Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 29(1): 97-116, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31691803

RESUMEN

Corneal endothelial cell (CEnC) loss is often associated with blinding endothelial corneal dystrophies: dominantly inherited, common (5%) Fuchs endothelial corneal dystrophy (FECD) and recessive, rare congenital hereditary endothelial dystrophy (CHED). Mutations of SLC4A11, an abundant corneal solute transporter, cause CHED and some cases of FECD. The link between defective SLC4A11 solute transport function and CEnC loss is, however, unclear. Cell adhesion assays using SLC4A11-transfected HEK293 cells and primary human CEnC revealed that SLC4A11 promotes adhesion to components of Descemet's membrane (DM), the basement membrane layer to which CEnC bind. An antibody against SLC4A11 extracellular loop 3 (EL3) suppressed cell adhesion, identifying EL3 as the DM-binding site. Earlier studies showed that some SLC4A11 mutations cause FECD and CHED by impairing solute transport activity or cell surface trafficking. Without affecting these functions, FECD-causing mutations in SLC4A11-EL3 compromised cell adhesion capacity. In an energy-minimized SLC4A11-EL3 three-dimensional model, these mutations cluster and are buried within the EL3 structure. A GST fusion protein of SLC4A11-EL3 interacts with principal DM protein, COL8A2, as identified by mass spectrometry. Engineered SLC4A11-EL3-containing protein, STIC (SLC4A11-EL3 Transmembrane-GPA Integrated Chimera), promotes cell adhesion in transfected HEK293 cells and primary human CEnC, confirming the cell adhesion role of EL3. Taken together, the data suggest that SLC4A11 directly binds DM to serve as a cell adhesion molecule (CAM). These data further suggest that cell adhesion defects contribute to FECD and CHED pathology. Observations with STIC point toward a new therapeutic direction in these diseases: replacement of lost cell adhesion capacity.


Asunto(s)
Proteínas de Transporte de Anión/metabolismo , Antiportadores/metabolismo , Adhesión Celular/fisiología , Distrofias Hereditarias de la Córnea/metabolismo , Proteínas de Transporte de Anión/genética , Antiportadores/genética , Adhesión Celular/genética , Células Cultivadas , Distrofias Hereditarias de la Córnea/genética , Distrofias Hereditarias de la Córnea/patología , Lámina Limitante Posterior/metabolismo , Células HEK293 , Humanos , Mutación/genética
2.
Artículo en Inglés | MEDLINE | ID: mdl-32789790

RESUMEN

The cornea, the eye's outermost layer, protects the eye from the environment. The cornea's innermost layer is an endothelium separating the stromal layer from the aqueous humor. A central role of the endothelium is to maintain stromal hydration state. Defects in maintaining this hydration can impair corneal clarity and thus visual acuity. Two endothelial corneal dystrophies, Fuchs Endothelial Corneal Dystrophy (FECD) and Congenital Hereditary Endothelial Dystrophy (CHED), are blinding corneal diseases with varied clinical presentation in patients across different age demographics. Recessive CHED with an early onset (typically age: 0-3 years) and dominantly inherited FECD with a late onset (age: 40-50 years) have similar phenotypes, although caused by defects in several different genes. A range of molecular mechanisms have been proposed to explain FECD and CHED pathology given the involvement of multiple causative genes. This critical review provides insight into the proposed molecular mechanisms underlying FECD and CHED pathology along with common pathways that may explain the link between the defective gene products and provide a new perspective to view these genetic blinding diseases.


Asunto(s)
Distrofias Hereditarias de la Córnea , Distrofia Endotelial de Fuchs , Adulto , Preescolar , Córnea/patología , Distrofias Hereditarias de la Córnea/genética , Distrofia Endotelial de Fuchs/genética , Humanos , Lactante , Recién Nacido , Persona de Mediana Edad
3.
Value Health ; 25(7): 1148-1156, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35779941

RESUMEN

OBJECTIVES: Value-based healthcare (VBHC) aims at improving patient outcomes while optimizing the use of hospitals' resources among medical personnel, administrations, and support services through an evidence-based, collaborative approach. In this article, we present a blueprint for the implementation of VBHC in hospitals, based on our experience as members of the European University Hospital Alliance. METHODS: The European University Hospital Alliance is a consortium of 9 large hospitals in Europe and aims at increasing the quality and efficiency of care to ultimately drive better outcomes for patients. RESULTS: The blueprint describes how to prepare hospitals for VBHC implementation; analyzes gaps, barriers, and facilitators; and explores the most effective ways to turn patient pathways into a process that results in high-value care. Using a patient-centric approach, we identified 4 core minimum components that must be established as cornerstones and 7 organizational enablers to waive the barriers to implementation and ensure sustainability. CONCLUSION: The blueprint guides through pathway implementation and establishment of key performance indicators in 6 phases, which hospitals can tailor to their current status on their way to implement VBHC.


Asunto(s)
Atención a la Salud , Personal de Salud , Consenso , Europa (Continente) , Hospitales Universitarios , Humanos
4.
Nat Rev Mol Cell Biol ; 11(1): 50-61, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19997129

RESUMEN

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.


Asunto(s)
Orgánulos/metabolismo , Transducción de Señal , ATPasas de Translocación de Protón Vacuolares/metabolismo , Animales , Metabolismo Energético , Homeostasis , Humanos , Concentración de Iones de Hidrógeno , Protones
5.
J Head Trauma Rehabil ; 36(2): 103-113, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32472832

RESUMEN

OBJECTIVE: To create novel Immediate Post-Concussion and Cognitive Testing (ImPACT)-based embedded validity indicators (EVIs) and to compare the classification accuracy to 4 existing EVIImPACT. METHOD: The ImPACT was administered to 82 male varsity football players during preseason baseline cognitive testing. The classification accuracy of existing EVIImPACT was compared with a newly developed index (ImPACT-5A and B). The ImPACT-5A represents the number of cutoffs failed on the 5 ImPACT composite scores at a liberal cutoff (0.85 specificity); ImPACT-5B is the sum of failures on conservative cutoffs (≥0.90 specificity). RESULTS: ImPACT-5A ≥1 was sensitive (0.81), but not specific (0.49) to invalid performance, consistent with EVIImPACT developed by independent researchers (0.68 sensitivity at 0.73-0.75 specificity). Conversely, ImPACT-5B ≥3 was highly specific (0.98), but insensitive (0.22), similar to Default EVIImPACT (0.04 sensitivity at 1.00 specificity). ImPACT-5A ≥3 or ImPACT-5B ≥2 met forensic standards of specificity (0.91-0.93) at 0.33 to 0.37 sensitivity. Also, the ImPACT-5s had the strongest linear relationship with clinically meaningful levels of invalid performance of existing EVIImPACT. CONCLUSIONS: The ImPACT-5s were superior to the standard EVIImPACT and comparable to existing aftermarket EVIImPACT, with the flexibility to optimize the detection model for either sensitivity or specificity. The wide range of ImPACT-5 cutoffs allows for a more nuanced clinical interpretation.


Asunto(s)
Conmoción Encefálica , Fútbol Americano , Humanos , Masculino , Pruebas Neuropsicológicas , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
6.
J Biol Chem ; 294(2): 593-607, 2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30446621

RESUMEN

Monocarboxylate transporters (MCTs) mediate the proton-coupled exchange of high-energy metabolites, including lactate and pyruvate, between cells and tissues. The transport activity of MCT1, MCT2, and MCT4 can be facilitated by the extracellular carbonic anhydrase IV (CAIV) via a noncatalytic mechanism. Combining physiological measurements in HEK-293 cells and Xenopus oocytes with pulldown experiments, we analyzed the direct interaction between CAIV and the two MCT chaperones basigin (CD147) and embigin (GP70). Our results show that facilitation of MCT transport activity requires direct binding of CAIV to the transporters chaperones. We found that this binding is mediated by the highly conserved His-88 residue in CAIV, which is also the central residue of the enzyme's intramolecular proton shuttle, and a charged amino acid residue in the Ig1 domain of the chaperone. Although the position of the CAIV-binding site in the chaperone was conserved, the amino acid residue itself varied among different species. In human CD147, binding of CAIV was mediated by the negatively charged Glu-73 and in rat CD147 by the positively charged Lys-73. In rat GP70, we identified the positively charged Arg-130 as the binding site. Further analysis of the CAIV-binding site revealed that the His-88 in CAIV can either act as H donor or H acceptor for the hydrogen bond, depending on the charge of the binding residue in the chaperone. Our results suggest that the CAIV-mediated increase in MCT transport activity requires direct binding between CAIV-His-88 and a charged amino acid in the extracellular domain of the transporter's chaperone.


Asunto(s)
Basigina/metabolismo , Anhidrasa Carbónica IV/metabolismo , Glicoproteínas/metabolismo , Glicoproteínas de Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Mapas de Interacción de Proteínas , Secuencia de Aminoácidos , Animales , Basigina/química , Células HEK293 , Humanos , Proteínas de la Membrana , Modelos Moleculares , Dominios Proteicos , Ratas , Alineación de Secuencia , Simportadores/metabolismo , Xenopus
7.
Biochem Cell Biol ; 97(3): 290-306, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30462520

RESUMEN

The human solute carrier 26 (SLC26) gene family of anion transporters consists of 10 members (SLC26A1-A11, A10 being a pseudogene) that encode membrane glycoproteins with 14 transmembrane segments and a C-terminal cytoplasmic sulfate transporter anti-sigma antagonist domain. Thus far, mutations in eight members of the SLC26 family (A1-A6, A8, and A9) have been linked to diseases in humans. Our goal is to characterize the role of N-glycosylation and the effect of mutations in SLC26A2 and A3 proteins on their functional expression in transfected HEK-293 cells. We found that certain mutants were retained in the endoplamic reticulum via an interaction with the lectin chaperone calnexin. Some could escape protein quality control and traffic to the cell surface upon removal of the N-glycosylation sites. Furthermore, we found that loss of N-glycosylation reduced expression of SLC26A2 at the cell surface. Loss of N-glycosylation had no effect on the stability of SLC26A3, yet resulted in a profound decrease in transport activity. Thus, N-glycosylation plays three roles in the functional expression of SLC26 proteins: (1) to retain misfolded proteins in the endoplamic reticulum, (2) to stabilize the protein at the cell surface, and (3) to maintain the transport protein in a functional state.


Asunto(s)
Antiportadores de Cloruro-Bicarbonato/metabolismo , Transportadores de Sulfato/metabolismo , Antiportadores de Cloruro-Bicarbonato/química , Antiportadores de Cloruro-Bicarbonato/genética , Retículo Endoplásmico/metabolismo , Glicosilación , Células HEK293 , Humanos , Modelos Moleculares , Mutación , Transportadores de Sulfato/química , Transportadores de Sulfato/genética
8.
J Head Trauma Rehabil ; 34(4): E20-E31, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30499932

RESUMEN

OBJECTIVE: To assess the prevalence of invalid performance on baseline neurocognitive testing using embedded measures within computerized tests and individually administered neuropsychological measures, and to examine the influence of incentive status and performance validity on neuropsychological test scores. SETTING: Sport-related concussion management program at a regionally accredited university. PARTICIPANTS: A total of 83 collegiate football athletes completing their preseason baseline assessment within the University's concussion management program and a control group of 140 nonathlete students. DESIGN: Cross-sectional design based on differential incentive status: motivated to do poorly to return to play more quickly after sustaining a concussion (athletes) versus motivated to do well due to incentivizing performance (students). MAIN MEASURES: Immediate Post-Concussion and Cognitive Testing (ImPACT), performance validity tests, and measures of cognitive ability. RESULTS: Half of the athletes failed at least 1 embedded validity indicator within ImPACT (51.8%), and the traditional neuropsychological tests (49.4%), with large effects for performance validity on cognitive test scores (d: 0.62-1.35), incentive status (athletes vs students; d: 0.36-1.15), and the combination of both factors (d: 1.07-2.20) on measures of attention and processing speed. CONCLUSION: Invalid performance on baseline assessment is common (50%), consistent across instruments (ImPACT or neuropsychological tests) and settings (one-on-one or group administration), increases as a function of incentive status (risk ratios: 1.3-4.0) and results in gross underestimates of the athletes' true ability level, complicating the clinical interpretation of the postinjury evaluation and potentially leading to premature return to play.


Asunto(s)
Traumatismos en Atletas/diagnóstico , Conmoción Encefálica/diagnóstico , Fútbol Americano/lesiones , Pruebas de Estado Mental y Demencia/estadística & datos numéricos , Trastornos Neurocognitivos/diagnóstico , Estudiantes/estadística & datos numéricos , Adolescente , Traumatismos en Atletas/epidemiología , Atención , Conmoción Encefálica/epidemiología , Estudios de Casos y Controles , Estudios Transversales , Humanos , Masculino , Motivación , Trastornos Neurocognitivos/epidemiología , Reproducibilidad de los Resultados , Adulto Joven
9.
Hum Mutat ; 39(5): 676-690, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29327391

RESUMEN

SLC4A11 mutations cause cases of congenital hereditary endothelial dystrophy (CHED), Harboyan syndrome (HS), and Fuchs endothelial corneal dystrophy (FECD). Defective water reabsorption from corneal stroma by corneal endothelial cells (CECs) leads to these corneal dystrophies. SLC4A11, in the CEC basolateral membrane, facilitates transmembrane movement of H2 O, NH3 , and H+ -equivalents. Some SLC4A11 disease mutants have impaired folding, leading to a failure to move to the cell surface, which in some cases can be corrected by the drug, glafenine. To identify SLC4A11 mutants that are targets for folding-correction therapy, we examined 54 SLC4A11 missense mutants. Cell-surface trafficking was assessed on immunoblots, by the level of mature, high molecular weight, cell surface-associated form, and using a bioluminescence resonance energy transfer assay. Low level of cell surface trafficking was found in four out of 18 (20%) of FECD mutants, 19/ out of 31 (61%) of CHED mutants, and three out of five (60%) of HS mutants. Amongst ER-retained mutants, 16 showed increased plasma membrane trafficking when grown at 30°C, suggesting that their defect has potential for rescue. CHED-causing point mutations mostly resulted in folding defects, whereas the majority of FECD missense mutations did not affect trafficking, implying functional impairment. We identified mutations that make patients candidates for folding correction of their corneal dystrophy.


Asunto(s)
Proteínas de Transporte de Anión/genética , Antiportadores/genética , Distrofia Endotelial de Fuchs/genética , Mutación Missense/genética , Medicina de Precisión , Secuencia de Aminoácidos , Animales , Proteínas de Transporte de Anión/química , Antiportadores/química , Membrana Celular/metabolismo , Frío , Perros , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Células de Riñón Canino Madin Darby , Modelos Moleculares , Fenotipo
10.
IUBMB Life ; 70(1): 32-40, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29240292

RESUMEN

Lacking protein synthesis machinery and organelles necessary for autophagy or apoptosis, aged red blood cells (RBCs) are marked by circulating auto-antibodies for macrophage-mediated clearance. The antigen recognized by these auto-antibodies is the major protein of the RBC membrane, Band 3. To ensure regulation and specificity in clearance, the molecular "clock" must mark senescent cells in a way that differentiates them from younger cells, to prevent premature clearance. Predominant models of Band 3 senescence signaling are reviewed, and merits are discussed in light of the recently published crystal structure of the Band 3 membrane domain. © 2017 IUBMB Life, 70(1):32-40, 2018.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/química , Autoanticuerpos/química , Autoantígenos/química , Epítopos/química , Eritrocitos/química , Proteínas Opsoninas/química , Proteína 1 de Intercambio de Anión de Eritrocito/sangre , Autoanticuerpos/sangre , Autoantígenos/sangre , Sitios de Unión de Anticuerpos , Senescencia Celular , Epítopos/sangre , Eritrocitos/citología , Eritrocitos/inmunología , Humanos , Transporte Iónico , Macrófagos/inmunología , Proteínas Opsoninas/sangre , Fagocitosis/fisiología , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Transducción de Señal , Factores de Tiempo
11.
Hum Mutat ; 38(3): 279-288, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27925686

RESUMEN

We studied the structural effects of point mutations of a membrane protein that cause genetic disease. SLC4A11 is a membrane transport protein (OH- /H+ /NH3 /H2 O) of basolateral corneal endothelium, whose mutations cause some cases of congenital hereditary endothelial dystrophy and Fuchs endothelial corneal dystrophy. We created a three-dimensional homology model of SLC4A11 membrane domain, using Band 3 (SLC4A1) crystal structure as template. The homology model was assessed in silico and by analysis of mutants designed on the basis of the model. Catalytic pathway mutants p.Glu675Gln, p.His724Arg, and p.His724Ala impaired SLC4A11 transport. p.Ala720Leu, in a region of extended structure of the proposed translocation pore, failed to mature to the cell surface. p.Gly509Lys, located in an open region at the core domain/gate domain interface, had wild-type level of transport function. The molecular phenotype of 37 corneal dystrophy-causing point mutants was rationalized, based on their location in the homology model. Four map to the substrate translocation pathway, 25 to regions of close transmembrane helix packing, three to the dimeric interface, and five lie in extramembraneous loops. The model provides a view of the spectrum of effects of disease mutations on membrane protein structure and provides a tool to analyze pathogenicity of additional newly discovered SLC4A11 mutants.


Asunto(s)
Proteínas de Transporte de Anión/química , Proteínas de Transporte de Anión/genética , Antiportadores/química , Antiportadores/genética , Distrofias Hereditarias de la Córnea/genética , Modelos Moleculares , Mutación , Conformación Proteica , Alelos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Proteínas de Transporte de Anión/metabolismo , Antiportadores/metabolismo , Transporte Biológico , Catálisis , Secuencia Conservada , Distrofias Hereditarias de la Córnea/metabolismo , Expresión Génica , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Dominios Proteicos , Dominios y Motivos de Interacción de Proteínas/genética , Multimerización de Proteína , Relación Estructura-Actividad
12.
Biochim Biophys Acta ; 1858(7 Pt A): 1507-32, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27058983

RESUMEN

The crystal structure of the dimeric membrane domain of human Band 3(1), the red cell chloride/bicarbonate anion exchanger 1 (AE1, SLC4A1), provides a structural context for over four decades of studies into this historic and important membrane glycoprotein. In this review, we highlight the key structural features responsible for anion binding and translocation and have integrated the following topological markers within the Band 3 structure: blood group antigens, N-glycosylation site, protease cleavage sites, inhibitor and chemical labeling sites, and the results of scanning cysteine and N-glycosylation mutagenesis. Locations of mutations linked to human disease, including those responsible for Southeast Asian ovalocytosis, hereditary stomatocytosis, hereditary spherocytosis, and distal renal tubular acidosis, provide molecular insights into their effect on Band 3 folding. Finally, molecular dynamics simulations of phosphatidylcholine self-assembled around Band 3 provide a view of this membrane protein within a lipid bilayer.


Asunto(s)
Desequilibrio Ácido-Base/sangre , Acidosis Tubular Renal/sangre , Anemia Hemolítica Congénita/sangre , Proteína 1 de Intercambio de Anión de Eritrocito/química , Eliptocitosis Hereditaria/sangre , Errores Innatos del Metabolismo/sangre , Esferocitosis Hereditaria/sangre , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Desequilibrio Ácido-Base/genética , Desequilibrio Ácido-Base/patología , Acidosis Tubular Renal/genética , Acidosis Tubular Renal/patología , Anemia Hemolítica Congénita/genética , Anemia Hemolítica Congénita/patología , Proteína 1 de Intercambio de Anión de Eritrocito/antagonistas & inhibidores , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Bicarbonatos/metabolismo , Eliptocitosis Hereditaria/genética , Eliptocitosis Hereditaria/patología , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Eritrocitos/patología , Eritrocitos Anormales/patología , Expresión Génica , Glicosilación , Humanos , Ligandos , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/patología , Mutación , Unión Proteica , Esferocitosis Hereditaria/genética , Esferocitosis Hereditaria/patología
13.
Am J Physiol Cell Physiol ; 310(2): C161-74, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26582474

RESUMEN

Large cytoplasmic domains (CD) are a common feature among integral membrane proteins. In virtually all cases, these CD have a function (e.g., binding cytoskeleton or regulatory factors) separate from that of the membrane domain (MD). Strong associations between CD and MD are rare. Here we studied SLC4A11, a membrane transport protein of corneal endothelial cells, the mutations of which cause genetic corneal blindness. SLC4A11 has a 41-kDa CD and a 57-kDa integral MD. One disease-causing mutation in the CD, R125H, manifests a catalytic defect, suggesting a role of the CD in transport function. Expressed in HEK-293 cells without the CD, MD-SLC4A11 is retained in the endoplasmic reticulum, indicating a folding defect. Replacement of CD-SLC4A11 with green fluorescent protein did not rescue MD-SLC4A11, suggesting some specific role of CD-SLC4A11. Homology modeling revealed that the structure of CD-SLC4A11 is similar to that of the Cl(-)/HCO3(-) exchange protein AE1 (SLC4A1) CD. Fusion to CD-AE1 partially rescued MD-SLC4A11 to the cell surface, suggesting that the structure of CD-AE1 is similar to that of CD-SLC4A11. The CD-AE1-MD-SLC4a11 chimera, however, had no functional activity. We conclude that CD-SLC4A11 has an indispensable role in the transport function of SLC4A11. CD-SLC4A11 forms insoluble precipitates when expressed in bacteria, suggesting that the domain cannot fold properly when expressed alone. Consistent with a strong association between CD-SLC4A11 and MD-SLC4A11, these domains specifically associate when coexpressed in HEK-293 cells. We conclude that SLC4A11 is a rare integral membrane protein in which the CD has strong associations with the integral MD, which contributes to membrane transport function.


Asunto(s)
Proteínas de Transporte de Anión/química , Proteínas de Transporte de Anión/metabolismo , Antiportadores/química , Antiportadores/metabolismo , Bicarbonatos/química , Bicarbonatos/metabolismo , Citoplasma/química , Citoplasma/metabolismo , Transporte Biológico Activo/fisiología , Células HEK293 , Humanos , Activación del Canal Iónico/fisiología , Estructura Terciaria de Proteína , Relación Estructura-Actividad
14.
Am J Physiol Cell Physiol ; 311(5): C735-C748, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27558157

RESUMEN

SLC4A11, a member of the SLC4 family of bicarbonate transporters, is a widely expressed integral membrane protein, abundant in kidney and cornea. Mutations of SLC4A11 cause some cases of the blinding corneal dystrophies, congenital hereditary endothelial dystrophy, and Fuchs endothelial corneal dystrophy. These diseases are marked by fluid accumulation in the corneal stroma, secondary to defective fluid reabsorption by the corneal endothelium. The role of SLC4A11 in these corneal dystrophies is not firmly established, as SLC4A11 function remains unclear. To clarify the normal function(s) of SLC4A11, we characterized the protein following expression in the simple, low-background expression system Xenopus laevis oocytes. Since plant and fungal SLC4A11 orthologs transport borate, we measured cell swelling associated with accumulation of solute borate. The plant water/borate transporter NIP5;1 manifested borate transport, whereas human SLC4A11 did not. SLC4A11 supported osmotically driven water accumulation that was electroneutral and Na+ independent. Studies in oocytes and HEK293 cells could not detect Na+-coupled HCO3- transport or Cl-/HCO3- exchange by SLC4A11. SLC4A11 mediated electroneutral NH3 transport in oocytes. Voltage-dependent OH- or H+ movement was not measurable in SLC4A11-expressing oocytes, but SLC4A11-expressing HEK293 cells manifested low-level cytosolic acidification at baseline. In mammalian cells, but not oocytes, OH-/H+ conductance may arise when SLC4A11 activates another protein or itself is activated by another protein. These data argue against a role of human SLC4A11 in bicarbonate or borate transport. This work provides additional support for water and ammonia transport by SLC4A11. When expressed in oocytes, SLC4A11 transported NH3, not NH3/H.


Asunto(s)
Proteínas de Transporte de Anión/genética , Proteínas de Transporte de Anión/metabolismo , Antiportadores/genética , Antiportadores/metabolismo , Córnea/metabolismo , Distrofias Hereditarias de la Córnea/genética , Distrofias Hereditarias de la Córnea/metabolismo , Proteínas de la Membrana/metabolismo , Mutación/genética , Animales , Bicarbonatos/metabolismo , Línea Celular , Células HEK293 , Humanos , Transporte Iónico/fisiología , Proteínas de la Membrana/genética , Oocitos/metabolismo , Sodio/metabolismo , Agua/metabolismo , Xenopus laevis/metabolismo
15.
Am J Physiol Renal Physiol ; 309(4): F383-92, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26041446

RESUMEN

Two-thirds of sodium filtered by the renal glomerulus is reabsorbed from the proximal tubule via a sodium/proton exchanger isoform 3 (NHE3)-dependent mechanism. Since sodium and bicarbonate reabsorption are coupled, we postulated that the molecules involved in their reabsorption [NHE3 and carbonic anhydrase II (CAII)] might physically and functionally interact. Consistent with this, CAII and NHE3 were closely associated in a renal proximal tubular cell culture model as revealed by a proximity ligation assay. Direct physical interaction was confirmed in solid-phase binding assays with immobilized CAII and C-terminal NHE3 glutathione-S-transferase fusion constructs. To assess the effect of CAII on NHE3 function, we expressed NHE3 in a proximal tubule cell line and measured NHE3 activity as the rate of intracellular pH recovery, following an acid load. NHE3-expressing cells had a significantly greater rate of intracellular pH recovery than controls. Inhibition of endogenous CAII activity with acetazolamide significantly decreased NHE3 activity, indicating that CAII activates NHE3. To ascertain whether CAII binding per se activates NHE3, we expressed NHE3 with wild-type CAII, a catalytically inactive CAII mutant (CAII-V143Y), or a mutant unable to bind other transporters (CAII-HEX). NHE3 activity increased upon wild-type CAII coexpression, but not in the presence of the CAII V143Y or HEX mutant. Together these studies support an association between CAII and NHE3 that alters the transporter's activity.


Asunto(s)
Bicarbonatos/metabolismo , Anhidrasa Carbónica II/metabolismo , Riñón/enzimología , Intercambiadores de Sodio-Hidrógeno/metabolismo , Sodio/metabolismo , Animales , Anhidrasa Carbónica II/genética , Inhibidores de Anhidrasa Carbónica/farmacología , Línea Celular , Humanos , Concentración de Iones de Hidrógeno , Inmunoprecipitación , Riñón/efectos de los fármacos , Cinética , Mutación , Zarigüeyas , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Proteínas Recombinantes/metabolismo , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/genética , Transfección
16.
Hum Mol Genet ; 22(22): 4579-90, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23813972

RESUMEN

Three genetic corneal dystrophies [congenital hereditary endothelial dystrophy type 2 (CHED2), Harboyan syndrome and Fuchs endothelial corneal dystrophy] arise from mutations of the SLC4a11 gene, which cause blindness from fluid accumulation in the corneal stroma. Selective transmembrane water conductance controls cell size, renal fluid reabsorption and cell division. All known water-channelling proteins belong to the major intrinsic protein family, exemplified by aquaporins (AQPs). Here we identified SLC4A11, a member of the solute carrier family 4 of bicarbonate transporters, as an unexpected addition to known transmembrane water movement facilitators. The rate of osmotic-gradient driven cell-swelling was monitored in Xenopus laevis oocytes and HEK293 cells, expressing human AQP1, NIP5;1 (a water channel protein from plant), hCNT3 (a human nucleoside transporter) and human SLC4A11. hCNT3-expressing cells swelled no faster than control cells, whereas SLC4A11-mediated water permeation at a rate about half that of some AQP proteins. SLC4A11-mediated water movement was: (i) similar to some AQPs in rate; (ii) uncoupled from solute-flux; (iii) inhibited by stilbene disulfonates (classical SLC4 inhibitors); (iv) inactivated in one CHED2 mutant (R125H). Localization of AQP1 and SLC4A11 in human and murine corneal (apical and basolateral, respectively) suggests a cooperative role in mediating trans-endothelial water reabsorption. Slc4a11(-/-) mice manifest corneal oedema and distorted endothelial cells, consistent with loss of a water-flux. Observed water-flux through SLC4A11 extends the repertoire of known water movement pathways and call for a re-examination of explanations for water movement in human tissues.


Asunto(s)
Distrofias Hereditarias de la Córnea/genética , Sustancia Propia/fisiopatología , Proteínas SLC4A/metabolismo , Agua/metabolismo , Animales , Acuaporina 1/metabolismo , Acuaporinas/metabolismo , Córnea/metabolismo , Distrofias Hereditarias de la Córnea/metabolismo , Distrofias Hereditarias de la Córnea/patología , Sustancia Propia/metabolismo , Sustancia Propia/patología , Células HEK293 , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/metabolismo , Pérdida Auditiva Sensorineural/patología , Humanos , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Noqueados , Modelos Animales , Oocitos/metabolismo , Fenotipo , Proteínas SLC4A/genética , Transducción de Señal/genética , Xenopus laevis
18.
Mol Membr Biol ; 31(7-8): 211-27, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25257781

RESUMEN

Anion exchanger 1 (AE1) is a 95 kDa glycoprotein that facilitates Cl(-)=HCO(-)(3) exchange across the erythrocyte plasma membrane. This transport activity resides in the 52 kDa C-terminal membrane domain (Gly(361)-Val(911)) predicted to span the membrane 14 times. To explore the role of tryptophan (Trp) residues in AE1 function, the seven endogenous Trp residues in the membrane domain were mutated individually to alanine (Ala) and phenylalanine (Phe). Expression levels, cell surface abundance, inhibitor binding and transport activities of the mutants were measured upon expression in HEK-293 cells. The seven Trp residues divided into three classes according the impact of mutations on the functional expression of AE1: Class 1, dramatically decreased expression (Trp(492) and Trp(496)); Class 2, decreased expression by Ala substitution but not Phe (Trp(648), Trp(662) and Trp(723)); and Class 3, normal expression (Trp(831) and Trp(848)). The results indicate that Trp residues play differential roles in AE1 expression and function depending on their location in the protein and that Trp mutants with low expression are misfolded and retained in the endoplasmic reticulum.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/química , Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Triptófano/metabolismo , Secuencias de Aminoácidos , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Sitios de Unión , Retículo Endoplásmico/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Pliegue de Proteína , Transporte de Proteínas , Triptófano/genética
19.
Nat Genet ; 38(7): 755-7, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16767101

RESUMEN

Congenital hereditary endothelial dystrophy (CHED) is a heritable, bilateral corneal dystrophy characterized by corneal opacification and nystagmus. We describe seven different mutations in the SLC4A11 gene in ten families with autosomal recessive CHED. Mutations in SLC4A11, which encodes a membrane-bound sodium-borate cotransporter, cause loss of function of the protein either by blocking its membrane targeting or nonsense-mediated decay.


Asunto(s)
Proteínas de Transporte de Anión/genética , Antiportadores/genética , Distrofias Hereditarias de la Córnea/genética , Mutación Puntual , Sustitución de Aminoácidos , Proteínas de Transporte de Anión/metabolismo , Antiportadores/metabolismo , Boratos/metabolismo , Distrofias Hereditarias de la Córnea/metabolismo , Femenino , Distrofia Endotelial de Fuchs/genética , Distrofia Endotelial de Fuchs/metabolismo , Genes Recesivos , Humanos , Masculino
20.
Hum Mutat ; 35(9): 1082-91, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24916015

RESUMEN

SLC4A11 mutations cause some cases of the corneal endothelial dystrophies, congenital hereditary endothelial corneal dystrophy type 2 (CHED2), Harboyan syndrome (HS), and Fuchs endothelial corneal dystrophy (FECD). SLC4A11 protein was recently identified as facilitating water flux across membranes. SLC4A11 point mutations usually cause SLC4A11 misfolding and retention in the endoplasmic reticulum (ER). We set about to test the feasibility of rescuing misfolded SLC4A11 protein to the plasma membrane as a therapeutic approach. Using a transfected HEK293 cell model, we measured functional activity present in cells expressing SLC4A11 variants in combinations representing the state found in CHED2 carriers, affected CHED2, FECD individuals, and unaffected individuals. These cells manifest respectively about 60%, 5%, and 25% of the water flux activity, relative to the unaffected (WT alone). ER-retained CHED2 mutant SLC4A11 protein could be rescued to the plasma membrane, where it conferred 25%-30% of WT water flux level. Further, some ER-retained CHED2 mutants expressed at 30°C supported increased water flux compared with 37°C cultures. Caspase activation and cell vitality assays revealed that expression of SLC4A11 mutants in HEK293 cells does not induce cell death. We conclude that therapeutics able to increase cell surface localization of ER-retained SLC4A11 mutants hold promise to treat CHED2 and FECD patients.


Asunto(s)
Distrofias Hereditarias de la Córnea/genética , Mutación , Proteínas SLC4A/genética , Apoptosis/genética , Caspasa 3/metabolismo , Línea Celular , Membrana Celular/metabolismo , Distrofias Hereditarias de la Córnea/metabolismo , Retículo Endoplásmico/metabolismo , Distrofia Endotelial de Fuchs/genética , Distrofia Endotelial de Fuchs/metabolismo , Expresión Génica , Células HEK293 , Humanos , Pliegue de Proteína , Multimerización de Proteína , Transporte de Proteínas , Deficiencias en la Proteostasis/genética , Proteínas SLC4A/química , Proteínas SLC4A/metabolismo , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA