Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Sci Data ; 11(1): 416, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38653806

ABSTRACT

Our sense of hearing is mediated by cochlear hair cells, of which there are two types organized in one row of inner hair cells and three rows of outer hair cells. Each cochlea contains 5-15 thousand terminally differentiated hair cells, and their survival is essential for hearing as they do not regenerate after insult. It is often desirable in hearing research to quantify the number of hair cells within cochlear samples, in both pathological conditions, and in response to treatment. Machine learning can be used to automate the quantification process but requires a vast and diverse dataset for effective training. In this study, we present a large collection of annotated cochlear hair-cell datasets, labeled with commonly used hair-cell markers and imaged using various fluorescence microscopy techniques. The collection includes samples from mouse, rat, guinea pig, pig, primate, and human cochlear tissue, from normal conditions and following in-vivo and in-vitro ototoxic drug application. The dataset includes over 107,000 hair cells which have been identified and annotated as either inner or outer hair cells. This dataset is the result of a collaborative effort from multiple laboratories and has been carefully curated to represent a variety of imaging techniques. With suggested usage parameters and a well-described annotation procedure, this collection can facilitate the development of generalizable cochlear hair-cell detection models or serve as a starting point for fine-tuning models for other analysis tasks. By providing this dataset, we aim to give other hearing research groups the opportunity to develop their own tools with which to analyze cochlear imaging data more fully, accurately, and with greater ease.


Subject(s)
Cochlea , Animals , Mice , Guinea Pigs , Humans , Rats , Swine , Hair Cells, Auditory , Microscopy, Fluorescence , Machine Learning
2.
bioRxiv ; 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37693382

ABSTRACT

Our sense of hearing is mediated by cochlear hair cells, localized within the sensory epithelium called the organ of Corti. There are two types of hair cells in the cochlea, which are organized in one row of inner hair cells and three rows of outer hair cells. Each cochlea contains a few thousands of hair cells, and their survival is essential for our perception of sound because they are terminally differentiated and do not regenerate after insult. It is often desirable in hearing research to quantify the number of hair cells within cochlear samples, in both pathological conditions, and in response to treatment. However, the sheer number of cells along the cochlea makes manual quantification impractical. Machine learning can be used to overcome this challenge by automating the quantification process but requires a vast and diverse dataset for effective training. In this study, we present a large collection of annotated cochlear hair-cell datasets, labeled with commonly used hair-cell markers and imaged using various fluorescence microscopy techniques. The collection includes samples from mouse, human, pig and guinea pig cochlear tissue, from normal conditions and following in-vivo and in-vitro ototoxic drug application. The dataset includes over 90'000 hair cells, all of which have been manually identified and annotated as one of two cell types: inner hair cells and outer hair cells. This dataset is the result of a collaborative effort from multiple laboratories and has been carefully curated to represent a variety of imaging techniques. With suggested usage parameters and a well-described annotation procedure, this collection can facilitate the development of generalizable cochlear hair cell detection models or serve as a starting point for fine-tuning models for other analysis tasks. By providing this dataset, we aim to supply other groups within the hearing research community with the opportunity to develop their own tools with which to analyze cochlear imaging data more fully, accurately, and with greater ease.

3.
Sci Rep ; 13(1): 2528, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36781873

ABSTRACT

The mechano-electrical transduction (MET) channel of the inner ear receptor cells, termed hair cells, is a protein complex that enables our senses of hearing and balance. Hair cell MET requires an elaborate interplay of multiple proteins that form the MET channel. One of the MET complex components is the transmembrane protein LHFPL5, which is required for hair cell MET and hearing. LHFPL5 is thought to form a multi-protein complex with other MET channel proteins, such as PCDH15, TMIE, and TMC1. Despite localizing to the plasma membrane of stereocilia, the mechanosensing organelles of hair cells, LHFPL5 requires its binding partner within the MET complex, PCDH15, to localize to the stereocilia tips in hair cells and to the plasma membrane in heterologous cells. Using the Aquaporin 3-tGFP reporter (AGR) for plasma membrane localization, we found that a region within extracellular loop 1, which interacts with PCDH15, precludes the trafficking of AGR reporter to the plasma membrane in heterologous cell lines. Our results suggest that the presence of protein partners may mask endoplasmic reticulum retention regions or enable the proper folding and trafficking of the MET complex components, to facilitate expression of the MET complex at the stereocilia membrane.


Subject(s)
Hair Cells, Auditory , Membrane Proteins , Hair Cells, Auditory/metabolism , Membrane Proteins/metabolism , Stereocilia/metabolism , Cell Membrane/metabolism , Hearing/physiology , Mechanotransduction, Cellular/physiology
4.
Sci Rep ; 12(1): 13764, 2022 08 12.
Article in English | MEDLINE | ID: mdl-35962067

ABSTRACT

During hair cell development, the mechanoelectrical transduction (MET) apparatus is assembled at the stereocilia tips, where it coexists with the stereocilia actin regulatory machinery. While the myosin-based tipward transport of actin regulatory proteins is well studied, isoform complexity and built-in redundancies in the MET apparatus have limited our understanding of how MET components are transported. We used a heterologous expression system to elucidate the myosin selective transport of isoforms of protocadherin 15 (PCDH15), the protein that mechanically gates the MET apparatus. We show that MYO7A selectively transports the CD3 isoform while MYO3A and MYO3B transports the CD2 isoform. Furthermore, MYO15A showed an insignificant role in the transport of PCDH15, and none of the myosins tested transport PCDH15-CD1. Our data suggest an important role for MYO3A, MYO3B, and MYO7A in the MET apparatus formation and highlight the intricate nature of MET and actin regulation during development and functional maturation of the stereocilia bundle.


Subject(s)
Protocadherins , Stereocilia , Actins/metabolism , Myosins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Stereocilia/metabolism
5.
Sci Adv ; 8(31): eabm5550, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35921424

ABSTRACT

The mechanoelectrical transduction (MET) channel in auditory hair cells converts sound into electrical signals, enabling hearing. Transmembrane-like channel 1 and 2 (TMC1 and TMC2) are implicated in forming the pore of the MET channel. Here, we demonstrate that inhibition of MET channels, breakage of the tip links required for MET, or buffering of intracellular Ca... induces pronounced phosphatidylserine externalization, membrane blebbing, and ectosome release at the hair cell sensory organelle, culminating in the loss of TMC1. Membrane homeostasis triggered by MET channel inhibition requires Tmc1 but not Tmc2, and three deafness-causing mutations in Tmc1 cause constitutive phosphatidylserine externalization that correlates with deafness phenotype. Our results suggest that, in addition to forming the pore of the MET channel, TMC1 is a critical regulator of membrane homeostasis in hair cells, and that Tmc1-related hearing loss may involve alterations in membrane homeostasis.


Subject(s)
Deafness , Mechanotransduction, Cellular , Hearing/physiology , Homeostasis , Humans , Mechanotransduction, Cellular/physiology , Membrane Proteins/metabolism , Phosphatidylserines
7.
J Assoc Res Otolaryngol ; 22(6): 601-608, 2021 12.
Article in English | MEDLINE | ID: mdl-34617206

ABSTRACT

Identification of the components of the mechanosensory transduction complex in hair cells has been a major research interest for many auditory and vestibular scientists and has attracted attention from outside the field. The past two decades have witnessed a number of significant advances with emergence of compelling evidence implicating at least a dozen distinct molecular components of the transduction machinery. Yet, how the pieces of this ensemble fit together and function in harmony to enable the senses of hearing and balance has not been clarified. The goal of this review is to summarize a 2021 symposium presented at the annual mid-winter meeting of the Association for Research in Otolaryngology. The symposium brought together the latest insights from within and beyond the field to examine individual components of the transduction complex and how these elements interact at molecular, structural, and biophysical levels to gate mechanosensitive channels and initiate sensory transduction in the inner ear. The review includes a brief historical background to set the stage for topics to follow that focus on structure, properties, and interactions of proteins such as CDH23, PCDH15, LHFPL5, TMIE, TMC1/2, and CIB2/3. We aim to present the diversity of ideas in this field and highlight emerging theories and concepts. This review will not only provide readers with a deeper appreciation of the components of the transduction apparatus and how they function together, but also bring to light areas of broad agreement, areas of scientific controversy, and opportunities for future scientific discovery.


Subject(s)
Hair Cells, Auditory/physiology , Hearing/physiology , Mechanotransduction, Cellular/physiology , Membrane Proteins/metabolism
8.
J Neurosci ; 41(20): 4378-4391, 2021 05 19.
Article in English | MEDLINE | ID: mdl-33824189

ABSTRACT

Transmembrane channel-like protein isoform 1 (TMC1) is a major component of the mechano-electrical transducer (MET) channel in cochlear hair cells and is subject to numerous mutations causing deafness. We report a new dominant human deafness mutation, TMC1 p.T422K, and have characterized the homologous mouse mutant, Tmc1 p.T416K, which caused deafness and outer hair cell (OHC) loss by the fourth postnatal week. MET channels showed decreased Ca2+ permeability and resting open probability, but no change in single-channel conductance or expression. Three adjacent deafness mutations are TMC1 p.L416R, p.G417R, and p.M418K, the last homologous to the mouse Beethoven that exhibits similar channel effects. All substitute a positive for a neutral residue, which could produce charge screening in the channel pore or influence binding of an accessory subunit. Channel properties were compared in mice of both sexes between dominant (Tmc1 p.T416K, Tmc1 p.D569N) and recessive (Tmc1 p.W554L, Tmc1 p.D528N) mutations of residues near the putative pore of the channel. Tmc1 p.W554L and p.D569N exhibit reduced maximum current with no effect on single-channel conductance, implying a smaller number of channels transported to the stereociliary tips; this may stem from impaired TMC1 binding to LHFPL5. Tmc1 p.D528N, located in the pore's narrowest region, uniquely caused large reductions in MET channel conductance and block by dihydrostreptomycin (DHS). For Tmc1 p.T416K and Tmc1 p.D528N, transduction loss occurred between P15 and P20. We propose two mechanisms linking channel mutations and deafness: decreased Ca2+ permeability, common to all mutants, and decreased resting open probability in low Ca2+, confined to dominant mutations.SIGNIFICANCE STATEMENT Transmembrane channel-like protein isoform 1 (TMC1) is thought to be a major component of the mechanotransducer channel in auditory hair cells, but the protein organization and channel structure are still uncertain. We made four mouse lines harboring Tmc1 point mutations that alter channel properties, causing hair cell degeneration and deafness. These include a mouse homolog of a new human deafness mutation pT416K that decreased channel Ca2+ permeability by introducing a positively-charged amino acid in the putative pore. All mutations are consistent with the channel structure predicted from modeling, but only one, p.D528N near the external face of the pore, substantially reduced channel conductance and Ca2+ permeability and virtually abolished block by dihydrostreptomycin (DHS), strongly endorsing its siting within the pore.


Subject(s)
Deafness/genetics , Deafness/metabolism , Hair Cells, Auditory/metabolism , Mechanotransduction, Cellular/genetics , Membrane Proteins/genetics , Adolescent , Adult , Animals , Child , Deafness/pathology , Female , Hair Cells, Auditory/pathology , Humans , Male , Mice , Mice, Mutant Strains , Middle Aged , Pedigree , Point Mutation
9.
Hear Res ; 404: 108212, 2021 05.
Article in English | MEDLINE | ID: mdl-33667877

ABSTRACT

The reporter mT/mG mice expressing a membrane-targeted fluorescent protein are becoming widely used to study the auditory and vestibular system due to its versatility. Here we show that high expression levels of the fluorescent mtdTomato reporter affect the function of the sensory hair cells and the auditory performance of mT/mG transgenic mice. Auditory brainstem responses and distortion product otoacoustic emissions revealed that adult mT/mG homozygous mice are profoundly deaf, whereas heterozygous mice present high frequency loss. We explore whether this line would be useful for studying and visualizing the membrane of auditory hair cells by airyscan super-resolution confocal microscopy. Membrane localization of the reporter was observed in hair cells of the cochlea, facilitating imaging of both cell bodies and stereocilia bundles without altering cellular architecture or the expression of the integral membrane motor protein prestin. Remarkably, hair cells from mT/mG homozygous mice failed to uptake the FM1-43 dye and to locate TMC1 at the stereocilia, indicating defective mechanotransduction machinery. Our work emphasizes that precautions must be considered when working with reporter mice and highlights the potential role of the cellular membrane in maintaining functional hair cells and ensuring proper hearing.


Subject(s)
Deafness , Hair Cells, Auditory , Mechanotransduction, Cellular , Animals , Deafness/genetics , Membrane Proteins/genetics , Mice , Stereocilia , Vestibular System
10.
Hepatología ; 2(1): 236-245, 2021. tab, graf
Article in Spanish | LILACS, COLNAL | ID: biblio-1396563

ABSTRACT

Introducción. La infección por el virus de la hepatitis C (VHC) es un problema de salud pública en el mundo. La Organización Mundial de la Salud (OMS) calcula en más de 70 a 100 millones las personas infectadas. La mayoría de ellas, a lo largo del mundo entero, no han sido diagnosticadas y permanecen sin tratamiento. Objetivo. Determinar la prevalencia de anticuerpos contra el VHC en pacientes con factores de riesgo para la infección, en tres hospitales del departamento de Cundinamarca, Colombia, mediante la prueba rápida SD Bioline en sangre capilar, y la confirmación de la infección por la prueba de ARN-PCR en tiempo real (PCR-RT). Metodología. Estudio de tipo observacional descriptivo donde se incluyeron pacientes adultos atendidos en los servicios de consulta externa de los hospitales de La Mesa, Ubaté y Zipaquirá. Se definieron como factores de riesgo para hepatitis C: antecedente transfusional antes de 1996, cirugías mayores (tórax, abdomen, ortopédicas), tatuajes, piercing. Se empleó como prueba rápida el kit comercial SD Bioline HCV (Standard Diagnostics, INC. Corea). Esta prueba contiene una membrana recubierta con antígenos recombinantes del VHC (core, NS3, NS4, NS5). Resultados. Entre enero y octubre de 2018 se tamizaron 1.856 pacientes, 1.531 mujeres (82%) y 325 (18%) hombres, con edad promedio de 45 años y rango de edad de 18 a 89 años. Los principales factores de riesgo identificados fueron los antecedentes de cirugía y las transfusiones antes de 1996. Se detectaron 2 pacientes mujeres positivas, mayores de 50 años, y como factor de riesgo el haber sido sometidas a cirugía mayor y el antecedente transfusional antes de 1996, confirmadas por la técnica de PCR-RT, lo que da una prevalencia global de la infección del 0,1%, pero del 0,05% en las personas con antecedente quirúrgico, 0,25% entre los mayores de 50 años, y 0,6% en los pacientes con antecedente transfusional antes de 1996. Conclusiones. Este estudio realizado en una población seleccionada por factores de riesgo asociados, mostró una prevalencia global de infección por VHC de 0,1%. Nuestros resultados de tamización con prueba rápida en una población seleccionada con factores de riesgo, sugieren que la estrategia de tamización se debe dirigir a personas mayores de 50 años con antecedente transfusional y cirugía mayor.


Introduction. Infection with hepatitis C virus (HCV) is a worldwide public health problem. The World Health Organization (WHO) estimates that more than 70 to 100 million people are infected. Most of them, throughout the world, have not been diagnosed and remain untreated. Objective. To determine the prevalence of antibodies against HCV in patients with risk factors for infection, in three hospitals in the department of Cundinamarca, Colombia, by means of the rapid SD Bioline test in capillary blood, and the confirmation of infection by real-time PCR (RT-PCR). Methodology. Adult patients treated in the outpatient services of the La Mesa, Ubaté and Zipaquirá hospitals. Risk factors for hepatitis C were defined as: transfusion history before 1996, major surgeries (chest, abdomen, orthopedic), tattoos, and piercing. The commercial SD Bioline HCV kit (Standard Diagnostics, INC. Korea) was used as the rapid test. This kit contains a membrane coated with recombinant HCV antigens (core, NS3, NS4, NS5). Results. Between January and October 2018, 1,856 patients were screened, 1,531 women (82%) and 325 (18%) men, with an average age of 45 years and an age range of 18 to 89 years. The main risk factors identified were a history of surgery and transfusions before 1996. Two positive female patients were detected, older than 50 years, and as a risk factor, having undergone major surgery and a history of transfusion before 1996, confirmed by the RT-PCR assay, which gives a global prevalence of infection of 0.1%, but of 0.05% in people with surgery history, 0.25% among those over 50 years of age, and 0.6% in patients with a transfusion history before 1996. Conclusions. This study, carried out in a population selected for associated risk factors, showed an overall prevalence of HCV infection of 0.1%. Our results suggest that screening with a rapid test in a selected population with risk factors should be directed at people over 50 years of age with a history of transfusion and major surgery.


Subject(s)
Humans , Adult , Middle Aged , Aged , Aged, 80 and over , Mass Screening , Hepatitis C , Risk Factors
11.
Reproduction ; 160(5): 685-694, 2020 11.
Article in English | MEDLINE | ID: mdl-33065543

ABSTRACT

In early equine pregnancy, a highly invasive trophoblast cell subpopulation, the chorionic girdle cells, invade the endometrium and form endometrial cups (EC). These cells express classical MHC molecules, thereby stimulating a humoral and cellular immune response, resulting in a massive accumulation of maternal CD4+ and CD8+ T cells around the EC. Nevertheless, no immediate destruction of endometrial cups by maternal lymphoid cells occurs, presumably due to immune tolerance. Although the environment of EC is rich in TGFB and in FOXP3+, CD4+ T cells, the mechanisms leading to tolerance have not been elucidated. Recently, we discovered that equine trophoblast cells secrete pregnancy-specific glycoproteins (PSGs). Since human and murine PSGs activate latent TGFB, we hypothesized that equine PSGs may have a similar activity. We performed plasmon surface resonance experiments to show that equine PSG CEACAM49 can directly bind to the latency-associated peptide (LAP) of both TGFB1 and TGFB2. We then found that the binding of CEACAM49 leads to the activation of TGFB1 as determined by both ELISA and cell-based assays. Furthermore, the activation of TGFB is a unique function of PSGs within the human CEA family, because CEACAM1, 3, 5, 6, 8 do not activate this cytokine. This finding further strengthens the classification of CEACAM49 as an equine PSG. Based on our results, we hypothesize that activation of latent TGFB in the EC environment by equine PSGs secreted by invasive trophoblast cells, could contribute to the generation of regulatory T cells (Tregs) to maintain immune tolerance.


Subject(s)
Carcinoembryonic Antigen/metabolism , Endometrium/metabolism , Glycoproteins/metabolism , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/metabolism , Trophoblasts/metabolism , Animals , Endometrium/immunology , Endometrium/pathology , Female , Horses , Pregnancy , Transforming Growth Factor beta1/genetics , Trophoblasts/immunology , Trophoblasts/pathology
12.
Reproduction ; 160(5): 737-750, 2020 11.
Article in English | MEDLINE | ID: mdl-33065549

ABSTRACT

We previously reported that binding to heparan sulfate (HS) is required for the ability of the placentally secreted pregnancy-specific glycoprotein 1 (PSG1) to induce endothelial tubulogenesis. PSG1 is composed of four immunoglobulin-like domains but which domains of the protein bind to HS remains unknown. To analyze the interaction of PSG1 with HS, we generated several recombinant proteins, including the individual domains, chimeric proteins between two PSG1 domains, and mutants. Using flow cytometric and surface plasmon resonance studies, we determined that the B2 domain of PSG1 binds to HS and that the positively charged amino acids encompassed between amino acids 43-59 are required for this interaction. Furthermore, we showed that the B2 domain of PSG1 is required for the increase in the formation of tubes by endothelial cells (EC) including a human endometrial EC line and two extravillous trophoblast (EVT) cell lines and for the pro-angiogenic activity of PSG1 observed in an aortic ring assay. PSG1 enhanced the migration of ECs while it increased the expression of matrix metalloproteinase-2 in EVTs, indicating that the pro-angiogenic effect of PSG1 on these two cell types may be mediated by different mechanisms. Despite differences in amino acid sequence, we observed that all human PSGs bound to HS proteoglycans and confirmed that at least two other members of the family, PSG6 and PSG9, induce tube formation. These findings contribute to a better understanding of the pro-angiogenic activity of human PSGs and strongly suggest conservation of this function among all PSG family members.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Endothelial Cells/metabolism , Glycoproteins/metabolism , Neovascularization, Physiologic , Placenta/metabolism , Pregnancy Proteins/metabolism , Trophoblasts/metabolism , Endothelial Cells/cytology , Female , Glycoproteins/genetics , Humans , Placenta/cytology , Pregnancy , Pregnancy Proteins/genetics , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Trophoblasts/cytology
13.
Glycobiology ; 30(11): 895-909, 2020 10 21.
Article in English | MEDLINE | ID: mdl-32280962

ABSTRACT

Pregnancy-specific beta 1 glycoprotein (PSG1) is secreted from trophoblast cells of the human placenta in increasing concentrations as pregnancy progresses, becoming one of the most abundant proteins in maternal serum in the third trimester. PSG1 has seven potential N-linked glycosylation sites across its four domains. We carried out glycomic and glycoproteomic studies to characterize the glycan composition of PSG1 purified from serum of pregnant women and identified the presence of complex N-glycans containing poly LacNAc epitopes with α2,3 sialyation at four sites. Using different techniques, we explored whether PSG1 can bind to galectin-1 (Gal-1) as these two proteins were previously shown to participate in processes required for a successful pregnancy. We confirmed that PSG1 binds to Gal-1 in a carbohydrate-dependent manner with an affinity of the interaction of 0.13 µM. In addition, we determined that out of the three N-glycosylation-carrying domains, only the N and A2 domains of recombinant PSG1 interact with Gal-1. Lastly, we observed that the interaction between PSG1 and Gal-1 protects this lectin from oxidative inactivation and that PSG1 competes the ability of Gal-1 to bind to some but not all of its glycoprotein ligands.


Subject(s)
Galectin 1/metabolism , Polysaccharides/metabolism , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Female , Galectin 1/chemistry , Humans , Ligands , Polysaccharides/chemistry , Pregnancy , Pregnancy-Specific beta 1-Glycoproteins/chemistry , Pregnancy-Specific beta 1-Glycoproteins/isolation & purification
14.
J Vis Exp ; (156)2020 02 08.
Article in English | MEDLINE | ID: mdl-32090986

ABSTRACT

The hair cell mechanotransduction (MET) channel plays an important role in hearing. However, the molecular identity and structural information of MET remain unknown. Electrophysiological studies of hair cells revealed that the MET channel has a large conductance and is permeable to relatively large fluorescent cationic molecules, including some styryl dyes and Texas Red-labeled aminoglycoside antibiotics. In this protocol, we describe a method to visualize and evaluate the uptake of fluorescent dextrans in hair cells of the organ of Corti explants that can be used to assay for functional MET channels. We found that 3 kDa Texas Red-labeled dextran specifically labels functional auditory hair cells after 1-2 h incubation. In particular, 3 kDa dextran labels the two shorter stereocilia rows and accumulates in the cell body in a diffuse pattern when functional MET channels are present. An additional vesicle-like pattern of labeling was observed in the cell body of hair cells and surrounding supporting cells. Our data suggest that 3 kDa Texas-Red dextran can be used to visualize and study two pathways for cellular dye uptake; a hair cell-specific entry route through functional MET channels and endocytosis, a pattern also available to larger dextran.


Subject(s)
Dextrans/pharmacology , Fluorescent Dyes/pharmacology , Hair Cells, Auditory/metabolism , Xanthenes/pharmacology , Animals , Endocytosis , Mechanotransduction, Cellular , Mice, Inbred C57BL , Stereocilia/metabolism
15.
Commun Biol ; 1: 50, 2018.
Article in English | MEDLINE | ID: mdl-30271933

ABSTRACT

Tight junctions consist of a network of sealing strands that create selective ion permeability barriers between adjoining epithelial or endothelial cells. The current model for tight junction strands consists of paired rows of claudins (Cldn) coupled by a cis interface (X-1) derived from crystalline Cldn15. Here we show that tight junction strands exhibit a broad range of lateral bending, indicating diversity in cis interactions. By combining protein-protein docking, coevolutionary analysis, molecular dynamics, and a mutagenesis screen, we identify a new Cldn-Cldn cis interface (Cis-1) that shares interacting residues with X-1 but has an ~ 17° lateral rotation between monomers. In addition, we found that a missense mutation in a Cldn14 that causes deafness and contributes stronger to Cis-1 than to X-1 prevents strand formation in cultured cells. Our results suggest that Cis-1 contributes to the inherent structural flexibility of tight junction strands and is required for maintaining permeability barrier function and hearing.

16.
Mol Hum Reprod ; 24(12): 602-612, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30371828

ABSTRACT

STUDY QUESTION: Do all 10 human pregnancy-specific beta 1-glycoproteins (PSGs) and murine PSG23 activate latent transforming growth factor-ß1 (TGF-ß1)? SUMMARY ANSWER: All human PSGs and murine PSG23 activated latent TGF-ß1. WHAT IS KNOWN ALREADY: Two of the 10 members of the PSG1 family, PSG1 and PSG9, were previously shown to activate the soluble small latent complex of TGF-ß1, a cytokine with potent immune suppressive functions. STUDY DESIGN, SIZE, DURATION: Recombinant PSGs were generated and tested for their ability to activate the small latent complex of TGF-ß1 in a cell-free ELISA-based assay and in a bioassay. In addition, we tested the ability of PSG1 and PSG4 to activate latent TGF-ß bound to the extracellular matrix (ECM) or on the membranes of the Jurkat human T-cell line. PARTICIPANTS/MATERIALS, SETTING, METHODS: Recombinant PSGs were generated by transient transfection and purified with a His-Trap column followed by gel filtration chromatography. The purified PSGs were compared to vehicle (PBS) used as control for their ability to activate the small latent complex of TGF-ß1. The concentration of active TGF-ß was measured in an ELISA using the TGF-ß receptor II as capture and a bioassay using transformed mink epithelial cells that express luciferase in response to active TGF-ß. The specificity of the signal was confirmed using a TGF-ß receptor inhibitor. We also measured the binding kinetics of some human PSGs for the latent-associated peptide (LAP) of TGF-ß using surface plasmon resonance and determined whether PSG1 and PSG4 could activate the large latent complex of TGF-ß1 bound to the ECM and latent TGF-ß1 bound to the cell membrane. All experiments were performed in triplicate wells and repeated three times. MAIN RESULTS AND THE ROLE OF CHANCE: All human PSGs activated the small latent complex of TGF-ß1 (P < 0.05 vs. control) and showed similar affinities (KD) for LAP. Despite the lack of sequence conservation with its human counterparts, the ability to activate latent TGF-ß1 was shared by a member of the murine PSG family. We found that PSG1 and PSG4 activated the latent TGF-ß stored in the ECM (P < 0.01) but did not activate latent TGF-ß1 bound to glycoprotein A repetitions predominant (GARP) on the surface of Jurkat T cells. LIMITATIONS, REASONS FOR CAUTION: The affinity of the interaction of LAP and PSGs was calculated using recombinant proteins, which may differ from the native proteins in their post-translational modifications. We also utilized a truncated form of murine PSG23 rather than the full-length protein. For the studies testing the ability of PSGs to activate membrane-bound TGF-ß1, we utilized the T-cell line Jurkat and Jurkat cells expressing GARP rather than primary T regulatory cells. All the studies were performed in vitro. WIDER IMPLICATIONS OF THE FINDINGS: Here, we show that all human PSGs activate TGF-ß1 and that this function is conserved in at least one member of the rodent PSG family. In vivo PSGs could potentially increase the availability of active TGF-ß1 from the soluble and matrix-bound latent forms of the cytokine contributing to the establishment of a tolerogenic environment during pregnancy. LARGE-SCALE DATA: None. STUDY FUNDING/COMPETING INTEREST(S): The research was supported by a grant from the Collaborative Health Initiative Research Program (CHIRP). No conflicts of interests are declared by the authors.


Subject(s)
Pregnancy-Specific beta 1-Glycoproteins/metabolism , Transforming Growth Factor beta1/metabolism , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix/metabolism , Female , Heparitin Sulfate , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Pregnancy , Pregnancy-Specific beta 1-Glycoproteins/genetics , Transforming Growth Factor beta1/genetics
17.
Elife ; 72018 07 31.
Article in English | MEDLINE | ID: mdl-30063209

ABSTRACT

The hair cell mechanotransduction (MET) channel complex is essential for hearing, yet it's molecular identity and structure remain elusive. The transmembrane channel-like 1 (TMC1) protein localizes to the site of the MET channel, interacts with the tip-link responsible for mechanical gating, and genetic alterations in TMC1 alter MET channel properties and cause deafness, supporting the hypothesis that TMC1 forms the MET channel. We generated a model of TMC1 based on X-ray and cryo-EM structures of TMEM16 proteins, revealing the presence of a large cavity near the protein-lipid interface that also harbors the Beethoven mutation, suggesting that it could function as a permeation pathway. We also find that hair cells are permeable to 3 kDa dextrans, and that dextran permeation requires TMC1/2 proteins and functional MET channels, supporting the presence of a large permeation pathway and the hypothesis that TMC1 is a pore forming subunit of the MET channel complex.


Subject(s)
Anoctamins/chemistry , Deafness/genetics , Mechanotransduction, Cellular/genetics , Membrane Proteins/chemistry , Animals , Anoctamins/genetics , Calcium/metabolism , Deafness/pathology , Dextrans/chemistry , Dextrans/genetics , Hair Cells, Auditory/chemistry , Hair Cells, Auditory/pathology , Hearing/genetics , Humans , Membrane Proteins/genetics , Mice , Mutation , Protein Conformation
18.
Proc Natl Acad Sci U S A ; 115(30): 7648-7650, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29980647

Subject(s)
Cell Membrane , Lipids
19.
Sci Rep ; 8(1): 8706, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29880844

ABSTRACT

Whole-exome sequencing of samples from affected members of two unrelated families with late-onset non-syndromic hearing loss revealed a novel mutation (c.2090 T > G; NM_017433) in MYO3A. The mutation was confirmed in 36 affected individuals, showing autosomal dominant inheritance. The mutation alters a single residue (L697W or p.Leu697Trp) in the motor domain of the stereocilia protein MYO3A, leading to a reduction in ATPase activity, motility, and an increase in actin affinity. MYO3A-L697W showed reduced filopodial actin protrusion initiation in COS7 cells, and a predominant tipward accumulation at filopodia and stereocilia when coexpressed with wild-type MYO3A and espin-1, an actin-regulatory MYO3A cargo. The combined higher actin affinity and duty ratio of the mutant myosin cause increased retention time at stereocilia tips, resulting in the displacement of the wild-type MYO3A protein, which may impact cargo transport, stereocilia length, and mechanotransduction. The dominant negative effect of the altered myosin function explains the dominant inheritance of deafness.


Subject(s)
Genes, Dominant , Genetic Diseases, Inborn/genetics , Hearing Loss/genetics , Mutation, Missense , Myosin Heavy Chains/genetics , Myosin Type III/genetics , Actins/genetics , Actins/metabolism , Adolescent , Adult , Aged , Amino Acid Substitution , Animals , Brazil , COS Cells , Cell Movement/genetics , Child , Chlorocebus aethiops , Female , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Hearing Loss/metabolism , Hearing Loss/pathology , Humans , Male , Middle Aged , Myosin Heavy Chains/metabolism , Myosin Type III/metabolism , Pseudopodia/genetics , Pseudopodia/metabolism , Pseudopodia/pathology , Stereocilia/genetics , Stereocilia/metabolism , Stereocilia/pathology
20.
Hum Vaccin Immunother ; 14(2): 345-360, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29135340

ABSTRACT

Pandemic outbreaks of influenza type A viruses have resulted in numerous fatalities around the globe. Since the conventional influenza vaccines (CIV) provide less than 20% protection for individuals with weak immune system, it has been considered that broadly cross-neutralizing antibodies may provide a better protection. Herein, we showed that a recently generated humanized mouse (DRAGA mouse; HLA-A2. HLA-DR4. Rag1KO. IL-2Rgc KO. NOD) that lacks the murine immune system and expresses a functional human immune system can be used to generate cross-reactive, human anti-influenza monoclonal antibodies (hu-mAb). DRAGA mouse was also found to be suitable for influenza virus infection, as it can clear a sub-lethal infection and sustain a lethal infection with PR8/A/34 influenza virus. The hu-mAbs were designed for targeting a human B-cell epitope (180WGIHHPPNSKEQ QNLY195) of hemagglutinin (HA) envelope protein of PR8/A/34 (H1N1) virus with high homology among seven influenza type A viruses. A single administration of HA180-195 specific hu-mAb in PR8-infected DRAGA mice significantly delayed the lethality by reducing the lung damage. The results demonstrated that DRAGA mouse is a suitable tool to (i) generate heterotype cross-reactive, anti-influenza human monoclonal antibodies, (ii) serve as a humanized mouse model for influenza infection, and (iii) assess the efficacy of anti-influenza antibody-based therapeutics for human use.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Orthomyxoviridae Infections/therapy , Amino Acid Sequence , Animals , Humans , Influenza A virus/classification , Influenza A virus/immunology , Mice , Mice, Knockout , Mice, Transgenic , Models, Molecular , Neutralization Tests , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...