Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Bioengineering (Basel) ; 3(4)2016 Nov 16.
Article in English | MEDLINE | ID: mdl-28952591

ABSTRACT

Cellular spheroids were studied to determine their use as "bioinks" in the biofabrication of tissue engineered constructs. Specifically, magnetic forces were used to mediate the cyclic longitudinal stretching of tissues composed of Janus magnetic cellular spheroids (JMCSs), as part of a post-processing method for enhancing the deposition and mechanical properties of an extracellular matrix (ECM). The purpose was to accelerate the conventional tissue maturation process via novel post-processing techniques that accelerate the functional, structural, and mechanical mimicking of native tissues. The results of a forty-day study of JMCSs indicated an expression of collagen I, collagen IV, elastin, and fibronectin, which are important vascular ECM proteins. Most notably, the subsequent exposure of fused tissue sheets composed of JMCSs to magnetic forces did not hinder the production of these key proteins. Quantitative results demonstrate that cyclic longitudinal stretching of the tissue sheets mediated by these magnetic forces increased the Young's modulus and induced collagen fiber alignment over a seven day period, when compared to statically conditioned controls. Specifically, the elastin and collagen content of these dynamically-conditioned sheets were 35- and three-fold greater, respectively, at seven days compared to the statically-conditioned controls at three days. These findings indicate the potential of using magnetic forces in tissue maturation, specifically through the cyclic longitudinal stretching of tissues.

2.
World J Biol Chem ; 6(3): 65-70, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26322165

ABSTRACT

Steadily increasing evidence supports the idea that genetic diversities in the vascular bed are, in addition to hemodynamic influences, a major contributing factor in determining region-specific cardiovascular disease susceptibility. Members of the phylogenetically highly conserved Hox gene family of developmental regulators have to be viewed as prime candidates for determining these regional genetic differences in the vasculature. During embryonic patterning, the regionally distinct and precisely choreographed expression patterns of HOX transcription factors are essential for the correct specification of positional identities. Apparently, these topographic patterns are to some degree retained in certain adult tissues, including the circulatory system. While an understanding of the functional significance of these localized Hox activities in adult blood vessels is only beginning to emerge, an argument can be made for a role of Hox genes in the maintenance of vessel wall homeostasis and functional integrity on the one hand, and in regulating the development and progression of regionally restricted vascular pathologies, on the other. Initial functional studies in animal models, as well as data from clinical studies provide some level of support for this view. The data suggest that putative genetic regulatory networks of Hox-dependent cardiovascular disease processes include genes of diverse functional categories (extracellular matrix remodeling, transmembrane signaling, cell cycle control, inflammatory response, transcriptional control, etc.), as potential targets in both vascular smooth muscle and endothelial cells, as well as cell populations residing in the adventitia.

3.
Acta Biomater ; 10(10): 4323-31, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24998183

ABSTRACT

Recent advances in three-dimensional (3-D) printing offer an excellent opportunity to address critical challenges faced by current tissue engineering approaches. Alginate hydrogels have been used extensively as bioinks for 3-D bioprinting. However, most previous research has focused on native alginates with limited degradation. The application of oxidized alginates with controlled degradation in bioprinting has not been explored. Here, a collection of 30 different alginate hydrogels with varied oxidation percentages and concentrations was prepared to develop a bioink platform that can be applied to a multitude of tissue engineering applications. The authors systematically investigated the effects of two key material properties (i.e. viscosity and density) of alginate solutions on their printabilities to identify a suitable range of material properties of alginates to be applied to bioprinting. Further, four alginate solutions with varied biodegradability were printed with human adipose-derived stem cells (hADSCs) into lattice-structured, cell-laden hydrogels with high accuracy. Notably, these alginate-based bioinks were shown to be capable of modulating proliferation and spreading of hADSCs without affecting the structure integrity of the lattice structures (except the highly degradable one) after 8days in culture. This research lays a foundation for the development of alginate-based bioink for tissue-specific tissue engineering applications.


Subject(s)
Adipose Tissue/metabolism , Alginates/chemistry , Cell Proliferation , Ink , Printing, Three-Dimensional , Stem Cells/metabolism , Tissue Engineering/methods , Adipose Tissue/cytology , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Hydrogels , Materials Testing/methods , Stem Cells/cytology
4.
Front Pharmacol ; 5: 141, 2014.
Article in English | MEDLINE | ID: mdl-24999331

ABSTRACT

Fibrocytes are bone marrow hematopoietic-derived cells that also express a mesenchymal cell marker (commonly collagen I) and participate in fibrotic diseases of multiple organs. Given their origin, they or their precursors must be circulating cells before recruitment into target tissues. While most previous studies focused on circulating fibrocytes, here we focus on the fibrocyte phenotype in fibrotic tissue. The study's relevance to human disease is heightened by use of a model in which bleomycin is delivered systemically, recapitulating several features of human scleroderma including multi-organ fibrosis not observed when bleomycin is delivered directly into the lungs. Using flow cytometry, we find in the fibrotic lung a large population of CD45(high) fibrocytes (called Region I) rarely found in vehicle-treated control mice. A second population of CD45+ fibrocytes (called Region II) is observed in both control and fibrotic lung. The level of CD45 in circulating fibrocytes is far lower than in either Region I or II lung fibrocytes. The chemokine receptors CXCR4 and CCR5 are expressed at higher levels in Region I than in Region II and are present at very low levels in all other lung cells including CD45+/collagen I- leucocytes. The collagen chaperone HSP47 is present at similar high levels in both Regions I and II, but at a higher level in fibrotic lung than in control lung. There is also a major population of HSP47(high)/CD45- cells in fibrotic lung not present in control lung. CD44 is present at higher levels in Region I than in Region II and at much lower levels in all other cells including CD45+/collagen I- leucocytes. When lung fibrosis is inhibited by restoring caveolin-1 activity using a caveolin-1 scaffolding domain peptide (CSD), a strong correlation is observed between fibrocyte number and fibrosis score. In summary, the distinctive phenotype of fibrotic lung fibrocytes suggests that fibrocyte differentiation occurs primarily within the target organ.

5.
Front Pharmacol ; 5: 140, 2014.
Article in English | MEDLINE | ID: mdl-24966836

ABSTRACT

In fibrotic diseases caveolin-1 underexpression in fibroblasts results in collagen overexpression and in monocytes leads to hypermigration. These profibrotic behaviors are blocked by the caveolin-1 scaffolding domain peptide (CSD) which compensates for caveolin-1 deficiency. Monocytes and fibroblasts are related in that monocytes are the progenitors of fibrocytes (CD45+/Collagen I+ cells) that, in turn, are the progenitors of many fibroblasts in fibrotic tissues. In an additional anti-fibrotic activity, CSD blocks monocyte differentiation into fibrocytes. We studied a mouse fibrosis model (Pump Model) involving systemic bleomycin delivery that closely models scleroderma (SSc) in several ways, the most important of which for this study is that fibrosis is observed in the lungs, skin, and internal organs. We show here that dermal thickness is increased 2-fold in the Pump Model and that this effect is almost completely blocked by CSD (p < 0.001). Concomitantly, the subcutaneous fat layer becomes >80% thinner. This effect is also blocked by CSD (p < 0.001). Even in mice receiving vehicle instead of bleomycin, CSD increases the thickness of the fat layer. To study the mechanisms of action of bleomycin and CSD, we examined the accumulation of the chemokine receptor CCR5 and its ligands MIP1α and MIP1ß in fibrotic tissue and their roles in monocyte migration. Fibrocytes and other leukocytes expressing CCR5 and its ligands were present at high levels in the fibrotic dermis of SSc patients and Pump Model mice while CSD blocked their accumulation in mouse dermis. Migration toward CCR5 ligands of SSc monocytes and Pump Model bone marrow cells was 3-fold greater than cells from control subjects. This enhanced migration was almost completely blocked by CSD. These results suggest that low monocyte caveolin-1 promotes fibrosis by enhancing the recruitment of fibrocytes and their progenitors into affected tissue.

6.
Biofabrication ; 6(2): 024111, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24717646

ABSTRACT

Tissue spheroids hold great potential in tissue engineering as building blocks to assemble into functional tissues. To date, agarose molds have been extensively used to facilitate fusion process of tissue spheroids. As a molding material, agarose typically requires low temperature plates for gelation and/or heated dispenser units. Here, we proposed and developed an alginate-based, direct 3D mold-printing technology: 3D printing microdroplets of alginate solution into biocompatible, bio-inert alginate hydrogel molds for the fabrication of scaffold-free tissue engineering constructs. Specifically, we developed a 3D printing technology to deposit microdroplets of alginate solution on calcium containing substrates in a layer-by-layer fashion to prepare ring-shaped 3D hydrogel molds. Tissue spheroids composed of 50% endothelial cells and 50% smooth muscle cells were robotically placed into the 3D printed alginate molds using a 3D printer, and were found to rapidly fuse into toroid-shaped tissue units. Histological and immunofluorescence analysis indicated that the cells secreted collagen type I playing a critical role in promoting cell-cell adhesion, tissue formation and maturation.


Subject(s)
Bioprinting/methods , Printing, Three-Dimensional , Tissue Engineering/methods , Alginates/chemistry , Algorithms , Cell Line , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Spheroids, Cellular/cytology
7.
Am J Physiol Lung Cell Mol Physiol ; 306(8): L736-48, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24583879

ABSTRACT

The interstitial lung diseases (ILD) include a large number of chronic, progressive, irreversible respiratory disorders involving pulmonary fibrosis, the most common of which are idiopathic pulmonary fibrosis and scleroderma lung disease (SSc ILD). Because bleomycin causes lung fibrosis when used in cancer chemotherapy, it is used to model human ILD in rodents. In most studies, bleomycin has been delivered directly into the lung by intratracheal or intraoral administration. Here we have compared the effects in mice of bleomycin delivered directly into the lungs (direct model) or systemically using osmotic minipumps (pump model) to determine which more closely resembles human ILD. The pump model is more similar to human SSc ILD in that: 1) lung injury/fibrosis is limited to the subpleural portion of the lung in the pump model and in SSc ILD, whereas the entire lung is affected in the direct model; 2) conversely, there is massive inflammation throughout the lung in the direct model, whereas inflammation is limited in the pump model and in SSc ILD; 3) hypertrophic type II alveolar epithelial cells are present at high levels in SSc ILD and in the pump model but not in the direct model; and 4) lung fibrosis is accompanied by dermal fibrosis. The pump model is also move convenient and humane than the direct model because there is less weight loss and mortality.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Bleomycin/administration & dosage , Drug Delivery Systems , Infusion Pumps , Lung Diseases, Interstitial/drug therapy , Scleroderma, Systemic/drug therapy , Animals , Caveolin 1/metabolism , Extracellular Matrix Proteins/metabolism , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Lung Diseases, Interstitial/metabolism , Lung Diseases, Interstitial/pathology , Male , Mice , Mice, Inbred C57BL , Osmosis , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Weight Loss/drug effects
8.
Arthritis Rheumatol ; 66(7): 1909-19, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24578173

ABSTRACT

OBJECTIVE: Interstitial lung disease (ILD) is the leading cause of death in patients with systemic sclerosis (SSc; scleroderma). Although SSc-related ILD is more common and severe in African Americans than in Caucasians, little is known about factors underlying this significant health disparity. The aim of this study was to examine the role that low expression of caveolin-1 might play in susceptibility to ILD among African Americans. METHODS: Assays of monocyte migration toward stromal cell-derived factor 1 (SDF-1) were performed using monocytes from Caucasian and African American healthy donors and patients with SSc. For fibrocyte differentiation studies, total peripheral blood mononuclear cells were incubated on fibronectin-coated plates. Protein expression was evaluated by immunohistochemistry and Western blotting. RESULTS: Monocytes from healthy African American donors and those from patients with SSc had low caveolin-1 levels, enhanced migration toward the CXCR4 ligand SDF-1, and enhanced differentiation to fibrocytes. Enhanced migration and differentiation of monocytes from African Americans and patients with SSc appeared to be attributable to the lack of caveolin-1, because restoring caveolin-1 function using a caveolin-1 scaffolding domain peptide inhibited these processes. Although they differed from monocytes from Caucasians, monocytes from both African Americans and patients with SSc were not identical, because SSc monocytes showed major increases from baseline in ERK, JNK, p38, and Smad2/3 activation, while monocytes from African Americans showed only limited ERK activation and no activation of JNK, p38, or Smad2/3. In contrast, SDF-1 exposure caused no additional ERK activation in SSc monocytes but did cause significant additional activation in monocytes from African Americans. CONCLUSION: African Americans may be predisposed to SSc-related ILD due to low baseline caveolin-1 levels in their monocytes, potentially affecting signaling, migration, and fibrocyte differentiation. The monocytes of African Americans may lack caveolin-1 due to high levels of transforming growth factor ß in their blood.


Subject(s)
Black or African American , Caveolin 1/deficiency , Lung Diseases, Interstitial/metabolism , Monocytes/cytology , Scleroderma, Systemic/metabolism , White People , Caveolin 1/metabolism , Cell Differentiation/immunology , Cell Movement/immunology , Cytoskeleton/metabolism , Fibroblasts/cytology , Humans , In Vitro Techniques , Lung Diseases, Interstitial/ethnology , Lung Diseases, Interstitial/immunology , MAP Kinase Signaling System/immunology , Monocytes/immunology , Receptors, CXCR4/metabolism , Risk Factors , Scleroderma, Systemic/ethnology , Scleroderma, Systemic/immunology , Transforming Growth Factor beta/metabolism
9.
Biomaterials ; 35(3): 949-60, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24183699

ABSTRACT

Cell aggregates, or spheroids, have been used as building blocks to fabricate scaffold-free tissues that can closely mimic the native three-dimensional in vivo environment for broad applications including regenerative medicine and high throughput testing of drugs. The incorporation of magnetic nanoparticles (MNPs) into spheroids permits the manipulation of spheroids into desired shapes, patterns, and tissues using magnetic forces. Current strategies incorporating MNPs often involve cellular uptake, and should therefore be avoided because it induces adverse effects on cell activity, viability, and phenotype. Here, we report a Janus structure of magnetic cellular spheroids (JMCS) with spatial control of MNPs to form two distinct domains: cells and extracellular MNPs. This separation of cells and MNPs within magnetic cellular spheroids was successfully incorporated into cellular spheroids with various cellular and extracellular compositions and contents. The amount of cells that internalized MNPs was quantified and showed that JMCSs resulted in significantly lower internalization (35%) compared to uptake spheroids (83%, p < 0.05). Furthermore, the addition of MNPs to cellular spheroids using the Janus method has no adverse effects on cellular viability up to seven weeks, with spheroids maintaining at least 82% viability over 7 weeks when compared to control spheroids without MNPs. By safely incorporating MNPs into cellular spheroids, results demonstrated that JMCSs were capable of magnetic manipulation, and that magnetic forces used during magnetic force assembly mediate fusion into controlled patterns and complex tissues. Finally, JMCSs were assembled and fused into a vascular tissue construct 5 mm in diameter using magnetic force assembly.


Subject(s)
Aorta/cytology , Magnetite Nanoparticles/chemistry , Myocytes, Smooth Muscle/cytology , Spheroids, Cellular/cytology , Tissue Engineering/methods , Animals , Cell Survival , Cells, Cultured , Fibroblasts/cytology , Humans , Magnetic Phenomena , Rats , Stem Cells/cytology
10.
J Biol Chem ; 289(11): 7856-72, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24324260

ABSTRACT

The hepatocyte growth factor (HGF) and the HGF receptor Met pathway are important in the pathogenesis of interstitial lung disease (ILD). Alternatively spliced isoforms of CD44 containing variable exon 6 (CD44v6) and its ligand hyaluronan (HA) alter cellular function in response to interaction between CD44v6 and HGF. TGF-ß1 is the crucial cytokine that induces fibrotic action in ILD fibroblasts (ILDFbs). We have identified an autocrine TGF-ß1 signaling that up-regulates both Met and CD44v6 mRNA and protein expression. Western blot analysis, flow cytometry, and immunostaining revealed that CD44v6 and Met colocalize in fibroblasts and in tissue sections from ILD patients and in lungs of bleomycin-treated mice. Interestingly, cell proliferation induced by TGF-ß1 is mediated through Met and CD44v6. Further, cell proliferation mediated by TGF-ß1/CD44v6 is ERK-dependent. In contrast, action of Met on ILDFb proliferation does not require ERK but does require p38(MAPK). ILDFbs were sorted into CD44v6(+)/Met(+) and CD44v6(-)/Met(+) subpopulations. HGF inhibited TGF-ß1-stimulated collagen-1 and α-smooth muscle cell actin expression in both of these subpopulations by interfering with TGF-ß1 signaling. HGF alone markedly stimulated CD44v6 expression, which in turn regulated collagen-1 synthesis. Our data with primary lung fibroblast cultures with respect to collagen-1, CD44v6, and Met expressions were supported by immunostaining of lung sections from bleomycin-treated mice and from ILD patients. These results define the relationships between CD44v6, Met, and autocrine TGF-ß1 signaling and the potential modulating influence of HGF on TGF-ß1-induced CD44v6-dependent fibroblast function in ILD fibrosis.


Subject(s)
Hyaluronan Receptors/metabolism , Lung Diseases, Interstitial/metabolism , Proto-Oncogene Proteins c-met/metabolism , Pulmonary Fibrosis/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism , Animals , Autocrine Communication , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Culture Media/chemistry , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/metabolism , Flow Cytometry , Gene Expression Regulation , Hepatocyte Growth Factor/metabolism , Humans , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/pathology , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction
11.
Acta Biomater ; 10(2): 623-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24176725

ABSTRACT

Magnetic nanoparticles (MNPs), primarily iron oxide nanoparticles, have been incorporated into cellular spheroids to allow for magnetic manipulation into desired shapes, patterns and 3-D tissue constructs using magnetic forces. However, the direct and long-term interaction of iron oxide nanoparticles with cells and biological systems can induce adverse effects on cell viability, phenotype and function, and remain a critical concern. Here we report the preparation of biological magnetic cellular spheroids containing magnetoferritin, a biological MNP, capable of serving as a biological alternative to iron oxide magnetic cellular spheroids as tissue engineered building blocks. Magnetoferritin NPs were incorporated into 3-D cellular spheroids with no adverse effects on cell viability up to 1 week. Additionally, cellular spheroids containing magnetoferritin NPs were magnetically patterned and fused into a tissue ring to demonstrate its potential for tissue engineering applications. These results present a biological approach that can serve as an alternative to the commonly used iron oxide magnetic cellular spheroids, which often require complex surface modifications of iron oxide NPs to reduce the adverse effects on cells.


Subject(s)
Magnetic Phenomena , Spheroids, Cellular/cytology , Tissue Engineering/methods , Animals , Apoferritins/chemical synthesis , Apoferritins/pharmacology , Apoferritins/ultrastructure , Cattle , Cell Survival/drug effects , Horses , Iron/pharmacology , Magnetite Nanoparticles/ultrastructure , Oxides/chemical synthesis , Oxides/pharmacology , Rats , Spheroids, Cellular/drug effects
12.
Respir Res ; 14: 90, 2013 Sep 08.
Article in English | MEDLINE | ID: mdl-24011378

ABSTRACT

BACKGROUND: In fibrotic lung diseases, expression of caveolin-1 is decreased in fibroblasts and monocytes. The effects of this deficiency are reversed by treating cells or animals with the caveolin-1 scaffolding domain peptide (CSD, amino acids 82-101 of caveolin-1) which compensates for the lack of caveolin-1. Here we compare the function of CSD subdomains (Cav-A, Cav-B, Cav-C, Cav-AB, and Cav-BC) and mutated versions of CSD (F92A and T90A/T91A/F92A). METHODS: Migration toward the chemokine CXCL12 and the associated expression of F-actin, CXCR4, and pSmad 2/3 were studied in monocytes from healthy donors and SSc patients. Fibrocyte differentiation was studied using PBMC from healthy donors and SSc patients. Collagen I secretion and signaling were studied in fibroblasts derived from the lung tissue of healthy subjects and SSc patients. RESULTS: Cav-BC and CSD at concentrations as low as 0.01 µM inhibited the hypermigration of SSc monocytes and TGFß-activated Normal monocytes and the differentiation into fibrocytes of SSc and Normal monocytes. While CSD also inhibited the migration of poorly migrating Normal monocytes, Cav-A (and other subdomains to a lesser extent) promoted the migration of Normal monocytes while inhibiting the hypermigration of TGFß-activated Normal monocytes. The effects of versions of CSD on migration may be mediated in part via their effects on CXCR4, F-actin, and pSmad 2/3 expression. Cav-BC was as effective as CSD in inhibiting fibroblast collagen I and ASMA expression and MEK/ERK signaling. Cav-C and Cav-AB also inhibited collagen I expression, but in many cases did not affect ASMA or MEK/ERK. Cav-A increased collagen I expression in scleroderma lung fibroblasts. Full effects on fibroblasts of versions of CSD required 5 µM peptide. CONCLUSIONS: Cav-BC retains most of the anti-fibrotic functions of CSD; Cav-A exhibits certain pro-fibrotic functions. Results obtained with subdomains and mutated versions of CSD further suggest that the critical functional residues in CSD depend on the cell type and readout being studied. Monocytes may be more sensitive to versions of CSD than fibroblasts and endothelial cells because the baseline level of caveolin-1 in monocytes is much lower than in these other cell types.


Subject(s)
Caveolin 1/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Lung/drug effects , Lung/metabolism , Scleroderma, Systemic/metabolism , Actins/metabolism , Adolescent , Adult , Aged , Case-Control Studies , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Female , Fibroblasts/pathology , Humans , Lung/pathology , Male , Matrix Attachment Regions , Middle Aged , Monocytes/drug effects , Monocytes/metabolism , Monocytes/pathology , Protein Structure, Tertiary , Receptors, CXCR4/metabolism , Scleroderma, Systemic/pathology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Young Adult
13.
Biotechnol Prog ; 29(5): 1298-306, 2013.
Article in English | MEDLINE | ID: mdl-23894049

ABSTRACT

Stem cells are needed for an increasing number of scientific applications, including both fundamental research and clinical disease treatment. To meet this rising demand, improved expansion methods to generate high quantities of high quality stem cells must be developed. Unfortunately, the bicarbonate buffering system - which relies upon an elevated CO2 environment - typically used to maintain pH in stem cell cultures introduces several unnecessary limitations in bioreactor systems. In addition to artificially high dissolved CO2 levels negatively affecting cell growth, but more importantly, the need to sparge CO2 into the system complicates the ability to control culture parameters. This control is especially important for stem cells, whose behavior and phenotype is highly sensitive to changes in culture conditions such as dissolved oxygen and pH. As a first step, this study developed a buffer to support expansion of mesenchymal stem cells (MSC) under an atmospheric CO2 environment in static cultures. MSC expanded under atmospheric CO2 with this buffer achieved equivalent growth rates without adaptation compared to those grown in standard conditions and also maintained a stem cell phenotype, self-renewal properties, and the ability to differentiate into multiple lineages after expansion.


Subject(s)
Carbon Dioxide/chemistry , Cell Culture Techniques , Mesenchymal Stem Cells/cytology , Animals , Bioreactors , Cell Differentiation , Cell Proliferation , Mice , Oxygen/chemistry
14.
Biol Open ; 1(5): 430-5, 2012 May 15.
Article in English | MEDLINE | ID: mdl-23213434

ABSTRACT

The distinct topographic Hox expression patterns observed in vascular smooth muscle cells (VSMCs) of the adult cardiovascular system suggest that these transcriptional regulators are critical for maintaining region-specific physiological properties of blood vessels. To test this proposition, we expanded the vascular Hoxc11 expression domain normally restricted to the lower limbs by utilizing an innovative integrated tetracycline regulatory system and Transgelin promoter elements to induce Hoxc11 expression universally in VSMCs of transgenic mice. Ectopic Hoxc11 expression in carotid arteries, aortic arch and descending aorta resulted in drastic vessel wall remodeling involving elastic laminae fragmentation, medial smooth muscle cell loss, and intimal lesion formation. None of these alterations were observed upon induction of Hoxc11 transgene expression in the femoral artery, i.e. the natural Hoxc11 activity domain, although this vessel was greatly enlarged, comparable to the topographically restricted vascular changes seen in Hoxc11(-/-) mice. To begin defining Hoxc11-controlled pathways of vascular remodeling, we performed immunolabeling studies in conjunction with co-transfection and chromatin immunoprecipitation (ChIP) assays using mouse vascular smooth muscle (MOVAS) cells. The results suggest direct transcriptional control of two members of the matrix metalloproteinase (Mmp) family, including Mmp2 and Mmp9 that are known as key players in the inception and progression of vascular remodeling events. In summary, the severe vascular abnormalities resulting from the induced dysregulated expression of a Hox gene with regional vascular patterning functions suggest that proper Hox function and regulation is critical for maintaining vascular functional integrity.

15.
Differentiation ; 84(1): 103-16, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22579502

ABSTRACT

Atrioventricular valve development commences with an EMT event whereby endocardial cells transform into mesenchyme. The molecular events that induce this phenotypic change are well understood and include many growth factors, signaling components, and transcription factors. Besides their clear importance in valve development, the role of these transformed mesenchyme and the function they serve in the developing prevalve leaflets is less understood. Indeed, we know that these cells migrate, but how and why do they migrate? We also know that they undergo a transition to a mature, committed cell, largely defined as an interstitial fibroblast due to their ability to secrete various matrix components including collagen type I. However, we have yet to uncover mechanisms by which the matrix is synthesized, how it is secreted, and how it is organized. As valve disease is largely characterized by altered cell number, cell activation, and matrix disorganization, answering questions of how the valves are built will likely provide us with information of real clinical relevance. Although expression profiling and descriptive or correlative analyses are insightful, to advance the field, we must now move past the simplicity of these assays and ask fundamental, mechanistic based questions aimed at understanding how valves are "built". Herein we review current understandings of atrioventricular valve development and present what is known and what isn't known. In most cases, basic, biological questions and hypotheses that were presented decades ago on valve development still are yet to be answered but likely hold keys to uncovering new discoveries with relevance to both embryonic development and the developmental basis of adult heart valve diseases. Thus, the goal of this review is to remind us of these questions and provide new perspectives on an old theme of valve development.


Subject(s)
Heart Valves/embryology , Animals , Cell Differentiation , Cell Lineage , Cell Movement , Chick Embryo , Collagen Type I/metabolism , Endocardial Cushions/cytology , Endocardium/cytology , Endothelial Cells/cytology , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Heart Defects, Congenital/embryology , Heart Valve Diseases/embryology , Heart Valve Diseases/etiology , Humans , Mesoderm/cytology , Mice , Mitral Valve/embryology , Mitral Valve/pathology , Tricuspid Valve/embryology , Tricuspid Valve/pathology
16.
J Mol Cell Cardiol ; 51(6): 955-65, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21871458

ABSTRACT

Advances in understanding of the maintenance of the cardiac valves during normal cardiac function and response to injury have led to several novel findings, including that there is contribution of extra-cardiac cells to the major cellular population of the valve: the valve interstitial cell (VIC). While suggested to occur in human heart studies, we have been able to experimentally demonstrate, using a mouse model, that cells of bone marrow hematopoietic stem cell origin engraft into the valves and synthesize collagen type I. Based on these initial findings, we sought to further characterize this cell population in terms of its similarity to VICs and begin to elucidate its contribution to valve homeostasis. To accomplish this, chimeric mice whose bone marrow was repopulated with enhanced green fluorescent protein (EGFP) expressing total nucleated bone marrow cells were used to establish a profile of EGFP(+) valve cells in terms of their expression of hematopoietic antigens, progenitor markers, fibroblast- and myofibroblast-related molecules, as well as their distribution within the valves. Using this profile, we show that normal (non-irradiated, non-transplanted) mice have BM-derived cell populations that exhibit identical morphology and phenotype to those observed in transplanted mice. Collectively, our findings establish that the engraftment of bone marrow-derived cells occurs as part of normal valve homeostasis. Further, our efforts demonstrate that the use of myeloablative irradiation, which is commonly employed in studies involving bone marrow transplantation, does not elicit changes in the bone marrow-derived VIC phenotype in recipient mice.


Subject(s)
Bone Marrow Cells/cytology , Heart Valves/cytology , Heart Valves/metabolism , AC133 Antigen , Animals , Antigens, CD/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation , Female , Glycoproteins/metabolism , Heart Valves/radiation effects , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Homeostasis , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptides/metabolism , Phenotype , Whole-Body Irradiation
17.
Fibrogenesis Tissue Repair ; 4(1): 15, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21722364

ABSTRACT

Interstitial lung disease (ILD) is a major cause of morbidity and mortality in scleroderma (systemic sclerosis, or SSc). Fibrocytes are a monocyte-derived cell population implicated in the pathogenesis of fibrosing disorders. Given the recently recognized importance of caveolin-1 in regulating function and signaling in SSc monocytes, in the present study we examined the role of caveolin-1 in the migration and/or trafficking and phenotype of monocytes and fibrocytes in fibrotic lung disease in human patients and an animal model. These studies fill a gap in our understanding of how monocytes and fibrocytes contribute to SSc-ILD pathology. We found that C-X-C chemokine receptor type 4-positive (CXCR4+)/collagen I-positive (ColI+), CD34+/ColI+ and CD45+/ColI+ cells are present in SSc-ILD lungs, but not in control lungs, with CXCR4+ cells being most prevalent. Expression of CXCR4 and its ligand, stromal cell-derived factor 1 (CXCL12), are also highly upregulated in SSc-ILD lung tissue. SSc monocytes, which lack caveolin-1 and therefore overexpress CXCR4, exhibit almost sevenfold increased migration toward CXCL12 compared to control monocytes. Restoration of caveolin-1 function by administering the caveolin scaffolding domain (CSD) peptide reverses this hypermigration. Similarly, transforming growth factor ß-treated normal monocytes lose caveolin-1, overexpress CXCR4 and exhibit 15-fold increased monocyte migration that is CSD peptide-sensitive. SSc monocytes exhibit a different phenotype than normal monocytes, expressing high levels of ColI, CD14 and CD34. Because ColI+/CD14+ cells are prevalent in SSc blood, we looked for such cells in lung tissue and confirmed their presence in SSc-ILD lungs but not in normal lungs. Finally, in the bleomycin model of lung fibrosis, we show that CSD peptide diminishes fibrocyte accumulation in the lungs. Our results suggest that low caveolin-1 in SSc monocytes contributes to ILD via effects on cell migration and phenotype and that the hyperaccumulation of fibrocytes in SSc-ILD may result from the altered phenotype and migratory activity of their monocyte precursors.

18.
J Tissue Eng Regen Med ; 4(8): 659-64, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20603872

ABSTRACT

Organ printing or computer-aided robotic layer-by-layer additive biofabrication of thick three-dimensional (3D) living tissue constructs employing self-assembling tissue spheroids is a rapidly evolving alternative to classic solid scaffold-based approaches in tissue engineering. However, the absence of effective methods of accelerated tissue maturation immediately after bioprinting is the main technological imperative and potential impediment for further progress in the development of this emerging organ printing technology. Identification of the optimal combination of factors and conditions that accelerate tissue maturation ('maturogenic' factors) is an essential and necessary endeavour. Screening of maturogenic factors would be most efficiently accomplished using high-throughput quantitative in vitro tissue maturation assays. We have recently reviewed the formation of solid scaffold-free tissue constructs through the fusion of bioprinted tissue spheroids that have measurable material properties. We hypothesize that the fusion kinetics of these tissue spheroids will provide an efficacious in vitro assay of the level of tissue maturation. We report here the results of experimental testing of two simple quantitative tissue spheroid fusion-based in vitro high-throughput screening assays of tissue maturation: (a) a tissue spheroid envelopment assay; and (b) a tissue spheroid fusion kinetics assay.


Subject(s)
Cell Fusion/methods , Spheroids, Cellular/cytology , Tissue Engineering/methods , Animals , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/metabolism , Dermis/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Fluorescent Dyes/metabolism , Mice , Spheroids, Cellular/metabolism
19.
Expert Opin Biol Ther ; 10(3): 409-20, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20132061

ABSTRACT

IMPORTANCE OF THE FIELD: Effective vascularization of thick three-dimensional engineered tissue constructs is a problem in tissue engineering. As in native organs, a tissue-engineered intra-organ vascular tree must be comprised of a network of hierarchically branched vascular segments. Despite this requirement, current tissue-engineering efforts are still focused predominantly on engineering either large-diameter macrovessels or microvascular networks. AREAS COVERED IN THIS REVIEW: We present the emerging concept of organ printing or robotic additive biofabrication of an intra-organ branched vascular tree, based on the ability of vascular tissue spheroids to undergo self-assembly. WHAT THE READER WILL GAIN: The feasibility and challenges of this robotic biofabrication approach to intra-organ vascularization for tissue engineering based on organ-printing technology using self-assembling vascular tissue spheroids including clinically relevantly vascular cell sources are analyzed. TAKE HOME MESSAGE: It is not possible to engineer 3D thick tissue or organ constructs without effective vascularization. An effective intra-organ vascular system cannot be built by the simple connection of large-diameter vessels and microvessels. Successful engineering of functional human organs suitable for surgical implantation will require concomitant engineering of a 'built in' intra-organ branched vascular system. Organ printing enables biofabrication of human organ constructs with a 'built in' intra-organ branched vascular tree.


Subject(s)
Blood Vessels , Tissue Engineering , Animals , Feasibility Studies , Humans , Robotics
20.
Biomaterials ; 30(12): 2164-74, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19176247

ABSTRACT

Organ printing can be defined as layer-by-layer additive robotic biofabrication of three-dimensional functional living macrotissues and organ constructs using tissue spheroids as building blocks. The microtissues and tissue spheroids are living materials with certain measurable, evolving and potentially controllable composition, material and biological properties. Closely placed tissue spheroids undergo tissue fusion - a process that represents a fundamental biological and biophysical principle of developmental biology-inspired directed tissue self-assembly. It is possible to engineer small segments of an intraorgan branched vascular tree by using solid and lumenized vascular tissue spheroids. Organ printing could dramatically enhance and transform the field of tissue engineering by enabling large-scale industrial robotic biofabrication of living human organ constructs with "built-in" perfusable intraorgan branched vascular tree. Thus, organ printing is a new emerging enabling technology paradigm which represents a developmental biology-inspired alternative to classic biodegradable solid scaffold-based approaches in tissue engineering.


Subject(s)
Spheroids, Cellular , Tissue Engineering/methods , Animals , Computer Simulation , Humans , Prostheses and Implants
SELECTION OF CITATIONS
SEARCH DETAIL
...