Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Gene ; 819: 146263, 2022 Apr 20.
Article in English | MEDLINE | ID: mdl-35121025

ABSTRACT

High temperature requirement A2 (HtrA2) contributes to regulating mitochondrial quality control and maintaining the balance between the death and survival of cells and living organisms. However, the molecular mechanism of HtrA2 in physiological and pathophysiological processes remains unclear. HtrA2 exhibits multifaceted characteristics according to the expression levels and acts opposite functions depending on its subcellular localization. Thus, innovative technologies and systems that can be freely manipulated at the quantitative, biochemical, molecular and cellular levels are needed to address not only the challenges faced by HtrA2 research but also the general obstacles to protein research. Here, we are the first to identify zebrafish HtrA2 (zHtrA2) as the true ortholog of human HtrA2 (hHtrA2), by in silico sequence analysis of genomic DNA and molecular biological techniques, which is highly conserved structurally and functionally as a serine protease and cell death regulator. The zHtrA2 protein is primarily localized in the mitochondria, where alanine-exposed mature zHtrA2 ((A)-zHtrA2) is generated by removing 111 residues at the N-terminus of pro-zHtrA2. The (A)-zHtrA2 released from the mitochondria into the cytosol induces the caspase cascade by binding to and inhibiting hXIAP, a cognate partner of hHtrA2. Notably, zHtrA2 has well conserved properties of serine protease that specifically cleaves hParkin, a cognate substrate of hHtrA2. Interestingly, cytosolic (M)-zHtrA2, which does not bind hXIAP, induces atypical cell death in a serine protease-dependent manner, as occurs in hHtrA2. Thus, the zebrafish-zHtrA2 system can be used to clarify the crucial role of HtrA2 in maintaining the survival of living organisms and provide an opportunity to develop novel therapeutics for HtrA2-associated diseases, such as neurodegenerative diseases and cancer, which are caused by dysregulation of HtrA2.


Subject(s)
High-Temperature Requirement A Serine Peptidase 2/genetics , Homeostasis , Mitochondria/genetics , Animals , Caspases/metabolism , Cell Death , Genes, Mitochondrial , HEK293 Cells , High-Temperature Requirement A Serine Peptidase 2/metabolism , Humans , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
2.
Korean J Physiol Pharmacol ; 26(1): 15-24, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34965992

ABSTRACT

The development of selective targeting of drug molecules towards the mitochondria is an important issue related to therapy efficacy. In this study, we report that gallic acid (GA)-mitochondria targeting sequence (MTS)-H3R9 exhibits a dual role as a mitochondria-targeting vehicle with antioxidant activity for disease therapy. In viability assays, GA-MTS-H3R9 showed a better rescue action compared to that of MTS-H3R9. GA-MTS-H3R9 dramatically exhibited cell penetration and intercellular uptake compared to MTS and fit escape from lysosome release to the cytosol. We demonstrated the useful targeting of GA-MTS-H3R9 towards mitochondria in AC16 cells. Also, we observed that the antioxidant properties of mitochondrial-accrued GA-MTSH3R9 alleviated cell damage by reactive oxygen species production and disrupted mitochondrial membrane potential. GA-MTS-H3R9 showed a very increased cytoprotective effect against anticancer activity compared to that of MTS-H3R9. We showed that GA-MTS-H3R9 can act as a vehicle for mitochondria-targeting and as a reagent for therapeutic applications intended for cardiovascular disease treatment.

3.
J Pediatr Endocrinol Metab ; 34(12): 1619-1622, 2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34416793

ABSTRACT

OBJECTIVES: Monogenic diabetes mellitus (DM) is a single gene disorder, primarily characterized by impairment in the development or function of pancreatic beta cells. CASE PRESENTATION: A 14-year-old girl was initially diagnosed with type 2 DM. The patient did not have any anti-islet autoantibody and showed acanthosis nigricans. She was managed with long-acting insulin and oral hypoglycemic agent, but HbA1c was still 9.3% after 1 year of management. Her mother already had type 2 DM at 46-year-old and was on medication. Under the possibility of familial monogenic DM, targeted exome sequencing was performed which included 29 genes associated with monogenic DM. Nonsense mutation of the gene RFX6 (c.2661T>A, p.Tyr887∗) was found. After adding Glucagon-like peptide-1 (GLP-1) receptor agonist, HbA1c improved from 8.8 to 6.8% and body mass index (BMI) also improved from 31.0 to 29.2 kg/m2. CONCLUSIONS: It may be worth investigating genetic etiology in early-onset autoantibody-negative DM for specific genetic diagnosis and better management.


Subject(s)
Codon, Nonsense , Diabetes Mellitus, Type 2/pathology , Regulatory Factor X Transcription Factors/genetics , Adolescent , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/genetics , Female , Humans , Hypoglycemic Agents/therapeutic use , Middle Aged , Prognosis
4.
J Int Med Res ; 48(9): 300060520959494, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32993389

ABSTRACT

Vasopressin local infiltration is useful in gynecological surgery because it can reduce hemorrhage. Depending on the activities of the sympathetic system and the renin-angiotensin system, reactions to vasopressin may differ and predicting its systemic effects is difficult. Because life-threatening complications can occur, infiltration with vasopressin should be administered with caution. A 42-year-old female patient was diagnosed with uterine leiomyomas. During a robot-assisted laparoscopic myomectomy, 50 U of vasopressin, which is ten-times the recommended dose, was accidentally infiltrated. Subsequently, bradycardia with a heart rate of 25 bpm occurred, which recovered within 3 minutes. Peripheral perfusion indices and the diameter of the radial and brachial arteries also decreased markedly and recovered within 1 hour. The surgery was concluded without additional events. The patient was discharged 2 days later with no abnormal findings. Because vasopressin infiltration can cause life-threatening complications, it is necessary to determine the extent of patient reactions to vasopressin using measures such as the peripheral perfusion index or radial and brachial artery diameters. These measures may also help to predict the occurrence of complications.


Subject(s)
Leiomyoma , Uterine Myomectomy , Adult , Female , Gynecologic Surgical Procedures , Hemodynamics , Humans , Vasopressins
6.
J Int Med Res ; 47(6): 2740-2745, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31068034

ABSTRACT

A 7-year-old child underwent surgical excision of a benign mesothelioma of the pleura near the right lower lung. Although insertion of a wire-reinforced endotracheal tube through the left main bronchus was attempted for one-lung ventilation to secure the surgical field of view, the attempt failed. Therefore, an endotracheal tube was inserted into the trachea, and an Arndt endobronchial blocker (Cook Medical, Bloomington, IN, USA) was placed in the right intermediate bronchus under bronchoscopic guidance to selectively block the right lower and middle lobes. The surgery was performed while ventilating the right upper lobe and left lung, and no specific intraoperative adverse events occurred.


Subject(s)
Bronchi/surgery , Intubation, Intratracheal/methods , Mesothelioma/surgery , One-Lung Ventilation/methods , Thoracic Surgery, Video-Assisted/methods , Child , Female , Humans , Mesothelioma/pathology , Prognosis
7.
Ann Rehabil Med ; 43(6): 700-706, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31918532

ABSTRACT

OBJECTIVE: To present the branching patterns and anatomical course of the common fibular nerve (CFN) and its relationship with fibular head (FH). METHODS: A total of 21 limbs from 12 fresh cadavers were dissected. The FH width (FH_width), distance between the FH and CFN (FH_CFN), and thickness of the nerve were measured. The ratio of the FH_CFN to FH_width was calculated as follows: <1, cross type and ≥1, posterior type. Angle between the CFN and vertical line of the lower limb 5 cm proximal to the tip of the FH was measured. Branching patterns of the lateral cutaneous nerve of the calf (LCNC) were classified into four types according to its origin and direction as follows: type 1a, lateral margin of the CFN; type 1b, medial margin of the CFN; type 2, lateral sural cutaneous nerve (LSCN); and type 3, CFN and LSCN. RESULTS: In the cross type (15 cases, 71.4%), the ratio of FH_CFN/FH_width was 0.83 and the angle was 13.0°. In the posterior type (6 cases, 28.6%), the ratio was 1.04 and the angle was 11.0°. In the branching patterns of LCNC, type 2 was the most common (10 cases), followed by types 1a and 1b (both, 5 cases). CONCLUSION: Location of the CFN around the FH might be related to the development of its neuropathy, especially in the cross type of CFN. The LCNC showed various branching patterns and direction, which could be associated with difficulties of electrophysiologic testing.

8.
Ann Rehabil Med ; 42(3): 483-487, 2018 Jun 27.
Article in English | MEDLINE | ID: mdl-29961748

ABSTRACT

Ulnar neuropathy at the wrist is an uncommon disease and pure ulnar sensory neuropathy at the wrist is even rarer. It is difficult to diagnose pure ulnar sensory neuropathy at the wrist by conventional methods. We report a case of pure ulnar sensory neuropathy at the hypothenar area. The lesion was localized between 3 cm and 5 cm distal to pisiform using orthodromic inching test of ulnar sensory nerve to stimulate at three points around the hypothenar area. Ultrasonographic examination confirmed compression of superficial sensory branch of the ulnar nerve. Further, surgical exploration reconfirmed compression of the ulnar nerve. This case report demonstrates the utility of orthodromic ulnar sensory inching test.

9.
J Inherit Metab Dis ; 2018 May 08.
Article in English | MEDLINE | ID: mdl-29740774

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.

10.
PM R ; 10(12): 1380-1384, 2018 12.
Article in English | MEDLINE | ID: mdl-29783066

ABSTRACT

BACKGROUND: Safe and accurate needle access to the rhomboid major (RM) during electromyography is challenging due to the overlying trapezius muscle and the risk of pneumothorax. OBJECTIVE: To investigate the RM anatomy associated with the trapezius using ultrasonography and to determine a safe and accurate needle insertion point for needle electromyography of the RM. DESIGN: Descriptive study. SETTING: Department of physical medicine and rehabilitation of a tertiary clinic center. PARTICIPANTS: Participants between 23 and 71 years of age without any diseases (N = 25; 13 men, 12 women; 50 scapulae) were included. INTERVENTIONS: Ultrasonography of the RM and trapezius muscles around the scapula. MAIN OUTCOME MEASURES: The point at which the lateral margin of the trapezius crosses the medial border of the scapula (point A) was determined. The probe was positioned at the level of the midpoint (point M) between point A and the inferior angle of the scapula. The horizontal distance from the point at which the RM was the thickest (point X) to point M was measured. At point X, the depth of the RM, RM thickness, and the depth of the pleura were measured. RESULTS: The mean age and body mass index were 37.4 ± 12.0 years and 22.3 ± 2.1 kg/m2, respectively. Point M was located at a mean distance of 3.9 ± 0.6 cm proximal to the inferior angle of the scapula. The mean distance between point X and point M was 1.0 ± 0.2 cm. At point X, the RM was at a mean depth of 9.7 ± 3.1 mm from the skin and had a mean thickness of 9.9 ± 1.8 mm. The pleura was observed at a mean depth of 28.4 ± 3.8 mm from the skin. CONCLUSION: Needle electromyographic examination of the RM can be performed easily and safely through the lower part of the RM that is not covered by the trapezius. LEVEL OF EVIDENCE: not applicable.


Subject(s)
Electromyography/methods , Needles , Superficial Back Muscles/anatomy & histology , Superficial Back Muscles/diagnostic imaging , Ultrasonography , Adult , Aged , Body Mass Index , Electromyography/instrumentation , Female , Humans , Male , Middle Aged , Scapula , Young Adult
11.
J Inherit Metab Dis ; 41(6): 1007-1014, 2018 11.
Article in English | MEDLINE | ID: mdl-29663270

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-ß. A deficiency in G6Pase-α causes GSD type Ia (GSD-Ia) and a deficiency in G6Pase-ß 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-ß 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.


Subject(s)
Blood Glucose/analysis , Genetic Therapy , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/therapy , Animals , Antiporters/genetics , Dependovirus/genetics , Genetic Vectors , Homeostasis , Humans , Mice , Mice, Knockout , Monosaccharide Transport Proteins/genetics , Mutation
12.
Biochem Biophys Res Commun ; 498(4): 925-931, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29545180

ABSTRACT

Glycogen storage disease type Ia (GSD-Ia) is caused by a deficiency in glucose-6-phosphatase-α (G6Pase-α or G6PC), a key enzyme in endogenous glucose production. This autosomal recessive disorder is characterized by impaired glucose homeostasis and long-term complications of hepatocellular adenoma/carcinoma (HCA/HCC). We have shown that hepatic G6Pase-α deficiency-mediated steatosis leads to defective autophagy that is frequently associated with carcinogenesis. We now show that hepatic G6Pase-α deficiency also leads to enhancement of hepatic glycolysis and hexose monophosphate shunt (HMS) that can contribute to hepatocarcinogenesis. The enhanced hepatic glycolysis is reflected by increased lactate accumulation, increased expression of many glycolytic enzymes, and elevated expression of c-Myc that stimulates glycolysis. The increased HMS is reflected by increased glucose-6-phosphate dehydrogenase activity and elevated production of NADPH and the reduced glutathione. We have previously shown that restoration of hepatic G6Pase-α expression in G6Pase-α-deficient liver corrects metabolic abnormalities, normalizes autophagy, and prevents HCA/HCC development in GSD-Ia. We now show that restoration of hepatic G6Pase-α expression normalizes both glycolysis and HMS in GSD-Ia. Moreover, the HCA/HCC lesions in L-G6pc-/- mice exhibit elevated levels of hexokinase 2 (HK2) and the M2 isoform of pyruvate kinase (PKM2) which play an important role in aerobic glycolysis and cancer cell proliferation. Taken together, hepatic G6Pase-α deficiency causes metabolic reprogramming, leading to enhanced glycolysis and elevated HMS that along with impaired autophagy can contribute to HCA/HCC development in GSD-Ia.


Subject(s)
Glycogen Storage Disease Type I/metabolism , Liver/metabolism , Animals , Autophagy , Carcinoma, Hepatocellular/etiology , Glycogen Storage Disease Type I/enzymology , Glycolysis , Humans , Liver/enzymology , Liver/pathology , Liver Neoplasms/etiology , Mice , Pentose Phosphate Pathway
13.
Front Immunol ; 9: 2881, 2018.
Article in English | MEDLINE | ID: mdl-30619261

ABSTRACT

Osteoarthritis (OA) is a degenerative disease that induces pain, cartilage deformation, and joint inflammation. Mesenchymal stem cells (MSCs) are potential therapeutic agents for treatment of OA. However, MSC therapy can cause excessive inflammation. Signal transducer and activator of transcription 3 (STAT3) modulates secretion of many proinflammatory cytokines. Experimental OA was induced by intra-articular (IA) injection of monosodium iodoacetate (MIA) to the right knee of rats. MSCs from OA patients (OA-MSCs) were treated with STA21, a small molecule that blocks STAT3 signaling, by IA or intravenous (IV) injection after MIA injection. Pain severity was quantified by assessment of secondary tactile allodynia using the von Frey assessment test. Cartilage degradation was measured by microcomputed tomography image analysis, histological analysis, and the Mankin score. Protein and gene expression was evaluated by enzyme-linked immunosorbent assay, immunohistochemistry, and real-time polymerase chain reaction. MSCs increased production of proinflammatory cytokines under inflammatory conditions. STA21 significantly decreased expression of these proinflammatory molecules via inhibition of STAT3 activity but increased gene expression of molecules related to migration potential and immunomodulation in OA-MSCs. STAT3-inhibited OA-MSCs administrated by IV or IA injection decreased pain severity and cartilage damage in rats with MIA-induced OA rats by decreasing proinflammatory cytokines in the joints. Combined IA and IV-injected STAT3-inhibited OA-MSCs had an additive effect of pain relief in MIA-induced OA rats. STAT3 inhibition may optimize the therapeutic activities of MSCs for treating OA by attenuating pain and progression of MIA by inhibiting inflammation and cartilage damage.


Subject(s)
Arthritis, Experimental/metabolism , Cartilage, Articular/metabolism , Mesenchymal Stem Cells/metabolism , Osteoarthritis/metabolism , Pain/metabolism , STAT3 Transcription Factor/metabolism , Administration, Intravenous , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/therapy , Cartilage, Articular/pathology , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Heterografts , Humans , Injections, Intra-Articular , Iodoacetic Acid , Male , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/drug effects , Osteoarthritis/chemically induced , Osteoarthritis/therapy , Pain/physiopathology , Pain/prevention & control , Rats, Wistar , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
14.
Hum Mol Genet ; 26(22): 4395-4405, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28973635

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.


Subject(s)
Genetic Therapy/methods , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/therapy , 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
15.
Exp Mol Med ; 49(7): e355, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28706297

ABSTRACT

The argon plasma jet (Ar-PJ) is widely used in medical fields such as dermatology and dentistry, and it is considered a promising tool for cancer therapy. However, the in vivo effects of Ar-PJ for medical uses have not yet been investigated, and there are no biological tools to determine the appropriate clinical dosages of Ar-PJ. In this study, we used the caudal fin and embryo of zebrafish as novel in vivo tools to evaluate the biosafety of Ar-PJ. Typically, Ar-PJ is known to induce cell death in two-dimensional (2D) cell culture systems. By contrast, no detrimental effects of Ar-PJ were shown in our 3D zebrafish systems composed of 2D cells. The Ar-PJ-treated caudal fins grew by an average length of 0.7 mm, similar to the length of the normally regenerating fins. Remarkably, Ar-PJ did not affect the expression patterns of Wnt8a and ß-Catenin, which play important roles in fin regeneration. In the embryo system, 85% of the Ar-PJ-treated embryos hatched, and the lateral length of these embryos was ~3.3 mm, which are equivalent to the lengths of normal embryos. In particular, vasculogenesis, which is the main cellular process during tissue regeneration and embryogenesis, occurred normally under the Ar-PJ dose used in this study. Therefore, our biosafety evaluation tools that use living model systems can be used to provide an experimental guideline to determine the clinically safe dosage of Ar-PJ.


Subject(s)
Argon Plasma Coagulation/adverse effects , Argon/adverse effects , Embryonic Development , Plasma Gases/adverse effects , Regeneration , Animal Fins , Animals , Cells, Cultured , Cytoskeletal Proteins/metabolism , Fibroblasts/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Models, Animal , Wnt Proteins/metabolism , Zebrafish , Zebrafish Proteins/metabolism , beta Catenin/metabolism
16.
PLoS Genet ; 13(5): e1006819, 2017 May.
Article in English | MEDLINE | ID: mdl-28558013

ABSTRACT

A deficiency in glucose-6-phosphatase-α (G6Pase-α) in glycogen storage disease type Ia (GSD-Ia) leads to impaired glucose homeostasis and metabolic manifestations including hepatomegaly caused by increased glycogen and neutral fat accumulation. A recent report showed that G6Pase-α deficiency causes impairment in autophagy, a recycling process important for cellular metabolism. However, the molecular mechanism underlying defective autophagy is unclear. Here we show that in mice, liver-specific knockout of G6Pase-α (L-G6pc-/-) leads to downregulation of sirtuin 1 (SIRT1) signaling that activates autophagy via deacetylation of autophagy-related (ATG) proteins and forkhead box O (FoxO) family of transcriptional factors which transactivate autophagy genes. Consistently, defective autophagy in G6Pase-α-deficient liver is characterized by attenuated expressions of autophagy components, increased acetylation of ATG5 and ATG7, decreased conjugation of ATG5 and ATG12, and reduced autophagic flux. We further show that hepatic G6Pase-α deficiency results in activation of carbohydrate response element-binding protein, a lipogenic transcription factor, increased expression of peroxisome proliferator-activated receptor-γ (PPAR-γ), a lipid regulator, and suppressed expression of PPAR-α, a master regulator of fatty acid ß-oxidation, all contributing to hepatic steatosis and downregulation of SIRT1 expression. An adenovirus vector-mediated increase in hepatic SIRT1 expression corrects autophagy defects but does not rectify metabolic abnormalities associated with G6Pase-α deficiency. Importantly, a recombinant adeno-associated virus (rAAV) vector-mediated restoration of hepatic G6Pase-α expression corrects metabolic abnormalities, restores SIRT1-FoxO signaling, and normalizes defective autophagy. Taken together, these data show that hepatic G6Pase-α deficiency-mediated down-regulation of SIRT1 signaling underlies defective hepatic autophagy in GSD-Ia.


Subject(s)
Autophagy , Glycogen Storage Disease Type I/metabolism , Signal Transduction , Sirtuin 1/metabolism , Animals , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Forkhead Transcription Factors/metabolism , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Glycogen Storage Disease Type I/genetics , Hepatocytes/metabolism , Mice , PPAR gamma/genetics , PPAR gamma/metabolism , Sirtuin 1/genetics
17.
Hum Mol Genet ; 26(10): 1890-1899, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28334808

ABSTRACT

Glycogen storage disease type Ia (GSD-Ia) is characterized by impaired glucose homeostasis and long-term risks of hepatocellular adenoma (HCA) and carcinoma (HCC). We have shown that the non-tumor-bearing (NT), recombinant adeno-associated virus (rAAV) vector-treated GSD-Ia mice (AAV-NT mice) expressing a wide range (0.9-63%) of normal hepatic glucose-6-phosphatase-α activity maintain glucose homeostasis and display physiologic features mimicking animals living under calorie restriction (CR). We now show that in AAV-NT mice, the signaling pathways of the CR mediators, AMP-activated protein kinase (AMPK) and sirtuin-1 are activated. AMPK/sirtuin-1 inhibit the activity of STAT3 (signal transducer and activator of transcription 3) and NFκB (nuclear factor κB), the pro-inflammatory and cancer-promoting transcription factors. Sirtuin-1 also inhibits cancer metastasis via increasing the expression of E-cadherin, a tumor suppressor, and decreasing the expression of mesenchymal markers. Consistently, in AAV-NT mice, hepatic levels of active STAT3 and NFκB-p65 were reduced as were expression of mesenchymal markers, STAT3 targets, NFκB targets and ß-catenin targets, all of which were consistent with the promotion of tumorigenesis. AAV-NT mice also expressed increased levels of E-cadherin and fibroblast growth factor 21 (FGF21), targets of sirtuin-1, and ß-klotho, which can acts as a tumor suppressor. Importantly, treating AAV-NT mice with a sirtuin-1 inhibitor markedly reversed many of the observed anti-inflammatory/anti-tumorigenic signaling pathways. In summary, activation of hepatic AMPK/sirtuin-1 and FGF21/ß-klotho signaling pathways combined with down-regulation of STAT3/NFκB-mediated inflammatory and tumorigenic signaling pathways can explain the absence of hepatic tumors in AAV-NT mice.


Subject(s)
Glucose-6-Phosphatase/metabolism , Liver/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Cadherins/genetics , Carcinogenesis/metabolism , Disease Models, Animal , Down-Regulation , Gene Expression , Genetic Therapy , Genetic Vectors , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/therapy , Inflammation/metabolism , Liver Neoplasms/metabolism , Mice , NF-kappa B , STAT3 Transcription Factor , Signal Transduction , Sirtuin 1/metabolism , beta Catenin/genetics
18.
Mol Genet Metab ; 120(3): 229-234, 2017 03.
Article in English | MEDLINE | ID: mdl-28096054

ABSTRACT

Glycogen storage disease type Ia (GSD-Ia), characterized by impaired glucose homeostasis and chronic risk of hepatocellular adenoma (HCA) and carcinoma (HCC), is caused by a deficiency in glucose-6-phosphatase-α (G6Pase-α or G6PC). We have previously shown that G6pc-/- mice receiving gene transfer mediated by rAAV-G6PC, a recombinant adeno-associated virus (rAAV) vector expressing G6Pase-α, and expressing 3-63% of normal hepatic G6Pase-α activity maintain glucose homeostasis and do not develop HCA/HCC. However, the threshold of hepatic G6Pase-α activity required to prevent tumor formation remained unknown. In this study, we constructed rAAV-co-G6PC, a rAAV vector expressing a codon-optimized (co) G6Pase-α and showed that rAAV-co-G6PC was more efficacious than rAAV-G6PC in directing hepatic G6Pase-α expression. Over an 88-week study, we showed that both rAAV-G6PC- and rAAV-co-G6PC-treated G6pc-/- mice expressing 3-33% of normal hepatic G6Pase-α activity (AAV mice) maintained glucose homeostasis, lacked HCA/HCC, and were protected against age-related obesity and insulin resistance. Of the eleven rAAV-G6PC/rAAV-co-G6PC-treated G6pc-/- mice harboring 0.9-2.4% of normal hepatic G6Pase-α activity (AAV-low mice), 3 expressing 0.9-1.3% of normal hepatic G6Pase-α activity developed HCA/HCC, while 8 did not (AAV-low-NT). Finally, we showed that the AAV-low-NT mice exhibited a phenotype indistinguishable from that of AAV mice expressing ≥3% of normal hepatic G6Pase-α activity. The results establish the threshold of hepatic G6Pase-α activity required to prevent HCA/HCC and show that GSD-Ia mice harboring <2% of normal hepatic G6Pase-α activity are at risk of tumor development.


Subject(s)
Adenoma, Liver Cell/prevention & control , Carcinoma, Hepatocellular/prevention & control , Genetic Therapy/methods , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/therapy , Liver Neoplasms/prevention & control , Adenoma, Liver Cell/enzymology , Animals , Carcinoma, Hepatocellular/enzymology , Dependovirus/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Genetic Vectors/administration & dosage , Glucose/metabolism , Glucose-6-Phosphatase/metabolism , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/enzymology , Homeostasis , Humans , Liver/enzymology , Liver Neoplasms/enzymology , Mice
19.
Liver Res ; 1(3): 174-180, 2017 Sep.
Article in English | MEDLINE | ID: mdl-29576889

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.

20.
Biochem Biophys Res Commun ; 482(4): 569-574, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27864142

ABSTRACT

Glycogen storage disease type Ib (GSD-Ib), characterized by impaired glucose homeostasis, neutropenia, and neutrophil dysfunction, is an inherited autosomal recessive disorder caused by a deficiency in the glucose-6-phosphate transporter (G6PT). Neutrophils play an essential role in the defense against invading pathogens. The recruitment of neutrophils towards the inflammation sites in response to inflammatory stimuli is a tightly regulated process involving rolling, adhesion, and transmigration. In this study, we investigated the role of G6PT in neutrophil adhesion and migration using in vivo and in vitro models. We showed that the GSD-Ib (G6pt-/-) mice manifested severe neutropenia in both blood and bone marrow, and treating G6pt-/- mice with granulocyte colony-stimulating factor (G-CSF) corrected neutropenia. However, upon thioglycolate challenge, neutrophils from both untreated and G-CSF-treated G6pt-/-mice exhibited decreased ability to migrate to the peritoneal cavity. In vitro migration and cell adhesion of G6PT-deficient neutrophils were also significantly impaired. Defects in cell migration were not due to enhanced apoptosis or altered fMLP receptor expression. Remarkably, the expression of the ß2 integrins CD11a and CD11b, which are critical for cell adhesion, was greatly decreased in G6PT-deficient neutrophils. This study suggests that deficiencies in G6PT cause impairment in neutrophil adhesion and migration via aberrant expression of ß2 integrins, and our finding should facilitate the development of novel therapies for GSD-Ib.


Subject(s)
Cell Adhesion , Cell Movement , Glycogen Storage Disease Type I/pathology , Neutropenia/pathology , Neutrophils/pathology , Animals , Antiporters/genetics , Apoptosis , Caco-2 Cells , Cells, Cultured , Gene Deletion , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/genetics , Humans , Mice , Monosaccharide Transport Proteins/genetics , Neutropenia/complications , Neutropenia/genetics , Neutrophils/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...