Trans ; 39(3): 781-7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21599649

ABSTRACT

As a major Ca2+ pump in the sarcoplasmic reticulum of the cardiomyocyte, SERCA2a (sarcoplasmic/endoplasmic reticulum Ca2+-ATPase 2a) controls the relaxation and contraction of the cardiomyocyte. It is meticulously regulated by adapting its expression levels and affinity for Ca2+ ions to the physiological demand of the heart. Dysregulation of the SERCA2a activity entails poor cardiomyocyte contractility, resulting in heart failure. Conversely, improving cardiac SERCA2a activity, e.g. by boosting its expression level or by increasing its affinity for Ca2+, is a promising strategy to rescue contractile dysfunction of the failing heart. The structures of the related SERCA1a Ca2+ pump and the Na+/K+-ATPase of the plasma membrane exposed the pumping mechanism and conserved domain architecture of these ion pumps. However, how the Ca2+ affinity of SERCA2a is regulated at the molecular level remained unclear. A structural and functional analysis of the closely related SERCA2b Ca2+ pump, i.e. the housekeeping Ca2+ pump found in the endoplasmic reticulum and the only SERCA isoform characterized by a high Ca2+ affinity, aimed to fill this gap. We demonstrated the existence of a novel and highly conserved site on the SERCA2 pump mediating Ca2+ affinity regulation by the unique C-terminus of SERCA2b (2b-tail). It differs from the earlier-described target site of the affinity regulator phospholamban. Targeting this novel site may provide a new approach to improve SERCA2a function in the failing heart. Strikingly, the intramembrane interaction site of the 2b-tail in SERCA2b shares sequence and structural homology with the binding site of the ß-subunit on the α Na+/K+-ATPase. Thus P-type ATPases seem to have developed related mechanisms of regulation, and it is a future challenge for us to discover these general principles of P-type regulation.


Subject(s)
Calcium/metabolism , Heart Failure/metabolism , Myocardium/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sarcoplasmic Reticulum/metabolism , Heart Failure/physiopathology , Isoenzymes/chemistry , Isoenzymes/metabolism , Models, Molecular , Myocardial Contraction/physiology , Protein Conformation , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Structure-Activity Relationship
15.
Biophys J ; 100(5): 1216-25, 2011 Mar 02.
Article in English | MEDLINE | ID: mdl-21354394

ABSTRACT

The SERCA2a isoform of the sarco/endoplasmic reticulum Ca(2+) pumps is specifically expressed in the heart, whereas SERCA2b is the ubiquitously expressed variant. It has been shown previously that replacement of SERCA2a by SERCA2b in mice (SERCA2(b/b) mice) results in only a moderate functional impairment, whereas SERCA activity is decreased by a 40% lower SERCA protein expression and by increased inhibition by phospholamban. To find out whether the documented kinetic differences in SERCA2b relative to SERCA2a (i.e., a twofold higher apparent Ca(2+) affinity, but twofold lower maximal turnover rate) can explain these compensatory changes, we simulated Ca(2+) dynamics in mouse ventricular myocytes. The model shows that the relative Ca(2+) transport capacity of SERCA2a and SERCA2b depends on the SERCA concentration. The simulations point to a dominant effect of SERCA2b's higher Ca(2+) affinity over its lower maximal turnover rate. The results suggest that increased systolic and decreased diastolic Ca(2+) levels in unstimulated conditions could contribute to the downregulation of SERCA in SERCA2(b/b) mice. In stress conditions, Ca(2+) handling is less efficient by SERCA2b than by SERCA2a, which might contribute to the observed hypertrophy in SERCA2(b/b) mice. Altogether, SERCA2a might be a better compromise between performance in basal conditions and performance during ß-adrenergic stress.


Subject(s)
Calcium/metabolism , Models, Biological , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Actin Cytoskeleton/metabolism , Animals , Biological Transport , Cell Compartmentation , Isoenzymes/metabolism , Kinetics , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/enzymology , Protein Binding , Sarcomeres/metabolism
16.
Article in English | MEDLINE | ID: mdl-21441596

ABSTRACT

The various splice variants of the three SERCA- and the two SPCA-pump genes in higher vertebrates encode P-type ATPases of the P(2A) group found respectively in the membranes of the endoplasmic reticulum and the secretory pathway. Of these, SERCA2b and SPCA1a represent the housekeeping isoforms. The SERCA2b form is characterized by a luminal carboxy terminus imposing a higher affinity for cytosolic Ca(2+) compared to the other SERCAs. This is mediated by intramembrane and luminal interactions of this extension with the pump. Other known affinity modulators like phospholamban and sarcolipin decrease the affinity for Ca(2+). The number of proteins reported to interact with SERCA is rapidly growing. Here, we limit the discussion to those for which the interaction site with the ATPase is specified: HAX-1, calumenin, histidine-rich Ca(2+)-binding protein, and indirectly calreticulin, calnexin, and ERp57. The role of the phylogenetically older and structurally simpler SPCAs as transporters of Ca(2+), but also of Mn(2+), is also addressed.


Subject(s)
Calcium-Transporting ATPases/physiology , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Animals , Calcium Signaling , Calcium-Transporting ATPases/chemistry , Calcium-Transporting ATPases/metabolism , Developmental Disabilities/genetics , Developmental Disabilities/metabolism , Endoplasmic Reticulum/chemistry , Gene Expression Regulation , Golgi Apparatus/chemistry , Humans , Pemphigus, Benign Familial/genetics , Pemphigus, Benign Familial/metabolism , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Isoforms/physiology , Protein Structure, Tertiary , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/physiology
17.
Biochim Biophys Acta ; 1813(5): 1118-27, 2011 May.
Article in English | MEDLINE | ID: mdl-21215281

ABSTRACT

The ubiquitous sarco(endo)plasmic reticulum (SR/ER) Ca(2+) ATPase (SERCA2b) and secretory-pathway Ca(2+) ATPase (SPCA1a) belong both to the P(2A)-type ATPase subgroup of Ca(2+) transporters and play a crucial role in the Ca(2+) homeostasis of respectively the ER and Golgi apparatus. They are ubiquitously expressed, but their low abundance precludes purification for crystallization. We have developed a new strategy for purification of recombinant hSERCA2b and hSPCA1a that is based on overexpression in yeast followed by a two-step affinity chromatography method biasing towards properly folded protein. In a first step, these proteins were purified with the aid of an analogue of the SERCA inhibitor thapsigargin (Tg) coupled to a matrix. Wild-type (WT) hSERCA2b bound efficiently to the gel, but its elution was hampered by the high affinity of SERCA2b for Tg. Therefore, a mutant was generated carrying minor modifications in the Tg-binding site showing a lower affinity for Tg. In a second step, reactive dye chromatography was performed to further purify and concentrate the properly folded pumps and to exchange the detergent to one more suitable for crystallization. A similar strategy was successfully applied to purify WT SPCA1a. This study shows that it is possible to purify functionally active intracellular Ca(2+) ATPases using successive thapsigargin and reactive dye affinity chromatography for future structural studies. This article is part of a Special Issue entitled: 11th European Symposium on Calcium.


Subject(s)
Calcium-Transporting ATPases/isolation & purification , Chromatography, Affinity/methods , Intracellular Space/enzymology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/isolation & purification , Thapsigargin/metabolism , Binding Sites , Calcium-Transporting ATPases/chemistry , Humans , Mutant Proteins/chemistry , Mutant Proteins/isolation & purification , Protein Structure, Secondary , Recombinant Proteins/isolation & purification , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Thapsigargin/chemistry
18.
Histochem Cell Biol ; 135(1): 11-20, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21120516

ABSTRACT

The neonatal isoform of the sarcoplasmic/endoplasmic reticulum Ca²(+) ATPase 1 (SERCA1b) is a dominant Ca²(+) pump in the young fibers of regenerating muscle. In vivo transfection of about 1% of the fibers with SERCA1b RNAi plasmid resulted in no apparent change in the transfected fibers, but enhanced the increase of fresh weight and fiber size in the whole regenerating rat soleus muscle, until the normal size was reached. Co-transfection of calcineurin inhibitor cain/cabin-1 with SERCA1b RNAi was sufficient to cut down the widespread growth stimulation, but the subsequent transfection of cain into the SERCA1b RNAi transfected muscle did not inhibit muscle growth. The SERCA1b RNAi preferably upregulated the expression of the NFAT reporter lacZ compared to controls when co-transfected into the fibers. Notably, perimuscular injection of interleukin-4 (IL-4) antibody but not that of an unrelevant antibody completely abolished the growth-promoting effect of SERCA1b RNAi. This indicates that silencing SERCA1b in a few fibers stimulates the calcineurin-NFAT-IL-4 pathway and fiber growth in the whole regenerating soleus. These results suggest the presence of an autocrine-paracrine coordination of growing muscle fibers, and put forward a new method to stimulate skeletal muscle regeneration.


Subject(s)
Gene Silencing , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiology , Regeneration/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Animals , Immunoblotting , Male , Protein Isoforms/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
19.
Biochim Biophys Acta ; 1798(8): 1512-21, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20363212

ABSTRACT

Lipid rafts are often considered as microdomains enriched in sphingomyelin and cholesterol, predominantly residing in the plasma membrane but which originate in earlier compartments of the cellular secretory pathway. Within this pathway, the membranes of the Golgi complex represent a transition stage between the cholesterol-poor membranes of the endoplasmic reticulum (ER) and the cholesterol-rich plasma membrane. The rafts are related to detergent-resistant membranes, which because of their ordered structure are poorly penetrated by cold non-ionic detergents and float in density gradient centrifugation. In this study the microdomain niche of the Golgi-resident SPCA Ca(2+)/Mn(2+) pumps was investigated in HT29 cells by Triton X-100 detergent extraction and density-gradient centrifugation. Similarly to cholesterol and the raft-resident flotillin-2, SPCA1 was found mainly in detergent-resistant fractions, while SERCA3 was detergent-soluble. Furthermore, cholesterol depletion of cells resulted in redistribution of flotillin-2 and SPCA1 to the detergent-soluble fractions of the density gradient. Additionally, the time course of solubilization by Triton X-100 was investigated in live COS-1 and HT29 cells expressing fluorescent SERCA2b, SPCA1d or SPCA2. In both cell types, the ER-resident SERCA2b protein was gradually solubilized, while SPCA1d resisted to detergent solubilization. SPCA2 was more sensitive to detergent extraction than SPCA1d. To investigate the functional impact of cholesterol on SPCA1, ATPase activity was monitored. Depletion of cholesterol inhibited the activity of SPCA1d, while SERCA2b function was not altered. From these results we conclude that SPCA1 is associated with cholesterol-rich domains of HT29 cells and that the cholesterol-rich environment is essential for the functioning of the pump.


Subject(s)
Adenocarcinoma/metabolism , Calcium-Transporting ATPases/chemistry , Calcium-Transporting ATPases/metabolism , Cholesterol/chemistry , Cholesterol/metabolism , Colonic Neoplasms/metabolism , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Animals , Base Sequence , COS Cells , Calcium-Transporting ATPases/genetics , Cell Line, Tumor , Chlorocebus aethiops , DNA Primers/genetics , Golgi Apparatus/metabolism , Humans , Immunohistochemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
20.
J Biol Chem ; 285(18): 13542-9, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20177054

ABSTRACT

In pulmonary arterial smooth muscle, Ca(2+) release from the sarcoplasmic reticulum (SR) via ryanodine receptors (RyRs) may induce constriction and dilation in a manner that is not mutually exclusive. We show here that the targeting of different sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPases (SERCA) and RyR subtypes to discrete SR regions explains this paradox. Western blots identified protein bands for SERCA2a and SERCA2b, whereas immunofluorescence labeling of isolated pulmonary arterial smooth muscle cells revealed striking differences in the spatial distribution of SERCA2a and SERCA2b and RyR1, RyR2, and RyR3, respectively. Almost all SERCA2a and RyR3 labeling was restricted to a region within 1.5 microm of the nucleus. In marked contrast, SERCA2b labeling was primarily found within 1.5 microm of the plasma membrane, where labeling for RyR1 was maximal. The majority of labeling for RyR2 lay in between these two regions of the cell. Application of the vasoconstrictor endothelin-1 induced global Ca(2+) waves in pulmonary arterial smooth muscle cells, which were markedly attenuated upon depletion of SR Ca(2+) stores by preincubation of cells with the SERCA inhibitor thapsigargin but remained unaffected after preincubation of cells with a second SERCA antagonist, cyclopiazonic acid. We conclude that functionally segregated SR Ca(2+) stores exist within pulmonary arterial smooth muscle cells. One sits proximal to the plasma membrane, receives Ca(2+) via SERCA2b, and likely releases Ca(2+) via RyR1 to mediate vasodilation. The other is located centrally, receives Ca(2+) via SERCA2a, and likely releases Ca(2+) via RyR3 and RyR2 to initiate vasoconstriction.


Subject(s)
Calcium/metabolism , Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Cell Membrane/metabolism , Endothelin-1/pharmacology , Male , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/cytology , Rats , Rats, Wistar , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...