Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 170(6): 1260-1260.e1, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28886388

RESUMEN

The connexin family of membrane proteins enable gap junction formation and homeostasis, supporting communication between adjacent cells. This SnapShot highlights mutations in different connexins associated with human pathologies and how they affect gap junction function.


Asunto(s)
Conexinas/metabolismo , Enfermedad/genética , Conexinas/química , Conexinas/genética , Uniones Comunicantes , Humanos , Mutación
2.
J Cell Sci ; 137(7)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38533727

RESUMEN

Connexins are channel-forming proteins that function to facilitate gap junctional intercellular communication. Here, we use dual cell voltage clamp and dye transfer studies to corroborate past findings showing that Cx31.1 (encoded by GJB5) is defective in gap junction channel formation, illustrating that Cx31.1 alone does not form functional gap junction channels in connexin-deficient mammalian cells. Rather Cx31.1 transiently localizes to the secretory pathway with a subpopulation reaching the cell surface, which is rarely seen in puncta reminiscent of gap junctions. Intracellular retained Cx31.1 was subject to degradation as Cx31.1 accumulated in the presence of proteasomal inhibition, had a faster turnover when Cx43 was present and ultimately reached lysosomes. Although intracellularly retained Cx31.1 was found to interact with Cx43, this interaction did not rescue its delivery to the cell surface. Conversely, the co-expression of Cx31 dramatically rescued the assembly of Cx31.1 into gap junctions where gap junction-mediated dye transfer was enhanced. Collectively, our results indicate that the localization and functional status of Cx31.1 is altered through selective interplay with co-expressed connexins, perhaps suggesting Cx31.1 is a key regulator of intercellular signaling in keratinocytes.


Asunto(s)
Conexinas , Animales , Comunicación Celular/fisiología , Conexina 43/genética , Conexina 43/metabolismo , Conexinas/genética , Conexinas/metabolismo , Uniones Comunicantes/metabolismo , Canales Iónicos/metabolismo , Queratinocitos/metabolismo , Mamíferos/metabolismo , Humanos
3.
J Biol Chem ; 299(11): 105263, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37734551

RESUMEN

Over 35 years ago the cell biology community was introduced to connexins as the subunit employed to assemble semicrystalline clusters of intercellular channels that had been well described morphologically as gap junctions. The decade that followed would see knowledge of the unexpectedly large 21-member human connexin family grow to reflect unique and overlapping expression patterns in all organ systems. While connexin biology initially focused on their role in constructing highly regulated intercellular channels, this was destined to change as discoveries revealed that connexin hemichannels at the cell surface had novel roles in many cell types, especially when considering connexin pathologies. Acceptance of connexins as having bifunctional channel properties was initially met with some resistance, which has given way in recent years to the premise that connexins have multifunctional properties. Depending on the connexin isoform and cell of origin, connexins have wide-ranging half-lives that vary from a couple of hours to the life expectancy of the cell. Diversity in connexin channel characteristics and molecular properties were further revealed by X-ray crystallography and single-particle cryo-EM. New avenues have seen connexins or connexin fragments playing roles in cell adhesion, tunneling nanotubes, extracellular vesicles, mitochondrial membranes, transcription regulation, and in other emerging cellular functions. These discoveries were largely linked to Cx43, which is prominent in most human organs. Here, we will review the evolution of knowledge on connexin expression in human adults and more recent evidence linking connexins to a highly diverse array of cellular functions.


Asunto(s)
Conexinas , Uniones Comunicantes , Humanos , Biología , Membrana Celular/metabolismo , Conexina 26/metabolismo , Conexinas/metabolismo , Uniones Comunicantes/metabolismo , Animales
4.
Int J Mol Sci ; 23(1)2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-35008913

RESUMEN

Although inherited GJA1 (encoding Cx43) gene mutations most often lead to oculodentodigital dysplasia and related disorders, four variants have been linked to erythrokeratodermia variabilis et progressiva (EKVP), a skin disorder characterized by erythematous and hyperkeratotic lesions. While two autosomal-dominant EKVP-linked GJA1 mutations have been shown to lead to augmented hemichannels, the consequence(s) of keratinocytes harboring a de novo P283L variant alone or in combination with a de novo T290N variant remain unknown. Interestingly, these variants reside within or adjacent to a carboxy terminus polypeptide motif that has been shown to be important in regulating the internalization and degradation of Cx43. Cx43-rich rat epidermal keratinocytes (REKs) or Cx43-ablated REKs engineered to express fluorescent protein-tagged P283L and/or T290N variants formed prototypical gap junctions at cell-cell interfaces similar to wildtype Cx43. Dye coupling and dye uptake studies further revealed that each variant or a combination of both variants formed functional gap junction channels, with no evidence of augmented hemichannel function or induction of cell death. Tracking the fate of EKVP-associated variants in the presence of the protein secretion blocker brefeldin A, or an inhibitor of protein synthesis cycloheximide, revealed that P283L or the combination of P283L and T290N variants either significantly extended Cx43 residency on the cell surface of keratinocytes or delayed its degradation. However, caution is needed in concluding that this modest change in the Cx43 life cycle is sufficient to cause EKVP, or whether an additional underlying mechanism or another unidentified gene mutation is contributing to the pathogenesis found in patients. This question will be resolved if further patients are identified where whole exome sequencing reveals a Cx43 P283L variant alone or, in combination with a T290N variant, co-segregates with EKVP across several family generations.


Asunto(s)
Conexina 43/química , Conexina 43/genética , Eritroqueratodermia Variable/genética , Mutación/genética , Animales , Colorantes , Retículo Endoplásmico/metabolismo , Uniones Comunicantes/metabolismo , Células HeLa , Humanos , Proteínas Mutantes/metabolismo , Proteolisis , Ratas
5.
Dev Dyn ; 250(12): 1810-1827, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34091987

RESUMEN

BACKGROUND: We compared skull shape and variation among genetically modified mice that exhibit different levels of connexin43 (Cx43) channel function, to determine whether Cx43 contributes to craniofacial phenotypic robustness. Specifically, we used two heterozygous mutant mouse models (G60S/+ and I130T/+) that, when compared to their wildtype counterparts, have an ~80% and ~50% reduction in Cx43 function, respectively. RESULTS: Both mutant strains showed significant differences in skull shape compared to wildtype littermates and while these differences were more severe in the G60S/+ mouse, shape differences were localized to similar regions of the skull in both mutants. However, increased skull shape variation was observed in G60S/+ mutants only. Additionally, covariation of skull structures was disrupted in the G60S/+ mutants only, indicating that while a 50% reduction in Cx43 function is sufficient to cause a shift in mean skull shape, the threshold for Cx43 function for disrupting craniofacial phenotypic robustness is lower. CONCLUSIONS: Collectively, our results indicate Cx43 can contribute to phenotypic robustness of the skull through a nonlinear relationship between Cx43 gap junctional function and phenotypic outcomes.


Asunto(s)
Conexina 43/fisiología , Dureza/fisiología , Cráneo/fisiología , Animales , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/patología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Tamaño de los Órganos/genética , Fenotipo , Embarazo , Cráneo/anatomía & histología , Cráneo/diagnóstico por imagen
6.
J Cell Sci ; 132(2)2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30559251

RESUMEN

Mutations in the genes that encode the gap junction proteins connexin 26 (Cx26, encoded by GJB2) and Cx30 (GJB6) are the leading cause of hereditary hearing loss. That said, the Cx30 p.Ala88Val (A88V) mutant causes Clouston syndrome, but not hearing loss. Here, we report that the Cx30-A88V mutant, despite being toxic to inner ear-derived HEI-OC1 cells, conferred remarkable long-term protection against age-related high frequency hearing loss in Cx30A88V/A88V mice. During early development, there were no overt structural differences in the cochlea between genotypes, including a normal complement of hair cells; however, the supporting cell Cx30 gap junction plaques in mutant mice were reduced in size. In adulthood, Cx30A88V/A88V mutant mice had a reduction of cochlear Cx30 mRNA and protein, yet a full complement of hair cells. Conversely, the age-related high frequency hearing loss in Cx30+/+ and Cx30+/A88V mice was due to extensive loss of outer hair cells. Our data suggest that the Cx30-A88V mutant confers long-term hearing protection and prevention of hair cell death, possibly via a feedback mechanism that leads to the reduction of total Cx30 gap junction expression in the cochlea.


Asunto(s)
Cóclea/metabolismo , Conexina 30/metabolismo , Uniones Comunicantes/metabolismo , Regulación de la Expresión Génica , Pérdida Auditiva/prevención & control , Mutación Missense , Sustitución de Aminoácidos , Animales , Línea Celular , Conexina 30/genética , Uniones Comunicantes/genética , Pérdida Auditiva/genética , Pérdida Auditiva/metabolismo , Ratones , Ratones Mutantes
7.
Stem Cells ; 38(1): 52-66, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31646713

RESUMEN

Characterization of the pluripotent "ground state" has led to a greater understanding of species-specific stem cell differences and has imparted an appreciation of the pluripotency continuum that exists in stem cells in vitro. Pluripotent stem cells are functionally coupled via connexins that serve in gap junctional intercellular communication (GJIC) and here we report that the level of connexin expression in pluripotent stem cells depends upon the state in which stem cells exist in vitro. Human and mouse pluripotent stem cells stabilized in a developmentally primitive or "naïve" state exhibit significantly less connexin expression compared with stem cells which are "primed" for differentiation. This dynamic connexin expression pattern may be governed, in part, by differential regulation by pluripotency transcription factors expressed in each cell state. Species-specific differences do exist, however, with mouse stem cells expressing several additional connexin transcripts not found in human pluripotent stem cells. Moreover, pharmacological inhibition of GJIC shows limited impact on naïve human stem cell survival, self-renewal, and pluripotency but plays a more significant role in primed human pluripotent stem cells. However, CRISPR-Cas9 gene ablation of Cx43 in human and mouse primed and naïve pluripotent stem cells reveals that Cx43 is dispensable in each of these four pluripotent stem cell types.


Asunto(s)
Conexinas/metabolismo , Células Madre Pluripotentes/metabolismo , Animales , Comunicación Celular , Diferenciación Celular , Humanos , Ratones
8.
J Cell Sci ; 131(9)2018 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-29618634

RESUMEN

Given the importance of connexin43 (Cx43, encoded by GJA1) function in the central nervous system and sensory organ processing, we proposed that it would also be crucial in auditory function. To that end, hearing was examined in two mouse models of oculodentodigital dysplasia that globally express GJA1 mutations resulting in mild or severe loss of Cx43 function. Although Cx43I130T/+ mutant mice, with ∼50% Cx43 channel function, did not have any hearing loss, Cx43G60S/+ mutant mice, with ∼20% Cx43 channel function, had severe hearing loss. There was no evidence of inner ear sensory hair cell loss, suggesting that the mechanism for Cx43-linked hearing loss lies downstream in the auditory pathway. Since evidence suggests that Cx26 function is essential for hearing and may be protective against noise-induced hearing loss, we challenged Cx43I130T/+ mice with a loud noise and found that they had a similar susceptibility to noise-induced hearing loss to that found in controls, suggesting that decreased Cx43 function does not sensitize the mice for environmentally induced hearing loss. Taken together, this study suggests that Cx43 plays an important role in baseline hearing and is essential for auditory processing.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Conexina 43/genética , Anomalías Craneofaciales/complicaciones , Anomalías Craneofaciales/genética , Anomalías del Ojo/complicaciones , Anomalías del Ojo/genética , Deformidades Congénitas del Pie/complicaciones , Deformidades Congénitas del Pie/genética , Pérdida Auditiva/etiología , Pérdida Auditiva/genética , Mutación , Sindactilia/complicaciones , Sindactilia/genética , Anomalías Dentarias/complicaciones , Anomalías Dentarias/genética , Animales , Tronco Encefálico/metabolismo , Tronco Encefálico/patología , Cóclea/metabolismo , Cóclea/patología , Conexina 43/metabolismo , Anomalías Craneofaciales/metabolismo , Anomalías Craneofaciales/patología , Modelos Animales de Enfermedad , Anomalías del Ojo/metabolismo , Anomalías del Ojo/patología , Deformidades Congénitas del Pie/metabolismo , Deformidades Congénitas del Pie/patología , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/patología , Pérdida Auditiva/patología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sindactilia/metabolismo , Sindactilia/patología , Anomalías Dentarias/metabolismo , Anomalías Dentarias/patología
9.
Calcif Tissue Int ; 107(6): 611-624, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32902679

RESUMEN

Mutations in the gene encoding the gap-junctional protein connexin43 (Cx43) are the cause of the human disease oculodentodigital dysplasia (ODDD). The mandible is often affected in this disease, with clinical reports describing both mandibular overgrowth and conversely, retrognathia. These seemingly opposing observations underscore our relative lack of understanding of how ODDD affects mandibular morphology. Using two mutant mouse models that mimic the ODDD phenotype (I130T/+ and G60S/+), we sought to uncover how altered Cx43 function may affect mandibular development. Specifically, mandibles of newborn mice were imaged using micro-CT, to enable statistical comparisons of shape. Tissue-level comparisons of key regions of the mandible were conducted using histomorphology, and we quantified the mRNA expression of several cartilage and bone cell differentiation markers. Both G60S/+ and I130T/+ mutant mice had altered mandibular morphology compared to their wildtype counterparts, and the morphological effects were similarly localized for both mutants. Specifically, the biggest phenotypic differences in mutant mice were focused in regions exposed to mechanical forces, such as alveolar bone, muscular attachment sites, and articular surfaces. Histological analyses revealed differences in ossification of the intramembranous bone of the mandibles of both mutant mice compared to their wildtype littermates. However, chondrocyte organization within the secondary cartilages of the mandible was unaffected in the mutant mice. Overall, our results suggest that the morphological differences seen in G60S/+ and I130T/+ mouse mandibles are due to delayed ossification and suggest that mechanical forces may exacerbate the effects of ODDD on the skeleton.


Asunto(s)
Conexina 43 , Anomalías Craneofaciales/patología , Anomalías del Ojo/patología , Deformidades Congénitas del Pie/patología , Mandíbula/patología , Osteogénesis , Sindactilia/patología , Anomalías Dentarias/patología , Animales , Conexina 43/metabolismo , Uniones Comunicantes , Ratones
10.
Exp Cell Res ; 381(2): 235-247, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31102595

RESUMEN

The channel-forming membrane glycoprotein pannexin 1 (Panx1) is best characterized as an ATP release channel. To investigate the trafficking and sorting of Panx1, we used polarized MDCK cells and non-polarized BICR-M1Rk cells to track the fate of GFP-tagged Panx1. In non-polarized cells, Panx1 was found throughout the plasma membrane, including the lamellipodia of the tumor cells and the cell surface-targeting domain was mapped to residues 307-379. Panx1 was preferentially enriched at the apical membrane domain of polarized MDCK cells grown as monolayer sheets or as spheroids. Residual Panx1 localized within basolateral membranes of polarized MDCK cells was independent of a putative dileucine sorting motif LL365/6 found within the C-terminal of Panx1. Unexpectedly, stable expression of a Panx1 mutant, where a putative tyrosine-based basolateral sorting motif (YxxØ) was mutated (Y308F), or a truncated Δ379 Panx1 mutant, caused MDCK cells to lose cell-cell contacts and their ability to polarize as they underwent a switch to a more fibroblast-like phenotype. We conclude that Panx1 is preferentially delivered to the apical domain of polarized epithelial cells, and Panx1 mutants drive phenotypic changes to MDCK cells preventing their polarization.


Asunto(s)
Polaridad Celular , Conexinas/metabolismo , Riñón/citología , Proteínas Mutantes/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Membrana Celular/metabolismo , Polaridad Celular/genética , Conexinas/genética , Perros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Riñón/metabolismo , Células de Riñón Canino Madin Darby , Proteínas Mutantes/genética , Proteínas del Tejido Nervioso/genética , Transporte de Proteínas , Transfección
11.
Pharmacol Rev ; 69(4): 396-478, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28931622

RESUMEN

Connexins are ubiquitous channel forming proteins that assemble as plasma membrane hemichannels and as intercellular gap junction channels that directly connect cells. In the heart, gap junction channels electrically connect myocytes and specialized conductive tissues to coordinate the atrial and ventricular contraction/relaxation cycles and pump function. In blood vessels, these channels facilitate long-distance endothelial cell communication, synchronize smooth muscle cell contraction, and support endothelial-smooth muscle cell communication. In the central nervous system they form cellular syncytia and coordinate neural function. Gap junction channels are normally open and hemichannels are normally closed, but pathologic conditions may restrict gap junction communication and promote hemichannel opening, thereby disturbing a delicate cellular communication balance. Until recently, most connexin-targeting agents exhibited little specificity and several off-target effects. Recent work with peptide-based approaches has demonstrated improved specificity and opened avenues for a more rational approach toward independently modulating the function of gap junctions and hemichannels. We here review the role of connexins and their channels in cardiovascular and neurovascular health and disease, focusing on crucial regulatory aspects and identification of potential targets to modify their function. We conclude that peptide-based investigations have raised several new opportunities for interfering with connexins and their channels that may soon allow preservation of gap junction communication, inhibition of hemichannel opening, and mitigation of inflammatory signaling.


Asunto(s)
Enfermedades Cardiovasculares/fisiopatología , Conexinas/antagonistas & inhibidores , Conexinas/fisiología , Enfermedades del Sistema Nervioso/fisiopatología , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Fenómenos Fisiológicos Cardiovasculares/efectos de los fármacos , Humanos , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Fenómenos Fisiológicos del Sistema Nervioso/efectos de los fármacos
12.
Semin Cell Dev Biol ; 50: 67-73, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26688333

RESUMEN

Gap junctions consist of clusters of intercellular channels composed of connexins that connect adjacent cells and allow the exchange of small molecules. While the 21 member multi-gene family of connexins are ubiquitously found in humans, only Cx39, Cx40, Cx43 and Cx45 have been documented in developing myoblasts and injured adult skeletal muscle while healthy adult skeletal muscle is devoid of connexins. The use of gap junctional blockers and cultured myoblast cell lines have suggested that these connexins play a critical role in myotube formation and muscle regeneration. More recent genetically-modified mouse models where Cx43 function is greatly compromized or ablated have further supported a role for Cx43 in regulating skeletal muscle development. In the last decade, we have become aware of a cohort of patients that have a development disorder known as oculodentodigital dysplasia (ODDD). These patients harbor either gain or loss of Cx43 function gene mutations that result in many organ anomalies raising questions as to whether they suffer from defects in skeletal muscle formation or regeneration upon injury. Interesting, some ODDD patients report muscle weakness and loss of limb control but it is not clear if this is neurogenic or myogenic in origin. This review will focus on the role connexins play in muscle development and repair and discuss the impact of Cx43 mutants on muscle function.


Asunto(s)
Conexinas/metabolismo , Desarrollo de Músculos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Enfermedades Musculares/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Cicatrización de Heridas
13.
J Biol Chem ; 292(23): 9721-9732, 2017 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-28428247

RESUMEN

Connexin26 (Cx26) is a gap junction protein that oligomerizes in the cell to form hexameric transmembrane channels called connexons. Cell surface connexons dock between adjacent cells to allow for gap junctional intercellular communication. Numerous autosomal dominant mutations in the Cx26-encoding GJB2 gene lead to many skin disorders and sensorineural hearing loss. Although some insights have been gained into the pathogenesis of these diseases, it is not fully understood how distinct GJB2 mutations result in hearing loss alone or in skin pathologies with comorbid hearing loss. Here we investigated five autosomal dominant Cx26 mutants (N14K, D50N, N54K, M163V, and S183F) linked to various syndromic or nonsyndromic diseases to uncover the molecular mechanisms underpinning these disease links. We demonstrated that when gap junction-deficient HeLa cells expressed the N14K and D50N mutants, they undergo cell death. The N54K mutant was retained primarily within intracellular compartments and displayed dominant or transdominant properties on wild-type Cx26 and coexpressed Cx30 and Cx43. The S183F mutant formed some gap junction plaques but was largely retained within the cell and exhibited only a mild transdominant reduction in gap junction communication when co-expressed with Cx30. The M163V mutant, which causes only hearing loss, exhibited impaired gap junction function and showed no transdominant interactions. These findings suggest that Cx26 mutants that promote cell death or exert transdominant effects on other connexins in keratinocytes will lead to skin diseases and hearing loss, whereas mutants having reduced channel function but exhibiting no aberrant effects on coexpressed connexins cause only hearing loss. Moreover, cell death-inducing GJB2 mutations lead to more severe syndromic disease.


Asunto(s)
Conexinas , Uniones Comunicantes , Pérdida Auditiva , Queratinocitos/metabolismo , Mutación Missense , Enfermedades Cutáneas Genéticas , Sustitución de Aminoácidos , Muerte Celular/genética , Conexina 26 , Conexina 30 , Conexina 43/genética , Conexina 43/metabolismo , Conexinas/genética , Conexinas/metabolismo , Uniones Comunicantes/genética , Uniones Comunicantes/metabolismo , Células HeLa , Pérdida Auditiva/genética , Pérdida Auditiva/metabolismo , Humanos , Enfermedades Cutáneas Genéticas/genética , Enfermedades Cutáneas Genéticas/metabolismo
14.
Biochim Biophys Acta Biomembr ; 1860(1): 237-243, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28655619

RESUMEN

This article is a report of the "International Colloquium on Gap junctions: 50Years of Impact on Cancer" that was held 8-9 September 2016, at the Amphitheater "Pôle Biologie Santé" of the University of Poitiers (Poitiers, France). The colloquium was organized by M Mesnil (Université de Poitiers, Poitiers, France) and C Naus (University of British Columbia, Vancouver, Canada) to celebrate the 50th anniversary of the seminal work published in 1966 by Loewenstein and Kanno [Intercellular communication and the control of tissue growth: lack of communication between cancer cells, Nature, 116 (1966) 1248-1249] which initiated studies on the involvement of gap junctions in carcinogenesis. During the colloquium, 15 participants presented reviews or research updates in the field which are summarized below.


Asunto(s)
Uniones Comunicantes/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Animales , Uniones Comunicantes/genética , Uniones Comunicantes/patología , Humanos , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patología
15.
J Biol Chem ; 291(24): 12432-12443, 2016 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-27129271

RESUMEN

Pannexin1 (PANX1) is probably best understood as an ATP release channel involved in paracrine signaling. Given its ubiquitous expression, PANX1 pathogenic variants would be expected to lead to disorders involving multiple organ systems. Using whole exome sequencing, we discovered the first patient with a homozygous PANX1 variant (c.650G→A) resulting in an arginine to histidine substitution at position 217 (p.Arg217His). The 17-year-old female has intellectual disability, sensorineural hearing loss requiring bilateral cochlear implants, skeletal defects, including kyphoscoliosis, and primary ovarian failure. Her consanguineous parents are each heterozygous for this variant but are not affected by the multiorgan syndromes noted in the proband. Expression of the p.Arg217His mutant in HeLa, N2A, HEK293T, and Ad293 cells revealed normal PANX1 glycosylation and cell surface trafficking. Dye uptake, ATP release, and electrophysiological measurements revealed p.Arg217His to be a loss-of-function variant. Co-expression of the mutant with wild-type PANX1 suggested the mutant was not dominant-negative to PANX1 channel function. Collectively, we demonstrate a PANX1 missense change associated with human disease in the first report of a "PANX1-related disorder."


Asunto(s)
Anomalías Múltiples/genética , Conexinas/genética , Mutación de Línea Germinal , Proteínas del Tejido Nervioso/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Adenosina Trifosfato/metabolismo , Adolescente , Animales , Línea Celular Tumoral , Conexinas/metabolismo , Consanguinidad , Salud de la Familia , Femenino , Células HEK293 , Células HeLa , Pérdida Auditiva Sensorineural/patología , Heterocigoto , Homocigoto , Humanos , Cifosis/patología , Masculino , Mutación Missense , Proteínas del Tejido Nervioso/metabolismo , Linaje , Insuficiencia Ovárica Primaria/patología , Escoliosis/patología , Síndrome
16.
J Cell Sci ; 128(21): 3947-60, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26359304

RESUMEN

In the present study we investigated the life cycle, trafficking, assembly and cell surface dynamics of a poorly characterized connexin family member, connexin 30 (Cx30; also known as GJB6), which plays a critical role in skin health and hearing. Unexpectedly, Cx30 localization at the cell surface and gap junctional intercellular communication was not affected by prolonged treatments with the endoplasmic reticulum (ER)-Golgi transport inhibitor brefeldin A or the protein synthesis inhibitor cycloheximide, whereas Cx43 (also known as GJA1) was rapidly cleared. Fluorescent recovery after photobleaching revealed that Cx30 plaques were rebuilt from the outer edges in keeping with older channels residing in the inner core of the plaque. Expression of a dominant-negative form of Sar1 GTPase led to the accumulation of Cx30 within the ER, in contrast to a report that Cx30 traffics via a Golgi-independent pathway. Co-expression of Cx30 with Cx43 revealed that these connexins segregate into distinct domains within common gap junction plaques, suggesting that their assembly is governed by different mechanisms. In summary, Cx30 was found to be an unusually stable, long-lived connexin (half-life >12 h), which may underlie its specific role in the epidermis and cochlea.


Asunto(s)
Conexinas/metabolismo , Uniones Comunicantes/metabolismo , Animales , Conexina 26 , Conexina 30 , Conexina 43/genética , Conexina 43/metabolismo , Conexinas/genética , Recuperación de Fluorescencia tras Fotoblanqueo , Células HeLa , Humanos , Queratinocitos/metabolismo , Ratones , Ratas
17.
Biochem J ; 473(24): 4665-4680, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27784763

RESUMEN

Hearing loss, including noise-induced hearing loss, is highly prevalent and severely hinders an individual's quality of life, yet many of the mechanisms that cause hearing loss are unknown. The pannexin (Panx) channel proteins, Panx1 and Panx3, are regionally expressed in many cell types along the auditory pathway, and mice lacking Panx1 in specific cells of the inner ear exhibit hearing loss, suggesting a vital role for Panxs in hearing. We proposed that Panx1 and/or Panx3 null mice would exhibit severe hearing loss and increased susceptibility to noise-induced hearing loss. Using the auditory brainstem response, we surprisingly found that Panx1-/- and Panx3-/- mice did not harbor hearing or cochlear nerve deficits. Furthermore, while Panx1-/- mice displayed no protection against loud noise-induced hearing loss, Panx3-/- mice exhibited enhanced 16- and 24-kHz hearing recovery 7 days after a loud noise exposure (NE; 12 kHz tone, 115 dB sound pressure level, 1 h). Interestingly, Cx26, Cx30, Cx43, and Panx2 were up-regulated in Panx3-/- mice compared with wild-type and/or Panx1-/- mice, and assessment of the auditory tract revealed morphological changes in the middle ear bones of Panx3-/- mice. It is unclear if these changes alone are sufficient to provide protection against loud noise-induced hearing loss. Contrary to what we expected, these data suggest that Panx1 and Panx3 are not essential for baseline hearing in mice tested, but the therapeutic targeting of Panx3 may prove protective against mid-high-frequency hearing loss caused by loud NE.


Asunto(s)
Conexinas/metabolismo , Pérdida Auditiva Provocada por Ruido/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Cóclea/metabolismo , Cóclea/fisiología , Conexina 26 , Conexina 30 , Conexina 43/metabolismo , Conexinas/genética , Potenciales Evocados Auditivos del Tronco Encefálico/genética , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Pérdida Auditiva Provocada por Ruido/genética , Immunoblotting , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
BMC Cell Biol ; 17 Suppl 1: 19, 2016 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-27230612

RESUMEN

Electron micrographs revealed the presence of gap junctions in osteoblastic cells over 40 years ago. These intercellular channels formed from connexins are present in bone forming osteoblasts, bone resorbing osteoclasts, and osteocytes (mature osteoblasts embedded in the mineralized bone matrix). More recently, genetic and pharmacologic studies revealed the role of connexins, and in particular Cx43, in the differentiation and function of all bone types. Furthermore, mutations in the gene encoding Cx43 were found to be causally linked to oculodentodigital dysplasia, a condition that results in an abnormal skeleton. Pannexins, molecules with similar structure and single-membrane channel forming potential as connexins when organized as hemichannels, are also expressed in osteoblastic cells. The function of pannexins in bone and cartilage is beginning to be uncovered, but more research is needed to determine the role of pannexins in bone development, adult bone mass and skeletal homeostasis. We describe here the current knowledge on the role of connexins and pannexins on skeletal health and disease.


Asunto(s)
Regeneración Ósea/fisiología , Huesos/metabolismo , Huesos/patología , Conexinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Conexinas/genética , Modelos Animales de Enfermedad , Humanos , Osteoartritis/metabolismo , Osteoartritis/patología
19.
J Cell Sci ; 127(Pt 8): 1751-64, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24522190

RESUMEN

Connexin 30 (Cx30), a member of the large gap-junction protein family, plays a role in the homeostasis of the epidermis and inner ear through gap junctional intercellular communication (GJIC). Here, we investigate the underlying mechanisms of four autosomal dominant Cx30 gene mutations that are linked to hearing loss and/or various skin diseases. First, the T5M mutant linked to non-syndromic hearing loss formed functional gap junction channels and hemichannels, similar to wild-type Cx30. The loss-of-function V37E mutant associated with Clouston syndrome or keratitis-ichthyosis-deafness syndrome was retained in the endoplasmic reticulum and significantly induced apoptosis. The G59R mutant linked to the Vohwinkel and Bart-Pumphrey syndromes was retained primarily in the Golgi apparatus and exhibited loss of gap junction channel and hemichannel function but did not cause cell death. Lastly, the A88V mutant, which is linked to the development of Clouston syndrome, also significantly induced apoptosis but through an endoplasmic-reticulum-independent mechanism. Collectively, we discovered that four unique Cx30 mutants might cause disease through different mechanisms that also likely include their selective trans-dominant effects on coexpressed connexins, highlighting the overall complexity of connexin-linked diseases and the importance of GJIC in disease prevention.


Asunto(s)
Conexinas/genética , Enfermedades de la Piel/genética , Animales , Apoptosis , Comunicación Celular , Conexina 26 , Conexina 30 , Conexina 43/metabolismo , Conexinas/metabolismo , Sordera/genética , Uniones Comunicantes/metabolismo , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Células HeLa , Humanos , Queratinocitos/metabolismo , Ratones , Mutación Missense , Ratas
20.
J Membr Biol ; 249(6): 713-741, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27586664

RESUMEN

It is now clear that connexin-based, gap junction "hemichannels" in an undocked state are capable of opening and connecting cytoplasm to the extracellular milieu. Varied studies also suggest that such channel activity plays a vital role in diverse cell processes and abnormal hemichannel activity contributes to pathogenesis. To pursue fundamental questions in this area, investigators require methods for studying hemichannel permeability and dynamics that are quantitative, sensitive, versatile, and available to most cellular and molecular laboratories. Here we first provide a theoretical background for this work, including the role of cellular membrane potentials. We then describe in detail our computer-assisted methods for both dye uptake and leakage along with illustrative results from different cell systems. A key feature of our protocol is the inclusion of a mechanical stimulation step. We describe dye uptake, interpreted as connexin dependent, that is shown to be enhanced with reduced extracellular Ca2+, mechanically responsive, inhibited by TPA, inhibited by EL186 antibodies for Cx43 and sustained for more than 15 min following mechanical stimulation. We describe dye leakage that displays these same properties, with estimates of hemichannel numbers per cell being derived from leakage rates. We also describe dye uptake that is shown to be unaffected by a reduction in external Ca2+, insensitive to EL186 antibodies and relatively short-lived following mechanical stimulation; this uptake may occur via pannexin 1 channels expressed in the cells studied here. It is unlikely that cell damage plays a significant role in dye uptake following mechanical stimulation, given compelling results from various control experiments.


Asunto(s)
Conexinas/metabolismo , Algoritmos , Animales , Transporte Biológico , Calcio/metabolismo , Línea Celular , Colorantes/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Conexinas/genética , Uniones Comunicantes/metabolismo , Expresión Génica , Humanos , Cinética , Ratones , Microscopía Fluorescente , Modelos Teóricos , Permeabilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA