Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
JCI Insight ; 1(3)2016.
Article in English | MEDLINE | ID: mdl-27152363

ABSTRACT

The capacity of pancreatic ß cells to maintain glucose homeostasis during chronic physiologic and immunologic stress is important for cellular and metabolic homeostasis. Insulin receptor substrate 2 (IRS2) is a regulated adapter protein that links the insulin and IGF1 receptors to downstream signaling cascades. Since strategies to maintain or increase IRS2 expression can promote ß cell growth, function, and survival, we conducted a screen to find small molecules that can increase IRS2 mRNA in isolated human pancreatic islets. We identified 77 compounds, including 15 that contained a tricyclic core. To establish the efficacy of our approach, one of the tricyclic compounds, trimeprazine tartrate, was investigated in isolated human islets and in mouse models. Trimeprazine is a first-generation antihistamine that acts as a partial agonist against the histamine H1 receptor (H1R) and other GPCRs, some of which are expressed on human islets. Trimeprazine promoted CREB phosphorylation and increased the concentration of IRS2 in islets. IRS2 was required for trimeprazine to increase nuclear Pdx1, islet mass, ß cell replication and function, and glucose tolerance in mice. Moreover, trimeprazine synergized with anti-CD3 Abs to reduce the progression of diabetes in NOD mice. Finally, it increased the function of human islet transplants in streptozotocin-induced (STZ-induced) diabetic mice. Thus, trimeprazine, its analogs, or possibly other compounds that increase IRS2 in islets and ß cells without adverse systemic effects might provide mechanism-based strategies to prevent the progression of diabetes.

2.
Best Pract Res Clin Endocrinol Metab ; 29(6): 821-31, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26696512

ABSTRACT

A key therapeutic approach for the treatment of Type 1 diabetes (T1D) is transplantation of functional islet ß-cells. Despite recent advances in generating stem cell-derived glucose-responsive insulin(+) cells, their further maturation to fully functional adult ß-cells still remains a daunting task. Conquering this hurdle will require a better understanding of the mechanisms driving maturation of embryonic insulin(+) cells into adult ß-cells, and the implementation of that knowledge to improve current differentiation protocols. Here, we will review our current understanding of ß-cell maturation, and discuss the contribution of key ß-cell transcription factor MafA, to this process. The fundamental importance of MafA in regulating adult ß-cell maturation and function indicates that enhancing MafA expression may improve the generation of definitive ß-cells for transplantation. Additionally, we suggest that the temporal control of MafA induction at a specific stage of ß-cell differentiation will be the next critical challenge for achieving optimum maturation of ß-cells.


Subject(s)
Cell Differentiation , Insulin-Secreting Cells/metabolism , Maf Transcription Factors, Large/metabolism , Animals , Humans , Insulin-Secreting Cells/cytology , Maf Transcription Factors, Large/genetics
3.
PLoS One ; 10(11): e0142286, 2015.
Article in English | MEDLINE | ID: mdl-26540252

ABSTRACT

Early in pancreatic development, epithelial cells of pancreatic buds function as primary multipotent progenitor cells (1°MPC) that specify all three pancreatic cell lineages, i.e., endocrine, acinar and duct. Bipotent "Trunk" progenitors derived from 1°MPC are implicated in directly regulating the specification of endocrine progenitors. It is unclear if this specification process is initiated in the 1°MPC where some 1°MPC become competent for later specification of endocrine progenitors. Previously we reported that in Pdx1tTA/+;tetOMafA (bigenic) mice inducing expression of transcription factor MafA in Pdx1-expressing (Pdx1+) cells throughout embryonic development inhibited the proliferation and differentiation of 1°MPC cells, resulting in reduced pancreatic mass and endocrine cells by embryonic day (E) 17.5. Induction of the transgene only until E12.5 in Pdx1+ 1°MPC was sufficient for this inhibition of endocrine cells and pancreatic mass at E17.5. However, by birth (P0), as we now report, such bigenic pups had significantly increased pancreatic and endocrine volumes with endocrine clusters containing all pancreatic endocrine cell types. The increase in endocrine cells resulted from a higher proliferation of tubular epithelial cells expressing the progenitor marker Glut2 in E17.5 bigenic embryos and increased number of Neurog3-expressing cells at E19.5. A BrdU-labeling study demonstrated that inhibiting proliferation of 1°MPC by forced MafA-expression did not lead to retention of those progenitors in E17.5 tubular epithelium. Our data suggest that the forced MafA expression in the 1°MPC inhibits their competency to specify endocrine progenitors only until E17.5, and after that compensatory proliferation of tubular epithelium gives rise to a distinct pool of endocrine progenitors. Thus, these bigenic mice provide a novel way to characterize the competency of 1°MPC for their ability to specify endocrine progenitors, a critical limitation in our understanding of endocrine differentiation.


Subject(s)
Epithelium/physiology , Pancreas/physiology , Stem Cells/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Cell Lineage/physiology , Endocrine Cells/metabolism , Endocrine Cells/physiology , Endocrine System/metabolism , Endocrine System/physiology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Epithelium/metabolism , Female , Gene Expression Regulation, Developmental/physiology , Glucose Transporter Type 2/metabolism , Maf Transcription Factors, Large/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Pancreas/metabolism , Pregnancy , Stem Cells/metabolism
4.
Dev Biol ; 385(1): 2-12, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24183936

ABSTRACT

Specification and maturation of insulin(+) cells accompanies a transition in expression of Maf family of transcription factors. In development, MafA is expressed after specification of insulin(+) cells that are expressing another Maf factor, MafB; after birth, these insulin(+) MafA(+) cells stop MafB expression and gain glucose responsiveness. Current differentiation protocols for deriving insulin-producing ß-cells from stem cells result in ß-cells lacking both MafA expression and glucose-stimulated insulin secretion. So driving expression of MafA, a ß-cell maturation factor in endocrine precursors could potentially generate glucose-responsive MafA(+) ß cells. Using inducible transgenic mice, we characterized the final stages of ß-cell differentiation and maturation with MafA pause/release experiments. We found that forcing MafA transgene expression, out of its normal developmental context, in Ngn3(+) endocrine progenitors blocked endocrine differentiation and prevented the formation of hormone(+) cells. However, this arrest was reversible such that with stopping the transgene expression, the cells resumed their differentiation to hormone(+) cells, including α-cells, indicating that the block likely occurred after progenitors had committed to a specific hormonal fate. Interestingly, this delayed resumption of endocrine differentiation resulted in a greater proportion of immature insulin(+)MafB(+) cells at P5, demonstrating that during maturation the inhibition of MafB in ß-cell transitioning from insulin(+)MafB(+) to insulin(+)MafB(-) stage is regulated by cell-autonomous mechanisms. These results demonstrate the importance of proper context of initiating MafA expression on the endocrine differentiation and suggest that generating mature Insulin(+)MafA(+) ß-cells will require the induction of MafA in a narrow temporal window to achieve normal endocrine differentiation.


Subject(s)
Endocrine Cells/metabolism , Maf Transcription Factors, Large/metabolism , MafB Transcription Factor/metabolism , Stem Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Endocrine Cells/cytology , Gene Expression Regulation, Developmental , Insulin/biosynthesis , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Maf Transcription Factors, Large/biosynthesis , Maf Transcription Factors, Large/genetics , MafB Transcription Factor/biosynthesis , MafB Transcription Factor/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Pancreas/cytology , Pancreas/metabolism
5.
Mol Endocrinol ; 27(7): 1078-90, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23660596

ABSTRACT

The reduction in the expression of glucose-responsive insulin gene transcription factor MafA accompanies the development of ß-cell dysfunction under oxidative stress/diabetic milieu. Humans with type 2 diabetes have reduced MafA expression, and thus preventing this reduction could overcome ß-cell dysfunction and diabetes. We previously showed that p38 MAPK, but not glycogen synthase kinase 3 (GSK3), is a major regulator of MafA degradation under oxidative stress. Here, we examined the mechanisms of this degradation and whether preventing MafA degradation under oxidative stress will overcome ß-cell dysfunction. We show that under oxidative and nonoxidative conditions p38 MAPK directly binds to MafA and triggers MafA degradation via ubiquitin proteasomal pathway. However, unlike nonoxidative conditions, MafA degradation under oxidative stress depended on p38 MAPK-mediated phosphorylation at threonine (T) 134, and not T57. Furthermore the expression of alanine (A) 134-MafA, but not A57-MafA, reduced the oxidative stress-mediated loss of glucose-stimulated insulin secretion, which was independent of p38 MAPK action on protein kinase D, a regulator of insulin secretion. Interestingly, the expression of proteasomal activator PA28γ that degrades GSK3-phosphorylated (including T57) MafA was reduced under oxidative stress, explaining the dominance of p38 MAPK over the GSK3 pathway in regulating MafA stability under oxidative stress. These results identify two distinct pathways mediating p38 MAPK-dependent MafA degradation under oxidative and nonoxidative conditions and show that inhibiting MafA degradation under oxidative stress ameliorates ß-cell dysfunction and could lead to novel therapies for diabetes.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Maf Transcription Factors, Large/metabolism , Oxidative Stress , Proteolysis , p38 Mitogen-Activated Protein Kinases/metabolism , Amino Acid Substitution , Animals , Autoantigens/metabolism , Enzyme Activation , Humans , Insulin/metabolism , Insulin Secretion , Mice , Models, Biological , Phosphorylation , Phosphothreonine/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Ubiquitin/metabolism
6.
Diabetes ; 62(5): 1569-80, 2013 May.
Article in English | MEDLINE | ID: mdl-23305647

ABSTRACT

Neonatal ß cells do not secrete glucose-responsive insulin and are considered immature. We previously showed the transcription factor MAFA is key for the functional maturation of ß cells, but the physiological regulators of this process are unknown. Here we show that postnatal rat ß cells express thyroid hormone (TH) receptor isoforms and deiodinases in an age-dependent pattern as glucose responsiveness develops. In vivo neonatal triiodothyronine supplementation and TH inhibition, respectively, accelerated and delayed metabolic development. In vitro exposure of immature islets to triiodothyronine enhanced the expression of Mafa, the secretion of glucose-responsive insulin, and the proportion of responsive cells, all of which are effects that were abolished in the presence of dominant-negative Mafa. Using chromatin immunoprecipitation and electrophoretic mobility shift assay, we show that TH has a direct receptor-ligand interaction with the Mafa promoter and, using a luciferase reporter, that this interaction was functional. Thus, TH can be considered a physiological regulator of functional maturation of ß cells via its induction of Mafa.


Subject(s)
Blood Glucose/analysis , Cell Differentiation , Insulin-Secreting Cells/cytology , Insulin/metabolism , Proto-Oncogene Proteins c-maf/metabolism , Triiodothyronine/metabolism , Animals , Animals, Newborn , Cell Nucleus/metabolism , Gene Expression Regulation, Developmental , Genes, Reporter , Insulin Secretion , Insulin-Secreting Cells/metabolism , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/growth & development , Islets of Langerhans/metabolism , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , Proto-Oncogene Proteins c-maf/genetics , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Recombinant Proteins/metabolism , Tissue Culture Techniques
7.
Diabetes Manag (Lond) ; 2(3): 213-222, 2012 May 01.
Article in English | MEDLINE | ID: mdl-23049620

ABSTRACT

Over the last decade, our knowledge of ß-cell biology has expanded with the use of new scientific techniques and strategies. Growth factors, hormones and small molecules have been shown to enhance ß-cell proliferation and function. Stem cell technology and research into the developmental biology of the pancreas have yielded new methods for in vivo and in vitro regeneration of ß cells from stem cells and endogenous progenitors as well as transdifferentiation of non-ß cells. Novel pharmacological approaches have been developed to preserve and enhance ß-cell function. Strategies to increase expression of insulin gene transcription factors in dysfunctional and immature ß cells have ameliorated these impairments. Hence, we suggest that strategies to minimize ß-cell loss and to increase their function and regeneration will ultimately lead to therapy for both Type 1 and 2 diabetes.

8.
Proc Natl Acad Sci U S A ; 106(34): 14460-5, 2009 Aug 25.
Article in English | MEDLINE | ID: mdl-19667185

ABSTRACT

Maturity-onset diabetes of the young (MODY) is a subtype of diabetes defined by an autosomal pattern of inheritance and a young age at onset, often before age 25. MODY is genetically heterogeneous, with 8 distinct MODY genes identified to date and more believed to exist. We resequenced 732 kb of genomic sequence at 8p23 in 6 MODY families unlinked to known MODY genes that showed evidence of linkage at that location. Of the 410 sequence differences that we identified, 5 had a frequency <1% in the general population and segregated with diabetes in 3 of the families, including the 2 showing the strongest support for linkage at this location. The 5 mutations were all placed within 100 kb corresponding to the BLK gene. One resulted in an Ala71Thr substitution; the other 4 were noncoding and determined decreased in vitro promoter activity in reporter gene experiments. We found that BLK--a nonreceptor tyrosine-kinase of the src family of proto-oncogenes--is expressed in beta-cells where it enhances insulin synthesis and secretion in response to glucose by up-regulating transcription factors Pdx1 and Nkx6.1. These actions are greatly attenuated by the Ala71Thr mutation. These findings point to BLK as a previously unrecognized modulator of beta-cell function, the deficit of which may lead to the development of diabetes.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Insulin-Secreting Cells/metabolism , Mutation , src-Family Kinases/genetics , Adolescent , Adult , Animals , Blotting, Western , Cell Line, Tumor , DNA Mutational Analysis , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Family Health , Female , Genetic Predisposition to Disease , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/pathology , Luciferases/genetics , Luciferases/metabolism , Male , Microscopy, Confocal , Middle Aged , Pedigree , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Young Adult , src-Family Kinases/metabolism
9.
Mol Endocrinol ; 23(8): 1281-90, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19407223

ABSTRACT

Mammalian MafA/RIPE3b1 is an important glucose-responsive transcription factor that regulates function, maturation, and survival of beta-cells. Increased expression of MafA results in improved glucose-stimulated insulin secretion and beta-cell function. Because MafA is a highly phosphorylated protein, we examined whether regulating activity of protein kinases can increase MafA expression by enhancing its stability. We demonstrate that MafA protein stability in MIN6 cells and isolated mouse islets is regulated by both p38 MAPK and glycogen synthase kinase 3. Inhibiting p38 MAPK enhanced MafA stability in cells grown under both low and high concentrations of glucose. We also show that the N-terminal domain of MafA plays a major role in p38 MAPK-mediated degradation; simultaneous mutation of both threonines 57 and 134 into alanines in MafA was sufficient to prevent this degradation. Under oxidative stress, a condition detrimental to beta-cell function, a decrease in MafA stability was associated with a concomitant increase in active p38 MAPK. Interestingly, inhibiting p38 MAPK but not glycogen synthase kinase 3 prevented oxidative stress-dependent degradation of MafA. These results suggest that the p38 MAPK pathway may represent a common mechanism for regulating MafA levels under oxidative stress and basal and stimulatory glucose concentrations. Therefore, preventing p38 MAPK-mediated degradation of MafA represents a novel approach to improve beta-cell function.


Subject(s)
Gene Expression Regulation, Enzymologic , Maf Transcription Factors, Large/metabolism , Oxidative Stress , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Glucose/metabolism , Glycogen Synthase Kinase 3/metabolism , Hyperglycemia/metabolism , Insulin-Secreting Cells/metabolism , Male , Mice , Models, Biological , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Am J Physiol Endocrinol Metab ; 291(4): E835-42, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16720626

ABSTRACT

During perinatal development, the regulation of IGF system appears to be growth hormone (GH) independent. By using highly purified primary fetal hepatocytes, we investigated the role of prolactin (PRL) in the regulation of IGF system and hepatocyte proliferation. We also analyzed the consequence of a maternal low-protein (LP) diet on the regulation of IGF, IGF-binding protein (IGFBP), and hepatocyte proliferation by prolactin. Pregnant Wistar rats were fed a control (C) diet (20% protein) or isocaloric (LP; 8%) diet throughout gestation. On day 21.5, fetal hepatocytes were cultured for 4 days and incubated with rat prolactin. In the C hepatocytes, PRL at 100 ng/ml decreased the abundance of IGFBP-1 and IGFBP-2 by 50 (P < 0.05) and 60% (P < 0.01), respectively. It also reduced by 70% the level of IGF-II mRNA (P < 0.01). By contrast, PRL failed to modulate IGFBP-1 and IGFBP-2 production by LP hepatocytes, and this was associated with reduced abundance of the short form of PRL receptor (P < 0.05). PRL had no effect on either the proliferation or the IGF-I production by C and LP hepatocytes, although it reduced the expression of IGF-II. These results suggest that prolactin influences hepatocyte proliferation in vitro by inhibiting IGFBP-1, IGFBP-2, and IGF-II levels, which may coincide with the decline of IGF-II observed in rodents during late gestation in vivo. On the other hand, maternal LP diet induces a resistance of fetal hepatocytes to PRL.


Subject(s)
Insulin-Like Growth Factor Binding Protein 1/metabolism , Insulin-Like Growth Factor Binding Protein 2/metabolism , Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor I/metabolism , Liver/metabolism , Malnutrition/metabolism , Prolactin/pharmacology , Animals , Blotting, Western , Cell Growth Processes/physiology , Culture Media, Conditioned/metabolism , Dietary Proteins/metabolism , Female , Fetal Development/physiology , Hepatocytes/cytology , Hepatocytes/metabolism , Insulin-Like Growth Factor Binding Protein 1/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 2/antagonists & inhibitors , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor II/antagonists & inhibitors , Insulin-Like Growth Factor II/genetics , Liver/cytology , Liver/embryology , Male , Pregnancy , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Random Allocation , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
11.
Dev Biol ; 293(2): 526-39, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16580660

ABSTRACT

Major insulin gene transcription factors, such as PDX-1 or NeuroD1, have equally important roles in pancreatic development and the differentiation of pancreatic endocrine cells. Previously, we identified and cloned another critical insulin gene transcription factor MafA (RIPE3b1) and reported that other Maf factors were expressed in pancreatic endocrine cells. Maf factors are important regulators of cellular differentiation; to understand their role in differentiation of pancreatic endocrine cells, we analyzed the expression pattern of large-Maf factors in the pancreas of embryonic and adult mice. Ectopically expressed large-Maf factors, MafA, MafB, or cMaf, induced expression from insulin and glucagon reporter constructs, demonstrating a redundancy in their function. Yet in adult pancreas, cMaf was expressed in both alpha- and beta-cells, and MafA and MafB showed selective expression in the beta- and alpha-cells, respectively. Interestingly, during embryonic development, a significant proportion of MafB-expressing cells also expressed insulin. In embryos, MafB is expressed before MafA, and our results suggest that the differentiation of beta-cells proceeds through a MafB+ MafA- Ins+ intermediate cell to MafB- MafA+ Ins+ cells. Furthermore, the MafB to MafA transition follows induction of PDX-1 expression (Pdx-1(high)) in MafB+ Ins+ cells. We suggest that MafB may have a dual role in regulating embryonic differentiation of both beta- and alpha-cells while MafA may regulate replication/survival and function of beta-cells after birth. Thus, this redundancy in the function and expression of the large-Maf factors may explain the normal islet morphology observed in the MafA knockout mice at birth.


Subject(s)
Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Maf Transcription Factors, Large/genetics , MafB Transcription Factor/genetics , Animals , Base Sequence , Cell Differentiation , Cell Proliferation , Female , Gene Expression Regulation, Developmental , Genes, Reporter , Glucagon/genetics , HeLa Cells , Humans , Insulin/biosynthesis , Insulin/genetics , Islets of Langerhans/embryology , Islets of Langerhans/growth & development , Mice , Mice, Inbred C57BL , Models, Biological , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection
12.
Am J Physiol Endocrinol Metab ; 285(5): E991-E1000, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12902319

ABSTRACT

We investigated the effect of an isocaloric maternal low-protein diet during pregnancy in rats on the proliferative capacity of cultured fetal hepatocytes. The potential roles of these changes on the IGF-IGF-binding protein (IGFBP) axis, and the role of insulin and glucocorticoids in liver growth retardation, were also evaluated. Pregnant Wistar rats were fed a control (C) diet (20% protein) or a low-protein (LP) diet (8%) throughout gestation. In primary culture, the DNA synthesis of hepatocytes derived from LP fetuses was decreased by approximately 30% compared with control hepatocytes (P < 0.05). In parallel, in vivo moderate protein restriction in the dam reduced the fetal liver weight and IGF-I level in fetal plasma (P < 0.01) and augmented the abundance of 29- to 32-kDa IGFBPs in fetal plasma (P < 0.01) and fetal liver (P < 0.01). By contrast, the abundance of IGF-II mRNA in liver of LP fetuses was unaffected by the LP diet. In vitro, the LP-derived hepatocytes produced less IGF-I (P < 0.01) and more 29- to 32-kDa IGFBPs (P < 0.01) than hepatocytes derived from control fetuses. These alterations still appeared after 3-4 days of culture, indicating some persistence in programming. Dexamethasone treatment of control-derived hepatocytes decreased cell proliferation (54 +/- 2.3%, P < 0.01) and stimulated 29- to 32-kDa IGFBPs, whereas insulin promoted fetal hepatocyte growth (127 +/- 5.5%, P < 0.01) and inhibited 29- to 32-kDa IGFBPs. These results show that liver growth and cell proliferation in association with IGF-I and IGFBP levels are affected in utero by fetal undernutrition. It also suggests that glucocorticoids and insulin may modulate these effects.


Subject(s)
Cell Division , Diet, Protein-Restricted , Fetus , Insulin-Like Growth Factor Binding Proteins/analysis , Insulin-Like Growth Factor I/analysis , Liver/embryology , Animals , Blotting, Western , Cells, Cultured , Dexamethasone/pharmacology , Female , Fetal Blood/chemistry , Glucocorticoids/pharmacology , Hepatocytes/cytology , Humans , Immunoenzyme Techniques , Insulin/pharmacology , Insulin-Like Growth Factor Binding Protein 1/analysis , Insulin-Like Growth Factor Binding Protein 1/pharmacology , Insulin-Like Growth Factor Binding Protein 2/analysis , Insulin-Like Growth Factor Binding Proteins/blood , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/genetics , Liver/chemistry , Liver/cytology , Maternal-Fetal Exchange , Organ Size , Pregnancy , RNA, Messenger/analysis , Rats , Rats, Wistar , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...