Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters










Publication year range
2.
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
3.
Cancer Discov ; 12(12): 2820-2837, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36122307

ABSTRACT

Isocitrate dehydrogenase (IDH) wild-type glioblastoma (GBM) has a dismal prognosis. A better understanding of tumor evolution holds the key to developing more effective treatment. Here we study GBM's natural evolutionary trajectory by using rare multifocal samples. We sequenced 61,062 single cells from eight multifocal IDH wild-type primary GBMs and defined a natural evolution signature (NES) of the tumor. We show that the NES significantly associates with the activation of transcription factors that regulate brain development, including MYBL2 and FOSL2. Hypoxia is involved in inducing NES transition potentially via activation of the HIF1A-FOSL2 axis. High-NES tumor cells could recruit and polarize bone marrow-derived macrophages through activation of the FOSL2-ANXA1-FPR1/3 axis. These polarized macrophages can efficiently suppress T-cell activity and accelerate NES transition in tumor cells. Moreover, the polarized macrophages could upregulate CCL2 to induce tumor cell migration. SIGNIFICANCE: GBM progression could be induced by hypoxia via the HIF1A-FOSL2 axis. Tumor-derived ANXA1 is associated with recruitment and polarization of bone marrow-derived macrophages to suppress the immunoenvironment. The polarized macrophages promote tumor cell NES transition and migration. This article is highlighted in the In This Issue feature, p. 2711.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/genetics , Glioblastoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Isocitrate Dehydrogenase/genetics , Prognosis , Hypoxia/genetics
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.
Acta Biomater ; 143: 310-319, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35278688

ABSTRACT

OBJECTIVE: Urogynecology meshes, typically manufactured from polypropylene, are widely used in the surgical treatment of stress urinary incontinence and pelvic organ prolapse. However, mesh-associated complications such as mesh exposure can develop in women undergoing mesh implantation, for which diabetes is an independent risk factor. We aimed to define the impact of diabetes on the vaginal immune response to mesh by comparing diabetic vs. normoglycemic conditions longitudinally in a rat sacrocolpopexy model. METHODS: Diabetes (blood glucose ≥ 300 mg/dL) was induced in middle-aged female Wistar rats with streptozotocin (STZ). A polypropylene mesh was implanted on the vagina via modified sacrocolpopexy following bilateral ovariectomy and supracervical hysterectomy for 3-, 7-, and 42-days. Sham-operated controls underwent the same procedures without mesh. Mesh-associated inflammation, immune cell populations and cytokine/chemokine profiles were examined in the excised vaginal tissues. RESULTS: Diabetes was reliably induced starting on the 3rd day following STZ injection. Under both normoglycemic and diabetic conditions, mesh caused a prolonged inflammatory response in the vagina with increased proinflammatory chemokines MCP-1 and MIP-1α as compared to Sham. Major differences between the two conditions were found at the later stage (42 days post-surgery), including an increased inflammation with larger foreign body granuloma and more giant cells at the mesh-tissue interface, increased fraction of macrophages in the immune cell population, and higher proinflammatory chemokine IP-10 in the diabetic group. CONCLUSION: Polypropylene mesh implanted on the vagina induces prolonged inflammation at the mesh-tissue interface. Diabetes increases the mesh-associated inflammation in the long term, which is related to a dysregulated macrophage response. STATEMENT OF SIGNIFICANCE: This study investigated the mechanism underlying the increased risk in women with diabetes for developing mesh complications such as mesh exposure. The significance includes: (1) it is the first study investigating vaginal host response to a prosthesis under the influence of diabetes; (2) the longitudinal study design elucidated the dynamic changes of vaginal immune response to mesh from very early to late stages; (3) our findings may inform future mechanistic studies and studies investigating preventive/therapeutic strategies to improve the outcomes of women with diabetes receiving vaginal implants.


Subject(s)
Diabetes Mellitus , Polypropylenes , Animals , Female , Humans , Immunity , Inflammation , Longitudinal Studies , Middle Aged , Rats , Rats, Wistar , Surgical Mesh , Vagina/surgery
6.
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
7.
J Vis Exp ; (175)2021 09 30.
Article in English | MEDLINE | ID: mdl-34661584

ABSTRACT

The pancreas is a bifunctional organ with both endocrine and exocrine components. A number of pathologies can afflict the pancreas, including diabetes, pancreatitis, and pancreatic cancer. All three of these diseases mark active areas of study, not only to develop immediate therapy, but also to better understand their pathophysiology. There are few tools to further these areas of study. Pancreatic duct infusion is an important technique that can allow for lineage tracing, gene introduction, and cell line-specific targeting. The technique requires the intricate dissection of the second portion of the duodenum and ampulla, followed by the occlusion of the bile duct and the cannulation of the pancreatic duct. Although the technique is technically challenging at first, the applications are myriad. Ambiguity in the specifics of the procedure between groups highlighted the need for a standard protocol. This work describes the expression of a green fluorescent protein (GFP) within the pancreas after the pancreatic duct infusion of a viral vector expressing GFP versus a sham surgery. The infusion and therefore expression is specific to the pancreas, without expression present in any other tissue type.


Subject(s)
Pancreatic Neoplasms , Pharmaceutical Preparations , Duodenum , Humans , Pancreas/surgery , Pancreatic Ducts
8.
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
10.
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
11.
Gastroenterology ; 159(3): 1036-1050.e8, 2020 09.
Article in English | MEDLINE | ID: mdl-32445858

ABSTRACT

BACKGROUND & AIMS: Calcineurin is a ubiquitously expressed central Ca2+-responsive signaling molecule that mediates acute pancreatitis, but little is known about its effects. We compared the effects of calcineurin expression by hematopoietic cells vs pancreas in mouse models of pancreatitis and pancreatitis-associated lung inflammation. METHODS: We performed studies with mice with hematopoietic-specific or pancreas-specific deletion of protein phosphatase 3, regulatory subunit B, alpha isoform (PPP3R1, also called CNB1), in mice with deletion of CNB1 (Cnb1UBC△/△) and in the corresponding controls for each deletion of CNB1. Acute pancreatitis was induced in mice by administration of caerulein or high-pressure infusion of radiocontrast into biliopancreatic ducts; some mice were also given intraductal infusions of an adeno-associated virus vector that expressed nuclear factor of activated T -cells (NFAT)-luciferase into pancreas. Pancreas, bone marrow, liver, kidney, heart, and lung were collected and analyzed by histopathology, immunohistochemistry, and immunoblots; levels of cytokines were measured in serum. Mouse and human primary pancreatic acinar cells were transfected with a vector that expressed NFAT-luciferase and incubated with an agent that blocks interaction of NFAT with calcineurin; cells were analyzed by immunofluorescence. Calcineurin-mediated neutrophil chemotaxis and reactive oxygen species production were measured in neutrophils from mice. RESULTS: Mice with hematopoietic-specific deletion of CNB1 developed the same level of local pancreatic inflammation as control mice after administration of caerulein or infusion of radiocontrast into biliopancreatic ducts. Cnb1UBC△/△ mice or mice with pancreas-specific deletion of CNB1 developed less severe pancreatitis and reduced pancreatic inflammation after administration of caerulein or infusion of radiocontrast into biliopancreatic ducts compared with control mice. NFAT was activated in pancreas of Swiss Webster mice given caerulein or infusions of radiocontrast into biliopancreatic ducts. Blocking the interaction between calcineurin and NFAT did not reduce pancreatic acinar cell necrosis in response to caerulein or infusions of radiocontrast. Mice with hematopoietic-specific deletion of CNB1 (but not mice with pancreas-specific deletion of CNB1) had reduced infiltration of lung tissues by neutrophils. Neutrophil chemotaxis and production of reactive oxygen species were decreased after incubation with a calcineurin inhibitor. CONCLUSIONS: Hematopoietic and neutrophil expression of calcineurin promotes pancreatitis-associated lung inflammation, whereas pancreatic calcineurin promotes local pancreatic inflammation. The findings indicate that the protective effects of blocking or deleting calcineurin on pancreatitis are mediated by the source of its expression. This information should be used in the development of strategies to inhibit calcineurin for the prevention of pancreatitis and pancreatitis-associated lung inflammation.


Subject(s)
Acute Lung Injury/immunology , Calcineurin Inhibitors/therapeutic use , Calcineurin/metabolism , Calcium-Binding Proteins/metabolism , Muscle Proteins/metabolism , Pancreatitis/immunology , Acinar Cells/metabolism , Acute Lung Injury/blood , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Calcineurin/genetics , Calcineurin/immunology , Calcium-Binding Proteins/genetics , Cells, Cultured , Ceruletide/administration & dosage , Ceruletide/toxicity , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Muscle Proteins/genetics , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Pancreas/cytology , Pancreas/immunology , Pancreas/metabolism , Pancreatitis/chemically induced , Pancreatitis/complications , Pancreatitis/drug therapy , Primary Cell Culture
12.
Article in English | MEDLINE | ID: mdl-32144129

ABSTRACT

OBJECTIVE: Pancreatic beta cells proliferate in response to metabolic requirements during pregnancy, while failure of this response may cause gestational diabetes. A member of the vascular endothelial growth factor family, placental growth factor (PlGF), typically plays a role in metabolic disorder and pathological circumstance. The expression and function of PlGF in the endocrine pancreas have not been reported and are addressed in the current study. RESEARCH DESIGN AND METHODS: PlGF levels in beta cells were determined by immunostaining or ELISA in purified beta cells in non-pregnant and pregnant adult mice. An adeno-associated virus (AAV) serotype 8 carrying a shRNA for PlGF under the control of a rat insulin promoter (AAV-rat insulin promoter (RIP)-short hairpin small interfering RNA for PlGF (shPlGF)) was prepared and infused into mouse pancreas through the pancreatic duct to specifically knock down PlGF in beta cells, and its effects on beta-cell growth were determined by beta-cell proliferation, beta-cell mass and insulin release. A macrophage-depleting reagent, clodronate, was coapplied into AAV-treated mice to study crosstalk between beta cells and macrophages. RESULTS: PlGF is exclusively produced by beta cells in the adult mouse pancreas. Moreover, PlGF expression in beta cells was significantly increased during pregnancy. Intraductal infusion of AAV-RIP-shPlGF specifically knocked down PlGF in beta cells, resulting in compromised beta-cell proliferation, reduced growth in beta-cell mass and impaired glucose tolerance during pregnancy. Mechanistically, PlGF depletion in beta cells reduced islet infiltration of trophic macrophages, which appeared to be essential for gestational beta-cell growth. CONCLUSIONS: Our study suggests that increased expression of PlGF in beta cells may trigger gestational beta-cell growth through recruited macrophages.


Subject(s)
Insulin-Secreting Cells/metabolism , Placenta Growth Factor/metabolism , Animals , Cell Enlargement , Cell Proliferation , Female , Glucose/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy
13.
Article in English | MEDLINE | ID: mdl-31421261

ABSTRACT

Asparaginase (ASNase) causes pancreatitis in approximately 10% of leukemia patients, and the mechanisms underlying this painful complication are not known. ASNase primarily depletes circulating asparagine, and the endogenously expressed enzyme, asparagine synthetase (ASNS), replenishes asparagine. ASNS was suggested previously to be highly expressed in the pancreas. In this study, we determined the expression pattern of ASNS in the pancreas and the mechanism for increased pancreatic ASNS abundance. Compared with other organs, ASNS was highly expressed in both the human and mouse pancreas, and, within the pancreas, ASNS was present primarily in the acinar cells. The high baseline pancreatic ASNS was associated with higher baseline activation of protein kinase R-like endoplasmic reticulum kinase (PERK) signaling in the pancreas, and inhibition of PERK in acinar cells lessened ASNS expression. ASNase exposure, but not the common pancreatitis triggers, uniquely up-regulated ASNS expression, indicating that the increase is mediated by nutrient stress. The up-regulation of acinar ASNS with ASNase exposure was owing to increased transcriptional rather than delayed degradation. Knockdown of ASNS in the 266-6 acinar cells provoked acinar cell injury and worsened ASNase-induced injury, whereas ASNS overexpression protected against ASNase-induced injury. In summary, ASNS is highly expressed in the pancreatic acinar cells through heightened basal activation of PERK, and ASNS appears to be crucial to maintaining acinar cell integrity. The implications are that ASNS is especially hardwired in the pancreas to protect against both baseline perturbations and nutrient deprivation stressors, such as during ASNase exposure.


Subject(s)
Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/metabolism , Pancreas/pathology , Pancreatitis/pathology , eIF-2 Kinase/metabolism , Acinar Cells/pathology , Animals , Asparaginase/administration & dosage , Asparaginase/metabolism , Asparagine/metabolism , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/genetics , Cell Line , Disease Models, Animal , Female , Gene Knockdown Techniques , Humans , Mice , Pancreas/cytology , Primary Cell Culture , Signal Transduction/drug effects , Up-Regulation , eIF-2 Kinase/antagonists & inhibitors
14.
Aging (Albany NY) ; 11(24): 12236-12245, 2019 12 21.
Article in English | MEDLINE | ID: mdl-31866580

ABSTRACT

Diabetic foot (DF) is a common complication of high severity for diabetes, a prevalent metabolic disorder that affects billions of people worldwide. Mesenchymal stem cells (MSCs) have a demonstrative therapeutic effect on DF, through their generation of pro-angiogenesis factors, like vascular endothelial growth factor (VEGF). Recently, genetically modified MSCs have been used in therapy and we have shown that depletion of micoRNA-205-5p (miR-205-5p) in human MSCs promotes VEGF-mediated therapeutic effects on DF. Here, we showed that a long non-coding RNA (lncRNA), MALAT1, is a competing endogenous RNA (ceRNA) for miR-205-5p, and is low expressed in human MSCs. Ectopic expression of MALAT1 in human MSCs significantly decreased miR-205-5p levels, resulting in upregulation of VEGF production and improved in vitro endothelial cell tube formation. In a DF model in immunodeficient NOD/SCID mice, transplantation of human miR-205-5p-depleted MSCs exhibited better therapeutic effects on DF recovery than control MSCs. Moreover, MALAT1-expressing MSCs showed even better therapeutic effects on DF recovery than miR-205-5p-depleted MSCs. This difference in DF recovery was shown to be associated with the levels of on-site vascularization. Together, our data suggest that MALAT1 functions as a sponge RNA for miR-205-5p to increase therapeutic effects of MSCs on DF.


Subject(s)
Diabetic Foot/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Animals , Diabetes Mellitus, Experimental/complications , Gene Expression Regulation , Humans , Male , Mice , RNA, Long Noncoding/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
15.
Development ; 146(13)2019 06 27.
Article in English | MEDLINE | ID: mdl-31160417

ABSTRACT

The Cre/loxP system has been used extensively in mouse models with a limitation of one lineage at a time. Differences in function and other properties among populations of adult ß-cells is termed ß-cell heterogeneity, which was recently associated with diabetic phenotypes. Nevertheless, the presence of a developmentally derived ß-cell heterogeneity is unclear. Here, we have developed a novel dual lineage-tracing technology, using a combination of two recombinase systems, Dre/RoxP and Cre/LoxP, to independently trace green fluorescent Pdx1-lineage cells and red fluorescent Ptf1a-lineage cells in the developing and adult mouse pancreas. We detected a few Pdx1+/Ptf1a- lineage cells in addition to the vast majority of Pdx1+/Ptf1a+ lineage cells in the pancreas. Moreover, Pdx1+/Ptf1a+ lineage ß-cells had fewer Ki-67+ proliferating ß-cells, and expressed higher mRNA levels of insulin, Glut2, Pdx1, MafA and Nkx6.1, but lower CCND1 and CDK4 levels, compared with Pdx1+/Ptf1a- lineage ß-cells. Furthermore, more TSQ-high, SSC-high cells were detected in the Pdx1+Ptf1a+ lineage population than in the Pdx1+Ptf1a- lineage population. Together, these data suggest that differential activation of Ptf1a in the developing pancreas may correlate with this ß-cell heterogeneity.


Subject(s)
Cell Lineage , Cell Tracking/methods , Insulin-Secreting Cells/cytology , Pancreas/cytology , Stem Cells/cytology , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Separation/methods , Cells, Cultured , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Imaging/methods , Organogenesis/genetics , Pancreas/embryology , Pancreas/growth & development , Pancreas/metabolism , Stem Cells/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Aging (Albany NY) ; 11(9): 2787-2796, 2019 05 06.
Article in English | MEDLINE | ID: mdl-31061237

ABSTRACT

Endoplasmic reticulum (ER) stress in beta cells induces a signaling network called the unfolded protein response (UPR), which plays a dual role in diabetes. A key regulator of ER-stress and UPR, the orosomucoid 1-like protein 3 (ORMDL3), has been shown to regulate airway remodeling through a major UPR protein, activating transcription factor 6 (ATF6), but the contribution of this regulatory axis to compensatory pancreatic beta cell proliferation in diabetes has not been studied. Here, we detected significantly lower levels of ORMDL3 mRNA in leukocytes of peripheral blood specimens from type 1 diabetes (T1D) children, compared to normal children. Moreover, these ORMDL3 levels in T1D children exhibited further decreases upon follow-up. ORMDL3 levels in islets from NOD mice, a mouse model for T1D in humans, showed a mild increase before diabetes onset, but a gradual decrease subsequently. In high glucose culture, beta cell proliferation, but not apoptosis, was increased by overexpression of ORMDL3 levels, likely mediated by its downstream factor ATF6. Mechanistically, ORMDL3 transcriptionally activated ATF6, which was confirmed in a promoter reporter assay. Together, our data suggest that ORMDL3 may increase beta cell proliferation through ATF6 as an early compensatory change in response to diabetes.


Subject(s)
Activating Transcription Factor 6/metabolism , Diabetes Mellitus, Type 1/metabolism , Insulin-Secreting Cells/metabolism , Membrane Proteins/metabolism , Activating Transcription Factor 6/genetics , Adolescent , Animals , Apoptosis Regulatory Proteins , Blood Glucose , Case-Control Studies , Cell Line, Tumor , Cell Proliferation/physiology , Diabetes Mellitus/metabolism , Female , Glycated Hemoglobin , Humans , Insulin/metabolism , Insulinoma , Leukocytes/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred NOD , Mitochondrial Proteins
17.
J Alzheimers Dis ; 68(3): 1051-1059, 2019.
Article in English | MEDLINE | ID: mdl-30909245

ABSTRACT

Aged people have a high chance to develop two prevalent diseases, diabetes and Alzheimer's disease (AD), which are characterized with hyperglycemia and neurodegeneration, respectively. Interestingly, recent evidence suggest that diabetes is a predisposing factor for AD. Nevertheless, the mechanisms underlying the association of diabetes with AD remain poorly defined. Here, we studied the effects of diabetes on AD in mice. The APP-PS1 mouse, an AD-prone strain, was administrated with streptozotocin (STZ) to destroy 75% beta cell mass to induce sustained hyperglycemia. We found that STZ-treated APP-PS1 mice exhibited poorer performance in the social recognition test, Morris water maze, and plus-maze discriminative avoidance task, compared to saline-treated normoglycemic APP-PS1 mice, likely resulting from increases in brain deposition of amyloid-ß peptide aggregates (Aß). Since formation of Aß is known to be induced by protein hyperphosphorylation mediated by calpain (CAPN)-induced cleavage of p35 into p25, we examined levels of these proteins in mouse brain. We detected not only increased p35-to-p25 conversion, but also enhanced CAPN1 activity via increased protein but not mRNA levels. The internal CAPN1 inhibitor, calpastatin (CAST), was downregulated in STZ-treated APP-PS1 mouse brain, as a basis for the increase in CAPN1. In vitro, a human neuronal cell line, HCN-2, increased CAPN1 activity and downregulated CAST levels when incubated for 8 days in high glucose level, resulting in increased cell apoptosis. Together, these data suggest that chronic hyperglycemia may promote AD development through downregulating CAST.


Subject(s)
Alzheimer Disease/etiology , Calcium-Binding Proteins/metabolism , Diabetes Mellitus, Experimental/complications , Animals , Brain/metabolism , Calpain/metabolism , Cell Line , Down-Regulation , Female , Humans , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism
18.
Methods Mol Biol ; 1950: 373-387, 2019.
Article in English | MEDLINE | ID: mdl-30783986

ABSTRACT

Genetic manipulation is a very powerful tool for studying diabetes, pancreatitis, and pancreatic cancer. Here we discuss the use of an adeno-associated virus (AAV) vector to modify gene expression, such as to introduce a green fluorescence protein (GFP) in wild-type mice, cre recombinase in loxP mice, or to inactivate a gene with shRNA. The use of viruses for genetic modification allows for time-specific genetic changes which have advantages over time-consuming and often complex cross-breeding strategies. Here we provide a detailed approach for this process from viral production and purification through pancreatic ductal infusion. Our protocol allows efficient delivery of AAV to mediate GFP or cre expression for cell lineage tracing in the mouse pancreas or for the delivery of transgenes under a specific promoter to these cells.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , Genetic Vectors/isolation & purification , Pancreas/metabolism , Gene Expression , Genes, Reporter , Genetic Vectors/administration & dosage , Humans , Injections , Pancreatic Ducts/metabolism , Transfection
19.
Aging (Albany NY) ; 11(2): 697-706, 2019 01 26.
Article in English | MEDLINE | ID: mdl-30684443

ABSTRACT

Diabetic patients are prone to developing Alzheimer's disease (AD), in which microglia play a critical role. However, the direct effect of high glucose (HG) on microglia and the role of extracellular-signal-regulated kinase 5 (ERK5) signaling in this interaction have not been examined before. Here, these questions were addressed in microglia cultured in HG versus normal glucose (NG) conditions. Initially, HG induced microglial differentiation into the M2a phenotype with concomitant ERK5 activation. However, longer exposure to HG further induced differentiation of microglia into the M2b-like phenotype, followed by the M1-like subtype, concomitant with a gradual loss of ERK5 activation. BIX021895, a specific inhibitor of ERK5 activation, prevented M2a- differentiation of microglia, but induced earlier M2b-like polarization followed by M1-like polarization. Transfection of microglia with a sustained activated form of MEK5 (MEK5DD) prolonged the duration of the M2a phenotype, and prevented later differentiation into the M2b/M1 subtype. Conditioned media from the M2a-polarized microglia reduced neuronal cell apoptosis in hypoxic condition, while media from M2b-like or M1-like microglia enhanced apoptosis. Together, our data suggest that chronic hyperglycemia may induce a gradual alteration of microglia polarization into an increasingly proinflammatory subtype, which could be suppressed by sustained activation of ERK5 signaling.


Subject(s)
Cell Differentiation/drug effects , Gene Expression Regulation/drug effects , Glucose/toxicity , Microglia/drug effects , Mitogen-Activated Protein Kinase 7/metabolism , Aniline Compounds/pharmacology , Cell Line , Humans , Indoles/pharmacology , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/genetics
20.
Int J Mol Sci ; 19(10)2018 Oct 12.
Article in English | MEDLINE | ID: mdl-30322036

ABSTRACT

Diabetes is a global epidemic and affects millions of individuals in the United States. Devising novel treatments for diabetes continues to be a great medical challenge. Postnatal beta cell growth or compensation is largely attributed to beta cell proliferation, which declines continuously with age. To boost beta cell proliferation to regenerate an adequate functional mass, there is a need to understand the signaling pathways that regulate beta cell proliferation for creating practical strategies to promote the process. Transforming growth factor ß (TGFß) belongs to a signaling superfamily that governs pancreatic development and the regeneration of beta cells after pancreatic diseases. TGFß exerts its functions by activation of downstream Smad proteins and through its crosstalk with other pathways. Accumulating data demonstrate that the TGFß receptor signaling pathway also participates in the control of beta cell proliferation. This review details the role of the TGFß receptor signaling pathway in beta cell proliferation physiologically and in the pathogenesis of diabetes.


Subject(s)
Insulin-Secreting Cells/cytology , Receptors, Transforming Growth Factor beta/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Adult , Animals , Cell Cycle , Cell Proliferation , Humans , Insulin-Secreting Cells/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...