Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Science ; 385(6705): 154-156, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38991082

ABSTRACT

Highlights from the Science family of journals.

2.
Science ; 382(6677): 1371-1373, 2023 Dec 22.
Article in English | MEDLINE | ID: mdl-38127760

ABSTRACT

Highlights from the Science family of journals.

3.
Sci Adv ; 8(8): eabd8056, 2022 02 25.
Article in English | MEDLINE | ID: mdl-35213226

ABSTRACT

Understanding the immune response to hydrogel implantation is critical for the design of immunomodulatory biomaterials. To study the progression of inflammation around poly(ethylene glycol) hydrogels presenting Arg-Gly-Asp (RGD) peptides and vascular endothelial growth factor, we used temporal analysis of high-dimensional flow cytometry data paired with intravital imaging, immunohistochemistry, and multiplexed proteomic profiling. RGD-presenting hydrogels created a reparative microenvironment promoting CD206+ cellular infiltration and revascularization in wounded dorsal skin tissue. Unbiased clustering algorithms (SPADE) revealed significant phenotypic transition shifts as a function of the cell-adhesion hydrogel properties. SPADE identified an intermediate macrophage subset functionally regulating in vivo cytokine secretion that was preferentially recruited for RGD-presenting hydrogels, whereas dendritic cell subsets were preferentially recruited to RDG-presenting hydrogels. Last, RGD-presenting hydrogels controlled macrophage functional cytokine secretion to direct polarization and vascularization. Our studies show that unbiased clustering of single-cell data provides unbiased insights into the underlying immune response to engineered materials.


Subject(s)
Hydrogels , Vascular Endothelial Growth Factor A , Biocompatible Materials/chemistry , Cluster Analysis , Cytokines , Hydrogels/chemistry , Immunity , Oligopeptides/chemistry , Proteomics
4.
Cytotherapy ; 24(6): 608-618, 2022 06.
Article in English | MEDLINE | ID: mdl-35190267

ABSTRACT

Cell therapies are expected to increase over the next decade owing to increasing demand for clinical applications. Mesenchymal stromal cells (MSCs) have been explored to treat a number of diseases, with some successes in early clinical trials. Despite early successes, poor MSC characterization results in lessened therapeutic capacity once in vivo. Here, we characterized MSCs derived from bone marrow (BM), adipose tissue and umbilical cord tissue for sphingolipids (SLs), a class of bioactive lipids, using liquid chromatography/tandem mass spectrometry. We found that ceramide levels differed based on the donor's sex in BM-MSCs. We detected fatty acyl chain variants in MSCs from all three sources. Linear discriminant analysis revealed that MSCs separated based on tissue source. Principal component analysis showed that interferon-γ-primed and unstimulated MSCs separated according to their SL signature. Lastly, we detected higher ceramide levels in low indoleamine 2,3-dioxygenase MSCs, indicating that sphingomyelinase or ceramidase enzymatic activity may be involved in their immune potency.


Subject(s)
Mesenchymal Stem Cells , Sphingolipids , Adipose Tissue , Bone Marrow Cells , Cell Differentiation , Cell Proliferation , Cells, Cultured , Ceramides , Humans , Lipidomics
5.
Stem Cell Res Ther ; 12(1): 565, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34736534

ABSTRACT

BACKGROUND: Human Mesenchymal stromal cells (hMSCs) from various tissue sources are widely investigated in clinical trials. These MSCs are often administered to patients immediately after thawing the cryopreserved product (out-of-thaw), yet little is known about the single-cell transcriptomic landscape and tissue-specific differences of out-of-thaw human MSCs. METHODS: 13 hMSC samples derived from 10 "healthy" donors were used to assess donor variability and tissue-of-origin differences in single-cell gene expression profiles. hMSCs derived and expanded from the bone marrow (BM) or cord tissue (CT) underwent controlled-rate freezing for 24 h. Cells were then transferred to the vapor phase of liquid nitrogen for cryopreservation. hMSCs cryopreserved for at least one week, were characterized immediately after thawing using a droplet-based single-cell RNA sequencing method. Data analysis was performed with SC3 and SEURAT pipelines followed by gene ontology analysis. RESULTS: scRNA-seq analysis of the hMSCs revealed two major clusters of donor profiles, which differ in immune-signaling, cell surface properties, abundance of cell-cycle related transcripts, and metabolic pathways of interest. Within-sample transcriptomic heterogeneity is low. We identified numerous differentially expressed genes (DEGs) that are associated with various cellular functions, such as cytokine signaling, cell proliferation, cell adhesion, cholesterol/steroid biosynthesis, and regulation of apoptosis. Gene-set enrichment analyses indicated different functional pathways in BM vs. CT hMSCs. In addition, MSC-batches showed significant variations in cell cycle status, suggesting different proliferative vs. immunomodulatory potential. Several potential transcript-markers for tissue source differences were identified for further investigation in future studies. In functional assays, both BM and CT MSCs suppressed macrophage TNFα secretion upon interferon stimulation. However, differences between donors, tissue-of-origin, and cell cycle are evident in both TNF suppression and cytokine secretion. CONCLUSIONS: This study shows that donor differences in hMSC transcriptome are minor relative to the intrinsic differences in tissue-of-origin. hMSCs with different transcriptomic profiles showed potential differences in functional characteristics. These findings contribute to our understanding of tissue origin-based differences in out-of-thaw therapeutic hMSC products and assist in the identification of cells with immune-regulatory or survival potential from a heterogeneous MSC population. Our results form the basis of future studies in correlating single-cell transcriptomic markers with immunomodulatory functions.


Subject(s)
Mesenchymal Stem Cells , Bone Marrow Cells , Cell Cycle/genetics , Cell Differentiation , Cell Proliferation/genetics , Cells, Cultured , Humans , Mesenchymal Stem Cells/metabolism , RNA-Seq , Tissue Donors
6.
Front Bioeng Biotechnol ; 9: 650289, 2021.
Article in English | MEDLINE | ID: mdl-33816455

ABSTRACT

Volumetric muscle loss (VML) injuries after extremity trauma results in an important clinical challenge often associated with impaired healing, significant fibrosis, and long-term pain and functional deficits. While acute muscle injuries typically display a remarkable capacity for regeneration, critically sized VML defects present a dysregulated immune microenvironment which overwhelms innate repair mechanisms leading to chronic inflammation and pro-fibrotic signaling. In this series of studies, we developed an immunomodulatory biomaterial therapy to locally modulate the sphingosine-1-phosphate (S1P) signaling axis and resolve the persistent pro-inflammatory injury niche plaguing a critically sized VML defect. Multiparameter pseudo-temporal 2D projections of single cell cytometry data revealed subtle distinctions in the altered dynamics of specific immune subpopulations infiltrating the defect that were critical to muscle regeneration. We show that S1P receptor modulation via nanofiber delivery of Fingolimod (FTY720) was characterized by increased numbers of pro-regenerative immune subsets and coincided with an enriched pool of muscle stem cells (MuSCs) within the injured tissue. This FTY720-induced priming of the local injury milieu resulted in increased myofiber diameter and alignment across the defect space followed by enhanced revascularization and reinnervation of the injured muscle. These findings indicate that localized modulation of S1P receptor signaling via nanofiber scaffolds, which resemble the native extracellular matrix ablated upon injury, provides great potential as an immunotherapy for bolstering endogenous mechanisms of regeneration following VML injury.

7.
Biomaterials ; 268: 120475, 2021 01.
Article in English | MEDLINE | ID: mdl-33321293

ABSTRACT

Inflammation after traumatic injury or surgical intervention is both a protective tissue response leading to regeneration and a potential cause of wound complications. One potentially successful strategy to harness to pro-regenerative roles of host inflammation is the localized delivery of bioactive materials to induce immune suppressive cellular responses by cells responding to injury. In this study, we designed a fully synthetic poly (ethylene) glycol (PEG)-based hydrogel to release the specialized pro-resolving lipid mediator aspirin-triggered resolvin-D1 (AT-RvD1) and recombinant human interleukin 10 (IL-10). We utilized a unique side-by-side internally controlled implant design wherein bioactive hydrogels were implanted adjacent to control hydrogels devoid of immune modulatory factors in the dorsal skinfold window chamber. We also explored single-immune cell data with unsupervised approaches such as SPADE. First, we show that RGD-presenting hydrogel delivery results in enhanced immune cell recruitment to the site of injury. We then use intra-vital imaging to assess cellular recruitment and microvascular remodeling to show an increase in the caliber and density of local microvessels. Finally, we show that the recruitment and re-education of mononuclear phagocytes by combined delivery IL-10 and AT-RvD1 localizes immune suppressive subsets to the hydrogel, including CD206+ macrophages (M2a/c) and IL-10 expressing dendritic cells in the context of chronic inflammation following surgical tissue disruption. These data demonstrate the potential of combined delivery on the recruitment of regenerative cell subsets involved in wound healing complications.


Subject(s)
Aspirin , Interleukin-10 , Biocompatible Materials , Humans , Hydrogels , Phenotype
8.
J Biomed Mater Res A ; 109(5): 695-712, 2021 05.
Article in English | MEDLINE | ID: mdl-32608188

ABSTRACT

Regeneration of skeletal muscle after volumetric injury is thought to be impaired by a dysregulated immune microenvironment that hinders endogenous repair mechanisms. Such defects result in fatty infiltration, tissue scarring, chronic inflammation, and debilitating functional deficits. Here, we evaluated the key cellular processes driving dysregulation in the injury niche through localized modulation of sphingosine-1-phosphate (S1P) receptor signaling. We employ dimensionality reduction and pseudotime analysis on single cell cytometry data to reveal heterogeneous immune cell subsets infiltrating preclinical muscle defects due to S1P receptor inhibition. We show that global knockout of S1P receptor 3 (S1PR3) is marked by an increase of muscle stem cells within injured tissue, a reduction in classically activated relative to alternatively activated macrophages, and increased bridging of regenerating myofibers across the defect. We found that local S1PR3 antagonism via nanofiber delivery of VPC01091 replicated key features of pseudotime immune cell recruitment dynamics and enhanced regeneration characteristic of global S1PR3 knockout. Our results indicate that local S1P receptor modulation may provide an effective immunotherapy for promoting a proreparative environment leading to improved regeneration following muscle injury.


Subject(s)
Cyclopentanes/therapeutic use , Immunotherapy/methods , Muscle, Skeletal/injuries , Regeneration/drug effects , Sphingosine-1-Phosphate Receptors/physiology , Animals , Cyclopentanes/pharmacology , Drug Liberation , Flow Cytometry , Leukopenia/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Atomic Force , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myeloid Cells/immunology , Nanofibers , Organ Size , Quadriceps Muscle/immunology , Quadriceps Muscle/injuries , Quadriceps Muscle/metabolism , Quadriceps Muscle/pathology , Signal Transduction/drug effects , Sphingosine-1-Phosphate Receptors/deficiency , Sphingosine-1-Phosphate Receptors/genetics , T-Lymphocyte Subsets/immunology , Tissue Scaffolds
9.
Tissue Eng Part A ; 26(23-24): 1259-1271, 2020 12.
Article in English | MEDLINE | ID: mdl-32628570

ABSTRACT

Current cell culture surfaces used for the expansion and production of mesenchymal stromal cells (MSCs) are not optimized for the production of highly secretory and nonsenescent cells. In this study, we used poly (ethylene glycol) hydrogel substrates with tunable mechanical and biochemical properties to screen the effect of culture surfaces on pro-regenerative secretome by multiplex enzyme-linked immunosorbent assay, proliferation by PicoGreen DNA analysis, and senescence by senescence-associated ß-galactosidase activity. We demonstrate that MSCs cultured on 30 kPa hydrogels, regardless of biochemical functionalization, broadly enhanced the secretion of immunomodulatory and regenerative factors versus stiffer 100 kPa or tissue culture plastic surfaces, but did not support robust proliferation. In contrast, culture on 100 kPa hydrogel surfaces promoted proliferation at a similar level and did not substantially alter the amount of secreted factors as compared with tissue culture plastic. Culture on integrin-engaging, cadherin-engaging, and hyaluronic acid-containing 30 kPa substrates enhanced MSC-conditioned media (CM) angiogenic activity in a human umbilical vein endothelial cell tube formation assay and human THP-1 monocyte chemoattraction in a transwell assay. However, 30 kPa substrate culture did not impact the myogenic activity of MSC CM in a C2C12 myoblast tube formation assay. Culture on selected 100 kPa surfaces enhanced CM angiogenic activity and monocyte chemotaxis, but not myogenic activity. Serial culture on 100 kPa RGD hydrogel surfaces significantly reduced senescence in MSCs versus tissue culture plastic, while maintaining the capacity of the cells to enhance their secretome in response to 30 kPa surfaces. Thus, hydrogel substrates that exhibit stiffness orders of magnitude lower than standard tissue culture plastic can serve as novel surfaces for the production of MSCs with an improved therapeutic secretory capacity and reduced senescence. Impact statement The success of mesenchymal stromal cell (MSC)-based therapies is dependent on the manufacture of a large number of cells with high therapeutic potency. Among the culture surfaces tested in this study, we demonstrate that substrate stiffness rather than biochemical functionalization predominantly guides changes in MSC proliferation and secretory capacity. We have identified substrate parameters to support MSC proliferation, enhance secretion of paracrine factors, and to reduce replicative senescence. By maximizing secretory capacity and reducing senescence through the choice of hydrogel culture materials, these findings have great potential to improve the large-scale production of therapeutic MSCs.


Subject(s)
Cellular Senescence , Hydrogels , Mesenchymal Stem Cells , Cell Differentiation , Cell Line , Cell Proliferation , Culture Media, Conditioned , Human Umbilical Vein Endothelial Cells , Humans , Mesenchymal Stem Cells/cytology
10.
Tissue Eng Part C Methods ; 25(2): 59-70, 2019 02.
Article in English | MEDLINE | ID: mdl-30648479

ABSTRACT

IMPACT STATEMENT: The goal of this study was to determine the threshold for a critically sized, nonhealing muscle defect by characterizing key components in the balance between fibrosis and regeneration as a function of injury size in the mouse quadriceps. There is currently limited understanding of what leads to a critically sized muscle defect and which muscle regenerative components are functionally impaired. With the substantial increase in preclinical VML models as testbeds for tissue engineering therapeutics, defining the critical threshold for VML injuries will be instrumental in characterizing therapeutic efficacy and potential for subsequent translation.


Subject(s)
Muscular Diseases/pathology , Muscular Diseases/therapy , Myofibrils/physiology , Neuromuscular Junction/cytology , Quadriceps Muscle/cytology , Quadriceps Muscle/injuries , Tissue Engineering , Animals , Female , Mice , Mice, Inbred C57BL , Quadriceps Muscle/physiology , Tissue Scaffolds , Wound Healing
11.
Cell Mol Bioeng ; 11(4): 241-253, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29983824

ABSTRACT

INTRODUCTION: Mesenchymal stem and progenitor cells (MSCs), which normally reside in the bone marrow, are critical to bone health and can be recruited to sites of traumatic bone injury, contributing to new bone formation. The ability to control the trafficking of MSCs provides therapeutic potential for improving traumatic bone healing and therapy for genetic bone diseases such as hypophosphatasia. METHODS: In this study, we explored the sphingosine-1-phosphate (S1P) signaling axis as a means to control the mobilization of MSCs into blood and possibly to recruit MSCs enhancing bone growth. RESULTS: Loss of S1P receptor 3 (S1PR3) leads to an increase in circulating CD45-/CD29+/CD90+/Sca1 putative mesenchymal progenitor cells, suggesting that blocking S1PR3 may stimulate MSCs to leave the bone marrow. Antagonism of S1PR3 with the small molecule VPC01091 stimulated acute migration of CD45-/CD29+/CD90+/Sca1+ MSCs into the blood as early as 1.5 hours after treatment. VPC01091 administration also increased ectopic bone formation induced by BMP-2 and significantly increased new bone formation in critically sized rat cranial defects, suggesting that mobilized MSCs may home to injuries to contribute to healing. We also explored the possibility of combining S1P manipulation of endogenous host cell occupancy with exogenous MSC transplantation for potential use in combination therapies. Importantly, reducing niche occupancy of host MSCs with VPC01091 does not impede engraftment of exogenous MSCs. CONCLUSIONS: Our studies suggest that MSC mobilization through S1PR3 antagonism is a promising strategy for endogenous tissue engineering and improving MSC delivery to treat bone diseases.

12.
ACS Biomater Sci Eng ; 4(4): 1241-1250, 2018 Apr 09.
Article in English | MEDLINE | ID: mdl-29682605

ABSTRACT

The immune response to biomaterial implants critically regulates functional outcomes such as vascularization, transplant integration/survival, and fibrosis. To create "immunologically smart" materials, the host-material response may be engineered to optimize the recruitment of pro-regenerative leukocyte subsets which mature into corresponding wound-healing macrophages. We have recently identified a unique feature of pro-regenerative Ly6Clow monocytes that is a higher expression of both the bioactive lipid receptor sphingosine-1-phosphate receptor 3 (S1PR3) and the stromal derived factor-1α (SDF-1α) receptor CXCR4. Therefore, we designed a bifunctional hydrogel to harnesses a mechanistic synergy between these signaling axes to enhance the recruitment of endogenous pro-regenerative monocytes. To overcome the challenge of codelivering two physiochemically distinct molecules-a large hydrophilic protein and hydrophobic small molecule-we engineered a dual affinity hydrogel that exploits the growth factor affinity of a heparin derivative (Hep-N) and lipid chaperone activity of albumin. The sphingosine analog FTY720 and SDF-1α are successfully loaded and coreleased from the Hep-N-functionalized PEG-DA hydrogels while maintaining bioactivity. Placement of these hydrogels into a murine partial thickness skin wound demonstrates that corelease of FTY720 and SDF-1α yields superior recruitment of myeloid cells to the implant interface compared to either factor alone. Although in vivo delivery of FTY720 or SDF-1α individually promotes the enhanced recruitment of Ly-6Clow anti-inflammatory monocytes, codelivery enhances the early accumulation and persistence of the differentiated wound healing CD206+ macrophages in the tissue surrounding the gel. Co-delivery similarly promoted the synergistic expansion of vasculature adjacent to the implant, a key step in tissue healing. Taken together, these findings suggest that the combination of chemotactic molecules may provide additional maturation signals to the infiltrating leukocytes to facilitate macrophage transition and vascular network expansion, thus, ultimately, potentiating tissue repair. The coupling of multiple pro-regenerative biological cues provides a foundation for more fine-tuned immunoregenerative modulation to facilitate tissue repair.

13.
Sci Rep ; 7(1): 447, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28348370

ABSTRACT

Successful tissue repair requires the activities of myeloid cells such as monocytes and macrophages that guide the progression of inflammation and healing outcome. Immunoregenerative materials leverage the function of endogenous immune cells to orchestrate complex mechanisms of repair; however, a deeper understanding of innate immune cell function in inflamed tissues and their subsequent interactions with implanted materials is necessary to guide the design of these materials. Blood monocytes exist in two primary subpopulations, characterized as classical inflammatory or non-classical. While classical monocytes extravasate into inflamed tissue and give rise to macrophages or dendritic cells, the recruitment kinetics and functional role of non-classical monocytes remains unclear. Here, we demonstrate that circulating non-classical monocytes are directly recruited to polymer films within skin injuries, where they home to a perivascular niche and generate alternatively activated, wound healing macrophages. Selective labeling of blood monocyte subsets indicates that non-classical monocytes are biased progenitors of alternatively activated macrophages. On-site delivery of the immunomodulatory small molecule FTY720 recruits S1PR3-expressing non-classical monocytes that support vascular remodeling after injury. These results elucidate a previously unknown role for blood-derived non-classical monocytes as contributors to alternatively activated macrophages, highlighting them as key regulators of inflammatory response and regenerative outcome.


Subject(s)
Macrophages/pathology , Monocytes/pathology , Soft Tissue Injuries/pathology , Stem Cells/pathology , Wound Healing , Adoptive Transfer , Animals , Antigens, CD/metabolism , Arterioles/drug effects , Arterioles/metabolism , Biocompatible Materials/pharmacology , Cell Differentiation/drug effects , Fingolimod Hydrochloride/pharmacology , Implants, Experimental , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Models, Biological , Monocytes/drug effects , Monocytes/metabolism , Skin/blood supply , Skin/pathology , Wound Healing/drug effects
14.
Stem Cells ; 35(4): 1040-1052, 2017 04.
Article in English | MEDLINE | ID: mdl-28026131

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) egress from bone marrow (BM) during homeostasis and at increased rates during stress; however, the mechanisms regulating their trafficking remain incompletely understood. Here we describe a novel role for lipid receptor, sphingosine-1-phosphate receptor 3 (S1PR3), in HSPC residence within the BM niche. HSPCs expressed increased levels of S1PR3 compared to differentiated BM cells. Pharmacological antagonism or knockout (KO) of S1PR3 mobilized HSPCs into blood circulation, suggesting that S1PR3 influences niche localization. S1PR3 antagonism suppressed BM and plasma SDF-1, enabling HSPCs to migrate toward S1P-rich plasma. Mobilization synergized with AMD3100-mediated antagonism of CXCR4, which tethers HSPCs in the niche, and recovered homing deficits of AMD3100-treated grafts. S1PR3 antagonism combined with AMD3100 improved re-engraftment and survival in lethally irradiated recipients. Our studies indicate that S1PR3 and CXCR4 signaling cooperate to maintain HSPCs within the niche under homeostasis. These results highlight an important role for S1PR3 in HSPC niche occupancy and trafficking that can be harnessed for both rapid clinical stem cell mobilization and re-engraftment strategies, as well as the opportunity to design novel therapeutics for control of recruitment, homing, and localization through bioactive lipid signaling. Stem Cells 2017;35:1040-1052.


Subject(s)
Hematopoietic Stem Cells/metabolism , Receptors, Lysosphingolipid/metabolism , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Adhesion/drug effects , Cellular Microenvironment/drug effects , Chemotaxis/drug effects , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Ligands , Lysophospholipids/pharmacology , Male , Mice, Inbred C57BL , Radiation, Ionizing , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Stem Cell Niche/drug effects
15.
Exp Biol Med (Maywood) ; 241(10): 1084-97, 2016 05.
Article in English | MEDLINE | ID: mdl-27229903

ABSTRACT

Monocytes and macrophages play a critical role in tissue development, homeostasis, and injury repair. These innate immune cells participate in guiding vascular remodeling, stimulation of local stem and progenitor cells, and structural repair of tissues such as muscle and bone. Therefore, there is a great interest in harnessing this powerful endogenous cell source for therapeutic regeneration through immunoregenerative biomaterial engineering. These materials seek to harness specific subpopulations of monocytes/macrophages to promote repair by influencing their recruitment, positioning, differentiation, and function within a damaged tissue. Monocyte and macrophage phenotypes span a continuum of inflammatory (M1) to anti-inflammatory or pro-regenerative cells (M2), and their heterogeneous functions are highly dependent on microenvironmental cues within the injury niche. Increasing evidence suggests that division of labor among subpopulations of monocytes and macrophages could allow for harnessing regenerative functions over inflammatory functions of myeloid cells; however, the complex balance between necessary functions of inflammatory versus regenerative myeloid cells remains to be fully elucidated. Historically, biomaterial-based therapies for promoting tissue regeneration were designed to minimize the host inflammatory response; although, recent appreciation for the roles that innate immune cells play in tissue repair and material integration has shifted this paradigm. A number of opportunities exist to exploit known signaling systems of specific populations of monocytes/macrophages to promote repair and to better understand the biological and pathological roles of myeloid cells. This review seeks to outline the characteristics of distinct populations of monocytes and macrophages, identify the role of these cells within diverse tissue injury niches, and offer design criteria for immunoregenerative biomaterials given the intrinsic inflammatory response to their implantation.


Subject(s)
Biocompatible Materials/pharmacology , Guided Tissue Regeneration/methods , Immunologic Factors/pharmacology , Macrophages/physiology , Monocytes/physiology , Wounds and Injuries/therapy , Animals , Biocompatible Materials/metabolism , Humans , Immunologic Factors/metabolism , Macrophages/drug effects , Monocytes/drug effects
16.
ACS Nano ; 8(12): 12080-91, 2014 Dec 23.
Article in English | MEDLINE | ID: mdl-25426706

ABSTRACT

Oxygenation in tissue scaffolds continues to be a limiting factor in regenerative medicine despite efforts to induce neovascularization or to use oxygen-generating materials. Unfortunately, many established methods to measure oxygen concentration, such as using electrodes, require mechanical disturbance of the tissue structure. To address the need for scaffold-based oxygen concentration monitoring, a single-component, self-referenced oxygen sensor was made into nanofibers. Electrospinning process parameters were tuned to produce a biomaterial scaffold with specific morphological features. The ratio of an oxygen sensitive phosphorescence signal to an oxygen insensitive fluorescence signal was calculated at each image pixel to determine an oxygenation value. A single component boron dye-polymer conjugate was chosen for additional investigation due to improved resistance to degradation in aqueous media compared to a boron dye polymer blend. Standardization curves show that in fully supplemented media, the fibers are responsive to dissolved oxygen concentrations less than 15 ppm. Spatial (millimeters) and temporal (minutes) ratiometric gradients were observed in vitro radiating outward from the center of a dense adherent cell grouping on scaffolds. Sensor activation in ischemia and cell transplant models in vivo show oxygenation decreases on the scale of minutes. The nanofiber construct offers a robust approach to biomaterial scaffold oxygen sensing.


Subject(s)
Boron/chemistry , Coloring Agents/chemistry , Nanofibers/chemistry , Nanotechnology/instrumentation , Oxygen/metabolism , Polyesters/chemistry , Animals , Cell Line , Islets of Langerhans/cytology , Lactic Acid/chemistry , Mice , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Prostheses and Implants , Spatio-Temporal Analysis , Tissue Engineering , Tissue Scaffolds/chemistry
17.
Acta Biomater ; 10(11): 4704-4714, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25128750

ABSTRACT

Biomaterial-mediated controlled release of soluble signaling molecules is a tissue engineering approach to spatially control processes of inflammation, microvascular remodeling and host cell recruitment, and to generate biochemical gradients in vivo. Lipid mediators, such as sphingosine 1-phosphate (S1P), are recognized for their essential roles in spatial guidance, signaling and highly regulated endogenous gradients. S1P and pharmacological analogs such as FTY720 are therapeutically attractive targets for their critical roles in the trafficking of cells between blood and tissue spaces, both physiologically and pathophysiologically. However, the interaction of locally delivered sphingolipids with the complex metabolic networks controlling the flux of lipid species in inflamed tissue has yet to be elucidated. In this study, complementary in vitro and in vivo approaches are investigated to identify relationships between polymer composition, drug release kinetics, S1P metabolic activity, signaling gradients and spatial positioning of circulating cells around poly(lactic-co-glycolic acid) biomaterials. Results demonstrate that biomaterial-based gradients of S1P are short-lived in the tissue due to degradation by S1P lyase, an enzyme that irreversibly degrades intracellular S1P. On the other hand, in vivo gradients of the more stable compound, FTY720, enhance microvascular remodeling by selectively recruiting an anti-inflammatory subset of monocytes (S1P3(high)) to the biomaterial. Results highlight the need to better understand the endogenous balance of lipid import/export machinery and lipid kinase/phosphatase activity in order to design biomaterial products that spatially control the innate immune environment to maximize regenerative potential.


Subject(s)
Inflammation/pathology , Microvessels/pathology , Microvessels/physiopathology , Receptors, Lysosphingolipid/metabolism , Tissue Engineering/methods , Vascular Remodeling , Animals , Fingolimod Hydrochloride , Kinetics , Lactic Acid/chemistry , Ligands , Lymphocytes/drug effects , Lysophospholipids , Male , Mice, Inbred C57BL , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Propylene Glycols , Prostheses and Implants , Receptors, Lysosphingolipid/agonists , Sphingolipids/metabolism , Sphingosine/analogs & derivatives
18.
Proc Natl Acad Sci U S A ; 110(34): 13785-90, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23918395

ABSTRACT

Endothelial cells play significant roles in conditioning tissues after injury by the production and secretion of angiocrine factors. At least two distinct subsets of monocytes, CD45(+)CD11b(+)Gr1(+)Ly6C(+) inflammatory and CD45(+)CD11b(+)Gr1(-)Ly6C(-) anti-inflammatory monocytes, respond differentially to these angiocrine factors and promote pathogen/debris clearance and arteriogenesis/tissue regeneration, respectively. We demonstrate here that local sphingosine 1-phosphate receptor 3 (S1P3) agonism recruits anti-inflammatory monocytes to remodeling vessels. Poly(lactic-co-glycolic acid) thin films were used to deliver FTY720, an S1P1/3 agonist, to inflamed and ischemic tissues, which resulted in a reduction in proinflammatory cytokine secretion and an increase in regenerative cytokine secretion. The altered balance of cytokine secretion results in preferential recruitment of anti-inflammatory monocytes from circulation. The chemotaxis of these cells, which express more S1P3 than inflammatory monocytes, toward SDF-1α was also enhanced with FTY720 treatment, but not in S1P3 knockout cells. FTY720 delivery enhanced arteriolar diameter expansion and increased length density of the local vasculature. This work establishes a role for S1P receptor signaling in the local conditioning of tissues by angiocrine factors that preferentially recruit regenerative monocytes that can enhance healing outcomes, tissue regeneration, and biomaterial implant functionality.


Subject(s)
Monocytes/physiology , Neovascularization, Physiologic/physiology , Propylene Glycols/pharmacology , Prostheses and Implants/adverse effects , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Tissue Engineering/methods , Vascular System Injuries/drug therapy , Analysis of Variance , Animals , Blotting, Western , Cells, Cultured , Chemotaxis/drug effects , Cytokines/metabolism , DNA Primers/genetics , Drug Carriers , Fingolimod Hydrochloride , Flow Cytometry , Humans , Immunohistochemistry , Lactic Acid , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microvessels/cytology , Monocytes/drug effects , Neovascularization, Physiologic/drug effects , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Propylene Glycols/administration & dosage , Real-Time Polymerase Chain Reaction , Receptors, Lysosphingolipid/agonists , Sphingosine/administration & dosage , Sphingosine/pharmacology , Vascular System Injuries/etiology
19.
Curr Pharm Des ; 19(19): 3403-19, 2013.
Article in English | MEDLINE | ID: mdl-23432670

ABSTRACT

Regeneration of bone requires the coordinated processes of angiogenesis and osteogenesis. These repair mechanisms are closely linked through both direct cell-cell contact and indirect paracrine signaling among osteoblasts, endothelial cells, and other cell types. The vasculature provides a source of nutrients, oxygen, metabolic substrates, and access for circulating cells that help to support new bone formation. The complexity of the endogenous signaling axis that promotes angiogenesis provides numerous opportunities for therapeutic intervention ranging from progenitor cell mobilization to endothelial proliferation and sprouting. Small molecules are particularly appealing for regenerative medicine applications because many exhibit extended in vivo stability, low cost, and scalable production. Innovative techniques for developing small molecules such as high throughput functional assays and broad-spectrum database analysis techniques have led to the development of new compounds and the identification of novel applications of existing drugs. In addition, rapid advances in biomaterials design and synthesis provide platforms to deliver therapeutic small molecules to sites of bone injury. This review presents an overview of current strategies for harnessing endogenous healing mechanisms using small molecules by targeting angiogenesis, osteogenesis, or both.


Subject(s)
Biological Products/pharmacology , Bone Regeneration/drug effects , Neovascularization, Physiologic/drug effects , Small Molecule Libraries/pharmacology , Animals , Biological Products/administration & dosage , Biological Products/chemistry , Biological Products/therapeutic use , Bone and Bones/blood supply , Fractures, Bone/drug therapy , Humans , Molecular Structure , Musculoskeletal Diseases/drug therapy , Osteogenesis/drug effects , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use
20.
Differentiation ; 86(4-5): 149-58, 2013.
Article in English | MEDLINE | ID: mdl-24480155

ABSTRACT

Embryonic stem (ES) cells and induced pluripotent stem (iPS) cells hold great promise in regenerative medicine for the treatment of neurodegenerative diseases. Current neuronal differentiation protocols however, are not optimized yet for the high scale production of neural precursors and terminally differentiated neurons. The present investigation reports a novel technique for the scalable production of highly uniformed neurospheres, neural precursors and terminal neurons from mouse ES and iPS cells using retinoic acid and a mechanical rotation procedure. We compared embryoid bodies (EB) and neurosphere morphology, yield of neural precursors and quality of neurons between rotary and static suspension cultures of mouse ES and iPS cells undergoing neural differentiation. Analysis of neurospheres formed under continuous rotation showed increased neurosphere uniformity and a high yield of neural precursors after neurosphere dissociation. Neurospheres formed under rotation conditions were relatively smaller, more uniform and had less dead cells and higher proliferation compared to those formed under static conditions. Neural precursors under rotation conditions matured faster, survived better, differentiated to functional neurons that stained positively for mature neuronal markers, and fired action potentials similar to the statically cultured neurons. This report thus provides a technique for the scalable production of neurons from ES and iPS cells and we suggest that rotation culture procedure can be a routine technique for stem cell neural and neuronal differentiation.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Animals , Cell Culture Techniques/methods , Mice , Regenerative Medicine , Stem Cells , Tretinoin/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...