Статті в журналах з теми "COCHLEAR PROGENITORS"

Щоб переглянути інші типи публікацій з цієї теми, перейдіть за посиланням: COCHLEAR PROGENITORS.

Оформте джерело за APA, MLA, Chicago, Harvard та іншими стилями

Оберіть тип джерела:

Ознайомтеся з топ-50 статей у журналах для дослідження на тему "COCHLEAR PROGENITORS".

Біля кожної праці в переліку літератури доступна кнопка «Додати до бібліографії». Скористайтеся нею – і ми автоматично оформимо бібліографічне посилання на обрану працю в потрібному вам стилі цитування: APA, MLA, «Гарвард», «Чикаго», «Ванкувер» тощо.

Також ви можете завантажити повний текст наукової публікації у форматі «.pdf» та прочитати онлайн анотацію до роботи, якщо відповідні параметри наявні в метаданих.

Переглядайте статті в журналах для різних дисциплін та оформлюйте правильно вашу бібліографію.

1

Udagawa, Tomokatsu, Patrick J. Atkinson, Beatrice Milon, Julia M. Abitbol, Yang Song, Michal Sperber, Elvis Huarcaya Najarro, et al. "Lineage-tracing and translatomic analysis of damage-inducible mitotic cochlear progenitors identifies candidate genes regulating regeneration." PLOS Biology 19, no. 11 (November 10, 2021): e3001445. http://dx.doi.org/10.1371/journal.pbio.3001445.

Повний текст джерела
Анотація:
Cochlear supporting cells (SCs) are glia-like cells critical for hearing function. In the neonatal cochlea, the greater epithelial ridge (GER) is a mitotically quiescent and transient organ, which has been shown to nonmitotically regenerate SCs. Here, we ablated Lgr5+ SCs using Lgr5-DTR mice and found mitotic regeneration of SCs by GER cells in vivo. With lineage tracing, we show that the GER houses progenitor cells that robustly divide and migrate into the organ of Corti to replenish ablated SCs. Regenerated SCs display coordinated calcium transients, markers of the SC subtype inner phalangeal cells, and survive in the mature cochlea. Via RiboTag, RNA-sequencing, and gene clustering algorithms, we reveal 11 distinct gene clusters comprising markers of the quiescent and damaged GER, and damage-responsive genes driving cell migration and mitotic regeneration. Together, our study characterizes GER cells as mitotic progenitors with regenerative potential and unveils their quiescent and damaged translatomes.
Стилі APA, Harvard, Vancouver, ISO та ін.
2

Li, Xiao-Jun, and Angelika Doetzlhofer. "LIN28B/let-7control the ability of neonatal murine auditory supporting cells to generate hair cells through mTOR signaling." Proceedings of the National Academy of Sciences 117, no. 36 (August 21, 2020): 22225–36. http://dx.doi.org/10.1073/pnas.2000417117.

Повний текст джерела
Анотація:
Mechano-sensory hair cells within the inner ear cochlea are essential for the detection of sound. In mammals, cochlear hair cells are only produced during development and their loss, due to disease or trauma, is a leading cause of deafness. In the immature cochlea, prior to the onset of hearing, hair cell loss stimulates neighboring supporting cells to act as hair cell progenitors and produce new hair cells. However, for reasons unknown, such regenerative capacity (plasticity) is lost once supporting cells undergo maturation. Here, we demonstrate that the RNA binding protein LIN28B plays an important role in the production of hair cells by supporting cells and provide evidence that the developmental drop in supporting cell plasticity in the mammalian cochlea is, at least in part, a product of declining LIN28B-mammalian target of rapamycin (mTOR) activity. Employing murine cochlear organoid and explant cultures to model mitotic and nonmitotic mechanisms of hair cell generation, we show that loss of LIN28B function, due to its conditional deletion, or due to overexpression of the antagonistic miRNAlet-7g, suppressed Akt-mTOR complex 1 (mTORC1) activity and renders young, immature supporting cells incapable of generating hair cells. Conversely, we found that LIN28B overexpression increased Akt-mTORC1 activity and allowed supporting cells that were undergoing maturation to de-differentiate into progenitor-like cells and to produce hair cells via mitotic and nonmitotic mechanisms. Finally, using the mTORC1 inhibitor rapamycin, we demonstrate that LIN28B promotes supporting cell plasticity in an mTORC1-dependent manner.
Стилі APA, Harvard, Vancouver, ISO та ін.
3

Munnamalai, Vidhya, Nabilah H. Sammudin, Caryl A. Young, Ankita Thawani, Richard J. Kuhn, and Donna M. Fekete. "Embryonic and Neonatal Mouse Cochleae Are Susceptible to Zika Virus Infection." Viruses 13, no. 9 (September 14, 2021): 1823. http://dx.doi.org/10.3390/v13091823.

Повний текст джерела
Анотація:
Congenital Zika Syndrome (CZS) is caused by vertical transmission of Zika virus (ZIKV) to the gestating human fetus. A subset of CZS microcephalic infants present with reduced otoacoustic emissions; this test screens for hearing loss originating in the cochlea. This observation leads to the question of whether mammalian cochlear tissues are susceptible to infection by ZIKV during development. To address this question using a mouse model, the sensory cochlea was explanted at proliferative, newly post-mitotic or maturing stages. ZIKV was added for the first 24 h and organs cultured for up to 6 days to allow for cell differentiation. Results showed that ZIKV can robustly infect proliferating sensory progenitors, as well as post-mitotic hair cells and supporting cells. Virus neutralization using ZIKV-117 antibody blocked cochlear infection. AXL is a cell surface molecule known to enhance the attachment of flavivirus to host cells. While Axl mRNA is widely expressed in embryonic cochlear tissues susceptible to ZIKV infection, it is selectively downregulated in the post-mitotic sensory organ by E15.5, even though these cells remain infectible. These findings may offer insights into which target cells could potentially contribute to hearing loss resulting from fetal exposure to ZIKV in humans.
Стилі APA, Harvard, Vancouver, ISO та ін.
4

Lin, Jizhen, Ling Feng, Shinji Fukudome, Yuki Hamajima, Tina Huang, and Samuel Levine. "Cochlear Stem Cells/Progenitors and Degenerative Hearing Disorders." Current Medicinal Chemistry 14, no. 27 (November 1, 2007): 2937–43. http://dx.doi.org/10.2174/092986707782360051.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
5

Feng, Ling. "Differentiation of cochlear neural progenitors with SV40 in vitro." Molecular and Cellular Pharmacology 1, no. 1 (February 10, 2009): 11–22. http://dx.doi.org/10.4255/mcpharmacol.09.03.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
6

Lin, Jizhen, Ling Feng, Yuki Hamajima, Masahiro Komori, Terry C. Burns, Shinji Fukudome, John Anderson, Dong Wang, Catherine M. Verfaillie, and Walter C. Low. "Directed differentiation of mouse cochlear neural progenitors in vitro." American Journal of Physiology-Cell Physiology 296, no. 3 (March 2009): C441—C452. http://dx.doi.org/10.1152/ajpcell.00324.2008.

Повний текст джерела
Анотація:
Multipotent cochlear neural progenitors (CNPs) in the organ of Corti hold the promise for cell replacement in degenerative hearing disorders. However, not much is known about the CNPs and the specific conditions for their differentiation. Here we isolate the CNPs from the postnatal day 1 organ of Corti in mice and demonstrate their capability to self-renew and to differentiate into hair cell-like and neuronal cell-like phenotypes under the guidance of sonic hedgehog (SHH), epidermal growth factor (EGF), retinoic acid (RA), and brain-derived neurotrophic factor (BDNF), herein termed SERB (abbreviation of SHH, EGF, RA, and BDNF) in an asymmetric or symmetric manner from clonal isolates. Differentiation of CNPs into hair cells by SERB was dependent on the ERK signaling pathway, whereas the differentiation of CNPs into neurons by SERB was not. This work develops a new in vitro methodology for the maintenance and self-regeneration of CNPs for future design of regenerative strategies for hearing disorders.
Стилі APA, Harvard, Vancouver, ISO та ін.
7

Otsuka, Kelly S., Christopher Nielson, Matthew A. Firpo, Albert H. Park, and Anna E. Beaudin. "Early Life Inflammation and the Developing Hematopoietic and Immune Systems: The Cochlea as a Sensitive Indicator of Disruption." Cells 10, no. 12 (December 20, 2021): 3596. http://dx.doi.org/10.3390/cells10123596.

Повний текст джерела
Анотація:
Emerging evidence indicates that perinatal infection and inflammation can influence the developing immune system and may ultimately affect long-term health and disease outcomes in offspring by perturbing tissue and immune homeostasis. We posit that perinatal inflammation influences immune outcomes in offspring by perturbing (1) the development and function of fetal-derived immune cells that regulate tissue development and homeostasis, and (2) the establishment and function of developing hematopoietic stem cells (HSCs) that continually generate immune cells across the lifespan. To disentangle the complexities of these interlinked systems, we propose the cochlea as an ideal model tissue to investigate how perinatal infection affects immune, tissue, and stem cell development. The cochlea contains complex tissue architecture and a rich immune milieu that is established during early life. A wide range of congenital infections cause cochlea dysfunction and sensorineural hearing loss (SNHL), likely attributable to early life inflammation. Furthermore, we show that both immune cells and bone marrow hematopoietic progenitors can be simultaneously analyzed within neonatal cochlear samples. Future work investigating the pathogenesis of SNHL in the context of congenital infection will therefore provide critical information on how perinatal inflammation drives disease susceptibility in offspring.
Стилі APA, Harvard, Vancouver, ISO та ін.
8

Kwan, Kelvin Y., and Patricia M. White. "Understanding the differentiation and epigenetics of cochlear sensory progenitors in pursuit of regeneration." Current Opinion in Otolaryngology & Head & Neck Surgery 29, no. 5 (August 9, 2021): 366–72. http://dx.doi.org/10.1097/moo.0000000000000741.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
9

Breuskin, Ingrid, Morgan Bodson, Nicolas Thelen, Marc Thiry, Laurence Borgs, Laurent Nguyen, Philippe P. Lefebvre, and Brigitte Malgrange. "Sox10 promotes the survival of cochlear progenitors during the establishment of the organ of Corti." Developmental Biology 335, no. 2 (November 2009): 327–39. http://dx.doi.org/10.1016/j.ydbio.2009.09.007.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
10

Gnedeva, Ksenia, Xizi Wang, Melissa M. McGovern, Matthew Barton, Litao Tao, Talon Trecek, Tanner O. Monroe, et al. "Organ of Corti size is governed by Yap/Tead-mediated progenitor self-renewal." Proceedings of the National Academy of Sciences 117, no. 24 (June 1, 2020): 13552–61. http://dx.doi.org/10.1073/pnas.2000175117.

Повний текст джерела
Анотація:
Precise control of organ growth and patterning is executed through a balanced regulation of progenitor self-renewal and differentiation. In the auditory sensory epithelium—the organ of Corti—progenitor cells exit the cell cycle in a coordinated wave between E12.5 and E14.5 before the initiation of sensory receptor cell differentiation, making it a unique system for studying the molecular mechanisms controlling the switch between proliferation and differentiation. Here we identify the Yap/Tead complex as a key regulator of the self-renewal gene network in organ of Corti progenitor cells. We show that Tead transcription factors bind directly to the putative regulatory elements of many stemness- and cell cycle-related genes. We also show that the Tead coactivator protein, Yap, is degraded specifically in the Sox2-positive domain of the cochlear duct, resulting in down-regulation of Tead gene targets. Further, conditional loss of theYapgene in the inner ear results in the formation of significantly smaller auditory and vestibular sensory epithelia, while conditional overexpression of a constitutively active version ofYap,Yap5SA, is sufficient to prevent cell cycle exit and to prolong sensory tissue growth. We also show that viral gene delivery ofYap5SAin the postnatal inner ear sensory epithelia in vivo drives cell cycle reentry after hair cell loss. Taken together, these data highlight the key role of the Yap/Tead transcription factor complex in maintaining inner ear progenitors during development, and suggest new strategies to induce sensory cell regeneration.
Стилі APA, Harvard, Vancouver, ISO та ін.
11

Kanzaki, Sho. "Gene Delivery into the Inner Ear and Its Clinical Implications for Hearing and Balance." Molecules 23, no. 10 (September 30, 2018): 2507. http://dx.doi.org/10.3390/molecules23102507.

Повний текст джерела
Анотація:
The inner ear contains many types of cell, including sensory hair cells and neurons. If these cells are damaged, they do not regenerate. Inner ear disorders have various etiologies. Some are related to aging or are idiopathic, as in sudden deafness. Others occur due to acoustic trauma, exposure to ototoxic drugs, viral infections, immune responses, or endolymphatic hydrops (Meniere’s disease). For these disorders, inner ear regeneration therapy is expected to be a feasible alternative to cochlear implants for hearing recovery. Recently, the mechanisms underlying inner ear regeneration have been gradually clarified. Inner ear cell progenitors or stem cells have been identified. Factors necessary for regeneration have also been elucidated from the mechanism of hair cell generation. Inducing differentiation of endogenous stem cells or inner ear stem cell transplantation is expected. In this paper, we discuss recent approaches to hair cell proliferation and differentiation for inner ear regeneration. We discuss the future road map for clinical application. The therapies mentioned above require topical administration of transgenes or drug onto progenitors of sensory cells. Developing efficient and safe modes of administration is clinically important. In this regard, we also discuss our development of an inner ear endoscope to facilitate topical administration.
Стилі APA, Harvard, Vancouver, ISO та ін.
12

Shi, Fuxin, Lingxiang Hu та Albert S. B. Edge. "Generation of hair cells in neonatal mice by β-catenin overexpression in Lgr5-positive cochlear progenitors". Proceedings of the National Academy of Sciences 110, № 34 (5 серпня 2013): 13851–56. http://dx.doi.org/10.1073/pnas.1219952110.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
13

Nayagam, Bryony A., Albert S. Edge, Karina Needham, Tomoko Hyakumura, Jessie Leung, David A. X. Nayagam, and Mirella Dottori. "An In Vitro Model of Developmental Synaptogenesis Using Cocultures of Human Neural Progenitors and Cochlear Explants." Stem Cells and Development 22, no. 6 (March 15, 2013): 901–12. http://dx.doi.org/10.1089/scd.2012.0082.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
14

Zhang, Yuan, Suo-qiang Zhai, Jianyong Shou, Wei Song, Jian-he Sun, Wei Guo, Gui-liang Zheng, Yin-yan Hu, and Wei-Qiang Gao. "Isolation, growth and differentiation of hair cell progenitors from the newborn rat cochlear greater epithelial ridge." Journal of Neuroscience Methods 164, no. 2 (August 2007): 271–79. http://dx.doi.org/10.1016/j.jneumeth.2007.05.009.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
15

Jan, T. A., R. Chai, Z. N. Sayyid, R. van Amerongen, A. Xia, T. Wang, S. T. Sinkkonen, et al. "Tympanic border cells are Wnt-responsive and can act as progenitors for postnatal mouse cochlear cells." Development 140, no. 6 (February 26, 2013): 1196–206. http://dx.doi.org/10.1242/dev.087528.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
16

Zine, Azel, Yassine Messat, and Bernd Fritzsch. "A human induced pluripotent stem cell-based modular platform to challenge sensorineural hearing loss." Stem Cells 39, no. 6 (February 8, 2021): 697–706. http://dx.doi.org/10.1002/stem.3346.

Повний текст джерела
Анотація:
Abstract The sense of hearing depends on a specialized sensory organ in the inner ear, called the cochlea, which contains the auditory hair cells (HCs). Noise trauma, infections, genetic factors, side effects of ototoxic drugs (ie, some antibiotics and chemotherapeutics), or simply aging lead to the loss of HCs and their associated primary neurons. This results in irreversible sensorineural hearing loss (SNHL) as in mammals, including humans; the inner ear lacks the capacity to regenerate HCs and spiral ganglion neurons. SNHL is a major global health problem affecting millions of people worldwide and provides a growing concern in the aging population. To date, treatment options are limited to hearing aids and cochlear implants. A major bottleneck for development of new therapies for SNHL is associated to the lack of human otic cell bioassays. Human induced pluripotent stem cells (hiPSCs) can be induced in two-dimensional and three-dimensional otic cells in vitro models that can generate inner ear progenitors and sensory HCs and could be a promising preclinical platform from which to work toward restoring SNHL. We review the potential applications of hiPSCs in the various biological approaches, including disease modeling, bioengineering, drug testing, and autologous stem cell based-cell therapy, that offer opportunities to understand the pathogenic mechanisms of SNHL and identify novel therapeutic strategies.
Стилі APA, Harvard, Vancouver, ISO та ін.
17

Li, Wenyan, Jingfang Wu, Jianming Yang, Shan Sun, Renjie Chai, Zheng-Yi Chen, and Huawei Li. "Notch inhibition induces mitotically generated hair cells in mammalian cochleae via activating the Wnt pathway." Proceedings of the National Academy of Sciences 112, no. 1 (December 22, 2014): 166–71. http://dx.doi.org/10.1073/pnas.1415901112.

Повний текст джерела
Анотація:
The activation of cochlear progenitor cells is a promising approach for hair cell (HC) regeneration and hearing recovery. The mechanisms underlying the initiation of proliferation of postnatal cochlear progenitor cells and their transdifferentiation to HCs remain to be determined. We show that Notch inhibition initiates proliferation of supporting cells (SCs) and mitotic regeneration of HCs in neonatal mouse cochlea in vivo and in vitro. Through lineage tracing, we identify that a majority of the proliferating SCs and mitotic-generated HCs induced by Notch inhibition are derived from the Wnt-responsive leucine-rich repeat-containing G protein-coupled receptor 5 (Lgr5+) progenitor cells. We demonstrate that Notch inhibition removes the brakes on the canonical Wnt signaling and promotes Lgr5+ progenitor cells to mitotically generate new HCs. Our study reveals a new function of Notch signaling in limiting proliferation and regeneration potential of postnatal cochlear progenitor cells, and provides a new route to regenerate HCs from progenitor cells by interrupting the interaction between the Notch and Wnt pathways.
Стилі APA, Harvard, Vancouver, ISO та ін.
18

Hu, Xiaohua, Jianmin Huang, Ling Feng, Shinji Fukudome, Yuki Hamajima, and Jizhen Lin. "Sonic hedgehog (SHH) promotes the differentiation of mouse cochlear neural progenitors via theMath1-Brn3.1 signaling pathway in vitro." Journal of Neuroscience Research 88, no. 5 (November 11, 2009): 927–35. http://dx.doi.org/10.1002/jnr.22286.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
19

Rousset, Francis, Giulia Schilardi, Stéphanie Sgroi, German Nacher-Soler, Rebecca Sipione, Sonja Kleinlogel та Pascal Senn. "WNT Activation and TGFβ-Smad Inhibition Potentiate Stemness of Mammalian Auditory Neuroprogenitors for High-Throughput Generation of Functional Auditory Neurons In Vitro". Cells 11, № 15 (5 серпня 2022): 2431. http://dx.doi.org/10.3390/cells11152431.

Повний текст джерела
Анотація:
Hearing loss affects over 460 million people worldwide and is a major socioeconomic burden. Both genetic and environmental factors (i.e., noise overexposure, ototoxic drug treatment and ageing), promote the irreversible degeneration of cochlear hair cells and associated auditory neurons, leading to sensorineural hearing loss. In contrast to birds, fish and amphibians, the mammalian inner ear is virtually unable to regenerate due to the limited stemness of auditory progenitors, and no causal treatment is able to prevent or reverse hearing loss. As of today, a main limitation for the development of otoprotective or otoregenerative therapies is the lack of efficient preclinical models compatible with high-throughput screening of drug candidates. Currently, the research field mainly relies on primary organotypic inner ear cultures, resulting in high variability, low throughput, high associated costs and ethical concerns. We previously identified and characterized the phoenix auditory neuroprogenitors (ANPGs) as highly proliferative progenitor cells isolated from the A/J mouse cochlea. In the present study, we aim at identifying the signaling pathways responsible for the intrinsic high stemness of phoenix ANPGs. A transcriptomic comparison of traditionally low-stemness ANPGs, isolated from C57Bl/6 and A/J mice at early passages, and high-stemness phoenix ANPGs was performed, allowing the identification of several differentially expressed pathways. Based on differentially regulated pathways, we developed a reprogramming protocol to induce high stemness in presenescent ANPGs (i.e., from C57Bl6 mouse). The pharmacological combination of the WNT agonist (CHIR99021) and TGFβ/Smad inhibitors (LDN193189 and SB431542) resulted in a dramatic increase in presenescent neurosphere growth, and the possibility to expand ANPGs is virtually limitless. As with the phoenix ANPGs, stemness-induced ANPGs could be frozen and thawed, enabling distribution to other laboratories. Importantly, even after 20 passages, stemness-induced ANPGs retained their ability to differentiate into electrophysiologically mature type I auditory neurons. Both stemness-induced and phoenix ANPGs resolve a main bottleneck in the field, allowing efficient, high-throughput, low-cost and 3R-compatible in vitro screening of otoprotective and otoregenerative drug candidates. This study may also add new perspectives to the field of inner ear regeneration.
Стилі APA, Harvard, Vancouver, ISO та ін.
20

Li, Guangfei, Yanbo Yin, Yaopeng Zhang, Jingfang Wu, and Shan Sun. "Electrospun regenerated silk fibroin is a promising biomaterial for the maintenance of inner ear progenitors in vitro." Journal of Biomaterials Applications 36, no. 7 (October 28, 2021): 1164–72. http://dx.doi.org/10.1177/08853282211051501.

Повний текст джерела
Анотація:
Objective We sought to determine the biocompatibility of electrospun regenerated silk fibroin (RSF) mats with inner ear progenitors, especially their effect on the differentiation of inner ear progenitors into hair cells. Methods Neonatal mouse cochleae (n = 20) were collected and digested and allowed to form spheres over several days. Cells digested from the spheres were then seeded onto aligned or random RSF mats, with laminin-coated coverslips serving as controls. The inner ear progenitor cell mortality was examined by TUNEL labeling, and the adhesion of cells to the RSF mats or coverslip was determined by scanning electron microscopy. Finally, the number of hair cells that differentiated from inner ear progenitors was determined by Myosin7a expression. Unpaired Student’s t-tests and one-way ANOVA followed by a Dunnett’s multiple comparisons test were used in this study ( p < 0.05). Results After 5 days of culture, the inner ear progenitors had good adhesion to both the aligned and random RSF mats and there was no significant difference in TUNEL+ cells between the mats compared to the coverslip ( p > 0.05). After 7 days of in vitro differentiation culture, the percentage of differentiated hair cells on the control, aligned, and random RSF mats was 2.5 ± 0.5%, 2.7 ± 0.4%, and 2.4 ± 0.2%, respectively, and there was no significant difference between Myosin7a+ cells on either RSF mat compared to controls ( p > 0.05). Conclusion The aligned and random RSF mats had excellent biocompatibility with inner ear progenitors and helped the inner ear progenitors maintain their stemness. Our results thus indicate that RSF mats represent a useful scaffold for the development of new strategies for inner ear tissue engineering research.
Стилі APA, Harvard, Vancouver, ISO та ін.
21

Savary, Etienne, Jean Charles Sabourin, Julien Santo, Jean Philippe Hugnot, Christian Chabbert, Thomas Van De Water, Alain Uziel, and Azel Zine. "Cochlear stem/progenitor cells from a postnatal cochlea respond to Jagged1 and demonstrate that notch signaling promotes sphere formation and sensory potential." Mechanisms of Development 125, no. 8 (August 2008): 674–86. http://dx.doi.org/10.1016/j.mod.2008.05.001.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
22

Liu, Quanwen, Yi Shen, Jiarong Chen, Jie Ding, Zihua Tang, Cui Zhang, Jianling Chen, Liang Li, Ping Chen, and Jinfu Wang. "Induction of Functional Hair-Cell-Like Cells from Mouse Cochlear Multipotent Cells." Stem Cells International 2016 (2016): 1–14. http://dx.doi.org/10.1155/2016/8197279.

Повний текст джерела
Анотація:
In this paper, we developed a two-step-induction method of generating functional hair cells from inner ear multipotent cells. Multipotent cells from the inner ear were established and induced initially into progenitor cells committed to the inner ear cell lineage on the poly-L-lysine substratum. Subsequently, the committed progenitor cells were cultured on the mitotically inactivated chicken utricle stromal cells and induced into hair-cell-like cells containing characteristic stereocilia bundles. The hair-cell-like cells exhibited rapid permeation of FM1-43FX. The whole-cell patch-clamp technique was used to measure the membrane currents of cells differentiated for 7 days on chicken utricle stromal cells and analyze the biophysical properties of the hair-cell-like cells by recording membrane properties of cells. The results suggested that the hair-cell-like cells derived from inner ear multipotent cells were functional following differentiation in an enabling environment.
Стилі APA, Harvard, Vancouver, ISO та ін.
23

Zhai, Suoqiang, Li Shi, Bu-er Wang, Guiliang Zheng, Wei Song, Yinyan Hu, and Wei-Qiang Gao. "Isolation and culture of hair cell progenitors from postnatal rat cochleae." Journal of Neurobiology 65, no. 3 (2005): 282–93. http://dx.doi.org/10.1002/neu.20190.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
24

Voelker, Johannes, Jonas Engert, Christine Voelker, Linda Bieniussa, Philipp Schendzielorz, Rudolf Hagen, and Kristen Rak. "Different Neurogenic Potential in the Subnuclei of the Postnatal Rat Cochlear Nucleus." Stem Cells International 2021 (April 5, 2021): 1–15. http://dx.doi.org/10.1155/2021/8871308.

Повний текст джерела
Анотація:
In patients suffering from hearing loss, the reduced or absent neural input induces morphological changes in the cochlear nucleus (CN). Neural stem cells have recently been identified in this first auditory relay. Afferent nerve signals and their impact on the immanent neural stem and progenitor cells already impinge upon the survival of early postnatal cells within the CN. This auditory brainstem nucleus consists of three different subnuclei: the anteroventral cochlear nucleus (AVCN), the posteroventral cochlear nucleus (PVCN), and the dorsal cochlear nucleus (DCN). Since these subdivisions differ ontogenetically and physiologically, the question arose whether regional differences exist in the neurogenic niche. CN from postnatal day nine Sprague-Dawley rats were microscopically dissected into their subnuclei and cultivated in vitro as free-floating cell cultures and as whole-mount organ cultures. In addition to cell quantifications, immunocytological and immunohistological studies of the propagated cells and organ preparations were performed. The PVCN part showed the highest mitotic potential, while the AVCN and DCN had comparable activity. Specific stem cell markers and the ability to differentiate into cells of the neural lineage were detected in all three compartments. The present study shows that in all subnuclei of rat CN, there is a postnatal neural stem cell niche, which, however, differs significantly in its potential. The results can be explained by the origin from different regions in the rhombic lip, the species, and the various analysis techniques applied. In conclusion, the presented results provide further insight into the neurogenic potential of the CN, which may prove beneficial for the development of new regenerative strategies for hearing loss.
Стилі APA, Harvard, Vancouver, ISO та ін.
25

Chen, Hsin-Chien, Jen-Tin Lee, Cheng-Ping Shih, Ting-Ting Chao, Huey-Kang Sytwu, Shiue-Li Li, Mei-Cho Fang, et al. "Hypoxia Induces a Metabolic Shift and Enhances the Stemness and Expansion of Cochlear Spiral Ganglion Stem/Progenitor Cells." BioMed Research International 2015 (2015): 1–12. http://dx.doi.org/10.1155/2015/359537.

Повний текст джерела
Анотація:
Previously, we demonstrated that hypoxia (1% O2) enhances stemness markers and expands the cell numbers of cochlear stem/progenitor cells (SPCs). In this study, we further investigated the long-term effect of hypoxia on stemness and the bioenergetic status of cochlear spiral ganglion SPCs cultured at low oxygen tensions. Spiral ganglion SPCs were obtained from postnatal day 1 CBA/CaJ mouse pups. The measurement of oxygen consumption rate, extracellular acidification rate (ECAR), and intracellular adenosine triphosphate levels corresponding to 20% and 5% oxygen concentrations was determined using a Seahorse XF extracellular flux analyzer. After low oxygen tension cultivation for 21 days, the mean size of the hypoxia-expanded neurospheres was significantly increased at 5% O2; this correlated with high-level expression of hypoxia-inducible factor-1 alpha (Hif-1α), proliferating cell nuclear antigen (PCNA), cyclin D1, Abcg2, nestin, and Nanog proteins but downregulated expression of p27 compared to that in a normoxic condition. Low oxygen tension cultivation tended to increase the side population fraction, with a significant difference found at 5% O2compared to that at 20% O2. In addition, hypoxia induced a metabolic energy shift of SPCs toward higher basal ECARs and higher maximum mitochondrial respiratory capacity but lower proton leak than under normoxia, where the SPC metabolism was switched toward glycolysis in long-term hypoxic cultivation.
Стилі APA, Harvard, Vancouver, ISO та ін.
26

Lopez-Juarez, Alejandra, Hanae Lahlou, Chantal Ripoll, Yves Cazals, Jean Michel Brezun, Quan Wang, Albert Edge, and Azel Zine. "Engraftment of Human Stem Cell-Derived Otic Progenitors in the Damaged Cochlea." Molecular Therapy 27, no. 6 (June 2019): 1101–13. http://dx.doi.org/10.1016/j.ymthe.2019.03.018.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
27

Wang, Junli, Yinglong Xu, Yuli Zhao, and Min Xu. "Different morphologic features of rat cochlea progenitor spheres and their implications." Journal of Medical Colleges of PLA 27, no. 6 (December 2012): 311–23. http://dx.doi.org/10.1016/s1000-1948(13)60001-5.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
28

Shi, F., J. S. Kempfle, and A. S. B. Edge. "Wnt-Responsive Lgr5-Expressing Stem Cells Are Hair Cell Progenitors in the Cochlea." Journal of Neuroscience 32, no. 28 (July 11, 2012): 9639–48. http://dx.doi.org/10.1523/jneurosci.1064-12.2012.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
29

Ho, Chin Chung, Tianli Qin, Boshi Wang, Elaine Y. M. Wong, Yuchen Liu, Chi-Chung Hui, and Mai Har Sham. "Sufu regulates the proliferation and differentiation of hair cell progenitors in mammalian cochlea." Mechanisms of Development 145 (July 2017): S118. http://dx.doi.org/10.1016/j.mod.2017.04.318.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
30

Volkenstein, S., K. Oshima, S. T. Sinkkonen, C. E. Corrales, S. P. Most, R. Chai, T. A. Jan, R. van Amerongen, A. G. Cheng, and S. Heller. "Transient, afferent input-dependent, postnatal niche for neural progenitor cells in the cochlear nucleus." Proceedings of the National Academy of Sciences 110, no. 35 (August 12, 2013): 14456–61. http://dx.doi.org/10.1073/pnas.1307376110.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
31

Zhong, Cuiping, Yu Han, Ji Ma, Xuan Zhang, Mengning Sun, Ye Wang, Jun Chen, Wenjuan Mi, Xuehai Xu, and Jianhua Qiu. "Viral-mediated expression of c-Myc and cyclin A2 induces cochlear progenitor cell proliferation." Neuroscience Letters 591 (March 2015): 93–98. http://dx.doi.org/10.1016/j.neulet.2015.02.027.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
32

Hei, Renyi, Jun Chen, Li Qiao, Xu Li, Xiaobo Mao, Jianhua Qiu, and Juan Qu. "Dynamic changes in microRNA expression during differentiation of rat cochlear progenitor cells in vitro." International Journal of Pediatric Otorhinolaryngology 75, no. 8 (August 2011): 1010–14. http://dx.doi.org/10.1016/j.ijporl.2011.05.005.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
33

Chen, Hsin-Chien, Chih-Hung Wang, Cheng-Ping Shih, Sheau-Huei Chueh, Shu-Fan Liu, Hang-Kang Chen, and Yi-Chun Lin. "TRPC1 is required for survival and proliferation of cochlear spiral ganglion stem/progenitor cells." International Journal of Pediatric Otorhinolaryngology 79, no. 12 (December 2015): 2290–94. http://dx.doi.org/10.1016/j.ijporl.2015.10.027.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
34

Sato, Eisuke, H. Elizabeth Shick, Richard M. Ransohoff, and Keiko Hirose. "Repopulation of cochlear macrophages in murine hematopoietic progenitor cell chimeras: The role of CX3CR1." Journal of Comparative Neurology 506, no. 6 (2007): 930–42. http://dx.doi.org/10.1002/cne.21583.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
35

Nishimura, Koji, Takayuki Nakagawa, Tatsunori Sakamoto, and Juichi Ito. "Fates of Murine Pluripotent Stem Cell-Derived Neural Progenitors following Transplantation into Mouse Cochleae." Cell Transplantation 21, no. 4 (April 2012): 763–71. http://dx.doi.org/10.3727/096368911x623907.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
36

Kubota, Marie, Mirko Scheibinger, Taha A. Jan, and Stefan Heller. "Greater epithelial ridge cells are the principal organoid-forming progenitors of the mouse cochlea." Cell Reports 34, no. 3 (January 2021): 108646. http://dx.doi.org/10.1016/j.celrep.2020.108646.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
37

Chen, Wei, Daniela I. Cacciabue-Rivolta, Harry D. Moore, and Marcelo N. Rivolta. "The human fetal cochlea can be a source for auditory progenitors/stem cells isolation." Hearing Research 233, no. 1-2 (November 2007): 23–29. http://dx.doi.org/10.1016/j.heares.2007.06.006.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
38

Doetzlhofer, Angelika, Patricia White, Yun-Shain Lee, Andrew Groves, and Neil Segil. "Prospective identification and purification of hair cell and supporting cell progenitors from the embryonic cochlea." Brain Research 1091, no. 1 (May 2006): 282–88. http://dx.doi.org/10.1016/j.brainres.2006.02.071.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
39

Chao, Ting-Ting, Chih-Hung Wang, Hsin-Chien Chen, Cheng-Ping Shih, Huey-Kang Sytwu, Kun-Lun Huang, and Shao-Yuan Chen. "Adherent culture conditions enrich the side population obtained from the cochlear modiolus-derived stem/progenitor cells." International Journal of Pediatric Otorhinolaryngology 77, no. 5 (May 2013): 779–84. http://dx.doi.org/10.1016/j.ijporl.2013.02.010.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
40

Boddy, Sarah L., Ricardo Romero-Guevara, Ae-Ri Ji, Christian Unger, Laura Corns, Walter Marcotti, and Marcelo N. Rivolta. "Generation of Otic Lineages from Integration-Free Human-Induced Pluripotent Stem Cells Reprogrammed by mRNAs." Stem Cells International 2020 (March 1, 2020): 1–10. http://dx.doi.org/10.1155/2020/3692937.

Повний текст джерела
Анотація:
Damage to the sensory hair cells and the spiral ganglion neurons of the cochlea leads to deafness. Induced pluripotent stem cells (iPSCs) are a promising tool to regenerate the cells in the inner ear that have been affected by pathology or have been lost. To facilitate the clinical application of iPSCs, the reprogramming process should minimize the risk of introducing undesired genetic alterations while conferring the cells the capacity to differentiate into the desired cell type. Currently, reprogramming induced by synthetic mRNAs is considered to be one of the safest ways of inducing pluripotency, as the transgenes are transiently delivered into the cells without integrating into the genome. In this study, we explore the ability of integration-free human-induced pluripotent cell lines that were reprogrammed by mRNAs, to differentiate into otic progenitors and, subsequently, into hair cell and neuronal lineages. hiPSC lines were induced to differentiate by culturing them in the presence of fibroblast growth factors 3 and 10 (FGF3 and FGF10). Progenitors were identified by quantitative microscopy, based on the coexpression of otic markers PAX8, PAX2, FOXG1, and SOX2. Otic epithelial progenitors (OEPs) and otic neuroprogenitors (ONPs) were purified and allowed to differentiate further into hair cell-like cells and neurons. Lineages were characterised by immunocytochemistry and electrophysiology. Neuronal cells showed inward Na+ (INa) currents and outward (Ik) and inward K+ (IK1) currents while hair cell-like cells had inward IK1 and outward delayed rectifier K+ currents, characteristic of developing hair cells. We conclude that human-induced pluripotent cell lines that have been reprogrammed using nonintegrating mRNAs are capable to differentiate into otic cell types.
Стилі APA, Harvard, Vancouver, ISO та ін.
41

Voelker, Johannes, Christine Voelker, Jonas Engert, Nikolas Goemann, Rudolf Hagen, and Kristen Rak. "Spontaneous Calcium Oscillations through Differentiation: A Calcium Imaging Analysis of Rat Cochlear Nucleus Neural Stem Cells." Cells 10, no. 10 (October 19, 2021): 2802. http://dx.doi.org/10.3390/cells10102802.

Повний текст джерела
Анотація:
Causal therapies for the auditory-pathway and inner-ear diseases are still not yet available for clinical application. Regenerative medicine approaches are discussed and examined as possible therapy options. Neural stem cells could play a role in the regeneration of the auditory pathway. In recent years, neural stem and progenitor cells have been identified in the cochlear nucleus, the second nucleus of the auditory pathway. The current investigation aimed to analyze cell maturation concerning cellular calcium activity. Cochlear nuclei from PND9 CD rats were microscopically dissected and propagated as neurospheres in free-floating cultures in stem-cell medium (Neurobasal, B27, GlutaMAX, EGF, bFGF). After 30 days, the dissociation and plating of these cells took place under withdrawal of the growth factors and the addition of retinoic acid, which induces neural cell differentiation. Calcium imaging analysis with BAPTA-1/Oregon Green was carried out at different times during the differentiation phase. In addition, the influence of different voltage-dependent calcium channels was analyzed through the targeted application of inhibitors of the L-, N-, R- and T-type calcium channels. For this purpose, comparative examinations were performed on CN NSCs, and primary CN neurons. As the cells differentiated, a significant increase in spontaneous neuronal calcium activity was demonstrated. In the differentiation stage, specific frequencies of the spontaneous calcium oscillations were measured in different regions of the individual cells. Initially, the highest frequency of spontaneous calcium oscillations was ascertainable in the maturing somata. Over time, these were overtaken by calcium oscillations in the axons and dendrites. Additionally, in the area of the growth cones, an increasing activity was determined. By inhibiting voltage-dependent calcium channels, their expression and function in the differentiation process were confirmed. A comparable pattern of maturation of these channels was found in CN NSCs and primary CN neurons. The present results show that neural stem cells of the rat cochlear nucleus differentiated not only morphologically but also functionally. Spontaneous calcium activities are of great relevance in terms of neurogenesis and integration into existing neuronal structures. These functional aspects of neurogenesis within the auditory pathway could serve as future targets for the exogenous control of neuronal regeneration.
Стилі APA, Harvard, Vancouver, ISO та ін.
42

Chen, P., and N. Segil. "p27(Kip1) links cell proliferation to morphogenesis in the developing organ of Corti." Development 126, no. 8 (April 15, 1999): 1581–90. http://dx.doi.org/10.1242/dev.126.8.1581.

Повний текст джерела
Анотація:
Strict control of cellular proliferation is required to shape the complex structures of the developing embryo. The organ of Corti, the auditory neuroepithelium of the inner ear in mammals, consists of two types of terminally differentiated mechanosensory hair cells and at least four types of supporting cells arrayed precisely along the length of the spiral cochlea. In mice, the progenitors of greater than 80% of both hair cells and supporting cells undergo their terminal division between embryonic day 13 (E13) and E14. As in humans, these cells persist in a non-proliferative state throughout the adult life of the animal. Here we report that the correct timing of cell cycle withdrawal in the developing organ of Corti requires p27(Kip1), a cyclin-dependent kinase inhibitor that functions as an inhibitor of cell cycle progression. p27(Kip1) expression is induced in the primordial organ of Corti between E12 and E14, correlating with the cessation of cell division of the progenitors of the hair cells and supporting cells. In wild-type animals, p27(Kip1) expression is downregulated during subsequent hair cell differentiation, but it persists at high levels in differentiated supporting cells of the mature organ of Corti. In mice with a targeted deletion of the p27(Kip1) gene, proliferation of the sensory cell progenitors continues after E14, leading to the appearance of supernumerary hair cells and supporting cells. In the absence of p27(Kip1), mitotically active cells are still observed in the organ of Corti of postnatal day 6 animals, suggesting that the persistence of p27(Kip1) expression in mature supporting cells may contribute to the maintenance of quiescence in this tissue and, possibly, to its inability to regenerate. Homozygous mutant mice are severely hearing impaired. Thus, p27(Kip1) provides a link between developmental control of cell proliferation and the morphological development of the inner ear.
Стилі APA, Harvard, Vancouver, ISO та ін.
43

Lou, Xiang-Xin, Takayuki Nakagawa, Hiroe Ohnishi, Koji Nishimura, and Juichi Ito. "Otospheres derived from neonatal mouse cochleae retain the progenitor cell phenotype after ex vivo expansions." Neuroscience Letters 534 (February 2013): 18–23. http://dx.doi.org/10.1016/j.neulet.2012.12.001.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
44

Massucci-Bissoli, M., K. Lezirovitz, J. Oiticica, and RF Bento. "Evidence of progenitor cells in the adult human cochlea: sphere formation and identification of ABCG2." Clinics 72, no. 11 (November 7, 2017): 714–17. http://dx.doi.org/10.6061/clinics/2017(11)11.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
45

Diensthuber, Marc, Kazuo Oshima, and Stefan Heller. "Stem/Progenitor Cells Derived from the Cochlear Sensory Epithelium Give Rise to Spheres with Distinct Morphologies and Features." Journal of the Association for Research in Otolaryngology 10, no. 2 (February 27, 2009): 173–90. http://dx.doi.org/10.1007/s10162-009-0161-3.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
46

Waqas, Muhammad, Luo Guo, Shasha Zhang, Yan Chen, Xiaoli Zhang, Lei Wang, Mingliang Tang, et al. "Characterization of Lgr5+ progenitor cell transcriptomes in the apical and basal turns of the mouse cochlea." Oncotarget 7, no. 27 (April 7, 2016): 41123–41. http://dx.doi.org/10.18632/oncotarget.8636.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
47

Savary, Etienne, Jean Philippe Hugnot, Yolaine Chassigneux, Cecile Travo, Christophe Duperray, Thomas Van De Water, and Azel Zine. "Distinct Population of Hair Cell Progenitors Can Be Isolated from the Postnatal Mouse Cochlea Using Side Population Analysis." Stem Cells 25, no. 2 (February 2007): 332–39. http://dx.doi.org/10.1634/stemcells.2006-0303.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
48

McLean, Will J., Dalton T. McLean, Ruth Anne Eatock, and Albert S. B. Edge. "Distinct capacity for differentiation to inner ear cell types by progenitor cells of the cochlea and vestibular organs." Development 143, no. 23 (October 27, 2016): 4381–93. http://dx.doi.org/10.1242/dev.139840.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
49

Chen, Hsin-Chien, Huey-Kang Sytwu, Junn-Liang Chang, Hsing-Won Wang, Hang-Kang Chen, Bor-Hwang Kang, Dai-Wei Liu, Chi-Huang Chen, Ting-Ting Chao, and Chih-Hung Wang. "Hypoxia enhances the stemness markers of cochlear stem/progenitor cells and expands sphere formation through activation of hypoxia-inducible factor-1alpha." Hearing Research 275, no. 1-2 (May 2011): 43–52. http://dx.doi.org/10.1016/j.heares.2010.12.004.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
50

Golden, Erin J., Ana Benito-Gonzalez, and Angelika Doetzlhofer. "The RNA-binding protein LIN28B regulates developmental timing in the mammalian cochlea." Proceedings of the National Academy of Sciences 112, no. 29 (July 2, 2015): E3864—E3873. http://dx.doi.org/10.1073/pnas.1501077112.

Повний текст джерела
Анотація:
Proper tissue development requires strict coordination of proliferation, growth, and differentiation. Strict coordination is particularly important for the auditory sensory epithelium, where deviations from the normal spatial and temporal pattern of auditory progenitor cell (prosensory cell) proliferation and differentiation result in abnormal cellular organization and, thus, auditory dysfunction. The molecular mechanisms involved in the timing and coordination of auditory prosensory proliferation and differentiation are poorly understood. Here we identify the RNA-binding protein LIN28B as a critical regulator of developmental timing in the murine cochlea. We show that Lin28b and its opposing let-7 miRNAs are differentially expressed in the auditory sensory lineage, with Lin28b being highly expressed in undifferentiated prosensory cells and let-7 miRNAs being highly expressed in their progeny—hair cells (HCs) and supporting cells (SCs). Using recently developed transgenic mouse models for LIN28B and let-7g, we demonstrate that prolonged LIN28B expression delays prosensory cell cycle withdrawal and differentiation, resulting in HC and SC patterning and maturation defects. Surprisingly, let-7g overexpression, although capable of inducing premature prosensory cell cycle exit, failed to induce premature HC differentiation, suggesting that LIN28B’s functional role in the timing of differentiation uses let-7 independent mechanisms. Finally, we demonstrate that overexpression of LIN28B or let-7g can significantly alter the postnatal production of HCs in response to Notch inhibition; LIN28B has a positive effect on HC production, whereas let-7 antagonizes this process. Together, these results implicate a key role for the LIN28B/let-7 axis in regulating postnatal SC plasticity.
Стилі APA, Harvard, Vancouver, ISO та ін.
Ми пропонуємо знижки на всі преміум-плани для авторів, чиї праці увійшли до тематичних добірок літератури. Зв'яжіться з нами, щоб отримати унікальний промокод!

До бібліографії