Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Circulation ; 143(2): 145-159, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33106031

ABSTRACT

BACKGROUND: Brown adipose tissue (BAT) is an important tissue for thermogenesis, making it a potential target to decrease the risks of obesity, type 2 diabetes, and cardiovascular disease, and recent studies have also identified BAT as an endocrine organ. Although BAT has been implicated to be protective in cardiovascular disease, to this point there are no studies that identify a direct role for BAT to mediate cardiac function. METHODS: To determine the role of BAT on cardiac function, we utilized a model of BAT transplantation. We then performed lipidomics and identified an increase in the lipokine 12,13-dihydroxy-9Z-octadecenoic acid (12,13-diHOME). We utilized a mouse model with sustained overexpression of 12,13-diHOME and investigated the role of 12,13-diHOME in a nitric oxide synthase type 1 deficient (NOS1-/-) mouse and in isolated cardiomyocytes to determine effects on function and respiration. We also investigated 12,13-diHOME in a cohort of human patients with heart disease. RESULTS: Here, we determined that transplantation of BAT (+BAT) improves cardiac function via the release of the lipokine 12,13-diHOME. Sustained overexpression of 12,13-diHOME using tissue nanotransfection negated the deleterious effects of a high-fat diet on cardiac function and remodeling, and acute injection of 12,13-diHOME increased cardiac hemodynamics via direct effects on the cardiomyocyte. Furthermore, incubation of cardiomyocytes with 12,13-diHOME increased mitochondrial respiration. The effects of 12,13-diHOME were absent in NOS1-/- mice and cardiomyocytes. We also provide the first evidence that 12,13-diHOME is decreased in human patients with heart disease. CONCLUSIONS: Our results identify an endocrine role for BAT to enhance cardiac function that is mediated by regulation of calcium cycling via 12,13-diHOME and NOS1.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/transplantation , Heart Failure/metabolism , Heart Failure/therapy , Lipidomics/methods , Oleic Acids/metabolism , Aged , Animals , Cells, Cultured , Cohort Studies , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Oleic Acids/administration & dosage , Physical Conditioning, Animal/methods , Physical Conditioning, Animal/physiology
2.
Biomater Sci ; 11(20): 6834-6847, 2023 Oct 10.
Article in English | MEDLINE | ID: mdl-37646133

ABSTRACT

Tumor-associated immune cells play a crucial role in cancer progression. Myeloid-derived suppressor cells (MDSCs), for example, are immature innate immune cells that infiltrate the tumor to exert immunosuppressive activity and protect cancer cells from the host's immune system and/or cancer-specific immunotherapies. While tumor-associated immune cells have emerged as a promising therapeutic target, efforts to counter immunosuppression within the tumor niche have been hampered by the lack of approaches that selectively target the immune cell compartment of the tumor, to effectively eliminate "tumor-protecting" immune cells and/or drive an "anti-tumor" phenotype. Here we report on a novel nanotechnology-based approach to target tumor-associated immune cells and promote "anti-tumor" responses in a murine model of breast cancer. Engineered extracellular vesicles (EVs) decorated with ICAM-1 ligands and loaded with miR-146a and Glut1, were biosynthesized (in vitro or in vivo) and administered to tumor-bearing mice once a week for up to 5 weeks. The impact of this treatment modality on the immune cell compartment and tumor progression was evaluated via RT-qPCR, flow cytometry, and histology. Our results indicate that weekly administration of the engineered EVs (i.e., ICAM-1-decorated and loaded with miR-146a and Glut1) hampered tumor progression compared to ICAM-1-decorated EVs with no cargo. Flow cytometry analyses of the tumors indicated a shift in the phenotype of the immune cell population toward a more pro-inflammatory state, which appeared to have facilitated the infiltration of tumor-targeting T cells, and was associated with a reduction in tumor size and decreased metastatic burden. Altogether, our results indicate that ICAM-1-decorated EVs could be a powerful platform nanotechnology for the deployment of immune cell-targeting therapies to solid tumors.

3.
Adv Healthc Mater ; 11(5): e2101619, 2022 03.
Article in English | MEDLINE | ID: mdl-34662497

ABSTRACT

Extracellular vesicles (EVs) have emerged as a promising carrier system for the delivery of therapeutic payloads in multiple disease models, including cancer. However, effective targeting of EVs to cancerous tissue remains a challenge. Here, it is shown that nonviral transfection of myeloid-derived suppressor cells (MDSCs) can be leveraged to drive targeted release of engineered EVs that can modulate transfer and overexpression of therapeutic anticancer genes in tumor cells and tissue. MDSCs are immature immune cells that exhibit enhanced tropism toward tumor tissue and play a role in modulating tumor progression. Current MDSC research has been mostly focused on mitigating immunosuppression in the tumor niche; however, the tumor homing abilities of these cells present untapped potential to deliver EV therapeutics directly to cancerous tissue. In vivo and ex vivo studies with murine models of breast cancer show that nonviral transfection of MDSCs does not hinder their ability to home to cancerous tissue. Moreover, transfected MDSCs can release engineered EVs and mediate antitumoral responses via paracrine signaling, including decreased invasion/metastatic activity and increased apoptosis/necrosis. Altogether, these findings indicate that MDSCs can be a powerful tool for the deployment of EV-based therapeutics to tumor tissue.


Subject(s)
Breast Neoplasms , Extracellular Vesicles , Myeloid-Derived Suppressor Cells , Animals , Breast Neoplasms/therapy , Female , Humans , Mice , Tumor Microenvironment
4.
Adv Healthc Mater ; 11(5): e2100805, 2022 03.
Article in English | MEDLINE | ID: mdl-35014204

ABSTRACT

Gene/oligonucleotide therapies have emerged as a promising strategy for the treatment of different neurological conditions. However, current methodologies for the delivery of neurogenic/neurotrophic cargo to brain and nerve tissue are fraught with caveats, including reliance on viral vectors, potential toxicity, and immune/inflammatory responses. Moreover, delivery to the central nervous system is further compounded by the low permeability of the blood brain barrier. Extracellular vesicles (EVs) have emerged as promising delivery vehicles for neurogenic/neurotrophic therapies, overcoming many of the limitations mentioned above. However, the manufacturing processes used for therapeutic EVs remain poorly understood. Here, we conducted a detailed study of the manufacturing process of neurogenic EVs by characterizing the nature of cargo and surface decoration, as well as the transfer dynamics across donor cells, EVs, and recipient cells. Neurogenic EVs loaded with Ascl1, Brn2, and Myt1l (ABM) are found to show enhanced neuron-specific tropism, modulate electrophysiological activity in neuronal cultures, and drive pro-neurogenic conversions/reprogramming. Moreover, murine studies demonstrate that surface decoration with glutamate receptors appears to mediate enhanced EV delivery to the brain. Altogether, the results indicate that ABM-loaded designer EVs can be a promising platform nanotechnology to drive pro-neuronal responses, and that surface functionalization with glutamate receptors can facilitate the deployment of EVs to the brain.


Subject(s)
Extracellular Vesicles , Animals , Blood-Brain Barrier , Cell Communication , Central Nervous System , Extracellular Vesicles/metabolism , Mice , Neurons
5.
Sci Adv ; 7(12)2021 03.
Article in English | MEDLINE | ID: mdl-33741587

ABSTRACT

Ischemic stroke causes vascular and neuronal tissue deficiencies that could lead to substantial functional impairment and/or death. Although progenitor-based vasculogenic cell therapies have shown promise as a potential rescue strategy following ischemic stroke, current approaches face major hurdles. Here, we used fibroblasts nanotransfected with Etv2, Foxc2, and Fli1 (EFF) to drive reprogramming-based vasculogenesis, intracranially, as a potential therapy for ischemic stroke. Perfusion analyses suggest that intracranial delivery of EFF-nanotransfected fibroblasts led to a dose-dependent increase in perfusion 14 days after injection. MRI and behavioral tests revealed ~70% infarct resolution and up to ~90% motor recovery for mice treated with EFF-nanotransfected fibroblasts. Immunohistological analysis confirmed increases in vascularity and neuronal cellularity, as well as reduced glial scar formation in response to treatment with EFF-nanotransfected fibroblasts. Together, our results suggest that vasculogenic cell therapies based on nanotransfection-driven (i.e., nonviral) cellular reprogramming represent a promising strategy for the treatment of ischemic stroke.


Subject(s)
Cellular Reprogramming , Ischemic Stroke , Animals , Cell Differentiation , Disease Models, Animal , Fibroblasts/metabolism , Ischemic Stroke/therapy , Mice
6.
Methods Mol Biol ; 2050: 79-84, 2020.
Article in English | MEDLINE | ID: mdl-31468481

ABSTRACT

Dendritic cells (DCs) are cells of the immune system that behave as antigen presenters and assist in T cell activation. DCs have recently been used in cell-based immunotherapies for the treatment of different diseases due to the lack of adverse nonspecific immune responses, typically elicited by other approaches. Genetically modified DCs, for example, have been used to stimulate CD4/CD8 antigen presenting immune responses. However, genetic manipulation of primary DCs remains a challenge. Here we describe a protocol for nonviral, benign transfection of primary DCs using nanochannel-based electroporation, and the subsequent collection of genetically modified exosomes for downstream applications.


Subject(s)
Dendritic Cells/cytology , Electroporation/instrumentation , Exosomes/genetics , Primary Cell Culture/methods , Transfection/instrumentation , Animals , Antigen Presentation , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cell Survival , Cells, Cultured , Dendritic Cells/immunology , Exosomes/transplantation , Humans , Mice , Nanotechnology
7.
Adv Biosyst ; 4(6): e2000049, 2020 06.
Article in English | MEDLINE | ID: mdl-32419350

ABSTRACT

Myeloid derived suppressor cells (MDSCs) have gained significant attention for their immunosuppressive role in cancer and their ability to contribute to tumor progression and metastasis. Understanding the role of MDSCs in driving cancer cell migration, a process fundamental to metastasis, is essential to fully comprehend and target MDSC-tumor cell interactions. This study employs microfabricated platforms, which simulate the structural cues present in the tumor microenvironment (TME) to elucidate the effects of MDSCs on the migratory phenotype of cancer cells at the single cell level. The results indicate that the presence of MDSCs enhances the motility of cancer-epithelial cells when directional cues (either topographical or spatial) are present. This behavior appears to be independent of cell-cell contact and driven by soluble byproducts from heterotypic interactions between MDSCs and cancer cells. Moreover, MDSC cell-motility is also impacted by the presence of cancer cells and the cancer cell secretome in the presence of directional cues. Epithelial dedifferentiation is the likely mechanism for changes in cancer cell motility in response to MDSCs. These results highlight the biochemical and biostructural conditions under which MDSCs can support cancer cell migration, and could therefore provide new avenues of research and therapy aimed at stemming cancer progression.


Subject(s)
Cell Communication , Cell Movement , Myeloid-Derived Suppressor Cells/metabolism , Neoplasms/metabolism , Tumor Microenvironment , Animals , Cell Line, Tumor , Female , Mice , Myeloid-Derived Suppressor Cells/pathology , Neoplasm Metastasis , Neoplasms/pathology
8.
Sci Rep ; 10(1): 1189, 2020 Jan 27.
Article in English | MEDLINE | ID: mdl-31988310

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are immune cells that exert immunosuppression within the tumor, protecting cancer cells from the host's immune system and/or exogenous immunotherapies. While current research has been mostly focused in countering MDSC-driven immunosuppression, little is known about the mechanisms by which MDSCs disseminate/infiltrate cancerous tissue. This study looks into the use of microtextured surfaces, coupled with in vitro and in vivo cellular and molecular analysis tools, to videoscopically evaluate the dissemination patterns of MDSCs under structurally guided migration, at the single-cell level. MDSCs exhibited topographically driven migration, showing significant intra- and inter-population differences in motility, with velocities reaching ~40 µm h-1. Downstream analyses coupled with single-cell migration uncovered the presence of specific MDSC subpopulations with different degrees of tumor-infiltrating and anti-inflammatory capabilities. Granulocytic MDSCs showed a ~≥3-fold increase in maximum dissemination velocities and traveled distances, and a ~10-fold difference in the expression of pro- and anti-inflammatory markers. Prolonged culture also revealed that purified subpopulations of MDSCs exhibit remarkable plasticity, with homogeneous/sorted subpopulations giving rise to heterogenous cultures that represented the entire hierarchy of MDSC phenotypes within 7 days. These studies point towards the granulocytic subtype as a potential cellular target of interest given their superior dissemination ability and enhanced anti-inflammatory activity.


Subject(s)
Breast Neoplasms/immunology , Cell Movement/genetics , Myeloid-Derived Suppressor Cells/immunology , Single-Cell Analysis/methods , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Plasticity/genetics , Female , Gene Expression , Humans , Inflammation/genetics , Mice , Mice, Nude , Phenotype , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
9.
Adv Biosyst ; 4(11): e2000157, 2020 11.
Article in English | MEDLINE | ID: mdl-32939985

ABSTRACT

While gene and cell therapies have emerged as promising treatment strategies for various neurological conditions, heavy reliance on viral vectors can hamper widespread clinical implementation. Here, the use of tissue nanotransfection as a platform nanotechnology to drive nonviral gene delivery to nerve tissue via nanochannels, in an effective, controlled, and benign manner is explored. TNT facilitates plasmid DNA delivery to the sciatic nerve of mice in a voltage-dependent manner. Compared to standard bulk electroporation (BEP), impairment in toe-spread and pinprick response is not caused by TNT, and has limited to no impact on electrophysiological parameters. BEP, however, induces significant nerve damage and increases macrophage immunoreactivity. TNT is subsequently used to deliver vasculogenic cell therapies to crushed nerves via delivery of reprogramming factor genes Etv2, Foxc2, and Fli1 (EFF). The results indicate the TNT-based delivery of EFF in a sciatic nerve crush model leads to increased vascularity, reduced macrophage infiltration, and improved recovery in electrophysiological parameters compared to crushed nerves that are TNT-treated with sham/empty plasmids. Altogether, the results indicate that TNT can be a powerful platform nanotechnology for localized nonviral gene delivery to nerve tissue, in vivo, and the deployment of reprogramming-based cell therapies for nerve repair/regeneration.


Subject(s)
Electroporation/methods , Gene Transfer Techniques , Nanomedicine/methods , Nanostructures , Animals , Disease Models, Animal , Female , Male , Mice , Peripheral Nerve Injuries/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/metabolism
10.
Trends Biotechnol ; 36(5): 549-561, 2018 05.
Article in English | MEDLINE | ID: mdl-29559164

ABSTRACT

Recent cancer research has more strongly emphasized the biophysical aspects of tumor development, progression, and microenvironment. In addition to genetic modifications and mutations in cancer cells, it is now well accepted that the physical properties of cancer cells such as stiffness, electrical impedance, and refractive index vary with tumor progression and can identify a malignant phenotype. Moreover, cancer heterogeneity renders population-based characterization techniques inadequate, as individual cellular features are lost in the average. Hence, platforms for fast and accurate characterization of biophysical properties of cancer cells at the single-cell level are required. Here, we highlight some of the recent advances in the field of cancer biophysics and the development of lab-on-a-chip platforms for single-cell biophysical analyses of cancer cells.


Subject(s)
Biophysical Phenomena , Lab-On-A-Chip Devices , Neoplasms/pathology , Neoplasms/physiopathology , Single-Cell Analysis/methods , Humans , Single-Cell Analysis/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL