Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
Add more filters










Publication year range
1.
Arterioscler Thromb Vasc Biol ; 42(7): 868-883, 2022 07.
Article in English | MEDLINE | ID: mdl-35510552

ABSTRACT

BACKGROUND: Arteriovenous fistulae (AVF) are the gold standard for vascular access for hemodialysis. Although the vein must thicken and dilate for successful hemodialysis, excessive wall thickness leads to stenosis causing AVF failure. Since TGF-ß (transforming growth factor-beta) regulates ECM (extracellular matrix) deposition and smooth muscle cell (SMC) proliferation-critical components of wall thickness-we hypothesized that disruption of TGF-ß signaling prevents excessive wall thickening during venous remodeling. METHODS: A mouse aortocaval fistula model was used. SB431542-an inhibitor of TGF-ß receptor I-was encapsulated in nanoparticles and applied to the AVF adventitia in C57BL/6J mice. Alternatively, AVFs were created in mice with conditional disruption of TGF-ß receptors in either SMCs or endothelial cells. Doppler ultrasound was performed serially to confirm patency and to measure vessel diameters. AVFs were harvested at predetermined time points for histological and immunofluorescence analyses. RESULTS: Inhibition of TGF-ß signaling with SB431542-containing nanoparticles significantly reduced p-Smad2-positive cells in the AVF wall during the early maturation phase (days 7-21) and was associated with decreased AVF wall thickness that showed both decreased collagen density and decreased SMC proliferation. SMC-specific TGF-ß signaling disruption decreased collagen density but not SMC proliferation or wall thickness. Endothelial cell-specific TGF-ß signaling disruption decreased both collagen density and SMC proliferation in the AVF wall and was associated with reduced wall thickness, increased outward remodeling, and improved AVF patency. CONCLUSIONS: Endothelial cell-targeted TGF-ß inhibition may be a translational strategy to improve AVF patency.


Subject(s)
Arteriovenous Fistula , Arteriovenous Shunt, Surgical , Animals , Collagen , Disease Models, Animal , Endothelial Cells , Mice , Mice, Inbred C57BL , Transforming Growth Factor beta , Transforming Growth Factors , Vascular Remodeling/physiology
2.
Nanomedicine ; 40: 102477, 2022 02.
Article in English | MEDLINE | ID: mdl-34740868

ABSTRACT

Magnetic resonance is a key imaging tool for the detection of prostate cancer; however, better tools focusing on cancer specificity are required to distinguish benign from cancerous regions. We found higher expression of claudin-3 (CLDN-3) and -4 (CLDN-4) in higher grade than lower-grade human prostate cancer biopsies (n = 174), leading to the design of functionalized nanoparticles (NPs) with a non-toxic truncated version of the natural ligand Clostridium perfringens enterotoxin (C-CPE) that has a strong binding affinity to Cldn-3 and Cldn-4 receptors. We developed a first-of-its-type, C-CPE-NP-based MRI detection tool in a prostate tumor-bearing mouse model. NPs with an average diameter of 152.9 ±â€¯15.7 nm (RS1) had a 2-fold enhancement of tumor specificity compared to larger (421.2 ±â€¯33.8 nm) NPs (RS4). There was a 1.8-fold (P < 0.01) and 1.6-fold (P < 0.01) upregulation of the tumor-to-liver signal intensities of C-RS1 and C-RS4 (functionalized NPs) compared to controls, respectively. Also, tumor specificity was 3.1-fold higher (P < 0.001) when comparing C-RS1 to C-RS4. This detection tool improved tumor localization of contrast-enhanced MRI, supporting potential clinical applicability.


Subject(s)
Nanoparticles , Prostatic Neoplasms , Animals , Enterotoxins/metabolism , Humans , Magnetic Resonance Imaging , Male , Mice , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/metabolism
4.
Nat Biomed Eng ; 5(9): 983-997, 2021 09.
Article in English | MEDLINE | ID: mdl-34616050

ABSTRACT

Oral formulations of insulin are typically designed to improve its intestinal absorption and increase its blood bioavailability. Here we show that polymerized ursodeoxycholic acid, selected from a panel of bile-acid polymers and formulated into nanoparticles for the oral delivery of insulin, restored blood-glucose levels in mice and pigs with established type 1 diabetes. The nanoparticles functioned as a protective insulin carrier and as a high-avidity bile-acid-receptor agonist, increased the intestinal absorption of insulin, polarized intestinal macrophages towards the M2 phenotype, and preferentially accumulated in the pancreas of the mice, binding to the islet-cell bile-acid membrane receptor TGR5 with high avidity and activating the secretion of glucagon-like peptide and of endogenous insulin. In the mice, the nanoparticles also reversed inflammation, restored metabolic functions and extended animal survival. When encapsulating rapamycin, they delayed the onset of diabetes in mice with chemically induced pancreatic inflammation. The metabolic and immunomodulatory functions of ingestible bile-acid-polymer nanocarriers may offer translational opportunities for the prevention and treatment of type 1 diabetes.


Subject(s)
Bile Acids and Salts , Diabetes Mellitus, Type 1 , Animals , Bile , Diabetes Mellitus, Type 1/drug therapy , Glucagon-Like Peptide 1 , Insulin , Mice , Polymers , Receptors, G-Protein-Coupled , Sirolimus , Swine
5.
Mol Pharm ; 18(3): 850-861, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33428414

ABSTRACT

Targeting different cell surface receptors with nanoparticle (NP)-based platforms can result in differential particle binding properties that may impact their localization, bioavailability, and, ultimately, the therapeutic efficacy of an encapsulated payload. Conventional in vitro assays comparing the efficacy of targeted NPs often do not adequately control for these differences in particle-receptor binding, potentially confounding their therapeutic readouts and possibly even limiting their experimental value. In this work, we characterize the conditions under which NPs loaded with Bruton's Tyrosine Kinase (BTK) inhibitor differentially suppress primary B cell activation when targeting either CD19 (internalizing) or B220 (noninternalizing) surface receptors. Surface binding of fluorescently labeled CD19- and B220-targeted NPs was analyzed and quantitatively correlated with the number of bound particles at given treatment concentrations. Using this binding data, suppression of B cell activation was directly compared for differentially targeted (CD19 vs B220) NPs loaded with a BTK inhibitor at a range of particle drug loading concentrations. When NPs were loaded with lower amounts of drug, CD19-mediated internalization demonstrated increased inhibition of B cell proliferation compared with B220 NPs. However, these differences were mitigated when particles were loaded with higher concentrations of BTK inhibitor and B220-mediated "paracrine-like" delivery demonstrated superior suppression of cellular activation when cells were bound to lower overall numbers of NPs. Taken together, these results demonstrate that inhibition of B cell activation can be optimized for NPs targeting either internalizing or noninternalizing surface receptors and that particle internalization is likely not a requisite endpoint when designing particles for delivery of BTK inhibitor to B cells.


Subject(s)
B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Nanoparticles/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Receptors, Antigen, B-Cell/metabolism , Receptors, Cell Surface/metabolism , Animals , Antigens, CD19/metabolism , Cell Proliferation/drug effects , Female , Leukocyte Common Antigens/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects
6.
J Colloid Interface Sci ; 587: 101-109, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33360882

ABSTRACT

The unique capability of fullerene (C60) to absorb light and generate reactive oxygen species (ROS) has been extensively studied for photosensitized water treatment and cancer therapy. Various material synthesis strategies have been proposed in parallel to overcome its intrinsic hydrophobicity and to enhance availability in water and physiological media. We present here a strikingly simple approach to make C60 available to these applications by hand-grinding dry C60 powder with nanodiamond (ND) using a mortar and pestle. The resulting ND-C60 composite was found to form a stable aqueous colloidal suspension and efficiently drive photosensitized production of ROS under visible light illumination. ND-C60 rapidly adsorbed and oxidized organic contaminants by photogenerated ROS. In the experiments for photodynamic cancer therapy, ND-C60 was internalized by cancer cells and induced cell apoptosis without noticeable toxicity. Treatment of tumor-bearing mice with ND-C60 and light irradiation resulted in tumor shrinkage and prolonged survival time.


Subject(s)
Fullerenes , Nanodiamonds , Neoplasms , Photochemotherapy , Water Purification , Animals , Mice , Neoplasms/drug therapy
7.
Front Med Technol ; 3: 640569, 2021.
Article in English | MEDLINE | ID: mdl-35047909

ABSTRACT

Multiple sclerosis (MS) is a demyelinating autoimmune disease that attacks the brain, with year-on-year loss of brain volume, starting late teens and becoming manifest late twenties. There is no cure, and current therapies are immunosuppressive only. LIF is a vital stem cell growth factor active throughout life-and essential for health of the central nervous system (CNS), being tolerogenic, myelinogenic, and neuroprotective. Nano-formulation of LIF (LIFNano) using FDA-approved PLGA captures LIF's compound therapeutic properties, increasing potency 1,000-fold when targeted to CD4 (LIFNano-CD4). Moreover, circulating CD4+ lymphocytes are themselves regulated by LIF to express the Treg phenotype, known to release T cell-derived LIF upon engagement with cognate antigen, perpetuating antigen-specific self-tolerance. With the longer-term aim of treating inflammatory lesions of MS, we asked, does LIFNano-CD4 cross the blood-brain barrier (BBB)? We measure pK and pD using novel methodologies, demonstrate crossing of the BBB, show LIF-cargo-specific anti-inflammatory efficacy in the frontal cortex of the brain, and show safety of intravenous delivery of LIFNano-CD4 at doses known to provide efficacious concentrations of LIF cargo behind the BBB.

8.
ACS Nano ; 14(7): 8646-8657, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32530598

ABSTRACT

The sensitivity and speed with which the immune system reacts to host disruption is unrivaled by any detection method for pathogenic biomarkers or infectious signatures. Engagement of cellular immunity in response to infections or cancer is contingent upon activation and subsequent cytotoxic activity by T cells. Thus, monitoring T cell activation can reliably serve as a metric for disease diagnosis as well as therapeutic prognosis. Rapid and direct quantification of T cell activation states, however, has been hindered by challenges associated with antigen target identification, labeling requirements, and assay duration. Here we present an electronic, label-free method for simultaneous separation and evaluation of T cell activation states. Our device utilizes a microfluidic design integrated with nanolayered electrode structures for dielectrophoresis (DEP)-driven discrimination of activated vs naïve T cells at single-cell resolution and demonstrates rapid (<2 min) separation of T cells at high single-pass efficiency as quantified by an on-chip Coulter counter module. Our device represents a microfluidic tool for electronic assessment of immune activation states and, hence, a portable diagnostic for quantitative evaluation of immunity and disease state. Further, its ability to achieve label-free enrichment of activated immune cells promises clinical utility in cell-based immunotherapies.


Subject(s)
Microfluidics , T-Lymphocytes , Biological Assay , Cell Separation , Electronics , Electrophoresis , Lymphocyte Activation
9.
Sci Adv ; 6(11): eaaz1580, 2020 03.
Article in English | MEDLINE | ID: mdl-32195350

ABSTRACT

Dendritic cells (DCs) are adept at cross-presentation and initiation of antigen-specific immunity. Clinically, however, DCs produced by in vitro differentiation of monocytes in the presence of exogenous cytokines have been met with limited success. We hypothesized that DCs produced in a physiological manner may be more effective and found that platelets activate a cross-presentation program in peripheral blood monocytes with rapid (18 hours) maturation into physiological DCs (phDCs). Differentiation of monocytes into phDCs was concomitant with the formation of an "adhesion synapse," a biophysical junction enriched with platelet P-selectin and monocyte P-selectin glycoprotein ligand 1, followed by intracellular calcium fluxing and nuclear localization of nuclear factor κB. phDCs were more efficient than cytokine-derived DCs in generating tumor-specific T cell immunity. Our findings demonstrate that platelets mediate a cytokine-independent, physiologic maturation of DC and suggest a novel strategy for DC-based immunotherapies.


Subject(s)
Antigen Presentation , Blood Platelets/immunology , Calcium Signaling/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Monocytes/immunology , P-Selectin/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Calcium Signaling/genetics , Cell Differentiation/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/immunology , P-Selectin/genetics , T-Lymphocytes/immunology
10.
Front Immunol ; 11: 583338, 2020.
Article in English | MEDLINE | ID: mdl-33391260

ABSTRACT

We recently reported that the treatment with nanoparticles (NPs) loaded with tolerogenic cytokines suppressed the manifestations of lupus-like disease induced by the transfer of donor CD4+ T cells from DBA/2 mice into (C57BL/6 × DBA/2)F1 (BDF1) mice. Although the protective effects were ascribed to the induction of adaptive CD4+ and CD8+ T regulatory cells, the results suggested that another population of immune cells could be involved. Here we report that NK cells critically contribute to the protection from lupus-like disease conferred by NPs to BDF1 mice, and that this effect is TGF-ß-dependent.


Subject(s)
CD2 Antigens/antagonists & inhibitors , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Lupus Erythematosus, Systemic/immunology , Transforming Growth Factor beta/immunology , Animals , Killer Cells, Natural/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Nanoparticles
11.
Trends Immunol ; 40(10): 888-908, 2019 10.
Article in English | MEDLINE | ID: mdl-31601519

ABSTRACT

During homeostasis, interactions between tolerogenic dendritic cells (DCs), self-reactive T cells, and T regulatory cells (Tregs) contribute to maintaining mammalian immune tolerance. In response to infection, immunogenic DCs promote the generation of proinflammatory effector T cell subsets. When complex homeostatic mechanisms maintaining the balance between regulatory and effector functions become impaired, autoimmune diseases can develop. We discuss some of the newest advances on the mechanisms of physiopathologic homeostasis that can be employed to develop strategies to restore a dysregulated immune equilibrium. Some of these designs are based on selectively activating regulators of immunity and inflammation instead of broadly suppressing these processes. Promising approaches include the use of nanoparticles (NPs) to restore Treg control over self-reactive cells, aiming to achieve long-term disease remission, and potentially to prevent autoimmunity in susceptible individuals.


Subject(s)
Autoimmune Diseases/immunology , Homeostasis/immunology , Animals , Autoimmunity/immunology , Dendritic Cells/immunology , Humans , Inflammation/immunology , T-Lymphocytes/immunology
12.
Arthritis Rheumatol ; 71(4): 632-640, 2019 04.
Article in English | MEDLINE | ID: mdl-30407752

ABSTRACT

OBJECTIVE: To develop a nanoparticle (NP) platform that can expand both CD4+ and CD8+ Treg cells in vivo for the suppression of autoimmune responses in systemic lupus erythematosus (SLE). METHODS: Poly(lactic-co-glycolic acid) (PLGA) NPs encapsulating interleukin-2 (IL-2) and transforming growth factor ß (TGFß) were coated with anti-CD2/CD4 antibodies and administered to mice with lupus-like disease induced by the transfer of DBA/2 T cells into (C57BL/6 × DBA/2)F1 (BDF1) mice. The peripheral frequency of Treg cells was monitored ex vivo by flow cytometry. Disease progression was assessed by measuring serum anti-double-stranded DNA antibody levels by enzyme-linked immunosorbent assay. Kidney disease was defined as the presence of proteinuria or renal histopathologic features. RESULTS: Anti-CD2/CD4 antibody-coated, but not noncoated, NPs encapsulating IL-2 and TGFß induced CD4+ and CD8+ FoxP3+ Treg cells in vitro. The optimal dosing regimen of NPs for expansion of CD4+ and CD8+ Treg cells was determined in in vivo studies in mice without lupus and then tested in BDF1 mice with lupus. The administration of anti-CD2/CD4 antibody-coated NPs encapsulating IL-2 and TGFß resulted in the expansion of CD4+ and CD8+ Treg cells, a marked suppression of anti-DNA antibody production, and reduced renal disease. CONCLUSION: This study shows for the first time that T cell-targeted PLGA NPs encapsulating IL-2 and TGFß can expand both CD4+ and CD8+ Treg cells in vivo and suppress murine lupus. This approach, which enables the expansion of Treg cells in vivo and inhibits pathogenic immune responses in SLE, could represent a potential new therapeutic modality in autoimmune conditions characterized by impaired Treg cell function associated with IL-2 deficiency.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Interleukin-2/administration & dosage , Lupus Erythematosus, Systemic/drug therapy , Nanoparticles/therapeutic use , T-Lymphocytes, Regulatory/drug effects , Transforming Growth Factor beta/administration & dosage , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred C57BL
13.
JCI Insight ; 3(16)2018 08 23.
Article in English | MEDLINE | ID: mdl-30135300

ABSTRACT

Defective DNA methylation in T cells leads to a series of T cell abnormalities in lupus; however, the full effect of T cell lineage-specific DNA methylation on disease expression has not been explored. Here, we show that 5-azacytidine, a DNA methyltransferase inhibitor, targeted to either CD4 or CD8 T cells in mice with established disease using a nanolipogel delivery system dramatically ameliorates lupus-related pathology through distinct mechanisms. In vivo targeted delivery of 5-azacytidine into CD4 T cells favors the expansion and function of Foxp3+ Tregs, whereas targeted delivery to CD8 T cells enhances the cytotoxicity and restrains the expansion of pathogenic TCR-αß+CD4-CD8- double-negative T cells. Our results signify the importance of cell-specific inhibition of DNA methylation in the treatment of established lupus.


Subject(s)
Azacitidine/administration & dosage , DNA Methylation/drug effects , Lupus Erythematosus, Systemic/drug therapy , Nanoconjugates/chemistry , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , DNA Methylation/immunology , DNA Modification Methylases/antagonists & inhibitors , Disease Models, Animal , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/immunology , Female , Humans , Immunoconjugates/chemistry , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Transgenic , Treatment Outcome
14.
J Clin Invest ; 128(8): 3445-3459, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29985166

ABSTRACT

Podocyte malfunction occurs in autoimmune and nonautoimmune kidney disease. Calcium signaling is essential for podocyte injury, but the role of Ca2+/calmodulin-dependent kinase (CaMK) signaling in podocytes has not been fully explored. We report that podocytes from patients with lupus nephritis and focal segmental glomerulosclerosis and lupus-prone and lipopolysaccharide- or adriamycin-treated mice display increased expression of CaMK IV (CaMK4), but not CaMK2. Mechanistically, CaMK4 modulated podocyte motility by altering the expression of the GTPases Rac1 and RhoA and suppressed the expression of nephrin, synaptopodin, and actin fibers in podocytes. In addition, it phosphorylated the scaffold protein 14-3-3ß, which resulted in the release and degradation of synaptopodin. Targeted delivery of a CaMK4 inhibitor to podocytes preserved their ultrastructure, averted immune complex deposition and crescent formation, and suppressed proteinuria in lupus-prone mice and proteinuria in mice exposed to lipopolysaccharide-induced podocyte injury by preserving nephrin/synaptopodin expression. In animals exposed to adriamycin, podocyte-specific delivery of a CaMK4 inhibitor prevented and reversed podocyte injury and renal disease. We conclude that CaMK4 is pivotal in immune and nonimmune podocyte injury and that its targeted cell-specific inhibition preserves podocyte structure and function and should have therapeutic value in lupus nephritis and podocytopathies, including focal segmental glomerulosclerosis.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Glomerulosclerosis, Focal Segmental/enzymology , Kidney Glomerulus/enzymology , Lupus Nephritis/enzymology , Signal Transduction , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 4/immunology , Cell Line, Transformed , Female , Glomerulosclerosis, Focal Segmental/immunology , Glomerulosclerosis, Focal Segmental/pathology , Humans , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred MRL lpr , Mice, Knockout , Proteinuria/enzymology , Proteinuria/immunology , Proteinuria/pathology
15.
ACS Appl Bio Mater ; 1(6): 1927-1941, 2018 Dec 17.
Article in English | MEDLINE | ID: mdl-34996256

ABSTRACT

Oncothermia, a special form of hyperthermia for oncological purposes, has been widely shown to be an effective mode of cancer therapy. However, its adoption among standard therapeutic practices has been limited by constraints in delivering sufficient thermal energy to tumor targets. To overcome these unique challenges in delivery presented by oncothermic therapeutics, we engineered a novel universal platform for hyperthermia cancer therapy utilizing versatile biocompatible materials. Herein, we show that Gold particle-in-particle (PIP), in which gold nanoparticles are physically confined within PLGA-PEG nanoparticles, significantly enhances thermal energy production by red-shifting the gold nanoparticle's absorption spectra via a mechanism in which we call nanoconfinement-induced therapeutic enhancement (NITE). NITE mediated Gold PIPs significantly suppress breast, skin, and multidrug resistant tumors and result in a multifold increase of heat shock protein expressed by cancer cells in vivo. Cotreatment of Gold PIP with doxorubicin shows a synergistic advantage. By using tumor-bearing mice, significant suppression of tumor growth by Gold PIPs shows the advantage of NITE mediated hyperthermia. Thus, we conclude that NITE mediated Gold PIP can be a strong anticancer therapy because of its sufficient amount of heat generation.

16.
Arterioscler Thromb Vasc Biol ; 38(1): 195-205, 2018 01.
Article in English | MEDLINE | ID: mdl-29146747

ABSTRACT

OBJECTIVE: Pseudoaneurysms remain a significant complication after vascular procedures. We hypothesized that TGF-ß (transforming growth factor-ß) signaling plays a mechanistic role in the development of pseudoaneurysms. APPROACH AND RESULTS: Rat aortic pericardial patch angioplasty was associated with a high incidence (88%) of pseudoaneurysms at 30 days, with increased smad2 phosphorylation in small pseudoaneurysms but not in large pseudoaneurysms; TGF-ß1 receptors were increased in small pseudoaneurysms and preserved in large pseudoaneurysms. Delivery of TGF-ß1 via nanoparticles covalently bonded to the patch stimulated smad2 phosphorylation both in vitro and in vivo and significantly decreased pseudoaneurysm formation (6.7%). Inhibition of TGF-ß1 signaling with SB431542 decreased smad2 phosphorylation both in vitro and in vivo and significantly induced pseudoaneurysm formation by day 7 (66.7%). CONCLUSIONS: Normal healing after aortic patch angioplasty is associated with increased TGF-ß1 signaling, and recruitment of smad2 signaling may limit pseudoaneurysm formation; loss of TGF-ß1 signaling is associated with the formation of large pseudoaneurysms. Enhancement of TGF-ß1 signaling may be a potential mechanism to limit pseudoaneurysm formation after vascular intervention.


Subject(s)
Aneurysm, False/prevention & control , Angioplasty/instrumentation , Aorta/surgery , Aortic Aneurysm/prevention & control , Coated Materials, Biocompatible , Pericardium/transplantation , Transforming Growth Factor beta1/administration & dosage , Wound Healing/drug effects , Aneurysm, False/etiology , Aneurysm, False/metabolism , Aneurysm, False/pathology , Angioplasty/adverse effects , Animals , Aorta/metabolism , Aorta/pathology , Aortic Aneurysm/etiology , Aortic Aneurysm/metabolism , Aortic Aneurysm/pathology , Cells, Cultured , Male , Mice , Nanoparticles , Phosphorylation , Prosthesis Design , Rats, Wistar , Signal Transduction/drug effects , Smad2 Protein/metabolism , Time Factors
17.
Methods Mol Biol ; 1530: 343-353, 2017.
Article in English | MEDLINE | ID: mdl-28150213

ABSTRACT

Artificial antigen-presenting cells (aAPCs) overcome many of the limitations of biologically based adoptive immunotherapy protocols. While these acellular systems can be designed with a variety of parameters, including material type, diameter, and proliferative signals for T cells, we outline methods to formulate and characterize a comprehensive polymeric microparticle aAPC platform. These aAPCs, which can be reproducibly fabricated in large quantities, efficiently stimulate antigen-specific T cell activation and proliferation by both paracrine cytokine signals and engagement of T cell surface proteins.


Subject(s)
Antigen Presentation , Artificial Cells/immunology , Immunotherapy , Antibodies/chemistry , Antibodies/immunology , Antigen-Presenting Cells/immunology , Avidin , Dendritic Cells/immunology , Dendritic Cells/metabolism , Histocompatibility Antigens/chemistry , Histocompatibility Antigens/immunology , Humans , Immunotherapy, Adoptive , Lactic Acid/chemistry , Microspheres , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Surface Properties , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
18.
Sci Rep ; 7: 40989, 2017 01 25.
Article in English | MEDLINE | ID: mdl-28120873

ABSTRACT

Patients with epithelial ovarian cancer have the best overall survival when maximal surgical effort is accomplished. However, despite numerous technological advances, surgery still relies primarily on white-light reflectance and the surgeon's vision. As such, micrometastases are usually missed and most patients clinically classified as a complete responder eventually recur and succumb to the disease. Our objective is to develop optical enhancers which can aid in the visualization of ovarian cancer micrometastasis. To this end we developed a nanoparticle (NP) platform, which is specifically targeted to the tumor microenvironment. Targeting is achieved by coating FDA-approved PLGA-PEG NP with the peptide sequence RGD, which binds with high affinity to αVß3 integrins present in both the tumor-associated neovasculature and on the surface of ovarian cancer cells. Administration of the NP platform carrying fluorescent dyes to mice bearing intraperitoneal ovarian cancer allowed visualization of tumor-associated vasculature and its contrast against normal blood vessels. More importantly, we demonstrate the visualization of intraperitoneal ovarian cancer micrometastasis as small as 100 µm with optimal resolution. Finally, we demonstrate that the fluorescent dye cargo was able to penetrate intra-tumorally. Such modality could be used to allow microscopic surgical debulking to assure maximal surgical effort.


Subject(s)
Neoplasm Micrometastasis/diagnosis , Optical Imaging/methods , Ovarian Neoplasms/secondary , Peritoneal Neoplasms/diagnosis , Staining and Labeling/methods , Animals , Disease Models, Animal , Female , Mice
19.
Sci Rep ; 7: 40142, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28071663

ABSTRACT

Prosthetic grafts and patches are commonly used in cardiovascular surgery, however neointimal hyperplasia remains a significant concern, especially under low flow conditions. We hypothesized that delivery of rapamycin from nanoparticles (NP) covalently attached to patches allows sustained site-specific delivery of therapeutic agents targeted to inhibit localized neointimal hyperplasia. NP were covalently linked to pericardial patches using EDC/NHS chemistry and could deliver at least 360 ng rapamycin per patch without detectable rapamycin in serum; nanoparticles were detectable in the liver, kidney and spleen but no other sites within 24 hours. In a rat venous patch angioplasty model, control patches developed robust neointimal hyperplasia on the patch luminal surface characterized by Eph-B4-positive endothelium and underlying SMC and infiltrating cells such as macrophages and leukocytes. Patches delivering rapamycin developed less neointimal hyperplasia, less smooth muscle cell proliferation, and had fewer infiltrating cells but retained endothelialization. NP covalently linked to pericardial patches are a novel composite delivery system that allows sustained site-specific delivery of therapeutics; NP delivering rapamycin inhibit patch neointimal hyperplasia. NP linked to patches may represent a next generation of tissue engineered cardiovascular implants.


Subject(s)
Angioplasty/methods , Growth Inhibitors/administration & dosage , Hyperplasia/prevention & control , Nanoparticles/administration & dosage , Neointima/pathology , Sirolimus/administration & dosage , Transplants/surgery , Animals , Drug Carriers/administration & dosage , Growth Inhibitors/pharmacokinetics , Histocytochemistry , Hyperplasia/pathology , Rats , Sirolimus/pharmacokinetics , Treatment Outcome
20.
Acta Biomater ; 57: 136-145, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28069499

ABSTRACT

The biophysical parameters governing nanoparticle (NP)-cell interactions significantly affect biological responses, particularly in the application of NP-based immunotherapeutics. Modulation of the surface biophysical character of NPs can be achieved via introduction of amino acids, which offer the ability to fine tune a range of biophysical parameters of interest. We employed this approach using monodisperse silica NPs coated with numerous poly(amino acid)s (PAAs). The NPs were incubated with dendritic cells (DCs) in conjunction with TLR ligands and production of IL-1ß from DCs and IFNγ from T cells primed by these DCs were measured. These key cytokines can prognosticate the efficacy of the NP platform as a potential vaccine or active cellular immunotherapy carrier. IL-1ß production showed a correlation with both NP size and degree of hydrophobicity. High IFNγ secretion from T cells was shown to be correlated with both the hydrophobicity and charge of the NPs used to activate the DCs. Other cytokines were also screened in order to compare the immune responses. The results of this study highlight the importance of nanoparticle biophysical parameters and the selection of TLR ligands to the rational design of nanoparticle-based vaccines and immunotherapies. STATEMENT OF SIGNIFICANCE: The manuscript describes a systematic investigation into the effects of biophysical parameters of nanoparticles (NPs) on immune cells. Modulation of the biophysical character of the NP surface can be achieved by introduction of amino acids on monodisperse silica NPs, introducing a range of tunable biophysical parameters of interest, i.e. distinct sizes, different surface charges and varying degrees of surface hydrophobicity. We examine internalization of the NP in dendritic cells (DCs) and measure a myriad of cytokines, including IL-1ß and IFNγ, which prognosticate the efficacy of the NPs as a potential vaccine (IL-1ß metric) or active cellular immunotherapy carrier (IFNγ metric). Two different TLR ligands (a viral TLR3 ligand and a bacterial TLR4 ligand) were used along with the PAA NPs to compare their costimulatory immunogenicity. We strongly believe that this study will provide crucial information to many readers of Acta Biomaterialia and further drive the use of nanoparticle platforms in modulating immune responses.


Subject(s)
Dendritic Cells/immunology , Interferon-gamma/immunology , Interleukin-1beta/immunology , Nanoparticles/chemistry , T-Lymphocytes/immunology , Toll-Like Receptor 3/agonists , Toll-Like Receptor 4/agonists , Animals , Female , Lignans , Mice , Toll-Like Receptor 3/immunology , Toll-Like Receptor 4/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...