Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 4489, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37563130

ABSTRACT

Lizards cannot naturally regenerate limbs but are the closest known relatives of mammals capable of epimorphic tail regrowth. However, the mechanisms regulating lizard blastema formation and chondrogenesis remain unclear. Here, single-cell RNA sequencing analysis of regenerating lizard tails identifies fibroblast and phagocyte populations linked to cartilage formation. Pseudotime trajectory analyses suggest spp1+-activated fibroblasts as blastema cell sources, with subsets exhibiting sulf1 expression and chondrogenic potential. Tail blastema, but not limb, fibroblasts express sulf1 and form cartilage under Hedgehog signaling regulation. Depletion of phagocytes inhibits blastema formation, but treatment with pericytic phagocyte-conditioned media rescues blastema chondrogenesis and cartilage formation in amputated limbs. The results indicate a hierarchy of phagocyte-induced fibroblast gene activations during lizard blastema formation, culminating in sulf1+ pro-chondrogenic populations singularly responsive to Hedgehog signaling. These properties distinguish lizard blastema cells from homeostatic and injury-stimulated fibroblasts and indicate potential actionable targets for inducing regeneration in other species, including humans.


Subject(s)
Hedgehog Proteins , Lizards , Humans , Animals , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Chondrogenesis , Lizards/physiology , Fibroblasts , Single-Cell Analysis , Tail/physiology , Mammals
2.
Sci Transl Med ; 15(688): eabq2395, 2023 03 22.
Article in English | MEDLINE | ID: mdl-36947594

ABSTRACT

Adult mammals are incapable of multitissue regeneration, and augmentation of this potential may shift current therapeutic paradigms. We found that a common co-receptor of interleukin 6 (IL-6) cytokines, glycoprotein 130 (gp130), serves as a major nexus integrating various context-specific signaling inputs to either promote regenerative outcomes or aggravate disease progression. Via genetic and pharmacological experiments in vitro and in vivo, we demonstrated that a signaling tyrosine 814 (Y814) within gp130 serves as a major cellular stress sensor. Mice with constitutively inactivated Y814 (F814) were resistant to surgically induced osteoarthritis as reflected by reduced loss of proteoglycans, reduced synovitis, and synovial fibrosis. The F814 mice also exhibited enhanced regenerative, not reparative, responses after wounding in the skin. In addition, pharmacological modulation of gp130 Y814 upstream of the SRC and MAPK circuit by a small molecule, R805, elicited a protective effect on tissues after injury. Topical administration of R805 on mouse skin wounds resulted in enhanced hair follicle neogenesis and dermal regeneration. Intra-articular administration of R805 to rats after medial meniscal tear and to canines after arthroscopic meniscal release markedly mitigated the appearance of osteoarthritis. Single-cell sequencing data demonstrated that genetic and pharmacological modulation of Y814 resulted in attenuation of inflammatory gene signature as visualized by the anti-inflammatory macrophage and nonpathological fibroblast subpopulations in the skin and joint tissue after injury. Together, our study characterized a molecular mechanism that, if manipulated, enhances the intrinsic regenerative capacity of tissues through suppression of a proinflammatory milieu and prevents pathological outcomes in injury and disease.


Subject(s)
Cytokines , Osteoarthritis , Mice , Rats , Animals , Dogs , Cytokine Receptor gp130 , Interleukin-6 , Proteoglycans , Mammals
3.
J Dev Biol ; 10(1)2022 Feb 10.
Article in English | MEDLINE | ID: mdl-35225965

ABSTRACT

(1) Background: Lizard tail regeneration provides a unique model of blastema-based tissue regeneration for large-scale appendage replacement in amniotes. Green anole lizard (Anolis carolinensis) blastemas contain fibroblastic connective tissue cells (FCTCs), which respond to hedgehog signaling to create cartilage in vivo. However, an in vitro model of the blastema has not previously been achieved in culture. (2) Methods: By testing two adapted tissue dissociation protocols and two optimized media formulations, lizard tail FCTCs were pelleted in vitro and grown in a micromass blastema organoid culture. Pellets were analyzed by histology and in situ hybridization for FCTC and cartilage markers alongside staged original and regenerating lizard tails. (3) Results: Using an optimized serum-free media and a trypsin- and collagenase II-based dissociation protocol, micromass blastema organoids were formed. Organoid cultures expressed FCTC marker CDH11 and produced cartilage in response to hedgehog signaling in vitro, mimicking in vivo blastema and tail regeneration. (4) Conclusions: Lizard tail blastema regeneration can be modeled in vitro using micromass organoid culture, recapitulating in vivo FCTC marker expression patterns and chondrogenic potential.

4.
Nat Commun ; 12(1): 6010, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34650077

ABSTRACT

Lizards regenerate amputated tails but fail to recapitulate the dorsoventral patterning achieved during embryonic development. Regenerated lizard tails form ependymal tubes (ETs) that, like embryonic tail neural tubes (NTs), induce cartilage differentiation in surrounding cells via sonic hedgehog (Shh) signaling. However, adult ETs lack characteristically roof plate-associated structures and express Shh throughout their circumferences, resulting in the formation of unpatterned cartilage tubes. Both NTs and ETs contain neural stem cells (NSCs), but only embryonic NSC populations differentiate into roof plate identities when protected from endogenous Hedgehog signaling. NSCs were isolated from parthenogenetic lizard embryos, rendered unresponsive to Hedgehog signaling via CRISPR/Cas9 gene knockout of smoothened (Smo), and implanted back into clonally-identical adults to regulate tail regeneration. Here we report that Smo knockout embryonic NSCs oppose cartilage formation when engrafted to adult ETs, representing an important milestone in the creation of regenerated lizard tails with dorsoventrally patterned skeletal tissues.


Subject(s)
Embryonic Stem Cells/physiology , Gene Editing , Lizards/genetics , Lizards/physiology , Neural Stem Cells/physiology , Regeneration/physiology , Tail/physiology , Animals , Body Patterning/genetics , Body Patterning/physiology , CRISPR-Cas Systems , Cartilage , Ependyma , Lizards/embryology , Signal Transduction/genetics , Smoothened Receptor/genetics , Spinal Cord/physiology
5.
Article in English | MEDLINE | ID: mdl-32337387

ABSTRACT

Lizards are the closest relatives of mammals capable of tail regeneration, but the specific determinants of amniote regenerative capabilities are currently unknown. Macrophages are phagocytic immune cells that play a critical role in wound healing and tissue regeneration in a wide range of species. We hypothesize that macrophages regulate the process of lizard tail regeneration, and that comparisons with mammalian cell populations will yield insight into the role phagocytes play in determining an organism's regenerative potential. Single cell RNA sequencing (scRNAseq) was used to profile lizard immune cells and compare with mouse counterparts to contrast cell types between the two species. Treatment with clodronate liposomes effectively inhibited lizard tail stump tissue ablation and subsequent regeneration, and scRNAseq was used to profile changes in lizard immune cell populations resulting from tail amputation as well as identifying specific cell types affected by clodronate treatment. ScRNAseq analysis of lizard bone marrow, peripheral blood, and tissue-resident phagocyte cell populations was used to trace marker progression during macrophage differentiation and activation. These results indicated that lizard macrophages are recruited to tail amputation injuries faster than mouse populations and express high levels of matrix metalloproteinases (MMPs). In turn, treatment with MMP inhibitors inhibited lizard tail regeneration. These results provide single cell sequencing data sets for evaluating and comparing lizard and mammalian immune cell populations, and identifying macrophage populations that are critical regulators of lizard tail regrowth.

6.
Stem Cell Res Ther ; 10(1): 254, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31412905

ABSTRACT

BACKGROUND: Stem cell-based bone tissue engineering shows promise for bone repair but faces some challenges, such as insufficient osteogenesis and limited architecture flexibility of the cell-delivery scaffold. METHODS: In this study, we first used lentiviral constructs to transduce ex vivo human bone marrow-derived stem cells with human bone morphogenetic protein-2 (BMP-2) gene (BMP-hBMSCs). We then introduced these cells into a hydrogel scaffold using an advanced visible light-based projection stereolithography (VL-PSL) technology, which is compatible with concomitant cell encapsulation and amenable to computer-aided architectural design, to fabricate scaffolds fitting local physical and structural variations in different bones and defects. RESULTS: The results showed that the BMP-hBMSCs encapsulated within the scaffolds had high viability with sustained BMP-2 gene expression and differentiated toward an osteogenic lineage without the supplement of additional BMP-2 protein. In vivo bone formation efficacy was further assessed using an intramuscular implantation model in severe combined immunodeficiency (SCID) mice. Microcomputed tomography (micro-CT) imaging indicated rapid bone formation by the BMP-hBMSC-laden constructs as early as 14 days post-implantation. Histological examination revealed a mature trabecular bone structure with considerable vascularization. Through tracking of the implanted cells, we also found that BMP-hBMSC were directly involved in the new bone formation. CONCLUSIONS: The robust, self-driven osteogenic capability and computer-designed architecture of the construct developed in this study should have potential applications for customized clinical repair of large bone defects or non-unions.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Hydrogels/chemistry , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Osteogenesis , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Bone Morphogenetic Protein 2/genetics , Cell Proliferation , Cells, Cultured , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Stereolithography
7.
Proc Natl Acad Sci U S A ; 115(35): E8256-E8265, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30104374

ABSTRACT

While lizards and salamanders both exhibit the ability to regenerate amputated tails, the outcomes achieved by each are markedly different. Salamanders, such as Ambystoma mexicanum, regenerate nearly identical copies of original tails. Regenerated lizard tails, however, exhibit important morphological differences compared with originals. Some of these differences concern dorsoventral patterning of regenerated skeletal and spinal cord tissues; regenerated salamander tail tissues exhibit dorsoventral patterning, while regrown lizard tissues do not. Additionally, regenerated lizard tails lack characteristically roof plate-associated structures, such as dorsal root ganglia. We hypothesized that differences in neural stem cells (NSCs) found in the ependyma of regenerated spinal cords account for these divergent regenerative outcomes. Through a combination of immunofluorescent staining, RT-PCR, hedgehog regulation, and transcriptome analysis, we analyzed NSC-dependent tail regeneration. Both salamander and lizard Sox2+ NSCs form neurospheres in culture. While salamander neurospheres exhibit default roof plate identity, lizard neurospheres exhibit default floor plate. Hedgehog signaling regulates dorsalization/ventralization of salamander, but not lizard, NSCs. Examination of NSC differentiation potential in vitro showed that salamander NSCs are capable of neural differentiation into multiple lineages, whereas lizard NSCs are not, which was confirmed by in vivo spinal cord transplantations. Finally, salamander NSCs xenogeneically transplanted into regenerating lizard tail spinal cords were influenced by native lizard NSC hedgehog signals, which favored salamander NSC floor plate differentiation. These findings suggest that NSCs in regenerated lizard and salamander spinal cords are distinct cell populations, and these differences contribute to the vastly different outcomes observed in tail regeneration.


Subject(s)
Cell Differentiation/physiology , Lizards/physiology , Neural Stem Cells/metabolism , Regeneration/physiology , Spinal Cord/physiology , Animals , Ependyma/metabolism , Species Specificity , Urodela
8.
Curr Pathobiol Rep ; 6(1): 61-69, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29967714

ABSTRACT

PURPOSE OF THE REVIEW: This manuscript discusses wound healing as a component of epimorphic regeneration and the role of the immune system in this process. RECENT FINDINGS: Epimorphic regeneration involves formation of a blastema, a mass of undifferentiated cells capable of giving rise to the regenerated tissues. The apical epithelial cap plays an important role in blastemal formation. SUMMARY: True regeneration is rarely observed in mammals. With the exception of transgenic strains, tissue repair in mammals usually leads to non-functional fibrotic tissue formation. In contrast, a number of lower order species including planarians, salamanders, and reptiles, have the ability to overcome the burden of scarring and tissue loss through complex adaptations that allow them to regenerate various anatomic structures through epimorphic regeneration. Blastemal cells have been suggested to originate via various mechanisms including de-differentiation, transdifferentiation, migration of pre-existing adult stem cell niches, and combinations of these.

9.
Article in English | MEDLINE | ID: mdl-29164111

ABSTRACT

INTRODUCTION: Human cartilage is an avascular tissue with limited capacity for repair. By contrast, certain lizards are capable of musculoskeletal tissue regeneration following tail loss throughout all stages of their lives. This extraordinary ability is the result of a complex process in which a blastema forms and gives rise to the tissues of the regenerate. Blastemal cells have been shown to originate either from dedifferentiated tissues or from existing progenitor cells in various species, but their origin has not been determined in lizards. As reptiles, lizards are the closest relatives to mammals with enhanced regenerative potential, and the origin of blastemal cells has important implications for the regenerative process. Hence, the aim of this study is to determine the cellular origin of regenerated cartilage and muscle tissues in reptiles using the mourning gecko lizard as the regenerative model. METHODS: To trace the fate and differentiation potential of cartilage during tail regeneration, cartilage cells pre-labeled with the fluorescent tracer Dil were injected into lizard tails, and the contribution of cartilage cells to regenerated tail tissues was assessed by histologic examination at 7, 14, and 21 days post-tail amputation. The contribution of muscle cells to regenerated tail tissues was evaluated using muscle creatine kinase promoter-driven Cre recombinase in conjunction with the Cre-responsive green-to-red fluorescence shift construct CreStoplight. 21 days after amputation, tail tissues were analyzed by histology for red fluorescent protein (RFP)-positive cells. RESULTS: At 7 days post-amputation, Dil-labeled cartilage cells localized to the subapical space contributing to the blastema. At 14 and 21 days post-amputation, Dil-labeled cells remained in the subapical space and colocalized with Collagen type II (Col2) staining in the cartilage tube and myosin heavy chain (MHC) staining in regenerated muscle. Lineage tracing of myocytes showed colocalization of RFP with Col2 and MHC in differentiated tissues at 21 days post-amputation. CONCLUSION: This study demonstrates that differentiated cartilage cells contribute to both regenerated muscle and cartilage tissues following tail loss, and in turn, differentiated muscle cells contribute to both tissue types as well. These findings suggest that dedifferentiation and/or transdifferentiation are at least partially responsible for the regenerative outcome in the mourning gecko.

10.
Sci Rep ; 7(1): 11327, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28900122

ABSTRACT

Currently, sustained in vivo delivery of active bone morphogenetic protein-2 (BMP-2) protein to responsive target cells, such as bone marrow-derived mesenchymal stem cells (BMSCs), remains challenging. Ex vivo gene transfer method, while efficient, requires additional operation for cell culture and therefore, is not compatible with point-of-care treatment. In this study, two lentiviral gene constructs - (1) Lv-BMP/GFP, containing human BMP-2 and green fluorescent protein (GFP) gene (BMP group); or (2) Lv-GFP, containing GFP gene (GFP group) - were incorporated with human BMSCs into a solution of photocrosslinkable gelatin, which was then subjected to visible light-based projection stereolithographic printing to form a scaffold with desired architectures. Upon in vitro culture, compared to the GFP group, cells from BMP group showed >1,000-fold higher BMP-2 release, and the majority of them stained intensely for alkaline phosphatase activity. Real-time RT-PCR also showed dramatically increased expression of osteogenesis marker genes only in the BMP group. 3.5 months post-implantation into SCID mice, the micro-computed tomography imaging showed detectable mineralized areas only in the BMP group, which was restricted within the scaffolds. Alizarin red staining and immunohistochemistry of GFP and osteocalcin further indicated that the grafted hBMSCs, not host cells, contributed primarily to the newly formed bone.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Bone Regeneration , Extracellular Matrix/metabolism , Tissue Engineering , Animals , Biomarkers , Bone Density , Bone Morphogenetic Protein 2/metabolism , Gene Expression , Genes, Reporter , Genetic Vectors/genetics , Humans , Immunohistochemistry , Lentivirus/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Models, Animal , Osteocalcin/genetics , Osteocalcin/metabolism , Osteogenesis/genetics , Transduction, Genetic
11.
Connect Tissue Res ; 58(2): 145-154, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27459585

ABSTRACT

The ability to regenerate damaged or lost tissues has remained the lofty goal of regenerative medicine. Unfortunately, humans, like most mammals, suffer from very minimal natural regenerative capabilities. Certain non-mammalian animal species, however, are not so limited in their healing capabilities, and several have attracted the attention of researchers hoping to recreate enhanced healing responses in humans. This review focuses on one such animal group with remarkable regenerative abilities, the lizards. As the closest relatives of mammals that exhibit enhanced regenerative abilities as adults, lizards potentially represent the most relevant model for direct comparison and subsequent improvement of mammalian healing. Lizards are able to regenerate amputated tails and exhibit adaptations that both limit tissue damage in response to injury and initiate coordinated regenerative responses. This review summarizes the salient aspects of lizard tail regeneration as they relate to the overall regenerative process and also presents the relevant information pertaining to regrowth of specific tissues, including skeletal, muscular, nervous, and vascular tissues. The goal of this review is to introduce the topic of lizard tail regeneration to new audiences with the hope of expanding the knowledge base of this underutilized but potentially powerful model organism.


Subject(s)
Lizards/physiology , Models, Biological , Regeneration/physiology , Tail , Animals , Humans
12.
Development ; 143(16): 2946-57, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27387871

ABSTRACT

Lizards are amniotes with the remarkable ability to regenerate amputated tails. The early regenerated lizard tail forms a blastema, and the regenerated skeleton consists of a cartilage tube (CT) surrounding the regenerated spinal cord. The proximal, but not distal, CT undergoes hypertrophy and ossifies. We hypothesized that differences in cell sources and signaling account for divergent cartilage development between proximal and distal CT regions. Exogenous spinal cord implants induced ectopic CT formation in lizard (Anolis carolinensis) blastemas. Regenerated spinal cords expressed Shh, and cyclopamine inhibited CT induction. Blastemas containing vertebrae with intact spinal cords formed CTs with proximal hypertrophic regions and distal non-hypertrophic regions, whereas removal of spinal cords resulted in formation of proximal CT areas only. In fate-mapping studies, FITC-labeled vertebra periosteal cells were detected in proximal, but not distal, CT areas. Conversely, FITC-labeled blastema cells were restricted to distal CT regions. Proximal cartilage formation was inhibited by removal of periosteum and could be recapitulated in vitro by periosteal cells treated with Ihh and BMP-2. These findings suggest that proximal CTs are directly derived from vertebra periosteal cells in response to BMP and Ihh signaling, whereas distal CTs form from blastema cells in response to Shh signals from regenerated spinal cords.


Subject(s)
Lizards/metabolism , Tail/physiology , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cartilage/metabolism , Cartilage/physiology , Cyclohexylamines/pharmacology , Fracture Healing/drug effects , Fracture Healing/genetics , Fracture Healing/physiology , Growth Plate/metabolism , Growth Plate/physiology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Regeneration/drug effects , Regeneration/genetics , Regeneration/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , Tail/metabolism , Thiophenes/pharmacology , Veratrum Alkaloids/pharmacology
13.
J Orthop Res ; 34(8): 1466-74, 2016 08.
Article in English | MEDLINE | ID: mdl-27246627

ABSTRACT

Neovascularization of intervertebral discs, a phenomenon considered pathological since normal discs are primarily avascular structures, occurs most frequently in annulus fibrosus (AF) of degenerated discs. Endothelial cells (ECs) are involved in this process, but the mechanism of the interaction between AF and endothelial cells is unclear. In this study, we evaluated the effects on matrix catabolic activity of AF cells by the extracellular endothelial microparticles (EMPs) and soluble protein factors (SUP fraction) produced from ECs. Passage 1 human AF cells grown in monolayer cultures were treated for 72 h with 250 µg of EMPs or SUP fraction isolated from culture of the microvascular endothelial cell line, HEMC-I. Live-cell imaging revealed uptake of EMPs by AF cells. RT-PCR analysis demonstrated increased mRNA expression of MMP-1 (50.3-fold), MMP-3 (4.5-fold) and MMP-13 (5.5-fold) in AF cell cultures treated with EMPs compared to untreated control. Western analysis also demonstrated increased MMP protein expression in EMP-treated AF cells. AF cells treated with the SUP fraction also exhibited a dramatic increase in MMP mRNA and protein expression. Increased MMP expression is primarily due to EMP or SUP stimulation of AF cells since EMPs or SUP fraction alone contained negligible amount of MMPs. Interestingly, MMP activity was elevated in AF cell cultures treated with EMPs but not with SUP. This study revealed enhanced matrix catabolism as a molecular consequence of action of ECs on AF cells via EMPs, which might be expected during neo-angiogenesis of degenerating disc. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1466-1474, 2016.


Subject(s)
Annulus Fibrosus/metabolism , Cell-Derived Microparticles , Endothelial Cells/physiology , Intervertebral Disc Degeneration/etiology , Neovascularization, Pathologic , Cell Line , Female , Humans , Intervertebral Disc Degeneration/metabolism , Male , Matrix Metalloproteinases/metabolism , Middle Aged
14.
Coluna/Columna ; 14(2): 77-81, Apr.-June 2015. tab, ilus
Article in English | LILACS | ID: lil-755841

ABSTRACT

OBJECTIVE:

To investigate the effects of proteins products of endothelial cells (ECs) on the annulus fibrosus (AF) cell metabolism in an in vitro culture.

METHODS:

Human AF cells were expanded in monolayer cultures and treated with proteins from the medium of cell line HMEC-1 (Human Microvascular Endothelial Cells) (125µg/ml). After 72h of treatment RNA was isolated from AF cells for analysis of gene expression and the culture medium was collected for protein expression analysis.

RESULTS:

The qRT-PCR analysis demonstrated increased gene expression of matrix metalloproteinases (MMPs) in AF cells treated with protein products of endothelial cells compared with cells from control group of AF cells: MMP-1 243.10 times (p<0.05), MMP-2 1.37 time (p<0.05), MMP-3 39.83 times (p<0.05) and MMP-13 5.70 times (p<0.05). In contrast, tissue inhibitors of metalloproteinases (TIMPs) were suppressed; TIMP-2 (0.55 time) (p<0.05) and TIMP-3 (0.60 time) (p<0.05) in the exposed groups. The expression of aggrecan gene (0.83 time) (p<0.05), an important extracellular matrix component, was also reduced. MMP-1 and MMP-3 detection was performed, confirming the results of PCR by Western Blot technique.

CONCLUSIONS:

In this study, we observed that the proteins produced by ECs induced the MMPs expression and suppressed the TIMPs as well as the aggrecan in primary cells of the human intervertebral disc, targeting the development of potential treatments for intervertebral disc degeneration and associated discogenic pain.

.

OBJETIVO:

Analisar o efeito de produtos proteicos de células endoteliais (CEs) sobre o metabolismo de células de ânulo fibroso (AF) em ambiente controlado de cultura celular in vitro.

MÉTODOS:

Células de AF humano foram expandidas em camada única e tratadas com proteínas obtidas a partir do meio de cultura de células da linhagem celular HMEC-1 (Human Microvascular Endothelial Cells) (125µg/ml). Após 72h de tratamento, isolou-se RNA das células de AF para análise da expressão gênica e coletou-se meio de cultura para análise de expressão proteica.

RESULTADOS:

A análise da qRT-PCR demonstrou aumento da expressão gênica das metaloproteinases de matriz (MMPs) nas células de AF tratadas com produtos proteicos das células endoteliais, em comparação com grupo controle de células de AF: MMP-1 243,10 vezes (p < 0,05), MMP-2 1,37 vezes (p < 0,05), MMP-3 39,83 vezes (p < 0,05) e MMP13 5,70 vezes (p < 0,05). Em contraste, os inibidores teciduais das metaloproteinases (TIMPs) apresentaram supressão da expressão gênica de TIMP-2 (0,55 vezes) (p < 0,05) e TIMP-3 (0,60 vezes) (p < 0,05) nos grupos expostos. A expressão do gene agrecan (0,83 vezes) (p < 0,05), componente importante da matriz extracelular, também estava diminuída. Foi realizada detecção de MMP-1 e MMP-3, confirmando os resultados de PCR através de técnica de Western Blot.

CONCLUSÕES:

Neste estudo observamos que proteínas produzidas pelas CEs induziram a expressão de MMPs e suprimiram a expressão de TIMPs e agrecan nas células primárias do disco intervertebral humano, objetivando desenvolvimento de potenciais terapias no tratamento da degeneração do disco intervertebral e dor discogênica associada.

.

OBJETIVO:

Analizar el efecto de los productos de proteína de las células endoteliales (CEs) en el metabolismo celular del anillo fibroso (AF) en sistema in vitro de cultivo controlado.

MÉTODOS:

Las células del AF humano se ampliaron en monocapa y se las trató con las proteínas obtenidas a partir de los medios de cultivo de la línea de células HMEC-1 (Human Microvascular Endothelial Cells) (125µg/ml). Después de 72h de tratamiento, se aisló el ARN de las células de AF para el análisis de la expresión génica y se recogió el medio de cultivo para el análisis de expresión de la proteína.

RESULTADOS:

El análisis de qRT-PCR demostró una mayor expresión génica de las metaloproteinasas de matriz (MMP) en las células tratadas con productos de proteína de AF en las células endoteliales, en comparación con el grupo de control de células AF: MMP-1 243,10 veces (p < 0,05), MMP-2 1,37 veces (p < 0,05), MMP-3 39,83 veces (p < 0,05) y MMP-13 5,70 veces (p < 0,05). En contraste, los inhibidores tisulares de las metaloproteinasas (TIMP), presentaron supresión de la expresión del gen TIMP-2 (0,55 veces) (p < 0,05) y TIMP-3 (0,60 veces) (p < 0,05) en los grupos expuestos. La expresión génica de agrecano (0,83 veces) (p < 0,05), importante componente de la matriz extracelular, también se redujo. La detección de MMP-1 y de MMP-3 fue realizada y se confirmaron los resultados de la PCR mediante la técnica Western Blot.

CONCLUSIONES:

En el presente estudio se observó que las proteínas producidas por las CEs indujeron la expresión de MMP y suprimieron la expresión del TIMP y de agrecano en células primarias del disco intervertebral humano, con el objetivo de desarrollar posibles tratamientos para la degeneración del disco intervertebral y el dolor discogénico asociado.

.


Subject(s)
Humans , Intervertebral Disc/cytology , Cell Culture Techniques , Matrix Metalloproteinases , Endothelial Cells
15.
Dev Biol ; 399(2): 249-62, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25596336

ABSTRACT

Lizards capable of caudal autotomy exhibit the remarkable ability to "drop" and then regenerate their tails. However, the regenerated lizard tail (RLT) is known as an "imperfect replicate" due to several key anatomical differences compared to the original tail. Most striking of these "imperfections" concerns the skeleton; instead of the vertebrae of the original tail, the skeleton of the RLT takes the form of an unsegmented cartilage tube (CT). Here we have performed the first detailed staging of skeletal development of the RLT CT, identifying two distinct mineralization events. CTs isolated from RLTs of various ages were analyzed by micro-computed tomography to characterize mineralization, and to correlate skeletal development with expression of endochondral ossification markers evaluated by histology and immunohistochemistry. During early tail regeneration, shortly after CT formation, the extreme proximal CT in direct contact with the most terminal vertebra of the original tail develops a growth plate-like region that undergoes endochondral ossification. Proximal CT chondrocytes enlarge, express hypertrophic markers, including Indian hedgehog (Ihh), apoptose, and are replaced by bone. During later stages of tail regeneration, the distal CT mineralizes without endochondral ossification. The sub-perichondrium of the distal CT expresses Ihh, and the perichondrium directly calcifies without cartilage growth plate formation. The calcified CT perichondrium also contains a population of stem/progenitor cells that forms new cartilage in response to TGF-ß stimulation. Treatment with the Ihh inhibitor cyclopamine inhibited both proximal CT ossification and distal CT calcification. Thus, while the two mineralization events are spatially, temporally, and mechanistically very different, they both involve Ihh. Taken together, these results suggest that Ihh regulates CT mineralization during two distinct stages of lizard tail regeneration.


Subject(s)
Calcification, Physiologic/physiology , Cartilage/physiology , Hedgehog Proteins/metabolism , Lizards/physiology , Osteogenesis/physiology , Regeneration/physiology , Tail/physiology , Animals , Blotting, Western , Osteoclasts/physiology , Signal Transduction/physiology , Tomography, X-Ray Computed
16.
J Cell Mol Med ; 18(12): 2372-84, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25250510

ABSTRACT

Tightly associated with blood vessels in their perivascular niche, human mesenchymal stem cells (MSCs) closely interact with endothelial cells (ECs). MSCs also home to tumours and interact with cancer cells (CCs). Microparticles (MPs) are cell-derived vesicles released into the extracellular environment along with secreted factors. MPs are capable of intercellular signalling and, as biomolecular shuttles, transfer proteins and RNA from one cell to another. Here, we characterize interactions among ECs, CCs and MSCs via MPs and secreted factors in vitro. MPs and non-MP secreted factors (Sup) were isolated from serum-free medium conditioned by human microvascular ECs (HMEC-1) or by the CC line HT1080. Fluorescently labelled MPs were prepared from cells treated with membrane dyes, and cytosolic GFP-containing MPs were isolated from cells transduced with CMV-GFP lentivirus. MSCs were treated with MPs, Sup, or vehicle controls, and analysed for MP uptake, proliferation, migration, activation of intracellular signalling pathways and cytokine release. Fluorescently labelled MPs fused with MSCs, transferring the fluorescent dyes to the MSC surface. GFP was transferred to and retained in MSCs incubated with GFP-MPs, but not free GFP. Thus, only MP-associated cellular proteins were taken up and retained by MSCs, suggesting that MP biomolecules, but not secreted factors, are shuttled to MSCs. MP and Sup treatment significantly increased MSC proliferation, migration, and MMP-1, MMP-3, CCL-2/MCP-1 and IL-6 secretion compared with vehicle controls. MSCs treated with Sup and MPs also exhibited activated NF-κB signalling. Taken together, these results suggest that MPs act to regulate MSC functions through several mechanisms.


Subject(s)
Cell Communication , Cell-Derived Microparticles/metabolism , Endothelial Cells/metabolism , Mesenchymal Stem Cells/metabolism , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell-Derived Microparticles/ultrastructure , Cells, Cultured , Cytokines/metabolism , Endothelial Cells/cytology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mesenchymal Stem Cells/cytology , Microscopy, Confocal , Microscopy, Electron, Transmission , NF-kappa B/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Time-Lapse Imaging/methods
17.
Exp Biol Med (Maywood) ; 239(9): 1080-95, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24994814

ABSTRACT

Tissue engineered constructs have the potential to function as in vitro pre-clinical models of normal tissue function and disease pathogenesis for drug screening and toxicity assessment. Effective high throughput assays demand minimal systems with clearly defined performance parameters. These systems must accurately model the structure and function of the human organs and their physiological response to different stimuli. Musculoskeletal tissues present unique challenges in this respect, as they are load-bearing, matrix-rich tissues whose functionality is intimately connected to the extracellular matrix and its organization. Of particular clinical importance is the osteochondral junction, the target tissue affected in degenerative joint diseases, such as osteoarthritis (OA), which consists of hyaline articular cartilage in close interaction with subchondral bone. In this review, we present an overview of currently available in vitro three-dimensional systems for bone and cartilage tissue engineering that mimic native physiology, and the utility and limitations of these systems. Specifically, we address the need to combine bone, cartilage and other tissues to form an interactive microphysiological system (MPS) to fully capture the biological complexity and mechanical functions of the osteochondral junction of the articular joint. The potential applications of three-dimensional MPSs for musculoskeletal biology and medicine are highlighted.


Subject(s)
Chondrogenesis , Osteoarthritis , Osteogenesis , Tissue Culture Techniques/instrumentation , Tissue Culture Techniques/methods , Animals , Extracellular Matrix/metabolism , Humans , Osteoarthritis/mortality , Osteoarthritis/pathology , Osteoarthritis/physiopathology
18.
Mol Pharm ; 11(7): 2203-12, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24830762

ABSTRACT

Osteoarthritis (OA) is a chronic degenerative disease of the articular joint that involves both bone and cartilage degenerative changes. An engineered osteochondral tissue within physiological conditions will be of significant utility in understanding the pathogenesis of OA and testing the efficacy of potential disease-modifying OA drugs (DMOADs). In this study, a multichamber bioreactor was fabricated and fitted into a microfluidic base. When the osteochondral construct is inserted, two chambers are formed on either side of the construct (top, chondral; bottom, osseous) that is supplied by different medium streams. These medium conduits are critical to create tissue-specific microenvironments in which chondral and osseous tissues will develop and mature. Human bone marrow stem cell (hBMSCs)-derived constructs were fabricated in situ and cultured within the bioreactor and induced to undergo spatially defined chondrogenic and osteogenic differentiation for 4 weeks in tissue-specific media. We observed tissue specific gene expression and matrix production as well as a basophilic interface suggesting a developing tidemark. Introduction of interleukin-1ß (IL-1ß) to either the chondral or osseous medium stream induced stronger degradative responses locally as well as in the opposing tissue type. For example, IL-1ß treatment of the osseous compartment resulted in a strong catabolic response in the chondral layer as indicated by increased matrix metalloproteinase (MMP) expression and activity, and tissue-specific gene expression. This induction was greater than that seen with IL-1ß application to the chondral component directly, indicative of active biochemical communication between the two tissue layers and supporting the osteochondral nature of OA. The microtissue culture system developed here offers novel capabilities for investigating the physiology of osteochondral tissue and pathogenic mechanisms of OA and serving as a high-throughput platform to test potential DMOADS.


Subject(s)
Chondrogenesis/physiology , Interleukin-1beta/metabolism , Osteogenesis/physiology , Stem Cells/physiology , Adult , Aged , Basophils/metabolism , Basophils/physiology , Bioreactors , Cartilage, Articular/metabolism , Cartilage, Articular/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Chondrogenesis/genetics , Female , Gene Expression/genetics , Humans , Interleukin-1beta/genetics , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Middle Aged , Osteogenesis/genetics , Stem Cells/metabolism , Tissue Engineering/methods
19.
Stem Cells Transl Med ; 3(2): 241-54, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24436440

ABSTRACT

Stem cells are promising candidate cells for regenerative applications because they possess high proliferative capacity and the potential to differentiate into other cell types. Mesenchymal stem cells (MSCs) are easily sourced but do not retain their proliferative and multilineage differentiative capabilities after prolonged ex vivo propagation. We investigated the use of hypoxia as a preconditioning agent and in differentiating cultures to enhance MSC function. Culture in 5% ambient O(2) consistently enhanced clonogenic potential of primary MSCs from all donors tested. We determined that enhanced clonogenicity was attributable to increased proliferation, increased vascular endothelial growth factor secretion, and increased matrix turnover. Hypoxia did not impact the incidence of cell death. Application of hypoxia to osteogenic cultures resulted in enhanced total mineral deposition, although this effect was detected only in MSCs preconditioned in normoxic conditions. Osteogenesis-associated genes were upregulated in hypoxia, and alkaline phosphatase activity was enhanced. Adipogenic differentiation was inhibited by exposure to hypoxia during differentiation. Chondrogenesis in three-dimensional pellet cultures was inhibited by preconditioning with hypoxia. However, in cultures expanded under normoxia, hypoxia applied during subsequent pellet culture enhanced chondrogenesis. Whereas hypoxic preconditioning appears to be an excellent way to expand a highly clonogenic progenitor pool, our findings suggest that it may blunt the differentiation potential of MSCs, compromising their utility for regenerative tissue engineering. Exposure to hypoxia during differentiation (post-normoxic expansion), however, appears to result in a greater quantity of functional osteoblasts and chondrocytes and ultimately a larger quantity of high-quality differentiated tissue.


Subject(s)
Adipocytes/cytology , Chondrocytes/cytology , Hypoxia/pathology , Ischemic Preconditioning/methods , Mesenchymal Stem Cells/cytology , Osteocytes/cytology , Cell Death/physiology , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Clone Cells/cytology , Cytokines/metabolism , Fibroblasts/cytology , Humans , Hypoxia/physiopathology , Immunophenotyping , Matrix Metalloproteinases/metabolism , Mesenchymal Stem Cells/metabolism , Oxygen/pharmacology
20.
Matrix Biol ; 34: 132-43, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24140982

ABSTRACT

Mesenchymal stem cells (MSCs) are attractive candidates for inclusion in cell-based therapies by virtue of their abilities to home to wound sites. However, in-depth characterization of the specific effects of MSCs on their microenvironments is needed to realize their full therapeutic potentials. Furthermore, since MSCs of varying properties can be isolated from a diverse spectrum of tissues, a strategic and rational approach in MSC sourcing for a particular application has yet to be achieved. For example, MSCs that activate their proteolytic environments may promote tissue remodeling, while those from different tissue sources may inhibit proteases and promote tissue stabilization. This study attempts to address these issues by analyzing MSCs isolated from three adult tissue sources in terms of their effects on their proteolytic microenvironments. Human bone marrow, adipose, and traumatized muscle derived MSCs were compared in their soluble and cellular-associated MMP components and activity. For all types of MSCs, MMP activity associated with the cell surface, but activity levels and MMP profiles differed with tissue source. All MSC types bound exogenous active MMPs at their surfaces. MSCs were also able to activate exogenous proMMP-2 and proMMP-13. This is in marked contrast to the MSC soluble compartment, which strongly inhibited MMPs via endogenous TIMPs. The exact TIMP used to inhibit the exogenous MMP differed with MSC type. Thus, MSCs saturate their environment with both MMPs and TIMPs. Since they bind and activate MMPs at their surfaces, the net result is a very controlled pericellular localization of MMP activities by MSCs.


Subject(s)
Matrix Metalloproteinase 13/metabolism , Mesenchymal Stem Cells/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Cells, Cultured , Cellular Microenvironment/genetics , Enzyme Precursors/metabolism , Gelatinases/metabolism , Humans , Protein Binding , Tissue Inhibitor of Metalloproteinases/metabolism , Tissue Inhibitor of Metalloproteinase-4
SELECTION OF CITATIONS
SEARCH DETAIL
...