Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
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
2.
Arterioscler Thromb Vasc Biol ; 40(5): 1220-1230, 2020 05.
Article in English | MEDLINE | ID: mdl-32160775

ABSTRACT

OBJECTIVE: Sickle cell anemia (SCA) causes chronic inflammation and multiorgan damage. Less understood are the arterial complications, most evident by increased strokes among children. Proteolytic mechanisms, biomechanical consequences, and pharmaceutical inhibitory strategies were studied in a mouse model to provide a platform for mechanistic and intervention studies of large artery damage due to sickle cell disease. Approach and Results: Townes humanized transgenic mouse model of SCA was used to test the hypothesis that elastic lamina and structural damage in carotid arteries increased with age and was accelerated in mice homozygous for SCA (sickle cell anemia homozygous genotype [SS]) due to inflammatory signaling pathways activating proteolytic enzymes. Elastic lamina fragmentation observed by 1 month in SS mice compared with heterozygous littermate controls (sickle cell trait heterozygous genotype [AS]). Positive immunostaining for cathepsin K, a powerful collagenase and elastase, confirmed accelerated proteolytic activity in SS carotids. Larger cross-sectional areas were quantified by magnetic resonance angiography and increased arterial compliance in SS carotids were also measured. Inhibiting JNK (c-jun N-terminal kinase) signaling with SP600125 significantly reduced cathepsin K expression, elastin fragmentation, and carotid artery perimeters in SS mice. By 5 months of age, continued medial thinning and collagen degradation was mitigated by treatment of SS mice with JNK inhibitor. CONCLUSIONS: Arterial remodeling due to SCA is mediated by JNK signaling, cathepsin proteolytic upregulation, and degradation of elastin and collagen. Demonstration in Townes mice establishes their utility for mechanistic studies of arterial vasculopathy, related complications, and therapeutic interventions for large artery damage due to SCA.


Subject(s)
Anemia, Sickle Cell/drug therapy , Anthracenes/pharmacology , Carotid Arteries/drug effects , Carotid Artery Diseases/prevention & control , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Vascular Remodeling/drug effects , Anemia, Sickle Cell/enzymology , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/physiopathology , Animals , Carotid Arteries/enzymology , Carotid Arteries/physiopathology , Carotid Artery Diseases/enzymology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/physiopathology , Cathepsin K/metabolism , Collagen/metabolism , Disease Models, Animal , Elastin/metabolism , Hemoglobins/genetics , Homozygote , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice, Transgenic , Mutation , Proteolysis , Signal Transduction , Time Factors
3.
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
4.
Blood ; 124(12): 1941-50, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25075126

ABSTRACT

Sphingolipids are a class of lipids containing a backbone of sphingoid bases that can be produced de novo through the reaction of palmitate and serine and further metabolized through the activity of various enzymes to produce intermediates with diverse roles in cellular processes and signal transduction. One of these intermediates, sphingosine 1-phosphate (S1P), is stored at high concentrations (1 µM) in red blood cells (RBCs) and directs a wide array of cellular processes mediated by 5 known G-protein coupled receptors (S1P1-S1P5). In this study, we show that RBC membrane alterations in sickle cell disease enhance the activation acid sphingomyelinase by 13%, resulting in increased production and storage of sphingosine (2.6-fold) and S1P (3.5-fold). We also show that acid sphingomyelinase enhances RBC-derived microparticle (MP) generation. These MPs are internalized by myeloid cells and promote proinflammatory cytokine secretion and endothelial cell adhesion, suggesting that potential crosstalk between circulating inflammatory cells and MPs may contribute to the inflammation-rooted pathogenesis of the disease. Treatment with amitriptyline reduces MP generation in vitro and in vivo and might be used to mitigate inflammatory processes in sickle cell disease.


Subject(s)
Anemia, Sickle Cell/blood , Anemia, Sickle Cell/enzymology , Cell-Derived Microparticles/enzymology , Erythrocytes, Abnormal/enzymology , Sphingomyelin Phosphodiesterase/blood , Anemia, Sickle Cell/etiology , Animals , Cell Adhesion , Cell Adhesion Molecules/blood , Disease Models, Animal , Endothelial Cells/pathology , Hemoglobin, Sickle/genetics , Humans , Inflammation/blood , Inflammation/enzymology , Lysophospholipids/blood , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Sphingolipids/blood , Sphingosine/analogs & derivatives , Sphingosine/blood
5.
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
6.
Tissue Eng Part A ; 30(1-2): 45-60, 2024 01.
Article in English | MEDLINE | ID: mdl-37897061

ABSTRACT

Rotator cuff tear is a significant problem that leads to poor clinical outcomes due to muscle degeneration after injury. The objective of this study was to synergistically increase the number of proregenerative cells recruited to injure rotator cuff muscle through a novel dual treatment system, consisting of a bone marrow mobilizing agent (VPC01091), hypothesized to "push" prohealing cells into the blood, and localized delivery of stromal cell-derived factor-1α (SDF-1α), to "pull" the cells to the injury site. Immediately after rotator cuff tendon injury in rat, the mobilizing agent was delivered systemically, and SDF-1α-loaded heparin-based microparticles were injected into the supraspinatus muscle. Regenerative and degenerative changes to supraspinatus muscle and the presence of inflammatory/immune cells, mesenchymal stem cells (MSCs), and satellite cells were assessed via flow cytometry and histology for up to 21 days. After dual treatment, significantly more MSCs (31.9 ± 8.0% single cells) and T lymphocytes (6.7 ± 4.3 per 20 × field of view) were observed in supraspinatus muscle 7 days after injury and treatment compared to injury alone (14.4 ± 6.5% single cells, 1.2 ± 0.7 per 20 × field of view), in addition to an elevated M2:M1 macrophage ratio (3.0 ± 0.5), an indicator of a proregenerative environment. These proregenerative cellular changes were accompanied by increased nascent fiber formation (indicated by embryonic myosin heavy chain staining) at day 7 compared to SDF-1α treatment alone, suggesting that this method may be a promising strategy to influence the early cellular response in muscle and promote a proregenerative microenvironment to increase muscle healing after severe rotator cuff tear.


Subject(s)
Rotator Cuff Injuries , Rotator Cuff , Rats , Animals , Rotator Cuff/pathology , Rotator Cuff Injuries/therapy , Rotator Cuff Injuries/pathology , Chemokine CXCL12/pharmacology , Bone Marrow , Muscle Fibers, Skeletal
7.
bioRxiv ; 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38328164

ABSTRACT

Cognitive deficit is a debilitating complication of SCD with multifactorial pathobiology. Here we show that neuroinflammation and dysregulation in lipidomics and transcriptomics profiles are major underlying mechanisms of social stress-induced cognitive deficit in SCD. Townes sickle cell (SS) mice and controls (AA) were exposed to social stress using the repeat social defeat (RSD) paradigm concurrently with or without treatment with minocycline. Mice were tested for cognitive deficit using novel object recognition (NOR) and fear conditioning (FC) tests. SS mice exposed to RSD without treatment had worse performance on cognitive tests compared to SS mice exposed to RSD with treatment or to AA controls, irrespective of their RSD or treatment disposition. Additionally, compared to SS mice exposed to RSD with treatment, SS mice exposed to RSD without treatment had significantly more cellular evidence of neuroinflammation coupled with a significant shift in the differentiation of neural progenitor cells towards astrogliogenesis. Additionally, brain tissue from SS mice exposed to RSD was significantly enriched for genes associated with blood-brain barrier dysfunction, neuron excitotoxicity, inflammation, and significant dysregulation in sphingolipids important to neuronal cell processes. We demonstrate in this study that neuroinflammation and lipid dysregulation are potential underlying mechanisms of social stress-related cognitive deficit in SS mice.

8.
Commun Biol ; 6(1): 749, 2023 07 19.
Article in English | MEDLINE | ID: mdl-37468760

ABSTRACT

Volumetric muscle loss (VML) results in permanent functional deficits and remains a substantial regenerative medicine challenge. A coordinated immune response is crucial for timely myofiber regeneration, however the immune response following VML has yet to be fully characterized. Here, we leveraged dimensionality reduction and pseudo-time analysis techniques to elucidate the cellular players underlying a functional or pathological outcome as a result of subcritical injury or critical VML in the murine quadriceps, respectively. We found that critical VML resulted in a sustained presence of M2-like and CD206hiLy6Chi 'hybrid' macrophages whereas subcritical defects resolved these populations. Notably, the retained M2-like macrophages from critical VML injuries presented with aberrant cytokine production which may contribute to fibrogenesis, as indicated by their co-localization with fibroadipogenic progenitors (FAPs) in areas of collagen deposition within the defect. Furthermore, several T cell subpopulations were significantly elevated in critical VML compared to subcritical injuries. These results demonstrate a dysregulated immune response in critical VML that is unable to fully resolve the chronic inflammatory state and transition to a pro-regenerative microenvironment within the first week after injury. These data provide important insights into potential therapeutic strategies which could reduce the immune cell burden and pro-fibrotic signaling characteristic of VML.


Subject(s)
Muscle, Skeletal , Muscular Diseases , Mice , Animals , Muscle, Skeletal/pathology , Regeneration , Muscular Diseases/pathology , Muscular Diseases/therapy , Regenerative Medicine , Collagen
9.
bioRxiv ; 2023 Dec 23.
Article in English | MEDLINE | ID: mdl-38187740

ABSTRACT

Orofacial clefts are the most common craniofacial congenital anomaly. Following cleft palate repair, up to 60% of surgeries have wound healing complications leading to oronasal fistula (ONF), a persistent connection between the roof of the mouth and the nasal cavity. The current gold standard methods for ONF repair use human allograft tissues; however, these procedures have risks of graft infection and/or rejection, requiring surgical revisions. Immunoregenerative therapies present a novel alternative approach to harness the body's immune response and enhance the wound healing environment. We utilized a repurposed FDA-approved immunomodulatory drug, FTY720, to reduce the egress of lymphocytes and induce immune cell fate switching toward pro-regenerative phenotypes. Here, we engineered a bilayer biomaterial system using Tegaderm™, a liquid-impermeable wound dressing, to secure and control the delivery of FTY720- nanofiber scaffolds (FTY720-NF). We optimized release kinetics of the bilayer FTY720-NF to sustain drug release for up to 7d with safe, efficacious transdermal absorption and tissue biodistribution. Through comprehensive immunophenotyping, our results illustrate a pseudotime pro-regenerative state transition in recruited hybrid immune cells to the wound site. Additional histological assessments established a significant difference in full thickness ONF closure in mice on Day 7 following treatment with bilayer FTY720-NF, compared to controls. These findings demonstrate the utility of immunomodulatory strategies for oral wound healing, better positing the field to develop more efficacious treatment options for pediatric patients. One Sentence Summary: Local delivery of bilayer FTY720-nanofiber scaffolds in an ONF mouse model promotes complete wound closure through modulation of pro-regenerative immune and stromal cells.

10.
Cell Tissue Res ; 347(3): 553-66, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21863314

ABSTRACT

Endogenous stem cell recruitment to the site of skeletal injury is key to enhanced osseous remodeling and neovascularization. To this end, this study utilized a novel bone allograft coating of poly(lactic-co-glycolic acid) (PLAGA) to sustain the release of FTY720, a selective agonist for sphingosine 1-phosphate (S1P) receptors, from calvarial allografts. Uncoated allografts, vehicle-coated, low dose FTY720 in PLAGA (1:200 w:w) and high dose FTY720 in PLAGA (1:40) were implanted into critical size calvarial bone defects. The ability of local FTY720 delivery to promote angiogenesis, maximize osteoinductivity and improve allograft incorporation by recruitment of bone progenitor cells from surrounding soft tissues and microcirculation was evaluated. FTY720 bioactivity after encapsulation and release was confirmed with sphingosine kinase 2 assays. HPLC-MS quantified about 50% loaded FTY720 release of the total encapsulated drug (4.5 µg) after 5 days. Following 2 weeks of defect healing, FTY720 delivery led to statistically significant increases in bone volumes compared to controls, with total bone volume increases for uncoated, coated, low FTY720 and high FTY720 of 5.98, 3.38, 7.2 and 8.9 mm(3), respectively. The rate and extent of enhanced bone growth persisted through week 4 but, by week 8, increases in bone formation in FTY720 groups were no longer statistically significant. However, micro-computed tomography (microCT) of contrast enhanced vascular ingrowth (MICROFIL®) and histological analysis showed enhanced integration as well as directed bone growth in both high and low dose FTY720 groups compared to controls.


Subject(s)
Bone Transplantation , Drug Delivery Systems/methods , Osteogenesis/drug effects , Propylene Glycols/administration & dosage , Propylene Glycols/pharmacology , Skull/drug effects , Sphingosine/analogs & derivatives , Actins/metabolism , Animals , Blood Vessels/drug effects , Blood Vessels/growth & development , Bone Remodeling/drug effects , Coated Materials, Biocompatible/chemistry , Female , Fingolimod Hydrochloride , Fluorescence , Lactic Acid/chemistry , Monocytes/drug effects , Monocytes/metabolism , Periosteum/blood supply , Periosteum/diagnostic imaging , Periosteum/drug effects , Periosteum/growth & development , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley , Skull/blood supply , Skull/pathology , Skull/surgery , Sphingosine/administration & dosage , Sphingosine/pharmacology , Transplantation, Homologous , X-Ray Microtomography
11.
Exp Biol Med (Maywood) ; 247(4): 368-374, 2022 02.
Article in English | MEDLINE | ID: mdl-35172625

ABSTRACT

Sickle cell disease (SCD) is characterized by vaso-occlusion, hemolysis, and systemic manifestations that form the hallmark of the disease. Apart from morbidity, SCD is also associated with increased mortality and decreased quality of life. Aging is a natural phenomenon that is associated with changes at cellular, tissue, and organ levels, in addition to the loss of physical fitness, increased susceptibility to diseases, and a higher likelihood of mortality. Some of the cellular mechanisms involved in normal (or physiological) aging include abnormalities of sphingolipids (ceramides) and reduced length of the telomere. These changes have also been documented in SCD. Cellular, organs, and physical manifestations of SCD resemble an accelerated aging syndrome. Sickle erythrocytes also acquire morphological features similar to that of aged normal erythrocytes and are thus picked up early by the macrophages for destruction. Brain, kidney, heart, innate and adaptive immune system, and musculoskeletal system of patients with SCD exhibit morphological and functional changes that are ordinarily seen in the elderly in the general population. Stroke, silent cerebral infarcts, cardiomegaly, heart failure, pulmonary hypertension, nephropathy with proteinuria, osteopenia, osteoporosis, osteonecrosis, gout, and infections are exceedingly common in SCD. In this review, we have attempted to draw parallels between SCD and accelerated aging syndromes.


Subject(s)
Anemia, Sickle Cell , Stroke , Aged , Aging , Anemia, Sickle Cell/complications , Humans , Quality of Life , Syndrome
12.
Blood Adv ; 6(5): 1381-1393, 2022 03 08.
Article in English | MEDLINE | ID: mdl-34547771

ABSTRACT

Sickle cell disease (SCD) is the most common hereditary blood disorder in the United States. SCD is frequently associated with osteonecrosis, osteoporosis, osteopenia, and other bone-related complications such as vaso-occlusive pain, ischemic damage, osteomyelitis, and bone marrow hyperplasia known as sickle bone disease (SBD). Previous SBD models have failed to distinguish the age- and sex-specific characteristics of bone morphometry. In this study, we use the Townes mouse model of SCD to assess the pathophysiological complications of SBD in both SCD and sickle cell trait. Changes in bone microarchitecture and bone development were assessed by using high-resolution quantitative micro-computed tomography and the three-dimensional reconstruction of femurs from male and female mice. Our results indicate that SCD causes bone loss and sex-dependent anatomical changes in bone. SCD female mice in particular are prone to trabecular bone loss, whereas cortical bone degradation occurs in both sexes. We also describe the impact of genetic knockdown of cathepsin K- and E-64-mediated cathepsin inhibition on SBD.


Subject(s)
Anemia, Sickle Cell , Bone Diseases, Metabolic , Osteoporosis , Anemia, Sickle Cell/pathology , Animals , Bone Diseases, Metabolic/etiology , Disease Models, Animal , Female , Male , Mice , Osteoporosis/etiology , X-Ray Microtomography
13.
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
14.
Adv Mater ; 34(10): e2108084, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34989049

ABSTRACT

Immunotherapy has emerged as one of the most powerful anti-cancer therapies but is stymied by the limits of existing preclinical models with respect to disease latency and reproducibility. Additionally, the influence of differing immune microenvironments within tumors observed clinically and associated with immunotherapeutic resistance cannot be tuned to facilitate drug testing workflows without changing model system or laborious genetic approaches. To address this testing platform gap in the immune oncology drug development pipeline, the authors deploy engineered biomaterials as scaffolds to increase tumor formation rate, decrease disease latency, and diminish variability of immune infiltrates into tumors formed from murine mammary carcinoma cell lines implanted into syngeneic mice. By altering synthetic gel formulations that reshape infiltrating immune cells within the tumor, responsiveness of the same tumor model to varying classes of cancer immunotherapies, including in situ vaccination with a molecular adjuvant and immune checkpoint blockade, diverge. These results demonstrate the significant role the local immune microenvironment plays in immunotherapeutic response. These engineered tumor immune microenvironments therefore improve upon the limitations of current breast tumor models used for immune oncology drug screening to enable immunotherapeutic testing relevant to the variability in tumor immune microenvironments underlying immunotherapeutic resistance seen in human patients.


Subject(s)
Immunotherapy , Neoplasms , Animals , Humans , Immunologic Factors/therapeutic use , Mice , Neoplasms/therapy , Reproducibility of Results , Tumor Microenvironment
15.
Clin Orthop Relat Res ; 469(9): 2665-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21161735

ABSTRACT

BACKGROUND: Osteonecrosis (ON) of the femoral head is a devastating disease affecting young patients at their most productive age, causing major socioeconomic burdens. ON is associated with various etiologic factors, and the pathogenesis of the disease is unknown. Most investigators believe the disease is the result of secondary microvascular compromise with subsequent bone and marrow cell death and defective bone repair. QUESTIONS/HYPOTHESES: We hypothesize that local delivery of vascular endothelial growth factor (VEGF) and bone morphogenetic protein-6 (BMP-6), which induces angiogenesis and osteogenesis respectively, will reverse the disease process and provide a treatment for precollapse ON. METHOD OF STUDY: We will use genetically engineered bone marrow stem cells, carrying VEGF and BMP-6 genes, to enhance angiogenesis and osteogenesis in necrotic bone of an animal model, by local delivery of growth factor in addition to the bone-forming property of the stem cells. The participation, localization, and fate of the stem cells in the repair process will be evaluated by tracing marker-gene product. Osteogenesis and angiogenesis will be assessed using high-resolution xray CT and immunohistomorphometry quantitatively. Mechanical properties of the repair tissue will be determined using an indentation test of the femoral head. SIGNIFICANCE: We envision that a deliverable or injectable bone graft substitute containing engineered stem cells and therapeutic growth factors will be developed through this proposed study and will provide a much needed treatment for ON.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Bone Morphogenetic Protein 6/biosynthesis , Femur Head Necrosis/therapy , Genetic Therapy , Stem Cell Transplantation , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Bone Morphogenetic Protein 6/genetics , Bone Regeneration , Cell Tracking , Chickens , Disease Models, Animal , Femur Head Necrosis/genetics , Femur Head Necrosis/metabolism , Femur Head Necrosis/pathology , Immunohistochemistry , Neovascularization, Physiologic , Osteogenesis , Research Design , Time Factors , Tomography, X-Ray Computed , Treatment Outcome , Vascular Endothelial Growth Factor A/genetics
16.
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
17.
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
18.
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
19.
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.

20.
Nat Commun ; 11(1): 114, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31913286

ABSTRACT

Stem cell therapies are limited by poor cell survival and engraftment. A hurdle to the use of materials for cell delivery is the lack of understanding of material properties that govern transplanted stem cell functionality. Here, we show that synthetic hydrogels presenting integrin-specific peptides enhance the survival, persistence, and osteo-reparative functions of human bone marrow-derived mesenchymal stem cells (hMSCs) transplanted in murine bone defects. Integrin-specific hydrogels regulate hMSC adhesion, paracrine signaling, and osteoblastic differentiation in vitro. Hydrogels presenting GFOGER, a peptide targeting α2ß1 integrin, prolong hMSC survival and engraftment in a segmental bone defect and result in improved bone repair compared to other peptides. Integrin-specific hydrogels have diverse pleiotropic effects on hMSC reparative activities, modulating in vitro cytokine secretion and in vivo gene expression for effectors associated with inflammation, vascularization, and bone formation. These results demonstrate that integrin-specific hydrogels improve tissue healing by directing hMSC survival, engraftment, and reparative activities.


Subject(s)
Bone Diseases/therapy , Integrin alpha2beta1/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Bone Diseases/metabolism , Bone Diseases/physiopathology , Bone Marrow/chemistry , Bone Marrow/metabolism , Bone Regeneration , Cell Adhesion , Cell Survival , Cell- and Tissue-Based Therapy , Humans , Hydrogels/chemistry , Integrin alpha2beta1/genetics , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Peptides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL