Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Biomedicines ; 10(9)2022 Aug 29.
Article in English | MEDLINE | ID: mdl-36140217

ABSTRACT

White adipose tissues are major endocrine organs that release factors, termed adipokines, which affect other major organ systems. The development and functions of adipose tissues depend largely upon the glycosaminoglycan heparan sulfate. Heparan sulfate proteoglycans (HSPGs) surround both adipocytes and vascular structures and facilitate the communication between these two components. This communication mediates the continued export of adipokines from adipose tissues. Heparan sulfates regulate cellular physiology and communication through a sulfation code that ionically interacts with heparan-binding regions on a select set of proteins. Many of these proteins are growth factors and chemokines that regulate tissue function and inflammation. Cells regulate heparan sulfate sulfation through the release of heparanases and sulfatases. It is now possible to tissue engineer vascularized adipose tissues that express heparan sulfate proteoglycans. This makes it possible to use these tissue constructs to study the role of heparan sulfates in the regulation of adipokine production and release. It is possible to regulate the production of heparanases and sulfatases in order to fine-tune experimental studies.

2.
J Orthop Res ; 36(6): 1757-1766, 2018 06.
Article in English | MEDLINE | ID: mdl-29194731

ABSTRACT

Regenerative medicine and tissue engineering studies are actively developing novel means to repair adult articular cartilage defects using biological approaches. One such approach is the harnessing of adult human therapeutic cells such as those referred to as mesenchymal stem cells. Upon exposure to chondrogenic signals, these cells differentiate and initiate the production of a complex and voluminous cartilaginous matrix that is crucial to both the structure and function of cartilage. Furthermore, this complexity requires the time-sensitive activation of a large number of genes to produce the components of this matrix. The current study analyzed the kinetics of matrix production in an aggregate culture model where adult human mesenchymal stem cells were induced to differentiate as chondrocytes. The results indicate the existence of a biphasic mode of differentiation and maturation during which matrix genes and molecules are differentially activated and secreted. These results have important implications for developing novel approaches for the creation of tissue engineered articular cartilage. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1757-1766, 2018.


Subject(s)
Chondrocytes/cytology , Extracellular Matrix/metabolism , Mesenchymal Stem Cells/cytology , Cell Aggregation , Cell Differentiation , Collagen/biosynthesis , Humans , Proteoglycans/biosynthesis
3.
Immunol Lett ; 168(2): 136-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26079607

ABSTRACT

We have evolved cellular mechanisms for protecting us from disabling intruders or injuries. The Mesenchymal Stem Cell, MSC, is derived from perivascular cells, pericytes that are liberated from their basement membrane tethers surrounding blood vessels upon injury or inflammation. These site-activated MSCs produce a curtain of immuno-modulation behind which slow and specific tissue regeneration takes place. In addition, the MSC senses the tissue microenvironment and adjusts the curtain and regenerative activity accordingly. This includes the production of antibiotic proteins like LL37 that both kills intruding bacteria on contact, but calls forth macrophage and other members of the hematopoietic system to further medicate the injury site. Indeed, MSCs appear to be local managers of the tissues' innate regenerative potential. Thus, I propose that MSC should denote "Medicinal Signaling Cells" since these cells are "drug stores" for sites of injury or inflammation.


Subject(s)
Cell Lineage/immunology , Immune System/immunology , Mesenchymal Stem Cells/immunology , Cellular Microenvironment/immunology , Hematopoiesis/immunology , Humans , Immune System/cytology , Immune System/physiology , Immunity, Innate/immunology , Mesenchymal Stem Cells/cytology , Pericytes/cytology , Pericytes/immunology , Regeneration/immunology , Signal Transduction/immunology
4.
Neurogastroenterol Motil ; 25(8): 700-5, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23663526

ABSTRACT

BACKGROUND: Increases in L-cell release of GLP-1 are proposed to serve as a negative feedback signal for postprandial changes in gastric emptying and/or motility. Previous ex vivo data suggests that direct electrical stimulation (E-stim) of ileal segments stimulates secretion of GLP-1. This suggests potential feed-forward increases in GLP-1 driven by intestinal neuronal and/or motor activity. METHODS: To determine if E-stim could increase GLP-1 levels in an in vivo setting, we administered E-stim and nutrients to male Long- Evans rats (300-350 g) under general anesthesia. KEY RESULTS: Nutrient infusion into the duodenum or ileum significantly increased plasma GLP-1 levels, but E-stim applied to these locations did not (P < 0.05). However, the combination of E-stim and nutrient infusion, in either the ileum or duodenum, significantly increased plasma GLP-1 when compared to nutrient infusion alone (P < 0.05), and this effect was not blocked by either norepinephrine or atropine. To test the impact of intestinal motor activity, the effect of extra-luminal mechanical stimulation (M-stim) on GLP-1 levels was assessed. In the duodenum, but not the ileum, M-stim plus nutrient infusion significantly increased GLP-1 over nutrient infusion or M-stim alone (P < 0.05). CONCLUSIONS & INFERENCES: Thus, both E- and M-stim of the duodenum, but only E-stim of the ileum augmented nutrient-stimulated GLP-1 release. These data demonstrate that factors beyond enteral nutrients could contribute to the regulation of GLP-1 secretion.


Subject(s)
Duodenum/metabolism , Food , Glucagon-Like Peptide 1/metabolism , Ileum/metabolism , Animals , Electric Stimulation/methods , Glucagon-Like Peptide 1/blood , Male , Rats , Rats, Long-Evans
5.
Endocrinology ; 153(6): 2647-54, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22518062

ABSTRACT

Leptin resistance is a feature of obesity that poses a significant therapeutic challenge. Any treatment that is effective to reduce body weight in obese patients must overcome or circumvent leptin resistance, which promotes the maintenance of excess body fat in obese individuals. Ciliary neurotrophic factor (CNTF) is unique in its ability to reduce food intake and body weight in obese, leptin-resistant humans and rodents. Although attempts to use CNTF as an obesity therapy failed due to the development of neutralizing antibodies to the drug, efforts to understand mechanisms for CNTF's anorectic effects provide an opportunity to develop new drugs for leptin-resistant individuals. CNTF and leptin share several structural, anatomic, and signaling properties, but it is not understood whether or how the two cytokines might interact to affect energy balance. Here, we conditionally deleted the CNTF receptor (CNTFR) subunit, CNTFRα, in cells expressing leptin receptors. We found that CNTFR signaling in leptin-responsive neurons is not required for endogenous maintenance of energy balance and is not required for the anorectic response to exogenous administration of a CNTF agonist. These results indicate that despite anatomical overlap for CNTF and leptin action, CNTF appears to act within a distinct neuronal population to elicit its potent anorectic effect.


Subject(s)
Appetite Depressants/pharmacology , Ciliary Neurotrophic Factor/pharmacology , Leptin/pharmacology , Neurons/drug effects , Animals , Body Weight/drug effects , Ciliary Neurotrophic Factor Receptor alpha Subunit/genetics , Ciliary Neurotrophic Factor Receptor alpha Subunit/metabolism , Diet, High-Fat , Eating/drug effects , Energy Metabolism/drug effects , Female , Immunohistochemistry , Male , Mice , Mice, Knockout , Mice, Transgenic , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Phosphorylation/drug effects , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
6.
Biomaterials ; 32(36): 9667-76, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21959010

ABSTRACT

An increased demand for soft-tissue substitutes has impelled the development of an in vitro adipose tissue. Ideally, such a tissue should contain a vascular network that can deliver blood throughout the construct following its engraftment. This study describes the in vitro fabrication of a pre-vascularized adipose tissue entirely using a self-assembly approach. Adult human adipose stromal cells (ASCs) provided the foundation for this construct. These cells were cultured at high density in the presence of elevated levels of ascorbate prior to adipocytic induction. Vascular support cells consisting of dermal fibroblasts, mixtures of adipose stromal cells and bone marrow mesenchymal stem cells (MSCs) were introduced to sustain an extensive vascular network formed by human umbilical vein endothelial cells (HUVECs). MSCs were introduced to serve as perivascular cells. The resulting construct contained a vascular-adipose tissue continuum that was held together by basement membrane molecules. This construct contains multiple cell types that are typically found in adipose tissue: adipocytes, pre-adipocytes, stem cells, fibroblasts, vascular cells, and perivascular support cells. As such, these constructs can be employed both for in vitro studies to assay cellular interactions between vasculature and other components of adipose tissue. Further, they can also be engrafted into athymic hosts to study vascular and adipocyte stability.


Subject(s)
Adipocytes/cytology , Adipose Tissue/blood supply , Adipose Tissue/cytology , Tissue Engineering/methods , Basement Membrane/metabolism , Coculture Techniques , Collagen Type IV/metabolism , Extracellular Matrix/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Neovascularization, Physiologic , Stromal Cells/cytology
7.
Glycobiology ; 21(2): 257-68, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20947661

ABSTRACT

Previous work has shown that versican, decorin and a catabolic fragment of decorin, termed decorunt, are the most abundant proteoglycans in human skin. Further analysis of versican indicates that four major core protein species are present in human skin at all ages examined from fetal to adult. Two of these are identified as the V0 and V1 isoforms, with the latter predominating. The other two species are catabolic fragments of V0 and V1, which have the amino acid sequence DPEAAE as their carboxyl terminus. Although the core proteins of human skin versican show no major age-related differences, the glycosaminoglycans (GAGs) of adult skin versican are smaller in size and show differences in their sulfation pattern relative to those in fetal skin versican. In contrast to human skin versican, human skin decorin shows minimal age-related differences in its sulfation pattern, although, like versican, the GAGs of adult skin decorin are smaller than those of fetal skin decorin. Analysis of the catabolic fragments of decorin from adult skin reveals the presence of other fragments in addition to decorunt, although the core proteins of these additional decorin catabolic fragments have not been identified. Thus, versican and decorin of human skin show age-related differences, versican primarily in the size and the sulfation pattern of its GAGs and decorin in the size of its GAGs. The catabolic fragments of versican are detected at all ages examined, but appear to be in lower abundance in adult skin compared with fetal skin. In contrast, the catabolic fragments of decorin are present in adult skin, but are virtually absent from fetal skin. Taken together, these data suggest that there are age-related differences in the catabolism of proteoglycans in human skin. These age-related differences in proteoglycan patterns and catabolism may play a role in the age-related changes in the physical properties and injury response of human skin.


Subject(s)
Aging , Decorin , Skin Aging , Skin , Versicans , Adult , Aging/metabolism , Amino Acid Sequence , Binding Sites, Antibody/genetics , Decorin/genetics , Decorin/metabolism , Drug Combinations , Electrophoresis, Polyacrylamide Gel , Fetus/metabolism , Humans , Immunoblotting , Middle Aged , Protein Isoforms/genetics , Protein Isoforms/metabolism , Skin/metabolism , Sulfamonomethoxine/metabolism , Trimethoprim/metabolism , Versicans/genetics , Versicans/metabolism , Young Adult
8.
Stem Cell Res Ther ; 1(4): 30, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20863417

ABSTRACT

Mesenchymal stem cells are a heterogeneous population of fibroblast-like cells found in most adult organs. However, most of our current knowledge is based on cells of bone marrow or interstitial adipose tissues. These cells are capable of differentiation along various mesenchymal lineages. In addition, they have demonstrated therapeutic characteristics in wounds and ischemic situations. The therapeutic characteristics of these cells are activated upon their entering wounds or other damaged tissues. A current problem is the development of strategies that ensure that these cells reach wound beds in a timely fashion and in sufficient numbers to maximize their therapeutic benefits. Currently, there are two basic delivery methods: systemic infusion of cells into the vascular circulation and direct application of therapeutic cells to wound sites. Skin wounds are optimal candidates for the topical delivery approach. However, the methods by which therapeutic cells are delivered to such wounds vary. This review outlines the basic methods used to deliver therapeutic cells to skin and other wounds. Upon entering wounds, therapeutic cells interact with other wound cells through paracrine mechanisms that are not yet well understood. Nonetheless, interactions with vascular endothelial cells and immunomodulation appear to play significant roles in accelerating wound healing and in reducing scar formation upon the completion of the healing process. Although the phenomenological body of evidence indicating the efficacy of therapeutic cells is substantial, considerable work is still required to better determine the molecular and cellular functions of these cells and to assess their fate and the long-term consequences of their application.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Wound Healing/physiology , Wounds and Injuries/therapy , Cell Differentiation , Drug Administration Routes , Endothelial Cells/cytology , Humans , Mesenchymal Stem Cells/metabolism
9.
Int Rev Cell Mol Biol ; 276: 161-214, 2009.
Article in English | MEDLINE | ID: mdl-19584013

ABSTRACT

The capacity of fibroblasts to produce and organize the extracellular matrix and to communicate with other cells makes them a central component of tissue biology. Even so, fibroblasts remain a somewhat enigmatic population. Our inability to fully comprehend these cells is in large part due to the paucity of unique cellular markers and to their pervasive diversity. Much of our understanding of fibroblast diversity has evolved from studies where subpopulations of these cells have been produced without resorting to cell surface markers. In this regard, cloning and mechanical separation of tissues prior to establishing cultures has provided multiple subpopulations. Nonetheless, in isolated situations, the expression or lack of expression of Thy-1/CD90 has been used to separate fibroblast subsets. The role of fibroblasts in intercellular communication is emerging through the implementation of organotypic studies in which three-dimensional fibroblast culture are combined with other populations of cells. Such studies have revealed critical paracrine loops that are essential for organ development and for wound repair. These studies also provide a backdrop for the emerging field of tissue engineering. The participation of fibroblasts in the regulation of tissue homeostasis and their contribution to the aging process are emerging issues that require better understanding. In short, fibroblasts represent a multifaceted, complex group of cells.


Subject(s)
Fibroblasts/physiology , Aging/physiology , Animals , Biomarkers/metabolism , Cell Culture Techniques , Cell Differentiation/physiology , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/physiology , Epithelial Cells/cytology , Epithelial Cells/physiology , Extracellular Matrix/metabolism , Fibroblasts/cytology , Hair Follicle/cytology , Humans , Neuropeptides/metabolism , Thy-1 Antigens/genetics , Thy-1 Antigens/metabolism , Tissue Engineering
10.
Tissue Eng Part A ; 15(7): 1751-61, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19196139

ABSTRACT

The effective delivery of bioactive molecules to wound sites hasten repair. Cellular therapies provide a means for the targeted delivery of a complex, multiple arrays of bioactive factors to wound sites. Thus, the identification of ideal therapeutic populations is an essential aspect of this approach. In vitro assays can provide an important first step toward this goal by selecting populations that are likely suitable for more expensive and time-consuming in vivo assays. In this study, bone marrow-derived mesenchymal stem cells (BM-MSCs) were integrated into a three-dimensional coculture system that supports the development and stabilization of vascular tube-like structures. The presence of a limited number of BM-MSCs resulted in their coalignment with vascular structures, and it further resulted in increased tubule numbers and complexity. Thus, these studies suggest that BM-MSCs functionally interacted with and were attracted to in vitro formed vascular structures. Further, these cells also provided sufficient bioactive factors and matrix molecules to support the formation of tubular arrays and the stabilization of these arrays. This in vitro system provides a means for assessing the function of BM-MSCs in aspects of the angiogenic component of wound repair.


Subject(s)
Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic , Adult , Coculture Techniques , Collagen/metabolism , Drug Combinations , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fibroblasts/cytology , Humans , Laminin/metabolism , Mesenchymal Stem Cells/metabolism , Proteoglycans/metabolism , Umbilical Veins/cytology
11.
Wound Repair Regen ; 16(2): 300-9, 2008.
Article in English | MEDLINE | ID: mdl-18318814

ABSTRACT

The superficial dermis of adult human skin contains a complex arcading microvasculature that provides nutrient support to the overlying epidermis. We propose that the unique subpopulations of dermal fibroblasts located in the superficial dermis contribute to the organization and maintenance of this elaborate microvasculature. This possibility was tested in a coculture system in which distinct subpopulations of adult human dermal fibroblasts were grown to form high-density lawns that were then seeded with human umbilical vein vascular endothelial cells (EC). The fibroblast subpopulation cultured specifically from the papillary dermis supported a robust array of highly branched tube-like structures. In contrast, fibroblasts cultured from the reticular dermis provided an anemic level of support for the formation of tube-like structures. These varied interactions with vascular EC were not due to the differential production of the potent pro-angiogenic factors vascular endothelial growth factor-A or fibroblast growth factor-2. Instead, the extracellular matrix and/or molecules bound to this matrix appeared to contain instructions that modulated these differential fibroblast-vascular EC interactions. One matrix-binding growth factor, hepatocyte growth factor/scatter factor, was identified that was both differentially expressed by papillary and reticular dermal fibroblasts and which was shown to be physiologically relevant in the coculture model. These studies highlight the importance of fibroblasts in supporting and maintaining vascular integrity. Furthermore, these studies have important implications for wound repair and may help to explain how fibroblasts contribute to the etiology of nonhealing wounds.


Subject(s)
Cell Communication , Dermis/cytology , Endothelial Cells/physiology , Extracellular Matrix/physiology , Fibroblasts/physiology , Adult , Angiogenesis Inducing Agents/pharmacology , Coculture Techniques , Fibroblast Growth Factor 2/pharmacology , Hepatocyte Growth Factor/pharmacology , Humans , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/pharmacology , Wound Healing/physiology
12.
Cells Tissues Organs ; 186(3): 157-68, 2007.
Article in English | MEDLINE | ID: mdl-17657137

ABSTRACT

The construction of vascularized connective tissues is an important goal in tissue engineering in that the presence of a patent bio-engineered vasculature should facilitate vascularization of an implant. Fibroblasts play an essential role in the angiogenic process through their production of extracellular matrix molecules and through their release of essential growth factors. Therefore, the aim of this study is to develop a thin 3-dimensional model in which fibroblasts support endothelial cells in the formation of tube-like structures. Macro- and microvascular endothelial cells were seeded onto confluent lawns of human fibroblasts and were cultured in the presence of high levels of ascorbate 2-phosphate to create a tissue-like structure in which endothelial cell organized into tube-like structures. The process was visualized in the culture dish through labeling of cells with a long-lasting fluorescent vital dye. Intact sheet-like structures were created in which endothelial cell tube-like structures were encased by fibroblasts and were surrounded by a basement membrane. These structures appeared to contain a lumen and remained stable for up to 5 weeks in culture. This culture system provides an in vitro method to study fibroblast-endothelial cell interactions and to study the effects of pro- and anti-angiogenic factors on endothelial cell differentiation. This system also provides an experimental basis for developing vascularized tissue-engineered connective tissue.


Subject(s)
Endothelial Cells/physiology , Endothelium, Vascular/physiology , Fibroblast Growth Factors/physiology , Fibroblasts/physiology , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A/physiology , Basement Membrane/ultrastructure , Capillaries/cytology , Capillaries/ultrastructure , Cell Differentiation/physiology , Cells, Cultured , Coculture Techniques/methods , Dermis/cytology , Extracellular Matrix/physiology , Humans , Tissue Engineering , Umbilical Veins/cytology
13.
Cell Tissue Res ; 327(3): 499-510, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17093925

ABSTRACT

The dermis of adult human skin contains a physiologically heterogeneous population of fibroblasts that interact to produce its unique architecture and that participate in inflammatory and wound repair functions in vivo. This heterogeneity has been well documented for fibroblasts located in the superficial papillary dermis and the deep reticular dermis. However, the existence of diverse fibroblast subpopulations within a given region of the dermis has not been explored. In this study, fibroblast cultures have been established from the superficial dermis following enzymatic dissociation of the tissue. These fibroblasts have been cloned by limiting dilution and initially selected on the basis of morphology and proliferation kinetics. Fibroblasts in some of the clones selected for study express alpha-smooth muscle actin, a myofibroblast characteristic. Significant differences for fibroblast clones obtained from the same piece of skin have been observed with regard to their rate of collagen lattice contraction, their ability to organize a fibronectin matrix, their release of specific growth factors/cytokines into culture medium, and their response to interleukin-1alpha. These differences in both morphological and physiological characteristics indicate that the superficial papillary dermis contains a heterogeneous population of fibroblasts. This heterogeneity might indicate that diverse subpopulations of fibroblasts are required to interact in both homeostatic and pathological situations in skin.


Subject(s)
Dermis/cytology , Fibroblasts/cytology , Actins/metabolism , Adult , Biomarkers/metabolism , Cell Culture Techniques , Cell Proliferation , Clone Cells , Dermis/metabolism , Fibroblast Growth Factor 7/metabolism , Fibroblasts/metabolism , Fibronectins/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-6/metabolism , Male
14.
Occup Med (Lond) ; 55(5): 345-8, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15930044

ABSTRACT

BACKGROUND: Advising on ill-health retirement is an important role of most practising occupational physicians. In recent years, the eligibility criteria and process for gaining early retirement benefits have changed in many pension schemes in the UK. AIM: To investigate the variation in rates of retirement due to ill-health in National Health Service (NHS) Trusts and Local Authorities and to update previously published guidance on ill-health retirement with specific reference to pension schemes with eligibility criteria that include permanence of incapacity due to ill-health. METHODS: Rates of retirement were calculated for 222 NHS Trusts and 132 Local Authorities with more than 1500 employees. Literature searches and consensus statements by the authors. RESULTS: Rates of retirement were widely distributed in the NHS Trusts and Local Authorities. The median rates of retirement were 2.11 (IQR 1.37-2.91)/1000 active members and 4.10 (IQR 3.01-6.10)/1000 employees, respectively (P<0.001). Difficulties in the doctor-patient relationship and in ascertaining the true functional ability of some patients were identified. CONCLUSION: There continues to be marked variation in rates of early retirement due to ill-health within and between organizations that warrants further investigation. The general and specific guidance that appears as an appendix in Supplementary data to this paper should help occupational physicians to make equitable recommendations when assessing applications for early retirement benefits and fitness to work.


Subject(s)
Disability Evaluation , Eligibility Determination , Occupational Medicine/standards , Practice Guidelines as Topic/standards , Retirement/statistics & numerical data , Health Personnel , Humans , Pensions , State Medicine
15.
J Cell Physiol ; 200(1): 134-45, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15137066

ABSTRACT

The interfollicular dermis of adult human skin is partitioned into histologically and physiologically distinct papillary and reticular zones. Each of these zones contains a unique population of fibroblasts that differ in respect to their proliferation kinetics, rates at which they contract type I collagen gels, and in their relative production of decorin and versican. Here, site-matched papillary and reticular dermal fibroblasts couples were compared to determine whether each population interacted with keratinocytes in an equivalent or different manner. Papillary and reticular fibroblasts grown in monolayer culture differed significantly from each other in their release of keratinocyte growth factor (KGF) and granulocyte-macrophage colony stimulating factor (GM-CSF) into culture medium. Some matched fibroblast couples also differed in their constitutive release of interleukin-6 (IL-6). Papillary fibroblasts produced a higher ratio of GM-CSF to KGF than did corresponding reticular fibroblasts. Interactions between site-matched papillary and reticular couples were also assayed in a three-dimensional culture system where fibroblasts and keratinocytes were randomly mixed, incorporated into type I collagen gels, and allowed to sort. Keratinocytes formed distinctive cellular masses in which the keratinocytes were organized such that the exterior most layer of cells exhibited characteristics of basal keratinocytes and the interior most cells exhibited characteristics of terminally differentiated keratinocytes. In the presence of papillary dermal fibroblasts, keratinocyte masses were highly symmetrical and cells expressed all levels of differentiation markers. In contrast, keratinocyte masses that formed in the presence of reticular fibroblasts tended to have irregular shapes, and terminal differentiation was suppressed. Furthermore, basement membrane formation was retarded in the presence of reticular cells. These studies indicate that site-matched papillary and reticular dermal fibroblasts qualitatively differ in their support of epidermal cells, with papillary cells interacting more effectively than corresponding reticular cells.


Subject(s)
Cytokines/metabolism , Fibroblasts/metabolism , Growth Substances/metabolism , Keratinocytes/metabolism , Skin/cytology , Adult , Aged , Biomarkers , Black People , Cells, Cultured , Coculture Techniques , Collagen Type I/metabolism , Culture Media , Female , Fibroblast Growth Factor 7 , Fibroblast Growth Factors/metabolism , Fibroblasts/cytology , Gels , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-6/metabolism , Keratinocytes/cytology , Male , Middle Aged , Organ Culture Techniques , White People
16.
J Cell Sci ; 117(Pt 5): 667-75, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14754903

ABSTRACT

Dermal fibroblasts are a dynamic and diverse population of cells whose functions in skin in many respects remain unknown. Normal adult human skin contains at least three distinct subpopulations of fibroblasts, which occupy unique niches in the dermis. Fibroblasts from each of these niches exhibit distinctive differences when cultured separately. Specific differences in fibroblast physiology are evident in papillary dermal fibroblasts, which reside in the superficial dermis, and reticular fibroblasts, which reside in the deep dermis. Both of these subpopulations of fibroblasts differ from the fibroblasts that are associated with hair follicles. Fibroblasts engage in fibroblast-epidermal interactions during hair development and in interfollicular regions of skin. They also play an important role in cutaneous wound repair and an ever-increasing role in bioengineering of skin. Bioengineered skin currently performs important roles in providing (1) a basic understanding of skin biology, (2) a vehicle for testing topically applied products and (3) a resource for skin replacement.


Subject(s)
Dermis/cytology , Fibroblasts/cytology , Fibroblasts/physiology , Basement Membrane/cytology , Basement Membrane/physiology , Dermis/physiology , Extracellular Matrix/physiology , Hair Follicle/cytology , Hair Follicle/physiology , Humans , Tissue Engineering
17.
Arch Dermatol Res ; 295(4): 160-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12861419

ABSTRACT

Cultured human dermal fibroblasts coexpress two cell surface ectopeptidases, aminopeptidase N (APN/CD13) and dipeptidyl peptidase IV (DPPIV/CD26). These enzymes catalyze the removal of a single amino acid or a dipeptide from the N-termini of oligopeptides, respectively. They are also localized in a differential pattern in normal, non-sun-exposed, adult skin, a finding that supports the supposition that these enzymes might have different functions in the skin, but relatively little is known about their functions in the skin. A better understanding of how the activities of these enzymes are regulated should increase our understanding of their functions in the skin. APN/CD13 was routinely expressed at higher levels on cultured fibroblasts than was DPPIV/CD26. Treatment of cultured fibroblasts with specific factors differentially modulated the activities of these enzymes. APN/CD13 was significantly upregulated by treatment with interleukin-4 (IL-4), interferon gamma (IFNgamma), and the glucocorticoids dexamethasone and hydrocortisone. In contrast, the regulation of DPPIV/CD26 activity was found to be different and more complex. This enzyme was consistently upregulated by IL-1alpha and IL-1beta, but consistently downregulated by glucocorticoids, tumor necrosis factor alpha (TNFalpha) and transforming growth factor beta(1) (TGFbeta(1)). Thus, although these two enzymes are expressed on the same populations of cultured cells, their activities are differentially regulated. This finding, along with their differential distribution in normal skin, suggests that APN/CD13 and DPPIV/CD26 have different functions in the skin.


Subject(s)
CD13 Antigens/metabolism , Cytokines/physiology , Dipeptidyl Peptidase 4/metabolism , Fibroblasts/enzymology , Glucocorticoids/physiology , Interleukins/physiology , Cells, Cultured , Cytokines/pharmacology , Dexamethasone/pharmacology , Down-Regulation , Glucocorticoids/pharmacology , Humans , Hydrocortisone/pharmacology , Interleukin-1/physiology , Interleukin-4/physiology , Interleukins/pharmacology , Skin/cytology , Transforming Growth Factor beta/physiology , Tumor Necrosis Factor-alpha/physiology , Up-Regulation
18.
Exp Dermatol ; 12(3): 315-23, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12823446

ABSTRACT

Epithelial-mesenchymal interactions play a critical role in skin development and differentiation, and similar interactions may also regulate the day-to-day proliferation and differentiation events of the epidermis that occur in normal adult skin. This study was directed at identifying molecules that are selectively located at the dermal-epidermal junction in normal adult skin as they may be involved in regulating these homeostatic events. To this end, monoclonal antibodies were raised against the crude cell membrane fraction of cultured human dermal fibroblasts. Screening of antibodies that recognized cell surface antigen on cultured human dermal fibroblasts was followed by determining which of these antibodies selectively localized cells at sites of epithelial-mesenchymal interactions. Antibody DF-5 fit these criteria and was further characterized. This antibody was found to recognize the cell surface ectopeptidase aminopeptidase N (APN), a molecule homologous to the cluster differentiation antigen CD13. Antibody DF-5 and anti-CD13 antibodies both identified cells at sites of epithelial-mesenchymal interactions in fetal, neonatal, and adult human skin, and the APN/CD13 enzyme activity was also identified at these sites. A second ectopeptidase, dipeptidyl peptidase IV (DPPIV) or CD26, presented a significantly different immunohistochemical and histochemical pattern in skin samples, confirming the specificity of the APN/CD13 studies. The function of APN/CD13 in skin has yet to be determined. Its invariant localization at sites of epithelial-mesenchymal interactions argues for a role particular to this region. It may play a role in regulating the activity of neuropeptides or other signaling peptides that are released in this region of skin or it may have an as yet undefined role in mediating communication between dermal and epidermal cells.


Subject(s)
Antibodies, Monoclonal/immunology , Dermis/cytology , Epithelial Cells/immunology , Adolescent , Adult , Biomarkers , CD13 Antigens/immunology , CD13 Antigens/metabolism , Cell Communication , Dipeptidyl Peptidase 4/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fetus , Fibroblasts/cytology , Humans , Immunohistochemistry , Mesoderm/cytology
19.
J Biol Chem ; 278(19): 17566-72, 2003 May 09.
Article in English | MEDLINE | ID: mdl-12621051

ABSTRACT

Dramatic changes occur in skin as a function of age, including changes in morphology, physiology, and mechanical properties. Changes in extracellular matrix molecules also occur, and these changes likely contribute to the overall age-related changes in the physical properties of skin. The major proteoglycans detected in extracts of human skin are decorin and versican. In addition, adult human skin contains a truncated form of decorin, whereas fetal skin contains virtually undetectable levels of this truncated decorin. Analysis of this molecule, herein referred to as decorunt, indicates that it is a catabolic fragment of decorin rather than a splice variant. With antibody probes to the core protein, decorunt is found to lack the carboxyl-terminal portion of decorin. Further analysis by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry shows that the carboxyl terminus of decorunt is at Phe(170) of decorin. This result indicates that decorunt represents the amino-terminal 43% of the mature decorin molecule. Such a structure is inconsistent with alternative splicing of decorin and suggests that decorunt is a catabolic fragment of decorin. A neoepitope antiserum, anti-VRKVTF, was generated against the carboxyl terminus of decorunt. This antiserum does not recognize intact decorin in any skin proteoglycan sample tested on immunoblots but recognizes every sample of decorunt tested. The results with anti-VRKVTF confirm the identification of the carboxyl terminus of decorunt. Analysis of collagen binding by surface plasmon resonance indicates that the affinity of decorunt for type I collagen is 100-fold less than that of decorin. This observation correlates with the structural analysis of decorunt, in that it lacks regions of decorin previously shown to be important for interaction with type I collagen. The detection of a catabolic fragment of decorin suggests the existence of a specific catabolic pathway for this proteoglycan. Because of the capacity of decorin to influence collagen fibrillogenesis, catabolism of decorin may have important functional implications with respect to the dermal collagen network.


Subject(s)
Aging/metabolism , Proteoglycans/metabolism , Skin/metabolism , Adult , Aged , Decorin , Extracellular Matrix Proteins , Humans , Immunohistochemistry , Middle Aged , Peptide Fragments , Proteoglycans/physiology , Skin Physiological Phenomena
20.
J Heart Valve Dis ; 10(4): 495-504, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11499597

ABSTRACT

BACKGROUND AND AIM OF THE STUDY: The use of flexible structures within cardiovascular prostheses such as valves, stents and vascular grafts has been proposed as a means of more closely modeling native mechanics, and thereby reducing the biomechanical problems associated with rigid materials. However, the design of such materials has been hampered by the paucity of quantitative information on the in-vivo behavior of such structures. The aim of this study was to explore the use of 3D ultrasound imaging coupled with finite element analysis (FEA) as a tool to estimate deformation and forces imposed in vivo on a novel bioprosthetic valve design. METHODS: The method was first tested using in-vitro static loading conditions, where good agreement between displacements seen on video and those obtained from application of the identical force within the finite element program was seen. The method was then tested in a porcine model with valves implanted in the mitral position. Images of the deforming annular ring were obtained over the cardiac cycle using 3D intravascular ultrasound; these images were fed into the FEA program for calculation of reaction forces. RESULTS: Results in vitro showed that a force of 2.7-8.0 Newtons (N) was required to produce a deformation of between 1.0 and 3.0 mm in the radial direction. A time history of deformation and force around the ring of the valve stent could be obtained for the in-vivo conditions. These results revealed a maximum deformation of 0.5-1.7 mm along the short axis (anteroposterior) of the mitral valve. Coupled to this, a peak reaction force of 4.4-13.9 N was found at the points corresponding to maximal deflection. Both deformation and reaction force reached maximum during atrial contraction. CONCLUSION: This method provides an accurate means of estimating deformation and corresponding forces imposed in vivo on intracardiac prostheses. The results provide information on the dynamic behavior of the mitral valve annulus. Such information should be useful in the design of flexible cardiovascular prostheses.


Subject(s)
Bioprosthesis , Echocardiography, Three-Dimensional , Finite Element Analysis , Heart Valve Prosthesis , Mitral Valve/diagnostic imaging , Ultrasonography, Interventional , Animals , Biomechanical Phenomena , Humans , In Vitro Techniques , Mitral Valve/transplantation , Research Design , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...