Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Glia ; 71(6): 1429-1450, 2023 06.
Article in English | MEDLINE | ID: mdl-36794545

ABSTRACT

Neonatal stroke is common and causes life-long motor and cognitive sequelae. Because neonates with stroke are not diagnosed until days-months after the injury, chronic targets for repair are needed. We evaluated oligodendrocyte maturity and myelination and assessed oligodendrocyte gene expression changes using single cell RNA sequencing (scRNA seq) at chronic timepoints in a mouse model of neonatal arterial ischemic stroke. Mice underwent 60 min of transient right middle cerebral artery occlusion (MCAO) on postnatal day 10 (p10) and received 5-ethynyl-2'-deoxyuridine (EdU) on post-MCAO days 3-7 to label dividing cells. Animals were sacrificed 14 and 28-30 days post-MCAO for immunohistochemistry and electron microscopy. Oligodendrocytes were isolated from striatum 14 days post-MCAO for scRNA seq and differential gene expression analysis. The density of Olig2+ EdU+ cells was significantly increased in ipsilateral striatum 14 days post-MCAO and the majority of oligodendrocytes were immature. Density of Olig2+ EdU+ cells declined significantly between 14 and 28 days post-MCAO without a concurrent increase in mature Olig2+ EdU+ cells. By 28 days post-MCAO there were significantly fewer myelinated axons in ipsilateral striatum. scRNA seq identified a cluster of "disease associated oligodendrocytes (DOLs)" specific to the ischemic striatum, with increased expression of MHC class I genes. Gene ontology analysis suggested decreased enrichment of pathways involved in myelin production in the reactive cluster. Oligodendrocytes proliferate 3-7 days post-MCAO and persist at 14 days, but fail to mature by 28 days. MCAO induces a subset of oligodendrocytes with reactive phenotype, which may be a therapeutic target to promote white matter repair.


Subject(s)
Infarction, Middle Cerebral Artery , Stroke , Mice , Animals , Infarction, Middle Cerebral Artery/complications , Animals, Newborn , Stroke/complications , Oligodendroglia , Myelin Sheath
2.
J Control Release ; 286: 85-93, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30030182

ABSTRACT

Decades of research into improving drug delivery to tumors has documented uptake of particulate delivery systems by resident macrophages in the lung, liver, and spleen, and correlated short circulation times with reduced tumor accumulation. An implicit assumption in these studies is that nanoparticles present in the blood are available for distribution to the tumor. This study documents significant levels of lipoplex uptake by circulating leukocytes, and its effect on distribution to the tumor and other organs. In agreement with previous studies, PEGylation dramatically extends circulation times and enhances tumor delivery. However, our studies suggest that this relationship is not straightforward, and that particle sequestration by leukocytes can significantly alter biodistribution, especially with non-PEGylated nanoparticle formulations. We conclude that leukocyte uptake should be considered in biodistribution studies, and that delivery to these circulating cells may present opportunities for treating viral infections and leukemia.


Subject(s)
Leukocytes/metabolism , Nanoparticles/metabolism , Polyethylene Glycols/metabolism , Animals , DNA/administration & dosage , DNA/pharmacokinetics , Female , Gene Transfer Techniques , Mice, Inbred BALB C , Mice, SCID , Neoplasms/metabolism , Plasmids/administration & dosage , Plasmids/pharmacokinetics , Tissue Distribution
3.
FASEB J ; 30(3): 1096-108, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26581599

ABSTRACT

White adipocytes in adults are typically derived from tissue resident mesenchymal progenitors. The recent identification of de novo production of adipocytes from bone marrow progenitor-derived cells in mice challenges this paradigm and indicates an alternative lineage specification that adipocytes exist. We hypothesized that alternative lineage specification of white adipocytes is also present in human adipose tissue. Bone marrow from transgenic mice in which luciferase expression is governed by the adipocyte-restricted adiponectin gene promoter was adoptively transferred to wild-type recipient mice. Light emission was quantitated in recipients by in vivo imaging and direct enzyme assay. Adipocytes were also obtained from human recipients of hematopoietic stem cell transplantation. DNA was isolated, and microsatellite polymorphisms were exploited to quantify donor/recipient chimerism. Luciferase emission was detected from major fat depots of transplanted mice. No light emission was observed from intestines, liver, or lungs. Up to 35% of adipocytes in humans were generated from donor marrow cells in the absence of cell fusion. Nontransplanted mice and stromal-vascular fraction samples were used as negative and positive controls for the mouse and human experiments, respectively. This study provides evidence for a nontissue resident origin of an adipocyte subpopulation in both mice and humans.


Subject(s)
Adipocytes, White/physiology , Adipose Tissue/physiology , Stem Cells/physiology , Animals , Bone Marrow Cells/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Fusion/methods , Cell Lineage/genetics , Cell Lineage/physiology , Hematopoietic Stem Cells/physiology , Humans , Male , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics
4.
Methods Enzymol ; 537: 281-96, 2014.
Article in English | MEDLINE | ID: mdl-24480352

ABSTRACT

Analysis and isolation of adipocytes via flow cytometry is particularly useful to study their biology. However, the adoption of this technology has often been hampered by the presence of stromal/vascular cells in adipocyte fractions prepared from collagenase-digested adipose tissue. Here, we describe a multistep staining method and gating strategy that effectively excludes stromal contaminants. Initially, we set a gate optimized to the size and internal complexity of adipocytes. Exclusion of cell aggregates is then performed based on fluorescence of a nuclear stain followed by positive selection to collect only those cell events containing lipid droplets. Lastly, negative selection of cells expressing stromal or vascular lineage markers removes any remaining stromal contaminants. These procedures are applicable to simple analysis of adipocytes and their subcellular constituents by flow cytometry as well as isolation of adipocytes by flow sorting.


Subject(s)
Adipocytes/cytology , Cell Lineage/genetics , Cell Separation/methods , Flow Cytometry/methods , Adipose Tissue/cytology , Biomarkers , Cell Differentiation/genetics , Humans
5.
Proc Natl Acad Sci U S A ; 107(33): 14781-6, 2010 Aug 17.
Article in English | MEDLINE | ID: mdl-20679227

ABSTRACT

It is generally assumed that white adipocytes arise from resident adipose tissue mesenchymal progenitor cells. We challenge this paradigm by defining a hematopoietic origin for both the de novo development of a subset of white adipocytes in adults and a previously uncharacterized adipose tissue resident mesenchymal progenitor population. Lineage and cytogenetic analysis revealed that bone marrow progenitor (BMP)-derived adipocytes and adipocyte progenitors arise from hematopoietic cells via the myeloid lineage in the absence of cell fusion. Global gene expression analysis indicated that the BMP-derived fat cells are bona fide adipocytes but differ from conventional white or brown adipocytes in decreased expression of genes involved in mitochondrial biogenesis and lipid oxidation, and increased inflammatory gene expression. The BMP-derived adipocytes accumulate with age, occur in higher numbers in visceral than in subcutaneous fat, and in female versus male mice. BMP-derived adipocytes may, therefore, account in part for adipose depot heterogeneity and detrimental changes in adipose metabolism and inflammation with aging and adiposity.


Subject(s)
Adipocytes, White/cytology , Adipose Tissue/cytology , Mesoderm/cytology , Myeloid Cells/cytology , Adipocytes, Brown/cytology , Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Adipose Tissue/metabolism , Age Factors , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , Cytogenetic Analysis , Female , Gene Expression Profiling , Male , Mesoderm/metabolism , Mice , Models, Biological , Myeloid Cells/metabolism , Oligonucleotide Array Sequence Analysis , Sex Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...