Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 285
Filter
1.
Biomaterials ; 286: 121544, 2022 07.
Article in English | MEDLINE | ID: mdl-35633592

ABSTRACT

Osteoarthritis (OA) is a progressive degenerative joint disease which results in chronic degeneration of articular cartilage and sclerosis of bone. While tendons and ligaments may heal to a limited extent, articular cartilage has poor intrinsic regenerative potential, and critical-sized bone defects and pathological fractures cannot regenerate spontaneously. OA represents a significant burden of disease globally, affecting 240 million people in the world. The objective of tissue engineering is to recapitulate the natural healing cascade and developmental process by transplanting stromal and progenitor cells which can act directly or indirectly. As the ultimate goal of regenerative medicine is to avoid in vitro expansion of cells and its associated complications, the adipose-derived stromal cell (ASC) is an attractive progenitor cell for tissue engineering for treatment of OA. While clinical studies are still in their infancy, ASCs together with novel scaffold materials represent promising treatment options for patients suffering from OA. How ASCs exert their regenerative potential is a topic of debate, whereby it may be a result of direct differentiation of ASCs into the desired regenerating tissue, and/or through paracrine activity. With the advancement of material science, it is increasingly possible to enhance engraftment of ASCs through the use of biomaterials or to direct progenitor cell fate by activating biophysical signals through designed material microstructures. There are currently over 180 completed or ongoing registered early stage clinical trials involving ASCs, with 17 completed studies reviewed herein detailing the use of ASCs in OA. In order for ASC therapy to become an "off-the-shelf" option for treating OA, several strategies are currently being explored such as ASC cryopreservation and use of allogeneic ASCs. Newer approaches, such as exosome therapy, allow for the use of acellular ASC-derived therapies and are also currently the focus of ongoing investigations.


Subject(s)
Osteoarthritis , Tissue Engineering , Adipose Tissue , Cell Differentiation , Humans , Osteoarthritis/therapy , Stem Cells , Stromal Cells , Tissue Engineering/methods
3.
Drug Deliv Transl Res ; 6(2): 159-73, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25990836

ABSTRACT

Unlike many other postnatal tissues, bone can regenerate and repair itself; nevertheless, this capacity can be overcome. Traditionally, surgical reconstructive strategies have implemented autologous, allogeneic, and prosthetic materials. Autologous bone--the best option--is limited in supply and also mandates an additional surgical procedure. In regenerative tissue engineering, there are myriad issues to consider in the creation of a functional, implantable replacement tissue. Importantly, there must exist an easily accessible, abundant cell source with the capacity to express the phenotype of the desired tissue, and a biocompatible scaffold to deliver the cells to the damaged region. A literature review was performed using PubMed; peer-reviewed publications were screened for relevance in order to identify key advances in stem and progenitor cell contribution to the field of bone tissue engineering. In this review, we briefly introduce various adult stem cells implemented in bone tissue engineering such as mesenchymal stem cells (including bone marrow- and adipose-derived stem cells), endothelial progenitor cells, and induced pluripotent stem cells. We then discuss numerous advances associated with their application and subsequently focus on technological advances in the field, before addressing key regenerative strategies currently used in clinical practice. Stem and progenitor cell implementation in bone tissue engineering strategies have the ability to make a major impact on regenerative medicine and reduce patient morbidity. As the field of regenerative medicine endeavors to harness the body's own cells for treatment, scientific innovation has led to great advances in stem cell-based therapies in the past decade.


Subject(s)
Adult Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Osteogenesis , Tissue Engineering/methods , Animals , Humans , Regenerative Medicine/methods , Stem Cell Transplantation , Tissue Scaffolds
4.
Br J Surg ; 102(2): e41-55, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25627135

ABSTRACT

BACKGROUND: Throughout history, surgeons have been prolific innovators, which is hardly surprising as most surgeons innovate daily, tailoring their intervention to the intrinsic uniqueness of each operation, each patient and each disease. Innovation can be defined as the application of better solutions that meet new requirements, unarticulated needs or existing market needs. In the past two decades, surgical innovation has significantly improved patient outcomes, complication rates and length of hospital stay. There is one key area that has great potential to change the face of surgical practice and which is still in its infancy: the realm of regenerative medicine and tissue engineering. METHODS: A literature review was performed using PubMed; peer-reviewed publications were screened for relevance in order to identify key surgical innovations influencing regenerative medicine, with a focus on osseous, cutaneous and soft tissue reconstruction. RESULTS: This review describes recent advances in regenerative medicine, documenting key innovations in osseous, cutaneous and soft tissue regeneration that have brought regenerative medicine to the forefront of the surgical imagination. CONCLUSION: Surgical innovation in the emerging field of regenerative medicine has the ability to make a major impact on surgery on a daily basis.


Subject(s)
Inventions/trends , Regenerative Medicine/trends , Surgical Procedures, Operative/trends , Tissue Engineering/trends , Adipose Tissue/transplantation , Bone Regeneration/physiology , Cell- and Tissue-Based Therapy/methods , Cell- and Tissue-Based Therapy/trends , Cicatrix/prevention & control , Connective Tissue/transplantation , Equipment Design/trends , Humans , Intercellular Signaling Peptides and Proteins/therapeutic use , Osteogenesis, Distraction/instrumentation , Osteogenesis, Distraction/methods , Osteogenesis, Distraction/trends , Skin Physiological Phenomena , Therapies, Investigational/methods , Therapies, Investigational/trends , Tissue Engineering/methods , Tissue Scaffolds/trends , Transplantation, Autologous/instrumentation , Transplantation, Autologous/methods , Transplantation, Autologous/trends
5.
J Dent Res ; 93(12): 1187-95, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25139365

ABSTRACT

Conditions such as congenital anomalies, cancers, and trauma can all result in devastating deficits of bone in the craniofacial skeleton. This can lead to significant alteration in function and appearance that may have significant implications for patients. In addition, large bone defects in this area can pose serious clinical dilemmas, which prove difficult to remedy, even with current gold standard surgical treatments. The craniofacial skeleton is complex and serves important functional demands. The necessity to develop new approaches for craniofacial reconstruction arises from the fact that traditional therapeutic modalities, such as autologous bone grafting, present myriad limitations and carry with them the potential for significant complications. While the optimal bone construct for tissue regeneration remains to be elucidated, much progress has been made in the past decade. Advances in tissue engineering have led to innovative scaffold design, complemented by progress in the understanding of stem cell-based therapy and growth factor enhancement of the healing cascade. This review focuses on the role of biomaterials for craniofacial bone engineering, highlighting key advances in scaffold design and development.


Subject(s)
Biocompatible Materials/therapeutic use , Plastic Surgery Procedures/methods , Skull/surgery , Tissue Engineering/methods , Tissue Scaffolds , Facial Bones/surgery , Humans , Intercellular Signaling Peptides and Proteins/therapeutic use , Prosthesis Design , Stem Cell Transplantation/methods
6.
Med Eng Phys ; 35(3): 392-402, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22784673

ABSTRACT

The primary cilium is an organelle that senses cues in a cell's local environment. Some of these cues constitute molecular signals; here, we investigate the extent to which primary cilia can also sense mechanical stimuli. We used a conditional approach to delete Kif3a in pre-osteoblasts and then employed a motion device that generated a spatial distribution of strain around an intra-osseous implant positioned in the mouse tibia. We correlated interfacial strain fields with cell behaviors ranging from proliferation through all stages of osteogenic differentiation. We found that peri-implant cells in the Col1Cre;Kif3a(fl/fl) mice were unable to proliferate in response to a mechanical stimulus, failed to deposit and then orient collagen fibers to the strain fields caused by implant displacement, and failed to differentiate into bone-forming osteoblasts. Collectively, these data demonstrate that the lack of a functioning primary cilium blunts the normal response of a cell to a defined mechanical stimulus. The ability to manipulate the genetic background of peri-implant cells within the context of a whole, living tissue provides a rare opportunity to explore mechanotransduction from a multi-scale perspective.


Subject(s)
Bone and Bones/pathology , Cilia/physiology , Osteogenesis , Animals , Bone and Bones/metabolism , Cell Proliferation , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Kinesins/metabolism , Male , Mice , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/metabolism , Poisson Distribution , Prostheses and Implants , Regenerative Medicine/methods , Signal Transduction , Stress, Mechanical , Tibia/pathology
7.
Oral Dis ; 17(6): 541-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21332599

ABSTRACT

Wound healing is a complex biological process that affects multiple tissue types. Wounds in the oral cavity are particularly challenging given the variety of tissue types that exist in close proximity to one another. The goal of regenerative medicine is to facilitate the rapid replacement of lost or damaged tissue with tissue that is functional, and physiologically similar to what previously existed. This review provides a general overview of wound healing and regenerative medicine, focusing specifically on how recent advances in the fields of stem cell biology, tissue engineering, and oral disease could translate into improved clinical outcomes.


Subject(s)
Mouth Diseases/therapy , Mouth Mucosa/physiology , Regeneration/physiology , Wound Healing/physiology , Adult Stem Cells/physiology , Cicatrix/prevention & control , Embryonic Stem Cells/physiology , Humans , Inflammation , Mesenchymal Stem Cells/physiology , Pluripotent Stem Cells/physiology , Stem Cells/physiology , Tissue Engineering
8.
Panminerva Med ; 51(1): 25-41, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19352307

ABSTRACT

Today, surgical intervention remains the mainstay of treatment to intervene upon a multitude of skeletal deficits and defects attributable to congenital malformations, oncologic resection, pathologic degenerative bone destruction, and post-traumatic loss. Despite this significant demand, the tools with which surgeons remain equipped are plagued with a surfeit of inadequacies, often resulting in less than ideal patient outcomes. The failings of current techniques largely arise secondary to their inability to produce a regenerate which closely resembles lost tissue. As such, focus has shifted to the potential of mesenchymal stem cell (MSC)-based skeletal tissue engineering. The successful development of such techniques would represent a paradigm shift from current approaches, carrying with it the potential to regenerate tissues which mimic the form and function of endogenous bone. Lessons learned from investigations probing the endogenous regenerative capacity of skeletal tissues have provided direction to early studies investigating the osteogenic potential of MSC. Additionally, increasing attention is being turned to the role of targeted molecular manipulations in augmenting MSC osteogenesis, as well as the development of an ideal scaffold ''vehicle'' with which to deliver progenitor cells. The following discussion presents the authors' current working knowledge regarding these critical aspects of MSC application in cell-based skeletal tissue engineering strategies, as well as provides insight towards what future steps must be taken to make their clinical translation a reality.


Subject(s)
Bone Diseases/surgery , Bone and Bones/surgery , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Regenerative Medicine , Tissue Engineering , Animals , Bone Diseases/metabolism , Bone Diseases/pathology , Bone Regeneration , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Differentiation , Cell Proliferation , Humans , Osteogenesis , Signal Transduction , Tissue Scaffolds
9.
J Hand Surg Eur Vol ; 33(6): 783-90, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18694919

ABSTRACT

Dupuytren's disease is characterised by nodular fibroblastic proliferation of the palmar fascia leading to contracture of the hand. Transforming growth factor beta (TGF-beta) is thought to play a role in its pathogenesis. We performed a cDNA microarray analysis of Dupuytren's diseased cord tissue with an emphasis on TGF-beta isoforms. Normal-appearing transverse ligament of the palmar fascia from adjacent to the diseased cord and palmar fascia from patients undergoing carpal tunnel release were used as controls. TGF-beta gene expression was confirmed by quantitative real-time polymerase chain reaction. Over 20 unique genes were found to be significantly up-regulated, including several previously reported genes. A dominant increase in TGF-beta2 expression was seen in the cord tissue, whereas TGF-beta1 and TGF-beta3 were found not to be significantly up-regulated. Quantitative real-time polymerase chain reaction confirmed these findings. This gene expression profile allows for further experiments that may eventually lead to gene therapy to block the development and progression of Dupuytren's disease clinically.


Subject(s)
Dupuytren Contracture/genetics , Transforming Growth Factor beta2/genetics , Aged , Female , Gene Expression Profiling , Humans , Male , Microarray Analysis , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
10.
Comput Methods Biomech Biomed Engin ; 11(5): 453-61, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18608339

ABSTRACT

It has been proposed that periosteal residual tensile strains influence periosteal bone apposition and endochondral ossification. The role of bone growth rates on the development of residual strains is not well known. This study examined the relationships between specific growth rate and residual strains in chick tibiotarsi. We measured length and circumference during embryonic days 11-20 using microCT. Bones grew faster in length, with longitudinal and circumferential specific growth rates decreasing from 17 to 9% and 14 to 8% per day, respectively. To calculate residual strains, opening dimensions of incisions through the periosteum were analysed using finite element techniques. Results indicate that Poisson's ratio for an isotropic material model is between 0 and 0.04. For the model with Poisson's ratio 0.03, longitudinal and circumferential residual strains decreased from 46.2 to 29.3% and 10.6 to 3.9%, respectively, during embryonic days 14-20. Specific growth rates and residual strains were positively correlated (p<0.05).


Subject(s)
Bone Development/physiology , Calcification, Physiologic , Osteogenesis/physiology , Periosteum/physiology , Tensile Strength/physiology , Aging/physiology , Animals , Bone and Bones/embryology , Bone and Bones/physiology , Chick Embryo , Finite Element Analysis , Periosteum/embryology , Stress, Mechanical , Tomography, X-Ray Computed/methods
11.
J Dent Res ; 85(11): 966-79, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17062735

ABSTRACT

Craniofacial tissue engineering promises the regeneration or de novo formation of dental, oral, and craniofacial structures lost to congenital anomalies, trauma, and diseases. Virtually all craniofacial structures are derivatives of mesenchymal cells. Mesenchymal stem cells are the offspring of mesenchymal cells following asymmetrical division, and reside in various craniofacial structures in the adult. Cells with characteristics of adult stem cells have been isolated from the dental pulp, the deciduous tooth, and the periodontium. Several craniofacial structures--such as the mandibular condyle, calvarial bone, cranial suture, and subcutaneous adipose tissue--have been engineered from mesenchymal stem cells, growth factor, and/or gene therapy approaches. As a departure from the reliance of current clinical practice on durable materials such as amalgam, composites, and metallic alloys, biological therapies utilize mesenchymal stem cells, delivered or internally recruited, to generate craniofacial structures in temporary scaffolding biomaterials. Craniofacial tissue engineering is likely to be realized in the foreseeable future, and represents an opportunity that dentistry cannot afford to miss.


Subject(s)
Mesenchymal Stem Cells , Periodontium/cytology , Regeneration/physiology , Skull/cytology , Tissue Engineering , Absorbable Implants , Adipose Tissue/cytology , Adult , Adult Stem Cells , Animals , Dental Pulp/cytology , Gene Transfer Techniques , Humans , Mandibular Condyle/cytology , Mesenchymal Stem Cell Transplantation , Temporomandibular Joint/cytology
12.
Orthod Craniofac Res ; 8(4): 259-66, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16238606

ABSTRACT

Reconstruction of craniofacial bony deficiencies, whether acquired through trauma or as a result of treatment for disease, is a chronic problem. Although numerous approaches utilizing a wide array of materials ranging from alloplastic materials to autogenous bone grafts have been employed to achieve bony replacement, no ideal clinical approach exists. In this brief review, we will provide an overview of current approaches to treating craniofacial bony defects. We will then discuss advances being made in the design of scaffolding materials and potential candidate cell types with which to design tissue-engineered constructs for craniofacial skeletal repair.


Subject(s)
Biomimetic Materials , Bone Regeneration , Craniotomy/methods , Membranes, Artificial , Stem Cell Transplantation , Tissue Engineering/methods , Adipose Tissue/cytology , Animals , Biomimetic Materials/pharmacology , Bone Marrow Cells , Bone Regeneration/drug effects , Craniofacial Abnormalities/surgery , Humans , Lactic Acid/pharmacology , Polyglycolic Acid/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers/pharmacology , Plastic Surgery Procedures
13.
J Pathol ; 207(2): 232-42, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16052471

ABSTRACT

Smad signalling plays important roles in developmental and cancer biology as well as in fibropathogenesis. Its role in keloid biology is not known. Epithelial-mesenchymal interactions, originally described in normal skin, have recently been established to play a significant role in keloid pathogenesis, and demonstrate the important influence of keratinocyte paracrine factor signalling on fibroblast behaviour. The present study investigated the role of downstream Smad cascade induction in this interaction. Normal fibroblasts (NF) and keloid fibroblasts (KF) were co-cultured in serum-free medium with normal keratinocytes (NK) or keloid keratinocytes (KK) for 5 days, after which fibroblast cell lysates were subjected to western blot and immunoprecipitation analysis to quantify the levels of Smad and Smad2/3/4 binding complex. In another set of experiments, wild-type (wt), Smad2-null (Smad2-/-) and Smad3-null (Smad3-/-) mouse embryonic fibroblasts (MEF) were assayed for cell proliferation and collagen production after serum-free co-culture with KK or exposure to conditioned media collected from serum-free KK/KF co-culture. Compared to normal skin, keloids expressed high basal levels of TGFbetaR1 and TGFbetaR2, Smad2, 3 and 4 and phospho-Smad2. Upregulation of TGFbetaR1 and TGFbetaR2, Smad3 and p-Smad2 was observed in KF co-cultured with KK, together with enhanced Smad3 phosphorylation and Smad2/3/4 binding complex production. When MEF-wt, MEF-Smad2-/- or MEF-Smad3-/- were co-cultured with KK or exposed to KK/KF co-culture conditioned media, enhanced proliferation and collagen production were seen in MEF-wt and MEF-Smad2-/- but not in MEF-Smad3-/- cells. The activation of Smad signalling, importantly that of Smad3, appears to be one facet of the complex epithelial-mesenchymal interactions in keloid pathogenesis, resulting in active KF proliferation and collagen-ECM production in co-culture with KK. This finding suggests the suppression of Smad signalling as a novel approach in keloid therapy.


Subject(s)
DNA-Binding Proteins/analysis , Keloid/metabolism , Signal Transduction/physiology , Trans-Activators/analysis , Adolescent , Adult , Benzamides/pharmacology , Burns/metabolism , Cell Division/drug effects , Child , Cicatrix/metabolism , Coculture Techniques/methods , Collagen Type I/analysis , Collagen Type III/analysis , Dioxoles/pharmacology , Epithelial Cells/metabolism , Female , Fibroblasts/metabolism , Fibronectins/analysis , Humans , Keratinocytes/metabolism , Male , Mesoderm/metabolism , Phosphorylation , Receptors, Transforming Growth Factor beta/analysis , Smad2 Protein , Smad3 Protein , Smad4 Protein
14.
Rev Stomatol Chir Maxillofac ; 105(1): 23-5, 2004 Feb.
Article in French | MEDLINE | ID: mdl-15041867

ABSTRACT

Distraction osteogenesis has become a mainstay in bone engineering and the recent application of this technique to the membranous craniofacial skeleton has significantly improved our armamentarium for reconstructive craniomaxillofacial procedures. However, if the biomechanical, histological and ultrastructural changes associated with distraction osteogenesis have been widely described, the molecular mechanisms governing the formation of new bone in the interfragmental gap of gradually distracted bone segments remain largely unclear. Recently, our laboratory has described a rat mandibular distraction model that provides an excellent environment for deciphering the molecular mechanisms that mediate distraction osteogenesis. In this Article, we present the hypotheses and current research that have furthered our knowledge of the molecular mechanisms that govern distraction osteogenesis. Recent studies have implicated a growing number of cytokines that are intimately involved in the regulation of bone synthesis and turnover. The gene regulation of numerous cytokines (Transforming Growth Factor-B, Bone Morphogenetic Proteins, Insulin-like Growth Factor-1, Fibroblast Growth Factor-2) during distraction osteogenesis have been best characterized and will be discussed in this text. We believe that novel systems like the rat model will facilitate our understanding of the biomolecular mechanisms that mediate membranous distraction osteogenesis and will ultimately guide the development of targeted-strategies designed to accelerate bone healing.


Subject(s)
Bone Remodeling/physiology , Growth Substances/genetics , Growth Substances/physiology , Osteogenesis, Distraction , Animals , Bone Morphogenetic Proteins/biosynthesis , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/physiology , Bone Remodeling/genetics , Fibroblast Growth Factor 2/biosynthesis , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/physiology , Gene Expression Regulation , Growth Substances/biosynthesis , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/physiology , Mandible/surgery , Models, Animal , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/physiology
15.
J Pediatr Surg ; 38(3): 296-300; discussion 296-300, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12632338

ABSTRACT

BACKGROUND/PURPOSE: Neonates with large congenital diaphragmatic hernias (CDH) require prosthetic patch closure of the defect because of the paucity of native diaphragmatic tissue. As the child grows, patch separation can occur necessitating reoperation. Use of vascularized autologous tissue may decrease the incidence of reherniation as tissue incorporation and growth may be improved. The authors report our early experience using a local muscle advancement flap with microneural anastomosis for those children in whom reherniation develops after prosthetic patch placement. METHODS: Seven patients with CDH (6 left and 1 right) whose synthetic diaphragmatic patch separated from the chest wall resulting in a clinically significant recurrent hernia were followed up with prospectively. After dissecting the ipsilateral latissimus dorsi off the chest wall and dividing the thoracodorsal neurovascular bundle (based on its lumbar blood supply), the synthetic patch was removed via an eighth intercostal incision. The muscle flap was placed into the hemithorax through the bed of the tenth rib and sutured in place over a Vicryl mesh scaffold. The thoracodorsal nerve was anastomosed to the phrenic nerve. Functional analysis of the flap was performed in 4 patients. RESULTS: Age at placement of the muscle graft ranged from 2 months to 48 months (median, 24 months). There has been no evidence of reherniation after placement of the muscle graft. Long-term outcome and functional analysis of the flap was available in 4 patients (mean, 19 months). Two infants had fluoroscopic and sonographic evidence of nonparadoxical neodiaphragmatic motion. In one of these, electromyographic evidence of function was documented with a phrenic nerve conduction velocity of 22 meters per second. The third infant showed no evidence of neodiaphragmatic motion, and the fourth infant had paradoxical motion. CONCLUSIONS: This is the first direct documentation of phrenic nerve function in an infant with CDH. An innervated reversed latissimus dorsi (RLD) flap reconstruction for recurrent CDH provides an alternative to prosthetic patch repair. This technique offers the advantages of autologous vascularized tissue with potential phrenic nerve innervation and physiologic neodiaphragmatic motion.


Subject(s)
Hernia, Diaphragmatic/surgery , Muscle, Skeletal/surgery , Surgical Flaps , Female , Follow-Up Studies , Hernias, Diaphragmatic, Congenital , Humans , Infant, Newborn , Male , Muscle, Skeletal/blood supply , Muscle, Skeletal/innervation , Nerve Transfer , Phrenic Nerve/surgery , Recurrence , Reoperation , Retrospective Studies , Surgical Mesh , Treatment Outcome
16.
Clin Plast Surg ; 28(4): 719-30, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11727856

ABSTRACT

Plastic surgery research affords tremendous opportunities in a variety of affluent mode systems. Only recently have researchers applied molecular biologic techniques to common plastic surgery problems. For example, investigating the fundamental biomolecular mechanisms of normal palate and cranial suture morphogenesis will improve the understanding of the etiopathogenesis of CLP and craniosynostosis and facilitate the development of biologically-based interventions. Furthermore, as interdisciplinary collaborations improve, surgeons can expect to see remarkable progress in de novo tissue synthesis, replacement, and repair. Ultimately, they may one day find that gene-modified endogenous tissue engineering will succeed today's biocompatible scaffolds and allogeneic or zenogeneic replacement strategies. In general, plastic surgeons can look forward to the development of highly effective biomolecular treatments for clinical problems such as complex wound repair, prolific scarring, bone deficits (or surpluses), and organ system replacement or repair. Researchers believe that biologically-based strategies like these will be combined with technical advances that harness minimally invasive approaches. Together, clinicians expect these new tactics will reduce morbidity and improve the results of clinical problems treated by plastic surgeons.


Subject(s)
Research/trends , Surgery, Plastic/trends , Animals , Biocompatible Materials/therapeutic use , Cleft Lip/surgery , Craniosynostoses/surgery , Disease Models, Animal , Humans , Osteogenesis, Distraction/methods , Tissue Engineering
17.
Yonsei Med J ; 42(6): 634-45, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11754147

ABSTRACT

Cutaneous wounding in adult humans and higher vertebrate animals results in scar formation. In contrast, both human and animal fetuses, at early gestational ages, exhibit skin wound healing without scarring. This distinction suggests that the repair of adult wounds by skin regeneration, rather than by fibrosis, may be achieved if adult wounds can be modified to mimic the healing process of fetal wounds. The development of gene therapy offers the possibility to specifically enhance or block the gene expression of cytokines and extracellular molecules, and thus convert adult wound healing into a healing process more similar to tissue regeneration. This article reviews the characteristics of fetal wound repair focusing on cytokine profiles and the inflammatory response to dermal injury. Also included are new developments in gene transfer techniques as well as their application in wound healing. Finally, the authors propose possible strategies of wound gene therapy, to reduce wound scarring and to promote tissue regeneration.


Subject(s)
Cicatrix/prevention & control , Genetic Therapy , Animals , Fetus/physiology , Humans , Wound Healing/physiology
18.
Yonsei Med J ; 42(6): 646-59, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11754148

ABSTRACT

Craniosynostosis occurs in approximately 1:2000 live births. It may affect the coronal, sagittal, metopic and lambdoid sutures in isolation or in combination. Although non-syndromic synostoses are more common, over 150 genetic syndromes have been identified. Recent advances in genetic mapping have linked chromosomal mutations with craniosynostotic syndromes. Despite the identification of these genetic mutations, the fundamental biomolecular mechanisms mediating cranial suture biology remain unknown. Today, many laboratories are investigating murine cranial suture biology as a model for human cranial suture development and fusion. Normal murine cranial suture biology is very complex, but evidence suggests that the dura mater provides the biomolecular blueprints (e.g. the soluble growth factors), which guide the fate of the pleuripotent osteogenic fronts. While our knowledge of these dura-derived signals has increased dramatically in the last decade, we have barely begun to understand the fundamental mechanisms that mediate cranial suture fusion or patency. Interestingly, recent advances in both premature human and programmed murine suture fusion have revealed unexpected results, and have generated more questions than answers.


Subject(s)
Craniosynostoses/etiology , Fetus/physiology , Animals , Craniosynostoses/genetics , Craniosynostoses/surgery , Embryonic and Fetal Development , Humans , Mutation
19.
J Craniofac Surg ; 12(6): 573-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11711826

ABSTRACT

Mandibular fractures, resulting from either trauma or reconstructive surgery, can be challenging craniofacial problems. The morbidity of failed fracture healing is significant and may require bone grafting. Donor site morbidity and finite amounts of autogenous bone are major drawbacks of autogenous bone grafting. Similarly, the use of allografts and xenografts may be associated with an increased risk of rejection, infection, and nonunion. To circumvent the limitations of bone grafting, research efforts have focused on formulating a suitable bone substitute. The purpose of our study was to evaluate the efficacy of type I collagen implants in repairing critical sized mandibular defects in rats. Twelve male Sprague-Dawley rats (200-300g) were divided equally into control and experimental groups. Full thickness, round, four millimeter in diameter defects were created in the ramus of the right mandible of all rats using an electrical burr at low speed. The defects were irrigated of all bone chips, and either filled with a precisely fitted disk of allogenic collagen type I gel (experimental animals) or left empty (control animals). Animals were killed 6 weeks after surgery and healing of the bone defects was assessed in a blinded fashion using radiologic and histologic analysis. Radiologic analysis of the control group revealed a clear circular right mandibular defect in all animals, whereas the collagen disk implant group revealed an indistinct to nonexistent right mandibular defect in all animals. Densitometric analysis revealed a significant difference between these groups (* P = 0.01). Similarly, gross analysis of control mandibles revealed a 4mm round, soft-tissue filled defect, while implanted defects demonstrated gross bone spanning the defect. Finally, histologic analysis of all control mandibles revealed clearly demarcated bony edges at the defect border with connective tissue spanning the defect. In contrast, histological analysis of all implanted mandibles revealed indistinct bony edges at the defect border with a thin layer of osteoblasts and viable bone spanning the defects. We have demonstrated the ability of type I collagen to promote healing of a membranous bony defect that would not otherwise heal at 6 weeks. The suitability of type I collagen as a carrier matrix provides ample opportunity for tissue-engineered approaches to further facilitate bony defect healing. Promoting bone formation through tissue engineering matrices offers great promise for skeletal healing and reconstruction.


Subject(s)
Bone Substitutes/therapeutic use , Collagen Type I/therapeutic use , Mandibular Diseases/surgery , Absorptiometry, Photon , Analysis of Variance , Animals , Coloring Agents , Connective Tissue/pathology , Disease Models, Animal , Drug Carriers , Fluorescent Dyes , Gels , Male , Mandibular Diseases/diagnostic imaging , Mandibular Diseases/pathology , Osteoblasts/pathology , Osteogenesis/physiology , Osteotomy , Rats , Rats, Sprague-Dawley , Single-Blind Method , Statistics as Topic , Tissue Engineering , Treatment Outcome , Wound Healing
20.
J Hand Surg Am ; 26(6): 1082-7, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11721255

ABSTRACT

Scar production and neuroma formation at nerve graft coaptation sites may limit axonal regeneration and impair functional outcome. Transforming growth factor beta (TGF-beta) is a family of growth factors that is involved in scar formation, wound healing, and nerve regeneration. Fifteen adult Sprague-Dawley rats underwent autogenous nerve grafting. The nerve grafts were analyzed by in situ hybridization to determine the temporal and spatial expression of TGF-beta1 and TGF-beta3 messenger RNA (mRNA). The grafted nerves showed increased expression of TGF-beta1 and TGF-beta3 mRNA in the nerve and the surrounding connective tissue during the first postoperative week. These data suggest that modulation of TGF-beta levels in the first postoperative week may be effective in helping to control scar formation and improve nerve regeneration.


Subject(s)
Nerve Regeneration/physiology , RNA, Messenger/genetics , Transforming Growth Factor beta/genetics , Wound Healing/physiology , Animals , Gene Expression , In Situ Hybridization , Nerve Transfer , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...