Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Stem Cell Res Ther ; 14(1): 53, 2023 03 29.
Article in English | MEDLINE | ID: mdl-36978104

ABSTRACT

National Eye Institute recently issued a new Strategic Plan outlining priority research areas for the next 5 years. Starting cell source for deriving stem cell lines is as an area with gaps and opportunities for making progress in regenerative medicine, a key area of emphasis within the NEI Strategic Plan. There is a critical need to understand how starting cell source affects the cell therapy product and what specific manufacturing capabilities and quality control standards are required for autologous vs allogeneic stem cell sources. With the goal of addressing some of these questions, in discussion with the community-at-large, NEI hosted a Town Hall at the Association for Research in Vision and Ophthalmology annual meeting in May 2022. This session leveraged recent clinical advances in autologous and allogeneic RPE replacement strategies to develop guidance for upcoming cell therapies for photoreceptors, retinal ganglion cells, and other ocular cell types. Our focus on stem cell-based therapies for RPE underscores the relatively advanced stage of RPE cell therapies to patients with several ongoing clinical trials. Thus, this workshop encouraged lessons learned from the RPE field to help accelerate progress in developing stem cell-based therapies in other ocular tissues. This report provides a synthesis of the key points discussed at the Town Hall and highlights needs and opportunities in ocular regenerative medicine.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Retinal Diseases , Humans , Retinal Diseases/therapy , Retinal Diseases/metabolism , Stem Cell Transplantation , Cell- and Tissue-Based Therapy , Induced Pluripotent Stem Cells/metabolism , Retinal Pigment Epithelium/metabolism
2.
ChemMedChem ; 15(16): 1562-1570, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32613743

ABSTRACT

Loss of ß-cell mass and function can lead to insufficient insulin levels and ultimately to hyperglycemia and diabetes mellitus. The mainstream treatment approach involves regulation of insulin levels; however, approaches intended to increase ß-cell mass are less developed. Promoting ß-cell proliferation with low-molecular-weight inhibitors of dual-specificity tyrosine-regulated kinase 1A (DYRK1A) offers the potential to treat diabetes with oral therapies by restoring ß-cell mass, insulin content and glycemic control. GNF4877, a potent dual inhibitor of DYRK1A and glycogen synthase kinase 3ß (GSK3ß) was previously reported to induce primary human ß-cell proliferation in vitro and in vivo. Herein, we describe the lead optimization that lead to the identification of GNF4877 from an aminopyrazine hit identified in a phenotypic high-throughput screening campaign measuring ß-cell proliferation.


Subject(s)
Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Insulin-Secreting Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Mice , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Structure-Activity Relationship , Dyrk Kinases
3.
J Med Chem ; 63(6): 2958-2973, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32077280

ABSTRACT

Autoimmune deficiency and destruction in either ß-cell mass or function can cause insufficient insulin levels and, as a result, hyperglycemia and diabetes. Thus, promoting ß-cell proliferation could be one approach toward diabetes intervention. In this report we describe the discovery of a potent and selective DYRK1A inhibitor GNF2133, which was identified through optimization of a 6-azaindole screening hit. In vitro, GNF2133 is able to proliferate both rodent and human ß-cells. In vivo, GNF2133 demonstrated significant dose-dependent glucose disposal capacity and insulin secretion in response to glucose-potentiated arginine-induced insulin secretion (GPAIS) challenge in rat insulin promoter and diphtheria toxin A (RIP-DTA) mice. The work described here provides new avenues to disease altering therapeutic interventions in the treatment of type 1 diabetes (T1D).


Subject(s)
Aza Compounds/chemistry , Aza Compounds/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Indoles/chemistry , Indoles/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Aza Compounds/pharmacokinetics , Cell Proliferation/drug effects , Cells, Cultured , Diabetes Mellitus, Type 1/metabolism , Humans , Hypoglycemic Agents/pharmacokinetics , Indoles/pharmacokinetics , Insulin Secretion/drug effects , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Molecular Docking Simulation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Dyrk Kinases
4.
Nature ; 569(7756): 368-373, 2019 05.
Article in English | MEDLINE | ID: mdl-31068696

ABSTRACT

In vitro differentiation of human stem cells can produce pancreatic ß-cells; the loss of this insulin-secreting cell type underlies type 1 diabetes. Here, as a step towards understanding this differentiation process, we report the transcriptional profiling of more than 100,000 human cells undergoing in vitro ß-cell differentiation, and describe the cells that emerged. We resolve populations that correspond to ß-cells, α-like poly-hormonal cells, non-endocrine cells that resemble pancreatic exocrine cells and a previously unreported population that resembles enterochromaffin cells. We show that endocrine cells maintain their identity in culture in the absence of exogenous growth factors, and that changes in gene expression associated with in vivo ß-cell maturation are recapitulated in vitro. We implement a scalable re-aggregation technique to deplete non-endocrine cells and identify CD49a (also known as ITGA1) as a surface marker of the ß-cell population, which allows magnetic sorting to a purity of 80%. Finally, we use a high-resolution sequencing time course to characterize gene-expression dynamics during the induction of human pancreatic endocrine cells, from which we develop a lineage model of in vitro ß-cell differentiation. This study provides a perspective on human stem-cell differentiation, and will guide future endeavours that focus on the differentiation of pancreatic islet cells, and their applications in regenerative medicine.


Subject(s)
Cell Differentiation , Insulin-Secreting Cells/cytology , Stem Cells/cytology , Animals , Biomarkers/metabolism , Cell Lineage , Cell Separation , Humans , Insulin/metabolism , Insulin-Secreting Cells/classification , Insulin-Secreting Cells/metabolism , Integrin alpha1/metabolism , Male , Mice , RNA-Seq , Single-Cell Analysis , Stem Cells/metabolism
5.
Cell Metab ; 29(3): 638-652.e5, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30581122

ABSTRACT

Small-molecule inhibitors of dual-specificity tyrosine-regulated kinase 1A (DYRK1A) induce human beta cells to proliferate, generating a labeling index of 1.5%-3%. Here, we demonstrate that combined pharmacologic inhibition of DYRK1A and transforming growth factor beta superfamily (TGFßSF)/SMAD signaling generates remarkable further synergistic increases in human beta cell proliferation (average labeling index, 5%-8%, and as high as 15%-18%), and increases in both mouse and human beta cell numbers. This synergy reflects activation of cyclins and cdks by DYRK1A inhibition, accompanied by simultaneous reductions in key cell-cycle inhibitors (CDKN1C and CDKN1A). The latter results from interference with the basal Trithorax- and SMAD-mediated transactivation of CDKN1C and CDKN1A. Notably, combined DYRK1A and TGFß inhibition allows preservation of beta cell differentiated function. These beneficial effects extend from normal human beta cells and stem cell-derived human beta cells to those from people with type 2 diabetes, and occur both in vitro and in vivo.


Subject(s)
Diabetes Mellitus, Type 2 , Harmine/pharmacology , Insulin-Secreting Cells , Monoamine Oxidase Inhibitors/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors , Adolescent , Adult , Aged , Animals , Cell Line , Cell Proliferation , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Female , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myeloid-Lymphoid Leukemia Protein/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Smad Proteins/antagonists & inhibitors , Stem Cells , Young Adult , Dyrk Kinases
6.
J Diabetes Res ; 2016: 8294805, 2016.
Article in English | MEDLINE | ID: mdl-27738640

ABSTRACT

Type 2 diabetes mellitus is prevalent especially in Gulf countries and poses serious long-term risks to patients. A multifaceted treatment approach can include nutritional supplements with antioxidant properties such as 5-aminolevulinic acid (5-ALA) with sodium ferrous citrate (SFC). This prospective, randomized, single-blind, placebo-controlled, dose escalating pilot clinical trial assessed the safety of 5-ALA with SFC at doses up to 200 mg 5-ALA/229.42 mg SFC per day in patients living in Bahrain with type 2 diabetes mellitus that was uncontrolled despite the use of one or more antidiabetic drugs. Fifty-three patients (n = 53) from 3 sites at one center were enrolled by Dr. Feryal (Site #01), Dr. Hesham (Site #02), and Dr. Waleed (Site #03) (n = 35, 5-ALA-SFC; n = 18, placebo). There was no significant difference in incidence of adverse events reported, and the most frequent events reported were gastrointestinal in nature, consistent with the known safety profile of 5-ALA in patients with diabetes. No significant changes in laboratory values and no difference in hypoglycemia between patients receiving 5-ALA and placebo were noted. Overall, the current results support that use of 5-ALA-SFC up to 200 mg per day taken as 2 divided doses is safe in patients taking concomitant oral antidiabetic medications and may offer benefits in the diabetic population. This trial is registered with ClinicalTrials.gov NCT02481141.


Subject(s)
Aminolevulinic Acid/administration & dosage , Diabetes Mellitus, Type 2/drug therapy , Ferrous Compounds/administration & dosage , Sodium/administration & dosage , Abdominal Pain/chemically induced , Aminolevulinic Acid/adverse effects , Bahrain , Blood Glucose/metabolism , Citric Acid , Cough/chemically induced , Diabetes Mellitus, Type 2/metabolism , Diarrhea/chemically induced , Female , Ferrous Compounds/adverse effects , Glycated Hemoglobin/metabolism , Humans , Male , Middle Aged , Phosphates/administration & dosage , Phosphates/adverse effects , Pilot Projects , Single-Blind Method , Sodium/adverse effects
7.
Nat Commun ; 6: 8372, 2015 Oct 26.
Article in English | MEDLINE | ID: mdl-26496802

ABSTRACT

Insufficient pancreatic ß-cell mass or function results in diabetes mellitus. While significant progress has been made in regulating insulin secretion from ß-cells in diabetic patients, no pharmacological agents have been described that increase ß-cell replication in humans. Here we report aminopyrazine compounds that stimulate robust ß-cell proliferation in adult primary islets, most likely as a result of combined inhibition of DYRK1A and GSK3B. Aminopyrazine-treated human islets retain functionality in vitro and after transplantation into diabetic mice. Oral dosing of these compounds in diabetic mice induces ß-cell proliferation, increases ß-cell mass and insulin content, and improves glycaemic control. Biochemical, genetic and cell biology data point to Dyrk1a as the key molecular target. This study supports the feasibility of treating diabetes with an oral therapy to restore ß-cell mass, and highlights a tractable pathway for future drug discovery efforts.


Subject(s)
Cell Proliferation , Glycogen Synthase Kinase 3/genetics , Insulin-Secreting Cells/cytology , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Animals , Cell Division/drug effects , Cell Proliferation/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Down-Regulation/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/enzymology , Male , Mice , Mice, Transgenic , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Pyridazines/pharmacology , Dyrk Kinases
8.
Am J Emerg Med ; 31(6): 928-34, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23685056

ABSTRACT

OBJECTIVE: To evaluate the cost-effectiveness of recombinant human hyaluronidase-facilitated subcutaneous (rHFSC) fluid administration compared to intravenous (IV) fluid administration in children with mild to moderate dehydration in the emergency department (ED). METHODS: A decision analytic model was created based on the results of a controlled clinical trial that compared the administration of isotonic fluids via rHFSC or IV for rehydration. The costs were determined from the hospital's perspective. The effectiveness unit was successful rehydration in the ED without the need for hospitalization for continued hydration. Mean estimates were determined for both the cost and effectiveness of each treatment. The incremental differences in costs and effectiveness were determined between treatments. Sensitivity analysis testing was also conducted. RESULTS: The treatment success rate was 93% with rHFSC fluids and 76% for IV fluids. Across all ages, the mean cost of rHFSC fluids was $722, compared to $889 for IV fluids. The difference in effectiveness was due to the larger number of patients for whom IV access could not be established, necessitating a rescue route of administration to deliver parenteral fluids. The difference in the overall cost was primarily due to the shorter time in the ED for patients receiving rHFSC fluids versus those treated with IV fluids. The cost-effectiveness of rHFSC compared to IV was most apparent in younger patients (<3 years of age), where IV access was more difficult to obtain. CONCLUSION: Analysis of this clinical trial data revealed that rHFSC fluid administration demonstrated greater treatment effectiveness and cost-effectiveness than traditional IV fluid administration in the ED. The primary reasons for this were the ease of obtaining parenteral access via rHFSC in young patients (especially those under 3) where IV access is difficult, and a shorter ED stay with rHFSC fluid administration.


Subject(s)
Fluid Therapy/economics , Fluid Therapy/methods , Hyaluronoglucosaminidase/therapeutic use , Age Factors , Child, Preschool , Cost-Benefit Analysis , Hospital Costs , Humans , Hyaluronoglucosaminidase/administration & dosage , Hyaluronoglucosaminidase/economics , Infant , Infant, Newborn , Infusions, Intravenous , Infusions, Subcutaneous , Recombinant Proteins/therapeutic use
9.
J Am Chem Soc ; 135(5): 1669-72, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23330637

ABSTRACT

The identification of factors that promote ß cell proliferation could ultimately move type 1 diabetes treatment away from insulin injection therapy and toward a cure. We have performed high-throughput, cell-based screens using rodent ß cell lines to identify molecules that induce proliferation of ß cells. Herein we report the discovery and characterization of WS6, a novel small molecule that promotes ß cell proliferation in rodent and human primary islets. In the RIP-DTA mouse model of ß cell ablation, WS6 normalized blood glucose and induced concomitant increases in ß cell proliferation and ß cell number. Affinity pulldown and kinase profiling studies implicate Erb3 binding protein-1 and the IκB kinase pathway in the mechanism of action of WS6.


Subject(s)
High-Throughput Screening Assays , Islets of Langerhans/drug effects , Urea/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Islets of Langerhans/cytology , Mice , Molecular Structure , Molecular Weight , Structure-Activity Relationship , Urea/analogs & derivatives , Urea/chemistry
10.
Clin Ther ; 34(11): 2232-45, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23062548

ABSTRACT

BACKGROUND: Alternative treatment of dehydration is needed when intravenous (IV) or oral rehydration therapy fails. Subcutaneous (SC) hydration facilitated by recombinant human hyaluronidase offers an alternative treatment for dehydration. This clinical trial is the first to compare recombinant human hyaluronidase-facilitated SC (rHFSC) rehydration with standard IV rehydration for use in dehydrated children. OBJECTIVE: This Phase IV noninferiority trial evaluated whether rHFSC fluid administration can be given safely and effectively, with volumes similar to those delivered intravenously, to children who have mild to moderate dehydration. METHODS: The study included mild to moderately dehydrated children (Gorelick dehydration score) aged 1 month to 10 years. They were randomized to receive 20 mL/kg of isotonic fluids using rHFSC or IV therapy over 1 hour and then as needed until clinically rehydrated. The primary outcome was total volume of fluid administered (emergency department [ED] plus inpatient hospitalization). Secondary outcomes included mean volume infused in the ED alone, postinfusion dehydration scores and weight changes, line placement success and time, safety, and provider and parent/guardian questionnaire. RESULTS: 148 patients (mean age, 2.3 [1.91] years]; white, 53.4%; black, 31.8%) were enrolled in the intention-to-treat population (73 rHFSC; 75 IV). The primary outcome, mean total volume infused, was 365.0 (324.6) mL in the rHFSC group over 3.1 hours versus 455.8 (597.4) mL in the IV group over 6.6 hours (P = 0.51). The secondary outcome of mean volume infused in the ED alone was 334.3 (226.40) mL in the rHFSC group versus 299.6 (252.33) mL in the IV group (P = 0.03). Dehydration scores and weight changes postinfusion were similar. Successful line placement occurred in all 73 rHFSC-treated patients and 59 of 75 (78.7%) IV-treated patients (P < 0.0001). All IV failures occurred in patients aged <3 years; rHFSC rescue was successful in all patients in whom it was attempted. Both treatments were well tolerated. Clinicians rated fluid administration as easy to perform in 94.5% (69 of 73) of the rHFSC group versus 65.3% (49 of 75) of the IV group (P < 0.001). Parents/caregivers were satisfied or very satisfied with fluid administration in 94.5% (69 of 73) of rHFSC-treated patients and 73.3% (55 of 75) of IV-treated patients. CONCLUSIONS: In mild to moderately dehydrated children, rHFSC was inferior to IV hydration for the primary outcome measure. However, rHFSC was noninferior in the ED phase of hydration. Additional benefits of rHFSC included time and success of line placement, ease of use, and satisfaction. SC hydration facilitated with recombinant human hyaluronidase represents a reasonable addition to the treatment options for children who have mild to moderate dehydration, especially those with difficult IV access. ClinicalTrials.gov identifier: NCT00773175.


Subject(s)
Antigens, Neoplasm/administration & dosage , Dehydration/therapy , Emergency Service, Hospital , Fluid Therapy/methods , Histone Acetyltransferases/administration & dosage , Hyaluronoglucosaminidase/administration & dosage , Hypodermoclysis , Isotonic Solutions/administration & dosage , Antigens, Neoplasm/adverse effects , Body Weight , Child , Child, Preschool , Dehydration/diagnosis , Female , Fluid Therapy/adverse effects , Histone Acetyltransferases/adverse effects , Hospitalization , Humans , Hyaluronoglucosaminidase/adverse effects , Hypodermoclysis/adverse effects , Infant , Infusions, Intravenous , Isotonic Solutions/adverse effects , Male , Prospective Studies , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Severity of Illness Index , Surveys and Questionnaires , Time Factors , Treatment Outcome , United States
11.
J Infus Nurs ; 34(5): 300-7, 2011.
Article in English | MEDLINE | ID: mdl-21915003

ABSTRACT

INTRODUCTION: Recombinant human hyaluronidase facilitates subcutaneous (SC) fluid delivery, but little is known about how various access sets influence ease of administration, technical challenges (TCs), or adverse events. METHODS: This randomized, open-label, parallel-group trial was performed to assess the impact of catheter size (20- and 24-gauge short peripheral intravenous catheter, 27-gauge SC button), catheter material (Teflon, polyurethane), and securement method (transparent semipermeable membrane dressing [TSM], double chevron with cloth or plastic tape) on hyaluronidase-facilitated SC fluid delivery. Healthy volunteers (N = 100) were randomized to 1 of 9 access groups using a factorial design. To minimize variability, treatment was performed at a single center and standardized to 150 units of SC recombinant human hyaluronidase (HYLENEX, Baxter Healthcare Corporation) followed by 1000 mL of lactated Ringer's solution. RESULTS: The first attempt at needle insertion succeeded in 98% of subjects; the median time for first catheter placement was less than 1 minute. The median infusion time was 6.8 hours. Overall, the incidence of TCs observed (catheter kinking, dislodgment, or pullout or infusion pump alarm) was low and comparable across groups (16.7%-27.3%); however, catheter kinking, dislodgment, and pullout occurred only in groups using double- chevron securement. Infusion-site reactions (pain, 20%-75%; erythema, 17%-36%; swelling, 0%-33%) were the most common adverse events. Pain was less frequent in groups using the 27-gauge SC button (27%) or the 24-gauge catheter (20%-36%) than with the 20-gauge catheter (50%-75%). DISCUSSION: Hyaluronidase-facilitated SC fluid administration with recombinant human hyaluronidase was generally well tolerated and successfully implemented using a range of access sets. Technical challenges were not common but were further minimized with TSM securement. Infusion-site pain was mostly mild and least common with 24-gauge or smaller catheter/needles.


Subject(s)
Hyaluronoglucosaminidase/administration & dosage , Humans , Infusions, Subcutaneous , Recombinant Proteins/administration & dosage
12.
Diabetes ; 59(12): 3131-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20876711

ABSTRACT

OBJECTIVE: Inducing human ß-cell growth while enhancing function is a major goal in the treatment of diabetes. Parathyroid hormone-related protein (PTHrP) enhances rodent ß-cell growth and function through the parathyroid hormone-1 receptor (PTH1R). Based on this, we hypothesized that PTH1R is expressed in human ß-cells and that PTHrP has the potential to enhance human ß-cell proliferation and/or function. RESEARCH DESIGN AND METHODS: PTH1R expression, ß-cell proliferation, glucose-stimulated insulin secretion (GSIS), and expression of differentiation and cell-cycle genes were analyzed in human islets transduced with adenoviral PTHrP constructs or treated with PTHrP peptides. The effect of overexpression of late G1/S cell cycle molecules was also assessed on human ß-cell proliferation. RESULTS: We found that human ß-cells express PTH1R. More importantly, overexpression of PTHrP causes a significant approximately threefold increase in human ß-cell proliferation. Furthermore, the amino terminus PTHrP(1-36) peptide is sufficient to increase replication as well as expression of the late G1/S cell-cycle proteins cyclin E and cyclin-dependent kinase 2 (cdk2) in human islets. Notably, PTHrP(1-36) also enhances GSIS. Finally, overexpression of cyclin E alone, but not cdk2, augments human ß-cell proliferation, and when both molecules are expressed simultaneously there is a further marked synergistic increase in replication. CONCLUSIONS: PTHrP(1-36) peptide enhances human ß-cell proliferation as well as function, with associated upregulation of two specific cell-cycle activators that together can induce human ß-cell proliferation several fold. The future therapeutic potential of PTHrP(1-36) for the treatment of diabetes is especially relevant given the complementary therapeutic efficacy of PTHrP(1-36) in postmenopausal osteoporosis.


Subject(s)
Cyclin E/genetics , Cyclin-Dependent Kinase 2/genetics , Insulin-Secreting Cells/physiology , Parathyroid Hormone-Related Protein/physiology , Receptor, Parathyroid Hormone, Type 1/genetics , Adolescent , Adult , Aged , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Division , Child , Child, Preschool , Female , Gene Expression Regulation , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Male , Middle Aged , Osteoporosis, Postmenopausal/drug therapy , Parathyroid Hormone-Related Protein/genetics , Parathyroid Hormone-Related Protein/pharmacology , Parathyroid Hormone-Related Protein/therapeutic use , Peptide Fragments/pharmacology
13.
Curr Med Res Opin ; 26(2): 279-88, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19947907

ABSTRACT

OBJECTIVE: To compare pharmacokinetics and safety of recombinant human hyaluronidase (rHuPH20)-facilitated subcutaneous (SC) ceftriaxone administration versus SC ceftriaxone preceded by SC saline placebo or intravenous (IV) ceftriaxone administration. RESEARCH DESIGN AND METHODS: This Phase I, two-part, placebo-controlled, crossover study was conducted in 54 healthy volunteers. In Part 1 (N = 24), subjects received 1 mL rHuPH20 (150 USP units) or placebo (0.9% sodium chloride) SC, followed by 1 or 2 g ceftriaxone (10-350 mg/mL). In Part 2 (N = 30), subjects received 1 g ceftriaxone at the Part 1 maximum tolerated concentration (MTC) administered either SC - preceded by SC rHuPH20 or placebo - or IV. Subjects were monitored for adverse events (AEs); blood samples were obtained (Part 2 only) during 48 hours post-dosing for ceftriaxone bioanalysis. MAIN OUTCOME MEASURES: Part 1 primary endpoint was the SC ceftriaxone (with or without rHuPH20) MTC. Pharmacokinetic parameters were determined in Part 2. Bioequivalence was based on maximum concentration (C(max)) and area under plasma concentration-time curve (AUC). RESULTS: The highest SC ceftriaxone concentration tested in Part 1 (350 mg/mL) was selected as the Part 2 MTC. In Part 2, median time to maximum concentration (t(max)) was 1 hour earlier (P < 0.0001), and C(max) was 12% higher (P < 0.0001) for ceftriaxone (350 mg/mL) administered via rHuPH20-facilitated SC versus SC preceded by placebo. IV ceftriaxone led to higher C(max) and shorter t(max) values than either SC treatment. Ceftriaxone exposure (AUC) was comparable among all three treatments. At least 1 AE was experienced by 100% of subjects after SC ceftriaxone and 76% after IV; most commonly reported AEs were infusion-site reactions. CONCLUSIONS: Ceftriaxone AUC did not differ significantly between the three administration routes. C(max) was higher and t(max) shorter with rHuPH20-facilitated SC than SC preceded by placebo. rHuPH20-facilitated SC ceftriaxone was generally well tolerated. This study is limited by evaluation of healthy adults and absence of repeated-dose groups.


Subject(s)
Antigens, Neoplasm/administration & dosage , Ceftriaxone/administration & dosage , Ceftriaxone/pharmacokinetics , Histone Acetyltransferases/administration & dosage , Histone Acetyltransferases/pharmacokinetics , Hyaluronoglucosaminidase/administration & dosage , Hyaluronoglucosaminidase/pharmacokinetics , Adolescent , Adult , Aged , Antigens, Neoplasm/adverse effects , Ceftriaxone/adverse effects , Cross-Over Studies , Double-Blind Method , Histone Acetyltransferases/adverse effects , Humans , Hyaluronoglucosaminidase/adverse effects , Injections, Intravenous , Injections, Subcutaneous , Middle Aged , Placebos , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacokinetics , Single-Blind Method , Young Adult
14.
Pediatrics ; 124(5): e858-67, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19805455

ABSTRACT

OBJECTIVES: The Increased Flow Utilizing Subcutaneously-Enabled (INFUSE)-Pediatric Rehydration Study was designed to assess efficacy, safety, and clinical utility of recombinant human hyaluronidase (rHuPH20)-facilitated subcutaneous rehydration in children 2 months to 10 years of age. METHODS: Patients with mild/moderate dehydration requiring parenteral treatment in US emergency departments were eligible for this phase IV, multicenter, single-arm study. They received subcutaneous injection of 1 mL rHuPH20 (150 U), followed by subcutaneous infusion of 20 mL/kg isotonic fluid over the first hour. Subcutaneous rehydration was continued as needed for up to 72 hours. Rehydration was deemed successful if it was attributed by the investigator primarily to subcutaneous fluid infusion and the child was discharged without requiring an alternative method of rehydration. RESULTS: Efficacy was evaluated in 51 patients (mean age: 1.9 years; mean weight: 11.2 kg). Initial subcutaneous catheter placement was achieved with 1 attempt for 46/51 (90.2%) of patients. Rehydration was successful for 43/51 (84.3%) of patients. Five patients (9.8%) were hospitalized but deemed to be rehydrated primarily through subcutaneous therapy, for a total of 48/51 (94.1%) of patients. No treatment-related systemic adverse events were reported, but 1 serious adverse event occurred (cellulitis at infusion site). Investigators found the procedure easy to perform for 96% of patients (49/51 patients), and 90% of parents (43/48 parents) were satisfied or very satisfied. CONCLUSIONS: rHuPH20-facilitated subcutaneous hydration seems to be safe and effective for young children with mild/moderate dehydration. Subcutaneous access is achieved easily, and the procedure is well accepted by clinicians and parents.


Subject(s)
Dehydration/therapy , Hyaluronoglucosaminidase/administration & dosage , Recombinant Proteins/administration & dosage , Rehydration Solutions/administration & dosage , Child , Child, Preschool , Female , Humans , Hyaluronoglucosaminidase/adverse effects , Infant , Infusion Pumps , Infusions, Subcutaneous/adverse effects , Injections, Subcutaneous , Male , Recombinant Proteins/adverse effects
15.
Diabetes ; 58(8): 1852-62, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19509021

ABSTRACT

OBJECTIVE: The retinoblastoma protein family (pRb, p130, p107) plays a central role in the regulation of cell cycle progression. Surprisingly, loss of pRb in the beta-cell has no discernible effect on cell cycle control. Therefore, we explored the effects of individual loss of either p130 or p107 in addition to the simultaneous loss of both pRb/p130 on the beta-cell. RESEARCH DESIGN AND METHODS: Adult mice deficient in either p130 or p107 or both pRb/p130 were examined for effects on beta-cell replication, function, and survival. The Cre-Lox system was also used to inactivate pRb in wild-type and p130-deficient beta-cells in vitro. RESULTS: In vivo loss of either p107 or p130 did not affect beta-cell replication or function. Combined pRb/p130 loss, however, resulted in dramatically accelerated proliferation as well as apoptotic cell death. Pancreas and beta-cell mass were significantly reduced in double mutants. Despite this, overall glucose tolerance was normal, except for mild postprandial hyperglycemia. Ex vivo, acute deletion of pRb in p130-deficient beta-cells also caused a striking increase in proliferation. The combined deletion of pRb/p130 upregulated islet expression of E2F2 but not E2F1. CONCLUSIONS: These studies define an essential role for the pocket proteins in controlling the G(1)/S transition in beta-cells. When deficient in both pRb and p130, beta-cells undergo unrestrained cell cycle reentry and activation of apoptosis. These studies underscore the central role of the pRb pathway in controlling beta-cell turnover and provide new cellular targets for beta-cell regeneration.


Subject(s)
Cell Cycle/physiology , G1 Phase/physiology , Insulin-Secreting Cells/cytology , Retinoblastoma Protein/physiology , Retinoblastoma-Like Protein p107/physiology , Retinoblastoma-Like Protein p130/physiology , S Phase/physiology , Animals , Apoptosis/physiology , Blood Glucose/metabolism , Cell Division , Glucose Tolerance Test , Mice , Mice, Knockout , Polymerase Chain Reaction , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/genetics , Retinoblastoma-Like Protein p107/deficiency , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p130/deficiency , Retinoblastoma-Like Protein p130/genetics
16.
J Autoimmun ; 32(2): 116-24, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19217258

ABSTRACT

Allogeneic islet transplantation is an option to treat diabetes however there are obstacles that are limiting its clinical use. We have examined whether mesenchymal stem cells (MSC) improve islet graft survival and whether such therapy allows for better graft acceptance with reduced requirement for immunosuppression. In vitro-expanded syngeneic bone marrow-derived MSC were co-transplanted with islets into omental pouch in a rat model of streptozotocin-induced diabetes. Marginal mass syngeneic islet transplantation into the omentum with MSC promoted sustained normoglycemia. Interestingly, allogeneic islets +MSC, but not islets alone, with short-term use of immunosuppression enhanced long-term islet graft survival, insulin expression in the grafts and induced normal serum insulin levels and normoglycemia. T cells from recipients transplanted with allogeneic islets +MSC produced low levels of IFN-gamma and TNF-alpha upon ex-vivo activation, and this transplantation protocol promoted the generation of IL-10-secreting CD4(+) T cells. These data encourage further preclinical and eventually, clinical MSC-based islet transplantation to improve the outcome of allogeneic islet transplantation in the treatment of diabetes.


Subject(s)
Glucose Metabolism Disorders/immunology , Graft Survival/immunology , Islets of Langerhans Transplantation/immunology , Mesenchymal Stem Cell Transplantation , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Glucose Metabolism Disorders/blood , Insulin/blood , Interleukin-10/metabolism , Male , Rats , Time Factors , Transplantation, Homologous/immunology
17.
Diabetes ; 58(4): 882-93, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19136653

ABSTRACT

OBJECTIVES: To comprehensively inventory the proteins that control the G1/S cell cycle checkpoint in the human islet and compare them with those in the murine islet, to determine whether these might therapeutically enhance human beta-cell replication, to determine whether human beta-cell replication can be demonstrated in an in vivo model, and to enhance human beta-cell function in vivo. RESEARCH DESIGN AND METHODS: Thirty-four G1/S regulatory proteins were examined in human islets. Effects of adenoviruses expressing cdk-6, cdk-4, and cyclin D1 on proliferation in human beta-cells were studied in both in vitro and in vivo models. RESULTS: Multiple differences between murine and human islets occur, most strikingly the presence of cdk-6 in human beta-cells versus its low abundance in the murine islet. Cdk-6 and cyclin D1 in vitro led to marked activation of retinoblastoma protein phosphorylation and cell cycle progression with no induction of cell death. Human islets transduced with cdk-6 and cyclin D1 were transplanted into diabetic NOD-SCID mice and markedly outperformed native human islets in vivo, maintaining glucose control for the entire 6 weeks of the study. CONCLUSIONS: The human G1/S proteome is described for the first time. Human islets are unlike their rodent counterparts in that they contain easily measurable cdk-6. Cdk-6 overexpression, alone or in combination with cyclin D1, strikingly stimulates human beta-cell replication, both in vitro as well as in vivo, without inducing cell death or loss of function. Using this model, human beta-cell replication can be induced and studied in vivo.


Subject(s)
Cyclin D1/physiology , Cyclin-Dependent Kinase 6/physiology , G1 Phase/genetics , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/physiology , Proteome , S Phase/genetics , Animals , Cell Cycle , Cell Division , Cyclin D1/genetics , Cyclin-Dependent Kinase 6/genetics , DNA Primers , Gene Expression Regulation , Glucose/pharmacology , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/transplantation , Kinetics , Mice , Mice, Inbred NOD , Mice, SCID , Phosphorylation , Retinoblastoma Protein/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity
18.
Diabetes ; 57(11): 3056-68, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18650366

ABSTRACT

OBJECTIVE: Rodent insulinoma cell lines may serve as a model for designing continuously replicating human beta-cell lines and provide clues as to the central cell cycle regulatory molecules in the beta-cell. RESEARCH DESIGN AND METHODS: We performed a comprehensive G1/S proteome analysis on the four most widely studied rodent insulinoma cell lines and defined their flow cytometric profiles and growth characteristics. RESULTS: 1) Despite their common T-antigen-derived origins, MIN6 and BTC3 cells display markedly different G1/S expression profiles; 2) despite their common radiation origins, RINm5F and INS1 cells display striking differences in cell cycle protein profiles; 3) phosphorylation of pRb is absent in INS1 and RINm5F cells; 4) cyclin D2 is absent in RINm5F and BTC3 cells and therefore apparently dispensable for their proliferation; 5) every cell cycle inhibitor is upregulated, presumably in a futile attempt to halt proliferation; 6) among the G1/S proteome members, seven are pro-proliferation molecules: cyclin-dependent kinase-1, -2, -4, and -6 and cyclins A, E, and D3; and 7) overexpression of the combination of these seven converts arrested proliferation rates in primary rat beta-cells to those in insulinoma cells. Unfortunately, this therapeutic overexpression appears to mildly attenuate beta-cell differentiation and function. CONCLUSIONS: These studies underscore the importance of characterizing the cell cycle at the protein level in rodent insulinoma cell lines. They also emphasize the hazards of interpreting data from rodent insulinoma cell lines as modeling normal cell cycle progression. Most importantly, they provide seven candidate targets for inducing proliferation in human beta-cells.


Subject(s)
Cell Cycle/physiology , Cell Proliferation , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Animals , Blotting, Western , Cell Cycle/genetics , Cell Line, Tumor , Cyclin A/genetics , Cyclin A/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Flow Cytometry , G1 Phase , Gene Expression Regulation, Neoplastic , Humans , Insulin-Secreting Cells/cytology , Insulinoma/genetics , Insulinoma/metabolism , Insulinoma/pathology , Islets of Langerhans/cytology , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , S Phase , Transfection
19.
Mol Ther ; 16(6): 1138-45, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18414479

ABSTRACT

Leptin injections evoke weight loss by causing a reduction in food consumption and an increase in energy expenditure. Also, the administration of leptin lowers blood glucose levels in some rodent models of diabetes and in humans with lipodystrophy. We explored the therapeutic potential of delivering leptin to obese, diabetic ob/ob mice and to mice fed on a high-fat diet (HFD), by transplanting gut-derived cells engineered to produce leptin, under the regulation of an inducing agent, mifepristone. These cells expressed and released leptin in a mifepristone dose-dependent and time-dependent manner. The engineered cells were either transplanted into the mice under the kidney capsule or were encapsulated in alginate and injected into the intraperitoneal cavity, while mifepristone was delivered by implanting 14-day release pellets. In ob/ob mice, leptin delivery by this method caused a significant reduction in food intake and profound weight loss, which was controllable by adjusting the dose of mifepristone. These transplants also achieved rapid and persistent amelioration of diabetes. However, mice fed on a HFD were resistant to the leptin therapy. These results indicate that gut cells can be modified to express leptin in an inducible manner and that the transplantation of these cells has a therapeutic effect in leptin-deficient mice, but not in mice fed on a HFD.


Subject(s)
Adipose Tissue/metabolism , Cell Transplantation/methods , Diabetes Mellitus, Experimental/therapy , Leptin/metabolism , Obesity/therapy , Animals , Body Weight , Dose-Response Relationship, Drug , Insulin/metabolism , Kidney/metabolism , Mice , Mice, Obese , Mifepristone/pharmacology , RNA, Messenger/metabolism , Transfection
20.
Endocrinology ; 149(7): 3679-87, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18372336

ABSTRACT

Closure of pancreatic beta-cell ATP-sensitive potassium (K(ATP)) channels links glucose metabolism to electrical activity and insulin secretion. It is now known that saturated, but not polyunsaturated, long-chain acyl-coenyzme A esters (acyl-CoAs) can potently activate K(ATP) channels when superfused directly across excised membrane patches, suggesting a plausible mechanism to account for reduced beta-cell excitability and insulin secretion observed in obesity and type 2 diabetes. However, reduced beta-cell excitability due to elevation of endogenous saturated acyl-CoAs has not been confirmed in intact pancreatic beta-cells. To test this notion directly, endogenous acyl-CoA levels were elevated within primary mouse beta-cells using virally delivered overexpression of long-chain acyl-CoA synthetase-1 (AdACSL-1), and the effects on beta-cell K(ATP) channel activity and cell excitability was assessed using the perforated whole-cell and cell-attached patch-clamp technique. Data indicated a significant increase in K(ATP) channel activity in AdACSL-1-infected beta-cells cultured in medium supplemented with palmitate/oleate but not with the polyunsaturated fat linoleate. No changes in the ATP/ADP ratio were observed in any of the groups. Furthermore, AdACSL-1-infected beta-cells (with palmitate/oleate) showed a significant decrease in electrical responsiveness to glucose and tolbutamide and a hyperpolarized resting membrane potential at 5 mm glucose. These results suggest a direct link between intracellular fatty ester accumulation and K(ATP) channel activation, which may contribute to beta-cell dysfunction in type 2 diabetes.


Subject(s)
Acyl Coenzyme A/physiology , Insulin-Secreting Cells/physiology , KATP Channels/physiology , Acyl Coenzyme A/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Blotting, Western , Cells, Cultured , Chromatography, High Pressure Liquid , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Membrane Potentials/drug effects , Mice , Mice, Inbred BALB C , Patch-Clamp Techniques , Tolbutamide/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...