Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
1.
CNS Neurosci Ther ; 30(2): e14594, 2024 02.
Article in English | MEDLINE | ID: mdl-38332538

ABSTRACT

BACKGROUND: With the rapidly increasing prevalence of metabolic diseases such as type 2 diabetes mellitus (T2DM), neuronal complications associated with these diseases have resulted in significant burdens on healthcare systems. Meanwhile, effective therapies have remained insufficient. A novel fatty acid called S-9-PAHSA has been reported to provide metabolic benefits in T2DM by regulating glucose metabolism. However, whether S-9-PAHSA has a neuroprotective effect in mouse models of T2DM remains unclear. METHODS: This in vivo study in mice fed a high-fat diet (HFD) for 5 months used fasting blood glucose, glucose tolerance, and insulin tolerance tests to examine the effect of S-9-PAHSA on glucose metabolism. The Morris water maze test was also used to assess the impact of S-9-PAHSA on cognition in the mice, while the neuroprotective effect of S-9-PAHSA was evaluated by measuring the expression of proteins related to apoptosis and oxidative stress. In addition, an in vitro study in PC12 cells assessed apoptosis, oxidative stress, and mitochondrial membrane potential with or without CAIII knockdown to determine the role of CAIII in the neuroprotective effect of S-9-PAHSA. RESULTS: S-9-PAHSA reduced fasting blood glucose levels significantly, increased insulin sensitivity in the HFD mice and also suppressed apoptosis and oxidative stress in the cortex of the mice and PC12 cells in a diabetic setting. By suppressing oxidative stress and apoptosis, S-9-PAHSA protected both neuronal cells and microvascular endothelial cells in in vivo and in vitro diabetic environments. Interestingly, this protective effect of S-9-PAHSA was reduced significantly when CAIII was knocked down in the PC12 cells, suggesting that CAIII has a major role in the neuroprotective effect of S-9-PAHSA. However, overexpression of CAIII did not significantly enhance the protective effect of S-9-PAHSA. CONCLUSION: S-9-PAHSA mediated by CAIII has the potential to exert a neuroprotective effect by suppressing apoptosis and oxidative stress in neuronal cells exposed to diabetic conditions. Furthermore, S-9-PAHSA has the capability to reduce fasting blood glucose and LDL levels and enhance insulin sensitivity in mice fed with HFD.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Neuroprotective Agents , Palmitic Acid , Stearic Acids , Animals , Mice , Rats , Apoptosis , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diet, High-Fat/adverse effects , Disease Models, Animal , Endothelial Cells/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress , Carbonic Anhydrase III/drug effects , Carbonic Anhydrase III/metabolism
2.
J Endocrinol Invest ; 46(7): 1449-1458, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36646964

ABSTRACT

PURPOSE: Adrenocortical carcinoma (ACC), a rare malignancy of the adrenocortex, is characterized by a crosstalk between the adipose microenvironment and tumor. Here, we assessed the involvement of carbonic anhydrase (CA) enzymes III and IX (CAIII and CAIX), in the metabolic alterations of the adipose tissue characterizing obesity and in the local crosstalk between the tumor adipose microenvironment and ACC. RESULTS/METHODS: CAIII and CAIX expression is altered in visceral adipose tissue (VAT) in obesity and in ACC. A significant CAIX upregulation was present in ACC at advanced stages (n = 14) (fold increase FI = 7.4 ± 0.1, P < 0.05) associated with lower CAIII levels (FI = 0.25 ± 0.06, P < 0.001), compared with lower stages (n = 9). In vitro coculture between visceral adipose stem cells (ASCs) and ACC cell lines, H295R and MUC-1, mimicking the interaction occurring between VAT and advanced ACC, showed a significant CAIX upregulation in H295R but not in MUC-1 cells, and a decreased expression of CAIII. The effect on adipose cells was different when cocultured with H295R or MUC-1 cells. Coculture did not modulate CAIII expression in ASCs, which, however, was significantly downregulated with H295R (FI = 0.34 ± 0.11, P < 0.05) and upregulated by MUC-1 when cocultured ASCs were induced to differentiate toward adipocytes, with an expression profile similar to what found in VAT of obese subjects. CAIX expression was markedly increased in ASCs cocultured with H295R and to a less extent following adipogenesis induction (FI = 150.9 ± 46.5 and FI = 4.6 ± 1.1, P < 0.01, respectively). CONCLUSION: Our findings highlight a modulation of CAIII and CAIX in the metabolic crosstalk between ACC and its local adipose microenvironment, suggesting that CAs might represent a potential target for novel anticancer therapies.


Subject(s)
Adrenal Cortex Neoplasms , Adrenocortical Carcinoma , Carbonic Anhydrase III , Carbonic Anhydrases , Humans , Carbonic Anhydrase IX , Antigens, Neoplasm/metabolism , Carbonic Anhydrases/metabolism , Obesity , Tumor Microenvironment
3.
FEBS Open Bio ; 12(4): 827-834, 2022 04.
Article in English | MEDLINE | ID: mdl-35108454

ABSTRACT

The abnormal lipid metabolism in the liver that occurs after high caloric intake is the main cause of nonalcoholic fatty liver disease (NAFLD). Differences between samples from healthy livers and livers from individuals with NAFLD indicate that changes in liver function occur during disease progression. Here, we examined changes in protein expression in a fatty liver model in the early stages of obesity to identify potential alterations in function. The proteins expressed in the liver tissue of pre-obese rats were separated via SDS/PAGE and stained with Coomassie brilliant blue-G250. Peptide mass fingerprinting indicated an increase in the expression of carbonic anhydrase 3 (CA3) relative to controls. Western blotting analysis confirmed the increase in CA3 expression, even in an early fat-accumulation state in which excessive weight gain had not yet occurred. In human hepatoma HepG2 cells, fat accumulation induced with oleic acid also resulted in increased CA3 expression. When the cells were in a state of fat accumulation, treating them with the CA3 inhibitors acetazolamide (ACTZ) or 6-ethoxyzolamide (ETZ) suppressed fat accumulation, but only ETZ somewhat reduced the fat-induced upregulation of CA3 expression. Expression of CA3 was therefore upregulated in response to the consumption of a high-fat diet, even in the absence of an increase in body weight. The suppression of CA3 activity by ACTZ or ETZ reduced fat accumulation in hepatocytes, suggesting that CA3 is involved in the development of fatty liver.


Subject(s)
Adipogenesis , Carbonic Anhydrase III , Non-alcoholic Fatty Liver Disease , Animals , Carbonic Anhydrase III/antagonists & inhibitors , Carbonic Anhydrase III/metabolism , Liver/enzymology , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity/drug therapy , Rats
4.
Mol Biol Rep ; 48(11): 7397-7404, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34651296

ABSTRACT

BACKGROUND: Carbonic anhydrases (CAs) play a significant role in maintaining pH balance by catalyzing the conversion of carbon dioxide to bicarbonate. The regulation of pH is critical for all living organisms. Although there are many studies in the literature on the biochemical, functional, and structural features of CAs, there is not sufficient information about the epigenetic regulation of CAs. METHODS AND RESULTS: The lysine acetyltransferase TIP60 (60 kDa Tat-interactive protein) was knocked out specifically in mouse liver using the Cre/loxP system, and knockout rate was shown as 83-88% by Southern blot analysis. The impact of Tip60 on the expression of Ca1, Ca3, and Ca7 was investigated at six Zeitgeber time (ZT) points in the control and liver-specific Tip60 knockout mice (mutant) groups by real-time PCR. In the control group, while Ca1 showed the highest expression at ZT8 and ZT12, the lowest expression profile was observed at ZT0 and ZT20. Hepatic Ca1 displayed robust circadian expression. However, hepatic Ca3 exhibited almost the same level of expression at all ZT points. The highest expression of Ca7 was observed at ZT12, and the lowest expression was determined at ZT4. Furthermore, hepatic Ca7 also showed robust circadian expression. The expression of Ca1 and Ca3 significantly decreased in mutant mice at all time periods, but the expression of Ca7 used as a negative control was not affected. CONCLUSIONS: It was suggested for the first time that Tip60 might be considered a candidate protein in the regulation of the Ca1 and Ca3 genes, possibly by acetylation.


Subject(s)
Carbonic Anhydrase III/metabolism , Carbonic Anhydrase I/metabolism , Circadian Rhythm , Liver/metabolism , Lysine Acetyltransferase 5/metabolism , Trans-Activators/metabolism , Acetylation , Animals , Lysine Acetyltransferase 5/genetics , Male , Mice , Mice, Knockout , Protein Processing, Post-Translational , Trans-Activators/genetics
5.
Metab Brain Dis ; 36(8): 2329-2341, 2021 12.
Article in English | MEDLINE | ID: mdl-34665375

ABSTRACT

Type 2 diabetes mellitus (T2DM) is one of the most common chronic diseases among the elderly people. The T2DM increases the risk of cardio-cerebrovascular disease (CCD), and the main pathological change of the CCD is atherosclerosis (AS). Meanwhile, the carbonic anhydrases (CAs) are involved in the formation and progression of plaques in AS. However, the exact physiological mechanism of carbonic anhydrase III (CAIII) has not been clear yet, and there are also no correlation study between CAIII protein and T2DM with CCD. The 8-week old diabetic mice (db/db-/- mice) and wild-type mice (wt mice) were feed by a normal diet till 32 weeks, and detected the carotid artery vascular opening angle using the method of biomechanics; The changes of cerebral cortex and myocardium were watched by the ultrastructure, and the autophagy were observed by electron microscope; The tissue structure, inflammation and cell injury were observed by Hematoxylin and eosin (HE) staining; The apoptosis of cells were observed by TUNEL staining; The protein levels of CAIII, IL-17, p53 were detected by immunohistochemical and Western Blot, and the Beclin-1, LC3, NF-κB were detected by Western Blot. All statistical analysis is performed using PRISM software. Compared with wt mice, db/db-/- mice' carotid artery open angle increased significantly. Electron microscope results indicated that autophagy in db/db-/- mice cerebral cortex and heart tissue decreased and intracellular organelle ultrastructure were damaged. HE staining indicated that, db/db-/- mice' cerebral cortex and heart tissue stained lighter, inflammatory cells infiltration, cell edema were obvious, myocardial fibers were disorder, and myocardial cells showed different degrees of degeneration. Compared with wt mice, TUNEL staining showed that there was obviously increase in db/db-/- mice cortex and heart tissue cell apoptosis. The results of immunohistochemistry and Western Blot indicated that CAIII, Beclin-1 and LC3II/I expression levels conspicuously decreased in cortex and heart tissue of db/db-/- mice, and the expression level of IL-17, NF-κB and p53 obviously increased. The carotid artery' vascular stiffness was increased and which was probably related with formation of AS in diabetic mice. And the autophagy participated in the occurrence and development of diabetic CCD. CAIII protein might somehow be involved in the regulation of autophagy probably through affecting cell apoptosis and inflammation, but the underlying mechanism remains to be further studied.


Subject(s)
Carbonic Anhydrase III , Cerebrovascular Disorders , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Animals , Autophagy , Mice
6.
Cardiovasc Toxicol ; 21(11): 914-926, 2021 11.
Article in English | MEDLINE | ID: mdl-34387844

ABSTRACT

Myocardial ischemia can cause insufficient oxygen and functional damage to myocardial cells. Carbonic anhydrase III (CAIII) has been found to be closely related to the abnormality of cardiomyocytes. To investigate the role of CAIII in the apoptosis of myocytes under hypoxic conditions and facilitate the strategy for treating hypoxia-induced damage, in vitro experiments in H9c2 were employed. The protein expression of CAIII in H9c2 cells after hypoxia or normoxia treatment was determined by western blotting and immunohistochemistry. MTT assay was employed for cells viability measurement and LDH release was monitored. The apoptotic cells were observed using immunofluorescence assay, flow cytometric analysis, and TUNEL assay. CAIII-overexpression or -knockdown cells were constructed to determine the role of CAIII in regulating apoptosis-related proteins caspase-3, Bax, Bcl-2, and anti-apoptosis pathway PI3K/Akt/mTOR. The mRNA levels of CAIII and genes related to CAIII synthesis including REN, IGHM, APOBEC 3F, and SKOR2 were significantly upregulated in hypoxia fetal sheep. The expression of CAIII protein and content of apoptotic H9c2 cells were increased at 1, 3, 6, and 12 h after hypoxia treatment. Overexpression of CAIII significantly upregulated Bcl2 level and downregulated Bax and caspase-3 cleavage levels, while its knockdown led to the contrary results. Overexpressed CAIII promoted the HIF-1α level and activated the PI3K/Akt/mTOR pathway, thereby exerting an inhibitory effect on hypoxia-induced apoptosis. In conclusion, our findings revealed that CAIII could protect cell from hypoxia-apoptosis of H9c2 cells, in which, activated PI3K/Akt/mTOR signaling pathway may be involved.


Subject(s)
Apoptosis , Carbonic Anhydrase III/metabolism , Fetal Heart/enzymology , Myocytes, Cardiac/enzymology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Carbonic Anhydrase III/genetics , Cell Hypoxia , Cell Line , Fetal Heart/pathology , Gestational Age , Myocytes, Cardiac/pathology , Rats , Sheep, Domestic , Signal Transduction
7.
Gastroenterology ; 160(7): 2467-2482.e3, 2021 06.
Article in English | MEDLINE | ID: mdl-33647280

ABSTRACT

BACKGROUNDS & AIMS: Squalene epoxidase (SQLE) is the rate-limiting enzyme for cholesterol biosynthesis. We elucidated the functional significance, molecular mechanisms, and clinical impact of SQLE in nonalcoholic steatohepatitis (NASH). METHODS: We performed studies with hepatocyte-specific Sqle overexpression transgenic (Sqle tg) mice and mice given high-fat high-cholesterol (HFHC) or methionine- and choline-deficient (MCD) diet to induce NASH. SQLE downstream target carbonic anhydrase III (CA3) was identified using co-immunoprecipitation and Western Blot. Some mice were given SQLE inhibitor (terbinafine) and CA3 inhibitor (acetazolamide) to study the therapeutic effects in NASH. Human samples (N = 217) including 65 steatoses, 80 NASH, and 72 healthy controls were analyzed for SQLE levels in liver tissue and in serum. RESULTS: SQLE is highly up-regulated in human NASH and mouse models of NASH. Sqle tg mice triggered spontaneous insulin resistance, hepatic steatosis, liver injury, and accelerated HFHC or MCD diet-induced NASH development. Mechanistically, SQLE tg mice caused hepatic cholesterol accumulation, thereby triggering proinflammatory nuclear factor-κB signaling and steatohepatitis. SQLE directly bound to CA3, which induced sterol regulatory element-binding protein 1C activation, acetyl-CoA carboxylase, fatty acid synthase, and stearoyl-CoA desaturase1 expression and de novo hepatic lipogenesis. Combined targeting SQLE (terbinafine) and CA3 (acetazolamide) synergistically ameliorated NASH in mice with superior efficacy to either drug alone. Serum SQLE with CA3 could distinguish patients with NASH from steatosis and healthy controls (area under the receiver operating characteristic curve, 0.815; 95% confidence interval, 0.758-0.871). CONCLUSIONS: SQLE drives the initiation and progression of NASH through inducing cholesterol biosynthesis, and SQLE/CA3 axis-mediated lipogenesis. Combined targeting of SQLE and CA3 confers therapeutic benefit in NASH. Serum SQLE and CA3 are novel biomarkers for the noninvasive diagnosis of patients with NASH.


Subject(s)
Carbonic Anhydrase III/metabolism , Cholesterol/biosynthesis , Non-alcoholic Fatty Liver Disease/metabolism , Squalene Monooxygenase/metabolism , Animals , Biomarkers/metabolism , Diet, High-Fat , Disease Models, Animal , Hepatocytes/metabolism , Humans , Insulin Resistance , Lipogenesis , Liver/metabolism , Mice , Mice, Transgenic , Non-alcoholic Fatty Liver Disease/etiology , Up-Regulation
8.
Ann Palliat Med ; 10(12): 12554-12565, 2021 Dec.
Article in English | MEDLINE | ID: mdl-35016406

ABSTRACT

BACKGROUND: Dilated cardiomyopathy (DCM) is a complex type of cardiomyopathy that is affected by both genetic and non-genetic factors. It is characterized by an enlargement of the left ventricle or bi-ventricle, and is often accompanied by cardiac systolic dysfunction. The main results include arrhythmia, heart failure (HF), and sudden death. The prognosis of this disease is usually poor, and the 5-year survival time is about 50%. Early diagnosis is very important for the treatment of DCM. Studies have shown that primary prevention after discovering the disease effectively reduces the mortality rate of the disease. However, there is currently no effective biomarker for the early diagnosis of DCM. The rapid development of omics in protein has promoted the "precise" study of modern medical research. In this article, the potential biomarkers for predicting and diagnosing DCM-related HF were studied by a plasma protein omics analysis. METHODS: Tandem mass tag-labeled quantitative proteomic studies were performed in 20 patients, comprising 10 DCM-associated HF patients, and 10 control patients who without clinical HF events. Further validation research was conducted by enzyme-linked immunosorbent assay (ELISA) with an expanded cohort (control group =40; HF group =48). RESULTS: Among the 854 identified proteins, the expression of 86 proteins was significantly upregulated, while the expression of 21 proteins was downregulated (with an expression difference >1.5-fold; P<0.05) in the 2 groups. The Gene Ontology, Kyoto Encyclopedia of Genes and Genomes pathway enrichment, and protein-protein interaction (PPI) networks analyses indicated that the bicarbonate transport process played a critical role in HF. Importantly, carbonic anhydrase 2 (CA2) and 3 (CA3), which play central roles in regulating the transport of bicarbonate, were highly expressed in the HF group. The ELISA validation results showed that the expression levels of CA2 and CA3 at admission were remarkably higher (P<0.0001 and P=0.0157) in the plasma of the HF patients than that of the control patients. CONCLUSIONS: The present study showed that two molecules (i.e., CA2 and CA3) are involved in the bicarbonate transport pathway, and are risk factors and potential biomarkers for the diagnosis of DCM patients with HF.


Subject(s)
Carbonic Anhydrase III , Carbonic Anhydrase II , Cardiomyopathy, Dilated , Heart Failure , Biomarkers , Cardiomyopathy, Dilated/genetics , Humans , Proteomics
9.
Gene ; 762: 145034, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32777521

ABSTRACT

Carbonic Anhydrase III (CAIII) belongs to a member of the alpha Carbonic Anhydrase (CA) family. Although some CA members are strongly up-regulated by HIF1-α, it is not known about the transcriptional regulation of CAIII in prostate cancer cells, PCa. Therefore, we aimed to identify regulatory regions important for the regulation of CAIII gene under hypoxic conditions in human prostate cancer cells (PC3). The present study, for the first time, demonstrated that the chemically mimicked hypoxic condition led to the induced CAIII mRNA and protein expression in prostate cancer cells. Transcriptional regulation of CAIII was investigated by transient transfection assay that indicates that the most active promoter activity was in the region of P2 -699/+86. Hypoxic condition also upregulates the basal activity of for P1;-941/+86 and P2;-699/+86 constructs containing putative Hypoxia Response Element (HRE) region located in -268/-252. EMSA analysis of HRE located in -268/-252 bases, showed one DNA-protein binding complexes. Competition assays indicated this complex is resulted from HIF1α interactions. In addition, site-directed mutagenesis of potential HIF1α binding sites diminished a DNA-protein complex. These findings suggest that CAIII is a hypoxia-regulated gene and valuable for targeting of prostate cancer tumors in hypoxic condition.


Subject(s)
Carbonic Anhydrase III/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Prostatic Neoplasms/metabolism , Carbonic Anhydrase III/metabolism , Cell Hypoxia , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , PC-3 Cells , Promoter Regions, Genetic , Up-Regulation
10.
J Enzyme Inhib Med Chem ; 35(1): 1483-1490, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32635773

ABSTRACT

A series of 17 compounds (12-16 b) with 2,4,5-trisubstitutedthiazole scaffold having 5-aryl group, 4-carboxylic acid/ester moiety, and 2-amino/amido/ureido functional groups were synthesised, characterised, and evaluated for their carbonic anhydrase (CA)-III inhibitory activities using the size exclusion Hummel-Dreyer method (HDM) of chromatography. Compound 12a with a free amino group at the 2-position, carboxylic acid moiety at the 4-position, and a phenyl ring at the 5-position of the scaffold was found to be the most potent CA-III inhibitor (Ki = 0.5 µM). The presence of a carboxylic acid group at the 4-position of the scaffold was found to be crucial for the CA-III inhibitory activity. Furthermore, replacement of the free amino group with an amide and urea group resulted in a significant reduction of activity (compounds 13c and 14c, Ki = 174.1 and 186.2 µM, respectively). Thus, compound 12a (2-amino-5-phenylthiazole-4-carboxylic acid) can be considered as the lead molecule for further modification and development of more potent CA-III inhibitors.


Subject(s)
Carbonic Anhydrase III/antagonists & inhibitors , Carbonic Anhydrase Inhibitors/pharmacology , Thiazoles/pharmacology , Animals , Carbonic Anhydrase III/metabolism , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/chemistry , Cattle , Dose-Response Relationship, Drug , Molecular Structure , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry
11.
Cells ; 9(3)2020 03 13.
Article in English | MEDLINE | ID: mdl-32183030

ABSTRACT

Epithelial-mesenchymal transition (EMT) is strongly correlated with tumor metastasis and contains several protein markers, such as E-cadherin. Carbonic anhydrase III (CA III) exhibits low carbon dioxide hydratase activity in cancer. However, the detailed mechanisms of CA III and their roles in oral cancer are still unknown. This study established a CA III-overexpressed stable clone and observed the expression of CA III protein in human SCC-9 and SAS oral cancer cell lines. The migration and invasion abilities were determined using a Boyden chamber assay. Our results showed that the overexpression of CA III protein significantly increased the migration and invasion abilities in oral cancer cells. Moreover, a whole genome array analysis revealed that CA III regulated epithelial-mesenchymal transition by reducing the expression of epithelial markers. Data from the GEO database also demonstrated that CA III mRNA is negatively correlated with CDH1 mRNA. Mechanistically, CA III increased the cell motility of oral cancer cells through the FAK/Src signaling pathway. In conclusion, this suggests that CA III promotes EMT and cell migration and is potentially related to the FAK/Src signaling pathway in oral cancer.


Subject(s)
Carbonic Anhydrase III/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Mouth Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck/pathology , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Carbonic Anhydrase III/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Promoter Regions, Genetic , Signal Transduction , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Transcription Factors/genetics , src-Family Kinases/metabolism
12.
Exp Cell Res ; 385(1): 111672, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31614133

ABSTRACT

Carbonic anhydrase III (CAIII) is selectively expressed in slow-twitch myofibers in skeletal muscle. The fast-twitch to slow-twitch transformation of myofibers following denervation is accompanied by increased CAIII expression, suggesting that the effects of nerve impulses on skeletal-muscle remodeling influence CAIII expression. Here, we determined the molecular mechanisms underlying the effects of nerve conduction on CAIII expression. The results indicated that changes in skeletal-muscle [Ca2+]i altered CAIII expression. Moreover, results from the RNA-interference and over-expression experiments identified myocyte enhancer factor 2C (MEF2C) as the key transcription factor regulating [Ca2+]i-mediated changes in CAIII transcription. Additionally, chromatin immunoprecipitation experiments and luciferase assays confirmed MEF2C interaction and direct binding of the CAIII promoter between -416 and -200 base pair. Investigations of upstream cytoplasmic signaling pathways responsible for MEF2C activation revealed Ca2+/calmodulin-dependent protein kinase II (CaMKII) as the key factor involved in MEF2C-mediated regulation of CAIII expression. This study demonstrates that the Ca2+-CaMKII-MEF2C signaling pathway is the key factor involved in regulating CAIII expression in skeletal muscle. These results provide a theoretical basis supporting further investigations of changes in CAIII levels under different pathophysiological conditions and will facilitate a broader understanding of the biological functions of CAIII.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Carbonic Anhydrase III/genetics , Muscle, Skeletal/metabolism , Signal Transduction/genetics , Animals , Calcium/metabolism , Cell Line , Cytoplasm/genetics , Gene Expression Regulation/genetics , MEF2 Transcription Factors/genetics , Mice , Promoter Regions, Genetic/genetics , RNA Interference/physiology , Transcription Factors/genetics , Transcription, Genetic/genetics
13.
Am J Physiol Cell Physiol ; 317(5): C922-C931, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31390226

ABSTRACT

Carbonic anhydrase III (CAIII) is abundant in liver, adipocytes, and skeletal muscles, but not heart. A cytosolic enzyme that catalyzes conversions between CO2 and HCO3- in the regulation of intracellular pH, its physiological role in myocytes is not fully understood. Mouse skeletal muscles lacking CAIII showed lower intracellular pH during fatigue, suggesting its function in stress tolerance. We created transgenic mice expressing CAIII in cardiomyocytes that lack endogenous CAIII. The transgenic mice showed normal cardiac development and life span under nonstress conditions. Studies of ex vivo working hearts under normal and acidotic conditions demonstrated that the transgenic and wild-type mouse hearts had similar pumping functions under normal pH. At acidotic pH, however, CAIII transgenic mouse hearts showed significantly less decrease in cardiac function than that of wild-type control as shown by higher ventricular pressure development, systolic and diastolic velocities, and stroke volume via elongating the time of diastolic ejection. In addition to the effect of introducing CAIII into cardiomyocytes on maintaining homeostasis to counter acidotic stress, the results demonstrate the role of carbonic anhydrases in maintaining intracellular pH in muscle cells as a potential mechanism to treat heart failure.


Subject(s)
Acidosis/enzymology , Carbonic Anhydrase III/biosynthesis , Gene Expression Regulation, Enzymologic , Myocardium/enzymology , Acidosis/genetics , Animals , Carbonic Anhydrase III/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
14.
ChemMedChem ; 13(19): 2024-2029, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30088334

ABSTRACT

Pseudomonas aeruginosa is a Gram-negative facultative anaerobe belonging to the Pseudomonadaceae family. It is a multidrug-resistant opportunistic human pathogen, a common cause of life-threatening nosocomial infections, and a key bacterial agent in cystic fibrosis and endocarditis. The bacterium exhibits intrinsic resistance to most antibacterial agents, including aminoglycosides and quinolones. Hence, the identification of new drug targets for P. aeruginosa is ongoing. PsCA3 is a ß-class carbonic anhydrase (ß-CA) that catalyzes the reversible hydration of carbon dioxide to bicarbonate and represents a new class of antimicrobial target. Previously, inhibitor screening studies of psCA3 have shown that a series of small anions including sulfamide (SFN), imidazole (IMD), and 4-methylimidazole (4MI), and thiocyanate (SCN) inhibit the enzyme with efficiencies in the micro- to millimolar range. Herein the X-ray crystal structures of these inhibitors in complex with psCA3 are presented and compared with human CA II. This structural survey into the binding modes of small anions forms the foundation for the development of inhibitors against ß-CAs and more selective inhibitors against P. aeruginosa.


Subject(s)
Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Carbonic Anhydrase III/metabolism , Carbonic Anhydrase Inhibitors/metabolism , Pseudomonas aeruginosa/enzymology , Anti-Bacterial Agents/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Carbonic Anhydrase II/chemistry , Carbonic Anhydrase II/metabolism , Carbonic Anhydrase III/chemistry , Carbonic Anhydrase III/isolation & purification , Carbonic Anhydrase Inhibitors/chemistry , Catalytic Domain , Crystallography, X-Ray , Humans , Imidazoles/chemistry , Imidazoles/metabolism , Kinetics , Protein Binding , Sulfonamides/chemistry , Sulfonamides/metabolism , Thiocyanates/chemistry , Thiocyanates/metabolism
15.
Oxid Med Cell Longev ; 2018: 2018306, 2018.
Article in English | MEDLINE | ID: mdl-30154947

ABSTRACT

Under oxidative stress conditions, several constitutive cellular defense systems are activated, which involve both enzymatic systems and molecules with antioxidant properties such as glutathione and vitamins. In addition, proteins containing reactive sulfhydryl groups may eventually undergo reversible redox modifications whose products act as protective shields able to avoid further permanent molecular oxidative damage either in stressful conditions or under pathological circumstances. After the recovery of normal redox conditions, the reduced state of protein sulfhydryl groups is restored. In this context, carbonic anhydrases (CAs) III and VII, which are human metalloenzymes catalyzing the reversible hydration of carbon dioxide to bicarbonate and proton, have been identified to play an antioxidant role in cells where oxidative damage occurs. Both proteins are mainly localized in tissues characterized by a high rate of oxygen consumption, and contain on their molecular surface two reactive cysteine residues eventually undergoing S-glutathionylation. Here, we will provide an overview on the molecular and functional features of these proteins highlighting their implications into molecular processes occurring during oxidative stress conditions.


Subject(s)
Antioxidants/chemistry , Carbonic Anhydrase III/metabolism , Carbonic Anhydrases/metabolism , Oxidative Stress/physiology , Animals , Humans , Isoenzymes/metabolism , Oxidation-Reduction
16.
Sci Rep ; 8(1): 4856, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29559661

ABSTRACT

The integrity of the avascular nucleus pulposus (NP) phenotype plays a crucial role in the maintenance of intervertebral disc health. While advances have been made to define the molecular phenotype of healthy NP cells, the functional relevance of several of these markers remains unknown. In this study, we test the hypothesis that expression of Carbonic Anhydrase III (CAIII), a marker of the notochordal NP, is hypoxia-responsive and functions as a potent antioxidant without a significant contribution to pH homeostasis. NP, but not annulus fibrosus or end-plate cells, robustly expressed CAIII protein in skeletally mature animals. Although CAIII expression was hypoxia-inducible, we did not observe binding of HIF-1α to select hypoxia-responsive-elements on Car3 promoter using genomic chromatin-immunoprecipitation. Similarly, analysis of discs from NP-specific HIF-1α null mice suggested that CAIII expression was independent of HIF-1α. Noteworthy, silencing CAIII in NP cells had no effect on extracellular acidification rate, CO2 oxidation rate, or intracellular pH, but rather sensitized cells to oxidative stress-induced death mediated through caspase-3. Our data clearly suggests that CAIII serves as an important antioxidant critical in protecting NP cells against oxidative stress-induced injury.


Subject(s)
Antioxidants/metabolism , Carbonic Anhydrase III/metabolism , Cell Death/physiology , Cell Hypoxia/physiology , Nucleus Pulposus/metabolism , Oxidative Stress/physiology , Adult , Analysis of Variance , Animals , Biomarkers/metabolism , Carbonic Anhydrase III/genetics , Caspase 3/metabolism , Cells, Cultured , Female , Gene Silencing , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Knockout , Nucleus Pulposus/embryology , Rats , Reactive Oxygen Species/metabolism
17.
FASEB J ; 32(1): 440-452, 2018 01.
Article in English | MEDLINE | ID: mdl-28928248

ABSTRACT

Osteocytes are master orchestrators of bone remodeling; they control osteoblast and osteoclast activities both directly via cell-to-cell communication and indirectly via secreted factors, and they are the main postnatal source of sclerostin and RANKL (receptor activator of NF-kB ligand), two regulators of osteoblast and osteoclast function. Despite progress in understanding osteocyte biology and function, much remains to be elucidated. Recently developed osteocytic cell lines-together with new genome editing tools-has allowed a closer look at the biology and molecular makeup of these cells. By using single-cell cloning, we identified genes that are associated with high Sost/sclerostin expression and analyzed their regulation and function. Unbiased transcriptome analysis of high- vs. low-Sost/sclerostin-expressing cells identified known and novel genes. Dmp1 (dentin matrix protein 1), Dkk1 (Dickkopf WNT signaling pathway inhibitor 1), and Phex were among the most up-regulated known genes, whereas Srpx2, Cd200, and carbonic anhydrase III (CAIII) were identified as novel markers of differentiated osteocytes. Aspn, Enpp2, Robo2, Nov, and Serpina3g were among the transcripts that were most significantly suppressed in high-Sost cells. Considering that CAII was recently identified as being regulated by Sost/sclerostin and capable of controlling mineral homeostasis, we focused our attention on CAIII. Here, we report that CAIII is highly expressed in osteocytes, is regulated by parathyroid hormone both in vitro and in vivo, and protects osteocytes from oxidative stress.-Shi, C., Uda, Y., Dedic, C., Azab, E., Sun, N., Hussein, A. I., Petty, C. A., Fulzele, K., Mitterberger-Vogt, M. C., Zwerschke, W., Pereira, R., Wang, K., Divieti Pajevic, P. Carbonic anhydrase III protects osteocytes from oxidative stress.


Subject(s)
Carbonic Anhydrase III/metabolism , Osteocytes/metabolism , Oxidative Stress , Adaptor Proteins, Signal Transducing , Animals , Bone Remodeling/genetics , Bone Remodeling/physiology , Carbonic Anhydrase III/genetics , Cell Line , Cell Survival , Glycoproteins/genetics , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins , Mice , Osteocytes/cytology , Osteocytes/drug effects , Teriparatide/pharmacology , Transcriptome
18.
Curr Mol Med ; 17(4): 258-269, 2017 Dec 07.
Article in English | MEDLINE | ID: mdl-29110605

ABSTRACT

BACKGROUND: Glutathione is a small antioxidant peptide in cells and it plays an important role in maintaining a reducing intracellular environment. Glutathione is also involved in the dynamic regulation of specific protein functions by reversible glutathiolation of certain proteins in response to oxidative stress. OBJECTIVE: The purpose of this work is to mechanistically investigate the effects of glutathiolation on the susceptibility of proteins to degradation by the ubiquitinproteasome pathway (UPP). METHODS AND RESULTS: The data show that γC-crystallin and carbonic anhydrase III were barely degraded by the UPP without modifications, but both were rapidly degraded by the UPP after glutathiolation. Modifications of sulfhydryls by other thiol-modification reagents, such as iodoacetamide, also increased the degradation of γC-crystallin, but not as effectively as glutathiolation. Biophysical analysis showed that glutathiolation caused reversible conformational changes of these proteins, including a significant increase in protein surface hydrophobicity and a decrease in thermal stability. The modified protein regained its native conformation and its resistance to degradation upon removal of the glutathione moiety. A cataract-causing T5P mutant γC-crystallin shares many biophysical characteristics as glutathiolated γC-crystallin, including increased surface hydrophobicity and decreased thermal stability. T5P mutant γC-crystallin was also rapidly degraded. Comparison of the conformational changes and the susceptibility to degradation of glutathiolated γC-crystallin with other forms of modified γC-crystallin suggests that the glutathiolation-induced exposure of hydrophobic patches, rather than the modification per se, serves as the signal for degradation by the UPP. Consistent with this hypothesis, masking the surface hydrophobicity of glutathiolated and T5P mutant γC-crystallins significantly reduced their susceptibility to degradation by the UPP. CONCLUSION: This work demonstrates that glutathiolation is a novel mechanism for the UPP to recognize substrates in response to oxidative stress.


Subject(s)
Carbonic Anhydrase III/chemistry , Proteasome Endopeptidase Complex/chemistry , Protein Processing, Post-Translational , Proteolysis , Ubiquitin/chemistry , gamma-Crystallins/chemistry , Carbonic Anhydrase III/metabolism , Glutathione , Humans , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , gamma-Crystallins/metabolism
19.
Mol Med Rep ; 16(6): 8289-8294, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28983629

ABSTRACT

Carbonic anhydrase III (CAIII) is expressed selectively in type I (slow­twitch) myofibers. To investigate the association between changes in the expression of CAIII and skeletal muscle structure following denervation, the present study stained adjacent sections of skeletal muscle for ATPase and immunohistochemically for CAIII. In addition, differences in the protein expression and phosphatase activity of CAIII were examined by western blot and phosphatase staining between rat soleus and extensol digitorum longus (EDL) muscles, which are composed of predominantly slow­ and fast­twitch fibers, respectively. Upon denervation, the EDL muscle showed more pronounced structural changes, compared with the soleus muscle. There was a transformation from fast to slow fibers, and a concomitant increase in fibers positive for CAIII. Following denervation, the protein expression of CAIII initially increased and then decreased in the soleus muscle, whereas the protein expression of CAIII in the EDL muscle increased gradually with time. In contrast to the protein changes, phosphatase activity in the soleus and EDL muscles decreased significantly following denervation. These results indicated that, following denervation, changes in the expression of CAIII were associated with myofiber remodeling. Specifically, the change in the expression of CAIII reflected the conversion to type I myofibers, suggesting the importance of CAIII in resistance to fatigue in skeletal muscle.


Subject(s)
Carbonic Anhydrase III/metabolism , Denervation , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Adenosine Triphosphatases/metabolism , Animals , Carbonic Anhydrase III/genetics , Gene Expression , Immunohistochemistry , Male , Muscle, Skeletal/pathology , Rats
20.
Autophagy ; 13(11): 1981-1994, 2017.
Article in English | MEDLINE | ID: mdl-28933591

ABSTRACT

Myasthenia gravis is an autoimmune disorder of the neuromuscular junction manifested as fatigable muscle weakness, which is typically caused by pathogenic autoantibodies against postsynaptic CHRN/AChR (cholinergic receptor nicotinic) in the endplate of skeletal muscle. Our previous studies have identified CA3 (carbonic anhydrase 3) as a specific protein insufficient in skeletal muscle from myasthenia gravis patients. In this study, we investigated the underlying mechanism of how CA3 insufficiency might contribute to myasthenia gravis. Using an experimental autoimmune myasthenia gravis animal model and the skeletal muscle cell C2C12, we find that inhibition of CAR3 (the mouse homolog of CA3) promotes CHRN internalization via a lipid raft-mediated pathway, leading to accelerated degradation of postsynaptic CHRN. Activation of CAR3 reduces CHRN degradation by suppressing receptor endocytosis. CAR3 exerts this effect by suppressing chaperone-assisted selective autophagy via interaction with BAG3 (BCL2-associated athanogene 3) and by dampening endoplasmic reticulum stress. Collectively, our study illustrates that skeletal muscle cell CAR3 is critical for CHRN homeostasis in the neuromuscular junction, and its deficiency leads to accelerated degradation of CHRN and development of myasthenia gravis, potentially revealing a novel therapeutic approach for this disorder.


Subject(s)
Autophagy , Carbonic Anhydrase III/metabolism , Endocytosis , Myasthenia Gravis/enzymology , Receptors, Nicotinic/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Cells, Cultured , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...