199 results on '"Chou, Janice Y."'
Search Results
152. Molecular characterization of Plasmodium falciparum S-adenosylmethionine synthetase
- Author
-
CHIANG, Peter K., primary, CHAMBERLIN, Margaret E., additional, NICHOLSON, Diarmuid, additional, SOUBES, Sandrine, additional, SU, Xin-zhuan, additional, SUBRAMANIAN, Gangadharan, additional, LANAR, David E., additional, PRIGGE, Sean T., additional, SCOVILL, John P., additional, MILLER, Louis H., additional, and CHOU, Janice Y., additional
- Published
- 1999
- Full Text
- View/download PDF
153. Pregnancy-Specific Glycoprotein Gene Expression in Recurrent Aborters: A Potential Correlation to Interleukin-10 Expression
- Author
-
Arnold, Lori L., primary, Doherty, T. Mark, additional, Flor, Armando W., additional, Simon, James A., additional, Chou, Janice Y., additional, Chan, Wai-Yee, additional, and Mansfield, Brian C., additional
- Published
- 1999
- Full Text
- View/download PDF
154. Consensus nomenclature for the mammalian methionine adenosyltransferase genes and gene products
- Author
-
Kotb, Malak, primary, Mudd, S.Harvey, additional, Mato, Jose M., additional, Geller, Arthur M., additional, Kredich, Nicholas M., additional, Chou, Janice Y., additional, and Cantoni, Gulio L., additional
- Published
- 1997
- Full Text
- View/download PDF
155. Two New Mutations in the Glucose-6-Phosphatase Gene Cause Glycogen Storage Disease in Hungarian Patients
- Author
-
Parvari, Ruti, primary, Lei, Ke-Jian, additional, Szonyi, Laszlo, additional, Narkis, Ginat, additional, Moses, Shimon, additional, and Chou, Janice Y., additional
- Published
- 1997
- Full Text
- View/download PDF
156. Cloning and characterization of the 5′-flanking region of the mouse tartrate-resistant acid phosphatase gene
- Author
-
Reddy, Sakamuri V., primary, Scarcez, Thierry, additional, Windle, Jolene J., additional, Leach, Robin J., additional, Hundley, Jeffrey E., additional, Chirgwin, John M., additional, Chou, Janice Y., additional, and Roodman, G. David, additional
- Published
- 1993
- Full Text
- View/download PDF
157. Differentiation and uteroglobin gene expression by novel rabbit endometrial cell lines
- Author
-
Mukherjee, Anil B., primary, Murty, Lalita C., additional, and Chou, Janice Y., additional
- Published
- 1993
- Full Text
- View/download PDF
158. Regulation of the Uteroferrin Gene Promoter in Endometrial Cells: Interactions among Estrogen, Progesterone, and Prolactin*
- Author
-
FLISS, ALBERT E., primary, MICHEL, FRANK J., additional, CHEN, CHAO-LING, additional, HOFIG, ANDREA, additional, BAZER, FULLER W., additional, CHOU, JANICE Y., additional, and SIMMEN, ROSALIA C. M., additional
- Published
- 1991
- Full Text
- View/download PDF
159. The glucose-6-phosphate transporter is a phosphate-linked antiporter deficient in glycogen storage disease type Ib and Ic.
- Author
-
Shih-Yin Che, Chi-Jiunn Pan, Nandigama, Krishnamachary, Mansfield, Brian C., Ambudkar, Suresh V., and Chou, Janice Y.
- Subjects
GLUCOSE-6-phosphatase ,GLYCOGEN storage disease ,ENDOPLASMIC reticulum ,VANADATES ,PHOSPHATES - Abstract
Glycogen storage disease type Ib (GSD-Ib) is caused by deficiencies in the glucose-6-phosphate (G6P) transporter (G6PT) that have been well characterized. Interestingly, deleterious mutations in the G6PT gene were identified in clinical cases of GSD type Ic (GSD-Ic) proposed to be deficient in an inorganic phosphate (P
i ) transporter. We hypothesized that G6PT is both the G6P and PPi transporter. Using reconstituted proteoliposomes we show that both G6P and PPi are efficiently taken up into PPi -loaded G6PT-proteoliposomes. The G6P uptake activity decreases as the internal:external PPi ratio decreases and the PPi uptake activity decreases in the presence of external G6P. Moreover, G6P or PPi uptake activity is not detectable in PPi -loaded proteoliposomes containing the p.R28H G6PT null mutant. The G6PT-proteoliposome-mediated G6P or PPi uptake is inhibited by cholorgenic acid and vanadate, both specific G6PT inhibitors. Glucose-6-phosphatase-α (G6Pase-α), which facilitates microsomal G6P uptake by G6PT, fails to stimulate G6P uptake in PPi -loaded G6PT-proteoliposomes, suggesting that the G6Pase-α-mediated stimulation is caused by decreasing G6P and increasing PPi concentrations in microsomes. Taken together, our results suggest that G6PT has a dual role as a G6P and a PPi transporter and that GSD-Ib and GSD-Ic are deficient in the same G6PT gene.--Chen, S.-Y., Pan, C.-.J, Nandigama, K., Mansfield, B., Ambudkar, S., Chou, J. The glucose-6-phosphate transporter is a phosphate-linked antiporter deficient in glycogen storage disease type Ib and and Ic. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
160. Induction of alkaline phosphatase in choriocarcinoma cells by 1-β-D-arabinofuranosyl-cytosine, mitomycin C, phleomycin, and cyclic nucleotides.
- Author
-
Chou, Janice Y. and Robinson, J. C.
- Published
- 1977
- Full Text
- View/download PDF
161. Cytomegalovirus Replication in Primary and Passaged Human Placental Cells.
- Author
-
Rosenthal, Leonard J., Panitz, Polly J., Crutchfield, D.B., and Chou, Janice Y.
- Published
- 1981
- Full Text
- View/download PDF
162. Oxidative stress mediates nephropathy in type Ia glycogen storage disease.
- Author
-
Wai Han Yiu, Mead, Paul A., Hyun Sik Jun, Mansfield, Brian C., and Chou, Janice Y.
- Published
- 2010
- Full Text
- View/download PDF
163. Human Trophoblasts: Cellular Source of Colony-Stimulating Activity in Placental Tissue
- Author
-
Ruscetti, Francis W., Chou, Janice Y., and Gallo, Robert C.
- Abstract
Colony-stimulating activity (CSA) is a class of protein factors that stimulates the in vitro growth and differentiation of hematopoietic committed stem cells into mature granulocytes and macrophages. In man, production of CSA is mediated by monocytes-macrophages, lymphocyte, and endothelial cells. One of the best studied and most potent sources of CSA is conditioned media derived from cultured human placenta. The cellular source of this placental CSA was studied using cloned term placental cell lines induced by a simian virus 40 (SV-40) Wild-Type (wt) or temperature sensitive (tsA) mutants. Conditioned media from SV-40 wt-transformed placental cells grown at 37°C and tsA-transformed placental cells grown at 33°C (the temperature at which the cells exhibit the transformed phenotype) and at 40°C (temperature at which the cells express a normal differentiated phenotype) contained high levels of CSA. At the restricted temperature (40°C), these cell lines expressed normal trophoblastic characteristics in that they produced human chorionic gonadotropin, pregnancy-specific-β1-glyco protein, and placental alkaline phosphatase, indicating that the SV-40 transformation has not interfered with normal cellular functions of these trophoblasts. The CSA released by these cell lines resemble those of CM from fresh placental cells, in that the level of activity is high and that formation of both granulocyte/ macrophage and eosinophil colonies is stimulated.
- Published
- 1982
- Full Text
- View/download PDF
164. HeLa cells secrete α subunit of glycoprotein tropic hormones.
- Author
-
LIEBLICH, JEFFREY M., WEINTRAUB, BRUCE D., ROSEN, SAUL W., CHOU, JANICE Y., and ROBINSON, J. C.
- Published
- 1976
- Full Text
- View/download PDF
165. Response letter.
- Author
-
Lee, Youngmok, Chou, Janice Y., and Weinstein, David A.
- Published
- 2018
- Full Text
- View/download PDF
166. Insulin receptors in a new human placenta cell line: Demonstration of negative cooperativity
- Author
-
Podskalny, Judith M., primary, Chou, Janice Y., additional, and Rechler, Matthew M., additional
- Published
- 1975
- Full Text
- View/download PDF
167. Induction of placental alkaline phosphatase in choriocarcinoma cells by 5-bromo-2′-deoxyuridine
- Author
-
Chou, Janice Y., primary and Robinson, J. C., additional
- Published
- 1977
- Full Text
- View/download PDF
168. Induction of alkaline phosphatase in choriocarcinoma cells by 1-?-D-arabinofuranosyl-cytosine, mitomycin C, phleomycin, and cyclic nucleotides
- Author
-
Chou, Janice Y., primary and Robinson, J. C., additional
- Published
- 1977
- Full Text
- View/download PDF
169. Effects of sodium butyrate on synthesis of human chorionic gonadotrophin in trophoblastic and non-trophoblastic tumours
- Author
-
CHOU, JANICE Y., primary, ROBINSON, J. C., additional, and WANG, SHAW-SHYAN, additional
- Published
- 1977
- Full Text
- View/download PDF
170. Phosphoprotein-phosphatase activity associated with human placental alkaline phosphatase
- Author
-
Huang, Kuo-Ping, primary, Robinson, J.C., additional, and Chou, Janice Y., additional
- Published
- 1976
- Full Text
- View/download PDF
171. Preface
- Author
-
Chou, Janice Y. and Raben, Nina
- Published
- 2002
172. Inhibition of Wnt/β-catenin signaling reduces renal fibrosis in murine glycogen storage disease type Ia.
- Author
-
Lee, Cheol, Pratap, Kunal, Zhang, Lisa, Chen, Hung Dar, Gautam, Sudeep, Arnaoutova, Irina, Raghavankutty, Mahadevan, Starost, Matthew F., Kahn, Michael, Mansfield, Brian C., and Chou, Janice Y.
- Subjects
- *
RENAL fibrosis , *GLYCOGEN storage disease , *EXTRACELLULAR matrix proteins , *ACUTE kidney failure , *KIDNEY cortex , *CATENINS , *WNT proteins - Abstract
Glycogen storage disease type Ia (GSD-Ia) is caused by a deficiency in the enzyme glucose-6-phosphatase-α (G6Pase-α or G6PC) that is expressed primarily in the gluconeogenic organs, namely liver, kidney cortex, and intestine. Renal G6Pase-α deficiency in GSD-Ia is characterized by impaired gluconeogenesis, nephromegaly due to elevated glycogen accumulation, and nephropathy caused, in part, by renal fibrosis, mediated by activation of the renin-angiotensin system (RAS). The Wnt/β-catenin signaling regulates the expression of a variety of downstream mediators implicated in renal fibrosis, including multiple genes in the RAS. Sustained activation of Wnt/β-catenin signaling is associated with the development and progression of renal fibrotic lesions that can lead to chronic kidney disease. In this study, we examined the molecular mechanism underlying GSD-Ia nephropathy. Damage to the kidney proximal tubules is known to trigger acute kidney injury (AKI) that can, in turn, activate Wnt/β-catenin signaling. We show that GSD-Ia mice have AKI that leads to activation of the Wnt/β-catenin/RAS axis. Renal fibrosis was demonstrated by increased renal levels of Snail1, α-smooth muscle actin (α-SMA), and extracellular matrix proteins, including collagen-Iα1 and collagen-IV. Treating GSD-Ia mice with a CBP/β-catenin inhibitor, ICG-001, significantly decreased nuclear translocated active β-catenin and reduced renal levels of renin, Snail1, α-SMA, and collagen-IV. The results suggest that inhibition of Wnt/β-catenin signaling may be a promising therapeutic strategy for GSD-Ia nephropathy. [Display omitted] • GSD-Ia mice display acute kidney injury. • GSD-Ia nephropathy is caused, in part, by activation of Wnt/β-catenin signaling. • Drug inhibition of the Wnt/β-catenin pathway reduces renal fibrosis in GSD-Ia. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
173. Molecular mechanisms of neutrophil dysfunction in glycogen storage disease type lb.
- Author
-
Jun, Hyun Sik, Weinstein, David A., Lee, Young Mok, Mansfield, Brian C., and Chou, Janice Y.
- Subjects
- *
NEUTROPHILS , *GLYCOGEN storage disease , *MOLECULES , *GLUCOSE , *HOMEOSTASIS - Abstract
Glycogen storage disease type lb (GSD-lb) is an autosomal-recessive syndrome characterized by neutropenia and impaired glucose homeostasis resulting from a deficiency in the glucose-6-phosphate (G6P) transporter (G6PT). The underlying cause of GSD-lb neutropenia is an enhanced neutrophil apoptosis, but patients also manifest neutrophil dysfunction of unknown etiology. Previously, we showed G6PT interacts with the enzyme glucose-6-phosphatase-β (G6Pase-β) to regulate the availability of G6P/glucose in neutrophils. A deficiency in G6Pase-p activity in neutrophils impairs both their energy homeostasis and function. We now show that G6PT-deficient neutrophils from GSD-lb patients are similarly impaired. Their energy impairment is characterized by decreased glucose uptake and reduced levels of intracellular G6P, lactate, adenosine triphosphate, and reduced NAD phosphate, whereas functional impairment is reflected in reduced neutrophil respiratory burst, chemotaxis, and calcium mobilization. We further show that the mechanism of neutrophil dysfunction in GSD-lb arises from activation of the hypoxia-inducible factor-Wperoxisome-proliterators-activated receptor pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
174. Glucose-6-phosphatase-β, implicated in a congenital neutropenia syndrome, is essential for macrophage energy homeostasis and functionality.
- Author
-
Hyun Sik Jun, Yuk Yin Cheung, Young Mok Lee, Mansfield, Brian C., and Chou, Janice Y.
- Subjects
- *
GLUCOSE phosphates , *NEUTROPENIA , *HOMEOSTASIS , *NEUTROPHILS , *GENE expression , *CHEMOTAXIS , *CHROMOSOMAL translocation - Abstract
Glucose-6-phosphatase-β (G6Pase-β or G6PC3) deficiency, also known as severe congenital neutropenia syndrome 4, is characterized not only by neutropenia but also by impaired neutrophil energy homeostasis and functionality. We now show the syndrome is also associated with macrophage dysfunction, with murine G6pc3-/- macrophages having impairments in their respiratory burst, Chemotaxis, calcium flux, and phagocytic activities. Consistent with a glucose-6-phosphate (G6P) metabolism deficiency, G6pc3-/- macrophages also have a lower glucose uptake and lower levels of G6P, lactate, and ATP than wild-type macrophages. Furthermore, the expression of NADPH oxidase subunits and membrane translocation of p47Phox are down-regulated, and G6pc3-/- macrophages exhibit repressed trafficking in vivo both during an inflammatory response and in pregnancy. During pregnancy, the absence of G6Pase-ß activity also leads to impaired energy homeostasis in the uterus and reduced fertility of G6pc3-/- mothers. Together these results show that immune deficiencies in this congenital neutropenia syndrome extend beyond neutrophil dysfunction. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
175. Treatment of newborn G6pc−/− mice with bone marrow-derived myelomonocytes induces liver repair
- Author
-
Resaz, Roberta, Emionite, Laura, Vanni, Cristina, Astigiano, Simonetta, Puppo, Maura, Lavieri, Rosa, Segalerba, Daniela, Pezzolo, Annalisa, Bosco, Maria Carla, Oberto, Alessandra, Eva, Carola, Chou, Janice Y., Varesio, Luigi, Barbieri, Ottavia, and Eva, Alessandra
- Subjects
- *
LIVER cells , *BONE marrow , *LIVER regeneration , *GLUCOSE-6-phosphatase , *IMMUNOHISTOCHEMISTRY , *CELL transplantation , *LABORATORY mice - Abstract
Background & Aims: Several studies have shown that bone marrow-derived committed myelomonocytic cells can repopulate diseased livers by fusing with host hepatocytes and can restore normal liver function. These data suggest that myelomonocyte transplantation could be a promising approach for targeted and well-tolerated cell therapy aimed at liver regeneration. We sought to determine whether bone marrow-derived myelomonocytic cells could be effective for liver reconstitution in newborn mice knock-out for glucose-6-phosphatase-α. Methods: Bone marrow-derived myelomonocytic cells obtained from adult wild type mice were transplanted in newborn knock-out mice. Tissues of control and treated mice were frozen for histochemical analysis, or paraffin-embedded and stained with hematoxylin and eosin for histological examination or analyzed by immunohistochemistry or fluorescent in situ hybridization. Results: Histological sections of livers of treated knock-out mice revealed areas of regenerating tissue consisting of hepatocytes of normal appearance and partial recovery of normal architecture as early as 1 week after myelomonocytic cells transplant. FISH analysis with X and Y chromosome paints indicated fusion between infused cells and host hepatocytes. Glucose-6-phosphatase activity was detected in treated mice with improved profiles of liver functional parameters. Conclusions: Our data indicate that bone marrow-derived myelomonocytic cell transplant may represent an effective way to achieve liver reconstitution of highly degenerated livers in newborn animals. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
176. Complete Normalization of Hepatic G6PC Deficiency in Murine Glycogen Storage Disease Type Ia Using Gene Therapy.
- Author
-
Wai Han Yiu, Young Mok Lee, Wen-Tao Peng, Chi-Jiunn Pan, Mead, Paul A., Mansfield, Brian C., and Chou, Janice Y.
- Subjects
- *
GLYCOGEN storage disease , *GENE therapy , *SEROTYPES , *TRANSGENE expression , *IMMUNE response , *BLOOD sugar , *THERAPEUTICS - Abstract
Glycogen storage disease type Ia (GSD-Ia) patients deficient in glucose-6-phosphatase-α (G6Pase-α or G6PC) manifest disturbed glucose homeostasis. We examined the efficacy of liver G6Pase-α delivery mediated by AAV-GPE, an adeno-associated virus (AAV) serotype 8 vector expressing human G6Pase-α directed by the human G6PC promoter/enhancer (GPE), and compared it to AAV-CBA, that directed murine G6Pase-α expression using a hybrid chicken β-actin (CBA) promoter/cytomegalovirus (CMV) enhancer. The AAV-GPE directed hepatic G6Pase-α expression in the infused G6pc−/− mice declined 12-fold from age 2 to 6 weeks but stabilized at wild-type levels from age 6 to 24 weeks. In contrast, the expression directed by AAV-CBA declined 95-fold over 24 weeks, demonstrating that the GPE is more effective in directing persistent in vivo hepatic transgene expression. We further show that the rapid decline in transgene expression directed by AAV-CBA results from an inflammatory immune response elicited by the AAV-CBA vector. The AAV-GPE-treated G6pc−/− mice exhibit normal levels of blood glucose, blood metabolites, hepatic glycogen, and hepatic fat. Moreover, the mice maintained normal blood glucose levels even after 6 hours of fasting. The complete normalization of hepatic G6Pase-α deficiency by the G6PC promoter/enhancer holds promise for the future of gene therapy in human GSD-Ia patients. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
177. Impaired neutrophil activity and increased susceptibility to bacterial infection in mice lacking glucose-6-phosphatase--β.
- Author
-
Yuk Yin Cheung, So Youn Kim, Wai Han Yiu, Chi-Jiunn Pan, Hyun-Sik Jun, Ruef, Robert A., Lee, Eric J., Westphal, Heiner, Mansfield, Brian C., and Chou, Janice Y.
- Subjects
- *
NEUTROPENIA , *GRANULOCYTOPENIA , *ETIOLOGY of diseases , *NEUTROPHILS , *GLUCOSE-6-phosphatase , *BACTERIAL diseases - Abstract
Neutropenia and neutrophil dysfunction are common in many diseases, although their etiology is often unclear. Previous views held that there was a single ER enzyme, glucose-6-phosphatase-α (G6Pase-α), whose activity — limited to the liver, kidney, and intestine — was solely responsible for the final stages of gluconeogenesis and glycogenolysis, in which glucose-6-phosphate (G6P) is hydrolyzed to glucose for release to the blood. Recently, we characterized a second G6Pase activity, that of G6Pase-β (also known as G6PC), which is also capable of hydrolyzing G6P to glucose but is ubiquitously expressed and not implicated in interprandial blood glucose homeostasis. We now report that the absence of G6Pase-β led to neutropenia; defects in neutrophil respiratory burst, chemotaxis, and calcium flux; and increased susceptibility to bacterial infection. Consistent with this, G6Pase-β-deficient (G6pc3-/-) mice with experimental peritonitis exhibited increased expression of the glucose-regulated proteins upregulated during ER stress in their neutrophils and bone marrow, and the G6pc3-/- neutrophils exhibited an enhanced rate of apoptosis. Our results define a molecular pathway to neutropenia and neutrophil dysfunction of previously unknown etiology, providing a potential model for the treatment of these conditions. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
178. CRISPR/Cas9-based double-strand oligonucleotide insertion strategy corrects metabolic abnormalities in murine glycogen storage disease type-Ia.
- Author
-
Samanta A, George N, Arnaoutova I, Chen HD, Mansfield BC, Hart C, Carlo T, and Chou JY
- Subjects
- Mice, Animals, CRISPR-Cas Systems, Liver metabolism, Glucose-6-Phosphatase genetics, Glucose-6-Phosphatase metabolism, Oligonucleotides metabolism, Glycogen Storage Disease Type I genetics, Glycogen Storage Disease Type I therapy, Glycogen Storage Disease Type I metabolism
- Abstract
Glycogen storage disease type-Ia (GSD-Ia), characterized by impaired blood glucose homeostasis, is caused by a deficiency in glucose-6-phosphatase-α (G6Pase-α or G6PC). Using the G6pc-R83C mouse model of GSD-Ia, we explored a CRISPR/Cas9-based double-strand DNA oligonucleotide (dsODN) insertional strategy that uses the nonhomologous end-joining repair mechanism to correct the pathogenic p.R83C variant in G6pc exon-2. The strategy is based on the insertion of a short dsODN into G6pc exon-2 to disrupt the native exon and to introduce an additional splice acceptor site and the correcting sequence. When transcribed and spliced, the edited gene would generate a wild-type mRNA encoding the native G6Pase-α protein. The editing reagents formulated in lipid nanoparticles (LNPs) were delivered to the liver. Mice were treated either with one dose of LNP-dsODN at age 4 weeks or with two doses of LNP-dsODN at age 2 and 4 weeks. The G6pc-R83C mice receiving successful editing expressed ~4% of normal hepatic G6Pase-α activity, maintained glucose homeostasis, lacked hypoglycemic seizures, and displayed normalized blood metabolite profile. The outcomes are consistent with preclinical studies supporting previous gene augmentation therapy which is currently in clinical trials. This editing strategy may offer the basis for a therapeutic approach with an earlier clinical intervention than gene augmentation, with the additional benefit of a potentially permanent correction of the GSD-Ia phenotype., (© 2023 SSIEM. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.)
- Published
- 2023
- Full Text
- View/download PDF
179. Gene therapy and genome editing for type I glycogen storage diseases.
- Author
-
Chou JY and Mansfield BC
- Abstract
Type I glycogen storage diseases (GSD-I) consist of two major autosomal recessive disorders, GSD-Ia, caused by a reduction of glucose-6-phosphatase-α (G6Pase-α or G6PC) activity and GSD-Ib, caused by a reduction in the glucose-6-phosphate transporter (G6PT or SLC37A4) activity. The G6Pase-α and G6PT are functionally co-dependent. Together, the G6Pase-α/G6PT complex catalyzes the translocation of G6P from the cytoplasm into the endoplasmic reticulum lumen and its subsequent hydrolysis to glucose that is released into the blood to maintain euglycemia. Consequently, all GSD-I patients share a metabolic phenotype that includes a loss of glucose homeostasis and long-term risks of hepatocellular adenoma/carcinoma and renal disease. A rigorous dietary therapy has enabled GSD-I patients to maintain a normalized metabolic phenotype, but adherence is challenging. Moreover, dietary therapies do not address the underlying pathological processes, and long-term complications still occur in metabolically compensated patients. Animal models of GSD-Ia and GSD-Ib have delineated the disease biology and pathophysiology, and guided development of effective gene therapy strategies for both disorders. Preclinical studies of GSD-I have established that recombinant adeno-associated virus vector-mediated gene therapy for GSD-Ia and GSD-Ib are safe, and efficacious. A phase III clinical trial of rAAV-mediated gene augmentation therapy for GSD-Ia (NCT05139316) is in progress as of 2023. A phase I clinical trial of mRNA augmentation for GSD-Ia was initiated in 2022 (NCT05095727). Alternative genetic technologies for GSD-I therapies, such as gene editing, are also being examined for their potential to improve further long-term outcomes., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Chou and Mansfield.)
- Published
- 2023
- Full Text
- View/download PDF
180. Molecular mechanism underlying impaired hepatic autophagy in glycogen storage disease type Ib.
- Author
-
Gautam S, Zhang L, Lee C, Arnaoutova I, Chen HD, Resaz R, Eva A, Mansfield BC, and Chou JY
- Subjects
- Humans, Sirtuin 1, AMP-Activated Protein Kinases genetics, Autophagy genetics, Carcinoma, Hepatocellular etiology, Liver Neoplasms genetics, Liver Neoplasms complications, Glycogen Storage Disease Type I metabolism
- Abstract
Type Ib glycogen storage disease (GSD-Ib) is caused by a deficiency in the glucose-6-phosphate (G6P) transporter (G6PT) that translocates G6P from the cytoplasm into the endoplasmic reticulum lumen, where the intraluminal G6P is hydrolyzed to glucose by glucose-6-phosphatase-α (G6Pase-α). Clinically, GSD-Ib patients manifest a metabolic phenotype of impaired blood glucose homeostasis and a long-term risk of hepatocellular adenoma/carcinoma (HCA/HCC). Studies have shown that autophagy deficiency contributes to hepatocarcinogenesis. In this study, we show that G6PT deficiency leads to impaired hepatic autophagy evident from attenuated expression of many components of the autophagy network, decreased autophagosome formation and reduced autophagy flux. The G6PT-deficient liver displayed impaired sirtuin 1 (SIRT1) and AMP-activated protein kinase (AMPK) signaling, along with reduced expression of SIRT1, forkhead boxO3a (FoxO3a), liver kinase B-1 (LKB1) and the active p-AMPK. Importantly, we show that overexpression of either SIRT1 or LKB1 in G6PT-deficient liver restored autophagy and SIRT1/FoxO3a and LKB1/AMPK signaling. The hepatosteatosis in G6PT-deficient liver decreased SIRT1 expression. LKB1 overexpression reduced hepatic triglyceride levels, providing a potential link between LKB1/AMPK signaling upregulation and the increase in SIRT1 expression. In conclusion, downregulation of SIRT1/FoxO3a and LKB1/AMPK signaling underlies impaired hepatic autophagy which may contribute to HCA/HCC development in GSD-Ib. Understanding this mechanism may guide future therapies., (Published by Oxford University Press 2022.)
- Published
- 2023
- Full Text
- View/download PDF
181. Correction of metabolic abnormalities in a mouse model of glycogen storage disease type Ia by CRISPR/Cas9-based gene editing.
- Author
-
Arnaoutova I, Zhang L, Chen HD, Mansfield BC, and Chou JY
- Subjects
- Animals, CRISPR-Cas Systems genetics, Dependovirus genetics, Disease Models, Animal, Genetic Vectors genetics, Glucose genetics, Glucose metabolism, Glycogen Storage Disease Type I genetics, Glycogen Storage Disease Type I metabolism, Glycogen Storage Disease Type I pathology, Humans, Liver metabolism, Liver pathology, Mice, Gene Editing, Genetic Therapy, Glucose-6-Phosphatase genetics, Glycogen Storage Disease Type I therapy
- Abstract
Glycogen storage disease type Ia (GSD-Ia), deficient in glucose-6-phosphatase-α (G6PC), is characterized by impaired glucose homeostasis and a hallmark of fasting hypoglycemia. We have developed a recombinant adeno-associated virus (rAAV) vector-mediated gene therapy for GSD-Ia that is currently in a phase I/II clinical trial. While therapeutic expression of the episomal rAAV-G6PC clinical vector is stable in mice, the long-term durability of expression in humans is currently being established. Here we evaluated CRISPR/Cas9-based in vivo genome editing technology to correct a prevalent pathogenic human variant, G6PC-p.R83C. We have generated a homozygous G6pc-R83C mouse strain and shown that the G6pc-R83C mice manifest impaired glucose homeostasis and frequent hypoglycemic seizures, mimicking the pathophysiology of GSD-Ia patients. We then used a CRISPR/Cas9-based gene editing system to treat newborn G6pc-R83C mice and showed that the treated mice grew normally to age 16 weeks without hypoglycemia seizures. The treated G6pc-R83C mice, expressing ≥ 3% of normal hepatic G6Pase-α activity, maintained glucose homeostasis, displayed normalized blood metabolites, and could sustain 24 h of fasting. Taken together, we have developed a second-generation therapy in which in vivo correction of a pathogenic G6PC-p.R83C variant in its native genetic locus could lead to potentially permanent, durable, long-term correction of the GSD-Ia phenotype., (Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
182. Molecular biology and gene therapy for glycogen storage disease type Ib.
- Author
-
Chou JY, Cho JH, Kim GY, and Mansfield BC
- Subjects
- Animals, Antiporters genetics, Dependovirus genetics, Genetic Vectors, Homeostasis, Humans, Mice, Mice, Knockout, Monosaccharide Transport Proteins genetics, Mutation, Blood Glucose analysis, Genetic Therapy, Glycogen Storage Disease Type I genetics, Glycogen Storage Disease Type I therapy
- Abstract
Glycogen storage disease type Ib (GSD-Ib) is caused by a deficiency in the ubiquitously expressed glucose-6-phosphate (G6P) transporter (G6PT or SLC37A4). The primary function of G6PT is to translocate G6P from the cytoplasm into the lumen of the endoplasmic reticulum (ER). Inside the ER, G6P is hydrolyzed to glucose and phosphate by either the liver/kidney/intestine-restricted glucose-6-phosphatase-α (G6Pase-α) or the ubiquitously expressed G6Pase-β. A deficiency in G6Pase-α causes GSD type Ia (GSD-Ia) and a deficiency in G6Pase-β causes GSD-I-related syndrome (GSD-Irs). In gluconeogenic organs, functional coupling of G6PT and G6Pase-α is required to maintain interprandial blood glucose homeostasis. In myeloid tissues, functional coupling of G6PT and G6Pase-β is required to maintain neutrophil homeostasis. Accordingly, GSD-Ib is a metabolic and immune disorder, manifesting impaired glucose homeostasis, neutropenia, and neutrophil dysfunction. A G6pt knockout mouse model is being exploited to delineate the pathophysiology of GSD-Ib and develop new clinical treatment options, including gene therapy. The safety and efficacy of several G6PT-expressing recombinant adeno-associated virus pseudotype 2/8 vectors have been examined in murine GSD-Ib. The results demonstrate that the liver-directed gene transfer and expression safely corrects metabolic abnormalities and prevents hepatocellular adenoma (HCA) development. However, a second vector system may be required to correct myeloid and renal dysfunction in GSD-Ib. These findings are paving the way to a safe and efficacious gene therapy for entering clinical trials.
- Published
- 2018
- Full Text
- View/download PDF
183. Sirtuin signaling controls mitochondrial function in glycogen storage disease type Ia.
- Author
-
Cho JH, Kim GY, Mansfield BC, and Chou JY
- Abstract
Glycogen storage disease type Ia (GSD-Ia) deficient in glucose-6-phosphatase-α (G6Pase-α) is a metabolic disorder characterized by impaired glucose homeostasis and a long-term complication of hepatocellular adenoma/carcinoma (HCA/HCC). Mitochondrial dysfunction has been implicated in GSD-Ia but the underlying mechanism and its contribution to HCA/HCC development remain unclear. We have shown that hepatic G6Pase-α deficiency leads to downregulation of sirtuin 1 (SIRT1) signaling that underlies defective hepatic autophagy in GSD-Ia. SIRT1 is a NAD
+ -dependent deacetylase that can deacetylate and activate peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α), a master regulator of mitochondrial integrity, biogenesis, and function. We hypothesized that downregulation of hepatic SIRT1 signaling in G6Pase-α-deficient livers impairs PGC-1α activity, leading to mitochondrial dysfunction. Here we show that the G6Pase-α-deficient livers display defective PGC-1α signaling, reduced numbers of functional mitochondria, and impaired oxidative phosphorylation. Overexpression of hepatic SIRT1 restores PGC-1α activity, normalizes the expression of electron transport chain components, and increases mitochondrial complex IV activity. We have previously shown that restoration of hepatic G6Pase-α expression normalized SIRT1 signaling. We now show that restoration of hepatic G6Pase-α expression also restores PGC-1α activity and mitochondrial function. Finally, we show that HCA/HCC lesions found in G6Pase-α-deficient livers contain marked mitochondrial and oxidative DNA damage. Taken together, our study shows that downregulation of hepatic SIRT1/PGC-1α signaling underlies mitochondrial dysfunction and that oxidative DNA damage incurred by damaged mitochondria may contribute to HCA/HCC development in GSD-Ia.- Published
- 2018
- Full Text
- View/download PDF
184. Liver-directed gene therapy for murine glycogen storage disease type Ib.
- Author
-
Kwon JH, Lee YM, Cho JH, Kim GY, Anduaga J, Starost MF, Mansfield BC, and Chou JY
- Subjects
- Animals, Antiporters genetics, Antiporters metabolism, Disease Models, Animal, Genetic Vectors, Glucose-6-Phosphatase genetics, Glucose-6-Phosphatase metabolism, Glucose-6-Phosphate genetics, Glucose-6-Phosphate metabolism, Glycogen Storage Disease Type I metabolism, Homeostasis, Humans, Insulin Resistance, Liver metabolism, Mice, Mice, Transgenic, Monosaccharide Transport Proteins genetics, Monosaccharide Transport Proteins metabolism, Promoter Regions, Genetic, Genetic Therapy methods, Glycogen Storage Disease Type I genetics, Glycogen Storage Disease Type I therapy
- Abstract
Glycogen storage disease type-Ib (GSD-Ib), deficient in the glucose-6-phosphate transporter (G6PT), is characterized by impaired glucose homeostasis, myeloid dysfunction, and long-term risk of hepatocellular adenoma (HCA). We examined the efficacy of G6PT gene therapy in G6pt-/- mice using recombinant adeno-associated virus (rAAV) vectors, directed by either the G6PC or the G6PT promoter/enhancer. Both vectors corrected hepatic G6PT deficiency in murine GSD-Ib but the G6PC promoter/enhancer was more efficacious. Over a 78-week study, using dose titration of the rAAV vectors, we showed that G6pt-/- mice expressing 3-62% of normal hepatic G6PT activity exhibited a normalized liver phenotype. Two of the 12 mice expressing < 6% of normal hepatic G6PT activity developed HCA. All treated mice were leaner and more sensitive to insulin than wild-type mice. Mice expressing 3-22% of normal hepatic G6PT activity exhibited higher insulin sensitivity than mice expressing 44-62%. The levels of insulin sensitivity correlated with the magnitudes of hepatic carbohydrate response element binding protein signaling activation. In summary, we established the threshold of hepatic G6PT activity required to prevent tumor formation and showed that mice expressing 3-62% of normal hepatic G6PT activity maintained glucose homeostasis and were protected against age-related obesity and insulin resistance., (Published by Oxford University Press 2017. This work is written by US Government employees and is in the public domain in the US.)
- Published
- 2017
- Full Text
- View/download PDF
185. Recent development and gene therapy for glycogen storage disease type Ia.
- Author
-
Chou JY, Kim GY, and Cho JH
- Abstract
Glycogen storage disease type Ia (GSD-Ia) is an autosomal recessive metabolic disorder caused by a deficiency in glucose-6-phosphatase-α (G6Pase-α or G6PC) that is expressed primarily in the liver, kidney, and intestine. G6Pase-α catalyzes the hydrolysis of glucose-6-phosphate (G6P) to glucose and phosphate in the terminal step of gluconeogenesis and glycogenolysis, and is a key enzyme for endogenous glucose production. The active site of G6Pase-α is inside the endoplasmic reticulum (ER) lumen. For catalysis, the substrate G6P must be translocated from the cytoplasm into the ER lumen by a G6P transporter (G6PT). The functional coupling of G6Pase-α and G6PT maintains interprandial glucose homeostasis. Dietary therapies for GSD-Ia are available, but cannot prevent the long-term complication of hepatocellular adenoma that may undergo malignant transformation to hepatocellular carcinoma. Animal models of GSD-Ia are now available and are being exploited to both delineate the disease more precisely and develop new treatment approaches, including gene therapy., Competing Interests: Conflict of interest The authors declare that they have no conflict of interest.
- Published
- 2017
- Full Text
- View/download PDF
186. Mice expressing reduced levels of hepatic glucose-6-phosphatase-α activity do not develop age-related insulin resistance or obesity.
- Author
-
Kim GY, Lee YM, Cho JH, Pan CJ, Jun HS, Springer DA, Mansfield BC, and Chou JY
- Subjects
- AMP-Activated Protein Kinases metabolism, Animals, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors, Dependovirus genetics, Disease Models, Animal, Energy Metabolism genetics, Genetic Therapy, Genetic Vectors administration & dosage, Genetic Vectors genetics, Glycogen Storage Disease Type I genetics, Glycogen Storage Disease Type I metabolism, Liver metabolism, Mice, Mice, Knockout, NAD metabolism, Nuclear Proteins metabolism, Obesity metabolism, Signal Transduction, Sirtuin 1 metabolism, Transcription Factors metabolism, Gene Expression, Glucose-6-Phosphatase genetics, Insulin Resistance genetics, Obesity genetics
- Abstract
Glycogen storage disease type-Ia (GSD-Ia) is caused by a lack of glucose-6-phosphatase-α (G6Pase-α or G6PC) activity. We have shown that gene therapy mediated by a recombinant adeno-associated virus (rAAV) vector expressing human G6Pase-α normalizes blood glucose homeostasis in the global G6pc knockout (G6pc(-/-)) mice for 70-90 weeks. The treated G6pc(-/-) mice expressing 3-63% of normal hepatic G6Pase-α activity (AAV mice) produce endogenous hepatic glucose levels 61-68% of wild-type littermates, have a leaner phenotype and exhibit fasting blood insulin levels more typical of young adult mice. We now show that unlike wild-type mice, the lean AAV mice have increased caloric intake and do not develop age-related obesity or insulin resistance. Pathway analysis shows that signaling by hepatic carbohydrate response element binding protein that improves glucose tolerance and insulin signaling is activated in AAV mice. In addition, several longevity factors in the calorie restriction pathway, including the NADH shuttle systems, NAD(+) concentrations and the AMP-activated protein kinase/sirtuin 1/peroxisome proliferator-activated receptor-γ coactivator 1α pathway are upregulated in the livers of AAV mice. The finding that partial restoration of hepatic G6Pase-α activity in GSD-Ia mice not only attenuates the phenotype of hepatic G6Pase-α deficiency but also prevents the development of age-related obesity and insulin resistance seen in wild-type mice may suggest relevance of the G6Pase-α enzyme to obesity and diabetes., (Published by Oxford University Press 2015. This work is written by (a) US Government employee(s) and is in the public domain in the US.)
- Published
- 2015
- Full Text
- View/download PDF
187. Type I glycogen storage diseases: disorders of the glucose-6-phosphatase/glucose-6-phosphate transporter complexes.
- Author
-
Chou JY, Jun HS, and Mansfield BC
- Subjects
- Animals, Dogs, Glycogen Storage Disease Type I therapy, Homeostasis, Humans, Mice, Models, Biological, Phenotype, Antiporters genetics, Genetic Therapy methods, Glucose-6-Phosphatase genetics, Glycogen Storage Disease Type I genetics, Granulocyte Colony-Stimulating Factor therapeutic use, Monosaccharide Transport Proteins genetics
- Abstract
Disorders of the glucose-6-phosphatase (G6Pase)/glucose-6-phosphate transporter (G6PT) complexes consist of three subtypes: glycogen storage disease type Ia (GSD-Ia), deficient in the liver/kidney/intestine-restricted G6Pase-α (or G6PC); GSD-Ib, deficient in a ubiquitously expressed G6PT (or SLC37A4); and G6Pase-β deficiency or severe congenital neutropenia syndrome type 4 (SCN4), deficient in the ubiquitously expressed G6Pase-β (or G6PC3). G6Pase-α and G6Pase-β are glucose-6-phosphate (G6P) hydrolases with active sites lying inside the endoplasmic reticulum (ER) lumen and as such are dependent upon the G6PT to translocate G6P from the cytoplasm into the lumen. The tissue expression profiles of the G6Pase enzymes dictate the disease's phenotype. A functional G6Pase-α/G6PT complex maintains interprandial glucose homeostasis, while a functional G6Pase-β/G6PT complex maintains neutrophil/macrophage energy homeostasis and functionality. G6Pase-β deficiency is not a glycogen storage disease but biochemically it is a GSD-I related syndrome (GSD-Irs). GSD-Ia and GSD-Ib patients manifest a common metabolic phenotype of impaired blood glucose homeostasis not shared by GSD-Irs. GSD-Ib and GSD-Irs patients manifest a common myeloid phenotype of neutropenia and neutrophil/macrophage dysfunction not shared by GSD-Ia. While a disruption of the activity of the G6Pase-α/G6PT complex readily explains why GSD-Ia and GSD-Ib patients exhibit impaired glucose homeostasis, the basis for neutropenia and myeloid dysfunction in GSD-Ib and GSD-Irs are only now starting to be understood. Animal models of all three disorders are now available and are being exploited to both delineate the disease more precisely and develop new treatment approaches, including gene therapy.
- Published
- 2015
- Full Text
- View/download PDF
188. The SLC37 family of sugar-phosphate/phosphate exchangers.
- Author
-
Chou JY and Mansfield BC
- Subjects
- Amino Acid Sequence, Animals, Cell Membrane metabolism, Humans, Membrane Transport Proteins chemistry, Molecular Sequence Data, Structure-Activity Relationship, Carbohydrate Metabolism, Membrane Transport Proteins metabolism, Phosphates metabolism
- Abstract
The SLC37 family members are endoplasmic reticulum (ER)-associated sugar-phosphate/phosphate (P(i)) exchangers. Three of the four members, SLC37A1, SLC37A2, and SLC37A4, function as Pi-linked glucose-6-phosphate (G6P) antiporters catalyzing G6P:P(i) and P(i):P(i) exchanges. The activity of SLC37A3 is unknown. SLC37A4, better known as the G6P transporter (G6PT), has been extensively characterized, functionally and structurally, and is the best characterized family member. G6PT contains 10 transmembrane helices with both N and C termini facing the cytoplasm. The primary in vivo function of the G6PT protein is to translocate G6P from the cytoplasm into the ER lumen where it couples with either the liver/kidney/intestine-restricted glucose-6-phosphatase-α (G6Pase-α or G6PC) or the ubiquitously expressed G6Pase-β (or G6PC3) to hydrolyze G6P to glucose and P(i). The G6PT/G6Pase-α complex maintains interprandial glucose homeostasis, and the G6PT/G6Pase-β complex maintains neutrophil energy homeostasis and functionality. G6PT is highly selective for G6P and is competitively inhibited by cholorogenic acid and its derivatives. Neither SLC37A1 nor SLC37A2 can couple functionally with G6Pase-α or G6Pase-β, and the antiporter activities of SLC37A1 or SLC37A2 are not inhibited by cholorogenic acid. Deficiencies in G6PT cause glycogen storage disease type Ib (GSD-Ib), a metabolic and immune disorder. To date, 91 separate SLC37A4 mutations, including 39 missense mutations, have been identified in GSD-Ib patients. Characterization of missense mutations has yielded valuable information on functionally important residues in the G6PT protein. The biological roles of the other SLC37 proteins remain to be determined and deficiencies have not yet been correlated to diseases., (© 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
189. Prevention of hepatocellular adenoma and correction of metabolic abnormalities in murine glycogen storage disease type Ia by gene therapy.
- Author
-
Lee YM, Jun HS, Pan CJ, Lin SR, Wilson LH, Mansfield BC, and Chou JY
- Subjects
- Animals, Antiporters genetics, Antiporters metabolism, Blood Glucose, Body Mass Index, Body Weight, Dependovirus genetics, Disease Models, Animal, Female, Genetic Vectors, Glucose Tolerance Test, Glucose-6-Phosphatase metabolism, Glycogen Storage Disease Type I genetics, Homeostasis, Insulin blood, Liver enzymology, Liver pathology, Male, Mice, Mice, Knockout, Monosaccharide Transport Proteins genetics, Monosaccharide Transport Proteins metabolism, Promoter Regions, Genetic, RNA, Messenger metabolism, Adenoma prevention & control, Genetic Therapy adverse effects, Glucose-6-Phosphatase genetics, Glycogen Storage Disease Type I enzymology, Glycogen Storage Disease Type I therapy, Liver metabolism, Liver Neoplasms prevention & control
- Abstract
Unlabelled: Glycogen storage disease type Ia (GSD-Ia), which is characterized by impaired glucose homeostasis and chronic risk of hepatocellular adenoma (HCA), is caused by deficiencies in the endoplasmic reticulum (ER)-associated glucose-6-phosphatase-α (G6Pase-α or G6PC) that hydrolyzes glucose-6-phosphate (G6P) to glucose. G6Pase-α activity depends on the G6P transporter (G6PT) that translocates G6P from the cytoplasm into the ER lumen. The functional coupling of G6Pase-α and G6PT maintains interprandial glucose homeostasis. We have shown previously that gene therapy mediated by AAV-GPE, an adeno-associated virus (AAV) vector expressing G6Pase-α directed by the human G6PC promoter/enhancer (GPE), completely normalizes hepatic G6Pase-α deficiency in GSD-Ia (G6pc(-/-) ) mice for at least 24 weeks. However, a recent study showed that within 78 weeks of gene deletion, all mice lacking G6Pase-α in the liver develop HCA. We now show that gene therapy mediated by AAV-GPE maintains efficacy for at least 70-90 weeks for mice expressing more than 3% of wild-type hepatic G6Pase-α activity. The treated mice displayed normal hepatic fat storage, had normal blood metabolite and glucose tolerance profiles, had reduced fasting blood insulin levels, maintained normoglycemia over a 24-hour fast, and had no evidence of hepatic abnormalities. After a 24-hour fast, hepatic G6PT messenger RNA levels in G6pc(-/-) mice receiving gene therapy were markedly increased. Because G6PT transport is the rate-limiting step in microsomal G6P metabolism, this may explain why the treated G6pc(-/-) mice could sustain prolonged fasts. The low fasting blood insulin levels and lack of hepatic steatosis may explain the absence of HCA., Conclusion: These results confirm that AAV-GPE-mediated gene transfer corrects hepatic G6Pase-α deficiency in murine GSD-Ia and prevents chronic HCA formation., (Copyright © 2012 American Association for the Study of Liver Diseases.)
- Published
- 2012
- Full Text
- View/download PDF
190. Glucose-6-phosphatase-β, implicated in a congenital neutropenia syndrome, is essential for macrophage energy homeostasis and functionality.
- Author
-
Jun HS, Cheung YY, Lee YM, Mansfield BC, and Chou JY
- Subjects
- Animals, Apoptosis, Blotting, Western, Calcium metabolism, Cell Proliferation, Chemotaxis, Congenital Bone Marrow Failure Syndromes, Cytokines metabolism, Female, Glucose metabolism, Glucose Transporter Type 1 genetics, Glucose Transporter Type 1 metabolism, Glucose Transporter Type 3 genetics, Glucose Transporter Type 3 metabolism, Immunoenzyme Techniques, Inflammation genetics, Inflammation metabolism, Macrophages cytology, Mice, Mice, Inbred C57BL, Mice, Knockout, NADPH Oxidases genetics, NADPH Oxidases metabolism, Neutropenia genetics, Neutropenia metabolism, Neutropenia pathology, Phagocytosis, Pregnancy, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Respiratory Burst, Signal Transduction, Syndrome, Glucose-6-Phosphatase physiology, Glucose-6-Phosphate metabolism, Homeostasis physiology, Inflammation pathology, Macrophages physiology, Neutropenia congenital
- Abstract
Glucose-6-phosphatase-β (G6Pase-β or G6PC3) deficiency, also known as severe congenital neutropenia syndrome 4, is characterized not only by neutropenia but also by impaired neutrophil energy homeostasis and functionality. We now show the syndrome is also associated with macrophage dysfunction, with murine G6pc3(-/-) macrophages having impairments in their respiratory burst, chemotaxis, calcium flux, and phagocytic activities. Consistent with a glucose-6-phosphate (G6P) metabolism deficiency, G6pc3(-/-) macrophages also have a lower glucose uptake and lower levels of G6P, lactate, and ATP than wild-type macrophages. Furthermore, the expression of NADPH oxidase subunits and membrane translocation of p47(phox) are down-regulated, and G6pc3(-/-) macrophages exhibit repressed trafficking in vivo both during an inflammatory response and in pregnancy. During pregnancy, the absence of G6Pase-β activity also leads to impaired energy homeostasis in the uterus and reduced fertility of G6pc3(-/-) mothers. Together these results show that immune deficiencies in this congenital neutropenia syndrome extend beyond neutrophil dysfunction.
- Published
- 2012
- Full Text
- View/download PDF
191. G-CSF improves murine G6PC3-deficient neutrophil function by modulating apoptosis and energy homeostasis.
- Author
-
Jun HS, Lee YM, Song KD, Mansfield BC, and Chou JY
- Subjects
- Animals, Apoptosis genetics, Cells, Cultured, Drug Evaluation, Preclinical, Energy Metabolism genetics, Glucose-6-Phosphatase physiology, Homeostasis drug effects, Homeostasis genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Neutrophils metabolism, Neutrophils physiology, Phosphatidylinositol Phosphates metabolism, Protein Subunits, Proto-Oncogene Proteins c-akt metabolism, Proto-Oncogene Proteins c-akt physiology, Signal Transduction drug effects, Signal Transduction genetics, eIF-2 Kinase metabolism, eIF-2 Kinase physiology, Apoptosis drug effects, Energy Metabolism drug effects, Glucose-6-Phosphatase genetics, Granulocyte Colony-Stimulating Factor pharmacology, Neutrophils drug effects
- Abstract
G6PC3 (or glucose-6-phosphatase-β) deficiency underlies a congenital neutropenia syndrome in which neutrophils exhibit enhanced endoplasmic reticulum (ER) stress, increased apoptosis, impaired energy homeostasis, and impaired functionality. Here we show that murine G6pc3(-/-) neutrophils undergoing ER stress activate protein kinase-like ER kinase and phosphatidylinositol 3,4,5-trisphosphate/Akt signaling pathways, and that neutrophil apoptosis is mediated in part by the intrinsic mitochondrial pathway. In G6PC3-deficient patients, granulocyte colony-stimulating factor (G-CSF) improves neutropenia, but its impact on neutrophil apoptosis and dysfunction is unknown. We now show that G-CSF delays neutrophil apoptosis in vitro by modulating apoptotic mediators. However, G6pc3(-/-) neutrophils in culture exhibit accelerated apoptosis compared with wild-type neutrophils both in the presence or absence of G-CSF. Limiting glucose (0.6mM) accelerates apoptosis but is more pronounced for wild-type neutrophils, leading to similar survival profiles for both neutrophil populations. In vivo G-CSF therapy completely corrects neutropenia and normalizes levels of p-Akt, phosphatidylinositol 3,4,5-trisphosphate, and active caspase-3. Neutrophils from in vivo G-CSF-treated G6pc3(-/-) mice exhibit increased glucose uptake and elevated intracellular levels of G6P, lactate, and adenosine-5'-triphosphate, leading to improved functionality. Together, the results strongly suggest that G-CSF improves G6pc3(-/-) neutrophil survival by modulating apoptotic mediators and rectifies function by enhancing energy homeostasis.
- Published
- 2011
- Full Text
- View/download PDF
192. Severe congenital neutropenia resulting from G6PC3 deficiency with increased neutrophil CXCR4 expression and myelokathexis.
- Author
-
McDermott DH, De Ravin SS, Jun HS, Liu Q, Priel DA, Noel P, Takemoto CM, Ojode T, Paul SM, Dunsmore KP, Hilligoss D, Marquesen M, Ulrick J, Kuhns DB, Chou JY, Malech HL, and Murphy PM
- Subjects
- Adolescent, Animals, Child, Female, Gene Expression, Homozygote, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mutation, Missense, Neutropenia enzymology, Neutrophils metabolism, Syndrome, Glucose-6-Phosphatase genetics, Glycogen Storage Disease Type I genetics, Neutropenia congenital, Neutropenia genetics, Receptors, CXCR4 genetics
- Abstract
Mutations in more than 15 genes are now known to cause severe congenital neutropenia (SCN); however, the pathologic mechanisms of most genetic defects are not fully defined. Deficiency of G6PC3, a glucose-6-phosphatase, causes a rare multisystem syndrome with SCN first described in 2009. We identified a family with 2 children with homozygous G6PC3 G260R mutations, a loss of enzymatic function, and typical syndrome features with the exception that their bone marrow biopsy pathology revealed abundant neutrophils consistent with myelokathexis. This pathologic finding is a hallmark of another type of SCN, WHIM syndrome, which is caused by gain-of-function mutations in CXCR4, a chemokine receptor and known neutrophil bone marrow retention factor. We found markedly increased CXCR4 expression on neutrophils from both our G6PC3-deficient patients and G6pc3(-/-) mice. In both patients, granulocyte colony-stimulating factor treatment normalized CXCR4 expression and neutrophil counts. In G6pc3(-/-) mice, the specific CXCR4 antagonist AMD3100 rapidly reversed neutropenia. Thus, myelokathexis associated with abnormally high neutrophil CXCR4 expression may contribute to neutropenia in G6PC3 deficiency and responds well to granulocyte colony-stimulating factor.
- Published
- 2010
- Full Text
- View/download PDF
193. Lack of glucose recycling between endoplasmic reticulum and cytoplasm underlies cellular dysfunction in glucose-6-phosphatase-beta-deficient neutrophils in a congenital neutropenia syndrome.
- Author
-
Jun HS, Lee YM, Cheung YY, McDermott DH, Murphy PM, De Ravin SS, Mansfield BC, and Chou JY
- Subjects
- Adenosine Triphosphate metabolism, Adolescent, Animals, Annexin A5 metabolism, Apoptosis, Caspase 3 metabolism, Child, Cytoplasm metabolism, Endoplasmic Reticulum metabolism, Female, Glucose Transporter Type 1 metabolism, Glucose-6-Phosphatase genetics, Glycogen Storage Disease Type I genetics, Glycogen Storage Disease Type I pathology, Humans, Lactic Acid metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, NADPH Oxidases metabolism, Neutropenia genetics, Neutropenia pathology, Neutrophils pathology, Stress, Physiological, Syndrome, Glucose metabolism, Glucose-6-Phosphatase metabolism, Glycogen Storage Disease Type I metabolism, Neutropenia congenital, Neutropenia metabolism, Neutrophils metabolism
- Abstract
G6PC3 deficiency, characterized by neutropenia and neutrophil dysfunction, is caused by deficiencies in the endoplasmic reticulum (ER) enzyme glucose-6-phosphatase-β (G6Pase-β or G6PC3) that converts glucose-6-phosphate (G6P) into glucose, the primary energy source of neutrophils. Enhanced neutrophil ER stress and apoptosis underlie neutropenia in G6PC3 deficiency, but the exact functional role of G6Pase-β in neutrophils remains unknown. We hypothesized that the ER recycles G6Pase-β-generated glucose to the cytoplasm, thus regulating the amount of available cytoplasmic glucose/G6P in neutrophils. Accordingly, a G6Pase-β deficiency would impair glycolysis and hexose monophosphate shunt activities leading to reductions in lactate production, adenosine-5'-triphosphate (ATP) production, and reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity. Using annexin V-depleted neutrophils, we show that glucose transporter-1 translocation is impaired in neutrophils from G6pc3(-/-) mice and G6PC3-deficient patients along with impaired glucose uptake in G6pc3(-/-) neutrophils. Moreover, levels of G6P, lactate, and ATP are markedly lower in murine and human G6PC3-deficient neutrophils, compared with their respective controls. In parallel, the expression of NADPH oxidase subunits and membrane translocation of p47(phox) are down-regulated in murine and human G6PC3-deficient neutrophils. The results establish that in nonapoptotic neutrophils, G6Pase-β is essential for normal energy homeostasis. A G6Pase-β deficiency prevents recycling of ER glucose to the cytoplasm, leading to neutrophil dysfunction.
- Published
- 2010
- Full Text
- View/download PDF
194. Complete normalization of hepatic G6PC deficiency in murine glycogen storage disease type Ia using gene therapy.
- Author
-
Yiu WH, Lee YM, Peng WT, Pan CJ, Mead PA, Mansfield BC, and Chou JY
- Subjects
- Animals, Dependovirus genetics, Genetic Vectors, Glucosephosphate Dehydrogenase genetics, Liver enzymology, Mice, Mice, Knockout, Genetic Therapy, Glycogen Storage Disease Type I therapy, Liver metabolism
- Abstract
Glycogen storage disease type Ia (GSD-Ia) patients deficient in glucose-6-phosphatase-alpha (G6Pase-alpha or G6PC) manifest disturbed glucose homeostasis. We examined the efficacy of liver G6Pase-alpha delivery mediated by AAV-GPE, an adeno-associated virus (AAV) serotype 8 vector expressing human G6Pase-alpha directed by the human G6PC promoter/enhancer (GPE), and compared it to AAV-CBA, that directed murine G6Pase-alpha expression using a hybrid chicken beta-actin (CBA) promoter/cytomegalovirus (CMV) enhancer. The AAV-GPE directed hepatic G6Pase-alpha expression in the infused G6pc(-/-) mice declined 12-fold from age 2 to 6 weeks but stabilized at wild-type levels from age 6 to 24 weeks. In contrast, the expression directed by AAV-CBA declined 95-fold over 24 weeks, demonstrating that the GPE is more effective in directing persistent in vivo hepatic transgene expression. We further show that the rapid decline in transgene expression directed by AAV-CBA results from an inflammatory immune response elicited by the AAV-CBA vector. The AAV-GPE-treated G6pc(-/-) mice exhibit normal levels of blood glucose, blood metabolites, hepatic glycogen, and hepatic fat. Moreover, the mice maintained normal blood glucose levels even after 6 hours of fasting. The complete normalization of hepatic G6Pase-alpha deficiency by the G6PC promoter/enhancer holds promise for the future of gene therapy in human GSD-Ia patients.
- Published
- 2010
- Full Text
- View/download PDF
195. Oxidative stress mediates nephropathy in type Ia glycogen storage disease.
- Author
-
Yiu WH, Mead PA, Jun HS, Mansfield BC, and Chou JY
- Subjects
- Animals, Antioxidants pharmacology, Catalase metabolism, Cyclic N-Oxides pharmacology, Disease Models, Animal, Down-Regulation, Kidney Diseases drug therapy, Mice, NADPH Oxidase 2, Oxidative Stress drug effects, Signal Transduction, Spin Labels, Superoxide Dismutase metabolism, Up-Regulation, Cytochrome b Group metabolism, Glycogen Storage Disease Type I physiopathology, Kidney Diseases physiopathology, Membrane Glycoproteins metabolism, NADPH Oxidases metabolism, Oxidative Stress physiology
- Abstract
Glycogen storage disease type Ia (GSD-Ia) patients, deficient in glucose-6-phosphatase-alpha, manifest disturbed glucose homeostasis with long-term renal disease. We have previously shown that renal fibrosis in GSD-Ia is mediated by the angiotensin/transforming growth factor-beta1 (TGF-beta1) pathway, which also elicits renal damage through oxidative stress. In this study, we further elucidate the mechanism of renal disease by showing that renal expression of Nox-2, p22(phox), and p47(phox), components of NADPH oxidase, are upregulated in GSD-Ia mice compared with controls. Akt/protein kinase B, a downstream mediator of angiotensin II and TGF-beta1, is also activated, leading to phosphorylation and inactivation of the Forkhead box O family of transcription factors. This in turn triggers downregulation of superoxide dismutase and catalase (CAT) activities that have essential roles in oxidative detoxification in mammals. Renal oxidative stress in GSD-Ia mice is shown by increased oxidation of dihydroethidium and by oxidative damage of DNA. Importantly, renal dysfunction, reflected by elevated serum levels of blood urea nitrogen, reduced renal CAT activity, and increased renal fibrosis, is improved in GSD-Ia mice treated with the antioxidant drug tempol. These data provide the first evidence that oxidative stress is one mechanism that underlies GSD-Ia nephropathy.
- Published
- 2010
- Full Text
- View/download PDF
196. Mutations in the glucose-6-phosphatase-alpha (G6PC) gene that cause type Ia glycogen storage disease.
- Author
-
Chou JY and Mansfield BC
- Subjects
- Glycogen Storage Disease Type I etiology, Humans, Mutation, Phenotype, Glucose-6-Phosphatase genetics, Glycogen Storage Disease Type I genetics
- Abstract
Glucose-6-phosphatase-alpha (G6PC) is a key enzyme in glucose homeostasis that catalyzes the hydrolysis of glucose-6-phosphate to glucose and phosphate in the terminal step of gluconeogenesis and glycogenolysis. Mutations in the G6PC gene, located on chromosome 17q21, result in glycogen storage disease type Ia (GSD-Ia), an autosomal recessive metabolic disorder. GSD-Ia patients manifest a disturbed glucose homeostasis, characterized by fasting hypoglycemia, hepatomegaly, nephromegaly, hyperlipidemia, hyperuricemia, lactic acidemia, and growth retardation. G6PC is a highly hydrophobic glycoprotein, anchored in the membrane of the endoplasmic reticulum with the active center facing into the lumen. To date, 54 missense, 10 nonsense, 17 insertion/deletion, and three splicing mutations in the G6PC gene have been identified in more than 550 patients. Of these, 50 missense, two nonsense, and two insertion/deletion mutations have been functionally characterized for their effects on enzymatic activity and stability. While GSD-Ia is not more prevalent in any ethnic group, mutations unique to Caucasian, Oriental, and Jewish populations have been described. Despite this, GSD-Ia patients exhibit phenotypic heterogeneity and a stringent genotype-phenotype relationship does not exist.
- Published
- 2008
- Full Text
- View/download PDF
197. Neutrophil stress and apoptosis underlie myeloid dysfunction in glycogen storage disease type Ib.
- Author
-
Kim SY, Jun HS, Mead PA, Mansfield BC, and Chou JY
- Subjects
- Animals, Antiporters genetics, Antiporters metabolism, Caspase 9 metabolism, Endoplasmic Reticulum metabolism, Endoplasmic Reticulum pathology, Glucose metabolism, Glucose-6-Phosphate metabolism, Glycogen Storage Disease Type I pathology, Mice, Mice, Mutant Strains, Mitochondria metabolism, Monosaccharide Transport Proteins genetics, Monosaccharide Transport Proteins metabolism, Reactive Oxygen Species metabolism, bcl-2-Associated X Protein metabolism, Apoptosis physiology, Glycogen Storage Disease Type I metabolism, Neutrophils metabolism, Neutrophils pathology, Oxidative Stress physiology
- Abstract
Glycogen storage disease type Ib (GSD-Ib) is caused by a deficiency in the glucose-6-phosphate (G6P) transporter (G6PT) that works with a liver/kidney/intestine-restricted glucose-6-phosphatase-alpha (G6Pase-alpha) to maintain glucose homeostasis between meals. Clinically, GSD-Ib patients manifest disturbed glucose homeostasis and neutrophil dysfunctions but the cause of the latter is unclear. Neutrophils express the ubiquitously expressed G6PT and G6Pase-beta that together transport G6P into the endoplasmic reticulum (ER) lumen and hydrolyze it to glucose. Because we expected G6PT-deficient neutrophils to be unable to produce endogenous glucose, we hypothesized this would lead to ER stress and increased apoptosis. Using GSD-Ib mice, we showed that GSD-Ib neutrophils exhibited increased production of ER chaperones and oxidative stress, consistent with ER stress, increased annexin V binding and caspase-3 activation, consistent with an increased rate of apoptosis. Bax activation, mitochondrial release of proapoptotic effectors, and caspase-9 activation demonstrated the involvement of the intrinsic mitochondrial pathway in these processes. The results demonstrate that G6P translocation and hydrolysis are required for normal neutrophil functions and support the hypothesis that neutrophil dysfunction in GSD-Ib is due, at least in part, to ER stress and increased apoptosis.
- Published
- 2008
- Full Text
- View/download PDF
198. Gene therapy for type I glycogen storage diseases.
- Author
-
Chou JY and Mansfield BC
- Subjects
- Adenoviridae genetics, Genetic Vectors, Glucose-6-Phosphatase genetics, Glucose-6-Phosphate deficiency, Humans, Genetic Therapy methods, Glycogen Storage Disease Type I therapy
- Abstract
The type I glycogen storage diseases (GSD-I) are a group of related diseases caused by a deficiency in the glucose-6-phosphatase-alpha (G6Pase-alpha) system, a key enzyme complex that is essential for the maintenance of blood glucose homeostasis between meals. The complex consists of a glucose-6-phosphate transporter (G6PT) that translocates glucose-6-phosphate from the cytoplasm into the lumen of the endoplasmic reticulum, and a G6Pase-alpha catalytic unit that hydrolyses the glucose-6-phosphate into glucose and phosphate. A deficiency in G6Pase-alpha causes GSD type Ia (GSD-Ia) and a deficiency in G6PT causes GSD type Ib (GSD-Ib). Both GSD-Ia and GSD-Ib patients manifest a disturbed glucose homeostasis, while GSD-Ib patients also suffer symptoms of neutropenia and myeloid dysfunctions. G6Pase-alpha and G6PT are both hydrophobic endoplasmic reticulum-associated transmembrane proteins that can not expressed in soluble active forms. Therefore protein replacement therapy of GSD-I is not an option. Animal models of GSD-Ia and GSD-Ib that mimic the human disorders are available. Both adenovirus- and adeno-associated virus (AAV)-mediated gene therapies have been evaluated for GSD-Ia in these model systems. While adenoviral therapy produces only short term corrections and only impacts liver expression of the gene, AAV-mediated therapy delivers the transgene to both the liver and kidney, achieving longer term correction of the GSD-Ia disorder, although there are substantial differences in efficacy depending on the AAV serotype used. Gene therapy for GSD-Ib in the animal model is still in its infancy, although an adenoviral construct has improved the metabolic profile and myeloid function. Taken together further refinements in gene therapy may hold long term benefits for the treatment of type I GSD disorders.
- Published
- 2007
- Full Text
- View/download PDF
199. Impaired neutrophil activity and increased susceptibility to bacterial infection in mice lacking glucose-6-phosphatase-beta.
- Author
-
Cheung YY, Kim SY, Yiu WH, Pan CJ, Jun HS, Ruef RA, Lee EJ, Westphal H, Mansfield BC, and Chou JY
- Subjects
- Animals, Bacterial Infections enzymology, Bacterial Infections immunology, Blood Glucose analysis, Disease Models, Animal, Genetic Predisposition to Disease, Glucose-6-Phosphatase analysis, Glucose-6-Phosphatase metabolism, Glucose-6-Phosphate metabolism, Hematopoiesis genetics, Homeostasis, Mice, Neutropenia enzymology, Neutrophils enzymology, Peritonitis enzymology, Peritonitis microbiology, Protein Subunits analysis, Protein Subunits metabolism, Bacterial Infections genetics, Glucose-6-Phosphatase genetics, Neutropenia genetics, Neutrophils immunology, Peritonitis genetics, Protein Subunits genetics
- Abstract
Neutropenia and neutrophil dysfunction are common in many diseases, although their etiology is often unclear. Previous views held that there was a single ER enzyme, glucose-6-phosphatase-alpha (G6Pase-alpha), whose activity--limited to the liver, kidney, and intestine--was solely responsible for the final stages of gluconeogenesis and glycogenolysis, in which glucose-6-phosphate (G6P) is hydrolyzed to glucose for release to the blood. Recently, we characterized a second G6Pase activity, that of G6Pase-beta (also known as G6PC), which is also capable of hydrolyzing G6P to glucose but is ubiquitously expressed and not implicated in interprandial blood glucose homeostasis. We now report that the absence of G6Pase-beta led to neutropenia; defects in neutrophil respiratory burst, chemotaxis, and calcium flux; and increased susceptibility to bacterial infection. Consistent with this, G6Pase-beta-deficient (G6pc3-/-) mice with experimental peritonitis exhibited increased expression of the glucose-regulated proteins upregulated during ER stress in their neutrophils and bone marrow, and the G6pc3-/- neutrophils exhibited an enhanced rate of apoptosis. Our results define a molecular pathway to neutropenia and neutrophil dysfunction of previously unknown etiology, providing a potential model for the treatment of these conditions.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.