Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Development ; 150(6)2023 03 15.
Article in English | MEDLINE | ID: mdl-36897571

ABSTRACT

Hormone secretion from pancreatic islets is essential for glucose homeostasis, and loss or dysfunction of islet cells is a hallmark of type 2 diabetes. Maf transcription factors are crucial for establishing and maintaining adult endocrine cell function. However, during pancreas development, MafB is not only expressed in insulin- and glucagon-producing cells, but also in Neurog3+ endocrine progenitor cells, suggesting additional functions in cell differentiation and islet formation. Here, we report that MafB deficiency impairs ß cell clustering and islet formation, but also coincides with loss of neurotransmitter and axon guidance receptor gene expression. Moreover, the observed loss of nicotinic receptor gene expression in human and mouse ß cells implied that signaling through these receptors contributes to islet cell migration/formation. Inhibition of nicotinic receptor activity resulted in reduced ß cell migration towards autonomic nerves and impaired ß cell clustering. These findings highlight a novel function of MafB in controlling neuronal-directed signaling events required for islet formation.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Islets of Langerhans , Mice , Adult , Animals , Humans , Glucagon/genetics , Glucagon/metabolism , Diabetes Mellitus, Type 2/metabolism , Islets of Langerhans/metabolism , Insulin/metabolism , Pancreas/metabolism , MafB Transcription Factor/genetics , MafB Transcription Factor/metabolism
2.
Sci Rep ; 10(1): 21778, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33311540

ABSTRACT

Sterile liver inflammation and fibrosis are associated with many liver disorders of different etiologies. Both type 1 and type 2 inflammatory responses have been reported to contribute to liver pathology. However, the mechanisms controlling the balance between these responses are largely unknown. Natural killer T (NKT) cells can be activated to rapidly secrete cytokines and chemokines associated with both type 1 and type 2 inflammatory responses. As these proteins have been reported to accumulate in different types of sterile liver inflammation, we hypothesized that these cells may play a role in this pathological process. We have found that a transgenic NKT (tgNKT) cell population produced in the immunodeficient 2,4αßNOD.Rag2-/- mice, but not in 2,4αßNOD.Rag2+/- control mice, promoted a type 1 inflammatory response with engagement of the NOD-, LRR- and pyrin domain-containing protein-3 (NLRP3) inflammasome. The induction of the type 1 inflammatory response was followed by an altered cytokine profile of the tgNKT cell population with a biased production of anti-inflammatory/profibrotic cytokines and development of liver fibrosis. These findings illustrate how the plasticity of NKT cells modulates the inflammatory response, suggesting a key role for the NKT cell population in the control of sterile liver inflammation.


Subject(s)
Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Natural Killer T-Cells/metabolism , Animals , Cytokines/metabolism , Disease Models, Animal , Fibrosis/metabolism , Hepatitis/pathology , Immunity, Cellular/physiology , Immunity, Innate/physiology , Inflammasomes/metabolism , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Diseases/pathology , Male , Mice , Mice, Inbred NOD , Natural Killer T-Cells/physiology
3.
J Vis Exp ; (160)2020 06 11.
Article in English | MEDLINE | ID: mdl-32597848

ABSTRACT

Imaging beta cells is a key step towards understanding islet transplantation. Although different imaging platforms for the recording of beta cell biology have been developed and utilized in vivo, they are limited in terms of allowing single cell resolution and continuous longitudinal recordings. Because of the transparency of the cornea, the anterior chamber of the eye (ACE) in mice is well suited to study human and mouse pancreatic islet cell biology. Here is a description of how this approach can be used to perform continuous longitudinal recordings of grafting and revascularization of individual human islet grafts. Human islet grafts are inserted into the ACE, using NOD.(Cg)-Gt(ROSA)26Sortm4-Rag2-/-mice as recipients. This allows for the investigation of the expansion of recipient versus donor cells and the contribution of recipient cells in promoting the encapsulation and vascularization of the graft. Further, a step-by-step approach for image analysis and quantification of the islet volume or segmented vasculature and islet capsule forming recipient cells is outlined.


Subject(s)
Anterior Chamber/cytology , Imaging, Three-Dimensional , Islets of Langerhans Transplantation , Islets of Langerhans/cytology , Animals , Humans , Insulin-Secreting Cells/cytology , Islets of Langerhans/surgery , Mice, Inbred NOD , Microscopy, Fluorescence, Multiphoton , Neovascularization, Physiologic
4.
Diabetologia ; 63(1): 137-148, 2020 01.
Article in English | MEDLINE | ID: mdl-31701200

ABSTRACT

AIMS/HYPOTHESIS: Rapid and adequate islet revascularisation and restoration of the islet-extracellular matrix (ECM) interaction are significant factors influencing islet survival and function of the transplanted islets in individuals with type 1 diabetes. Because the ECM encapsulating the islets is degraded during islet isolation, understanding the process of revascularisation and engraftment after transplantation is essential and needs further investigation. METHODS: Here we apply a longitudinal and high-resolution imaging approach to investigate the dynamics of the pancreatic islet engraftment process up to 11 months after transplantation. Human and mouse islet grafts were inserted into the anterior chamber of the mouse eye, using a NOD.ROSA-tomato.Rag2-/- or B6.ROSA-tomato host allowing the investigation of the expansion of host vs donor cells and the contribution of host cells to aspects such as promoting the encapsulation and vascularisation of the graft. RESULTS: A fibroblast-like stromal cell population of host origin rapidly migrates to ensheath the transplanted islet and aid in the formation of a basement membrane-like structure. Moreover, we show that the vessel network, while reconstituted by host endothelial cells, still retains the overall architecture of the donor islets. CONCLUSIONS/INTERPRETATION: In this transplantation situation the fibroblast-like stromal cells appear to take over as main producers of ECM or act as a scaffold for other ECM-producing cells to reconstitute a peri-islet-like basement membrane. This may have implications for our understanding of long-term graft rejection and for the design of novel strategies to interfere with this process.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Animals , Diabetes Mellitus, Type 1/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Graft Survival/physiology , Humans , Immunohistochemistry , Islets of Langerhans Transplantation , Longitudinal Studies , Mice
5.
Sci Rep ; 9(1): 9074, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31235823

ABSTRACT

Maf transcription factors are critical regulators of beta-cell function. We have previously shown that reduced MafA expression in human and mouse islets is associated with a pro-inflammatory gene signature. Here, we investigate if the loss of Maf transcription factors induced autoimmune processes in the pancreas. Transcriptomics analysis showed expression of pro-inflammatory as well as immune cell marker genes. However, clusters of CD4+ T and B220+ B cells were associated primarily with adult MafA-/-MafB+/-, but not MafA-/- islets. MafA expression was detected in the thymus, lymph nodes and bone marrow suggesting a novel role of MafA in regulating immune-cell function. Analysis of pancreatic lymph node cells showed activation of CD4+ T cells, but lack of CD8+ T cell activation which also coincided with an enrichment of naïve CD8+ T cells. Further analysis of T cell marker genes revealed a reduction of T cell receptor signaling gene expression in CD8, but not in CD4+ T cells, which was accompanied with a defect in early T cell receptor signaling in mutant CD8+ T cells. These results suggest that loss of MafA impairs both beta- and T cell function affecting the balance of peripheral immune responses against islet autoantigens, resulting in local inflammation in pancreatic islets.


Subject(s)
Gene Expression Regulation , Islets of Langerhans/pathology , Maf Transcription Factors, Large/metabolism , MafB Transcription Factor/metabolism , Animals , Antigen-Presenting Cells/metabolism , Autoimmunity , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/metabolism , Gene Knockout Techniques , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Islets of Langerhans/immunology , Maf Transcription Factors, Large/deficiency , Maf Transcription Factors, Large/genetics , MafB Transcription Factor/deficiency , MafB Transcription Factor/genetics , Mice , Mutation , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
6.
Front Immunol ; 10: 455, 2019.
Article in English | MEDLINE | ID: mdl-30936870

ABSTRACT

The apparition of adaptive immunity in Gnathostomata correlates with the expansion of the E-protein family to encompass E2-2, HEB, and E2A. Within the family, E2-2 and HEB are more closely evolutionarily related but their concerted action in hematopoiesis remains to be explored. Here we show that the combined disruption of E2-2 and HEB results in failure to express the early lymphoid program in Common lymphoid precursors (CLPs) and a near complete block in B-cell development. In the thymus, Early T-cell progenitors (ETPs) were reduced and T-cell development perturbed, resulting in reduced CD4 T- and increased γδ T-cell numbers. In contrast, hematopoietic stem cells (HSCs), erythro-myeloid progenitors, and innate immune cells were unaffected showing that E2-2 and HEB are dispensable for the ancestral hematopoietic lineages. Taken together, this E-protein dependence suggests that the appearance of the full Gnathostomata E-protein repertoire was critical to reinforce the gene regulatory circuits that drove the emergence and expansion of the lineages constituting humoral immunity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Gene Expression Regulation/physiology , Immunity, Humoral/physiology , Leukopoiesis/physiology , Lymphoid Progenitor Cells/pathology , Transcription Factor 4/physiology , Vertebrates/immunology , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/immunology , Biological Evolution , Cell Lineage , Evolution, Molecular , Gene Duplication , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Lymphocyte Subsets/pathology , Mice , Mice, Inbred C57BL , Multigene Family , Phylogeny , Sequence Alignment , Sequence Homology, Amino Acid , Spleen/pathology , Transcription Factor 4/deficiency , Transcription Factor 4/immunology
7.
Acta Neuropathol ; 137(4): 657-673, 2019 04.
Article in English | MEDLINE | ID: mdl-30830316

ABSTRACT

The TCF4 gene encodes for the basic helix-loop-helix transcription factor 4 (TCF4), which plays an important role in the development of the central nervous system (CNS). Haploinsufficiency of TCF4 was found to cause Pitt-Hopkins syndrome (PTHS), a severe neurodevelopmental disorder. Recently, the screening of a large cohort of medulloblastoma (MB), a highly aggressive embryonal brain tumor, revealed almost 20% of adult patients with MB of the Sonic hedgehog (SHH) subtype carrying somatic TCF4 mutations. Interestingly, many of these mutations have previously been detected as germline mutations in patients with PTHS. We show here that overexpression of wild-type TCF4 in vitro significantly suppresses cell proliferation in MB cells, whereas mutant TCF4 proteins do not to the same extent. Furthermore, RNA sequencing revealed significant upregulation of multiple well-known tumor suppressors upon expression of wild-type TCF4. In vivo, a prenatal knockout of Tcf4 in mice caused a significant increase in apoptosis accompanied by a decreased proliferation and failed migration of cerebellar granule neuron precursor cells (CGNP), which are thought to be the cells of origin for SHH MB. In contrast, postnatal in vitro and in vivo knockouts of Tcf4 with and without an additional constitutive activation of the SHH pathway led to significantly increased proliferation of CGNP or MB cells. Finally, publicly available data from human MB show that relatively low expression levels of TCF4 significantly correlate with a worse clinical outcome. These results not only point to time-specific roles of Tcf4 during cerebellar development but also suggest a functional linkage between TCF4 mutations and the formation of SHH MB, proposing that TCF4 acts as a tumor suppressor during postnatal stages of cerebellar development.


Subject(s)
Hedgehog Proteins/genetics , Medulloblastoma/genetics , Mutation , Transcription Factor 4/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Facies , Hedgehog Proteins/metabolism , Humans , Hyperventilation/genetics , Hyperventilation/metabolism , Hyperventilation/pathology , Intellectual Disability/genetics , Intellectual Disability/metabolism , Intellectual Disability/pathology , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice , Mice, Knockout , Transcription Factor 4/metabolism
8.
PLoS One ; 13(9): e0203228, 2018.
Article in English | MEDLINE | ID: mdl-30183741

ABSTRACT

Quinoline-3-carboxamides (Q substances) are small molecule compounds with anti-inflammatory properties. In this study, we used one of these substances, Paquinimod, to treat a novel model for chronic liver inflammation and liver fibrosis, the NOD-Inflammation Fibrosis (N-IF) mouse. We show that treatment of N-IF mice significantly reduced inflammation and resulted in the regression of fibrosis, even when the treatment was initiated after onset of disease. The reduced disease phenotype was associated with a systemic decrease in the number and reduced activation of disease-promoting transgenic natural killer T (NKT)-II cells and their type 2-cytokine expression profile. Paquinimod treatment also led to a reduction of CD115+ Ly6Chi monocytes and CD11b+ F4/80+ CD206+ macrophages.


Subject(s)
Immunologic Factors/pharmacology , Liver Cirrhosis/drug therapy , Quinolines/pharmacology , Animals , Cytokines/metabolism , Disease Models, Animal , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred NOD , Mice, Transgenic , Monocytes/drug effects , Monocytes/metabolism , Monocytes/pathology , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/pathology
9.
PLoS One ; 13(5): e0196598, 2018.
Article in English | MEDLINE | ID: mdl-29742113

ABSTRACT

Quinoline-3-carboxamides (Q compounds) are immunomodulatory compounds that have shown efficacy both in autoimmune disease and cancer. We have in here investigated the impact of one such compound, paquinimod, on the development of diabetes in the NOD mouse model for type I diabetes (T1D). In cohorts of NOD mice treated with paquinimod between weeks 10 to 20 of age and followed up until 40 weeks of age, we observed dose-dependent reduction in incidence of disease as well as delayed onset of disease. Further, in contrast to untreated controls, the majority of NOD mice treated from 15 weeks of age did not develop diabetes at 30 weeks of age. Importantly, these mice displayed significantly less insulitis, which correlated with selectively reduced number of splenic macrophages and splenic Ly6Chi inflammatory monocytes at end point as compared to untreated controls. Collectively, these results demonstrate that paquinimod treatment can significantly inhibit progression of insulitis to T1D in the NOD mouse. We propose that the effect of paquinimod on disease progression may be related to the reduced number of these myeloid cell populations. Our finding also indicates that this compound could be a candidate for clinical development towards diabetes therapy in humans.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Immunosuppressive Agents/therapeutic use , Quinolines/therapeutic use , Animals , Diabetes Mellitus, Type 1/pathology , Female , Glycosuria/prevention & control , Immunosuppressive Agents/pharmacology , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , Macrophages/drug effects , Mice , Mice, Inbred NOD , Monocytes/drug effects , Myeloid Cells/drug effects , Quinolines/pharmacology
10.
J Neuroimmune Pharmacol ; 13(2): 265-276, 2018 06.
Article in English | MEDLINE | ID: mdl-29550892

ABSTRACT

Multiple sclerosis is a neuroinflammatory degenerative disease, caused by activated immune cells infiltrating the CNS. The disease etiology involves both genetic and environmental factors. The mouse genetic locus, Eae27, linked to disease development in the experimental autoimmune encephalomyelitis (EAE) model for multiple sclerosis, was studied in order to identify contributing disease susceptibility factors and potential drug targets for multiple sclerosis. Studies of an Eae27 congenic mouse strain, revealed that genetic variation within Eae27 influences EAE development. The Abl2 gene, encoding the non-receptor tyrosine kinase Arg, is located in the 4,1 megabase pair long Eae27 region. The Arg protein plays an important role in cellular regulation and is, in addition, involved in signaling through the B- and T-cell receptors, important for the autoimmune response. The presence of a single nucleotide polymorphism causing an amino acid change in a near actin-interacting domain of Arg, in addition to altered lymphocyte activation in the congenic mice upon immunization with myelin antigen, makes Abl2/Arg a candidate gene for EAE. Here we demonstrate that the non-synonymous SNP does not change Arg's binding affinity for F-actin but suggest a role for Abl kinases in CNS inflammation pathogenesis by showing that pharmacological inhibition of Abl kinases ameliorates EAE, but not experimental arthritis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Protein-Tyrosine Kinases/genetics , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Mutant Strains , Polymorphism, Single Nucleotide , Protein-Tyrosine Kinases/immunology , Protein-Tyrosine Kinases/metabolism
11.
Diabetologia ; 60(10): 2033-2041, 2017 10.
Article in English | MEDLINE | ID: mdl-28660492

ABSTRACT

AIMS/HYPOTHESIS: Obesity is associated with glucose intolerance and insulin resistance and is closely linked to the increasing prevalence of type 2 diabetes. In mouse models of diet-induced obesity (DIO) and type 2 diabetes, an increased fat intake results in adipose tissue expansion and the secretion of proinflammatory cytokines. The innate immune system not only plays a crucial role in obesity-associated chronic low-grade inflammation but it is also proposed to play a role in modulating energy metabolism. However, little is known about how the modulation of metabolism by the immune system may promote increased adiposity in the early stages of increased dietary intake. Here we aimed to define the role of type I IFNs in DIO and insulin resistance. METHODS: Mice lacking the receptor for IFN-α (IFNAR-/-) and deficient in plasmacytoid dendritic cells (pDCs) (B6.E2-2 fl/fl .Itgax-cre) were fed a diet with a high fat content or normal chow. The mice were analysed in vivo and in vitro using cellular, biochemical and molecular approaches. RESULTS: We found that the development of obesity was inhibited by an inability to respond to type I IFNs. Furthermore, the development of obesity and insulin resistance in this model was associated with pDC recruitment to the fatty tissues and liver of obese mice (a 4.3-fold and 2.7-fold increase, respectively). Finally, we demonstrated that the depletion of pDCs protects mice from DIO and from developing obesity-associated metabolic complications. CONCLUSIONS/INTERPRETATION: Our results provide genetic evidence that pDCs, via type I IFNs, regulate energy metabolism and promote the development of obesity.


Subject(s)
Dendritic Cells/metabolism , Insulin Resistance/physiology , Interferon Type I/metabolism , Obesity/metabolism , Signal Transduction/physiology , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Diet, High-Fat , Male , Mice , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism
12.
Biomed Opt Express ; 8(12): 5637-5650, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29296493

ABSTRACT

In recent years, three-dimensional mesoscopic imaging has gained significant importance in life sciences for fundamental studies at the whole-organ level. In this manuscript, we present an optical projection tomography (OPT) method designed for imaging of the intact mouse brain. The system features an isotropic resolution of ~50 µm and an acquisition time of four to eight minutes, using a 3-day optimized clearing protocol. Imaging of the brain autofluorescence in 3D reveals details of the neuroanatomy, while the use of fluorescent labels displays the vascular network and amyloid deposition in 5xFAD mice, an important model of Alzheimer's disease (AD). Finally, the OPT images are compared with histological slices.

13.
Biomed Opt Express ; 7(11): 4569-4580, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27895996

ABSTRACT

In diabetes, pancreatic ß-cells play a key role. These cells are clustered within structures called islets of Langerhans inside the pancreas and produce insulin, which is directly secreted into the blood stream. The dense vascularization of islets of Langerhans is critical for maintaining a proper regulation of blood glucose homeostasis and is known to be affected from the early stage of diabetes. The deep localization of these islets inside the pancreas in the abdominal cavity renders their in vivo visualization a challenging task. A fast label-free imaging method with high spatial resolution is required to study the vascular network of islets of Langerhans. Based on these requirements, we developed a label-free and three-dimensional imaging method for observing islets of Langerhans using extended-focus Fourier domain Optical Coherence Microscopy (xfOCM). In addition to structural imaging, this system provides three-dimensional vascular network imaging and dynamic blood flow information within islets of Langerhans. We propose our method to deepen the understanding of the interconnection between diabetes and the evolution of the islet vascular network.

14.
BMC Med Genet ; 17(1): 70, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-27716086

ABSTRACT

BACKGROUND: T1D and AITD are autoimmune disorders commonly occurring in the same family and even in the same individual. The genetic contribution to these disorders is complex making uncovering of susceptibility genes very challenging. The general aim of this study was to identify loci and genes contributing to T1D/AITD susceptibility. Our strategy was to perform linkage and association studies in the relatively genetically homogenous population of northern Sweden. We performed a GWLS to find genomic regions linked to T1D/AITD in families from northern Sweden and we performed an association study in the families to test for association between T1D/AITD and variants in previously published candidate genes as well as a novel candidate gene, CD247. METHODS: DNA prepared from 459 individuals was used to perform a linkage and an association study. The ABI PRISM Linkage Mapping Set v2.5MD10 was employed for an initial 10-cM GWLS, and additional markers were added for fine mapping. Merlin was used for linkage calculations. For the association analysis, a GoldenGate Custom Panel from Illumina containing 79 SNPs of interest was used and FBAT was used for association calculations. RESULTS: Our study revealed linkage to two previously identified chromosomal regions, 4q25 and 6p22, as well as to a novel chromosomal region, 1q23. The association study replicated association to PTPN22, HLA-DRB1, INS, IFIH1, CTLA4 and C12orf30. Evidence in favor of association was also found for SNPs in the novel susceptibility gene CD247. CONCLUSIONS: Several risk loci for T1D/AITD identified in published association studies were replicated in a family material, of modest size, from northern Sweden. This provides evidence that these loci confer disease susceptibility in this population and emphasizes that small to intermediate sized family studies in this population can be used in a cost-effective manner for the search of genes involved in complex diseases. The linkage study revealed a chromosomal region in which a novel T1D/AITD susceptibility gene, CD247, is located. The association study showed association between T1D/AITD and several variants in this gene. These results suggests that common susceptibility genes act in concert with variants of CD247 to generate genetic risk for T1D/AITD in this population.


Subject(s)
CD3 Complex/genetics , Diabetes Mellitus, Type 1/genetics , Polymorphism, Single Nucleotide , Thyroiditis, Autoimmune/genetics , Genetic Association Studies , Genetic Linkage , Genetic Predisposition to Disease , Humans , Sequence Analysis, DNA , Sweden , White People/genetics
15.
PLoS One ; 11(7): e0159850, 2016.
Article in English | MEDLINE | ID: mdl-27441847

ABSTRACT

Here we characterize a new animal model that spontaneously develops chronic inflammation and fibrosis in multiple organs, the non-obese diabetic inflammation and fibrosis (N-IF) mouse. In the liver, the N-IF mouse displays inflammation and fibrosis particularly evident around portal tracts and central veins and accompanied with evidence of abnormal intrahepatic bile ducts. The extensive cellular infiltration consists mainly of macrophages, granulocytes, particularly eosinophils, and mast cells. This inflammatory syndrome is mediated by a transgenic population of natural killer T cells (NKT) induced in an immunodeficient NOD genetic background. The disease is transferrable to immunodeficient recipients, while polyclonal T cells from unaffected syngeneic donors can inhibit the disease phenotype. Because of the fibrotic component, early on-set, spontaneous nature and reproducibility, this novel mouse model provides a unique tool to gain further insight into the underlying mechanisms mediating transformation of chronic inflammation into fibrosis and to evaluate intervention protocols for treating conditions of fibrotic disorders.


Subject(s)
Hepatitis, Chronic/etiology , Hepatitis, Chronic/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/pathology , Adoptive Transfer , Animals , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/pathology , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Hepatitis, Chronic/metabolism , Inflammation Mediators/metabolism , Liver Cirrhosis/metabolism , Lymphocyte Activation/immunology , Mice , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
16.
Diabetologia ; 59(3): 550-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26613896

ABSTRACT

AIMS/HYPOTHESIS: It is generally accepted that structural and functional quantitative imaging of individual islets would be beneficial to elucidate the pathogenesis of type 1 diabetes. We here introduce functional optical coherence imaging (FOCI) for fast, label-free monitoring of beta cell destruction and associated alterations of islet vascularisation. METHODS: NOD mouse and human islets transplanted into the anterior chamber of the eye (ACE) were imaged with FOCI, in which the optical contrast of FOCI is based on intrinsic variations of the index of refraction resulting in a faster tomographic acquisition. In addition, the phase sensitivity allows simultaneous label-free acquisition of vascularisation. RESULTS: We demonstrate that FOCI allows longitudinal quantification of progressive autoimmune insulitis, including the three-dimensional quantification of beta cell volume, inflammation and vascularisation. The substantially increased backscattering of islets is dominated by the insulin-zinc nanocrystals in the beta cell granules. This translates into a high specificity for the functional beta cell volume of islets. Applying FOCI to a spontaneous mouse model of type 1 diabetes, we quantify the modifications of the pancreatic microvasculature accompanying the progression of diabetes and reveal a strong correlation between increasing insulitis and density of the vascular network of the islet. CONCLUSIONS/INTERPRETATION: FOCI provides a novel imaging technique for investigating functional and structural diabetes-induced alterations of the islets. The label-free detection of beta cell volume and infiltration together with vascularisation offers a unique extension to study ACE-transplanted human islets. These results are contributing to a deeper understanding of human islet transplant rejection and label-free in vivo monitoring of drug efficacy.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Animals , Disease Models, Animal , Genotype , Humans , Insulin-Secreting Cells/pathology , Islets of Langerhans/pathology , Islets of Langerhans Transplantation , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout
17.
PLoS One ; 10(12): e0144090, 2015.
Article in English | MEDLINE | ID: mdl-26624013

ABSTRACT

Autoimmune diabetes is a consequence of immune-cell infiltration and destruction of pancreatic ß-cells in the islets of Langerhans. We analyzed the cellular composition of the insulitic lesions in the autoimmune-prone non-obese diabetic (NOD) mouse and observed a peak in recruitment of plasmacytoid dendritic cells (pDCs) to NOD islets around 8-9 weeks of age. This peak coincides with increased spontaneous expression of type-1-IFN response genes and CpG1585 induced production of IFN-α from NOD islets. The transcription factor E2-2 is specifically required for the maturation of pDCs, and we show that knocking out E2-2 conditionally in CD11c+ cells leads to a reduced recruitment of pDCs to pancreatic islets and reduced CpG1585 induced production of IFN-α during insulitis. As a consequence, insulitis has a less aggressive expression profile of the Th1 cytokine IFN-γ and a markedly reduced diabetes incidence. Collectively, these observations demonstrate a disease-promoting role of E2-2 dependent pDCs in the pancreas during autoimmune diabetes in the NOD mouse.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Dendritic Cells/metabolism , Diabetes Mellitus, Type 1/metabolism , Animals , Female , Interferon-alpha/metabolism , Interferon-gamma/metabolism , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Pancreas/metabolism , Transcription Factor 4 , Tumor Necrosis Factor-alpha/metabolism
18.
Gene ; 545(1): 80-7, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24797614

ABSTRACT

The Cd247 gene encodes for a transmembrane protein important for the expression and assembly of TCR/CD3 complex on the surface of T lymphocytes. Down-regulation of CD247 has functional consequences in systemic autoimmunity and has been shown to be associated with Type 1 Diabetes in NOD mouse. In this study, we have utilized the wealth of high-throughput sequencing data produced during the Encyclopedia of DNA Elements (ENCODE) project to identify spatially conserved regulatory elements within the Cd247 gene from human and mouse. We show the presence of two transcription factor binding sites, supported by histone marks and ChIP-seq data, that specifically have features of an enhancer and a promoter, respectively. We also identified a putative long non-coding RNA from the characteristically long first intron of the Cd247 gene. The long non-coding RNA annotation is supported by manual annotations from the GENCODE project in human and our expression quantification analysis performed in NOD and B6 mice using qRT-PCR. Furthermore, 17 of the 23 SNPs already known to be implicated with T1D were observed within the long non-coding RNA region in mouse. The spatially conserved regulatory elements identified in this study have the potential to enrich our understanding of the role of Cd247 gene in autoimmune diabetes.


Subject(s)
CD3 Complex/genetics , RNA, Long Noncoding , Regulatory Sequences, Nucleic Acid , Animals , Base Sequence , Binding Sites , Conserved Sequence , Evolution, Molecular , Female , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Transcription Factors/genetics , Transcription Factors/metabolism
19.
PLoS One ; 8(10): e76330, 2013.
Article in English | MEDLINE | ID: mdl-24124545

ABSTRACT

Multiple Sclerosis (MS) is a progressive autoimmune inflammatory and demyelinating disease of the central nervous system (CNS). T cells play a key role in the progression of neuroinflammation in MS and also in the experimental autoimmune encephalomyelitis (EAE) animal models for the disease. A technology for quantitative and 3 dimensional (3D) spatial assessment of inflammation in this and other CNS inflammatory conditions is much needed. Here we present a procedure for 3D spatial assessment and global quantification of the development of neuroinflammation based on Optical Projection Tomography (OPT). Applying this approach to the analysis of rodent models of MS, we provide global quantitative data of the major inflammatory component as a function of the clinical course. Our data demonstrates a strong correlation between the development and progression of neuroinflammation and clinical disease in several mouse and a rat model of MS refining the information regarding the spatial dynamics of the inflammatory component in EAE. This method provides a powerful tool to investigate the effect of environmental and genetic forces and for assessing the therapeutic effects of drug therapy in animal models of MS and other neuroinflammatory/neurodegenerative disorders.


Subject(s)
Imaging, Three-Dimensional/methods , Multiple Sclerosis/diagnosis , Tomography, Optical/methods , Animals , Central Nervous System/immunology , Central Nervous System/pathology , Demyelinating Diseases/diagnosis , Demyelinating Diseases/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/diagnosis , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/diagnosis , Inflammation/immunology , Mice , Multiple Sclerosis/immunology , Rats , T-Lymphocyte Subsets/metabolism
20.
Diabetologia ; 56(12): 2669-78, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23963325

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to visualise the dynamics and interactions of the cells involved in autoimmune-driven inflammation in type 1 diabetes. METHODS: We adopted the anterior chamber of the eye (ACE) transplantation model to perform non-invasive imaging of leucocytes infiltrating the endocrine pancreas during initiation and progression of insulitis in the NOD mouse. Individual, ACE-transplanted islets of Langerhans were longitudinally and repetitively imaged by stereomicroscopy and two-photon microscopy to follow fluorescently labelled leucocyte subsets. RESULTS: We demonstrate that, in spite of the immune privileged status of the eye, the ACE-transplanted islets develop infiltration and beta cell destruction, recapitulating the autoimmune insulitis of the pancreas, and exemplify this by analysing reporter cell populations expressing green fluorescent protein under the Cd11c or Foxp3 promoters. We also provide evidence that differences in morphological appearance of subpopulations of infiltrating leucocytes can be correlated to their distinct dynamic behaviour. CONCLUSIONS/INTERPRETATION: Together, these findings demonstrate that the kinetics and dynamics of these key cellular components of autoimmune diabetes can be elucidated using this imaging platform for single cell resolution, non-invasive and repetitive monitoring of the individual islets of Langerhans during the natural development of autoimmune diabetes.


Subject(s)
Anterior Chamber/pathology , Autoimmunity , CD11c Antigen/immunology , Diabetes Mellitus, Type 1/pathology , Forkhead Transcription Factors/immunology , Inflammation/pathology , Islets of Langerhans/pathology , Prediabetic State/pathology , Animals , Anterior Chamber/immunology , Autoantibodies/blood , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/immunology , Disease Progression , Female , Flow Cytometry , Inflammation/immunology , Islets of Langerhans/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Microscopy, Fluorescence , Prediabetic State/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...