Nasal chondrocytes

Last updated

Nasal chondrocytes (NC) are present in the hyaline cartilage of the nasal septum and in fact are the only cell type within the tissue. Similar to chondrocytes present in articular cartilage, NC express extracellular matrix proteins such as glycosaminoglycans and collagen. [1]

Contents

In their natural environment

At birth the nasal septum, except for the vomer ‘’anlage” is completely cartilaginous. In the second half of the first year of life, the septum progressively ossifies in posterior-anterior direction by a process of endochondral ossification. [2] The remaining cartilaginous part (characterized as hyaline cartilage) of the human nasal septum has a specific three-dimensional organization with regards to local differences in cell size and the amounts of extracellular matrix. In the outer region of the cartilage, NC are numerous, small, flat and oriented parallel to the surface. In the intermediate and central regions, NC are spheroid in shape, less dense, and are aligned perpendicular to the cartilage surface. [3] The cartilage matrix is mainly composed of type II collagen (90-95%), although small amounts of type IX and XI collagen are also found. [4] Nasal cartilage is tightly connected to perichondrium, consisting of several layers of connective tissue fibers (mainly based on type I collagen) running parallel to the cartilage. [5]

Tissue engineering applications

Articular chondrocytes have typically been the cell type used for cartilage tissue engineering strategies for articular cartilage repair. Since NC can also express hyaline cartilage specific extracellular matrix proteins such as glycosaminoglycans and collagen, NC have recently also been used for the in vitro engineering of cartilage tissues.

Cell isolation

Harvesting of nasal cartilage is minimally invasive, can be performed in an outpatient procedure under local anesthesia and is associated with minimal morbidity. [6] NC can be isolated from nasal septal cartilage biopsies by enzymatic digestion using collagenase type I, II or IV (at different combination and concentration – varying from 0.15% to 0.6% –) alone or after an initial short pre-incubation phase with pronase (0.2% - 1%). [7] [8] [9] [10] [11] [12] Cell yield after enzymatic digestion of the nasal cartilage was estimated to be 2,100 - 3,700 cells/mg of tissue. [7] [12] Alternatively, NC can be isolated by outgrowth culture of nasal cartilage fragments. [8]

Cell expansion

After isolation from septal cartilage biopsies, NC can be extensively expanded in numbers by conventional in vitro cell culture methods (monolayer culture in flasks or Petri dishes). The proliferation rate of NC was reported to be increased in the presence of specific growth factors such as TGF-beta and FGF-2 [12] [13] or culture supplements like Insulin-Transferrin-Selenium. [7] NC cultured in medium containing autologous serum exhibit similar proliferation rates to NC cultured with medium supplemented with fetal bovine serum. [13] Although articular chondrocytes derived from older individuals have been shown to have a lower proliferation capacity than from younger donors, NC have been shown to have significantly less age-dependence. [13] [14]

Differentiation

Similar to other chondrocytes from hyaline cartilage tissues in other locations in the human body, NC undergo a process of cell de-differentiation during monolayer culture. NC de-differentiation can be characterized by a gradual acquisition of a fibroblastic morphology, the expression of proteins associated with an undifferentiated mesenchymal cell phenotype (e.g., type I collagen and versican), and decreased expression of hyaline cartilage proteins (e.g., type II collagen and aggrecan). [9] However, NC can re-differentiate when transferred back into a more physiological three-dimensional environment. Abundant production of cartilage specific matrix has been reported by expanded NC when induced to re-differentiate in micromass pellets, [11] [12] [14] [15] alginate beads, [16] [17] [18] [19] [20] hydrogels, [7] [21] or into porous scaffolds based on polyglycolic acid, [9] [19] [22] polyethylene glycol terephthalate/polybuthylene terephalate, [23] collagen, [24] or hyaluronic acid. [1] [14] [25] Supplementation with specific growth factors (e.g., TGF-beta, IGF-1, and GDF-5) during re-differentiation has been shown to enhance the accumulation of glycosaminoglycans (GAG) and type II collagen as well as the biomechanical properties of the generated constructs. [20] [26]

Autologous serum has also been used during NC re-differentiation in place of fetal bovine serum with similar efficacy. [14] [26] [27] Studies that directly compared the re-differentiation of articular chondrocytes to NC have shown that the cartilage forming capacity of NC was higher and more reproducible than that of articular chondrocytes [24] [25] [28] with lower donor-related dependency. [13] [14] Additionally NC have been recently shown to exhibit features of self-renewal capability, being able to form cartilage tissue following serial cloning possibly due to their neuro-ectodermal origin. [29]

Animal studies

Pre-clinical investigations have been undertaken using various animal models to provide proof-of-principle of the clinical potential of NC-based tissue engineered constructs for cartilage reconstruction. The maturation of human NC engineered grafts has often been assessed in the subcutaneous pocket of nude mice, i.e., an environment highly vascularized and permissive to, but not inductive of, chondrogenesis. [30] [31] The extent of cartilage matrix production and the mechanical properties of NC-based constructs have been reported to increase in such ectopic in vivo models. [21] [13] [18] [22] [32] [33] [34] An ectopic mouse model was also used in order to test the effects of different production methods for generating large clinically relevant-sized NC-based tissue grafts. [26]

Although these models can yield insightful results, nude mice are not capable of eliciting a significant immune response, and therefore these studies cannot predict the prognosis of implanted engineered septal cartilage in an immunocompetent host. As an alternative, an orthotopic rat model has been established to study nasal septum repair. In this model, septal cartilage was first perforated to create a defect, and subsequently, an engineered cartilage graft implanted into the defect during the same surgical procedure [35]

Using an orthotopic large animal model to study repair of articular cartilage defects, engineered NC-based cartilage grafts were implanted into the condyle of goats. In this study, it was determined that NC directly contributed to the repair of the articular cartilage defects and resulted in a superior outcome as compared to engineered articular chondrocyte-based grafts. [36]

Clinical applications

Clinical application of autologous NC-based cartilage tissues Nose to knee cartilage tissue engineering approach.tif
Clinical application of autologous NC-based cartilage tissues

Engineered cartilage tissue, based on autologous NC, has recently been used by plastic surgeons for the reconstruction of nasal cartilage defects. Tissue engineered cartilage grafts based on NC as autologous grafts for the reconstruction of the alar lobule of the nose after skin tumor resection in a first-in-man clinical trial (ClinicalTrials.gov, number NCT01242618) [29] (see figure on the right). Their results demonstrated that the engineered grafts could lead to complete structural, functional and aesthetic satisfaction. Moreover, since harvesting of the nasal cartilage biopsy was minimally invasive, it could be performed in an outpatient procedure under local anesthesia, and was therefore associated with minimal morbidity. [6]

Several studies have demonstrated that NC are compatible with the environmental features typical of the injured knee (e.g., in terms of response to inflammatory molecules, mechanical loading and genetic molecular signature). [24] [25] [30] Thus nasal chondrocytes have been proposed as an alternative cell source for the repair of articular cartilage defects. A phase I clinical trial (ClinicalTrials.gov, number NCT 01605201) was conducted to test the safety and feasibility of implanting a tissue engineered cartilage graft based on autologous nasal chondrocytes for the regeneration of knee cartilage defects. The clinical observations of the first 10 patients of this study indicated not only safety and feasibility of the procedure, but together with Magnetic Resonance Imaging (MRI) and delayed gadolinium-enhanced magnetic resonance imaging of cartilage (dGEMRIC) data, also showed promising results for efficacy of the treatment as indicated by significant improvement in clinical scores and regeneration of hyaline repair tissue after 24 months. [37] Based on this study, a multi-center phase II clinical trial was initiated and is currently ongoing to assess the efficacy of the NC-based cartilage grafts for the repair of traumatic knee cartilage defects (BIO-CHIP; funded by the European Union through the Horizon 2020 program, grant number 681103).

Related Research Articles

<span class="mw-page-title-main">Cartilage</span> Resilient and smooth elastic tissue present in animals

Cartilage is a resilient and smooth type of connective tissue. It is a semi-transparent and non-porous type of tissue. It is usually covered by a tough and fibrous membrane called perichondrium. In tetrapods, it covers and protects the ends of long bones at the joints as articular cartilage, and is a structural component of many body parts including the rib cage, the neck and the bronchial tubes, and the intervertebral discs. In other taxa, such as chondrichthyans, but also in cyclostomes, it may constitute a much greater proportion of the skeleton. It is not as hard and rigid as bone, but it is much stiffer and much less flexible than muscle. The matrix of cartilage is made up of glycosaminoglycans, proteoglycans, collagen fibers and, sometimes, elastin. It usually grows quicker than bone.

<span class="mw-page-title-main">Rhinoplasty</span> Surgical procedure to enhance or reconstruct a human nose

Rhinoplasty, commonly called nose job, medically called nasal reconstruction, is a plastic surgery procedure for altering and reconstructing the nose. There are two types of plastic surgery used – reconstructive surgery that restores the form and functions of the nose and cosmetic surgery that changes the appearance of the nose. Reconstructive surgery seeks to resolve nasal injuries caused by various traumas including blunt, and penetrating trauma and trauma caused by blast injury. Reconstructive surgery can also treat birth defects, breathing problems, and failed primary rhinoplasties. Rhinoplasty may remove a bump, narrow nostril width, change the angle between the nose and the mouth, or address injuries, birth defects, or other problems that affect breathing, such as a deviated nasal septum or a sinus condition. Surgery only on the septum is called a septoplasty.

<span class="mw-page-title-main">Tissue engineering</span> Biomedical engineering discipline

Tissue engineering is a biomedical engineering discipline that uses a combination of cells, engineering, materials methods, and suitable biochemical and physicochemical factors to restore, maintain, improve, or replace different types of biological tissues. Tissue engineering often involves the use of cells placed on tissue scaffolds in the formation of new viable tissue for a medical purpose, but is not limited to applications involving cells and tissue scaffolds. While it was once categorized as a sub-field of biomaterials, having grown in scope and importance, it can is considered as a field of its own.

<span class="mw-page-title-main">Nasal septum</span> Separator of the left and right airways in the nose

The nasal septum separates the left and right airways of the nasal cavity, dividing the two nostrils.

<span class="mw-page-title-main">Hyaline cartilage</span> Type of cartilage in animals

Hyaline cartilage is the glass-like (hyaline) and translucent cartilage found on many joint surfaces. It is also most commonly found in the ribs, nose, larynx, and trachea. Hyaline cartilage is pearl-gray in color, with a firm consistency and has a considerable amount of collagen. It contains no nerves or blood vessels, and its structure is relatively simple.

<span class="mw-page-title-main">Chondrocyte</span> Cell that makes up cartilage

Chondrocytes are the only cells found in healthy cartilage. They produce and maintain the cartilaginous matrix, which consists mainly of collagen and proteoglycans. Although the word chondroblast is commonly used to describe an immature chondrocyte, the term is imprecise, since the progenitor of chondrocytes can differentiate into various cell types, including osteoblasts.

<span class="mw-page-title-main">Mechanotransduction</span> Conversion of mechanical stimulus of a cell into electrochemical activity

In cellular biology, mechanotransduction is any of various mechanisms by which cells convert mechanical stimulus into electrochemical activity. This form of sensory transduction is responsible for a number of senses and physiological processes in the body, including proprioception, touch, balance, and hearing. The basic mechanism of mechanotransduction involves converting mechanical signals into electrical or chemical signals.

<span class="mw-page-title-main">Chondroblast</span> Mesenchymal progenitor cell that forms a chondrocyte

Chondroblasts, or perichondrial cells, is the name given to mesenchymal progenitor cells in situ which, from endochondral ossification, will form chondrocytes in the growing cartilage matrix. Another name for them is subchondral cortico-spongious progenitors. They have euchromatic nuclei and stain by basic dyes.

<span class="mw-page-title-main">Chondrogenesis</span>

Chondrogenesis is the biological process through which cartilage tissue is formed and developed. This intricate and tightly regulated cellular differentiation pathway plays a crucial role in skeletal development, as cartilage serves as a fundamental component of the embryonic skeleton. The term "chondrogenesis" is derived from the Greek words "chondros," meaning cartilage, and "genesis," meaning origin or formation.

Articular cartilage, most notably that which is found in the knee joint, is generally characterized by very low friction, high wear resistance, and poor regenerative qualities. It is responsible for much of the compressive resistance and load bearing qualities of the knee joint and, without it, walking is painful to impossible. Osteoarthritis is a common condition of cartilage failure that can lead to limited range of motion, bone damage and invariably, pain. Due to a combination of acute stress and chronic fatigue, osteoarthritis directly manifests itself in a wearing away of the articular surface and, in extreme cases, bone can be exposed in the joint. Some additional examples of cartilage failure mechanisms include cellular matrix linkage rupture, chondrocyte protein synthesis inhibition, and chondrocyte apoptosis. There are several different repair options available for cartilage damage or failure.

<span class="mw-page-title-main">Nasal cartilages</span> Supportive structures in the nose

The nasal cartilages are structures within the nose that provide form and support to the nasal cavity. The nasal cartilages are made up of a flexible material called hyaline cartilage in the distal portion of the nose. There are five individual cartilages that make up the nasal cavity: septal nasal cartilage, lateral nasal cartilage, major alar cartilage, minor alar cartilage, and vomeronasal cartilage.

Articular cartilage repair treatment involves the repair of the surface of the articular joint's hyaline cartilage, though these solutions do not perfectly restore the articular cartilage. These treatments have been shown to have positive results for patients who have articular cartilage damage. They can provide some measure of pain relief, while slowing down the accumulation of damage, or delaying the need for joint replacement surgery.

Autologous chondrocyte implantation is a biomedical treatment that repairs damages in articular cartilage. ACI provides pain relief while at the same time slowing down the progression or considerably delaying partial or total joint replacement surgery.

Mesenchymal stem cells (MSCs) are multipotent cells found in multiple human adult tissues, including bone marrow, synovial tissues, and adipose tissues. Since they are derived from the mesoderm, they have been shown to differentiate into bone, cartilage, muscle, and adipose tissue. MSCs from embryonic sources have shown promise scientifically while creating significant controversy. As a result, many researchers have focused on adult stem cells, or stem cells isolated from adult humans that can be transplanted into damaged tissue.

A fibrin scaffold is a network of protein that holds together and supports a variety of living tissues. It is produced naturally by the body after injury, but also can be engineered as a tissue substitute to speed healing. The scaffold consists of naturally occurring biomaterials composed of a cross-linked fibrin network and has a broad use in biomedical applications.

Autologous matrix-induced chondrogenesis (AMIC) is a treatment for articular cartilage damage. It combines microfracture surgery with the application of a bi-layer collagen I/III membrane. There is tentative short to medium term benefits as of 2017.

Cartilage repair techniques are the current focus of large amounts of research. Many different strategies have been proposed as solutions for cartilage defects. Surgical techniques currently being studied include:

Gene therapy for osteoarthritis is the application of gene therapy to treat osteoarthritis (OA). Unlike pharmacological treatments which are administered locally or systemically as a series of interventions, gene therapy aims to establish sustained therapeutic effect after a single, local injection.

The in vivo bioreactor is a tissue engineering paradigm that uses bioreactor methodology to grow neotissue in vivo that augments or replaces malfunctioning native tissue. Tissue engineering principles are used to construct a confined, artificial bioreactor space in vivo that hosts a tissue scaffold and key biomolecules necessary for neotissue growth. Said space often requires inoculation with pluripotent or specific stem cells to encourage initial growth, and access to a blood source. A blood source allows for recruitment of stem cells from the body alongside nutrient delivery for continual growth. This delivery of cells and nutrients to the bioreactor eventually results in the formation of a neotissue product. 

Artificial cartilage is a synthetic material made of hydrogels or polymers that aims to mimic the functional properties of natural cartilage in the human body. Tissue engineering principles are used in order to create a non-degradable and biocompatible material that can replace cartilage. While creating a useful synthetic cartilage material, certain challenges need to be overcome. First, cartilage is an avascular structure in the body and therefore does not repair itself. This creates issues in regeneration of the tissue. Synthetic cartilage also needs to be stably attached to its underlying surface i.e. the bone. Lastly, in the case of creating synthetic cartilage to be used in joint spaces, high mechanical strength under compression needs to be an intrinsic property of the material.

References

  1. 1 2 Candrian C, Vonwil D, Barbero A, Bonacina E, Miot S, Farhadi J, et al. (January 2008). "Engineered cartilage generated by nasal chondrocytes is responsive to physical forces resembling joint loading". Arthritis and Rheumatism. 58 (1): 197–208. doi:10.1002/art.23155. PMID   18163475.
  2. Pelttari K, Mumme M, Barbero A, Martin I (October 2017). "Nasal chondrocytes as a neural crest-derived cell source for regenerative medicine". Current Opinion in Biotechnology. 47: 1–6. doi:10.1016/j.copbio.2017.05.007. PMID   28551498.
  3. Schultz-Coulon HJ, Eckermeier L (January 1976). "Zum Postnatalen Wachstum Der Nasenscheidewand" [Postnatal growth of nasal septum]. Acta Oto-Laryngologica (in German). 82 (1–2): 131–142. doi:10.3109/00016487609120872. PMID   948977.
  4. Popko M, Bleys RL, De Groot JW, Huizing EH (June 2007). "Histological structure of the nasal cartilages and their perichondrial envelope. I. The septal and lobular cartilage". Rhinology. 45 (2): 148–152. PMID   17708463.
  5. Holden PK, Liaw LH, Wong BJ (July 2008). "Human nasal cartilage ultrastructure: characteristics and comparison using scanning electron microscopy". The Laryngoscope. 118 (7): 1153–1156. doi:10.1097/MLG.0b013e31816ed5ad. PMC   4151991 . PMID   18438266.
  6. 1 2 Aksoy F, Yildirim YS, Demirhan H, Özturan O, Solakoglu S (January 2012). "Structural characteristics of septal cartilage and mucoperichondrium". The Journal of Laryngology and Otology. 126 (1): 38–42. doi:10.1017/S0022215111002404. PMID   21888752.
  7. 1 2 3 4 Siegel NS, Gliklich RE, Taghizadeh F, Chang Y (February 2000). "Outcomes of septoplasty". Otolaryngology–Head and Neck Surgery. 122 (2): 228–232. doi:10.1016/S0194-5998(00)70244-0. PMID   10652395. S2CID   43019892.
  8. 1 2 Chua KH, Aminuddin BS, Fuzina NH, Ruszymah BH (June 2005). "Insulin-transferrin-selenium prevent human chondrocyte dedifferentiation and promote the formation of high quality tissue engineered human hyaline cartilage". European Cells & Materials. 9: 58–67, discussion 67. doi:10.22203/ecm.v009a08. PMID   15962238.
  9. 1 2 3 Elsaesser AF, Schwarz S, Joos H, Koerber L, Brenner RE, Rotter N (2016). "Characterization of a migrative subpopulation of adult human nasoseptal chondrocytes with progenitor cell features and their potential for in vivo cartilage regeneration strategies". Cell & Bioscience. 6: 11. doi: 10.1186/s13578-016-0078-6 . PMC   4752797 . PMID   26877866.
  10. Homicz MR, Schumacher BL, Sah RL, Watson D (November 2002). "Effects of serial expansion of septal chondrocytes on tissue-engineered neocartilage composition". Otolaryngology–Head and Neck Surgery. 127 (5): 398–408. doi:10.1067/mhn.2002.129730. PMID   12447233. S2CID   24779895.
  11. 1 2 Liese J, Marzahn U, El Sayed K, Pruss A, Haisch A, Stoelzel K (June 2013). "Cartilage tissue engineering of nasal septal chondrocyte-macroaggregates in human demineralized bone matrix". Cell and Tissue Banking. 14 (2): 255–266. doi:10.1007/s10561-012-9322-4. PMID   22714645. S2CID   254381658.
  12. 1 2 3 4 Shafiee A, Kabiri M, Ahmadbeigi N, Yazdani SO, Mojtahed M, Amanpour S, Soleimani M (December 2011). "Nasal septum-derived multipotent progenitors: a potent source for stem cell-based regenerative medicine". Stem Cells and Development. 20 (12): 2077–2091. doi:10.1089/scd.2010.0420. PMID   21401444.
  13. 1 2 3 4 5 Wolf F, Haug M, Farhadi J, Candrian C, Martin I, Barbero A (February 2008). "A low percentage of autologous serum can replace bovine serum to engineer human nasal cartilage". European Cells & Materials. 15: 1–10. doi:10.22203/ecm.v015a01. PMID   18247273.
  14. 1 2 3 4 5 Vinatier C, Magne D, Moreau A, Gauthier O, Malard O, Vignes-Colombeix C, et al. (January 2007). "Engineering cartilage with human nasal chondrocytes and a silanized hydroxypropyl methylcellulose hydrogel". Journal of Biomedical Materials Research. Part A. 80 (1): 66–74. doi:10.1002/jbm.a.30867. PMID   16958048.
  15. Bujía J, Pitzke P, Kastenbauer E, Wilmes E, Hammer C (1996). "Effect of growth factors on matrix synthesis by human nasal chondrocytes cultured in monolayer and in agar". European Archives of Oto-Rhino-Laryngology. 253 (6): 336–340. doi:10.1007/BF00178288. PMID   8858257. S2CID   12243849.
  16. Peñuela L, Wolf F, Raiteri R, Wendt D, Martin I, Barbero A (June 2014). "Atomic force microscopy to investigate spatial patterns of response to interleukin-1beta in engineered cartilage tissue elasticity". Journal of Biomechanics. 47 (9): 2157–2164. doi:10.1016/j.jbiomech.2013.10.056. PMID   24290139.
  17. Alexander TH, Sage AB, Schumacher BL, Sah RL, Watson D (September 2006). "Human serum for tissue engineering of human nasal septal cartilage". Otolaryngology–Head and Neck Surgery. 135 (3): 397–403. doi:10.1016/j.otohns.2006.05.029. PMID   16949971. S2CID   383736.
  18. 1 2 Rotter N, Bonassar LJ, Tobias G, Lebl M, Roy AK, Vacanti CA (August 2002). "Age dependence of biochemical and biomechanical properties of tissue-engineered human septal cartilage". Biomaterials. 23 (15): 3087–3094. doi:10.1016/s0142-9612(02)00031-5. PMID   12102179.
  19. 1 2 Chang AA, Reuther MS, Briggs KK, Schumacher BL, Williams GM, Corr M, et al. (January 2012). "In vivo implantation of tissue-engineered human nasal septal neocartilage constructs: a pilot study". Otolaryngology–Head and Neck Surgery. 146 (1): 46–52. doi:10.1177/0194599811425141. PMC   4352411 . PMID   22031592.
  20. 1 2 Homicz MR, Chia SH, Schumacher BL, Masuda K, Thonar EJ, Sah RL, Watson D (January 2003). "Human septal chondrocyte redifferentiation in alginate, polyglycolic acid scaffold, and monolayer culture". The Laryngoscope. 113 (1): 25–32. doi:10.1097/00005537-200301000-00005. PMID   12514377. S2CID   24972306.
  21. 1 2 Tay AG, Farhadi J, Suetterlin R, Pierer G, Heberer M, Martin I (2004). "Cell yield, proliferation, and postexpansion differentiation capacity of human ear, nasal, and rib chondrocytes". Tissue Engineering. 10 (5–6): 762–770. doi:10.1089/1076327041348572. PMID   15265293.
  22. 1 2 van Osch GJ, Marijnissen WJ, van der Veen SW, Verwoerd-Verhoef HL (2001). "The potency of culture-expanded nasal septum chondrocytes for tissue engineering of cartilage". American Journal of Rhinology. 15 (3): 187–192. doi:10.2500/105065801779954166. PMID   11453506. S2CID   24763664.
  23. Naumann A, Rotter N, Bujía J, Aigner J (1998). "Tissue engineering of autologous cartilage transplants for rhinology". American Journal of Rhinology. 12 (1): 59–63. doi:10.2500/105065898782102972. PMID   9513661. S2CID   37632669.
  24. 1 2 3 Malda J, Kreijveld E, Temenoff JS, van Blitterswijk CA, Riesle J (December 2003). "Expansion of human nasal chondrocytes on macroporous microcarriers enhances redifferentiation". Biomaterials. 24 (28): 5153–5161. doi:10.1016/s0142-9612(03)00428-9. PMID   14568432.
  25. 1 2 3 Scotti C, Osmokrovic A, Wolf F, Miot S, Peretti GM, Barbero A, Martin I (February 2012). "Response of human engineered cartilage based on articular or nasal chondrocytes to interleukin-1β and low oxygen". Tissue Engineering. Part A. 18 (3–4): 362–372. doi:10.1089/ten.TEA.2011.0234. PMC   3267974 . PMID   21902467.
  26. 1 2 3 Farhadi J, Fulco I, Miot S, Wirz D, Haug M, Dickinson SC, et al. (December 2006). "Precultivation of engineered human nasal cartilage enhances the mechanical properties relevant for use in facial reconstructive surgery". Annals of Surgery. 244 (6): 978–85, discussion 985. doi:10.1097/01.sla.0000247057.16710.be. PMC   1856618 . PMID   17122623.
  27. do Amaral RJ, Pedrosa C, Kochem MC, Silva KR, Aniceto M, Claudio-da-Silva C, et al. (March 2012). "Isolation of human nasoseptal chondrogenic cells: a promise for cartilage engineering". Stem Cell Research. 8 (2): 292–299. doi:10.1016/j.scr.2011.09.006. PMID   22099383.
  28. Barandun M, Iselin LD, Santini F, Pansini M, Scotti C, Baumhoer D, et al. (August 2015). "Generation and characterization of osteochondral grafts with human nasal chondrocytes". Journal of Orthopaedic Research. 33 (8): 1111–1119. doi:10.1002/jor.22865. PMID   25994595. S2CID   206142529.
  29. 1 2 Fulco I, Miot S, Haug MD, Barbero A, Wixmerten A, Feliciano S, et al. (July 2014). "Engineered autologous cartilage tissue for nasal reconstruction after tumour resection: an observational first-in-human trial". Lancet. 384 (9940): 337–346. doi:10.1016/S0140-6736(14)60544-4. PMID   24726477. S2CID   12217253.
  30. 1 2 Pelttari K, Pippenger B, Mumme M, Feliciano S, Scotti C, Mainil-Varlet P, et al. (August 2014). "Adult human neural crest-derived cells for articular cartilage repair". Science Translational Medicine. 6 (251): 251ra119. doi:10.1126/scitranslmed.3009688. PMID   25163479. S2CID   5520982.
  31. Dell'Accio F, De Bari C, Luyten FP (July 2001). "Molecular markers predictive of the capacity of expanded human articular chondrocytes to form stable cartilage in vivo". Arthritis and Rheumatism. 44 (7): 1608–1619. doi:10.1002/1529-0131(200107)44:7<1608::AID-ART284>3.0.CO;2-T. PMID   11465712.
  32. Scott MA, Levi B, Askarinam A, Nguyen A, Rackohn T, Ting K, et al. (March 2012). "Brief review of models of ectopic bone formation". Stem Cells and Development. 21 (5): 655–667. doi:10.1089/scd.2011.0517. PMC   3295855 . PMID   22085228.
  33. Bichara DA, Zhao X, Hwang NS, Bodugoz-Senturk H, Yaremchuk MJ, Randolph MA, Muratoglu OK (October 2010). "Porous poly(vinyl alcohol)-alginate gel hybrid construct for neocartilage formation using human nasoseptal cells". The Journal of Surgical Research. 163 (2): 331–336. doi:10.1016/j.jss.2010.03.070. PMID   20538292.
  34. Dobratz EJ, Kim SW, Voglewede A, Park SS (2009). "Injectable cartilage: using alginate and human chondrocytes". Archives of Facial Plastic Surgery. 11 (1): 40–47. doi:10.1001/archfacial.2008.509. PMID   19153292.
  35. Kim SW, Dobratz EJ, Ballert JA, Voglewede AT, Park SS (January 2009). "Subcutaneous implants coated with tissue-engineered cartilage". The Laryngoscope. 119 (1): 62–66. doi:10.1002/lary.20025. PMID   19117288. S2CID   22733319.
  36. Bermueller C, Schwarz S, Elsaesser AF, Sewing J, Baur N, von Bomhard A, et al. (October 2013). "Marine collagen scaffolds for nasal cartilage repair: prevention of nasal septal perforations in a new orthotopic rat model using tissue engineering techniques". Tissue Engineering. Part A. 19 (19–20): 2201–2214. doi:10.1089/ten.TEA.2012.0650. PMC   3762606 . PMID   23621795.
  37. Elsaesser AF, Bermueller C, Schwarz S, Koerber L, Breiter R, Rotter N (June 2014). "In vitro cytotoxicity and in vivo effects of a decellularized xenogeneic collagen scaffold in nasal cartilage repair". Tissue Engineering. Part A. 20 (11–12): 1668–1678. doi:10.1089/ten.TEA.2013.0365. PMID   24372309.