Статті в журналах з теми "Cartilage cells"

Щоб переглянути інші типи публікацій з цієї теми, перейдіть за посиланням: Cartilage cells.

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

Оберіть тип джерела:

Ознайомтеся з топ-50 статей у журналах для дослідження на тему "Cartilage cells".

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

Åberg, Thomas, Ritva Rice, David Rice, Irma Thesleff, and Janna Waltimo-Sirén. "Chondrogenic Potential of Mouse Calvarial Mesenchyme." Journal of Histochemistry & Cytochemistry 53, no. 5 (May 2005): 653–63. http://dx.doi.org/10.1369/jhc.4a6518.2005.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Facial and calvarial bones form intramembranously without a cartilagenous model; however, cultured chick calvarial mesenchyme cells may differentiate into both osteoblasts and chondroblasts and, in rodents, small cartilages occasionally form at the sutures in vivo. Therefore, we wanted to investigate what factors regulate normal differentiation of calvarial mesenchymal cells directly into osteoblasts. In embryonic mouse heads and in cultured tissue explants, we analyzed the expression of selected transcription factors and extracellular matrix molecules associated with bone and cartilage development. Cartilage markers Sox9 and type II collagen were expressed in all craniofacial cartilages. In addition, Msx2 and type I collagen were expressed in sense capsule cartilages. We also observed that the undifferentiated calvarial mesenchyme and the osteogenic fronts in the jaw expressed Co∗∗∗l2A1. Moreover, we found that cultured mouse calvarial mesenchyme could develop into cartilage. Of the 49 explants that contained mesenchyme, intramembranous ossification occurred in 35%. Only cartilage formed in 4%, and both cartilage and bone formed in 4%. Our study confirms that calvarial mesenchyme, which normally gives rise to intramembranous bone, also has chondrogenic potential.
2

Holmbeck, Kenn, Paolo Bianco, Kali Chrysovergis, Susan Yamada, and Henning Birkedal-Hansen. "MT1-MMP–dependent, apoptotic remodeling of unmineralized cartilage." Journal of Cell Biology 163, no. 3 (November 10, 2003): 661–71. http://dx.doi.org/10.1083/jcb.200307061.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Skeletal tissues develop either by intramembranous ossification, where bone is formed within a soft connective tissue, or by endochondral ossification. The latter proceeds via cartilage anlagen, which through hypertrophy, mineralization, and partial resorption ultimately provides scaffolding for bone formation. Here, we describe a novel and essential mechanism governing remodeling of unmineralized cartilage anlagen into membranous bone, as well as tendons and ligaments. Membrane-type 1 matrix metalloproteinase (MT1-MMP)–dependent dissolution of unmineralized cartilages, coupled with apoptosis of nonhypertrophic chondrocytes, mediates remodeling of these cartilages into other tissues. The MT1-MMP deficiency disrupts this process and uncouples apoptotic demise of chondrocytes and cartilage degradation, resulting in the persistence of “ghost” cartilages with adverse effects on skeletal integrity. Some cells entrapped in these ghost cartilages escape apoptosis, maintain DNA synthesis, and assume phenotypes normally found in the tissues replacing unmineralized cartilages. The coordinated apoptosis and matrix metalloproteinase-directed cartilage dissolution is akin to metamorphosis and may thus represent its evolutionary legacy in mammals.
3

Yi, Hee-Gyeong, Yeong-Jin Choi, Jin Woo Jung, Jinah Jang, Tae-Ha Song, Suhun Chae, Minjun Ahn, Tae Hyun Choi, Jong-Won Rhie, and Dong-Woo Cho. "Three-dimensional printing of a patient-specific engineered nasal cartilage for augmentative rhinoplasty." Journal of Tissue Engineering 10 (January 2019): 204173141882479. http://dx.doi.org/10.1177/2041731418824797.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Autologous cartilages or synthetic nasal implants have been utilized in augmentative rhinoplasty to reconstruct the nasal shape for therapeutic and cosmetic purposes. Autologous cartilage is considered to be an ideal graft, but has drawbacks, such as limited cartilage source, requirements of additional surgery for obtaining autologous cartilage, and donor site morbidity. In contrast, synthetic nasal implants are abundantly available but have low biocompatibility than the autologous cartilages. Moreover, the currently used nasal cartilage grafts involve additional reshaping processes, by meticulous manual carving during surgery to fit the diverse nose shape of each patient. The final shapes of the manually tailored implants are highly dependent on the surgeons’ proficiency and often result in patient dissatisfaction and even undesired separation of the implant. This study describes a new process of rhinoplasty, which integrates three-dimensional printing and tissue engineering approaches. We established a serial procedure based on computer-aided design to generate a three-dimensional model of customized nasal implant, and the model was fabricated through three-dimensional printing. An engineered nasal cartilage implant was generated by injecting cartilage-derived hydrogel containing human adipose-derived stem cells into the implant containing the octahedral interior architecture. We observed remarkable expression levels of chondrogenic markers from the human adipose-derived stem cells grown in the engineered nasal cartilage with the cartilage-derived hydrogel. In addition, the engineered nasal cartilage, which was implanted into mouse subcutaneous region, exhibited maintenance of the exquisite shape and structure, and striking formation of the cartilaginous tissues for 12 weeks. We expect that the developed process, which combines computer-aided design, three-dimensional printing, and tissue-derived hydrogel, would be beneficial in generating implants of other types of tissue.
4

Mazor, Marija, Annabelle Cesaro, Mazen Ali, Thomas M. Best, Eric Lespessailles, and Hechmi Toumi. "Progenitor Cells From Cartilage." Medicine & Science in Sports & Exercise 49, no. 5S (May 2017): 681. http://dx.doi.org/10.1249/01.mss.0000518798.14205.0d.

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

Benjamin, M., C. W. Archer, and J. R. Ralphs. "Cytoskeleton of cartilage cells." Microscopy Research and Technique 28, no. 5 (August 1, 1994): 372–77. http://dx.doi.org/10.1002/jemt.1070280503.

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

Suchorska, Wiktoria Maria, Ewelina Augustyniak, Magdalena Richter, Magdalena Łukjanow, Violetta Filas, Jacek Kaczmarczyk, and Tomasz Trzeciak. "Modified methods for efficiently differentiating human embryonic stem cells into chondrocyte-like cells." Postępy Higieny i Medycyny Doświadczalnej 71, no. 1 (June 19, 2017): 0. http://dx.doi.org/10.5604/01.3001.0010.3831.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Human articular cartilage has a poor regenerative capacity. This often results in the serious joint disease- osteoarthritis (OA) that is characterized by cartilage degradation. An inability to self-repair provided extensive studies on AC regeneration. The cell-based cartilage tissue engineering is a promising approach for cartilage regeneration. So far, numerous cell types have been reported to show chondrogenic potential, among others human embryonic stem cells (hESCs).
7

Le, Hanxiang, Weiguo Xu, Xiuli Zhuang, Fei Chang, Yinan Wang, and Jianxun Ding. "Mesenchymal stem cells for cartilage regeneration." Journal of Tissue Engineering 11 (January 2020): 204173142094383. http://dx.doi.org/10.1177/2041731420943839.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Cartilage injuries are typically caused by trauma, chronic overload, and autoimmune diseases. Owing to the avascular structure and low metabolic activities of chondrocytes, cartilage generally does not self-repair following an injury. Currently, clinical interventions for cartilage injuries include chondrocyte implantation, microfracture, and osteochondral transplantation. However, rather than restoring cartilage integrity, these methods only postpone further cartilage deterioration. Stem cell therapies, especially mesenchymal stem cell (MSCs) therapies, were found to be a feasible strategy in the treatment of cartilage injuries. MSCs can easily be isolated from mesenchymal tissue and be differentiated into chondrocytes with the support of chondrogenic factors or scaffolds to repair damaged cartilage tissue. In this review, we highlighted the full success of cartilage repair using MSCs, or MSCs in combination with chondrogenic factors and scaffolds, and predicted their pros and cons for prospective translation to clinical practice.
8

Zhang, Hong, Xiaopeng Zhao, Zhiguang Zhang, Weiwei Chen, and Xinli Zhang. "An Immunohistochemistry Study of Sox9, Runx2, and Osterix Expression in the Mandibular Cartilages of Newborn Mouse." BioMed Research International 2013 (2013): 1–11. http://dx.doi.org/10.1155/2013/265380.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
The purpose of this study is to investigate the spacial expression pattern and functional significance of three key transcription factors related to bone and cartilage formation, namely, Sox9, Runx2, and Osterix in cartilages during the late development of mouse mandible. Immunohistochemical examinations of Sox9, Runx2, and Osterix were conducted in the mandibular cartilages of the 15 neonatal C57BL/6N mice. In secondary cartilages, both Sox9 and Runx2 were weakly expressed in the polymorphic cell zone, strongly expressed in the flattened cell zone and throughout the entire hypertrophic cell zone. Similarly, both transcriptional factors were weakly expressed in the uncalcified Meckel’s cartilage while strongly expressed in the rostral cartilage. Meanwhile, Osterix was at an extremely low level in cells of the flattened cell zone and the upper hypertrophic cell zone in secondary cartilages. Surprisingly, Osterix was intensely expressed in hypertrophic chondrocytes in the center of the uncalcified Meckel’s cartilage while moderately expressed in part of hypertrophic chondrocytes in the rostral process. Consequently, it is suggested that Sox9 is a main and unique positive regulator in the hypertrophic differentiation process of mandibular secondary cartilages, in addition to Runx2. Furthermore, Osterix is likely responsible for phenotypic conversion of Meckel’s chondrocytes during its degeneration.
9

Hayes, Anthony J., John Whitelock, and James Melrose. "Regulation of FGF-2, FGF-18 and Transcription Factor Activity by Perlecan in the Maturational Development of Transitional Rudiment and Growth Plate Cartilages and in the Maintenance of Permanent Cartilage Homeostasis." International Journal of Molecular Sciences 23, no. 4 (February 9, 2022): 1934. http://dx.doi.org/10.3390/ijms23041934.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
The aim of this study was to highlight the roles of perlecan in the regulation of the development of the rudiment developmental cartilages and growth plate cartilages, and also to show how perlecan maintains permanent articular cartilage homeostasis. Cartilage rudiments are transient developmental templates containing chondroprogenitor cells that undergo proliferation, matrix deposition, and hypertrophic differentiation. Growth plate cartilage also undergoes similar changes leading to endochondral bone formation, whereas permanent cartilage is maintained as an articular structure and does not undergo maturational changes. Pericellular and extracellular perlecan-HS chains interact with growth factors, morphogens, structural matrix glycoproteins, proteases, and inhibitors to promote matrix stabilization and cellular proliferation, ECM remodelling, and tissue expansion. Perlecan has mechanotransductive roles in cartilage that modulate chondrocyte responses in weight-bearing environments. Nuclear perlecan may modulate chromatin structure and transcription factor access to DNA and gene regulation. Snail-1, a mesenchymal marker and transcription factor, signals through FGFR-3 to promote chondrogenesis and maintain Acan and type II collagen levels in articular cartilage, but prevents further tissue expansion. Pre-hypertrophic growth plate chondrocytes also express high Snail-1 levels, leading to cessation of Acan and CoI2A1 synthesis and appearance of type X collagen. Perlecan differentially regulates FGF-2 and FGF-18 to maintain articular cartilage homeostasis, rudiment and growth plate cartilage growth, and maturational changes including mineralization, contributing to skeletal growth.
10

Schilling, T. F., C. Walker, and C. B. Kimmel. "The chinless mutation and neural crest cell interactions in zebrafish jaw development." Development 122, no. 5 (May 1, 1996): 1417–26. http://dx.doi.org/10.1242/dev.122.5.1417.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
During vertebrate development, neural crest cells are thought to pattern many aspects of head organization, including the segmented skeleton and musculature of the jaw and gills. Here we describe mutations at the gene chinless, chn, that disrupt the skeletal fates of neural crest cells in the head of the zebrafish and their interactions with muscle precursors. chn mutants lack neural-crest-derived cartilage and mesoderm-derived muscles in all seven pharyngeal arches. Fate mapping and gene expression studies demonstrate the presence of both undifferentiated cartilage and muscle precursors in mutants. However, chn blocks differentiation directly in neural crest, and not in mesoderm, as revealed by mosaic analyses. Neural crest cells taken from wild-type donor embryos can form cartilage when transplanted into chn mutant hosts and rescue some of the patterning defects of mutant pharyngeal arches. In these cases, cartilage only forms if neural crest is transplanted at least one hour before its migration, suggesting that interactions occur transiently in early jaw precursors. In contrast, transplanted cells in paraxial mesoderm behave according to the host genotype; mutant cells form jaw muscles in a wild-type environment. These results suggest that chn is required for the development of pharyngeal cartilages from cranial neural crest cells and subsequent crest signals that pattern mesodermally derived myocytes.
11

Mackie, E. J., I. Thesleff, and R. Chiquet-Ehrismann. "Tenascin is associated with chondrogenic and osteogenic differentiation in vivo and promotes chondrogenesis in vitro." Journal of Cell Biology 105, no. 6 (December 1, 1987): 2569–79. http://dx.doi.org/10.1083/jcb.105.6.2569.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
The tissue distribution of the extracellular matrix glycoprotein, tenascin, during cartilage and bone development in rodents has been investigated by immunohistochemistry. Tenascin was present in condensing mesenchyme of cartilage anlagen, but not in the surrounding mesenchyme. In fully differentiated cartilages, tenascin was only present in the perichondrium. In bones that form by endochondral ossification, tenascin reappeared around the osteogenic cells invading the cartilage model. Tenascin was also present in the condensing mesenchyme of developing bones that form by intramembranous ossification and later was present around the spicules of forming bone. Tenascin was absent from mature bone matrix but persisted on periosteal and endosteal surfaces. Immunofluorescent staining of wing bud cultures from chick embryos showed large amounts of tenascin in the forming cartilage nodules. Cultures grown on a substrate of tenascin produced more cartilage nodules than cultures grown on tissue culture plastic. Tenascin in the culture medium inhibited the attachment of wing bud cells to fibronectin-coated substrates. We propose that tenascin plays an important role in chondrogenesis by modulating fibronectin-cell interactions and causing cell rounding and condensation.
12

Jian, Quan-Liang, Wei-Chun HuangFu, Yen-Hua Lee, and I.-Hsuan Liu. "Age, but not short-term intensive swimming, affects chondrocyte turnover in zebrafish vertebral cartilage." PeerJ 6 (October 1, 2018): e5739. http://dx.doi.org/10.7717/peerj.5739.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Both age and intensive exercise are generally considered critical risk factors for osteoarthritis. In this work, we intend to establish zebrafish models to assess the role of these two factors on cartilage homeostasis. We designed a swimming device for zebrafish intensive exercise. The body measurements, bone mineral density (BMD) and the histology of spinal cartilages of 4- and 12-month-old zebrafish, as well the 12-month-old zebrafish before and after a 2-week exercise were compared. Our results indicate that both age and exercise affect the body length and body weight, and the micro-computed tomography reveals that both age and exercise affect the spinal BMD. However, quantitative analysis of immunohistochemistry and histochemistry indicate that short-term intensive exercise does not affect the extracellular matrix (ECM) of spinal cartilage. On the other hand, the cartilage ECM significantly grew from 4 to 12 months of age with an increase in total chondrocytes. dUTP nick end labeling staining shows that the percentages of apoptotic cells significantly increase as the zebrafish grows, whereas the BrdU labeling shows that proliferative cells dramatically decrease from 4 to 12 months of age. A 30-day chase of BrdU labeling shows some retention of labeling in cells in 4-month-old spinal cartilage but not in cartilage from 12-month-old zebrafish. Taken together, our results suggest that zebrafish chondrocytes are actively turned over, and indicate that aging is a critical factor that alters cartilage homeostasis. Zebrafish vertebral cartilage may serve as a good model to study the maturation and homeostasis of articular cartilage.
13

Kurenkova, Anastasiia D., Irina A. Romanova, Pavel D. Kibirskiy, Peter Timashev, and Ekaterina V. Medvedeva. "Strategies to Convert Cells into Hyaline Cartilage: Magic Spells for Adult Stem Cells." International Journal of Molecular Sciences 23, no. 19 (September 22, 2022): 11169. http://dx.doi.org/10.3390/ijms231911169.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Damaged hyaline cartilage gradually decreases joint function and growing pain significantly reduces the quality of a patient’s life. The clinically approved procedure of autologous chondrocyte implantation (ACI) for treating knee cartilage lesions has several limits, including the absence of healthy articular cartilage tissues for cell isolation and difficulties related to the chondrocyte expansion in vitro. Today, various ACI modifications are being developed using autologous chondrocytes from alternative sources, such as the auricles, nose and ribs. Adult stem cells from different tissues are also of great interest due to their less traumatic material extraction and their innate abilities of active proliferation and chondrogenic differentiation. According to the different adult stem cell types and their origin, various strategies have been proposed for stem cell expansion and initiation of their chondrogenic differentiation. The current review presents the diversity in developing applied techniques based on autologous adult stem cell differentiation to hyaline cartilage tissue and targeted to articular cartilage damage therapy.
14

Kirasirova, E. A., N. V. Lafutkina, R. A. Rezakov, R. F. Mamedov, and I. F. Al-Assaf. "PATHOMORPHOLOGICAL CHANGES IN THE CARTILAGE OF THE TRACHEA DEPENDING ON TERMS OF THE INTUBATION." Folia Otorhinolaryngologiae et Pathologiae Respiratoriae 25, no. 3 (2019): 87–93. http://dx.doi.org/10.33848/foliorl23103825-2019-25-3-87-93.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Objective - to study the nature and prevalence of pathological changes in the cartilage of the trachea depending on the duration of intubation according to the results of pathomorphological studies. Materials and methods. Pathomorphological study of cartilage of the anterior tracheal wall was carried out on 37 patients at different times of mechanical ventilation. Depending on the timing of the ventilator before the tracheostomy, all patients were divided into three groups. In 10 people, the duration of ventilation until tracheostomy was no more than 3 days, in 15 people - 4 -7 days and in 12 people - more than 7 days. Results. On the 1st day of mechanical ventilation, dystrophic changes are determined, with the loss of cartilage cells. On the 2nd day of mechanical ventilation in the cartilage of the trachea significant dystrophic changes with pycnosis of the nuclei in chondrocytes were revealed. By the third day of mechanical ventilation, detachment of perichondria occurred, the surface of cartilage lacking perichondria was usurized, covered with fibrinous overlays, the cartilage no longer contained cartilage cells; by the 7th day of mechanical ventilation, deeper dystrophic and destructive changes were revealed in the tracheal cartilages - there was no perichondria, the cartilage surface was usurized, foci of chondronecrosis were observed in the areas adjacent to the damaged perichondria; by the tenth days of mechanical ventilation, the pathomorphological picture indicates the death of the cartilage and its replacement with granulation tissue, the presence of regeneration processes, expressed in focal proliferation of chondrocytes, thickening and fibrosis of the perichondria, the appearance and sequestration of dead cartilage. Conclusion. The severity of pathomorphological changes in the cartilage of the trachea due to the duration of intubation
15

Wang, Mingjie, Zhiguo Yuan, Ning Ma, Chunxiang Hao, Weimin Guo, Gengyi Zou, Yu Zhang, et al. "Advances and Prospects in Stem Cells for Cartilage Regeneration." Stem Cells International 2017 (2017): 1–16. http://dx.doi.org/10.1155/2017/4130607.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
The histological features of cartilage call attention to the fact that cartilage has a little capacity to repair itself owing to the lack of a blood supply, nerves, or lymphangion. Stem cells have emerged as a promising option in the field of cartilage tissue engineering and regenerative medicine and could lead to cartilage repair. Much research has examined cartilage regeneration utilizing stem cells. However, both the potential and the limitations of this procedure remain controversial. This review presents a summary of emerging trends with regard to using stem cells in cartilage tissue engineering and regenerative medicine. In particular, it focuses on the characterization of cartilage stem cells, the chondrogenic differentiation of stem cells, and the various strategies and approaches involving stem cells that have been used in cartilage repair and clinical studies. Based on the research into chondrocyte and stem cell technologies, this review discusses the damage and repair of cartilage and the clinical application of stem cells, with a view to increasing our systematic understanding of the application of stem cells in cartilage regeneration; additionally, several advanced strategies for cartilage repair are discussed.
16

Soliman, Soha A., Basma Mohamed Kamal, and Hanan H. Abd-Elhafeez. "Cellular Invasion and Matrix Degradation, a Different Type of Matrix-Degrading Cells in the Cartilage of Catfish (Clarias gariepinus) and Japanese Quail Embryos (Coturnix coturnix japonica)." Microscopy and Microanalysis 25, no. 05 (October 2019): 1283–92. http://dx.doi.org/10.1017/s1431927619014892.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
AbstractWe previously studied the phenomena of the mesenchymal cell-dependent mode of cartilage growth in quail and catfish. Thus, we selected the two cartilage models in which mesenchymal cells participate in their growth. In such models, cartilage degradation occurred to facilitate cellular invasion. The studies do not explain the nature of the cartilage degrading cells. The current study aims to explore the nature of the cartilage-degrading cells using transmission electron microscopy (TEM) and immunohistochemistry. Samples of cartilage have been isolated from the air-breathing organ of catfish and the cartilage of the prospective occipital bone of quail embryos. Samples have been processed for TEM and immunohistochemistry. We found that two different cell types are involved in cartilage degradation; the macrophage in the cartilage of catfish and mesenchymal cells in the cartilage of the quail. Areas of cellular invasion in both catfish cartilage and quail embryo cartilage had an immunological affinity for MMP-9. In catfish, cartilage-degrading cells had identical morphological features of macrophages, whereas in quail embryos, cartilage-degrading cells were mesenchymal-like cells which had cell processes rich in vesicles and expressed CD117. Further study should consider the role of macrophage and mesenchymal cells during cartilage degradation. This could be valuable to be applied to remove the defective cartilage matrix formed in osteoarthritic patients to improve cartilage repair strategies.
17

Peng, Haining, Yi Zhang, Zhongkai Ren, Ziran Wei, Renjie Chen, Yingze Zhang, Xiaohong Huang, and Tengbo Yu. "Cartilaginous Metabolomics Reveals the Biochemical-Niche Fate Control of Bone Marrow-Derived Stem Cells." Cells 11, no. 19 (September 21, 2022): 2951. http://dx.doi.org/10.3390/cells11192951.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Joint disorders have become a global health issue with the growth of the aging population. Screening small active molecules targeting chondrogenic differentiation of bone marrow-derived stem cells (BMSCs) is of urgency. In this study, microfracture was employed to create a regenerative niche in rabbits (n = 9). Cartilage samples were collected four weeks post-surgery. Microfracture-caused morphological (n = 3) and metabolic (n = 6) changes were detected. Non-targeted metabolomic analysis revealed that there were 96 differentially expressed metabolites (DEMs) enriched in 70 pathways involved in anti-inflammation, lipid metabolism, signaling transduction, etc. Among the metabolites, docosapentaenoic acid 22n-3 (DPA) and ursodeoxycholic acid (UDCA) functionally facilitated cartilage defect healing, i.e., increasing the vitality and adaptation of the BMSCs, chondrogenic differentiation, and chondrocyte functionality. Our findings firstly reveal the differences in metabolomic activities between the normal and regenerated cartilages and provide a list of endogenous biomolecules potentially involved in the biochemical-niche fate control for chondrogenic differentiation of BMSCs. Ultimately, the biomolecules may serve as anti-aging supplements for chondrocyte renewal or as drug candidates for cartilage regenerative medicine.
18

Zhang, Jianying, Shiwu Dong, Wesley Sivak, Hui Bin Sun, and Kai Tao. "Stem Cells in Cartilage Regeneration." Stem Cells International 2017 (2017): 1–2. http://dx.doi.org/10.1155/2017/7034726.

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

Kistler, Andreas, Brigitta Galli, and Herbert Kuhn. "Retinoic acid-induced cartilage degradation is caused by cartilage cells." Roux's Archives of Developmental Biology 199, no. 7 (July 1991): 377–86. http://dx.doi.org/10.1007/bf01705847.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
20

CHEN, JING, CHUNGEN GUO, HONGSHENG LI, XIAOQIN ZHU, SHUYUAN XIONG, and JIANXIN CHEN. "NONLINEAR SPECTRAL IMAGING OF ELASTIC CARTILAGE IN RABBIT EARS." Journal of Innovative Optical Health Sciences 06, no. 03 (July 2013): 1350024. http://dx.doi.org/10.1142/s1793545813500247.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Elastic cartilage in the rabbit external ear is an important animal model with attractive potential value for researching the physiological and pathological states of cartilages especially during wound healing. In this work, nonlinear optical microscopy based on two-photon excited fluorescence and second harmonic generation were employed for imaging and quantifying the intact elastic cartilage. The morphology and distribution of main components in elastic cartilage including cartilage cells, collagen and elastic fibers were clearly observed from the high-resolution two-dimensional nonlinear optical images. The areas of cell nuclei, a parameter related to the pathological changes of normal or abnormal elastic cartilage, can be easily quantified. Moreover, the three-dimensional structure of chondrocytes and matrix were displayed by constructing three-dimensional image of cartilage tissue. At last, the emission spectra from cartilage were obtained and analyzed. We found that the different ratio of collagen over elastic fibers can be used to locate the observed position in the elastic cartilage. The redox ratio based on the ratio of nicotinamide adenine dinucleotide (NADH) over flavin adenine dinucleotide (FAD) fluorescence can also be calculated to analyze the metabolic state of chondrocytes in different regions. Our results demonstrated that this technique has the potential to provide more accurate and comprehensive information for the physiological states of elastic cartilage.
21

Longo, Umile Giuseppe, Stefano Petrillo, Edoardo Franceschetti, Alessandra Berton, Nicola Maffulli, and Vincenzo Denaro. "Stem Cells and Gene Therapy for Cartilage Repair." Stem Cells International 2012 (2012): 1–9. http://dx.doi.org/10.1155/2012/168385.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Cartilage defects represent a common problem in orthopaedic practice. Predisposing factors include traumas, inflammatory conditions, and biomechanics alterations. Conservative management of cartilage defects often fails, and patients with this lesions may need surgical intervention. Several treatment strategies have been proposed, although only surgery has been proved to be predictably effective. Usually, in focal cartilage defects without a stable fibrocartilaginous repair tissue formed, surgeons try to promote a natural fibrocartilaginous response by using marrow stimulating techniques, such as microfracture, abrasion arthroplasty, and Pridie drilling, with the aim of reducing swelling and pain and improving joint function of the patients. These procedures have demonstrated to be clinically useful and are usually considered as first-line treatment for focal cartilage defects. However, fibrocartilage presents inferior mechanical and biochemical properties compared to normal hyaline articular cartilage, characterized by poor organization, significant amounts of collagen type I, and an increased susceptibility to injury, which ultimately leads to premature osteoarthritis (OA). Therefore, the aim of future therapeutic strategies for articular cartilage regeneration is to obtain a hyaline-like cartilage repair tissue by transplantation of tissues or cells. Further studies are required to clarify the role of gene therapy and mesenchimal stem cells for management of cartilage lesions.
22

Ranger, Ann M., Louis C. Gerstenfeld, Jinxi Wang, Tamiyo Kon, Hyunsu Bae, Ellen M. Gravallese, Melvin J. Glimcher, and Laurie H. Glimcher. "The Nuclear Factor of Activated T Cells (Nfat) Transcription Factor Nfatp (Nfatc2) Is a Repressor of Chondrogenesis." Journal of Experimental Medicine 191, no. 1 (January 3, 2000): 9–22. http://dx.doi.org/10.1084/jem.191.1.9.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Nuclear factor of activated T cells (NFAT) transcription factors regulate gene expression in lymphocytes and control cardiac valve formation. Here, we report that NFATp regulates chondrogenesis in the adult animal. In mice lacking NFATp, resident cells in the extraarticular connective tissues spontaneously differentiate to cartilage. These cartilage cells progressively differentiate and the tissue undergoes endochondral ossification, recapitulating the development of endochondral bone. Proliferation of already existing articular cartilage cells also occurs in some older animals. At both sites, neoplastic changes in the cartilage cells occur. Consistent with these data, NFATp expression is regulated in mesenchymal stem cells induced to differentiate along a chondrogenic pathway. Lack of NFATp in articular cartilage cells results in increased expression of cartilage markers, whereas overexpression of NFATp in cartilage cell lines extinguishes the cartilage phenotype. Thus, NFATp is a repressor of cartilage cell growth and differentiation and also has the properties of a tumor suppressor.
23

Bae, Jung Yoon, Kazuaki Matsumura, Shigeyuki Wakitani, Amu Kawaguchi, Sadami Tsutsumi, and Suong-Hyu Hyon. "Beneficial Storage Effects of Epigallocatechin-3-O-Gallate on the Articular Cartilage of Rabbit Osteochondral Allografts." Cell Transplantation 18, no. 5-6 (May 2009): 505–12. http://dx.doi.org/10.1177/096368970901805-604.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
A fresh osteochondral allograft is one of the most effective treatments for cartilage defects of the knee. Despite the clinical success, fresh osteochondral allografts have great limitations in relation to the short storage time that cartilage tissues can be well-preserved. Fresh osteochondral grafts are generally stored in culture medium at 4°C. While the viability of articular cartilage stored in culture medium is significantly diminished within 1 week, appropriate serology testing to minimize the chances for the disease transmission requires a minimum of 2 weeks. (–)-Epigallocatechin-3- O-gallate (EGCG) has differential effects on the proliferation of cancer and normal cells, thus a cytotoxic effect on various cancer cells, but a cytopreservative effect on normal cells. Therefore, a storage solution containing EGCG might extend the storage duration of articular cartilages. Rabbit osteochondral allografts were performed with osteochondral grafts stored at 4°C in culture medium containing EGCG for 2 weeks and then the clinical effects were examined with macroscopic and histological assessment after 4 weeks. The cartilaginous structure of an osteochondral graft stored with EGCG was well-preserved with high cell viability and glycosaminoglycan (GAG) content of the extracellular matrix (ECM). After an osteochondral allograft, the implanted osteochondral grafts stored with EGCG also provided a significantly better retention of the articular cartilage with viability and metabolic activity. These data suggest that EGCG can be an effective storage agent that allows long-term preservation of articular cartilage under cold storage conditions.
24

A Soliman, Soha. "MMP-9 Expression in Normal Rabbit Chondrocytes." Cytology & Histology International Journal 5, no. 1 (2021): 1–9. http://dx.doi.org/10.23880/chij-16000131.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Chondrocytes regulate anabolic and catabolic processes to maintain the extracellular matrix components. Catabolic activities depend on the proteolytic action of the matrix -degrading enzymes including ADAMTS (A disintegrin and metalloproteinases) and MMP (Matrix Metalloproteinase). The current study explored the distribution of MMP-9 in normal articular cartilages of the embryos rabbit. Articular cartilage has grown by appositional growth that the perichondrial stem cells differentiate into chondrocytes. MMP-9 positive perichondrial stem cells or chondroblasts and early chondrocytes. Mature chondrocytes exhibited weak immunoaffinity for MMP-9. In conclusion, MMP-9 was essential during chondrocytes growth. The current study alludes to the potential role of MMP-9 during the growth of the articular cartilage.
25

Sun, GW, H. Kobayashi, M. Suzuki, N. Kanayama, and T. Terao. "Production of cartilage link protein by human granulosa-lutein cells." Journal of Endocrinology 175, no. 2 (November 1, 2002): 505–15. http://dx.doi.org/10.1677/joe.0.1750505.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Link protein (LP), an extracellular matrix protein in cartilage, stabilizes aggregates of hyaluronic acid (HA) and proteoglycans, including aggrecan and inter-alpha-trypsin inhibitor (ITI). We have shown previously that cartilage LP is present in the maturing rat and mouse ovary. In the present study, we have employed immunohistochemistry to examine the anatomical distribution of cartilage LP in the human ovary. The expression of cartilage LP was selectively detected in the cells within the granulosa compartment of the preovulatory dominant follicle. The HA-positive granulosa-lutein cells were found to be a cartilage LP-positive subpopulation. We subsequently studied the in vitro expression of cartilage LP in cultured human granulosa-lutein cells obtained at oocyte retrieval for in vitro fertilization. Analysis of cultured cells by enzyme-linked immunoaffinity assay, Western blotting and immunofluorescence microscopy revealed that gonadotropin stimulates cartilage LP production. Time-course studies indicated that the cartilage LP production was induced as early as with gonadotropin stimulation for 2 h, and the effect was sustained up to 8 h. Western blot analysis further revealed the presence of the macroaggregates composed of HA, ITI and cartilage LP in the gonadotropin-stimulated granulosa-lutein cell extracts. Collectively, the present results raise the possibility that cartilage LP forms extracellular structures that may have a regulatory function in the developing follicle in the human ovary.
26

Sen, Rwik, Sofia Pezoa, Lomeli Carpio Shull, Laura Hernandez-Lagunas, Lee Niswander, and Kristin Artinger. "Kat2a and Kat2b Acetyltransferase Activity Regulates Craniofacial Cartilage and Bone Differentiation in Zebrafish and Mice." Journal of Developmental Biology 6, no. 4 (November 12, 2018): 27. http://dx.doi.org/10.3390/jdb6040027.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Cranial neural crest cells undergo cellular growth, patterning, and differentiation within the branchial arches to form cartilage and bone, resulting in a precise pattern of skeletal elements forming the craniofacial skeleton. However, it is unclear how cranial neural crest cells are regulated to give rise to the different shapes and sizes of the bone and cartilage. Epigenetic regulators are good candidates to be involved in this regulation, since they can exert both broad as well as precise control on pattern formation. Here, we investigated the role of the histone acetyltransferases Kat2a and Kat2b in craniofacial development using TALEN/CRISPR/Cas9 mutagenesis in zebrafish and the Kat2ahat/hat (also called Gcn5) allele in mice. kat2a and kat2b are broadly expressed during embryogenesis within the central nervous system and craniofacial region. Single and double kat2a and kat2b zebrafish mutants have an overall shortening and hypoplastic nature of the cartilage elements and disruption of the posterior ceratobranchial cartilages, likely due to smaller domains of expression of both cartilage- and bone-specific markers, including sox9a and col2a1, and runx2a and runx2b, respectively. Similarly, in mice we observe defects in the craniofacial skeleton, including hypoplastic bone and cartilage and altered expression of Runx2 and cartilage markers (Sox9, Col2a1). In addition, we determined that following the loss of Kat2a activity, overall histone 3 lysine 9 (H3K9) acetylation, the main epigenetic target of Kat2a/Kat2b, was decreased. These results suggest that Kat2a and Kat2b are required for growth and differentiation of craniofacial cartilage and bone in both zebrafish and mice by regulating H3K9 acetylation.
27

Schilling, T. F., and C. B. Kimmel. "Musculoskeletal patterning in the pharyngeal segments of the zebrafish embryo." Development 124, no. 15 (August 1, 1997): 2945–60. http://dx.doi.org/10.1242/dev.124.15.2945.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
The head skeleton and muscles of the zebrafish develop in a stereotyped pattern in the embryo, including seven pharyngeal arches and a basicranium underlying the brain and sense organs. To investigate how individual cartilages and muscles are specified and organized within each head segment, we have examined their early differentiation using Alcian labeling of cartilage and expression of several molecular markers of muscle cells. Zebrafish larvae begin feeding by four days after fertilization, but cartilage and muscle precursors develop in the pharyngeal arches up to 2 days earlier. These chondroblasts and myoblasts lie close together within each segment and differentiate in synchrony, perhaps reflecting the interdependent nature of their patterning. Initially, cells within a segment condense and gradually become subdivided into individual dorsal and ventral structures of the differentiated arch. Cartilages or muscles in one segment show similar patterns of condensation and differentiation as their homologues in another, but vary in size and shape in the most anterior (mandibular and hyoid) and posterior (tooth-bearing) arches, possibly as a consequence of changes in the timing of their development. Our results reveal a segmental scaffold of early cartilage and muscle precursors and suggest that interactions between them coordinate their patterning in the embryo. These data provide a descriptive basis for genetic analyses of craniofacial patterning.
28

McBurney, Kim M., and Glenda M. Wright. "Chondrogenesis of a non-collagen-based cartilage in the sea lamprey, Petromyzon marinus." Canadian Journal of Zoology 74, no. 12 (December 1, 1996): 2118–30. http://dx.doi.org/10.1139/z96-241.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Chondrogenesis of the trabeculae, non-collagen-based cartilages in prolarval stages of the sea lamprey, Petromyzon marinus, was examined by light and electron microscopy. Chondrogenesis of the trabecular cartilages in prolarval lampreys commenced with the formation of mesenchymal condensations. Two peaks in mesenchymal cell density occurred, one prior to condensation formation and a second immediately before cartilage differentiation. The possibility of inductive influences by epithelio-mesenchymal interactions on the initiation of chondrogenesis is discussed. Bilateral condensations first appeared by day 17 post fertilization ventromedial to the eyes in a band of tightly packed yolk-laden mesenchymal cells that represent neural crest derived tissue. Cartilage differentiation occurred by day 19 post fertilization and was indicated by the presence of matrix-synthesizing organelles and the first ultrastructural appearance in the extracellular matrix of lamprin, a structural protein unique to lamprey cartilage. Lamprin was initially deposited as discrete 15- to 40-nm globules. Subsequently, lamprin appeared as fibrils aggregated into branching and parallel arrays arranged in pericellular, territorial, and interterritorial zones. Lengthening of the trabecular cartilages was primarily by appositional growth at the rostral end. The timing of the appearance of trabecular cartilages in prolarval stages likely reflects the functional importance of these structures for supporting the brain as the lamprey initiates burrowing behaviour.
29

Rim, Yeri Alice, Yoojun Nam, and Ji Hyeon Ju. "Application of Cord Blood and Cord Blood-Derived Induced Pluripotent Stem Cells for Cartilage Regeneration." Cell Transplantation 28, no. 5 (September 25, 2018): 529–37. http://dx.doi.org/10.1177/0963689718794864.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Regeneration of articular cartilage is of great interest in cartilage tissue engineering since articular cartilage has a low regenerative capacity. Due to the difficulty in obtaining healthy cartilage for transplantation, there is a need to develop an alternative and effective regeneration therapy to treat degenerative or damaged joint diseases. Stem cells including various adult stem cells and pluripotent stem cells are now actively used in tissue engineering. Here, we provide an overview of the current status of cord blood cells and induced pluripotent stem cells derived from these cells in cartilage regeneration. The abilities of these cells to undergo chondrogenic differentiation are also described. Finally, the technical challenges of articular cartilage regeneration and future directions are discussed.
30

Kotaka, Shinji, Shigeyuki Wakitani, Akira Shimamoto, Naosuke Kamei, Mikiya Sawa, Nobuo Adachi, and Mituo Ochi. "Magnetic Targeted Delivery of Induced Pluripotent Stem Cells Promotes Articular Cartilage Repair." Stem Cells International 2017 (December 26, 2017): 1–7. http://dx.doi.org/10.1155/2017/9514719.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Cartilage regeneration treatments using stem cells are associated with problems due to the cell source and the difficulty of delivering the cells to the cartilage defect. We consider labeled induced pluripotent stem (iPS) cells to be an ideal source of cells for tissue regeneration, and if iPS cells could be delivered only into cartilage defects, it would be possible to repair articular cartilage. Consequently, we investigated the effect of magnetically labeled iPS (m-iPS) cells delivered into an osteochondral defect by magnetic field on the repair of articular cartilage. iPS cells were labeled magnetically and assessed for maintenance of pluripotency by their ability to form embryoid bodies in vitro and to form teratomas when injected subcutaneously into nude rats. These cells were delivered specifically into cartilage defects in nude rats using a magnetic field. The samples were graded according to the histologic grading score for cartilage regeneration. m-iPS cells differentiated into three embryonic germ layers and formed teratomas in the subcutaneous tissue. The histologic grading score was significantly better in the treatment group compared to the control group. m-iPS cells maintained pluripotency, and the magnetic delivery system proved useful and safe for cartilage repair using iPS cells.
31

Wang, L., M. Lazebnik, and M. S. Detamore. "Hyaline cartilage cells outperform mandibular condylar cartilage cells in a TMJ fibrocartilage tissue engineering application." Osteoarthritis and Cartilage 17, no. 3 (March 2009): 346–53. http://dx.doi.org/10.1016/j.joca.2008.07.004.

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

Schmitt, Andreas, Martijn van Griensven, Andreas B. Imhoff, and Stefan Buchmann. "Application of Stem Cells in Orthopedics." Stem Cells International 2012 (2012): 1–11. http://dx.doi.org/10.1155/2012/394962.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Stem cell research plays an important role in orthopedic regenerative medicine today. Current literature provides us with promising results from animal research in the fields of bone, tendon, and cartilage repair. While early clinical results are already published for bone and cartilage repair, the data about tendon repair is limited to animal studies. The success of these techniques remains inconsistent in all three mentioned areas. This may be due to different application techniques varying from simple mesenchymal stem cell injection up to complex tissue engineering. However, the ideal carrier for the stem cells still remains controversial. This paper aims to provide a better understanding of current basic research and clinical data concerning stem cell research in bone, tendon, and cartilage repair. Furthermore, a focus is set on different stem cell application techniques in tendon reconstruction, cartilage repair, and filling of bone defects.
33

Zheng, Min. "Stem Cells Promote the Regeneration of Knee Joint Degenerative Bone and Articular Cartilage." Journal of Healthcare Engineering 2022 (March 24, 2022): 1–7. http://dx.doi.org/10.1155/2022/9533211.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Cartilage damage has a certain ability to spontaneously repair, but the repaired tissue often shows the phenomenon of cartilage terminal differentiation, which causes irreversible damage to its structure and function and seriously affects the quality of life and work of patients. It is of great significance to study the problems encountered in the process of cartilage damage repair. This article mainly studied stem cells to promote the regeneration of knee joint degenerative bone articular cartilage. First, the animal articular cartilage defect is modeled, 10 ml of animal venous blood is drawn, 0.5 ml of PRP is collected by centrifugation, mixed with cartilage fragments, and transplanted into the defect area into a gel. In the BMSCs group, 1 ml of BMSCs with a cell concentration of 107 cells/ml was injected intra-articularly. The histological chromosomes were observed after 6 weeks and 12 weeks, and the effect of cartilage tissue repair was analyzed and evaluated, and the related data were statistically analyzed. We evaluated the spontaneous repair ability of partial cartilage damage, full-thickness cartilage damage, and osteochondral damage. Furthermore, for partial cartilage damage repair, by using the cartilage damage in vitro model and biomaterials to simulate the in vivo microenvironment, the adhesion and cell morphology on the surface of partial- and full-thickness cartilage damage were evaluated, and the experiments were further used to evaluate the exogenous and internal induced migration effect of source on cultured cells in vitro. In the cell concentration study, the cartilage repair effect increased with the increase in concentration within a certain range, and the tissue repair ability remained stable when the concentration exceeded 107 cells/ml. Using ECM-oriented scaffolds to compound autologous BMSCs, tissue-engineered cartilage was successfully constructed, which had the histological and biochemical characteristics of normal cartilage tissue, and better repaired the damaged articular cartilage of large animals.
34

Enomura, Masahiro, Soichiro Murata, Yuri Terado, Maiko Tanaka, Shinji Kobayashi, Takayoshi Oba, Shintaro Kagimoto, et al. "Development of a Method for Scaffold-Free Elastic Cartilage Creation." International Journal of Molecular Sciences 21, no. 22 (November 11, 2020): 8496. http://dx.doi.org/10.3390/ijms21228496.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Microtia is a congenital aplasia of the auricular cartilage. Conventionally, autologous costal cartilage grafts are collected and shaped for transplantation. However, in this method, excessive invasion occurs due to limitations in the costal cartilage collection. Due to deformation over time after transplantation of the shaped graft, problems with long-term morphological maintenance exist. Additionally, the lack of elasticity with costal cartilage grafts is worth mentioning, as costal cartilage is a type of hyaline cartilage. Medical plastic materials have been transplanted as alternatives to costal cartilage, but transplant rejection and deformation over time are inevitable. It is imperative to create tissues for transplantation using cells of biological origin. Hence, cartilage tissues were developed using a biodegradable scaffold material. However, such materials suffer from transplant rejection and biodegradation, causing the transplanted cartilage tissue to deform due to a lack of elasticity. To address this problem, we established a method for creating elastic cartilage tissue for transplantation with autologous cells without using scaffold materials. Chondrocyte progenitor cells were collected from perichondrial tissue of the ear cartilage. By using a multilayer culture and a three-dimensional rotating suspension culture vessel system, we succeeded in creating scaffold-free elastic cartilage from cartilage progenitor cells.
35

Kaplan, David. "Role of cartilage-forming cells in regenerative medicine for cartilage repair." Orthopedic Research and Reviews Volume 2 (September 2010): 85–94. http://dx.doi.org/10.2147/orr.s7194.

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

Messaoudi, Océane, Christel Henrionnet, Kevin Bourge, Damien Loeuille, Pierre Gillet, and Astrid Pinzano. "Stem Cells and Extrusion 3D Printing for Hyaline Cartilage Engineering." Cells 10, no. 1 (December 22, 2020): 2. http://dx.doi.org/10.3390/cells10010002.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Hyaline cartilage is deficient in self-healing properties. The early treatment of focal cartilage lesions is a public health challenge to prevent long-term degradation and the occurrence of osteoarthritis. Cartilage tissue engineering represents a promising alternative to the current insufficient surgical solutions. 3D printing is a thriving technology and offers new possibilities for personalized regenerative medicine. Extrusion-based processes permit the deposition of cell-seeded bioinks, in a layer-by-layer manner, allowing mimicry of the native zonal organization of hyaline cartilage. Mesenchymal stem cells (MSCs) are a promising cell source for cartilage tissue engineering. Originally isolated from bone marrow, they can now be derived from many different cell sources (e.g., synovium, dental pulp, Wharton’s jelly). Their proliferation and differentiation potential are well characterized, and they possess good chondrogenic potential, making them appropriate candidates for cartilage reconstruction. This review summarizes the different sources, origins, and densities of MSCs used in extrusion-based bioprinting (EBB) processes, as alternatives to chondrocytes. The different bioink constituents and their advantages for producing substitutes mimicking healthy hyaline cartilage is also discussed.
37

Wang, Ketao, Ji Li, Zhongli Li, Bin Wang, Yuanyuan Qin, Ning Zhang, Hao Zhang, Xiangzheng Su, Yuxing Wang, and Heng Zhu. "Chondrogenic Progenitor Cells Exhibit Superiority Over Mesenchymal Stem Cells and Chondrocytes in Platelet-Rich Plasma Scaffold-Based Cartilage Regeneration." American Journal of Sports Medicine 47, no. 9 (June 13, 2019): 2200–2215. http://dx.doi.org/10.1177/0363546519854219.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Background: Platelet-rich plasma (PRP) has been considered a promising tool for cartilage regeneration. However, increasing evidence has demonstrated the controversial effects of PRP on tissue regeneration, partially due to the unsatisfactory cell source. Chondrogenic progenitor cells (CPCs) have gained increasing attention as a potential cell source due to their self-renewal and multipotency, especially toward the chondrogenic lineage, and, thus, may be an appropriate alternative for cartilage engineering. Purpose: To compare the effects of PRP on CPC, mesenchymal stem cell (MSC), and chondrocyte proliferation, chondrogenesis, and cartilage regeneration. Study Design: Controlled laboratory study. Methods: Whole blood samples were obtained from 5 human donors to create PRPs (0, 1000 × 109, and 2000 × 109 platelets per liter). The proliferation and chondrogenesis of CPCs, bone marrow–derived MSCs (BMSCs), and chondrocytes were evaluated via growth kinetic and CCK-8 assays. Immunofluorescence, cytochemical staining, and gene expression analyses were performed to assess chondrogenic differentiation and cartilaginous matrix formation. The in vivo effects of CPCs, BMSCs, and chondrocytes on cartilage regeneration after PRP treatment were measured by use of histopathological, biochemical, and biomechanical techniques in a cartilage defect model involving mature male New Zealand White rabbits (critical size, 5 mm). Results: The CPCs possessed migration abilities and proliferative capacities superior to those of the chondrocytes, while exhibiting a chondrogenic predisposition stronger than that of the BMSCs. The growth kinetic, CCK-8, cytochemical staining, and biochemical analyses revealed that the CPCs simultaneously displayed a higher cell density than the chondrocytes and stronger chondrogenesis than the BMSCs after PRP stimulation. In addition, the in vivo study demonstrated that the PRP+CPC construct yielded better histological (International Cartilage Repair Society [ICRS] score, mean ± SEM, 1197.2 ± 163.2) and biomechanical (tensile modulus, 1.523 ± 0.194) results than the PRP+BMSC (701.1 ± 104.9, P < .05; 0.791 ± 0.151, P < .05) and PRP+chondrocyte (541.6 ± 98.3, P < .01; 0.587 ± 0.142, P < .01) constructs at 12 weeks after implantation. Conclusion: CPCs exhibit superiority over MSCs and chondrocytes in PRP scaffold-based cartilage regeneration, and PRP+CPC treatment may be a favorable strategy for cartilage repair. Clinical Relevance: These findings provide evidence highlighting the preferable role of CPCs as a cell source in PRP-mediated cartilage regeneration and may help researchers address the problem of unsatisfactory cell sources in cartilage engineering.
38

Chen, Yawen, Xinli Ouyang, Yide Wu, Shaojia Guo, Yongfang Xie, and Guohui Wang. "Co-culture and Mechanical Stimulation on Mesenchymal Stem Cells and Chondrocytes for Cartilage Tissue Engineering." Current Stem Cell Research & Therapy 15, no. 1 (March 19, 2020): 54–60. http://dx.doi.org/10.2174/1574888x14666191029104249.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Defects in articular cartilage injury and chronic osteoarthritis are very widespread and common, and the ability of injured cartilage to repair itself is limited. Stem cell-based cartilage tissue engineering provides a promising therapeutic option for articular cartilage damage. However, the application of the technique is limited by the number, source, proliferation, and differentiation of stem cells. The co-culture of mesenchymal stem cells and chondrocytes is available for cartilage tissue engineering, and mechanical stimulation is an important factor that should not be ignored. A combination of these two approaches, i.e., co-culture of mesenchymal stem cells and chondrocytes under mechanical stimulation, can provide sufficient quantity and quality of cells for cartilage tissue engineering, and when combined with scaffold materials and cytokines, this approach ultimately achieves the purpose of cartilage repair and reconstruction. In this review, we focus on the effects of co-culture and mechanical stimulation on mesenchymal stem cells and chondrocytes for articular cartilage tissue engineering. An in-depth understanding of the impact of co-culture and mechanical stimulation of mesenchymal stem cells and chondrocytes can facilitate the development of additional strategies for articular cartilage tissue engineering.
39

Joe, Su Mee, In Seon Lee, Yong Tae Lee, Jun Hyuk Lee, and Byung Tae Choi. "Suppression of Collagen-Induced Arthritis in Rats by Continuous Administration of Dae-Bang-Poong-Tang (Da-Fang-Feng-Tang)." American Journal of Chinese Medicine 29, no. 02 (January 2001): 355–65. http://dx.doi.org/10.1142/s0192415x0100037x.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Although Dae-Bang-Poong-Tang (an herbal formula of 15 herbs)-treated rats exhibited a mild inflammation, the significant histological changes including a marked infiltration of inflammatory cells in the synovium and damaged articular cartilages were not observed. The staining abilities of the cartilage such as periodic acid Schiff's reaction in the interterritorial matrix of hyaline cartilage, alcian blue and aldehyde fuchsin staining in the capsule of chondrocytes and in the interterritorial matrix of articular cartilage and Con A, sWGA and BSL-1 affinities of chondrocytes tended to decrease in the rats with collagent-induced arthritis compared with normal rats. Decreased stainabilities and affinities were almost recovered in the Dae-Bang-Poong-Tang-treated rats. In the collagen-induced rats, iNOS expression in the synovial lining cells and subsynovial tissue were significantly increased and numerous strong immunoreactive cells were demostrated in the subsynovial tissue. Somewhat decreased immunoreaction of iNOS was shown in the synovial lining cells and subsynovial tissue of Dae-Bang-Poong-Tang-treated rats. It was concluded that Dae-Bang-Poong-Tang showed a notable protection against histological changes and histochemical staining, and it acted as an inhibitor of iNOS expression. Dae-Bang-Poong-Tang may be used as a complementary therapeutic agent to alleviate the rheumatoid arthritis.
40

Li, Lu, Yuehui Ma, Xianglong Li, Xiangchen Li, Chunyu Bai, Meng Ji, Shuang Zhang, Weijun Guan, and Junjie Li. "Isolation, Culture, and Characterization of Chicken Cartilage Stem/Progenitor Cells." BioMed Research International 2015 (2015): 1–11. http://dx.doi.org/10.1155/2015/586290.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
A chondrocyte progenitor population isolated from the surface zone of articular cartilage has become a promising cell source for cell-based cartilage repair. The cartilage-derived stem/progenitor cells are multipotent stem cells, which can differentiate into three cell types in vitro including adipocytes, osteoblasts, and chondrocytes. Much work has been done on cartilage stem/progenitor cells (CSPCs) from people, horses, and cattle, but the relatively little literature has been published about these cells in chickens. In our work, CSPCs were isolated from chicken embryos in incubated eggs for 20 days. In order to inquire into the biological characteristics of chicken CSPCs, immunofluorescence, reverse transcription-polymerase chain reaction (RT-PCR), and flow cytometry were adopted to detect the characteristic surface markers of CSPCs. Primary CSPCs were subcultured to passage 22 and, for purpose of knowing the change of cell numbers, we drew the growth curves. Isolated CSPCs were induced to adipocytes, osteoblasts, and chondrocytes. Our results suggest that we have identified and characterised a novel cartilage progenitor population resident in chicken articular cartilage and CSPCs isolated from chickens possess similar biological characteristics to those from other species, which will greatly benefit future cell-based cartilage repair therapies.
41

Buhrmann, Constanze, Ali Honarvar, Mohsen Setayeshmehr, Saeed Karbasi, Mehdi Shakibaei, and Ali Valiani. "Herbal Remedies as Potential in Cartilage Tissue Engineering: An Overview of New Therapeutic Approaches and Strategies." Molecules 25, no. 13 (July 6, 2020): 3075. http://dx.doi.org/10.3390/molecules25133075.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
It is estimated that by 2023, approximately 20% of the population of Western Europe and North America will suffer from a degenerative joint disease commonly known as osteoarthritis (OA). During the development of OA, pro-inflammatory cytokines are one of the major causes that drive the production of inflammatory mediators and thus of matrix-degrading enzymes. OA is a challenging disease for doctors due to the limitation of the joint cartilage’s capacity to repair itself. Though new treatment approaches, in particular with mesenchymal stem cells (MSCs) that integrate the tissue engineering (TE) of cartilage tissue, are promising, they are not only expensive but more often do not lead to the regeneration of joint cartilage. Therefore, there is an increasing need for novel, safe, and more effective alternatives to promote cartilage joint regeneration and TE. Indeed, naturally occurring phytochemical compounds (herbal remedies) have a great anti-inflammatory, anti-oxidant, and anabolic potential, and they have received much attention for the development of new therapeutic strategies for the treatment of inflammatory diseases, including the prevention of age-related OA and cartilage TE. This paper summarizes recent research on herbal remedies and their chondroinductive and chondroprotective effects on cartilage and progenitor cells, and it also emphasizes the possibilities that exist in this research area, especially with regard to the nutritional support of cartilage regeneration and TE, which may not benefit from non-steroidal anti-inflammatory drugs (NSAIDs).
42

Shiraishi, Katsunori, Naosuke Kamei, Shunsuke Takeuchi, Shinobu Yanada, Hisashi Mera, Shigeyuki Wakitani, Nobuo Adachi, and Mitsuo Ochi. "Quality Evaluation of Human Bone Marrow Mesenchymal Stem Cells for Cartilage Repair." Stem Cells International 2017 (2017): 1–9. http://dx.doi.org/10.1155/2017/8740294.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Quality evaluation of mesenchymal stem cells (MSCs) based on efficacy would be helpful for their clinical application. In this study, we aimed to find the factors of human bone marrow MSCs relating to cartilage repair. The expression profiles of humoral factors, messenger RNAs (mRNAs), and microRNAs (miRNAs) were analyzed in human bone marrow MSCs from five different donors. We investigated the correlations of these expression profiles with the capacity of the MSCs for proliferation, chondrogenic differentiation, and cartilage repair in vivo. The mRNA expression of MYBL1 was positively correlated with proliferation and cartilage differentiation. By contrast, the mRNA expression of RCAN2 and the protein expression of TIMP-1 and VEGF were negatively correlated with proliferation and cartilage differentiation. However, MSCs from all five donors had the capacity to promote cartilage repair in vivo regardless of their capacity for proliferation and cartilage differentiation. The mRNA expression of HLA-DRB1 was positively correlated with cartilage repair in vivo. Meanwhile, the mRNA expression of TMEM155 and expression of miR-486-3p, miR-148b, miR-93, and miR-320B were negatively correlated with cartilage repair. The expression analysis of these factors might help to predict the ability of bone marrow MSCs to promote cartilage repair.
43

He, Yuan-Jia, Shuang Lin, and Qiang Ao. "Research Progress of Tissue-Engineered Cartilage in Repairing Cartilage Defects." Science of Advanced Materials 12, no. 1 (January 1, 2020): 66–74. http://dx.doi.org/10.1166/sam.2020.3704.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Due to the unsatisfactory outcome of current clinical treatment, tissue engineering technology has become a promising approach for the treatment of cartilage defects. Typical cartilage tissue engineering uses seed cells that have been expanded in vitro to implant into various biomaterial scaffolds that are biocompatible and are gradually degraded and absorbed in the body, with or without physical/chemical factors mimicking the cartilage microenvironment, to regenerate cartilage tissue with similar biochemical and biomechanical properties to natural cartilage tissue. Therefore, we summarise the three aspects of seed cells, biological scaffolds, and factors/signals.
44

Cui, Dixin, Hongyu Li, Xin Xu, Ling Ye, Xuedong Zhou, Liwei Zheng, and Yachuan Zhou. "Mesenchymal Stem Cells for Cartilage Regeneration of TMJ Osteoarthritis." Stem Cells International 2017 (2017): 1–11. http://dx.doi.org/10.1155/2017/5979741.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Temporomandibular joint osteoarthritis (TMJ OA) is a degenerative disease, characterized by progressive cartilage degradation, subchondral bone remodeling, synovitis, and chronic pain. Due to the limited self-healing capacity in condylar cartilage, traditional clinical treatments have limited symptom-modifying and structure-modifying effects to restore impaired cartilage as well as other TMJ tissues. In recent years, stem cell-based therapy has raised much attention as an alternative approach towards tissue repair and regeneration. Mesenchymal stem cells (MSCs), derived from the bone marrow, synovium, and even umbilical cord, play a role as seed cells for the cartilage regeneration of TMJ OA. MSCs possess multilineage differentiation potential, including chondrogenic differentiation as well as osteogenic differentiation. In addition, the trophic modulations of MSCs exert anti-inflammatory and immunomodulatory effects under aberrant conditions. Furthermore, MSCs combined with appropriate scaffolds can form cartilaginous or even osseous compartments to repair damaged tissue and impaired function of TMJ. In this review, we will briefly discuss the pathogenesis of cartilage degeneration in TMJ OA and emphasize the potential sources of MSCs and novel approaches for the cartilage regeneration of TMJ OA, particularly focusing on the MSC-based therapy and tissue engineering.
45

Clark, Andrea L., Linda Mills, David A. Hart, and Walter Herzog. "MUSCLE-INDUCED PATELLOFEMORAL JOINT LOADING RAPIDLY AFFECTS CARTILAGE mRNA LEVELS IN A SITE SPECIFIC MANNER." Journal of Musculoskeletal Research 08, no. 01 (March 2004): 1–12. http://dx.doi.org/10.1142/s0218957704001223.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Mechanical loading of articular cartilage affects the synthesis and degradation of matrix macromolecules. Much of the work in this area has involved mechanical loading of articular cartilage explants or cells in vitro and assessing biological responses at the mRNA and protein levels. In this study, we developed a new experimental technique to load an intact patellofemoral joint in vivo using muscle stimulation. The articular cartilages were cyclically loaded for one hour in a repeatable and measurable manner. Cartilage was harvested from central and peripheral regions of the femoral groove and patella, either immediately after loading or after a three hour recovery period. Total RNA was isolated from the articular cartilage and biological responses were assessed on the mRNA level using the reverse transcriptase-polymerase chain reaction. Articular cartilage from intact patellofemoral joints demonstrated heterogeneity at the mRNA level for six of the genes assessed independent of the loading protocol. Cyclical loading of cartilage in its native environment led to alterations in mRNA levels for a subset of molecules when assessed immediately after the loading period. However, the increases in TIMP-1 and decreases in bFGF mRNA levels were transient; being present immediately after load application but not after a three hour recovery period.
46

Deng, Zhantao, Jiewen Jin, Jianning Zhao, and Haidong Xu. "Cartilage Defect Treatments: With or without Cells? Mesenchymal Stem Cells or Chondrocytes? Traditional or Matrix-Assisted? A Systematic Review and Meta-Analyses." Stem Cells International 2016 (2016): 1–14. http://dx.doi.org/10.1155/2016/9201492.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Articular cartilage defects have been addressed by using multiple strategies. In the last two decades, promising new strategies by using assorted scaffolds and cell sources to induce tissue regeneration have emerged, such as autologous chondrocyte implantation (ACI) and mesenchymal stem cell implantation (MSCI). However, it is still controversial in the clinical strategies when to choose these treatments. Thus, we conducted a systematic review and meta-analyses to compare the efficacy and safety of different cartilage treatments. In our study, 17 studies were selected to compare different treatments for cartilage defects. The results of meta-analyses indicated that cell-based cartilage treatments showed significant better efficacy than cell-free treatments did (OR: 4.27, 95% CI: 2.19–8.34; WMD: 10.11, 95% CI: 2.69–16.53). Another result indicated that MACT had significant better efficacy than traditional ACI did (OR: 0.49, 95% CI: 0.30–0.82). Besides, the incidence of graft hypertrophy of MACT was slightly lower than that of traditional ACI (OR: 2.43, 95% CI: 1.00–5.94). Current data showed that the cell-based treatments and MACT are better options for cartilage treatments, but more well-designed comparative studies are still needed to enhance our understanding of different treatments for cartilage defects.
47

Huselstein, C., Y. Li, and X. He. "Mesenchymal stem cells for cartilage engineering." Bio-Medical Materials and Engineering 22, no. 1-3 (2012): 69–80. http://dx.doi.org/10.3233/bme-2012-0691.

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

Min, Byoung-Hyun, Hyun Jung Lee, and Young Jick Kim. "Cartilage Repair Using Mesenchymal Stem Cells." Journal of the Korean Medical Association 52, no. 11 (2009): 1077. http://dx.doi.org/10.5124/jkma.2009.52.11.1077.

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

Savkovic, Vuk, Hanluo Li, Jong-Keun Seon, Michael Hacker, Sandra Franz, and Jan-Christoph Simon. "Mesenchymal Stem Cells in Cartilage Regeneration." Current Stem Cell Research & Therapy 9, no. 6 (September 22, 2014): 469–88. http://dx.doi.org/10.2174/1574888x09666140709111444.

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

Liao, Jinfeng, and Yunfeng Lin. "Stem Cells and Cartilage Tissue Engineering." Current Stem Cell Research & Therapy 13, no. 7 (August 29, 2018): 489. http://dx.doi.org/10.2174/1574888x1307180803122513.

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

До бібліографії