Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Am J Transplant ; 24(2): 177-189, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37813189

ABSTRACT

Present-day islet culture methods provide short-term maintenance of cell viability and function, limiting access to islet transplantation. Attempts to lengthen culture intervals remain unsuccessful. A new method was developed to permit the long-term culture of islets. Human islets were embedded in polysaccharide 3D-hydrogel in cell culture inserts or gas-permeable chambers with serum-free CMRL 1066 supplemented media for up to 8 weeks. The long-term cultured islets maintained better morphology, cell mass, and viability at 4 weeks than islets in conventional suspension culture. In fact, islets cultured in the 3D-hydrogel retained ß cell mass and function on par with freshly isolated islets in vitro and, when transplanted into diabetic mice, restored glucose balance similar to fresh islets. Using gas-permeable chambers, the 3D-hydrogel culture method was scaled up over 10-fold and maintained islet viability and function, although the cell mass recovery rate was 50%. Additional optimization of scale-up methods continues. If successful, this technology could afford flexibility and expand access to islet transplantation, especially single-donor islet-after-kidney transplantation.


Subject(s)
Diabetes Mellitus, Experimental , Islets of Langerhans Transplantation , Islets of Langerhans , Humans , Mice , Animals , Cell Culture Techniques , Hydrogels , Insulin , Cell Survival
2.
Pharmaceuticals (Basel) ; 16(10)2023 Oct 17.
Article in English | MEDLINE | ID: mdl-37895951

ABSTRACT

Recently, a G-protein coupled receptor 44 (GPR44) was discovered to play a significant role in the process of inflammation-related diseases, including cancer and diabetes. However, the precise role of GPR44 has yet to be fully elucidated. Currently, there is a strong and urgent need for the development of GPR44 radiotracers as a non-invasive methodology to explore the exact mechanism of GPR44 on inflammation-related diseases and monitor the progress of therapy. TM-30089 is a potent GPR44 antagonist that exhibits a high specificity and selectivity for GPR44. Its structure contains a fluorine nuclide, which could potentially be replaced with 18F. In the present study, we successfully took a highly effective synthesis strategy that pretreated the unprotected carboxylic acid group of the precursor and developed a feasible one-step automatic radiosynthesis strategy for [18F]TM-30089 with a high radiochemical purity and a good radiochemical yield. We further evaluated this radiotracer using mice models implanted with 1.1 B4 cell lines (GPR44-enriched cell lines) and human islets (high GPR44 expression), respectively. The results revealed the persistent and specific uptake of [18F]TM-30089 in GPR44 region, indicating that [18F]TM-30089 is a promising candidate for targeting GPR44. Further evaluation is ongoing.

3.
J Immunol Methods ; 501: 113199, 2022 02.
Article in English | MEDLINE | ID: mdl-34871593

ABSTRACT

Graves' disease (GD) is an autoimmune disease caused by antibodies to the thyroid stimulating hormone receptor (TSHR). The FDA-cleared Thyretain™ TSI bioassay is a highly specific method to detect thyroid stimulating antibodies (TSAb/TSI) in the blood of patients with autoimmune thyroid disease (AITD), particularly GD. To simplify the workflow of this bioassay and to support a semi-quantitative result, we have generated a stable CHO-K1 cell line expressing both a chimeric TSH receptor (TSHR-Mc4) and a luciferase-based homogeneous cAMP biosensor (GS luciferase). Here, we describe a rapid, real-time, homogenous bioassay (Turbo™ TSI Bioassay) to directly assess the functional activity of TSI and produce results in International Units of IU/L. The Turbo™ TSI bioassay works by measuring changes in the intracellular cAMP level induced by a G-protein coupled receptor (G-PCR) signaling cascade which is triggered by the binding of TSI to the TSHR. Upon binding to cAMP, the GS luciferase reporter is activated through conformational changes and generates light that can be measured in intact cells with a luminometer. The LoD and LoQ of the assay were determined to be 0.016 IU/L and 0.03 IU/L, respectively and the preliminary assay cutoff was determined to be 0.024 IU/L by ROC analysis using the Thyretain™ TSI bioassay results as reference. The analytical performance of the Turbo™ TSI bioassay is comparable to the Thyretain™ TSI bioassay as evidenced by similar EC50 values for a TSHR stimulating monoclonal antibody (M22). The specificity of the Turbo™ TSI bioassay was demonstrated by showing no response to a high concentration of a human monoclonal TSHR blocking antibody (K1-70). The precision of the assay was excellent with an overall within-laboratory precision <15% CV. When testing 198 clinical samples, the positive and negative percent agreement between the Turbo™ TSI and the Thyretain™ TSI bioassays were 98.7% and 93.5%, respectively. While both bioassays yield equivalent analytical and clinical performances, the Turbo™ TSI bioassay is much simpler to perform. It does not require cell culture, sample dilution, washing or cell lysis steps, resulting in a dramatically reduced turnaround time from about 21 h to 60 min. In addition, the same cell line showed its capability of detecting thyroid blocking antibodies (TBAb/TBI) in a competitive format. The Turbo™ TSI bioassay is user-friendly and is a very promising advancement to aid the diagnosis of autoimmune thyroid disease (AITD).


Subject(s)
Biosensing Techniques , Cyclic AMP/metabolism , Graves Disease/diagnosis , Immunoassay , Immunoglobulins, Thyroid-Stimulating/blood , Animals , Biological Assay , Biomarkers/blood , CHO Cells , Cricetulus , Graves Disease/blood , Graves Disease/immunology , Humans , Luciferases/genetics , Luciferases/metabolism , Predictive Value of Tests , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism , Reproducibility of Results , Workflow
4.
Sci Rep ; 9(1): 5705, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30952975

ABSTRACT

[68Ga]DO3A-VS-Cys40-Exendin-4, a glucagon-like peptide 1 receptor agonist, was evaluated as a potential PET tracer for the quantitation of human islets transplanted to the liver. The short-lived PET radionuclide 68Ga, available on a regular basis from a 68Ge/68Ga generator, is an attractive choice. Human C-peptide was measured to evaluate human islet function post-transplantation and prior to microPET imaging. [68Ga]DO3A-VS-Cys40-Exendin-4 was radiosynthesized and evaluated for PET imaging of transplanted human islets in the liver of healthy NOD/SCID mice. The biodistribution of the tracer was evaluated to determine the uptake into various organs, and qPCR of liver samples was conducted to confirm engrafted islet numbers after PET imaging. Measurement of human C-peptide indicated that higher engrafted islet mass resulted in higher human C-peptide levels in post-transplantation. The microPET imaging yielded high resolution images of liver-engrafted islets and also showed significant retention in mouse livers at 8 weeks post-transplantation. Biodistribution studies in mice revealed that liver uptake of [68Ga]DO3A-VS-Cys40-Exendin-4 was approximately 6-fold higher in mice that received 1000 islet equivalent (IEQ) than in non-transplanted mice. qPCR analysis of insulin expression suggested that islet engraftment numbers were close to 1000 IEQ transplanted. In conclusion, human islets transplanted into the livers of mice exhibited significant uptake of [68Ga]DO3A-VS-Cys40-Exendin-4 compared to the livers of untreated mice; and imaging of the mice using PET showed the human islets clearly with high contrast against liver tissue, enabling accurate quantitation of islet mass. Further validation of [68Ga]DO3A-VS-Cys40-Exendin-4 as an islet imaging probe for future clinical application is ongoing.


Subject(s)
Exenatide/pharmacokinetics , Gallium Radioisotopes/pharmacokinetics , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Islets of Langerhans/diagnostic imaging , Liver/diagnostic imaging , Positron-Emission Tomography/methods , Vinyl Compounds/pharmacokinetics , Animals , Humans , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation , Mice , Mice, Inbred NOD , Mice, SCID , Tissue Distribution
5.
PLoS One ; 10(6): e0126924, 2015.
Article in English | MEDLINE | ID: mdl-26083576

ABSTRACT

Trefoil factor 3 (TFF3), also called intestinal trefoil factor or Itf, is a 59 amino acid peptide found as a homodimer predominantly along the gastrointestinal tract and in serum. TFF3 expression is elevated during gastrointestinal adenoma progression and has been shown to promote mucosal wound healing. Here we show that in contrast to other trefoil factor family members, TFF1 and TFF2, TFF3 is highly expressed in mouse duodenum, jejunum and ileum and that its expression is regulated by food intake. Overexpression of TFF3 using a recombinant adeno-associated virus (AAV) vector, or daily administration of recombinant TFF3 protein in vivo improved glucose tolerance in a diet-induced obesity mouse model. Body weight, fasting insulin, triglyceride, cholesterol and leptin levels were not affected by TFF3 treatment. Induction of mucinous metaplasia was observed in mice with AAV-mediated TFF3 overexpression, however, no such adverse histological effect was seen after the administration of recombinant TFF3 protein. Altogether these results suggest that the therapeutic potential of targeting TFF3 to treat T2D may be limited.


Subject(s)
Blood Glucose/metabolism , Eating/genetics , Genetic Vectors/adverse effects , Metaplasia/genetics , Mucins/genetics , Obesity/genetics , Animals , Cholesterol/blood , Dependovirus/genetics , Diet, High-Fat , Duodenum/metabolism , Duodenum/pathology , Gene Expression , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glucose Tolerance Test , Humans , Ileum/metabolism , Ileum/pathology , Insulin/blood , Jejunum/metabolism , Jejunum/pathology , Leptin/blood , Male , Metaplasia/etiology , Metaplasia/metabolism , Metaplasia/pathology , Mice , Mucins/administration & dosage , Mucins/metabolism , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Trefoil Factor-2 , Trefoil Factor-3 , Triglycerides/blood
6.
Mol Cell Endocrinol ; 369(1-2): 119-29, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23403053

ABSTRACT

FFA1 (GPR40) and GPR120 are G-protein-coupled receptors activated by long-chain fatty acids. FFA1 is expressed in pancreatic ß-cells, where it regulates glucose-dependent insulin secretion, and GPR120 has been implicated in mediating GLP-1 secretion. We show here that FFA1 co-localizes with GLP-1 in enteroendocrine cells and plays a critical role in glucose management by mediating GLP-1 secretion in vivo. Corn oil induces GLP-1 secretion in wild type mice and in GPR120-/- mice, but not in FFA1-/- mice. α-Linolenic acid, an endogenous ligand of FFA1, induces GLP-1 secretion in GLUTag cells and in primary fetal mouse intestinal cells. Synthetic partial FFA1 agonists do not stimulate GLP-1 secretion in mice, but partial and full agonists combined function cooperatively to enhance receptor activation and GLP-1 secretion both in vitro and in vivo. We conclude that allosterism at FFA1 can contribute to postprandial glucose management by stimulating insulin secretion via an extrapancreatic mechanism of action, and that GPR120 in GLP-1 secretion requires further investigation.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Receptors, G-Protein-Coupled/physiology , Animals , CHO Cells , Cells, Cultured , Cloning, Molecular , Cricetinae , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
7.
ACS Med Chem Lett ; 4(6): 551-5, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-24900707

ABSTRACT

GPR40 (FFAR1 or FFA1) is a target of high interest being pursued to treat type II diabetes due to its unique mechanism leading to little risk of hypoglycemia. We recently reported the discovery of AM-1638 (2), a potent full agonist of GPR40. In this report, we present the discovery of GPR40 full agonists containing conformationally constrained tricyclic spirocycles and their structure-activity relationships leading to more potent agonists such as AM-5262 (26) with improved rat PK profile and general selectivity profile. AM-5262 enhanced glucose stimulated insulin secretion (mouse and human islets) and improved glucose homeostasis in vivo (OGTT in HF/STZ mice) when compared to AM-1638.

8.
PLoS One ; 7(10): e46300, 2012.
Article in English | MEDLINE | ID: mdl-23056280

ABSTRACT

Type 2 diabetes is characterized by impaired glucose homeostasis due to defects in insulin secretion, insulin resistance and the incretin response. GPR40 (FFAR1 or FFA1) is a G-protein-coupled receptor (GPCR), primarily expressed in insulin-producing pancreatic ß-cells and incretin-producing enteroendocrine cells of the small intestine. Several GPR40 agonists, including AMG 837 and TAK-875, have been disclosed, but no GPR40 synthetic agonists have been reported that engage both the insulinogenic and incretinogenic axes. In this report we provide a molecular explanation and describe the discovery of a unique and potent class of GPR40 full agonists that engages the enteroinsular axis to promote dramatic improvement in glucose control in rodents. GPR40 full agonists AM-1638 and AM-6226 stimulate GLP-1 and GIP secretion from intestinal enteroendocrine cells and increase GSIS from pancreatic islets, leading to enhanced glucose control in the high fat fed, streptozotocin treated and NONcNZO10/LtJ mouse models of type 2 diabetes. The improvement in hyperglycemia by AM-1638 was reduced in the presence of the GLP-1 receptor antagonist Ex(9-39)NH(2).


Subject(s)
Blood Glucose/metabolism , Receptors, G-Protein-Coupled/agonists , Animals , CHO Cells , Cricetinae , Cricetulus , Insulin/metabolism , Insulin Secretion , Mice , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Second Messenger Systems
9.
J Clin Microbiol ; 50(2): 396-400, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22075584

ABSTRACT

The recent association of certain influenza A virus subtypes with clinically relevant phenotypes has led to the increasing importance of subtyping by clinical virology laboratories. To provide clinical laboratories with a definitive immunofluorescence assay for the subtyping of influenza A virus isolates, we generated a panel of monoclonal antibodies (MAbs) against the major circulating influenza A virus subtypes using multiple inactivated H1N1, H3N2, and 2009 H1N1 strains individually as immunogens. Eleven MAbs that target hemagglutinin (HA) of H1N1 and H3N2 subtypes were selected. These MAbs were combined into three subtype-specific reagents, one each for pan-H1 (seasonal and 2009 strains), H3, and 2009 H1, for the subtyping of influenza A virus-positive specimens by indirect immunofluorescence assay (IFA). Each subtype-specific reagent was tested on 21 prototype influenza A virus strains and confirmed to be specific for its intended subtype. In addition, the subtyping reagents did not cross-react with any of 40 other viruses. The clinical performance of the subtyping reagents was evaluated with 75 archived clinical samples collected between 2006 and 2009 using the D(3) Ultra DFA influenza A virus identification reagent (Diagnostic Hybrids, Inc., Athens, OH) and the influenza A virus subtyping reagents by IFA simultaneously. Sixty-four samples grew virus and were subtyped as follows: 30 as H3N2, 9 as seasonal H1N1, and 25 as 2009 H1N1. RT-PCR was used to confirm the influenza A virus subtyping of these samples, and there was 100% agreement with IFA. This subtyping IFA provides clinical laboratories with a cost-effective diagnostic tool for better management of influenza virus infection and surveillance of influenza virus activity.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Influenza A virus/classification , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/isolation & purification , Cross Reactions , Fluorescent Antibody Technique, Indirect/methods , Humans , Influenza A virus/isolation & purification , Influenza, Human/virology , Mice , Mice, Inbred BALB C , Sensitivity and Specificity
10.
J Clin Microbiol ; 48(8): 2677-82, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20519459

ABSTRACT

To develop an immunofluorescence assay for identification of the 2009 H1N1 influenza A virus, we generated a number of monoclonal antibodies (MAbs) by using inactivated H1N1 2009 virus (A/California/07/2009) as the immunogen. Two MAbs that target two different epitopes of the 2009 H1N1 hemagglutinin (HA) were selected to make the D(3) Ultra 2009 H1N1 Influenza A ID kit (2009 H1N1 ID kit; Diagnostic Hybrids, Inc., Athens, OH), which is intended for the identification of the 2009 H1N1 virus by indirect immunofluorescence assay (IFA). The kit does not detect any of 14 seasonal H1N1 or H3N2 prototype influenza virus strains and is also not reactive with seven other major respiratory viruses. Clinical respiratory specimens were evaluated using both the 2009 H1N1 ID kit and the CDC human influenza virus real-time reverse transcription-PCR swine flu panel (CDC rRT-PCR) and showed 100% agreement between the two assays. Four of these clinical specimens, however, were positive by the 2009 H1N1 ID kit but were identified as presumptively positive by the CDC rRT-PCR by virtue of showing threshold cycle (C(T)) values only with universal InfA and swInfA primers, not with swH1 primers. Sequence analysis of the HA genes of these four specimens revealed point mutations in both the primer and probe regions. In addition, unlike the CDC rRT-PCR, the 2009 H1N1 ID kit can differentiate the 2009 H1N1 virus from a swine-derived H1 influenza A virus (A/New Jersey/8/76). The 2009 H1N1 ID kit offers clinical laboratories an alternative to RT-PCR for the identification of the 2009 H1N1 influenza A virus.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza, Human/diagnosis , Influenza, Human/virology , Virology/methods , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/isolation & purification , DNA Primers/genetics , Hemagglutinins, Viral/immunology , Humans , Immunoassay/methods , Influenza A Virus, H1N1 Subtype/immunology , Mice , Mice, Inbred BALB C , Oligonucleotide Probes/genetics , Point Mutation , RNA, Viral/genetics , Reagent Kits, Diagnostic , Reverse Transcriptase Polymerase Chain Reaction/methods , Sensitivity and Specificity
11.
J Clin Microbiol ; 47(10): 3108-13, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19710278

ABSTRACT

Enteroviruses (EVs) are common seasonal viruses that are associated with a variety of diseases. High-quality monoclonal antibodies (MAbs) are needed to improve the accuracy of EV diagnosis in clinical laboratories. In the present study, the full-length VP1 genes of poliovirus 1 (Polio 1) and coxsackievirus B3 (Cox B3) were cloned, and the encoded proteins were expressed and used as antigens in an attempt to raise broad-spectrum MAbs to EVs. Two pan-EV MAbs were isolated: one raised against Polio 1 VP1 and the other against Cox B3 VP1. The binding sites of both pan-EV MAbs were mapped to an amino acid sequence within a conserved region in the N terminus of Polio 1 VP1 by peptide and competition enzyme-linked immunosorbent assay. Two additional MAbs, an EV70-specific MAb and an EV71/Cox A16-bispecific MAb, developed against EV70 and 71 VP1 proteins, were pooled with the two pan-EV MAbs (pan-EV MAb mix) and tested for their sensitivity and specificity in the staining of various virus-infected cells. The pan-EV MAb mix detected all 40 prototype EVs tested and showed no cross-reactivity to 18 different non-EV human viruses. Compared with two commercially available EV tests, the pan-EV MAb mix exhibited higher specificity than one test and broader spectrum reactivity than the other. Thus, our study demonstrates that full-length Polio 1 VP1 and Cox B3 VP1 can serve as effective antigens for developing a pan-EV MAb and that the pan-EV MAb mix can be used for the laboratory diagnosis of a wide range of EV infections.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Capsid Proteins/immunology , Enterovirus Infections/diagnosis , Viral Structural Proteins/immunology , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/isolation & purification , Capsid Proteins/genetics , Cloning, Molecular , Conserved Sequence/immunology , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Gene Expression , Humans , Immunohistochemistry , Microscopy, Fluorescence , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sensitivity and Specificity , Viral Structural Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...