Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Biol Rep ; 51(1): 203, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38270651

ABSTRACT

BACKGROUND: Recovery from a foot ulcer is compromised in a diabetic status, due to the impaired tissue microenvironment that consists of altered inflammation, angiogenesis and fibrosis. Phenotypic alterations in both macrophages and fibroblasts have been detected in the diabetic wound. Recently, a fibroblast subpopulation that expresses high matrix metalloproteinase 1 (MMP1), MMP3, MMP11 and Chitinase-3-Like Protein 1 (CHI3L1) was associated with a successful diabetic wound healing. However, it is not known whether these healing-associated fibroblasts are regulated by macrophages. METHODS AND RESULTS: We used bioinformatic tools to analyze selected public databases on normal and diabetic skin from patients, and identified genes significantly altered in diabetes. In a mouse model for diabetic wound healing, we detected not only a loss of the spatiotemporal changes in interleukin 1ß (IL1ß), IL6, IL10 and vascular endothelial growth factor A (VEGF-A) in wound macrophages, but also a compromised expression of MMP1, MMP3, MMP11, CHI3L1 and VEGF-A in healing-associated wound fibroblasts in a diabetic status. Co-culture with diabetic macrophages significantly reduced the expression of MMP1, MMP3, MMP11, CHI3L1 and VEGF-A in fibroblasts from non-diabetic wound. Co-culture with non-diabetic macrophages or diabetic macrophages supplied with IL6 significantly increased the expression of MMP1, MMP3, MMP11, CHI3L1 and VEGF-A in fibroblasts from diabetic wound. Moreover, macrophage-specific expression of IL6 significantly improved wound healing and angiogenesis in diabetic mice. CONCLUSIONS: Macrophages may induce the activation of wound-healing-associated fibroblasts, while the defective macrophages in diabetes may be corrected with IL6 treatment as a promising therapy for diabetic foot disease.


Subject(s)
Diabetes Mellitus, Experimental , Vascular Endothelial Growth Factor A , Humans , Animals , Mice , Matrix Metalloproteinase 3 , Matrix Metalloproteinase 1 , Matrix Metalloproteinase 11 , Interleukin-6 , Wound Healing
2.
Nat Cell Biol ; 25(3): 493-507, 2023 03.
Article in English | MEDLINE | ID: mdl-36849558

ABSTRACT

How abnormal neurodevelopment relates to the tumour aggressiveness of medulloblastoma (MB), the most common type of embryonal tumour, remains elusive. Here we uncover a neurodevelopmental epigenomic programme that is hijacked to induce MB metastatic dissemination. Unsupervised analyses of integrated publicly available datasets with our newly generated data reveal that SMARCD3 (also known as BAF60C) regulates Disabled 1 (DAB1)-mediated Reelin signalling in Purkinje cell migration and MB metastasis by orchestrating cis-regulatory elements at the DAB1 locus. We further identify that a core set of transcription factors, enhancer of zeste homologue 2 (EZH2) and nuclear factor I X (NFIX), coordinates with the cis-regulatory elements at the SMARCD3 locus to form a chromatin hub to control SMARCD3 expression in the developing cerebellum and in metastatic MB. Increased SMARCD3 expression activates Reelin-DAB1-mediated Src kinase signalling, which results in a MB response to Src inhibition. These data deepen our understanding of how neurodevelopmental programming influences disease progression and provide a potential therapeutic option for patients with MB.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Humans , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Medulloblastoma/genetics , Phosphorylation , Epigenomics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion Molecules, Neuronal/pharmacology , Cerebellar Neoplasms/genetics , Epigenesis, Genetic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism
3.
Front Immunol ; 14: 1084056, 2023.
Article in English | MEDLINE | ID: mdl-36733483

ABSTRACT

The contribution of bone-marrow derived cells (BMCs) to a newly formed beta-cell population in adults is controversial. Previous studies have only used models of bone marrow transplantation from sex-mismatched donors (or other models of genetic labeling) into recipient animals that had undergone irradiation. This approach suffers from the significant shortcoming of the off-target effects of irradiation. Partial pancreatic duct ligation (PDL) is a mouse model of acute pancreatitis with a modest increase in beta-cell number. However, the possibility that recruited BMCs in the inflamed pancreas may convert into beta-cells has not been examined. Here, we used an irradiation-free model to track the fate of the BMCs from the donor mice. A ROSA-mTmG red fluorescent mouse was surgically joined to an INS1Cre knock-in mouse by parabiosis to establish a mixed circulation. PDL was then performed in the INS1Cre mice 2 weeks after parabiosis, which was one week after establishment of the stable blood chimera. The contribution of red cells from ROSA-mTmG mice to beta-cells in INS1Cre mouse was evaluated based on red fluorescence, while cell fusion was evaluated by the presence of green fluorescence in beta-cells. We did not detect any red or green insulin+ cells in the INS1Cre mice, suggesting that there was no contribution of BMCs to the newly formed beta-cells, either by direct differentiation, or by cell fusion. Thus, the contribution of BMCs to beta-cells in the inflamed pancreas should be minimal, if any.


Subject(s)
Bone Marrow , Pancreatitis , Mice , Animals , Acute Disease , Bone Marrow Cells , Pancreas
4.
Diabetes Obes Metab ; 24(9): 1721-1733, 2022 09.
Article in English | MEDLINE | ID: mdl-35546452

ABSTRACT

AIM: To show that depletion of pancreatic macrophages impairs gestational beta cell proliferation and leads to glucose intolerance. MATERIALS AND METHODS: Genetic animal models were applied to study the effects of depletion of pancreatic macrophges on gestational beta-cell proliferaiton and glucose response. The crosstalk between macrophages and beta-cells was studied in vivo using beta-cell-specific extracellular-signal-regulated kinase 5 (ERK5) knockout and epidermal growth receptor (EGFR) knockout mice, and in vitro using a co-culture system. RESULTS: Beta cell-derived placental growth factor (PlGF) recruited naïve macrophages and polarized them towards an M2-like phenotype. These macrophages then secreted epidermal growth factor (EGF), which activated extracellular signal-regulated kinase 5 (ERK5) signalling in beta cells to promote gestational beta cell proliferation. On the other hand, activation of ERK5 signalling in beta cells likely, in turn, enhanced the production and secretion of PlGF by beta cells. CONCLUSIONS: Our study shows a regulatory loop between macrophages and beta cells through PlGF/EGF/ERK5 signalling cascades to regulate gestational beta cell growth.


Subject(s)
Epidermal Growth Factor , Mitogen-Activated Protein Kinase 7 , Animals , Cell Proliferation , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Female , Macrophages/metabolism , Mice , Mitogen-Activated Protein Kinase 7/metabolism , Placenta Growth Factor/metabolism
5.
Lab Invest ; 102(3): 290-297, 2022 03.
Article in English | MEDLINE | ID: mdl-34764436

ABSTRACT

Clinical evidence suggests that patients with chronic pancreatitis (CP) are prone to development of diabetes (chronic pancreatitis-related diabetes; CPRD), whereas the underlying mechanisms are not fully determined. Recently, we showed that the gradual loss of functional beta-cells in a mouse model for CPRD, partial pancreatic duct ligation (PDL), results from a transforming growth factor ß1 (TGFß1)-triggered beta-cell epithelial-mesenchymal transition (EMT), rather than from apoptotic beta-cell death. Here, the role of angiogenesis in CPRD-associated beta-cell EMT was addressed. We detected enhanced angiogenesis in the inflamed pancreas from CP patients by bioinformatic analysis and from PDL-mice. Inhibition of angiogenesis by specific antisera for vascular endothelial growth factor receptor 2 (VEGFR2), DC101, did not alter the loss of beta-cells and the fibrotic process in PDL-pancreas. However, DC101-mediated inhibition of angiogenesis abolished pancreatitis-induced beta-cell EMT and rendered it to apoptotic beta-cell death. Thus, our data suggest that angiogenesis promotes beta-cell survival in the inflamed pancreas, while suppression of angiogenesis turns beta-cell EMT into apoptotic beta-cell death. This finding could be informative during development of intervention therapies for CPRD.


Subject(s)
Diabetes Mellitus/genetics , Epithelial-Mesenchymal Transition/genetics , Insulin-Secreting Cells/metabolism , Neovascularization, Pathologic/genetics , Pancreatitis, Chronic/genetics , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Profiling/methods , Humans , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/prevention & control , Pancreatitis, Chronic/complications , Pancreatitis, Chronic/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
6.
Front Immunol ; 12: 736153, 2021.
Article in English | MEDLINE | ID: mdl-34659227

ABSTRACT

Diabetic foot disease (DFD) is a common and serious complication for diabetes and is characterized with impaired angiogenesis. In addition to the well-defined role of vascular endothelial growth factor (VEGF) -A and its defect in the pathogenesis of DFD, another VEGF family member, placental growth factor (PlGF), was also recently found to alter expression pattern in the DFD patients with undetermined mechanisms. This question was thus addressed in the current study. We detected attenuated PlGF upregulation in a mouse DFD model. In addition, the major cell types at the wound to express the unique PlGF receptor, VEGF receptor 1 (VEGFR1), were macrophages and endothelial cells. To assess how PlGF regulates DFD-associated angiogenesis, we injected recombinant PlGF and depleted VEGF1R specifically in macrophages by local injection of an adeno-associated virus (AAV) carrying siRNA for VEGFR1 under a macrophage-specific CD68 promoter. We found that the angiogenesis and recovery of the DFD were both improved by PlGF injection. The PlGF-induced improvement in angiogenesis and the recovery of skin injury were largely attenuated by macrophage-specific depletion of VEGF1R, likely resulting from reduced macrophage number and reduced M2 polarization. Together, our data suggest that reduced PlGF compromises angiogenesis in DFD at least partially through macrophages.


Subject(s)
Diabetic Foot/metabolism , Foot/blood supply , Macrophages/metabolism , Neovascularization, Pathologic , Placenta Growth Factor/metabolism , Angiogenesis Inducing Agents/pharmacology , Animals , Dependovirus/genetics , Diabetic Foot/drug therapy , Diabetic Foot/genetics , Diabetic Foot/pathology , Disease Models, Animal , Down-Regulation , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Genetic Vectors , Macrophages/drug effects , Male , Mice, Inbred C57BL , Placenta Growth Factor/genetics , Placenta Growth Factor/pharmacology , RNA Interference , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Wound Healing
8.
Exp Mol Med ; 53(4): 605-614, 2021 04.
Article in English | MEDLINE | ID: mdl-33820959

ABSTRACT

The adult pancreatic ductal system was suggested to harbor facultative beta-cell progenitors similar to the embryonic pancreas, and the appearance of insulin-positive duct cells has been used as evidence for natural duct-to-beta-cell reprogramming. Nevertheless, the phenotype and fate of these insulin-positive cells in ducts have not been determined. Here, we used a cell-tagging dye, CFDA-SE, to permanently label pancreatic duct cells through an intraductal infusion technique. Representing a time when significant increases in beta-cell mass occur, pregnancy was later induced in these CFDA-SE-treated mice to assess the phenotype and fate of the insulin-positive cells in ducts. We found that a small portion of CFDA-SE-labeled duct cells became insulin-positive, but they were not fully functional beta-cells based on the in vitro glucose response and the expression levels of key beta-cell genes. Moreover, these insulin-positive cells in ducts expressed significantly lower levels of genes associated with extracellular matrix degradation and cell migration, which may thus prevent their budding and migration into preexisting islets. A similar conclusion was reached through analysis of the Gene Expression Omnibus database for both mice and humans. Together, our data suggest that the contribution of duct cells to normal beta-cells in adult islets is minimal at best.


Subject(s)
Cell Movement , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Pancreatic Ducts/cytology , Animals , Biomarkers , Cell Differentiation , Computational Biology/methods , Extracellular Matrix , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunophenotyping , Mice , Mice, Transgenic , Pregnancy
9.
Growth Factors ; 37(3-4): 170-177, 2019 08.
Article in English | MEDLINE | ID: mdl-31576764

ABSTRACT

Lowe syndrome is an x-linked disorder characterized by congenital cataracts, nervous system abnormalities and renal tubular dysfunction. With the rising number of reported cases, more patients are found to suffer from endocrine abnormalities. Hereby, three Chinese patients with typical symptoms and extremely short stature were described. The OCRL gene was analyzed. A combination of blood biochemistry and radiological examinations were performed. Growth hormone provocation test was taken in one patient. Nucleotide sequence analysis revealed a de novo novel hemizygous mutation (c.2290_2291delinsCT) in exon 21 in an adolescent boy. As indicated by the growth hormone provocation test, the boy had growth hormone deficiency. The other two patients were brothers with extremely short stature, and manifested the same hemizygous mutation (c.2581G > A) in exon 23. It was speculated that the mutation of OCRL gene could lead to deficiency of growth hormone, for which an early growth hormone intervention may be beneficial.


Subject(s)
Child Development/physiology , Human Growth Hormone/deficiency , Oculocerebrorenal Syndrome/genetics , Phosphoric Monoester Hydrolases/genetics , Adolescent , Base Sequence , Body Height/genetics , Child , Child, Preschool , China , Humans , Male , Mutation/genetics , Oculocerebrorenal Syndrome/pathology , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL