Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Physiol Rev ; 103(1): 515-606, 2023 01 01.
Article in English | MEDLINE | ID: mdl-35981302

ABSTRACT

The protonation state of soluble and membrane-associated macromolecules dictates their charge, conformation, and functional activity. In addition, protons (H+ or their equivalents) partake in numerous metabolic reactions and serve as a source of electrochemical energy to drive the transmembrane transport of both organic and inorganic substrates. Stringent regulation of the intracellular pH is therefore paramount to homeostasis. Although the regulation of the cytosolic pH has been studied extensively, our understanding of the determinants of the H+ concentration ([H+]) of intracellular organelles has developed more slowly, limited by their small size and inaccessibility. Recently, however, targeting of molecular probes to the organellar lumen together with advances in genomic, proteomic, and electrophysiological techniques have led to the identification and characterization of unique pumps, channels, and transporters responsible for the establishment and maintenance of intraorganellar pH. These developments and their implications for cellular function in health and disease are the subject of this review.


Subject(s)
Vacuolar Proton-Translocating ATPases , Humans , Hydrogen-Ion Concentration , Molecular Probes , Organelles/metabolism , Proteomics , Protons
2.
Nature ; 623(7985): 38-40, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37880527
3.
Nat Rev Mol Cell Biol ; 11(1): 50-61, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19997129

ABSTRACT

Protons dictate the charge and structure of macromolecules and are used as energy currency by eukaryotic cells. The unique function of individual organelles therefore depends on the establishment and stringent maintenance of a distinct pH. This, in turn, requires a means to sense the prevailing pH and to respond to deviations from the norm with effective mechanisms to transport, produce or consume proton equivalents. A dynamic, finely tuned balance between proton-extruding and proton-importing processes underlies pH homeostasis not only in the cytosol, but in other cellular compartments as well.


Subject(s)
Organelles/metabolism , Signal Transduction , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Energy Metabolism , Homeostasis , Humans , Hydrogen-Ion Concentration , Protons
4.
J Biol Chem ; 295(20): 7075-7095, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32277048

ABSTRACT

Genetic screening has identified numerous variants of the endosomal solute carrier family 9 member A6 (SLC9A6)/(Na+,K+)/H+ exchanger 6 (NHE6) gene that cause Christianson syndrome, a debilitating X-linked developmental disorder associated with a range of neurological, somatic, and behavioral symptoms. Many of these variants cause complete loss of NHE6 expression, but how subtler missense substitutions or nonsense mutations that partially truncate its C-terminal cytoplasmic regulatory domain impair NHE6 activity and endosomal function are poorly understood. Here, we describe the molecular and cellular consequences of six unique mutations located in the N-terminal cytoplasmic segment (A9S), the membrane ion translocation domain (L188P and G383D), and the C-terminal regulatory domain (E547*, R568Q, and W570*) of human NHE6 that purportedly cause disease. Using a heterologous NHE6-deficient cell expression system, we show that the biochemical, catalytic, and cellular properties of the A9S and R568Q variants were largely indistinguishable from those of the WT transporter, which obscured their disease significance. By contrast, the L188P, G383D, E547*, and W570* mutants exhibited variable deficiencies in biosynthetic post-translational maturation, membrane sorting, pH homeostasis in recycling endosomes, and cargo trafficking, and they also triggered apoptosis. These findings broaden our understanding of the molecular dysfunctions of distinct NHE6 variants associated with Christianson syndrome.


Subject(s)
Ataxia , Endosomes , Epilepsy , Genetic Diseases, X-Linked , Intellectual Disability , Microcephaly , Mutation, Missense , Ocular Motility Disorders , Sodium-Hydrogen Exchangers , Amino Acid Substitution , Animals , Ataxia/genetics , Ataxia/metabolism , Cricetinae , Endosomes/chemistry , Endosomes/genetics , Endosomes/metabolism , Epilepsy/genetics , Epilepsy/metabolism , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , HeLa Cells , Humans , Hydrogen-Ion Concentration , Intellectual Disability/genetics , Intellectual Disability/metabolism , Microcephaly/genetics , Microcephaly/metabolism , Ocular Motility Disorders/genetics , Ocular Motility Disorders/metabolism , Protein Domains , Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism
5.
Hum Mol Genet ; 28(4): 598-614, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30335141

ABSTRACT

We report two unrelated families with multigenerational nonsyndromic intellectual disability (ID) segregating with a recurrent de novo missense variant (c.1543C>T:p.Leu515Phe) in the alkali cation/proton exchanger gene SLC9A7 (also commonly referred to as NHE7). SLC9A7 is located on human X chromosome at Xp11.3 and has not yet been associated with a human phenotype. The gene is widely transcribed, but especially abundant in brain, skeletal muscle and various secretory tissues. Within cells, SLC9A7 resides in the Golgi apparatus, with prominent enrichment in the trans-Golgi network (TGN) and post-Golgi vesicles. In transfected Chinese hamster ovary AP-1 cells, the Leu515Phe mutant protein was correctly targeted to the TGN/post-Golgi vesicles, but its N-linked oligosaccharide maturation as well as that of a co-transfected secretory membrane glycoprotein, vesicular stomatitis virus G (VSVG) glycoprotein, was reduced compared to cells co-expressing SLC9A7 wild-type and VSVG. This correlated with alkalinization of the TGN/post-Golgi compartments, suggestive of a gain-of-function. Membrane trafficking of glycosylation-deficient Leu515Phe and co-transfected VSVG to the cell surface, however, was relatively unaffected. Mass spectrometry analysis of patient sera also revealed an abnormal N-glycosylation profile for transferrin, a clinical diagnostic marker for congenital disorders of glycosylation. These data implicate a crucial role for SLC9A7 in the regulation of TGN/post-Golgi pH homeostasis and glycosylation of exported cargo, which may underlie the cellular pathophysiology and neurodevelopmental deficits associated with this particular nonsyndromic form of X-linked ID.


Subject(s)
Genetic Diseases, X-Linked/genetics , Golgi Apparatus/genetics , Intellectual Disability/genetics , Sodium-Hydrogen Exchangers/genetics , Acids/metabolism , Animals , CHO Cells , Cell Membrane/genetics , Cricetinae , Cricetulus , Gene Expression Regulation/genetics , Genetic Diseases, X-Linked/metabolism , Genetic Diseases, X-Linked/pathology , Glycosylation , Golgi Apparatus/metabolism , Humans , Intellectual Disability/metabolism , Intellectual Disability/pathology , Membrane Glycoproteins/genetics , Mutation, Missense/genetics , Protein Transport/genetics , Transfection , Viral Envelope Proteins/genetics , trans-Golgi Network/genetics
6.
Neurobiol Dis ; 130: 104490, 2019 10.
Article in English | MEDLINE | ID: mdl-31175985

ABSTRACT

Christianson Syndrome is a rare but increasingly diagnosed X-linked intellectual disability disorder that arises from mutations in SLC9A6/NHE6, a pH-regulating transporter that localizes to early and recycling endosomes. We have recently reported that one of the originally identified disease-causing mutations in NHE6 (p.E287-S288del, or ΔES) resulted in a loss of its pH regulatory function. However, the impact of this mutation upon neuronal synapse formation and plasticity is unknown. Here, we investigate the consequences of the ΔES mutant upon mouse hippocampal pyramidal neurons by expressing a fluorescently-labeled ΔES NHE6 construct into primary hippocampal neurons. Neurons expressing the ΔES mutant showed significant reductions in mature dendritic spine density with a concurrent increase in immature filopodia. Furthermore, compared to wild-type (WT), ΔES-containing endosomes are redirected away from early and recycling endosomes toward lysosomes. In parallel, the ΔES mutant reduced the trafficking of glutamatergic AMPA receptors to excitatory synapses and increased their accumulation within lysosomes for potential degradation. Upon long-term potentiation (LTP), neurons expressing ΔES failed to undergo significant structural and functional changes as observed in controls and WT transfectants. Interestingly, synapse density and LTP-induced synaptic remodeling in ΔES-expressing neurons were partially restored by bafilomycin, a vesicular alkalinisation agent, or by leupeptin, an inhibitor of lysosomal proteolytic degradation. Overall, our results demonstrate that the ∆ES mutation attenuates synapse density and structural and functional plasticity in hippocampal neurons. These deficits may be partially due to the mistargeting of AMPA receptors and other cargos to lysosomes, thereby preventing their trafficking during synaptic remodeling. This mechanism may contribute to the cognitive learning deficits observed in patients with Christianson Syndrome and suggests a potential therapeutic strategy for treatment.


Subject(s)
Ataxia/genetics , Epilepsy/genetics , Genetic Diseases, X-Linked/genetics , Hippocampus/metabolism , Hippocampus/pathology , Intellectual Disability/genetics , Microcephaly/genetics , Neuronal Plasticity/genetics , Ocular Motility Disorders/genetics , Sodium-Hydrogen Exchangers/genetics , Animals , Dendritic Spines/metabolism , Dendritic Spines/pathology , Mice , Mutation , Protein Transport/genetics , Receptors, AMPA/metabolism
7.
Neurobiol Dis ; 121: 187-204, 2019 01.
Article in English | MEDLINE | ID: mdl-30296617

ABSTRACT

Loss-of-function mutations in the recycling endosomal (Na+,K+)/H+ exchanger gene SLC9A6/NHE6 result in overacidification and dysfunction of endosomal-lysosomal compartments, and cause a neurodevelopmental and degenerative form of X-linked intellectual disability called Christianson Syndrome (CS). However, knowledge of the disease heterogeneity of CS is limited. Here, we describe the clinical features and underlying molecular and cellular mechanisms associated with a CS patient carrying a de novo missense variant (p.Gly218Arg; G218R) of a conserved residue in its ion translocation domain that results in a potential gain-of-function. The patient manifested several core symptoms typical of CS, including pronounced cognitive impairment, mutism, epilepsy, ataxia and microcephaly; however, deterioration of motor function often observed after the first decade of life in CS children with total loss of SLC9A6/NHE6 function was not evident. In transfected non-neuronal cells, complex glycosylation and half-life of the G218R were significantly decreased compared to the wild-type transporter. This correlated with elevated ubiquitination and partial proteasomal-mediated proteolysis of G218R. However, a major fraction was delivered to the plasma membrane and endocytic pathways. Compared to wild-type, G218R-containing endosomes were atypically alkaline and showed impaired uptake of recycling endosomal cargo. Moreover, instead of accumulating in recycling endosomes, G218R was redirected to multivesicular bodies/late endosomes and ejected extracellularly in exosomes rather than progressing to lysosomes for degradation. Attenuated acidification and trafficking of G218R-containing endosomes were also observed in transfected hippocampal neurons, and correlated with diminished dendritic branching and density of mature mushroom-shaped spines and increased appearance of filopodia-like protrusions. Collectively, these findings expand our understanding of the genetic diversity of CS and further elucidate a critical role for SLC9A6/NHE6 in fine-tuning recycling endosomal pH and cargo trafficking, processes crucial for the maintenance of neuronal polarity and mature synaptic structures.


Subject(s)
Ataxia/genetics , Ataxia/pathology , Endosomes/metabolism , Epilepsy/genetics , Epilepsy/pathology , Gain of Function Mutation , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/pathology , Intellectual Disability/genetics , Intellectual Disability/pathology , Microcephaly/genetics , Microcephaly/pathology , Neurons/pathology , Ocular Motility Disorders/genetics , Ocular Motility Disorders/pathology , Sodium-Hydrogen Exchangers/genetics , Adult , Animals , Atrophy , Cricetulus , Dendrites/pathology , Extracellular Vesicles/metabolism , HeLa Cells , Hippocampus/pathology , Humans , Male , Mutation, Missense , Sodium-Hydrogen Exchangers/chemistry , Young Adult
8.
Plant Cell Environ ; 41(9): 2169-2182, 2018 09.
Article in English | MEDLINE | ID: mdl-29520811

ABSTRACT

Natural genetic variations in waterlogging tolerance are controlled by multiple genes mapped as quantitative trait loci (QTLs) in major crops, including soybean (Glycine max L.). In this research, 2 novel QTLs associated with waterlogging tolerance were mapped from an elite/exotic soybean cross. The subsequent research was focused on a major QTL (qWT_Gm03) with the tolerant allele from the exotic parent. This QTL was isolated into near-isogenic backgrounds, and its effects on waterlogging tolerance were validated in multiple environments. Fine mapping narrowed qWT_Gm03 into a genomic region of <380 Kbp excluding Rps1 gene for Phytophthora sojae resistance. The tolerant allele of qWT_Gm03 promotes root growth under nonstress conditions and favourable root plasticity under waterlogging, resulting in improved waterlogging tolerance, yield, and drought tolerance-related traits, possibly through more efficient water/nutrient uptakes. Meanwhile, involvement of auxin pathways was also identified in the regulation of waterlogging tolerance, as the genotypic differences of qWT_Gm03 in waterlogging tolerance and formation of adventitious/aerial roots can be complemented by an exogenous auxin-biosynthesis inhibitor. These findings provided genetic resources to address the urgent demand of improving waterlogging tolerance in soybean and revealed the determinant roles of root architecture and plasticity in the plant adaptation to waterlogging.


Subject(s)
Glycine max/genetics , Plant Roots/anatomy & histology , Quantitative Trait Loci , Alleles , Chromosome Mapping , Genetic Variation , Indoleacetic Acids/metabolism , Plant Roots/genetics , Plant Roots/growth & development , Glycine max/physiology , Water/metabolism
9.
J Biol Chem ; 290(29): 18173-18186, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26063808

ABSTRACT

Mammalian Na(+)/H(+) exchangers (NHEs) regulate numerous physiological processes and are involved in the pathogenesis of several diseases, including tissue ischemia and reperfusion injuries, cardiac hypertrophy and failure, and cancer progression. Hence, NHEs are being targeted for pharmaceutical-based clinical therapies, but pertinent information regarding the structural elements involved in cation translocation and drug binding remains incomplete. Molecular manipulations of the prototypical NHE1 isoform have implicated several predicted membrane-spanning (M) helices, most notably M4, M9, and M11, as important determinants of cation permeation and drug sensitivity. Here, we have used substituted-cysteine accessibility mutagenesis and thiol-modifying methanethiosulfonate (MTS) reagents to further probe the involvement of evolutionarily conserved sites within M9 (residues 342-363) and the adjacent exofacial re-entrant loop 5 between M9 and M10 (EL5; residues 364-415) of a cysteine-less variant of rat NHE1 on its kinetic and pharmacological properties. MTS treatment significantly reduced the activity of mutants containing substitutions within M9 (H353C, S355C, and G356C) and EL5 (G403C and S405C). In the absence of MTS, mutants S355C, G403C, and S405C showed modest to significant decreases in their apparent affinities for Na(+) o and/or H(+) i. In addition, mutations Y370C and E395C within EL5, whereas failing to confer sensitivity to MTS, nevertheless, reduced the affinity for Na(+) o, but not for H(+) i. The Y370C mutant also exhibited higher affinity for ethylisopropylamiloride, a competitive antagonist of Na(+) o transport. Collectively, these results further implicate helix M9 and EL5 of NHE1 as important elements involved in cation transport and inhibitor sensitivity, which may inform rational drug design.


Subject(s)
Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Cations/metabolism , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Drug Resistance , Mesylates/chemistry , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Secondary , Rats , Sequence Alignment , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/genetics , Substrate Specificity
10.
J Biol Chem ; 289(30): 20879-97, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24936055

ABSTRACT

Strict regulation of intra- and extracellular pH is an important determinant of nervous system function as many voltage-, ligand-, and H(+)-gated cationic channels are exquisitely sensitive to transient fluctuations in pH elicited by neural activity and pathophysiologic events such as hypoxia-ischemia and seizures. Multiple Na(+)/H(+) exchangers (NHEs) are implicated in maintenance of neural pH homeostasis. However, aside from the ubiquitous NHE1 isoform, their relative contributions are poorly understood. NHE5 is of particular interest as it is preferentially expressed in brain relative to other tissues. In hippocampal neurons, NHE5 regulates steady-state cytoplasmic pH, but intriguingly the bulk of the transporter is stored in intracellular vesicles. Here, we show that NHE5 is a direct target for phosphorylation by the AMP-activated protein kinase (AMPK), a key sensor and regulator of cellular energy homeostasis in response to metabolic stresses. In NHE5-transfected non-neuronal cells, activation of AMPK by the AMP mimetic AICAR or by antimycin A, which blocks aerobic respiration and causes acidification, increased cell surface accumulation and activity of NHE5, and elevated intracellular pH. These effects were effectively blocked by the AMPK antagonist compound C, the NHE inhibitor HOE694, and mutation of a predicted AMPK recognition motif in the NHE5 C terminus. This regulatory pathway was also functional in primary hippocampal neurons, where AMPK activation of NHE5 protected the cells from sustained antimycin A-induced acidification. These data reveal a unique role for AMPK and NHE5 in regulating the pH homeostasis of hippocampal neurons during metabolic stress.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Hippocampus/metabolism , Neurons/metabolism , Sodium-Hydrogen Exchangers/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/genetics , Amino Acid Motifs , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/metabolism , Animals , Antifungal Agents/pharmacology , Antimycin A/pharmacology , Cell Line , Guanidines/pharmacology , Hippocampus/cytology , Humans , Hydrogen-Ion Concentration , Mice , Neurons/cytology , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Structure, Tertiary , Ribonucleotides/genetics , Ribonucleotides/metabolism , Sodium-Hydrogen Exchangers/genetics , Stress, Physiological/drug effects , Stress, Physiological/genetics , Sulfones/pharmacology
11.
EMBO J ; 30(4): 679-91, 2011 Feb 16.
Article in English | MEDLINE | ID: mdl-21245831

ABSTRACT

The Na(+)/H(+) exchanger NHE3 plays a central role in intravascular volume and acid-base homeostasis. Ion exchange activity is conferred by its transmembrane domain, while regulation of the rate of transport by a variety of stimuli is dependent on its cytosolic C-terminal region. Liposome- and cell-based assays employing synthetic or recombinant segments of the cytosolic tail demonstrated preferential association with anionic membranes, which was abrogated by perturbations that interfere with electrostatic interactions. Resonance energy transfer measurements indicated that segments of the C-terminal domain approach the bilayer. In intact cells, neutralization of basic residues in the cytosolic tail by mutagenesis or disruption of electrostatic interactions inhibited Na(+)/H(+) exchange activity. An electrostatic switch model is proposed to account for multiple aspects of the regulation of NHE3 activity.


Subject(s)
Cell Membrane/physiology , Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/physiology , Static Electricity , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Cells, Cultured , Dogs , Electrophysiological Phenomena , Models, Biological , Molecular Sequence Data , Sequence Homology, Amino Acid , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/metabolism , Structure-Activity Relationship , Surface Properties
12.
J Neurosci ; 33(31): 12656-69, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23904602

ABSTRACT

Axon degeneration is a critical pathological feature of many neurodegenerative diseases. Misregulation of local axonal ion homeostasis has been recognized as an important yet understudied mechanism for axon degeneration. Here we report a chemically induced, recessive mouse mutation, vacillator (vac), which causes ataxia and concomitant axon degeneration of cerebellar Purkinje cells. By positional cloning, we identified vac as a point mutation in the calcineurin-like EF hand protein 1 (Chp1) gene that resulted in the production of mutant CHP1 isoforms with an amino acid substitution in a functional EF-hand domain or a truncation of this motif by aberrant splicing and significantly reduced protein levels. CHP1 has been previously shown to interact with the sodium hydrogen exchanger 1 (NHE1), a major regulator of intracellular pH. We demonstrated that CHP1 assists in the full glycosylation of NHE1 that is necessary for the membrane localization of this transporter and that truncated isoforms of CHP1 were defective in stimulating NHE1 biosynthetic maturation. Consistent with this, membrane localization of NHE1 at axon terminals was greatly reduced in Chp1-deficient Purkinje cells before axon degeneration. Furthermore, genetic ablation of Nhe1 also resulted in Purkinje cell axon degeneration, pinpointing the functional convergence of the two proteins. Our findings clearly demonstrate that the polarized presynaptic localization of NHE/CHP1 is an important feature of neuronal axons and that selective disruption of NHE1-mediated proton homeostasis in axons can lead to degeneration, suggesting that local regulation of pH is pivotal for axon survival.


Subject(s)
Axons/physiology , Calcium-Binding Proteins/genetics , Cation Transport Proteins/biosynthesis , Homeostasis/genetics , Nerve Degeneration/pathology , Purkinje Cells/cytology , Sodium-Hydrogen Exchangers/biosynthesis , Age Factors , Alkylating Agents/pharmacology , Animals , Ataxia/genetics , Ataxia/pathology , Axons/drug effects , Axons/pathology , Axons/ultrastructure , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Cells, Cultured , Cerebellum/cytology , Cerebellum/pathology , Cricetinae , Disease Models, Animal , Ethylnitrosourea/pharmacology , Female , Gene Expression Regulation, Developmental/genetics , Homeostasis/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Degeneration/genetics , Nerve Tissue Proteins/metabolism , Point Mutation , Purkinje Cells/drug effects , Sodium-Hydrogen Exchanger 1
13.
J Neurosci ; 33(2): 595-610, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23303939

ABSTRACT

Postsynaptic endosomal trafficking has emerged as a principal regulatory mechanism of structural and functional plasticity of glutamatergic synapses. Recycling endosomes perform activity-dependent transport of AMPA receptors (AMPARs) and lipids to the postsynaptic membrane, activities that are known to contribute to long-term synaptic potentiation and hypothesized to subserve learning and memory processes in the brain. Recently, genetic defects in a widely expressed vesicular pH-regulating transporter, the Na(+)/H(+) exchanger NHE6 isoform, have been implicated in neurodevelopmental disorders including severe X-linked mental retardation and autism. However, little information is available regarding the cellular properties of this transporter in the CNS. Here, we show by quantitative light microscopy that the protein abundance of NHE6 is developmentally regulated in area CA1 of the mouse hippocampus. Within pyramidal neurons, NHE6 was found to localize to discrete puncta throughout the soma and neurites, with noticeable accumulation at dendritic spines and presynaptic terminals. Dual immunolabeling of dendritic spines revealed that NHE6 partially colocalizes with typical markers of early and recycling endosomes as well as with the AMPAR subunit GluA1. Significantly, NHE6-containing vesicles exhibited enhanced translocation to dendritic spine heads during NMDA receptor (NMDAR)-dependent long-term potentiation. These data suggest that NHE6 may play a unique, previously unrecognized, role at glutamatergic synapses that are important for learning and memory.


Subject(s)
Dendritic Spines/metabolism , Endosomes/metabolism , Hippocampus/metabolism , Long-Term Potentiation/physiology , Pyramidal Cells/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Recruitment, Neurophysiological/physiology , Sodium-Hydrogen Exchangers/metabolism , Animals , Blotting, Western , CHO Cells , Cells, Cultured , Centrifugation , Cricetinae , Cricetulus , Dendritic Spines/ultrastructure , Endosomes/ultrastructure , Fluorescent Antibody Technique , Glycine/physiology , Hippocampus/cytology , Hippocampus/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron , Organ Culture Techniques , Pyramidal Cells/ultrastructure , Receptors, AMPA/genetics , Receptors, AMPA/physiology
14.
Curr Opin Cell Biol ; 19(4): 483-92, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17646094

ABSTRACT

The regulated movement of monovalent cations such as H(+), Li(+), Na(+) and K(+) across biological membranes influences a myriad of cellular processes and is fundamental to all living organisms. This is accomplished by a multiplicity of ion channels, pumps and transporters. Our insight into their molecular, cellular and physiological diversity has increased greatly in the past few years with the advent of genome sequencing, genetic manipulation and sophisticated imaging techniques. One of the revelations from these studies is the emergence of novel alkali cation/protons exchangers that are present in endomembranes, where they function to regulate not only intraorganellar pH but also vesicular biogenesis, trafficking and other aspects of cellular homeostasis.


Subject(s)
Homeostasis , Ion Transport , Metals, Alkali/metabolism , Organelles/physiology , Sodium-Hydrogen Exchangers/physiology , Animals , Endosomes/physiology , Humans , Protons
15.
Cell Metab ; 36(6): 1169-1171, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38838638

ABSTRACT

In this issue of Cell Metabolism, Fang et al.1 report a novel pH-sensitive cellular signaling mechanism involving the transcription factor SMAD5 that regulates the vesicular secretion of insulin from pancreatic ß cells in response to dietary challenges. Dysregulation of this pathway may contribute to metabolic disorders such as type 2 diabetes mellitus.


Subject(s)
Insulin Secretion , Insulin-Secreting Cells , Insulin , Signal Transduction , Smad5 Protein , Insulin/metabolism , Animals , Insulin-Secreting Cells/metabolism , Smad5 Protein/metabolism , Humans , Diabetes Mellitus, Type 2/metabolism , Mice , Hydrogen-Ion Concentration
16.
J Biol Chem ; 287(44): 36883-95, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22984264

ABSTRACT

Calcineurin B homologous proteins (CHP) are N-myristoylated, EF-hand Ca(2+)-binding proteins that regulate multiple cellular processes, including intracellular pH homeostasis. Previous work has shown that the heart-enriched isoform, CHP3, regulates the plasmalemmal Na(+)/H(+) exchanger NHE1 isoform by enhancing its rate of oligosaccharide maturation and exocytosis as well as its half-life and transport activity at the cell surface (Zaun, H. C., Shrier, A., and Orlowski, J. (2008) J. Biol. Chem. 283, 12456-12467). However, the molecular basis for this effect is not well understood. In this report, we investigated whether the N-myristoylation and Ca(2+)-binding domains of CHP3 are important elements for regulating NHE1. Mutation of residues essential for either N-myristoylation (G2A) or calcium binding (D123A) did not prevent the interaction of CHP3 with NHE1, although the D123A mutant no longer showed elevated binding to NHE1 in the presence of Ca(2+) when assessed using in vitro binding assays. Disruption of either site also did not impair the ability of CHP3 to stimulate the biosynthetic processing and trafficking of NHE1 to the plasma membrane nor did it affect the H(+) sensitivity of the exchanger. However, they did significantly reduce the cell surface half-life and near maximal transport velocity of NHE1 to a similar extent. Simultaneous mutation of both sites (G2A/D123A) gave results identical to the individual substitutions. This finding suggests that both domains in CHP3 are interdependent and may function cooperatively as a Ca(2+)-myristoyl switch mechanism to selectively stabilize the NHE1·CHP3 complex at the cell surface in a conformation that promotes optimal transport activity.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/metabolism , Cation Transport Proteins/metabolism , Protein Processing, Post-Translational , Sodium-Hydrogen Exchangers/metabolism , Amino Acid Substitution , Animals , CHO Cells , Calcium-Binding Proteins/genetics , Cell Membrane/metabolism , Cricetinae , Glycosylation , Half-Life , Hydrogen-Ion Concentration , Mutagenesis, Site-Directed , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Protein Transport , Sodium-Hydrogen Exchanger 1
17.
J Biol Chem ; 287(21): 17530-17545, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22474287

ABSTRACT

Mammalian target of rapamycin complex 1 (mTORC1) signaling is frequently dysregulated in cancer. Inhibition of mTORC1 is thus regarded as a promising strategy in the treatment of tumors with elevated mTORC1 activity. We have recently identified niclosamide (a Food and Drug Administration-approved antihelminthic drug) as an inhibitor of mTORC1 signaling. In the present study, we explored possible mechanisms by which niclosamide may inhibit mTORC1 signaling. We tested whether niclosamide interferes with signaling cascades upstream of mTORC1, the catalytic activity of mTOR, or mTORC1 assembly. We found that niclosamide does not impair PI3K/Akt signaling, nor does it inhibit mTORC1 kinase activity. We also found that niclosamide does not interfere with mTORC1 assembly. Previous studies in helminths suggest that niclosamide disrupts pH homeostasis of the parasite. This prompted us to investigate whether niclosamide affects the pH balance of cancer cells. Experiments in both breast cancer cells and cell-free systems demonstrated that niclosamide possesses protonophoric activity in cells and in vitro. In cells, niclosamide dissipated protons (down their concentration gradient) from lysosomes to the cytosol, effectively lowering cytoplasmic pH. Notably, analysis of five niclosamide analogs revealed that the structural features of niclosamide required for protonophoric activity are also essential for mTORC1 inhibition. Furthermore, lowering cytoplasmic pH by means other than niclosamide treatment (e.g. incubation with propionic acid or bicarbonate withdrawal) recapitulated the inhibitory effects of niclosamide on mTORC1 signaling, lending support to a possible role for cytoplasmic pH in the control of mTORC1. Our data illustrate a potential mechanism for chemical inhibition of mTORC1 signaling involving modulation of cytoplasmic pH.


Subject(s)
Antinematodal Agents/pharmacology , Niclosamide/pharmacology , Proteins/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Humans , Hydrogen-Ion Concentration , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases
18.
Adv Exp Med Biol ; 961: 397-410, 2013.
Article in English | MEDLINE | ID: mdl-23224898

ABSTRACT

The pH milieu of the central and peripheral nervous systems is an important determinant of neuronal excitability, function, and survival. In mammals, neural acid-base homeostasis is coordinately regulated by ion transporters belonging to the Na(+)/H(+) exchanger (NHE) and bicarbonate transporter gene families. However, the relative contributions of individual isoforms within the respective families are not fully understood. This report focuses on the NHE family, specifically the plasma membrane-type NHE5 which is preferentially transcribed in brain, but the distribution of the native protein has not been extensively characterized. To this end, we generated a rabbit polyclonal antibody that specifically recognizes NHE5. In both central (cortex, hippocampus) and peripheral (superior cervical ganglia, SCG) nervous tissue of mice, NHE5 immunostaining was punctate and highly concentrated in the somas and to lesser amounts in the dendrites of neurons. Very little signal was detected in axons. Similarly, in primary cultures of differentiated SCG neurons, NHE5 localized predominantly to vesicles in the somatodendritic compartment, though some immunostaining was also evident in punctate vesicles along the axons. NHE5 was also detected predominantly in intracellular vesicles of cultured SCG glial cells. Dual immunolabeling of SCG neurons showed that NHE5 did not colocalize with markers for early endosomes (EEA1) or synaptic vesicles (synaptophysin), but did partially colocalize with the transferrin receptor, a marker of recycling endosomes. Collectively, these data suggest that NHE5 partitions into a unique vesicular pool in neurons that shares some characteristics of recycling endosomes where it may serve as an important regulated store of functional transporters required to maintain cytoplasmic pH homeostasis.


Subject(s)
Axons/metabolism , Brain/metabolism , Endosomes/metabolism , Nerve Tissue Proteins/metabolism , Neuroglia/metabolism , Sodium-Hydrogen Exchangers/metabolism , Synaptic Vesicles/metabolism , Animals , Brain/cytology , Cells, Cultured , Endosomes/genetics , Hydrogen-Ion Concentration , Mice , Nerve Tissue Proteins/genetics , Neuroglia/cytology , Rabbits , Sodium-Hydrogen Exchangers/genetics , Synaptic Vesicles/genetics , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
19.
J Biol Chem ; 286(13): 11456-68, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21296876

ABSTRACT

Internalization of the Na(+)/H(+) exchanger NHE5 into recycling endosomes is enhanced by the endocytic adaptor proteins ß-arrestin1 and -2, best known for their preferential recognition of ligand-activated G protein-coupled receptors (GPCRs). However, the mechanism underlying their atypical association with non-GPCRs, such as NHE5, is unknown. In this study, we identified a highly acidic, serine/threonine-rich, di-isoleucine motif (amino acids 697-723) in the cytoplasmic C terminus of NHE5 that is recognized by ß-arrestin2. Gross deletions of this site decreased the state of phosphorylation of NHE5 as well as its binding and responsiveness to ß-arrestin2 in intact cells. More refined in vitro analyses showed that this site was robustly phosphorylated by the acidotropic protein kinase CK2, whereas other kinases, such as CK1 or the GPCR kinase GRK2, were considerably less potent. Simultaneous mutation of five Ser/Thr residues within 702-714 to Ala ((702)ST/AA(714)) abolished phosphorylation and binding of ß-arrestin2. In transfected cells, the CK2 catalytic α subunit formed a complex with NHE5 and decreased wild-type but not (702)ST/AA(714) NHE5 activity, further supporting a regulatory role for this kinase. The rate of internalization of (702)ST/AA(714) was also diminished and relatively insensitive to overexpression of ß-arrestin2. However, unlike in vitro, this mutant retained its ability to form a complex with ß-arrestin2 despite its lack of responsiveness. Additional mutations of two di-isoleucine-based motifs (I697A/L698A and I722A/I723A) that immediately flank the acidic cluster, either separately or together, were required to disrupt their association. These data demonstrate that discrete elements of an elaborate sorting signal in NHE5 contribute to ß-arrestin2 binding and trafficking along the recycling endosomal pathway.


Subject(s)
Arrestins/metabolism , Endocytosis/physiology , Endosomes/metabolism , Protein Sorting Signals/physiology , Sodium-Hydrogen Exchangers/metabolism , Amino Acid Motifs , Amino Acid Substitution , Arrestins/genetics , Casein Kinase I/genetics , Casein Kinase I/metabolism , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Line , Endosomes/genetics , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , Humans , Mutation, Missense , Protein Binding/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Protein Transport/physiology , Sodium-Hydrogen Exchangers/genetics , beta-Arrestins
20.
Front Physiol ; 13: 892196, 2022.
Article in English | MEDLINE | ID: mdl-35547574

ABSTRACT

Endomembrane alkali cation (Na+, K+)/proton (H+) exchangers (eNHEs) are increasingly associated with neurological disorders. These eNHEs play integral roles in regulating the luminal pH, processing, and trafficking of cargo along the secretory (Golgi and post-Golgi vesicles) and endocytic (early, recycling, and late endosomes) pathways, essential regulatory processes vital for neuronal development and plasticity. Given the complex morphology and compartmentalization of multipolar neurons, the contribution of eNHEs in maintaining optimal pH homeostasis and cargo trafficking is especially significant during periods of structural and functional development and remodeling. While the importance of eNHEs has been demonstrated in a variety of non-neuronal cell types, their involvement in neuronal function is less well understood. In this review, we will discuss their emerging roles in excitatory synaptic function, particularly as it pertains to cellular learning and remodeling. We will also explore their connections to neurodevelopmental conditions, including intellectual disability, autism, and attention deficit hyperactivity disorders.

SELECTION OF CITATIONS
SEARCH DETAIL