Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
JGH Open ; 8(4): e13061, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38617108

ABSTRACT

Hypertriglyceridemia-induced acute pancreatitis (HTG-AP) is the third most common cause of AP after gallstones and alcohol. Supportive measures, intravenous insulin, and plasmapheresis are possible treatment modalities for HTG-AP; however, definitive guidelines evaluating the best therapeutic approach are not clearly established. We present a rare case of a 42-year-old male without known comorbidities who was found to have HTG-AP. Despite early initiation of intravenous insulin and plasmapheresis and the initial decline in his triglycerides level, his condition was complicated by necrotizing pancreatitis and subsequent multi-organ failure. Future studies are warranted to evaluate the role of plasmapheresis in HTG-AP and its efficacy.

2.
J Neurosci ; 43(47): 8043-8057, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37722850

ABSTRACT

The malignant brain cancer glioblastoma (GBM) contains groups of highly invasive cells that drive tumor progression as well as recurrence after surgery and chemotherapy. The molecular mechanisms that enable these GBM cells to exit the primary mass and disperse throughout the brain remain largely unknown. Here we report using human tumor specimens and primary spheroids from male and female patients that glial cell adhesion molecule (GlialCAM), which has normal roles in brain astrocytes and is mutated in the developmental brain disorder megalencephalic leukoencephalopathy with subcortical cysts (MLC), is differentially expressed in subpopulations of GBM cells. High levels of GlialCAM promote cell-cell adhesion and a proliferative GBM cell state in the tumor core. In contrast, GBM cells with low levels of GlialCAM display diminished proliferation and enhanced invasion into the surrounding brain parenchyma. RNAi-mediated inhibition of GlialCAM expression leads to activation of proinvasive extracellular matrix adhesion and signaling pathways. Profiling GlialCAM-regulated genes combined with cross-referencing to single-cell transcriptomic datasets validates functional links among GlialCAM, Mlc1, and aquaporin-4 in the invasive cell state. Collectively, these results reveal an important adhesion and signaling axis comprised of GlialCAM and associated proteins including Mlc1 and aquaporin-4 that is critical for control of GBM cell proliferation and invasion status in the brain cancer microenvironment.SIGNIFICANCE STATEMENT Glioblastoma (GBM) contains heterogeneous populations of cells that coordinately drive proliferation and invasion. We have discovered that glial cell adhesion molecule (GlialCAM)/hepatocyte cell adhesion molecule (HepaCAM) is highly expressed in proliferative GBM cells within the tumor core. In contrast, GBM cells with low levels of GlialCAM robustly invade into surrounding brain tissue along blood vessels and white matter. Quantitative RNA sequencing identifies various GlialCAM-regulated genes with functions in cell-cell adhesion and signaling. These data reveal that GlialCAM and associated signaling partners, including Mlc1 and aquaporin-4, are key factors that determine proliferative and invasive cell states in GBM.


Subject(s)
Aquaporins , Glioblastoma , Female , Humans , Male , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Membrane Proteins/metabolism , Tumor Microenvironment , Cell Proliferation , Neoplasm Invasiveness
3.
Development ; 150(6)2023 03 15.
Article in English | MEDLINE | ID: mdl-36960827

ABSTRACT

The blood-brain barrier (BBB) is a vascular endothelial cell boundary that partitions the circulation from the central nervous system to promote normal brain health. We have a limited understanding of how the BBB is formed during development and maintained in adulthood. We used quantitative transcriptional profiling to investigate whether specific adhesion molecules are involved in BBB functions, with an emphasis on understanding how astrocytes interact with endothelial cells. Our results reveal a striking enrichment of multiple genes encoding laminin subunits as well as the laminin receptor gene Itga7, which encodes the alpha7 integrin subunit, in astrocytes. Genetic ablation of Itga7 in mice led to aberrant BBB permeability and progressive neurological pathologies. Itga7-/- mice also showed a reduction in laminin protein expression in parenchymal basement membranes. Blood vessels in the Itga7-/- brain showed separation from surrounding astrocytes and had reduced expression of the tight junction proteins claudin 5 and ZO-1. We propose that the alpha7 integrin subunit in astrocytes via adhesion to laminins promotes endothelial cell junction integrity, all of which is required to properly form and maintain a functional BBB.


Subject(s)
Astrocytes , Blood-Brain Barrier , Mice , Animals , Blood-Brain Barrier/metabolism , Laminin/metabolism , Endothelial Cells/metabolism , Integrins/metabolism , Tight Junctions/metabolism
4.
Development ; 149(6)2022 03 15.
Article in English | MEDLINE | ID: mdl-35217866

ABSTRACT

In the developing mammalian brain, neuroepithelial cells interact with blood vessels to regulate angiogenesis, blood-brain barrier maturation and other key neurovascular functions. Genetic studies in mice have shown that neurovascular development is controlled, in part, by Itgb8, which encodes the neuroepithelial cell-expressed integrin ß8 subunit. However, these studies have involved complete loss-of-function Itgb8 mutations, and have not discerned the relative roles for the ß8 integrin extracellular matrix (ECM) binding region versus the intracellular signaling tail. Here, Cre/lox strategies have been employed to selectively delete the cytoplasmic tail of murine Itgb8 without perturbing its transmembrane and extracellular domains. We report that the ß8 integrin cytoplasmic domain is essential for inside-out modulation of adhesion, including activation of latent-TGFßs in the ECM. Quantitative sequencing of the brain endothelial cell transcriptome identifies TGFß-regulated genes with putative links to blood vessel morphogenesis, including several genes linked to Wnt/ß-catenin signaling. These results reveal that the ß8 integrin cytoplasmic domain is essential for the regulation of TGFß-dependent gene expression in endothelial cells and suggest that cross-talk between TGFßs and Wnt pathways is crucial for neurovascular development.


Subject(s)
Endothelial Cells , Integrin beta Chains , Animals , Brain/metabolism , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Integrins/genetics , Integrins/metabolism , Mammals/metabolism , Mice , Transforming Growth Factor beta/metabolism
5.
J Neurosci ; 42(8): 1406-1416, 2022 02 23.
Article in English | MEDLINE | ID: mdl-34965971

ABSTRACT

In the mammalian brain, perivascular astrocytes (PAs) closely juxtapose blood vessels and are postulated to have important roles in the control of vascular physiology, including regulation of the blood-brain barrier (BBB). Deciphering specific functions for PAs in BBB biology, however, has been limited by the ability to distinguish these cells from other astrocyte populations. In order to characterize selective roles for PAs in vivo, a new mouse model has been generated in which the endogenous megalencephalic leukoencephalopathy with subcortical cysts 1 (Mlc1) gene drives expression of Cre fused to a mutated estrogen ligand-binding domain (Mlc1-T2A-CreERT2). This knock-in mouse model, which we term MLCT, allows for selective identification and tracking of PAs in the postnatal brain. We also demonstrate that MLCT-mediated ablation of PAs causes severe defects in BBB integrity, resulting in premature death. PA loss results in aberrant localization of Claudin 5 and -VE-Cadherin in endothelial cell junctions as well as robust microgliosis. Collectively, these data reveal essential functions for Mlc1-expressing PAs in regulating endothelial barrier integrity in mice and indicate that primary defects in astrocytes that cause BBB breakdown may contribute to human neurologic disorders.SIGNIFICANCE STATEMENT Interlaced among the billions of neurons and glia in the mammalian brain is an elaborate network of blood vessels. Signals from the brain parenchyma control the unique permeability properties of cerebral blood vessels known as the blood-brain barrier (BBB). However, we understand very little about the relative contributions of different neural cell types in the regulation of BBB functions. Here, we show that a specific subpopulation of astrocyte is essential for control of BBB integrity, with ablation of these cells leading to defects in endothelial cell junctions, BBB breakdown, and resulting neurologic deficits.


Subject(s)
Astrocytes , Hereditary Central Nervous System Demyelinating Diseases , Animals , Astrocytes/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Claudin-5/genetics , Cysts , Disease Models, Animal , Hereditary Central Nervous System Demyelinating Diseases/genetics , Hereditary Central Nervous System Demyelinating Diseases/metabolism , Mammals , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice
6.
Oncogene ; 39(50): 7253-7264, 2020 12.
Article in English | MEDLINE | ID: mdl-33040087

ABSTRACT

Glioblastoma (GBM), or grade IV astrocytoma, is a malignant brain cancer that contains subpopulations of proliferative and invasive cells that coordinately drive primary tumor growth, progression, and recurrence after therapy. Here, we have analyzed functions for megalencephalic leukoencephalopathy with subcortical cysts 1 (Mlc1), an eight-transmembrane protein normally expressed in perivascular brain astrocyte end feet that is essential for neurovascular development and physiology, in the pathogenesis of GBM. We show that Mlc1 is expressed in human stem-like GBM cells (GSCs) and is linked to the development of primary and recurrent GBM. Genetically inhibiting MLC1 in GSCs using RNAi-mediated gene silencing results in diminished growth and invasion in vitro as well as impaired tumor initiation and progression in vivo. Biochemical assays identify the receptor tyrosine kinase Axl and its intracellular signaling effectors as important for MLC1 control of GSC invasive growth. Collectively, these data reveal key functions for MLC1 in promoting GSC growth and invasion, and suggest that targeting the Mlc1 protein or its associated signaling effectors may be a useful therapy for blocking tumor progression in patients with primary or recurrent GBM.


Subject(s)
Brain Neoplasms/pathology , Brain/pathology , Glioblastoma/pathology , Membrane Proteins/metabolism , Tumor Microenvironment , Animals , Cell Line, Tumor , Cell Polarity , Cell Proliferation , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction
7.
Development ; 147(18)2020 09 23.
Article in English | MEDLINE | ID: mdl-32895288

ABSTRACT

The central nervous system (CNS) contains a complex network of blood vessels that promote normal tissue development and physiology. Abnormal control of blood vessel morphogenesis and maturation is linked to the pathogenesis of various neurodevelopmental diseases. The CNS-specific genes that regulate blood vessel morphogenesis in development and disease remain largely unknown. Here, we have characterized functions for the gene encoding prion protein 2 (Prnd) in CNS blood vessel development and physiology. Prnd encodes the glycosylphosphatidylinositol (GPI)-linked protein doppel, which is expressed on the surface of angiogenic vascular endothelial cells, but is absent in quiescent endothelial cells of the adult CNS. During CNS vascular development, doppel interacts with receptor tyrosine kinases and activates cytoplasmic signaling pathways involved in endothelial cell survival, metabolism and migration. Analysis of mice genetically null for Prnd revealed impaired CNS blood vessel morphogenesis and associated endothelial cell sprouting defects. Prnd-/- mice also displayed defects in endothelial barrier integrity. Collectively, these data reveal novel mechanisms underlying doppel control of angiogenesis in the developing CNS, and may provide new insights about dysfunctional pathways that cause vascular-related CNS disorders.


Subject(s)
Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/physiology , Prion Proteins/metabolism , Animals , Central Nervous System/metabolism , Cytoplasm/metabolism , GPI-Linked Proteins/metabolism , Mice , Morphogenesis/physiology , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology
8.
Cancer Res ; 78(14): 3809-3822, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29743287

ABSTRACT

Glioblastoma (GBM) is an invasive brain cancer with tumor cells that disperse from the primary mass, escaping surgical resection and invariably giving rise to lethal recurrent lesions. Here we report that PTP-PEST, a cytoplasmic protein tyrosine phosphatase, controls GBM cell invasion by physically bridging the focal adhesion protein Crk-associated substrate (Cas) to valosin-containing protein (Vcp), an ATP-dependent protein segregase that selectively extracts ubiquitinated proteins from multiprotein complexes and targets them for degradation via the ubiquitin proteasome system. Both Cas and Vcp are substrates for PTP-PEST, with the phosphorylation status of tyrosine 805 (Y805) in Vcp impacting affinity for Cas in focal adhesions and controlling ubiquitination levels and protein stability. Perturbing PTP-PEST-mediated phosphorylation of Cas and Vcp led to alterations in GBM cell-invasive growth in vitro and in preclinical mouse models. Collectively, these data reveal a novel regulatory mechanism involving PTP-PEST, Vcp, and Cas that dynamically balances phosphorylation-dependent ubiquitination of key focal proteins involved in GBM cell invasion.Significance: PTP-PEST balances GBM cell growth and invasion by interacting with the ATP-dependent ubiquitin segregase Vcp/p97 and regulating phosphorylation and stability of the focal adhesion protein p130Cas.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/14/3809/F1.large.jpg Cancer Res; 78(14); 3809-22. ©2018 AACR.


Subject(s)
Focal Adhesions/genetics , Glioblastoma/genetics , Phosphorylation/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 12/genetics , Ubiquitination/genetics , Animals , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Crk-Associated Substrate Protein/genetics , Glioblastoma/pathology , HEK293 Cells , Humans , Mice , Mice, Nude , Neoplasm Invasiveness/genetics , Tyrosine/genetics , Valosin Containing Protein/genetics
9.
Sci Rep ; 8(1): 8267, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29844613

ABSTRACT

Disruption of the blood-brain barrier (BBB) by cancer cells is linked to metastatic tumor initiation and progression; however, the pathways that drive these events remain poorly understood. Here, we have developed novel patient-derived xenograft (PDX) models of brain metastases that recapitulate pathological growth features found in original patient samples, thus allowing for analysis of BBB disruption by tumor cells. We report that the BBB is selectively disrupted in brain metastases, in part, via inhibition of the endothelial cell-expressed docosahexaenoic acid (DHA) transporter, major facilitator superfamily domain 2a (Mfsd2a). Loss of Mfsd2a expression in the tumor endothelium results in enhanced BBB leakage, but reduced DHA transport and altered lipid metabolism within metastases. Mfsd2a expression in normal cerebral endothelial cells is cooperatively regulated by TGFß and bFGF signaling pathways, and these pathways are pathologically diminished in the brain metastasis endothelium. These results not only reveal a fundamental pathway underlying BBB disruption by metastatic cancer cells, but also suggest that restoring DHA metabolism in the brain tumor microenvironment may be a novel therapeutic strategy to block metastatic cell growth and survival.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Biological Transport , Brain/metabolism , Disease Models, Animal , Docosahexaenoic Acids/metabolism , Endothelial Cells/metabolism , Fatty Acid Transport Proteins/metabolism , Fatty Acids/metabolism , Female , Humans , Ion Transport , Lipid Metabolism/physiology , Male , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Nude , Neoplasm Metastasis/physiopathology , Signal Transduction , Symporters , Tumor Microenvironment , Tumor Suppressor Proteins/physiology , Xenograft Model Antitumor Assays
10.
PLoS One ; 12(9): e0185065, 2017.
Article in English | MEDLINE | ID: mdl-28938007

ABSTRACT

Glioblastoma (GBM) is a rapidly progressive brain cancer that exploits the neural microenvironment, and particularly blood vessels, for selective growth and survival. Anti-angiogenic agents such as the vascular endothelial growth factor-A (VEGF-A) blocking antibody bevacizumab yield short-term benefits to patients due to blood vessel regression and stabilization of vascular permeability. However, tumor recurrence is common, and this is associated with acquired resistance to bevacizumab. The mechanisms that drive acquired resistance and tumor recurrence in response to anti-angiogenic therapy remain largely unknown. Here, we report that Neuropilin-1 (Nrp1) regulates GBM growth and invasion by balancing tumor cell responses to VEGF-A and transforming growth factor ßs (TGFßs). Nrp1 is expressed in GBM cells where it promotes TGFß receptor internalization and signaling via Smad transcription factors. GBM that recur after bevacizumab treatment show down-regulation of Nrp1 expression, indicating that altering the balance between VEGF-A and TGFß signaling is one mechanism that promotes resistance to anti-angiogenic agents. Collectively, these data reveal that Nrp1 plays a critical role in balancing responsiveness to VEGF-A versus TGFß to regulate GBM growth, progression, and recurrence after anti-vascular therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Bevacizumab/pharmacology , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Neuropilin-1/metabolism , Transforming Growth Factor beta/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/diagnostic imaging , Glioblastoma/pathology , HEK293 Cells , Humans , Male , Mice, Nude , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm Transplantation , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism
11.
Mol Cancer Res ; 14(12): 1277-1287, 2016 12.
Article in English | MEDLINE | ID: mdl-27655131

ABSTRACT

Glioblastoma is a primary brain cancer that is resistant to all treatment modalities. This resistance is due, in large part, to invasive cancer cells that disperse from the main tumor site, escape surgical resection, and contribute to recurrent secondary lesions. The adhesion and signaling mechanisms that drive glioblastoma cell invasion remain enigmatic, and as a result there are no effective anti-invasive clinical therapies. Here we have characterized a novel adhesion and signaling pathway comprised of the integrin αvß8 and its intracellular binding partner, Spinophilin (Spn), which regulates glioblastoma cell invasion in the brain microenvironment. We show for the first time that Spn binds directly to the cytoplasmic domain of ß8 integrin in glioblastoma cells. Genetically targeting Spn leads to enhanced invasive cell growth in preclinical models of glioblastoma. Spn regulates glioblastoma cell invasion by modulating the formation and dissolution of invadopodia. Spn-regulated invadopodia dynamics are dependent, in part, on proper spatiotemporal activation of the Rac1 GTPase. Glioblastoma cells that lack Spn showed diminished Rac1 activities, increased numbers of invadopodia, and enhanced extracellular matrix degradation. Collectively, these data identify Spn as a critical adhesion and signaling protein that is essential for modulating glioblastoma cell invasion in the brain microenvironment. IMPLICATIONS: Tumor cell invasion is a major clinical obstacle in glioblastoma and this study identifies a new signaling pathway regulated by Spinophilin in invasive glioblastoma. Mol Cancer Res; 14(12); 1277-87. ©2016 AACR.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Integrins/metabolism , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Podosomes/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Binding Sites , Brain Neoplasms/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement , Glioblastoma/metabolism , Humans , Integrins/chemistry , Mice , Microfilament Proteins/chemistry , Neoplasm Invasiveness , Neoplasm Transplantation , Nerve Tissue Proteins/chemistry , Protein Binding , Signal Transduction
12.
Development ; 142(24): 4363-73, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26586223

ABSTRACT

Angiogenesis in the developing central nervous system (CNS) is regulated by neuroepithelial cells, although the genes and pathways that couple these cells to blood vessels remain largely uncharacterized. Here, we have used biochemical, cell biological and molecular genetic approaches to demonstrate that ß8 integrin (Itgb8) and neuropilin 1 (Nrp1) cooperatively promote CNS angiogenesis by mediating adhesion and signaling events between neuroepithelial cells and vascular endothelial cells. ß8 integrin in the neuroepithelium promotes the activation of extracellular matrix (ECM)-bound latent transforming growth factor ß (TGFß) ligands and stimulates TGFß receptor signaling in endothelial cells. Nrp1 in endothelial cells suppresses TGFß activation and signaling by forming intercellular protein complexes with ß8 integrin. Cell type-specific ablation of ß8 integrin, Nrp1, or canonical TGFß receptors results in pathological angiogenesis caused by defective neuroepithelial cell-endothelial cell adhesion and imbalances in canonical TGFß signaling. Collectively, these data identify a paracrine signaling pathway that links the neuroepithelium to blood vessels and precisely balances TGFß signaling during cerebral angiogenesis.


Subject(s)
Brain/blood supply , Brain/metabolism , Integrin beta Chains/metabolism , Neovascularization, Physiologic , Neuropilin-1/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Actins/metabolism , Animals , Brain/pathology , Cell Adhesion , Embryo Loss/pathology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Deletion , Male , Mice , Models, Biological , Neuroepithelial Cells/cytology , Neuroepithelial Cells/metabolism , Zebrafish
13.
Stem Cells Transl Med ; 4(10): 1234-45, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26285657

ABSTRACT

UNLABELLED: Human embryonic stem cells (hESCs) are a promising source of cells for tissue regeneration, yet histoincompatibility remains a major challenge to their clinical application. Because the human leukocyte antigen class I (HLA-I) molecules are the primary mediators of immune rejection, we hypothesized that cells derived from a hESC line lacking HLA-I expression could be transplanted without evoking a robust immune response from allogeneic recipients. In the present study, we used the replacement targeting strategy to delete exons 2 and 3 of ß2-microglobulin on both gene alleles in hESCs. Because ß2-microglobulin serves as the HLA-I light chain, disruption of the ß2-microglobulin gene led to complete HLA-I deficiency on the cell surface of hESCs and their derivatives. Therefore, these cells were resistant to CD8+ T-cell-mediated destruction. Although interferon-γ (IFN-γ) treatment significantly induced ß2-microglobulin expression, promoting CD8+ T cell-mediated killing of control hESCs and their derivatives, CD8+ T-cell-mediated cytotoxicity was barely observed with ß2-microglobulin-null hESCs and their derivatives treated with IFN-γ. This genetic manipulation to disrupt HLA-I expression did not affect the self-renewal capacity, genomic stability, or pluripotency of hESCs. Despite being relatively sensitive to natural killer (NK) cell-mediated killing due to the lack of HLA-I expression, when transplanted into NK cell-depleted immunocompetent mice, ß2-microglobulin-null hESCs developed into tumors resembling those derived from control hESCs in severe combined immunodeficiency mice. These results demonstrate that ß2-microglobulin-null hESCs significantly reduce immunogenicity to CD8+ T cells and might provide a renewable source of cells for tissue regeneration without the need for HLA matching in the future. SIGNIFICANCE: This study reports the generation of a novel ß2-microglobulin (B2M)-/- human embryonic stem cell (hESC) line. Differentiated mature cells from this line do not express cell surface human leukocyte antigen molecules even after interferon-γ stimulation and are resistant to alloreactive CD8+ T cells. Moreover, this B2M-/- hESC line contains no off-target integration or cleavage events, is devoid of stable B2M mRNA, exhibits a normal karyotype, and retains its self-renewal capacity, genomic stability, and pluripotency. Although B2M-/- hESC-derived cells are more susceptible to natural killer (NK) cells, murine transplantation studies have indicated that they are, overall, much less immunogenic than normal hESCs. Thus, these data show for the first time that, in vivo, the advantages provided by B2M-/- hESC-derived cells in avoiding CD8+ T-cell killing appear significantly greater than any disadvantage caused by increased susceptibility to NK cells.


Subject(s)
Gene Knockdown Techniques , Human Embryonic Stem Cells/immunology , beta 2-Microglobulin/genetics , Alleles , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Self Renewal , Cell Survival , Cell Transplantation/adverse effects , Cytotoxicity, Immunologic , Exons/genetics , Gene Expression Profiling , Genetic Vectors , Genomic Instability , Graft Rejection/prevention & control , HLA Antigens , Heterografts , Histocompatibility , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/transplantation , Humans , Interferon-gamma/pharmacology , Killer Cells, Natural/immunology , Mice , Mice, SCID , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/immunology , Pluripotent Stem Cells/transplantation , Sequence Deletion , Teratoma/etiology , Teratoma/immunology , beta 2-Microglobulin/physiology
14.
PLoS Genet ; 11(6): e1005356, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26121667

ABSTRACT

Lipophorin, the main Drosophila lipoprotein, circulates in the hemolymph transporting lipids between organs following routes that must adapt to changing physiological requirements. Lipophorin receptors expressed in developmentally dynamic patterns in tissues such as imaginal discs, oenocytes and ovaries control the timing and tissular distribution of lipid uptake. Using an affinity purification strategy, we identified a novel ligand for the lipophorin receptors, the circulating lipoprotein Lipid Transfer Particle (LTP). We show that specific isoforms of the lipophorin receptors mediate the extracellular accumulation of LTP in imaginal discs and ovaries. The interaction requires the LA-1 module in the lipophorin receptors and is strengthened by a contiguous region of 16 conserved amino acids. Lipophorin receptor variants that do not interact with LTP cannot mediate lipid uptake, revealing an essential role of LTP in the process. In addition, we show that lipophorin associates with the lipophorin receptors and with the extracellular matrix through weak interactions. However, during lipophorin receptor-mediated lipid uptake, LTP is required for a transient stabilization of lipophorin in the basolateral plasma membrane of imaginal disc cells. Together, our data suggests a molecular mechanism by which the lipophorin receptors tether LTP to the plasma membrane in lipid acceptor tissues. LTP would interact with lipophorin particles adsorbed to the extracellular matrix and with the plasma membrane, catalyzing the exchange of lipids between them.


Subject(s)
Apolipoproteins/metabolism , Carrier Proteins/metabolism , Cell Membrane/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Lipid Metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Animals, Genetically Modified , Apolipoproteins/genetics , Carrier Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian/metabolism , Female , Hemolymph/metabolism , Lipoproteins/blood , Lipoproteins/genetics , Ovary/metabolism , Protein Isoforms , Protein Structure, Tertiary , Receptors, Cytoplasmic and Nuclear/metabolism
15.
J Immunol ; 193(10): 5099-107, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25297874

ABSTRACT

Listeria monocytogenes is a major cause of mortality resulting from food poisoning in the United States. In mice, C5 has been genetically linked to host resistance to listeriosis. Despite this genetic association, it remains poorly understood how C5 and its activation products, C5a and C5b, confer host protection to this Gram-positive intracellular bacterium. In this article, we show in a systemic infection model that the major receptor for C5a, C5aR1, is required for a normal robust host immune response against L. monocytogenes. In comparison with wild-type mice, C5aR1(-/-) mice had reduced survival and increased bacterial burden in their livers and spleens. Infected C5aR1(-/-) mice exhibited a dramatic reduction in all major subsets of splenocytes, which was associated with elevated caspase-3 activity and increased TUNEL staining. Because type 1 IFN has been reported to impede the host response to L. monocytogenes through the promotion of splenocyte death, we examined the effect of C5aR1 on type 1 IFN expression in vivo. Indeed, serum levels of IFN-α and IFN-ß were significantly elevated in L. monocytogenes-infected C5aR1(-/-) mice. Similarly, the expression of TRAIL, a type 1 IFN target gene and a proapoptotic factor, was elevated in NK cells isolated from infected C5aR1(-/-) mice. Treatment of C5aR1(-/-) mice with a type 1 IFNR blocking Ab resulted in near-complete rescue of L. monocytogenes-induced mortality. Thus, these findings reveal a critical role for C5aR1 in host defense against L. monocytogenes through the suppression of type 1 IFN expression.


Subject(s)
Interferon-alpha/genetics , Interferon-beta/genetics , Listeria monocytogenes/immunology , Listeriosis/immunology , Spleen/immunology , Anaphylatoxins/immunology , Animals , Antibodies/pharmacology , Apoptosis , Bacterial Load , Caspase 3/genetics , Caspase 3/immunology , Complement C5a/genetics , Complement C5a/immunology , Complement C5b/genetics , Complement C5b/immunology , Gene Expression , Interferon-alpha/immunology , Interferon-beta/immunology , Listeriosis/drug therapy , Listeriosis/microbiology , Listeriosis/mortality , Liver/immunology , Liver/microbiology , Liver/pathology , Lymphocytes/immunology , Lymphocytes/microbiology , Lymphocytes/pathology , Male , Mice , Mice, Knockout , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/immunology , Receptors, Interferon/antagonists & inhibitors , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Spleen/microbiology , Spleen/pathology , Survival Analysis , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/immunology
16.
J Immunol ; 193(3): 1278-89, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24981453

ABSTRACT

Listeria monocytogenes is a Gram-positive intracellular bacterium that is acquired through tainted food and may lead to systemic infection and possible death. Despite the importance of the innate immune system in fighting L. monocytogenes infection, little is known about the role of complement and its activation products, including the potent C3a anaphylatoxin. In a model of systemic L. monocytogenes infection, we show that mice lacking the receptor for C3a (C3aR(-/-)) are significantly more sensitive to infection compared with wild-type mice, as demonstrated by decreased survival, increased bacterial burden, and increased damage to their livers and spleens. The inability of the C3aR(-/-) mice to clear the bacterial infection was not caused by defective macrophages or by a reduction in cytokines/chemokines known to be critical in the host response to L. monocytogenes, including IFN-γ and TNF-α. Instead, TUNEL staining, together with Fas, active caspase-3, and Bcl-2 expression data, indicates that the increased susceptibility of C3aR(-/-) mice to L. monocytogenes infection was largely caused by increased L. monocytogenes-induced apoptosis of myeloid and lymphoid cells in the spleen that are required for ultimate clearance of L. monocytogenes, including neutrophils, macrophages, dendritic cells, and T cells. These findings reveal an unexpected function of C3a/C3aR signaling during the host immune response that suppresses Fas expression and caspase-3 activity while increasing Bcl-2 expression, thereby providing protection to both myeloid and lymphoid cells against L. monocytogenes-induced apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/immunology , Complement C3a/metabolism , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/pathology , Receptors, Complement/physiology , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Caspase 3/metabolism , Caspase Inhibitors/pharmacology , Complement C3a/immunology , Disease Models, Animal , Down-Regulation/genetics , Down-Regulation/immunology , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Listeria monocytogenes/pathogenicity , Listeriosis/genetics , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Receptors, Complement/deficiency , Receptors, Complement/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Up-Regulation/genetics , Up-Regulation/immunology , fas Receptor/antagonists & inhibitors , fas Receptor/biosynthesis
17.
Mol Ther ; 18(3): 625-34, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20087316

ABSTRACT

Respiratory diseases are a major cause of mortality and morbidity worldwide. Current treatments offer no prospect of cure or disease reversal. Transplantation of pulmonary progenitor cells derived from human embryonic stem cells (hESCs) may provide a novel approach to regenerate endogenous lung cells destroyed by injury and disease. Here, we examine the therapeutic potential of alveolar type II epithelial cells derived from hESCs (hES-ATIICs) in a mouse model of acute lung injury. When transplanted into lungs of mice subjected to bleomycin (BLM)-induced acute lung injury, hES-ATIICs behaved as normal primary ATIICs, differentiating into cells expressing phenotypic markers of alveolar type I epithelial cells. Without experiencing tumorigenic side effects, lung injury was abrogated in mice transplanted with hES-ATIICs, demonstrated by recovery of body weight and arterial blood oxygen saturation, decreased collagen deposition, and increased survival. Therefore, transplantation of hES-ATIICs shows promise as an effective therapeutic to treat acute lung injury.


Subject(s)
Acute Lung Injury/metabolism , Embryonic Stem Cells/cytology , Epithelial Cells/cytology , Pulmonary Alveoli/metabolism , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Humans , Mice , Oxygen/metabolism , Phenotype , Promoter Regions, Genetic , Stem Cell Transplantation/methods , Transfection , Transgenes
18.
J Immunol ; 182(10): 6533-9, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19414808

ABSTRACT

Carboxypeptidase N (CPN) is a plasma zinc metalloprotease, which consists of two enzymatically active small subunits (CPN1) and two large subunits (CPN2) that protect the protein from degradation. Historically, CPN has been implicated as a major regulator of inflammation by its enzymatic cleavage of functionally important arginine and lysine amino acids from potent phlogistic molecules, such as the complement anaphylatoxins C3a and C5a. Because of no known complete CPN deficiencies, the biological impact of CPN in vivo has been difficult to evaluate. Here, we report the generation of a mouse with complete CPN deficiency by targeted disruption of the CPN1 gene. CPN1(-/-) mice were hypersensitive to lethal anaphylactic shock due to acute complement activation by cobra venom factor. This hypersensitivity was completely resolved in CPN1(-/-)/C5aR(-/-) but not in CPN1(-/-)/C3aR(-/-) mice. Moreover, CPN1(-/-) mice given C5a i.v., but not C3a, experienced 100% mortality. This C5a-induced mortality was reduced to 20% when CPN1(-/-) mice were treated with an antihistamine before C5a challenge. These studies describe for the first time a complete deficiency of CPN and demonstrate 1) that CPN plays a requisite role in regulating the lethal effects of anaphylatoxin-mediated shock, 2) that these lethal effects are mediated predominantly by C5a-induced histamine release, and 3) that C3a does not contribute significantly to shock following acute complement activation.


Subject(s)
Complement C5a/metabolism , Lysine Carboxypeptidase/genetics , Shock/genetics , Animals , Blotting, Southern , Complement C3a/immunology , Complement C3a/metabolism , Complement C5a/immunology , Complement Inactivating Agents/toxicity , Disease Susceptibility/immunology , Elapid Venoms/toxicity , Female , Histamine/immunology , Histamine/metabolism , Humans , Lysine Carboxypeptidase/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Shock/immunology
SELECTION OF CITATIONS
SEARCH DETAIL