Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters

Publication year range
1.
Arterioscler Thromb Vasc Biol ; 39(8): 1652-1666, 2019 08.
Article in English | MEDLINE | ID: mdl-31294623

ABSTRACT

OBJECTIVE: We examined the pathogenic significance of VEGF (vascular endothelial growth factor)-A in experimental abdominal aortic aneurysms (AAAs) and the translational value of pharmacological VEGF-A or its receptor inhibition in aneurysm suppression. Approaches and Results: AAAs were created in male C57BL/6J mice via intra-aortic elastase infusion. Soluble VEGFR (VEGF receptor)-2 extracellular ligand-binding domain (delivered in Ad [adenovirus]-VEGFR-2), anti-VEGF-A mAb (monoclonal antibody), and sunitinib were used to sequester VEGF-A, neutralize VEGF-A, and inhibit receptor tyrosine kinase activity, respectively. Influences on AAAs were assessed using ultrasonography and histopathology. In vitro transwell migration and quantitative reverse transcription polymerase chain reaction assays were used to assess myeloid cell chemotaxis and mRNA expression, respectively. Abundant VEGF-A mRNA and VEGF-A-positive cells were present in aneurysmal aortae. Sequestration of VEGF-A by Ad-VEGFR-2 prevented AAA formation, with attenuation of medial elastolysis and smooth muscle depletion, mural angiogenesis and monocyte/macrophage infiltration. Treatment with anti-VEGF-A mAb prevented AAA formation without affecting further progression of established AAAs. Sunitinib therapy substantially mitigated both AAA formation and further progression of established AAAs, attenuated aneurysmal aortic MMP2 (matrix metalloproteinase) and MMP9 protein expression, inhibited inflammatory monocyte and neutrophil chemotaxis to VEGF-A, and reduced MMP2, MMP9, and VEGF-A mRNA expression in macrophages and smooth muscle cells in vitro. Additionally, sunitinib treatment reduced circulating monocytes in aneurysmal mice. CONCLUSIONS: VEGF-A and its receptors contribute to experimental AAA formation by suppressing mural angiogenesis, MMP and VEGF-A production, myeloid cell chemotaxis, and circulating monocytes. Pharmacological inhibition of receptor tyrosine kinases by sunitinib or related compounds may provide novel opportunities for clinical aneurysm suppression.


Subject(s)
Aortic Aneurysm, Abdominal/etiology , Pancreatic Elastase/pharmacology , Receptors, Vascular Endothelial Growth Factor/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Aortic Aneurysm, Abdominal/drug therapy , Aortic Aneurysm, Abdominal/metabolism , Chemotaxis/drug effects , Disease Models, Animal , Disease Progression , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 9/analysis , Mice , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Sunitinib/therapeutic use , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/antagonists & inhibitors
2.
J Vasc Surg ; 67(2): 573-584.e2, 2018 02.
Article in English | MEDLINE | ID: mdl-28434702

ABSTRACT

OBJECTIVE: Angiotensin (Ang) II type 1 receptor (AT1) activation is essential for the development of exogenous Ang II-induced abdominal aortic aneurysms (AAAs) in hyperlipidemic animals. Experimental data derived from this modeling system, however, provide limited insight into the role of endogenous Ang II in aneurysm pathogenesis. Consequently, the potential translational value of AT1 inhibition in clinical AAA disease management remains incompletely understood on the basis of the existing literature. METHODS: AAAs were created in wild-type (WT) and AT1a knockout (KO) mice by intra-aortic infusion of porcine pancreatic elastase (PPE). WT mice were treated with the AT1 receptor antagonist telmisartan, 10 mg/kg/d in chow, or the peroxisome proliferator-activated receptor γ (PPARγ) antagonist GW9662, 3 mg/kg/d through oral gavage, beginning 1 week before or 3 days after PPE infusion. Influences on aneurysm progression as well as mechanistic insights into AT1-mediated pathogenic processes were determined using noninvasive ultrasound imaging, histopathology, aortic gene expression profiling, and flow cytometric analysis. RESULTS: After PPE infusion, aortic enlargement was almost completely abrogated in AT1a KO mice compared with WT mice. As defined by a ≥50% increase in aortic diameter, no PPE-infused, AT1a KO mouse actually developed an AAA. On histologic evaluation, medial smooth muscle cellularity and elastic lamellae were preserved in AT1a KO mice compared with WT mice, with marked attenuation of mural angiogenesis and leukocyte infiltration. In WT mice, telmisartan administration effectively suppressed aneurysm pathogenesis after PPE infusion as well, regardless of whether treatment was initiated before or after aneurysm creation or continued for a limited or extended time. Telmisartan treatment was associated with reduced messenger RNA levels for CCL5 and matrix metalloproteinases 2 and 9 in aneurysmal aortae, with no apparent effect on PPARγ-regulated gene expression. Administration of the PPARγ antagonist GW9662 failed to "rescue" the aneurysm phenotype in telmisartan-treated, PPE-infused WT mice. Neither effector T-cell differentiation nor regulatory T-cell cellularity was affected by telmisartan treatment status. CONCLUSIONS: Telmisartan effectively suppresses the progression of elastase-induced AAAs without apparent effect on PPARγ activation or T-cell differentiation. These findings reinforce the critical importance of endogenous AT1 activation in experimental AAA pathogenesis and reinforce the translational potential of AT1 inhibition in medical aneurysm disease management.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Aorta, Abdominal/drug effects , Aortic Aneurysm, Abdominal/prevention & control , Benzimidazoles/pharmacology , Benzoates/pharmacology , Pancreatic Elastase , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/deficiency , Animals , Aorta, Abdominal/diagnostic imaging , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Dilatation, Pathologic , Disease Models, Animal , Gene Expression Regulation , Genetic Predisposition to Disease , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptor, Angiotensin, Type 1/genetics , Signal Transduction/drug effects , Telmisartan , Time Factors , Transcriptome
3.
J Vasc Surg ; 68(5): 1538-1550.e2, 2018 11.
Article in English | MEDLINE | ID: mdl-29242064

ABSTRACT

OBJECTIVE: Mural angiogenesis and macrophage accumulation are two pathologic hallmarks of abdominal aortic aneurysm (AAA) disease. The heterodimeric transcription factor hypoxia-inducible factor 1 (HIF-1) is an essential regulator of angiogenesis and macrophage function. In this study, we investigated HIF-1 expression and activity in clinical and experimental AAA disease. METHODS: Human aortic samples were obtained from 24 AAA patients and six organ donors during open abdominal surgery. Experimental AAAs were created in 10-week-old male C57BL/6J mice by transient intra-aortic infusion of porcine pancreatic elastase (PPE). Expression of HIF-1α and its target gene messenger RNA (mRNA) levels were assessed in aneurysmal and control aortae. The HIF-1α inhibitors 2-methoxyestradiol and digoxin, the prolyl hydroxylase domain-containing protein (PHD) inhibitors cobalt chloride and JNJ-42041935, and the vehicle alone as control were administered daily to mice at varying time points beginning before or after PPE infusion. Influences on experimental AAA formation and progression were assessed by serial transabdominal ultrasound measurements of aortic diameter and histopathologic analysis at sacrifice. RESULTS: The mRNA levels for HIF-1α, vascular endothelial growth factor A, glucose transporter 1, and matrix metalloproteinase 2 were significantly increased in both human and experimental aneurysm tissue. Tissue immunostaining detected more HIF-1α protein in both human and experimental aneurysmal aortae compared with respective control aortae. Treatment with either HIF-1α inhibitor, beginning before or after PPE infusion, prevented enlargement of experimental aneurysms. Both HIF-1α inhibition regimens attenuated medial elastin degradation, smooth muscle cell depletion, and mural angiogenesis and the accumulation of macrophages, T cells, and B cells. Whereas mRNA levels for PHD1 and PHD2 were elevated in experimental aneurysmal aortae, pharmacologic inhibition of PHDs had limited effect on experimental aneurysm progression. CONCLUSIONS: Expression of HIF-1α and its target genes is increased in human and experimental AAAs. Treatment with HIF-1α inhibitors limits experimental AAA progression, with histologic evidence of attenuated mural leukocyte infiltration and angiogenesis. These findings underscore the potential significance of HIF-1α in aneurysm pathogenesis and as a target for pharmacologic suppression of AAA disease.


Subject(s)
Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , 2-Methoxyestradiol/pharmacology , Aged , Animals , Aorta, Abdominal/drug effects , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/prevention & control , CD4-Positive T-Lymphocytes/metabolism , Case-Control Studies , Chemotaxis, Leukocyte , Digoxin/pharmacology , Disease Models, Animal , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Male , Mice, Inbred C57BL , Middle Aged , Neovascularization, Pathologic , Pancreatic Elastase , Procollagen-Proline Dioxygenase/metabolism , Signal Transduction , Up-Regulation
4.
J Pharmacol Exp Ther ; 354(1): 73-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25962391

ABSTRACT

Interspecies differences have limited the predictive utility of toxicology studies performed using animal species. A drug that could be a safe and effective treatment in humans could cause toxicity in animals, preventing it from being used in humans. We investigated whether the use of thymidine kinase (TK)-NOG mice with humanized livers could prevent this unfortunate outcome (i.e., "rescue" a drug for use in humans). A high dose of furosemide is known to cause severe liver toxicity in mice, but it is a safe and effective treatment in humans. We demonstrate that administration of a high dose of furosemide (200 mg/kg i.p.) causes extensive hepatotoxicity in control mice but not in humanized TK-NOG mice. This interspecies difference results from a higher rate of production of the toxicity-causing metabolite by mouse liver. Comparison of their survival curves indicated that the humanized mice were more resistant than control mice to the hepatotoxicity caused by high doses of furosemide. In this test case, humanized TK-NOG mouse studies indicate that humans could be safely treated with a high dose of furosemide.


Subject(s)
Drug Evaluation, Preclinical/methods , Furosemide/toxicity , Hepatocytes/pathology , Liver/drug effects , Thymidine Kinase/genetics , Animals , Dose-Response Relationship, Drug , Furosemide/administration & dosage , Furosemide/pharmacokinetics , Hepatocytes/transplantation , Humans , Liver/pathology , Male , Mice , Necrosis , Species Specificity , Tissue Distribution
5.
J Pharmacol Exp Ther ; 352(2): 274-80, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25424997

ABSTRACT

Due to the substantial interspecies differences in drug metabolism and disposition, drug-induced liver injury (DILI) in humans is often not predicted by studies performed in animal species. For example, a drug (bosentan) used to treat pulmonary artery hypertension caused unexpected cholestatic liver toxicity in humans, which was not predicted by preclinical toxicology studies in multiple animal species. In this study, we demonstrate that NOG mice expressing a thymidine kinase transgene (TK-NOG) with humanized livers have a humanized profile of biliary excretion of a test (cefmetazole) drug, which was shown by an in situ perfusion study to result from interspecies differences in the rate of biliary transport and in liver retention of this drug. We also found that readily detectable cholestatic liver injury develops in TK-NOG mice with humanized livers after 1 week of treatment with bosentan (160, 32, or 6 mg/kg per day by mouth), whereas liver toxicity did not develop in control mice after 1 month of treatment. The laboratory and histologic features of bosentan-induced liver toxicity in humanized mice mirrored that of human subjects. Because DILI has become a significant public health problem, drug safety could be improved if preclinical toxicology studies were performed using humanized TK-NOG.


Subject(s)
Cefmetazole/pharmacokinetics , Chemical and Drug Induced Liver Injury/metabolism , Cholestasis/metabolism , Disease Models, Animal , Mice, Transgenic , Thymidine Kinase/genetics , Animals , Bosentan , Chemical and Drug Induced Liver Injury/complications , Chemical and Drug Induced Liver Injury/pathology , Cholestasis/etiology , Cholestasis/pathology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Ganciclovir/administration & dosage , Ganciclovir/pharmacology , Hepatocytes/metabolism , Hepatocytes/physiology , Hepatocytes/transplantation , Humans , Metabolic Clearance Rate , Species Specificity , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Sulfonamides/toxicity , Thymidine Kinase/metabolism , Tissue Distribution , Transgenes
6.
Infect Immun ; 82(7): 2826-39, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24752515

ABSTRACT

Reactivation of chronic infection with Toxoplasma gondii can cause life-threatening toxoplasmic encephalitis in immunocompromised individuals. We examined the role of VCAM-1/α4ß1 integrin interaction in T cell recruitment to prevent reactivation of the infection in the brain. SCID mice were infected and treated with sulfadiazine to establish a chronic infection. VCAM-1 and ICAM-1 were the endothelial adhesion molecules detected on cerebral vessels of the infected SCID and wild-type animals. Immune T cells from infected wild-type mice were treated with anti-α4 integrin or control antibodies and transferred into infected SCID or nude mice, and the animals received the same antibody every other day. Three days later, sulfadiazine was discontinued to initiate reactivation of infection. Expression of mRNAs for CD3δ, CD4, CD8ß, gamma interferon (IFN-γ), and inducible nitric oxide synthase (NOS2) (an effector molecule to inhibit T. gondii growth) and the numbers of CD4(+) and CD8(+) T cells in the brain were significantly less in mice treated with anti-α4 integrin antibody than in those treated with control antibody at 3 days after sulfadiazine discontinuation. At 6 days after sulfadiazine discontinuation, cerebral tachyzoite-specific SAG1 mRNA levels and numbers of inflammatory foci associated with tachyzoites were markedly greater in anti-α4 integrin antibody-treated than in control antibody-treated animals, even though IFN-γ and NOS2 mRNA levels were higher in the former than in the latter. These results indicate that VCAM-1/α4ß1 integrin interaction is crucial for prompt recruitment of immune T cells and induction of IFN-γ-mediated protective immune responses during the early stage of reactivation of chronic T. gondii infection to control tachyzoite growth.


Subject(s)
Encephalitis/parasitology , Integrin alpha4beta1/metabolism , T-Lymphocytes/physiology , Toxoplasmosis, Animal/immunology , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Brain/blood supply , Brain/cytology , Chronic Disease , Encephalitis/immunology , Female , Gene Expression Regulation/immunology , Integrin alpha4beta1/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , T-Lymphocytes/classification , Toxoplasma , Vascular Cell Adhesion Molecule-1/genetics
7.
PLoS Med ; 11(4): e1001628, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24736310

ABSTRACT

BACKGROUND: Seven of 15 clinical trial participants treated with a nucleoside analogue (fialuridine [FIAU]) developed acute liver failure. Five treated participants died, and two required a liver transplant. Preclinical toxicology studies in mice, rats, dogs, and primates did not provide any indication that FIAU would be hepatotoxic in humans. Therefore, we investigated whether FIAU-induced liver toxicity could be detected in chimeric TK-NOG mice with humanized livers. METHODS AND FINDINGS: Control and chimeric TK-NOG mice with humanized livers were treated orally with FIAU 400, 100, 25, or 2.5 mg/kg/d. The response to drug treatment was evaluated by measuring plasma lactate and liver enzymes, by assessing liver histology, and by electron microscopy. After treatment with FIAU 400 mg/kg/d for 4 d, chimeric mice developed clinical and serologic evidence of liver failure and lactic acidosis. Analysis of liver tissue revealed steatosis in regions with human, but not mouse, hepatocytes. Electron micrographs revealed lipid and mitochondrial abnormalities in the human hepatocytes in FIAU-treated chimeric mice. Dose-dependent liver toxicity was detected in chimeric mice treated with FIAU 100, 25, or 2.5 mg/kg/d for 14 d. Liver toxicity did not develop in control mice that were treated with the same FIAU doses for 14 d. In contrast, treatment with another nucleotide analogue (sofosbuvir 440 or 44 mg/kg/d po) for 14 d, which did not cause liver toxicity in human trial participants, did not cause liver toxicity in mice with humanized livers. CONCLUSIONS: FIAU-induced liver toxicity could be readily detected using chimeric TK-NOG mice with humanized livers, even when the mice were treated with a FIAU dose that was only 10-fold above the dose used in human participants. The clinical features, laboratory abnormalities, liver histology, and ultra-structural changes observed in FIAU-treated chimeric mice mirrored those of FIAU-treated human participants. The use of chimeric mice in preclinical toxicology studies could improve the safety of candidate medications selected for testing in human participants. Please see later in the article for the Editors' Summary.


Subject(s)
Antiviral Agents/toxicity , Arabinofuranosyluracil/analogs & derivatives , Liver Failure, Acute/chemically induced , Liver/drug effects , Animals , Arabinofuranosyluracil/toxicity , Chimera , Drug Evaluation, Preclinical , Female , Humans , Liver Failure, Acute/physiopathology , Male , Mice , Models, Animal , Toxicity Tests
8.
Biomolecules ; 13(8)2023 07 31.
Article in English | MEDLINE | ID: mdl-37627259

ABSTRACT

Lymphocyte function-associated antigen-1 (LFA-1) and its endothelial ligand intercellular adhesion molecule-1 (ICAM-1) are important for the migration of lymphocytes from blood vessels into lymph nodes. However, it is largely unknown whether these molecules mediate the homeostatic migration of lymphocytes from peripheral tissues into lymph nodes through lymphatic vessels. In this study, we find that, in naive mice, ICAM-1 is expressed on the sinus endothelia of lymph nodes, but not on the lymphatic vessels of peripheral tissues. In in vivo lymphocyte migration assays, memory CD4+ T cells migrated to lymph nodes from peripheral tissues much more efficiently than from blood vessels, as compared to naive CD4+ T cells. Moreover, ICAM-1 deficiency in host mice significantly inhibited the migration of adoptively transferred wild-type donor lymphocytes from peripheral tissues, but not from blood vessels, into lymph nodes. The migration of LFA-1-deficient donor lymphocytes from peripheral tissues into the lymph nodes of wild-type host mice was also significantly reduced as compared to wild-type donor lymphocytes. Furthermore, the number of memory T cells in lymph nodes was significantly reduced in the absence of ICAM-1 or LFA-1. Thus, our study extends the functions of the LFA-1/ICAM-1 adhesion pathway, indicating its novel role in controlling the homeostatic migration of lymphocytes from peripheral tissues into lymph nodes and maintaining memory T cellularity in lymph nodes.


Subject(s)
Lymphatic Vessels , Lymphocyte Function-Associated Antigen-1 , Animals , Mice , Intercellular Adhesion Molecule-1 , Lymphocytes , Lymph Nodes
9.
BMC Musculoskelet Disord ; 13: 119, 2012 Jul 03.
Article in English | MEDLINE | ID: mdl-22759918

ABSTRACT

BACKGROUND: Sjögren's syndrome is a tissue-specific autoimmune disease that affects exocrine tissues, especially salivary glands and lacrimal glands. Despite a large body of evidence gathered over the past 60 years, significant gaps still exist in our understanding of Sjögren's syndrome. The goal of this study was to develop a database that collects and organizes gene and protein expression data from the existing literature for comparative analysis with future gene expression and proteomic studies of Sjögren's syndrome. DESCRIPTION: To catalog the existing knowledge in the field, we used text mining to generate the Sjögren's Syndrome Knowledge Base (SSKB) of published gene/protein data, which were extracted from PubMed using text mining of over 7,700 abstracts and listing approximately 500 potential genes/proteins. The raw data were manually evaluated to remove duplicates and false-positives and assign gene names. The data base was manually curated to 477 entries, including 377 potential functional genes, which were used for enrichment and pathway analysis using gene ontology and KEGG pathway analysis. CONCLUSIONS: The Sjögren's syndrome knowledge base ( http://sskb.umn.edu) can form the foundation for an informed search of existing knowledge in the field as new potential therapeutic targets are identified by conventional or high throughput experimental techniques.


Subject(s)
Autoimmunity/genetics , Data Mining , Databases, Nucleic Acid , Databases, Protein , Knowledge Bases , Sjogren's Syndrome/genetics , Sjogren's Syndrome/metabolism , Bibliometrics , Gene Expression Regulation , Gene Regulatory Networks , Genetic Markers , Genetic Predisposition to Disease , Genomics , Humans , Phenotype , Proteomics , PubMed , Sjogren's Syndrome/immunology
10.
Am J Pathol ; 176(4): 1607-13, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20167872

ABSTRACT

Chronic infection with Toxoplasma gondii is one of the most common parasitic infections in humans. Formation of tissue cysts is the basis of persistence of the parasite in infected hosts, and this cyst stage has generally been regarded as untouchable. Here we provide the first evidence that the immune system can eliminate T. gondii cysts from the brains of infected hosts when immune T cells are transferred into infected immunodeficient animals that have already developed large numbers of cysts. This T cell-mediated immune process was associated with accumulation of microglia and macrophages around tissue cysts. CD8(+) immune T cells possess a potent activity to remove the cysts. The initiation of this process by CD8(+) T cells does not require their production of interferon-gamma, the major mediator to prevent proliferation of tachyzoites during acute infection, but does require perforin. These results suggest that CD8(+) T cells induce elimination of T. gondii cysts through their perforin-mediated cytotoxic activity. Our findings provide a new mechanism of the immune system to fight against chronic infection with T. gondii and suggest a possibility of developing a novel vaccine to eliminate cysts from patients with chronic infection and to prevent the establishment of chronic infection after a newly acquired infection.


Subject(s)
Brain/pathology , CD8-Positive T-Lymphocytes/parasitology , Toxoplasma/metabolism , Animals , Female , Immune System , Interferon-gamma/metabolism , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Microglia/pathology , Models, Biological , Toxoplasmosis, Animal/immunology , Toxoplasmosis, Animal/parasitology
11.
Proc Natl Acad Sci U S A ; 105(34): 12439-44, 2008 Aug 26.
Article in English | MEDLINE | ID: mdl-18716002

ABSTRACT

With the goal of identifying changes in gene expression in CD4(+) T cells during the development of diabetes in the nonobese diabetic (NOD) mouse, we used DNA microarrays to analyze gene expression in CD4(+) T cells from the pancreatic draining lymph nodes of NOD/BDC 2.5 T cell receptor transgenic and WT NOD mice at different ages. At 4 and 6 weeks of age, we found up-regulation of a number of genes that are known to be induced by IFN-alpha. IFN-alpha levels and IFN-alpha-producing plasmacytoid dendritic cells were increased in the PLNs of 3- to 4-week-old NOD mice. Moreover, blockade of IFN-alpha receptor 1 in NOD mice by a neutralizing antibody at 2-3 weeks of age significantly delayed the onset and decreased the incidence of type 1 diabetes, increased the relative number of immature dendritic cells in the PLNs, and enhanced the ability of spleen CD4(+) T cells to produce IL-4 and IL-10. These findings demonstrate that IFN-alpha in the PLNs is an essential initiator in the pathogenesis of type 1 diabetes in NOD mice.


Subject(s)
Diabetes Mellitus, Type 1/etiology , Interferon-alpha/physiology , Receptor, Interferon alpha-beta/physiology , Animals , CD4-Positive T-Lymphocytes/metabolism , Dendritic Cells/cytology , Diabetes Mellitus, Type 1/pathology , Gene Expression Profiling , Interferon-alpha/analysis , Interferon-alpha/pharmacology , Interleukin-10/biosynthesis , Interleukin-4/biosynthesis , Mice , Mice, Inbred NOD , Mice, Transgenic , Pancreas/cytology , Pancreas/immunology , Up-Regulation/drug effects
12.
J Exp Med ; 197(10): 1255-67, 2003 May 19.
Article in English | MEDLINE | ID: mdl-12756264

ABSTRACT

Bronchus-associated lymphoid tissue (BALT) participates in airway immune responses. However, little is known about the lymphocyte-endothelial adhesion cascades that recruit lymphocytes from blood into BALT. We show that high endothelial venules (HEVs) in BALT express substantial levels of VCAM-1, in marked contrast to HEVs in other secondary lymphoid tissues. BALT HEVs also express the L-selectin ligand PNAd. Anti-L-selectin, anti-PNAd, and anti-LFA-1 mAbs almost completely block the homing of B and T lymphocytes into BALT, whereas anti-alpha4 integrin and anti-VCAM-1 mAbs inhibit homing by nearly 40%. alpha4beta7 integrin and MAdCAM-1 are not involved. Importantly, we found that mAbs against alpha4 integrin and VCAM-1 significantly block the migration of total T cells (80% memory phenotype) but not naive T and B cells to BALT. These results suggest that an adhesion cascade, which includes L-selectin/PNAd, alpha4beta1 integrin/VCAM-1, and LFA-1, targets specific lymphocyte subsets to BALT. This high level of involvement of alpha4beta1 integrin/VCAM-1 is unique among secondary lymphoid tissues, and may help unify lymphocyte migration pathways and immune responses in BALT and other bronchopulmonary tissues.


Subject(s)
Antigens, Surface/physiology , Bronchi/immunology , Integrin alpha4beta1/physiology , L-Selectin/physiology , Lymphocytes/physiology , Lymphoid Tissue/immunology , Vascular Cell Adhesion Molecule-1/physiology , Animals , Cell Movement , Immunohistochemistry , Integrins/physiology , Membrane Proteins , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Rats , Rats, Sprague-Dawley
13.
J Autoimmun ; 35(2): 124-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20488663

ABSTRACT

Although B cells are crucial antigen-presenting cells in the initiation of T cell autoimmunity to islet beta cell autoantigens in type 1 diabetes (T1D), adhesion molecules that control migration of B cells into pancreatic lymph nodes (PanLN) in the nonobese diabetic (NOD) mouse model of human T1D have not been defined. In this study, we found that B cells from PanLN of 3-4-week-old female NOD mice expressed high levels of alpha(4) integrin and LFA-1 and intermediate levels of beta(7) integrin; half of B cells were L-selectin(high). In short-term in vivo lymphocyte migration assays, B cells migrated from the bloodstream into PanLN more efficiently than into peripheral LNs. Moreover, antibodies to mucosal addressin cell adhesion molecule 1 (MAdCAM-1) and alpha(4)beta(7) integrin inhibited >90% of B cell migration into PanLN. In contrast, antibodies to peripheral node addressin, L-selectin or LFA-1 partially inhibited B cell migration into PanLN. Furthermore, one intraperitoneal injection of anti-MAdCAM-1 antibody into 3-week-old NOD mice significantly inhibited entry of B cells into PanLN for at least 2 weeks. Taken together, these results indicate that the alpha(4)beta(7) integrin/MAdCAM-1 adhesion pathway plays a predominant role in migration of B cells into PanLN in NOD mice. Thus, specific blockage of alpha(4)beta(7) integrin/MAdCAM-1 adhesion pathway-mediated B cell migration may be a potential treatment for T1D.


Subject(s)
B-Lymphocytes/metabolism , Cell Adhesion Molecules/metabolism , Diabetes Mellitus, Type 1/immunology , Integrins/metabolism , Animals , Antibodies, Blocking/administration & dosage , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Adhesion/drug effects , Cell Adhesion Molecules/immunology , Cell Movement/drug effects , Cells, Cultured , Diabetes Mellitus, Type 1/drug therapy , Female , Humans , Integrins/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Inbred NOD , Mucoproteins , Pancreas/pathology , Signal Transduction/drug effects , Signal Transduction/immunology
14.
J Cell Biol ; 171(6): 931-7, 2005 Dec 19.
Article in English | MEDLINE | ID: mdl-16365160

ABSTRACT

Keratins 8 and 18 (K8/18) are major constituents of Mallory bodies (MBs), which are hepatocyte cytoplasmic inclusions seen in several liver diseases. K18-null but not K8-null or heterozygous mice form MBs, which indicates that K8 is important for MB formation. Early stages in MB genesis include K8/18 hyperphosphorylation and overexpression. We used transgenic mice that overexpress K8, K18, or K8/18 to test the importance of K8 and/or K18 in MB formation. MBs were induced by feeding 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC). Livers of young K8 or K8/K18 overexpressors had no histological abnormalities despite increased keratin protein and phosphorylation. In aging mice, only K8-overexpressing livers spontaneously developed small "pre-MB" aggregates. Only K8-overexpressing young mice are highly susceptible to MB formation after short-term DDC feeding. Thus, the K8 to K18 ratio, rather than K8/18 overexpression by itself, plays an essential role in MB formation. K8 overexpression is sufficient to form pre-MB and primes animals to accumulate MBs upon DDC challenge, which may help explain MB formation in human liver diseases.


Subject(s)
Gene Expression Regulation/physiology , Hepatocytes/ultrastructure , Inclusion Bodies/ultrastructure , Keratins/metabolism , Proteins/metabolism , Animals , Humans , Keratin-18 , Keratin-8 , Liver/enzymology , Liver/metabolism , Mice , Mice, Transgenic , Microscopy, Fluorescence , Models, Biological , RNA, Messenger/metabolism
15.
Cancer Immunol Immunother ; 58(10): 1577-86, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19238383

ABSTRACT

Tumor-associated macrophages (TAMs) are frequently found in glioblastomas and a high degree of macrophage infiltration is associated with a poor prognosis for glioblastoma patients. However, it is unclear whether TAMs in glioblastomas promote tumor growth. In this study, we found that folate receptor beta (FR beta) was expressed on macrophages in human glioblastomas and a rat C6 glioma implanted subcutaneously in nude mice. To target FR beta-expressing TAMs, we produced a recombinant immunotoxin consisting of immunoglobulin heavy and light chain Fv portions of an anti-mouse FR beta monoclonal antibody and Pseudomonas exotoxin A. Injection of the immunotoxin into C6 glioma xenografts in nude mice significantly depleted TAMs and reduced tumor growth. The immunotoxin targeting FR beta-expressing macrophages will provide a therapeutic tool for human glioblastomas.


Subject(s)
ADP Ribose Transferases/therapeutic use , Bacterial Toxins/therapeutic use , Carrier Proteins/immunology , Exotoxins/therapeutic use , Glioblastoma/therapy , Immunotoxins/therapeutic use , Macrophages, Peritoneal/immunology , Receptors, Cell Surface/immunology , Recombinant Fusion Proteins/therapeutic use , Virulence Factors/therapeutic use , Animals , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , Female , Folate Receptors, GPI-Anchored , Glioblastoma/immunology , Glioblastoma/pathology , Immunoenzyme Techniques , Immunoglobulin Variable Region/immunology , Mastocytoma/immunology , Mastocytoma/therapy , Mice , Mice, Inbred BALB C , Mice, Nude , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Nitric Oxide/metabolism , Rats , Survival Rate , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
16.
Respir Res ; 10: 97, 2009 Oct 22.
Article in English | MEDLINE | ID: mdl-19845971

ABSTRACT

BACKGROUND: Bronchus-associated lymphoid tissue (BALT) is the secondary lymphoid tissue in bronchial mucosa and is involved in the development of bronchopulmonary immune responses. Although migration of lymphocytes from blood vessels into secondary lymphoid tissues is critical for the development of appropriate adaptive immunity, the endothelia and lymphocyte adhesion molecules that recruit specific subsets of lymphocytes into human BALT are not known. The aim of this study was to determine which adhesion molecules are expressed on lymphocytes and high endothelial venules (HEVs) in human BALT. METHODS: We immunostained frozen sections of BALT from lobectomy specimens from 17 patients with lung carcinoma with a panel of monoclonal antibodies to endothelia and lymphocyte adhesion molecules. RESULTS: Sections of BALT showed B cell follicles surrounded by T cells. Most BALT CD4+ T cells had a CD45RO+ memory phenotype. Almost all BALT B cells expressed alpha4 integrin and L-selectin. In contrast, 43% of BALT T cells expressed alpha4 integrin and 20% of BALT T cells expressed L-selectin. Almost all BALT lymphocytes expressed LFA-1. HEVs, which support the migration of lymphocytes from the bloodstream into secondary lymphoid tissues, were prominent in BALT. All HEVs expressed peripheral node addressin, most HEVs expressed vascular cell adhesion molecule-1, and no HEVs expressed mucosal addressin cell adhesion molecule-1. CONCLUSION: Human BALT expresses endothelia and lymphocyte adhesion molecules that may be important in recruiting naive and memory/effector lymphocytes to BALT during protective and pathologic bronchopulmonary immune responses.


Subject(s)
Bronchi/immunology , Cell Adhesion Molecules/analysis , Lung Neoplasms/immunology , Lymphatic Vessels/immunology , Lymphocytes/immunology , Lymphoid Tissue/immunology , Adult , Antigens, Surface/analysis , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Humans , Immunity, Innate , Immunity, Mucosal , Immunoglobulins/analysis , Immunohistochemistry , Immunologic Memory , Immunophenotyping , Integrin alpha4/analysis , L-Selectin/analysis , Lung Neoplasms/surgery , Lymphocyte Function-Associated Antigen-1/analysis , Membrane Proteins/analysis , Mucoproteins/analysis , Pneumonectomy , Vascular Cell Adhesion Molecule-1/analysis
17.
J Interferon Cytokine Res ; 27(4): 329-38, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17477820

ABSTRACT

Interferon-gamma (IFNgamma) is essential for preventing reactivation of chronic infection with Toxoplasma gondii in the brain. We examined the role of IFNgamma on lymphocyte and endothelial adhesion molecule expression and T cell recruitment into the brain during chronic infection with T. gondii in IFNgamma knockout (IFNgamma(-/-)) and wild-type (WT) mice. Although the number of cerebral vessels expressing intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) increased in both WT and IFNgamma(-/-) mice following infection, there were more VCAM-1(+) vessels in brains of infected WT than of infected IFNgamma(-/-) mice; in contrast, numbers of ICAM-1(+) vessels did not differ between strains. We did not detect endothelial E-selectin, P-selectin, MAdCAM-1, or PNAd in any of the brains. Significantly fewer CD8(+) T cells were recruited into brains of infected IFNgamma(-/-) than WT mice. Treatment of infected IFNgamma(-/-) mice with recombinant IFN-gamma restored the expression of VCAM-1 on their cerebral vessels and recruitment of CD8(+) T cells into their brains, confirming an importance of this cytokine for upregulation of VCAM-1 expression and CD8(+) T cell trafficking. In infected WT and IFNgamma(-/-) animals, almost all cerebral CD8(+) T cells were lymphocyte function-associated antigen-1 (LFA-1)(high), CD44(high), and CD62L(neg), and approximately 38% were alpha4beta1 integrin(+). In adoptive transfer of immune spleen cells, pretreatment of the cells with a monoclonal antibody (mAb) against alpha4 integrin markedly inhibited recruitment of CD8(+) T cells into the brain of chronically infected WT mice. These results indicate that IFN-gamma-induced expression of endothelial VCAM-1 and its binding to alpha4beta1 integrin on CD8(+) T cells is important for recruitment of the T cells into the brain during the chronic stage of T. gondii infection, although LFA-1/ICAM-1 interaction may also be involved in this process.


Subject(s)
Brain , CD8-Positive T-Lymphocytes/immunology , Endothelium/metabolism , Interferon-gamma/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Vascular Cell Adhesion Molecule-1/metabolism , Adoptive Transfer , Animals , Brain/cytology , Brain/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , Cell Movement , Female , Integrin alpha4beta1/metabolism , Intercellular Adhesion Molecule-1/metabolism , Interferon-gamma/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Spleen/cytology , Spleen/immunology
18.
Diabetes Res Clin Pract ; 75(1): 14-26, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16765472

ABSTRACT

An unique isoform of hormone-sensitive lipase (HSL) is expressed in beta-cells. Recent findings suggest that HSL could be involved in the regulation of glucose stimulated insulin secretion (GSIS), however, these findings are controversial. To test the hypothesis that HSL is involved in control of normal GSIS via changes in its expression and/or activity in response to stimuli, we examined the effects of free fatty acid (FFA) loading and glucagon like peptide-1 (GLP-1) stimulation on the regulation of HSL expression and activity. With prolonged FFA loading, there was increased expression of beta-cell HSL and increased HSL hydrolytic activity in clonal beta-cells. Short-term treatment with GLP-1 increased HSL activity without changing the expression of the beta-cell isoform of HSL. Basal insulin secretion was increased, whereas GLP-1 potentiation of GSIS was decreased in islets isolated from HSL-/- mice, as compared to islets from wild type mice. Furthermore, using PancChip 2.2 cDNA microarrays (NIDDK consortium), the gene expression profile in the islets of HSL-/- mice was compared with wild type mice. Results showed changes in several metabolic pathways due to changes in lipid homeostasis caused by inactivation of HSL. Quantitative PCR for selected genes also revealed changes in genes that are related to insulin secretion, such as UCP-2. Therefore, these results suggest that the beta-cell isoform of HSL is involved in maintaining lipid homeostasis in islets and contributes to the proper control of GSIS.


Subject(s)
Insulin-Secreting Cells/enzymology , Sterol Esterase/metabolism , Animals , Base Sequence , DNA Primers , Fatty Acids, Nonesterified/metabolism , Gene Expression Regulation, Enzymologic , Genotype , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Sterol Esterase/deficiency , Sterol Esterase/genetics
19.
Toxicol Mech Methods ; 17(8): 475-81, 2007.
Article in English | MEDLINE | ID: mdl-20020874

ABSTRACT

ABSTRACT Chromium is a common human contact allergen, but it is not known whether chromates cause contact hypersensitivity by immunological mechanisms similar to those induced by strong haptens. To understand the immunological events of contact hypersensitivity to chromates, we investigated whether and how chromate sensitization alters lymphocyte subsets in draining lymph nodes (DLNs), blood, and spleens in mice. BALB/c mice were sensitized by painting their ears with 0.5% potassium dichromate or vehicle alone on 3 consecutive days. Flow cytometric analysis of lymphocyte surface antigens showed that the chromate exposure significantly increased the percentage of B cells and decreased the percentages of T cells in the DLNs. This was accompanied by a relative increase in T cells and a relative decrease in B cells in peripheral blood. In contrast to the chromate, sodium dodecyl sulfate (a skin irritant) did not affect B cells or T cells in the three compartments. Moreover, sensitization to the chromate led to dose-dependent decreases in the percentages of CD4(+) T cells and CD8(+) T cells in the DLNs. However, CD4(+) and CD8(+) memory T cells were significantly increased in the blood and DLNs of the chromate-sensitized mice. Additionally, the percentage of B cells in the DLNs but not blood was dose-dependently increased in the chromate-sensitized mice. Histologically, B-cell areas were dramatically enlarged in the DLNs of the chromate-sensitized mice. Thus, this report provides basic information to further elucidate the role of individual lymphocyte subsets in contact hypersensitivity to chromates.

20.
J Tissue Eng Regen Med ; 11(3): 887-895, 2017 03.
Article in English | MEDLINE | ID: mdl-25619945

ABSTRACT

The success of pancreatic islet (PI) transplantation is challenged by PI functional damage during the peritransplantation period. A silk-based encapsulation platform including mesenchymal stromal cells (MSCs) was evaluated for islet cell delivery in vivo. Islet equivalents (IEQs) were transplanted into the epididymal fat pads of mice with streptozotocin-induced diabetes. Three PI combinations were tested: (A) co-encapsulated in silk with MSCs; (b) encapsulated in silk alone; or (c) pelleted. Blood glucose levels were monitored and intraperitoneal glucose tolerance test (IPGTT) was performed upon return to euglycaemia. Grafts were removed for histology and cytokine content analysis. Mice with PI grafts in silk showed a prompt return to euglycaemia. IPGTT was significantly improved with PI in silk with MSCs, compared to PI in silk alone or pelleted. Both Th1 and Th2 cytokines were increased in PI grafts in silk, but Th1 cytokines were decreased significantly with PI and MSC co-encapsulation. Histological analysis showed osteogenesis and chondrogenesis in the silk grafts containing MSCs. Future studies will evaluate MSC stability and function in vivo and improve silk biocompatibility for applications in islet transplantation. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Cells, Immobilized/cytology , Islets of Langerhans Transplantation , Islets of Langerhans/physiology , Silk/pharmacology , Animals , Blood Glucose/metabolism , Cells, Immobilized/drug effects , Chemokines/metabolism , Glucose Tolerance Test , Inflammation Mediators/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Islets of Langerhans/drug effects , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL