Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol Res ; 2021: 6880036, 2021.
Article in English | MEDLINE | ID: mdl-34646889

ABSTRACT

Tobacco smoke is an established risk factor for thoracic aortic aneurysms and dissections (TAAD). However, little is known about its underlying mechanisms due to the lack of validated animal models. The present study developed a mouse model that may be utilized to investigate exacerbation of TAAD formation by mimetics of tobacco smoke. TAADs were created via inducible deletion of smooth muscle cell-specific Tgfbr2 receptors. Using this model, the first set of experiments evaluated the efficacy of nicotine salt (34.0 mg/kg/day), nicotine free base (NFB, 5.0 mg 90-day pellets), and cigarette smoke extract (0.1 ml/mouse/day). Compared with their respective control groups, only NFB pellets promoted TAAD dilation (23 ± 3% vs. 12 ± 2%, P = 0.014), and this efficacy was achieved at a cost of >50% acute mortality. Infusion of NFB with osmotic minipumps at extremely high, but nonlethal, doses (15.0 or 45.0 mg/kg/day) failed to accelerate TAAD dilation. Interestingly, costimulation with ß-aminopropionitrile (BAPN) promoted TAAD dilation and aortic rupture at dosages of 3.0 and 45.0 mg/kg/day, respectively, indicating that BAPN sensitizes the response of TAADs to NFB. In subsequent analyses, the detrimental effects of NFB were associated with clustering of macrophages, neutrophils, and T-cells in areas with structural destruction, enhanced matrix metalloproteinase- (MMP-) 2 production, and pathological angiogenesis with attenuated fibrosis in the adventitia. In conclusion, modeling nicotine exacerbation of TAAD formation requires optimization of chemical form, route of delivery, and dosage of the drug as well as the pathologic complexity of TAADs. Under the optimized conditions of the present study, chronic inflammation and adventitial mal-remodeling serve as critical pathways through which NFB exacerbates TAAD formation.


Subject(s)
Aortic Aneurysm, Thoracic/etiology , Aortic Dissection/etiology , Cigarette Smoking/adverse effects , Nicotine/toxicity , Receptor, Transforming Growth Factor-beta Type II/deficiency , Aortic Dissection/pathology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/pathology , Aortic Aneurysm, Thoracic/pathology , Disease Models, Animal , Humans , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Receptor, Transforming Growth Factor-beta Type II/genetics
3.
Nature ; 587(7832): 115-120, 2020 11.
Article in English | MEDLINE | ID: mdl-33087928

ABSTRACT

The immune system uses two distinct defence strategies against infections: microbe-directed pathogen destruction characterized by type 1 immunity1, and host-directed pathogen containment exemplified by type 2 immunity in induction of tissue repair2. Similar to infectious diseases, cancer progresses with self-propagating cancer cells inflicting host-tissue damage. The immunological mechanisms of cancer cell destruction are well defined3-5, but whether immune-mediated cancer cell containment can be induced remains poorly understood. Here we show that depletion of transforming growth factor-ß receptor 2 (TGFBR2) in CD4+ T cells, but not CD8+ T cells, halts cancer progression as a result of tissue healing and remodelling of the blood vasculature, causing cancer cell hypoxia and death in distant avascular regions. Notably, the host-directed protective response is dependent on the T helper 2 cytokine interleukin-4 (IL-4), but not the T helper 1 cytokine interferon-γ (IFN-γ). Thus, type 2 immunity can be mobilized as an effective tissue-level defence mechanism against cancer.


Subject(s)
Neoplasms/immunology , Neoplasms/pathology , Signal Transduction/immunology , Th2 Cells/immunology , Transforming Growth Factor beta/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Death/drug effects , Cell Hypoxia , Cell Line , Disease Progression , Female , Gene Expression Regulation/immunology , Humans , Interferon-gamma/immunology , Interleukin-4/immunology , Male , Mice , Mice, Inbred C57BL , Neoplasms/blood supply , Neoplasms/metabolism , Receptor, Transforming Growth Factor-beta Type II/deficiency , Signal Transduction/drug effects , Stromal Cells/cytology , Stromal Cells/immunology , Th2 Cells/metabolism , Transforming Growth Factor beta/antagonists & inhibitors
4.
Cell Biochem Funct ; 37(5): 320-330, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31172564

ABSTRACT

GDF15 plays a paradoxical role during carcinogenesis; it inhibits tumour growth in the early stages and promotes tumour cell proliferation in the late stages of cancer. Besides, GDF15 can induce apoptosis in some cancer cells including A549 but not in some others. Moreover, as a potential receptor for GDF15, TGFBR2 is inactivated during carcinogenesis in many types of cancers, and it is not present in cells with no GDF15 induced apoptosis. Thus, we tested whether GDF15 overexpression and/or TGFBR2 silencing can affect the GDF15 induced apoptosis in A549 cells. The full and mature forms of GDF15 were cloned and overexpressed in A549 cells. The TGFBR2 was silenced using specific siRNA and confirmed by real-time PCR. Results indicated that overexpression of full and mature forms of GDF15 as well as TGFBR2 knocked down reduced A549 cell viability in 24 and 48 hours. Flow cytometric analysis of annexin V/PI indicated induction of apoptosis in A549 cells by overexpression of GDF15 or silencing TGFBR2. Interestingly, the silencing of TGFBR2 inhibited the GDF15 induced cytotoxicity and apoptosis in A549 cells. Overexpression of GDF15 activated caspase-9 and caspase-3 and inhibited ERK1/2 and p38 phosphorylation in A549 cells. TGFBR2 knocked down inhibited GDF15 effects on caspases, ERK1/2, and p38MAPK activation. Our results indicated that the effect of GDF15 on apoptosis and activation of MAPK in A549 cells depends on TGFBR2 expression. These findings may point to mechanisms in which GDF15 exerts dual effect during carcinogenesis with regard to TGFBR2 expression. SIGNIFICANCE OF THE STUDY: GDF15 plays a tumour suppressor or promotor roles during carcinogenesis. The expression of GDF15 induced cytotoxicity, apoptosis, and inhibition of MAPK in A549 cells. All these effects were blocked by silencing TGFBR2 expression. These findings may point to mechanisms in which GDF15 exerts dual effect during carcinogenesis with regard to TGFBR2 expression.


Subject(s)
Apoptosis/drug effects , Growth Differentiation Factor 15/pharmacology , Receptor, Transforming Growth Factor-beta Type II/genetics , A549 Cells , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Profiling , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptor, Transforming Growth Factor-beta Type II/deficiency , Receptor, Transforming Growth Factor-beta Type II/metabolism , Tumor Cells, Cultured
5.
J Neuroinflammation ; 16(1): 69, 2019 Apr 02.
Article in English | MEDLINE | ID: mdl-30940161

ABSTRACT

BACKGROUND: Acute liver failure resulting from drug-induced liver injury can lead to the development of neurological complications called hepatic encephalopathy (HE). Hepatic transforming growth factor beta 1 (TGFß1) is upregulated due to liver failure in mice and inhibiting circulating TGFß reduced HE progression. However, the specific contributions of TGFß1 on brain cell populations and neuroinflammation during HE are not known. Therefore, the aim of this study was to characterize hepatic and brain TGFß1 signaling during acute liver failure and its contribution to HE progression using a combination of pharmacological and genetic approaches. METHODS: C57Bl/6 or neuron-specific transforming growth factor beta receptor 2 (TGFßR2) null mice (TGFßR2ΔNeu) were treated with azoxymethane (AOM) to induce acute liver failure and HE. The activity of circulating TGFß1 was inhibited in C57Bl/6 mice via injection of a neutralizing antibody against TGFß1 (anti-TGFß1) prior to AOM injection. In all mouse treatment groups, liver damage, neuroinflammation, and neurological deficits were assessed. Inflammatory signaling between neurons and microglia were investigated in in vitro studies through the use of pharmacological inhibitors of TGFß1 signaling in HT-22 and EOC-20 cells. RESULTS: TGFß1 was expressed and upregulated in the liver following AOM injection. Pharmacological inhibition of TGFß1 after AOM injection attenuated neurological decline, microglia activation, and neuroinflammation with no significant changes in liver damage. TGFßR2ΔNeu mice administered AOM showed no effect on liver pathology but significantly reduced neurological decline compared to control mice. Microglia activation and neuroinflammation were attenuated in mice with pharmacological inhibition of TGFß1 or in TGFßR2ΔNeu mice. TGFß1 increased chemokine ligand 2 (CCL2) and decreased C-X3-C motif ligand 1 (CX3CL1) expression in HT-22 cells and reduced interleukin-1 beta (IL-1ß) expression, tumor necrosis factor alpha (TNFα) expression, and phagocytosis activity in EOC-20 cells. CONCLUSION: Increased circulating TGFß1 following acute liver failure results in activation of neuronal TGFßR2 signaling, driving neuroinflammation and neurological decline during AOM-induced HE.


Subject(s)
Cerebral Cortex/pathology , Hepatic Encephalopathy/etiology , Liver Failure, Acute/complications , Liver Failure, Acute/pathology , Neurons/metabolism , Receptor, Transforming Growth Factor-beta Type II/deficiency , Transforming Growth Factor beta1/blood , Animals , Antibodies/therapeutic use , Azoxymethane/toxicity , Benzamides/pharmacology , Carcinogens/toxicity , Cell Line, Transformed , Disease Models, Animal , Hepatic Encephalopathy/drug therapy , Inflammation/drug therapy , Inflammation/etiology , Isoquinolines/pharmacology , Liver/metabolism , Liver/pathology , Liver Failure, Acute/chemically induced , Liver Failure, Acute/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Neurons/drug effects , Phagocytosis/drug effects , Phagocytosis/genetics , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrroles/pharmacology , Receptor, Transforming Growth Factor-beta Type II/genetics , Signal Transduction/drug effects , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/immunology , Up-Regulation/drug effects , Up-Regulation/genetics
6.
J Clin Invest ; 129(4): 1654-1670, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30855280

ABSTRACT

In tumors, extravascular fibrin forms provisional scaffolds for endothelial cell (EC) growth and motility during angiogenesis. We report that fibrin-mediated angiogenesis was inhibited and tumor growth delayed following postnatal deletion of Tgfbr2 in the endothelium of Cdh5-CreERT2 Tgfbr2fl/fl mice (Tgfbr2iECKO mice). ECs from Tgfbr2iECKO mice failed to upregulate the fibrinolysis inhibitor plasminogen activator inhibitor 1 (Serpine1, also known as PAI-1), due in part to uncoupled TGF-ß-mediated suppression of miR-30c. Bypassing TGF-ß signaling with vascular tropic nanoparticles that deliver miR-30c antagomiRs promoted PAI-1-dependent tumor growth and increased fibrin abundance, whereas miR-30c mimics inhibited tumor growth and promoted vascular-directed fibrinolysis in vivo. Using single-cell RNA-Seq and a NanoString miRNA array, we also found that subtypes of ECs in tumors showed spectrums of Serpine1 and miR-30c expression levels, suggesting functional diversity in ECs at the level of individual cells; indeed, fresh EC isolates from lung and mammary tumor models had differential abilities to degrade fibrin and launch new vessel sprouts, a finding that was linked to their inverse expression patterns of miR-30c and Serpine1 (i.e., miR-30chi Serpine1lo ECs were poorly angiogenic and miR-30clo Serpine1hi ECs were highly angiogenic). Thus, by balancing Serpine1 expression in ECs downstream of TGF-ß, miR-30c functions as a tumor suppressor in the tumor microenvironment through its ability to promote fibrin degradation and inhibit blood vessel formation.


Subject(s)
Endothelial Cells/metabolism , Mammary Neoplasms, Experimental/metabolism , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , Plasminogen Activator Inhibitor 1/metabolism , RNA, Neoplasm/metabolism , Transforming Growth Factor beta/metabolism , Animals , Endothelial Cells/pathology , Female , Gene Deletion , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , MicroRNAs/genetics , Neoplasm Proteins/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Plasminogen Activator Inhibitor 1/genetics , RNA, Neoplasm/genetics , Receptor, Transforming Growth Factor-beta Type II/deficiency , Receptor, Transforming Growth Factor-beta Type II/metabolism , Transforming Growth Factor beta/genetics
7.
J Pathol ; 248(2): 217-229, 2019 06.
Article in English | MEDLINE | ID: mdl-30714146

ABSTRACT

Proliferation of pancreatic acinar cells is a critical process in the pathophysiology of pancreatic diseases, because limited or defective proliferation is associated with organ dysfunction and patient morbidity. In this context, elucidating the signalling pathways that trigger and sustain acinar proliferation is pivotal to develop therapeutic interventions promoting the regenerative process of the organ. In this study we used genetic and pharmacological approaches to manipulate both local and systemic levels of thyroid hormones to elucidate their role in acinar proliferation following caerulein-mediated acute pancreatitis in mice. In addition, molecular mechanisms mediating the effects of thyroid hormones were identified by genetic and pharmacological inactivation of selected signalling pathways.In this study we demonstrated that levels of the thyroid hormone 3,3',5-triiodo-l-thyronine (T3) transiently increased in the pancreas during acute pancreatitis. Moreover, by using genetic and pharmacological approaches to manipulate both local and systemic levels of thyroid hormones, we showed that T3 was required to promote proliferation of pancreatic acinar cells, without affecting the extent of tissue damage or inflammatory infiltration.Finally, upon genetic and pharmacological inactivation of selected signalling pathways, we demonstrated that T3 exerted its mitogenic effect on acinar cells via a tightly controlled action on different molecular effectors, including histone deacetylase, AKT, and TGFß signalling.In conclusion, our data suggest that local availability of T3 in the pancreas is required to promote acinar cell proliferation and provide the rationale to exploit thyroid hormone signalling to enhance pancreatic regeneration. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Acinar Cells/metabolism , Cell Proliferation , Hyperthyroidism/metabolism , Pancreas, Exocrine/metabolism , Pancreatitis/metabolism , Triiodothyronine/metabolism , Acinar Cells/pathology , Animals , Ceruletide , Disease Models, Animal , Histone Deacetylases/metabolism , Hyperthyroidism/genetics , Hyperthyroidism/pathology , Iodide Peroxidase/deficiency , Iodide Peroxidase/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Pancreas, Exocrine/pathology , Pancreatitis/chemically induced , Pancreatitis/genetics , Pancreatitis/pathology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Transforming Growth Factor-beta Type II/deficiency , Receptor, Transforming Growth Factor-beta Type II/genetics , Signal Transduction , Thyroxine/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation
8.
J Autoimmun ; 95: 47-57, 2018 12.
Article in English | MEDLINE | ID: mdl-30340822

ABSTRACT

Gut microbiota and bacterial translocation have been implicated as significant contributors to mucosal immune responses and tolerance; alteration of microbial molecules, termed pathogen-associated molecular patterns (PAMP) and bacterial translocation are associated with immune pathology. However, the mechanisms by which dysregulated gut microbiota promotes autoimmunity is unclear. We have taken advantage of a well-characterized murine model of primary biliary cholangitis, dnTGFßRII mice, and an additional unique construct, toll-like receptor 2 (TLR2)-deficient dnTGFßRII mice coined dnTGFßRIITLR2-/- mice to investigate the influences of gut microbiota on autoimmune cholangitis. Firstly, we report that dnTGFßRII mice manifest altered composition of gut microbiota and that alteration of this gut microbiota by administration of antibiotics significantly alleviates T-cell-mediated infiltration and bile duct damage. Second, toll-like receptor 2 (TLR2)-deficient dnTGFßRII mice demonstrate significant exacerbation of autoimmune cholangitis when their epithelial barrier integrity was disrupted. Further, TLR2-deficiency mediates downregulated expression of tight junction-associated protein ZO-1 leading to increased gut permeability and bacterial translocation from gut to liver; use of antibiotics reduces microbiota translocation to liver and also decreases biliary pathology. In conclusion, our data demonstrates the important role of gut microbiota and bacterial translocation in the pathogenesis of murine autoimmune cholangitis.


Subject(s)
Autoimmune Diseases/microbiology , Bacterial Translocation/immunology , Bile Ducts/immunology , Liver Cirrhosis, Biliary/immunology , Receptor, Transforming Growth Factor-beta Type II/immunology , Toll-Like Receptor 2/immunology , Ampicillin/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , Bacterial Translocation/drug effects , Bile Ducts/drug effects , Bile Ducts/microbiology , Bile Ducts/pathology , Colon/drug effects , Colon/immunology , Colon/microbiology , Colon/pathology , Feces/microbiology , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Gene Expression Regulation , Immunity, Mucosal/drug effects , Liver/drug effects , Liver/immunology , Liver/microbiology , Liver/pathology , Liver Cirrhosis, Biliary/drug therapy , Liver Cirrhosis, Biliary/microbiology , Liver Cirrhosis, Biliary/pathology , Metronidazole/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neomycin/pharmacology , Receptor, Transforming Growth Factor-beta Type II/deficiency , Receptor, Transforming Growth Factor-beta Type II/genetics , Signal Transduction , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...