Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Dis Model Mech ; 8(12): 1625-33, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26398949

ABSTRACT

Human umbilical cord matrix-derived stem cells (uMSCs), owing to their cellular and procurement advantages compared with mesenchymal stem cells derived from other tissue sources, are in clinical trials to treat type 1 (T1D) and type 2 diabetes (T2D). However, the therapeutic basis remains to be fully understood. The immunomodulatory property of uMSCs could explain the use in treating T1D; however, the mere immune modulation might not be sufficient to support the use in T2D. We thus tested whether uMSCs could exert direct trophic effects on ß-cells. Infusion of uMSCs into chemically induced diabetic rats prevented hyperglycemic progression with a parallel preservation of islet size and cellularity, demonstrating the protective effect of uMSCs on ß-cells. Mechanistic analyses revealed that uMSCs engrafted long-term in the injured pancreas and the engraftment markedly activated the pancreatic PI3K pathway and its downstream anti-apoptotic machinery. The pro-survival pathway activation was associated with the expression and secretion of ß-cell growth factors by uMSCs, among which insulin-like growth factor 1 (IGF1) was highly abundant. To establish the causal relationship between the uMSC-secreted factors and ß-cell survival, isolated rat islets were co-cultured with uMSCs in the transwell system. Co-culturing improved the islet viability and insulin secretion. Furthermore, reduction of uMSC-secreted IGF1 via siRNA knockdown diminished the protective effects on islets in the co-culture. Thus, our data support a model whereby uMSCs exert trophic effects on islets by secreting ß-cell growth factors such as IGF1. The study reveals a novel therapeutic role of uMSCs and suggests that multiple mechanisms are employed by uMSCs to treat diabetes.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/therapy , Extracellular Matrix/metabolism , Insulin-Secreting Cells/pathology , Stem Cell Transplantation , Stem Cells/cytology , Umbilical Cord/cytology , Animals , Cell Survival , Coculture Techniques , Diabetes Mellitus, Experimental/complications , Disease Progression , Gene Expression Regulation , Humans , Hyperglycemia/complications , Hyperglycemia/pathology , Hyperglycemia/therapy , Insulin-Like Growth Factor I/metabolism , Insulin-Secreting Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rats, Sprague-Dawley , Streptozocin , Tissue Survival , Weight Loss
2.
BMC Pediatr ; 15: 51, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25939992

ABSTRACT

BACKGROUND: To investigate the attention-deficit/hyperactivity disorder (ADHD) status among preschool-aged children in Guiyu, an electronic waste (e-waste) recycling town in Guangdong, China. METHODS: Two hundred and forty-three parents were surveyed regarding ADHD behaviors in their children (aged 3-7 years) based solely on the DSM-IV criteria. The peripheral blood samples were taken from these children to measure blood lead levels (BLLs) and blood cadmium levels (BCLs). RESULTS: 12.8% of children met the criteria for ADHD, of which the inattentive, hyperactive/impulsive and combined subtypes were 4.5%, 5.3% and 2.9% respectively. Of all children, 28.0% had BLLs ≥ 10 ug/dL and only 1.2% had BCLs ≥ 2 ug/L, levels conventionally considered high. Either modeled by univariate or multivariable analysis, the three ADHD scores (inattentive, hyperactive/impulsive and total scores) calculated from the Parent Rating Scale showed strong positive correlations with BLLs but not with BCLs. Furthermore, children with high BLLs had 2.4 times higher risk of ADHD than those with low BLLs (OR: 2.4 [95% CI: 1.1-5.2]). When each of the 18 categories on the Parent Rating Scale was separately analyzed, children with high BLLs had significant higher risks for positive ADHD symptoms than those with low BLLs in 12 of the 18 categories (ORs ranged from 2.1 [95% CI: 1.1-3.9] to 3.6 [95% CI: 1.7-7.5]). CONCLUSIONS: This study suggests that environmental lead contamination due to e-waste recycling has an impact on neurobehavioral development of preschool children in Guiyu.


Subject(s)
Attention Deficit Disorder with Hyperactivity/diagnosis , Environmental Pollutants/adverse effects , Lead/adverse effects , Waste Disposal Facilities , Attention Deficit Disorder with Hyperactivity/epidemiology , Cadmium/adverse effects , Cadmium/blood , Child , Child, Preschool , China/epidemiology , Cross-Sectional Studies , Diagnostic and Statistical Manual of Mental Disorders , Environmental Exposure/adverse effects , Female , Humans , Lead/blood , Male , Prevalence
3.
Mol Med Rep ; 12(1): 819-28, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25815600

ABSTRACT

Our previous study demonstrated that human umbilical cord mesenchymal stem cells (HUMSCs) were capable of differentiation into germ cells in vitro. To assess this potential in vivo, HUMSCs were microinjected into the lumen of seminiferous tubules of immunocompetent mice, which were treated with busulfan to destroy endogenous spermatogenesis. Bromodeoxyuridine labeling studies demonstrated that HUMSCs survived in the tubule for at least 120 days, exhibited a round cell shape typical of proliferating or differentiating germ cells, migrated to the basement of the tubule, where proliferating spermatogonia reside and returned to the luminal compartment, where differentiating spermatids and spermatozoa reside. The migration pattern resembled that of germ cell development in vivo. Immunohistochemical and colocalization studies revealed that transplanted HUMSCs expressed the germ cell markers octamer-binding transcription factor 4, α6 integrin, C-kit and VASA, confirming the germ cell differentiation. In addition, it was observed that tubules transplanted with HUMSCs exhibited marked improvement in the histological features damaged by the chemotherapeutic busulfan, as judged by morphology and quantitative histology. Taken together, these data demonstrated the capacity of HUMSCs to form germ cells in the testes and to repair testicular tissue. These findings suggest a potential utility of HUMSCs to treat the infertility and testicular insufficiency caused by cancer therapeutics.


Subject(s)
Cell Differentiation/genetics , Mesenchymal Stem Cells/cytology , Seminiferous Tubules/growth & development , Spermatogenesis/genetics , Animals , Germ Cells/cytology , Germ Cells/growth & development , Humans , Male , Mice , Spermatozoa/growth & development , Spermatozoa/metabolism , Testis/growth & development , Umbilical Cord/cytology , Umbilical Cord/growth & development
4.
J Mol Endocrinol ; 54(2): 125-35, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25624461

ABSTRACT

Hypothalamic glucose-sensing neurons regulate the expression of genes encoding feeding-related neuropetides POMC, AgRP, and NPY - the key components governing metabolic homeostasis. AMP-activated protein kinase (AMPK) is postulated to be the molecular mediator relaying glucose signals to regulate the expression of these neuropeptides. Whether other signaling mediator(s) plays a role is not clear. In this study, we investigated the role of ERK1/2 using primary hypothalamic neurons as the model system. The primary neurons were differentiated from hypothalamic progenitor cells. The differentiated neurons possessed the characteristic neuronal cell morphology and expressed neuronal post-mitotic markers as well as leptin-regulated orexigenic POMC and anorexigenic AgRP/NPY genes. Treatment of cells with glucose dose-dependently increased POMC and decreased AgRP/NPY expression with a concurrent suppression of AMPK phosphorylation. In addition, glucose treatment dose-dependently increased the ERK1/2 phosphorylation. Blockade of ERK1/2 activity with its specific inhibitor PD98059 partially (approximately 50%) abolished glucose-induced POMC expression, but had little effect on AgRP/NPY expression. Conversely, blockade of AMPK activity with its specific inhibitor produced a partial (approximately 50%) reversion of low-glucose-suppressed POMC expression, but almost completely blunted the low-glucose-induced AgRP/NPY expression. The results indicate that ERK1/2 mediated POMC but not AgRP/NPY expression. Confirming the in vitro findings, i.c.v. administration of PD98059 in rats similarly attenuated glucose-induced POMC expression in the hypothalamus, but again had little effect on AgRP/NPY expression. The results are indicative of a novel role of ERK1/2 in glucose-regulated POMC expression and offer new mechanistic insights into hypothalamic glucose sensing.


Subject(s)
Gene Expression Regulation/drug effects , Glucose/pharmacology , Hypothalamus/cytology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/enzymology , Pro-Opiomelanocortin/genetics , AMP-Activated Protein Kinases/metabolism , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Cell Differentiation , Cells, Cultured , Enzyme Activation/drug effects , Feeding Behavior/drug effects , Female , Flavonoids/pharmacology , Injections, Intraventricular , Neurons/cytology , Neurons/drug effects , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Phosphorylation , Pro-Opiomelanocortin/metabolism , Rats, Sprague-Dawley
5.
Dis Model Mech ; 7(6): 723-30, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24764190

ABSTRACT

Major depression disorder (MDD) or depression is highly prevalent in individuals with diabetes, and the depressive symptoms are more severe and less responsive to antidepressant therapies in these patients. The underlying mechanism is little understood. We hypothesized that the pathophysiology of comorbid depression was more complex than that proposed for MDD and that neural cell death played a role in the disease severity. To test this hypothesis, we generated streptozotocin (STZ)-induced diabetic mice. These mice had blood glucose levels threefold above controls and exhibited depressive phenotypes as judged by a battery of behavioral tests, thus confirming the comorbidity in mice. Immunohistological studies showed markedly increased TUNEL-positive cells in the frontal cortex and hippocampus of the comorbid mice, indicating apoptosis. This finding was supported by increased caspase-3 and decreased Bcl-2 proteins in these brain regions. In addition, the serum brain-derived neurotrophic factor (BDNF) level of comorbid mice was reduced compared with controls, further supporting the neurodegenerative change. Mechanistic analyses showed an increased expression of mitochondrial fission genes fission protein 1 (Fis1) and dynamin-related protein 1 (Drp1), and a decreased expression of mitochondrial fusion genes mitofusin 1 (Mfn1), mitofusin 2 (Mfn2) and optical atrophy 1 (Opa1). Representative assessment of the proteins Drp1 and Mfn2 mirrored the mRNA changes. The data demonstrated that neural cell death was associated with the depressive phenotype of comorbid mice and that a fission-dominant expression of genes and proteins mediating mitochondrial dynamics played a role in the hyperglycemia-induced cell death. The study provides new insight into the disease mechanism and could aid the development of novel therapeutics aimed at providing neuroprotection by modulating mitochondrial dynamics to treat comorbid depression with diabetes.


Subject(s)
Cell Death , Depressive Disorder, Major/complications , Diabetes Mellitus, Experimental/pathology , Neurons/cytology , Animals , Apoptosis , Behavior, Animal , Brain-Derived Neurotrophic Factor/metabolism , Diabetes Mellitus, Experimental/complications , Frontal Lobe/pathology , Hippocampus/pathology , In Situ Nick-End Labeling , Mice , Streptozocin
6.
Int J Mol Med ; 33(2): 263-70, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24297321

ABSTRACT

Islet transplantation involves the transplantation of pancreatic islets from the pancreas of a donor to another individual. It has proven to be an effective method for the treatment of type 1 diabetes. However, islet transplantation is hampered by immune rejection, as well as the shortage of donor islets. Human umbilical cord Wharton's jelly-derived mesenchymal stem cells (HUMSCs) are an ideal cell source for use in transplantation due to their biological characteristics and their use does not provoke any ethical issues. In this study, we investigated the immunological characteristics of HUMSCs and their effects on lymphocyte proliferation and the secretion of interferon (IFN)-γ, and explored whether direct cell-to-cell interactions and soluble factors, such as IFN-γ were important for balancing HUMSC-mediated immune regulation. We transplanted HUMSCs into diabetic rats to investigate whether these cells can colonize in vivo and differentiate into pancreatic ß-cells, and whether the hyperglycemia of diabetic rats can be improved by transplantation. Our results revealed that HUMSCs did not stimulate the proliferation of lymphocytes and did not induce allogeneic or xenogeneic immune cell responses. qRT-PCR demonstrated that the HUMSCs produced an immunosuppressive isoform of human leukocyte antigen (HLA-I) and did not express HLA-DR. Flow cytometry revealed that the HUMSCs did not express immune response-related surface antigens such as, CD40, CD40L, CD80 and CD86. IFN-γ secretion by human peripheral blood lymphocytes was reduced when the cells were co-cultured with HUMSCs. These results suggest that HUMSCs are tolerated by the host in an allogeneic transplant. We transplanted HUMSCs into diabetic rats, and the cells survived in the liver and pancreas. Hyperglycemia of the diabetic rats was improved and the destruction of pancreatic cells was partly repaired by HUMSC transplantation. Hyperglycemic improvement may be related to the immunomodulatory effects of HUMSCs. However, the exact mechanisms involved remain to be further clarified.


Subject(s)
Hyperglycemia/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Umbilical Cord/cytology , Animals , Antigens, CD/metabolism , Cell Communication , Cell Proliferation , Cells, Cultured , Coculture Techniques , Disease Models, Animal , HLA-A Antigens/metabolism , HLA-DR Antigens/metabolism , Humans , Hyperglycemia/immunology , Insulin-Secreting Cells/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mesenchymal Stem Cells/immunology , Phenotype , Rats
7.
PLoS One ; 8(12): e81844, 2013.
Article in English | MEDLINE | ID: mdl-24339974

ABSTRACT

Human umbilical cord mesenchymal stem cells (HUMSCs) are highly proliferative and can be induced to differentiate into advanced derivatives of all three germ layers. Thus, HUMSCs are considered to be a promising source for cell-targeted therapies and tissue engineering. However there are reports on spontaneous transformation of mesenchymal stem cells (MSCs) derived from human bone marrows. The capacity for HUMSCs to undergo malignant transform spontaneously or via induction by chemical carcinogens is presently unknown. Therefore, we isolated HUMSCs from 10 donors and assessed their transformation potential either spontaneously or by treating them with 3-methycholanthrene (3-MCA), a DNA-damaging carcinogen. The malignant transformation of HUMSCs in vitro was evaluated by morphological changes, proliferation rates, ability to enter cell senescence, the telomerase activity, chromosomal abnormality, and the ability to form tumors in vivo. Our studies showed that HUMSCs from all 10 donors ultimately entered senescence and did not undergo spontaneous malignant transformation. However, HUMSCs from two of the 10 donors treated with 3-MCA displayed an increased proliferation rate, failed to enter senescence, and exhibited an altered cell morphology. When these cells (tHUMSCs) were injected into immunodeficient mice, they gave rise to sarcoma-like or poorly differentiated tumors. Moreover, in contrast to HUMSCs, tHUMSCs showed a positive expression of human telomerase reverse transcriptase (hTERT) and did not exhibit a shortening of the relative telomere length during the long-term culture in vitro. Our studies demonstrate that HUMSCs are not susceptible to spontaneous malignant transformation. However, the malignant transformation could be induced by chemical carcinogen 3-MCA.


Subject(s)
Carcinogens/pharmacology , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Chromosome Aberrations/chemically induced , Fetal Blood/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cellular Senescence/drug effects , DNA Damage , Humans , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred BALB C
8.
PLoS One ; 8(12): e82502, 2013.
Article in English | MEDLINE | ID: mdl-24324800

ABSTRACT

In this study, we attempted to design a model using Sprague-Dawley rats to better reproduce perinatal systemic hypoxic-ischemic encephalopathy (HIE) in early preterm newborns. On day 21 of gestation, the uterus of pregnant rats were exposed and the blood supply to the fetuses of neonatal HIE groups were thoroughly abscised by hemostatic clamp for 5, 10 or 15 min. Thereafter, fetuses were moved from the uterus and manually stimulated to initiate breathing in an incubator at 37 °C for 1 hr in air. We showed that survival rates of offspring rats were decreased with longer hypoxic time. TUNEL staining showed that apoptotic cells were significant increased in the brains of offspring rats from the 10 min and 15 min HIE groups as compared to the offspring rats in the control group at postnatal day (PND) 1, but there was no statistical difference between the offspring rats in the 5 min HIE and control groups. The perinatal hypoxic treatment resulted in decreased neurons and increased cleaved caspase-3 protein levels in the offspring rats from all HIE groups at PND 1. Platform crossing times and the percentage of the time spent in the target quadrant of Morris Water Maze test were significantly reduced in the offspring rats of all HIE groups at PND 30, which were associated with decreased brain-derived neurotrophic factor levels and neuronal cells in the hippocampus of offspring rats at PND 35. These data demonstrated that perinatal ischemic injury led to the death of neuronal cells and long-lasting impairment of memory. This model reproduced hypoxic ischemic encephalopathy in early preterm newborns and may be appropriate for investigating therapeutic interventions.


Subject(s)
Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Premature Birth , Age Factors , Animals , Animals, Newborn , Apoptosis , Brain/metabolism , Brain/pathology , Brain-Derived Neurotrophic Factor/metabolism , Caspase 3/metabolism , Disease Models, Animal , Female , Hippocampus/metabolism , Hypoxia-Ischemia, Brain/mortality , Male , Maze Learning , Memory , Neurons/metabolism , Pregnancy , Rats
9.
Mol Med Rep ; 8(3): 769-74, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23900717

ABSTRACT

Human umbilical cord mesenchymal stem cells (HUMSCs) are candidates for tissue engineering and may potentially be used for transdifferentiation into pancreatic endocrine cells. The adenoviral vector is effective in transducing genes into stem cells that are refractory to gene delivery by non­viral approaches. qPCR was used to detect the pancreatic endogenous gene expression of HUMSCs transfected by islet cell-specific transcription factors (TFs). In the present study, using adenoviruses, the mouse TFs, pancreatic and duodenal homeobox 1 (pdx1), V-maf musculoaponeurotic fibrosarcoma oncogene homolog A (mafa) and class B basic helix­loop­helix factor neurogenin 3 (ngn3), which are essential for pancreatic cell development, were introduced into HUMSCs to assess the expression of the pancreatic genes, glucagon, pdx1 and nk2 homeobox 2 (nkx2.2). When pdx1, mafa and ngn3 were cotransduced into HUMSCs, the expression of glucagon increased by 21­fold at days 3 and 7 following transduction, while the endogenous pdx1 gene expression was increased by 15­fold at day 3 and decreased by 70% at day 7. When mafa and ngn3 were cotransduced into HUMSCs, there was a 5­fold increase in pdx1 gene expression at day 7, but no activation was observed at day 3. When mafa alone was introduced into HUMSCs, the pdx1 gene expression was elevated by 6­fold at day 3 and decreased by 3­fold at day 7. Transduction of ngn3 alone into HUMSCs induced nkx2.2 gene expression at day 3 but the expression levels were decreased at day 7. However, when pdx1 and ngn3 were cotransduced into HUMSCs, the expression levels of glucagon, pdx1 and nks2.2 were all lower than those observed with pdx1 or ngn3 transduction alone. These results suggested that the transduction of pdx1, mafa and ngn3 genes into HUMSCs induced the expression of the pancreatic genes, glucagon, pdx­1 and nkx2.2, and that the expression was time dependent. In addition, different combinations of the TFs may demonstrate synergistic or antagonistic effects. This data may be beneficial for guiding future studies obtaining mature pancreatic endocrine cells from HUMSCs.


Subject(s)
Cellular Reprogramming , Mesenchymal Stem Cells/cytology , Pancreas/metabolism , Umbilical Cord/cytology , Adenoviridae/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Transdifferentiation , Gene Expression Regulation , Genetic Vectors/metabolism , Glucagon/genetics , Glucagon/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins , Pancreas/cytology , Phenotype , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors
10.
BMC Neurosci ; 14: 67, 2013 Jul 08.
Article in English | MEDLINE | ID: mdl-23829597

ABSTRACT

BACKGROUND: There is increasing evidence that maternal stress may have long-term effects on brain development in the offspring. In this study, we examined whether pre-gestational stress might affect offspring rats on the medial prefrontal cortical (mPFC) dopaminergic activity in response to acute stress in puberty and if so, whether such effects exhibited hemispheric asymmetry or sexual dimorphism. RESULTS: We used behavioral tests to assess the model of chronic unpredictable stress (CUS). We found that the activity in the open field test and sucrose intake test were lower for maternal rats in the CUS group than those in the control group. Offspring rats in the CUS group floated more and swam or climbed less as compared to the offsprings in the control group in the forced swimming test. The floating time was longer and swimming or climbing time was shorter in the female offspring rats than those in the males. Serum corticosterone and corticotrophin-releasing hormone levels were significantly higher for CUS maternal rats and their offsprings than the respective controls. The ratio of dihydroxy-phenyl acetic acid (DOPAC) to dopamine (DA), DA transporter (DAT), norepinephrine transporter (NET) were lower in the mPFC of offspring rats in the CUS group than the control group. Levels of catechol-O-methyltransferase (COMT) in the left mPFC of female offspring rats and in the right mPFC of both female and male offspring rats were lower in the CUS group than those in the controls, but there was no difference in the left mPFC of male offspring between the CUS and control groups. DOPAC, the ratio of DOPAC to DA, NET and COMT were lower in the right mPFC than in the left mPFC of offspring rats in the CUS group. The ratio of DOPAC to DA in the right mPFC was lower in the female offspring rats than male offspring rats in the CUS group. The NET and COMT levels in both left and right mPFC were lower in the female offspring rats than those of the male offsprings in the CUS group. CONCLUSION: Our data provide evidence that the effect of pre-gestational stress on the mPFC dopaminergic activity in response to acute stress exhibited hemispheric asymmetry and sexual dimorphism in the pubertal offspring rats.


Subject(s)
Functional Laterality/physiology , Prefrontal Cortex/pathology , Prenatal Exposure Delayed Effects/physiopathology , Sex Characteristics , Stress, Psychological/etiology , Stress, Psychological/pathology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Analysis of Variance , Animals , Animals, Newborn , Body Weight , Catechol O-Methyltransferase/metabolism , Catecholamine Plasma Membrane Transport Proteins/metabolism , Corticosterone/blood , Corticotropin-Releasing Hormone/blood , Cues , Dopamine/metabolism , Eating , Exploratory Behavior , Female , Gene Expression Regulation, Developmental , Male , Movement/physiology , Prefrontal Cortex/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Rats , Rats, Sprague-Dawley , Stress, Psychological/blood , Swimming/psychology
11.
Pharmacology ; 79(2): 114-21, 2007.
Article in English | MEDLINE | ID: mdl-17179741

ABSTRACT

In this study, we investigated the activation of the serum response element (SRE) by the D2 dopamine receptor (D2R) agonist quinpirole. Stimulation of CHO cells expressing the D2R by quinpirol evoked a dose-dependent SRE activation, which was completely blocked by overnight treatment of pertussis toxin or by co-expression of the beta-adrenergic receptor kinase C-terminus, implicating the involvement of Galpha(i )and Gbetagamma in the signal transduction. Furthermore, using MEK inhibitors and dominant negative mutants of RhoA, Rac1, and Cdc42, we showed that the Gbetagamma-mediated activation of the SRE in CHO cells utilizes both MAPK and Rho pathways. Expression of either regulator of G protein signaling 2 or 4 (RGS2 or RGS4) proteins significantly attenuated the quinpirole-induced SRE activation. These results delineate the signaling pathways which couple D2 receptor to the transcriptional activation of SRE and demonstrate a modulatory role for RGS proteins in these processes.


Subject(s)
Heterotrimeric GTP-Binding Proteins/physiology , Mitogen-Activated Protein Kinases/physiology , RGS Proteins/physiology , Receptors, Dopamine D2/physiology , Serum Response Element , rho GTP-Binding Proteins/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits/physiology , GTP-Binding Protein beta Subunits/physiology , GTP-Binding Protein gamma Subunits/physiology , Genes, Reporter , Pertussis Toxin/pharmacology , Quinpirole/pharmacology , Receptors, Dopamine D2/agonists , Signal Transduction , beta-Adrenergic Receptor Kinases/metabolism
12.
J Biol Chem ; 278(18): 15935-40, 2003 May 02.
Article in English | MEDLINE | ID: mdl-12594226

ABSTRACT

Loss-of-function mutations in the human melanocortin-4 receptor (MC4R) are associated with obesity. Previous work has implicated a C-terminal di-isoleucine motif at residues 316/317 in MC4R cell surface targeting. It was therefore of interest to examine function and cell surface expression of an MC4R mutation found in an obese proband in which one of these isoleucines was substituted by threonine (I317T). Single mutant (I316T or I317T) and double mutant (I316T,I317T) forms of MC4R were constructed by oligonucleotide-directed mutagenesis and tested for function and cell surface expression in transfected cells. Function was assessed using assays for agonist, [Nle(4)-d-Phe(7)]alpha-melanocyte-stimulating hormone (NDP-alpha-MSH) or forskolin-stimulated cAMP accumulation. Cell surface expression was determined by whole-cell binding of [(125)I]NDP-alpha-MSH, fluorescence immunocytochemistry and fluorescence-activated cell sorting. Maximal cAMP generation of the single mutants was reduced by 40% of wild-type receptor; the double mutant further reduced function to 40% of control, effects that were mirrored by decreases in cell-surface expression. Quantitative RT-PCR showed that, relative to wild-type receptor, transcript levels for the mutated receptors were not reduced. The results further implicate the C-terminal di-isoleucines in cell surface expression of MC4R and suggest that mutations of residues 316 or 317 would predict MC4R hypofunction.


Subject(s)
Codon , Receptors, Corticotropin/chemistry , Receptors, Corticotropin/genetics , Amino Acid Sequence , Animals , COS Cells , Cell Membrane/chemistry , Cyclic AMP/metabolism , Humans , Isoleucine , Microscopy, Fluorescence , Molecular Sequence Data , RNA, Messenger/analysis , Receptor, Melanocortin, Type 4 , Structure-Activity Relationship , Transfection
13.
J Biol Chem ; 277(50): 48325-32, 2002 Dec 13.
Article in English | MEDLINE | ID: mdl-12379657

ABSTRACT

Regulator of G protein signaling (RGS) proteins constitute a family of over 20 proteins that negatively regulate heterotrimeric G protein-coupled receptor signaling pathways by enhancing endogenous GTPase activities of G protein alpha subunits. RGSZ1, one of the RGS proteins specifically localized to the brain, has been cloned previously and described as a selective GTPase accelerating protein for Galpha(z) subunit. Here, we employed several methods to provide new evidence that RGSZ1 interacts not only with Galpha(z,) but also with Galpha(i), as supported by in vitro binding assays and functional studies. Using glutathione S-transferase fusion protein pull-down assays, glutathione S-transferase-RGSZ1 protein was shown to bind (35)S-labeled Galpha(i1) protein in an AlF(4)(-)dependent manner. The interaction between RGSZ1 and Galpha(i) was confirmed further by co-immunoprecipitation studies and yeast two-hybrid experiments using a quantitative luciferase reporter gene. Extending these observations to functional studies, RGSZ1 accelerated endogenous GTPase activity of Galpha(i1) in single-turnover GTPase assays. Human RGSZ1 functionally regulated GPA1 (a yeast Galpha(i)-like protein)-mediated yeast pheromone response when expressed in a SST2 (yeast RGS protein) knockout strain. In PC12 cells, transfected RGSZ1 blocked mitogen-activated protein kinase activity induced by UK14304, an alpha(2)-adrenergic receptor agonist. Furthermore, RGSZ1 attenuated D2 dopamine receptor agonist-induced serum response element reporter gene activity in Chinese hamster ovary cells. In summary, these data suggest that RGSZ1 serves as a GTPase accelerating protein for Galpha(i) and regulates Galpha(i)-mediated signaling, thus expanding the potential role of RGSZ1 in G protein-mediated cellular activities.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTPase-Activating Proteins , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction/physiology , Adrenergic alpha-2 Receptor Antagonists , Animals , Enzyme Activation , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Humans , Membrane Proteins/physiology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Nerve Tissue Proteins/physiology , PC12 Cells , Precipitin Tests , Protein Binding , RGS Proteins , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...