201
|
Morishita A, Tadokoro T, Fujihara S, Iwama H, Oura K, Fujita K, Tani J, Takuma K, Nakahara M, Shi T, Haba R, Okano K, Nishiyama A, Ono M, Himoto T, Masaki T. Ipragliflozin attenuates non-alcoholic steatohepatitis development in an animal model. PLoS One 2022; 17:e0261310. [PMID: 35192632 PMCID: PMC8863244 DOI: 10.1371/journal.pone.0261310] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/30/2021] [Indexed: 01/02/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a common chronic liver disease with no decisive treatment. The sodium glucose cotransporter 2 (SGLT2) inhibitor ipragliflozin was developed as a new oral hypoglycemic drug, which can improve NASH via an insulin-independent glucose-lowering effect by inhibiting glucose reabsorption in the renal proximal tubules. However, ipragliflozin appears to modulate steatosis or inflammation via different pathways. To elucidate the new mechanism of ipragliflozin for the treatment of NASH, we evaluated its effects in a NASH mouse model (STAM mice) with beta cell depletion, and compared the expression of microRNAs (miRNAs) in STAM mice treated with or without ipragliflozin (16.7 μg/day for 5 weeks). Ipragliflozin reduced aspartate transaminase and alanine aminotransferase levels, along with reduced hepatic steatosis, hepatocyte ballooning, lobular inflammation, and liver fibrosis. In addition, ipragliflozin upregulated mitochondrial transport-related and antioxidant defensive system-related genes in the liver. Among 2555 mouse miRNA probes, miR-19b-3p was commonly differentially expressed with ipragliflozin treatment for 5 weeks in both the liver and serum but in different directions, with a decrease in the liver and increase in the serum. Therefore, ipragliflozin can improve NASH development likely through the antioxidative stress pathway and by regulating miR-19b-3p.
Collapse
Affiliation(s)
- Asahiro Morishita
- Department of Gastroenterology and Neurology, Kagawa, Japan
- * E-mail:
| | | | | | | | - Kyoko Oura
- Department of Gastroenterology and Neurology, Kagawa, Japan
| | - Koji Fujita
- Department of Gastroenterology and Neurology, Kagawa, Japan
| | - Joji Tani
- Department of Gastroenterology and Neurology, Kagawa, Japan
| | - Kei Takuma
- Department of Gastroenterology and Neurology, Kagawa, Japan
| | - Mai Nakahara
- Department of Gastroenterology and Neurology, Kagawa, Japan
| | - Tingting Shi
- Department of Gastroenterology and Neurology, Kagawa, Japan
| | | | - Keiichi Okano
- Department of Gastroenterological Surgery, Kagawa, Japan
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University Faculty of Medicine, Kagawa, Japan
| | - Masafumi Ono
- Department of Gastroenterology and Neurology, Kagawa, Japan
| | - Takashi Himoto
- Department of Medical Technology, Kagawa Prefectual University of Health Sciences, Kagawa, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Kagawa, Japan
| |
Collapse
|
202
|
Salas JR, Clark PM. SIGNALING PATHWAYS THAT DRIVE 18F-FDG ACCUMULATION IN CANCER. J Nucl Med 2022; 63:659-663. [PMID: 35241480 DOI: 10.2967/jnumed.121.262609] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/22/2022] [Indexed: 11/16/2022] Open
Abstract
2-18F-fluoro-2-deoxy-D-glucose (18F-FDG) measures glucose consumption and is an integral part of cancer management. Most cancer types upregulate their glucose consumption, yielding elevated 18F-FDG PET accumulation in those cancer cells. The biochemical pathway through which 18F-FDG accumulates in cancer cells is well-established. However, beyond well-known regulators such as c-Myc, PI3K/Akt, and HIF1α, the proteins and signaling pathways that cancer cells modulate to activate the facilitated glucose transporters (GLUTs) and hexokinase enzymes that drive elevated 18F-FDG accumulation are less well-understood. Understanding these signaling pathways could yield additional biological insights from 18F-FDG PET scans and could suggest new uses of 18F-FDG PET in the management of cancer. Work over the past five years, building on studies from years prior, has identified new proteins and signaling pathways that drive glucose consumption in cancer. Here we review these recent studies and discuss current limitations to our understanding of glucose consumption in cancer.
Collapse
Affiliation(s)
| | - Peter M Clark
- University of California, Los Angeles, United States
| |
Collapse
|
203
|
Barreto YB, Alencar AM. Random-walk model of the sodium-glucose transporter SGLT2 with stochastic steps and inhibition. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2022; 34:184004. [PMID: 35090150 DOI: 10.1088/1361-648x/ac4fea] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/28/2022] [Indexed: 06/14/2023]
Abstract
Random-walk models are frequently used to model distinct natural phenomena such as diffusion processes, stock-market fluctuations, and biological systems. Here, we present a random-walk model to describe the dynamics of glucose uptake by the sodium-glucose transporter of type 2, SGLT2. Our starting point is the canonical alternating-access model, which suggests the existence of six states for the transport cycle. We propose the inclusion of two new states to this canonical model. The first state is added to implement the recent discovery that the Na+ion can exit before the sugar is released into the proximal tubule epithelial cells. The resulting model is a seven-state mechanism with stochastic steps. Then we determined the transition probabilities between these seven states and used them to write a set of master equations to describe the time evolution of the system. We showed that our model converges to the expected equilibrium configuration and that the binding of Na+and glucose to SGLT2 in the inward-facing conformation must be necessarily unordered. After that, we added another state to implement inhibition in the model. Our results reproduce the experimental dependence of glucose uptake on the inhibitor concentration and they reveal that the inhibitors act by decreasing the number of available SGLT2s, which increases the chances of glucose escaping reabsorption.
Collapse
Affiliation(s)
- Yan B Barreto
- Instituto de Física, Universidade de São Paulo, 05508-090 São Paulo, São Paulo, Brazil
| | - Adriano M Alencar
- Instituto de Física, Universidade de São Paulo, 05508-090 São Paulo, São Paulo, Brazil
| |
Collapse
|
204
|
Rhodes RS, Singh SK, Rajendran VM, Walk ST, Coon SD. Regulation of Glucose Insulinotropic Peptide and Intestinal Glucose Transporters in the Diet-Induced Obese Mouse. J Diabetes Res 2022; 2022:5636499. [PMID: 35224107 PMCID: PMC8872650 DOI: 10.1155/2022/5636499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/12/2022] [Accepted: 01/15/2022] [Indexed: 12/13/2022] Open
Abstract
Our recent studies have shown that glucose-dependent insulinotropic polypeptide (GIP), but not glucagon-like peptide 1 (GLP-1), augments Na-glucose transporter 1- (SGLT1-) mediated glucose absorption in mouse jejunum. Na-dependent glucose absorption sharply rose and peaked in 3 months of high-fat (i.e., obese) compared to normal (i.e., normal weight) diet fed animals. Previous studies have shown that GIP-augmented SGLT1 and PEPT1 (peptide transporter 1) are regulated by protein kinase A (PKA) signaling in mouse jejunum. Additional studies have indicated that cAMP and PI3 kinase signaling augment PEPT1 through EPAC and AKT activation pathways, respectively, through increased apical PEPT1 trafficking in intestinal epithelial cells. However, little is known about how the signaling glucose transport paradigm is altered over a long period. Early on, increased glucose absorption occurs through SGLT1, but as the obesity and diabetes progress, there is a dramatic shift towards a Na-independent mechanism. Surprisingly, at the peak of glucose absorption during the fifth month of the progression of obesity, the SGLT1 activity was severely depressed, while a Na-independent glucose absorptive process begins to appear. Since glucose transporter 2 (GLUT2) is expressed on the apical membrane of the small intestine in obese patients and animal models of obesity, it was hypothesized to be the new more efficient route. Western blot analyses and biotinylation of the apical membrane revealed that the GIP expression increases in the obese animals and its trafficking to the apical membrane increases with the GIP treatment.
Collapse
Affiliation(s)
| | - Satish K. Singh
- Section of Gastroenterology, Boston University School of Medicine, USA
- Section of Gastroenterology Veterans Affairs Boston Healthcare System Boston, Massachusetts, USA
| | - Vazhaikkurichi M. Rajendran
- Department of Biochemistry Robert C Byrd Health Sciences Center and Section of Digestive Diseases, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Seth T. Walk
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Steven D. Coon
- Fort Peck Community College, Poplar, Montana, USA
- Section of Gastroenterology, Boston University School of Medicine, USA
- Section of Gastroenterology Veterans Affairs Boston Healthcare System Boston, Massachusetts, USA
- Boston University Clinical and Translational Science Institute, Boston, Massachusetts, USA
| |
Collapse
|
205
|
Berghaus C, Groh AC, Breljak D, Ciarimboli G, Sabolić I, Pavenstädt H, Weide T. Impact of Pals1 on Expression and Localization of Transporters Belonging to the Solute Carrier Family. Front Mol Biosci 2022; 9:792829. [PMID: 35252349 PMCID: PMC8888964 DOI: 10.3389/fmolb.2022.792829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Pals1 is part of the evolutionary conserved Crumbs polarity complex and plays a key role in two processes, the formation of apicobasal polarity and the establishment of cell-cell contacts. In the human kidney, up to 1.5 million nephrons control blood filtration, as well as resorption and recycling of inorganic and organic ions, sugars, amino acids, peptides, vitamins, water and further metabolites of endogenous and exogenous origin. All nephron segments consist of polarized cells and express high levels of Pals1. Mice that are functionally haploid for Pals1 develop a lethal phenotype, accompanied by heavy proteinuria and the formation of renal cysts. However, on a cellular level, it is still unclear if reduced cell polarization, incomplete cell-cell contact formation, or an altered Pals1-dependent gene expression accounts for the renal phenotype. To address this, we analyzed the transcriptomes of Pals1-haploinsufficient kidneys and the littermate controls by gene set enrichment analysis. Our data elucidated a direct correlation between TGFβ pathway activation and the downregulation of more than 100 members of the solute carrier (SLC) gene family. Surprisingly, Pals1-depleted nephrons keep the SLC's segment-specific expression and subcellular distribution, demonstrating that the phenotype is not mainly due to dysfunctional apicobasal cell polarization of renal epithelia. Our data may provide first hints that SLCs may act as modulating factors for renal cyst formation.
Collapse
Affiliation(s)
- Carmen Berghaus
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Münster, Germany
| | - Ann-Christin Groh
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Münster, Germany
| | - Davorka Breljak
- Molecular Toxicology, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Giuliano Ciarimboli
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Münster, Germany
| | - Ivan Sabolić
- Molecular Toxicology, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Hermann Pavenstädt
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Münster, Germany
| | - Thomas Weide
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Münster, Germany
| |
Collapse
|
206
|
Elferink H, Titulaer WHC, Derks MGN, Veeneman GH, Rutjes FPJT, Boltje TJ. Chloromethyl Glycosides as Versatile Synthons to Prepare Glycosyloxymethyl-Prodrugs. Chemistry 2022; 28:e202103910. [PMID: 35045197 PMCID: PMC9304170 DOI: 10.1002/chem.202103910] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Indexed: 11/21/2022]
Abstract
This work investigates the addition of monosaccharides to marketed drugs to improve their pharmacokinetic properties for oral absorption. To this end, a set of chloromethyl glycoside synthons were developed to prepare a variety of glycosyloxymethyl-prodrugs derived from 5-fluorouracil, thioguanine, propofol and losartan. Drug release was studied in vitro using β-glucosidase confirming rapid conversion of the monosaccharide prodrugs to release the parent drug, formaldehyde and the monosaccharide. To showcase this prodrug approach, a glucosyloxymethyl conjugate of the tetrazole-containing drug losartan was used for in vivo experiments and showed complete release of the drug in a dog-model.
Collapse
Affiliation(s)
- Hidde Elferink
- Synthetic Organic Chemistry Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525AJ NijmegenThe Netherlands
| | - Willem H. C. Titulaer
- Synthetic Organic Chemistry Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525AJ NijmegenThe Netherlands
| | - Maik G. N. Derks
- Synthetic Organic Chemistry Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525AJ NijmegenThe Netherlands
| | | | - Floris P. J. T. Rutjes
- Synthetic Organic Chemistry Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525AJ NijmegenThe Netherlands
| | - Thomas J. Boltje
- Synthetic Organic Chemistry Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525AJ NijmegenThe Netherlands
| |
Collapse
|
207
|
Wu H, Liu S, Su P, Xie Z, Gui T, Zhao L, Liu Y, Chen L. Molecular insight into coordination sites for substrates and their coupling kinetics in Na
+
/HCO
3
−
cotransporter NBCe1. J Physiol 2022; 600:3083-3111. [DOI: 10.1113/jp282034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 02/03/2022] [Indexed: 11/08/2022] Open
Affiliation(s)
- Han Wu
- Key Laboratory of Molecular Biophysics of Ministry of Education School of Life Science & Technology Huazhong University of Science & Technology Wuhan 430074 China
| | - Shiyong Liu
- School of Physics Huazhong University of Science and Technology Wuhan 430074 China
| | - Pan Su
- Key Laboratory of Molecular Biophysics of Ministry of Education School of Life Science & Technology Huazhong University of Science & Technology Wuhan 430074 China
| | - Zhang‐Dong Xie
- Key Laboratory of Molecular Biophysics of Ministry of Education School of Life Science & Technology Huazhong University of Science & Technology Wuhan 430074 China
| | - Tian‐Xiang Gui
- Key Laboratory of Molecular Biophysics of Ministry of Education School of Life Science & Technology Huazhong University of Science & Technology Wuhan 430074 China
| | - Lei Zhao
- Department of Obstetrics Maternal and Child Health Hospital of Hubei Province Wuhan 430070 China
| | - Ying Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education School of Life Science & Technology Huazhong University of Science & Technology Wuhan 430074 China
| | - Li‐Ming Chen
- Key Laboratory of Molecular Biophysics of Ministry of Education School of Life Science & Technology Huazhong University of Science & Technology Wuhan 430074 China
| |
Collapse
|
208
|
Weinrauch AM, Clifford AM, Folkerts EJ, Schaefer CM, Giacomin M, Goss GG. Molecular identification and post-prandial regulation of glucose carrier proteins in the hindgut of Pacific hagfish, Eptatretus stoutii. Am J Physiol Regul Integr Comp Physiol 2022; 322:R336-R345. [PMID: 35138949 DOI: 10.1152/ajpregu.00003.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hagfish are an excellent model species in which to draw inferences on the evolution of transport systems in early-vertebrates owing to their basal position in vertebrate phylogeny. Glucose is a ubiquitous cellular energy source that is transported into cells via two classes of carrier proteins: sodium-glucose linked transporters (Sglt; Slc5a) and glucose transporters (Glut; Slc2a). While previous pharmacological evidence has suggested the presence of both sodium-dependent and -independent transport mechanisms in the hagfish, the molecular identities were heretofore unconfirmed. We have identified and phylogenetically characterized both a Slc5a1-like and Slc2a-like gene in the Pacific hagfish (Eptatretus stoutii); the latter sharing common ancestry with other glucose-transporting isoforms of the Slc2a family. To assess the potential post-prandial regulation of these glucose transporters, we examined the abundance and localization of these transporters with qPCR and immunohistochemistry alongside functional studies using radiolabeled 14C-D-glucose. The effects of glucose- or insulin-injection on glucose transport rate and transporter expression were also examined to determine their potential role(s) in the regulation of intestinal glucose carrier proteins. Feeding prompted an increase in glucose uptake across the hindgut at both 0.5 mM (~84%) and 1 mM (~183%) concentrations. Concomitant increases were observed in hindgut Slc5a1 protein expression. These effects were not observed following either of glucose- or insulin-injection, indicating these post-prandial factors are not the driving force for transporter regulation over this timeframe. We conclude that Pacific hagfish utilize evolutionarily-conserved mechanisms of glucose uptake and so represent a useful model to understand early vertebrate evolution of glucose uptake and regulation.
Collapse
Affiliation(s)
- Alyssa M Weinrauch
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.,Bamfield Marine Sciences Centre, 100 Pachena Rd, Bamfield, BC, Canada
| | - Alexander M Clifford
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.,Bamfield Marine Sciences Centre, 100 Pachena Rd, Bamfield, BC, Canada.,Marine Biology Research Division, Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA, United States
| | - Erik J Folkerts
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Christina M Schaefer
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.,Bamfield Marine Sciences Centre, 100 Pachena Rd, Bamfield, BC, Canada
| | - Marina Giacomin
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.,Bamfield Marine Sciences Centre, 100 Pachena Rd, Bamfield, BC, Canada
| | - Greg G Goss
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.,Bamfield Marine Sciences Centre, 100 Pachena Rd, Bamfield, BC, Canada
| |
Collapse
|
209
|
Associations of Plasma Concentration Profiles of Dapagliflozin, a Selective Inhibitor of Sodium–Glucose Co-Transporter Type 2, with Its Effects in Japanese Patients with Type 2 Diabetes Mellitus. Pharmaceuticals (Basel) 2022; 15:ph15020203. [PMID: 35215315 PMCID: PMC8880045 DOI: 10.3390/ph15020203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 12/04/2022] Open
Abstract
This study was conducted to evaluate the long-term plasma concentration profiles of dapagliflozin and its effects on the glycated hemoglobin (HbA1c) level, body weight, and estimated glomerular filtration rate (eGFR) in 72 Japanese outpatients with type 2 diabetes mellitus (T2DM) receiving metformin and a dipeptidyl peptidase-4 inhibitor. At baseline, HbA1c level, body weight, and eGFR were 6.9 ± 0.6%, 77.9 ± 13.5 kg, and 78.8 ± 20.7 mL/min/1.73 m2, respectively. A once-daily oral dose of 5 mg dapagliflozin was administered, and its trough plasma concentrations were evaluated at 1, 3, 6, 9, and 12 months. In this study, the patients with stable dapagliflozin concentrations were defined, based on a well-organized clinical trial, as those with average plasma concentrations of 2–5 ng/mL with a coefficient of variation <30%; these values were achieved if patients complied with their once-daily dosage. Multivariate analysis showed a significant decrease in the HbA1c levels among patients with stable concentrations (−0.6 ± 0.4%, p < 0.01), which was greater than the mean change among all 72 patients (−0.2 ± 0.5%, p < 0.01). The patients’ mean body weight also decreased (−2.3 ± 4.0 kg, p = 0.060). Average plasma concentrations ranged from 1.6 to 11.8 ng/mL; however, multivariate analysis indicated it was unrelated to the HbA1c-lowering effect. In conclusion, the long-term stability of plasma dapagliflozin concentration was important in lowering HbA1c level, and a once-daily oral dose of 5 mg was sufficient in achieving this effect.
Collapse
|
210
|
Hailat M, Zakaraya Z, Al-Ani I, Meanazel OA, Al-Shdefat R, Anwer MK, Saadh MJ, Abu Dayyih W. Pharmacokinetics and Bioequivalence of Two Empagliflozin, with Evaluation in Healthy Jordanian Subjects under Fasting and Fed Conditions. Pharmaceuticals (Basel) 2022; 15:ph15020193. [PMID: 35215305 PMCID: PMC8879246 DOI: 10.3390/ph15020193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023] Open
Abstract
The current study is a randomized, open-label, two-period, two-sequence, two-way crossover pharmacokinetic study in healthy Jordanian subjects to evaluate the pharmacokinetics and bioequivalence profile of two cases of empagliflozin 10 mg under fasting and fed conditions. The plasma concentrations of empagliflozin were determined using an HPLC-MS/MS method. Tolerability and safety were assessed throughout the study. This study included 26 subjects, 26 in both fasting and fed groups.The pharmacokinetic parameters, which included the area under the concentration–time curve from time zero to infinity (AUC0–inf) and the final quantifiable concentration (AUC0–last), maximum serum concentration (Cmax), and time to reach the maximum drug concentration (Tmax) were found to be within an equivalence margin of 80.00–125.00%. The pharmacokinetic profiles show that the empagliflozin test and parent reference cases were bioequivalent in healthy subjects. The two treatments’ safety evaluations were also comparable.
Collapse
Affiliation(s)
- Mohammad Hailat
- Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan;
| | - Zainab Zakaraya
- Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan; (Z.Z.); (I.A.-A.)
| | - Israa Al-Ani
- Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan; (Z.Z.); (I.A.-A.)
| | - Osaid Al Meanazel
- Michael Sayegh Faculty of Pharmacy, Aqaba University of Technology, Aqaba 77110, Jordan;
| | | | - Md. Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Mohamed J. Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan;
| | - Wael Abu Dayyih
- Faculty of Pharmacy, Mutah University, Al-Karak 61710, Jordan
- Correspondence:
| |
Collapse
|
211
|
Schicker K, Farr CV, Boytsov D, Freissmuth M, Sandtner W. Optimizing the Substrate Uptake Rate of Solute Carriers. Front Physiol 2022; 13:817886. [PMID: 35185619 PMCID: PMC8850955 DOI: 10.3389/fphys.2022.817886] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/10/2022] [Indexed: 11/16/2022] Open
Abstract
The diversity in solute carriers arose from evolutionary pressure. Here, we surmised that the adaptive search for optimizing the rate of substrate translocation was also shaped by the ambient extracellular and intracellular concentrations of substrate and co-substrate(s). We explored possible solutions by employing kinetic models, which were based on analytical expressions of the substrate uptake rate, that is, as a function of the microscopic rate constants used to parameterize the transport cycle. We obtained the defining terms for five reaction schemes with identical transport stoichiometry (i.e., Na+: substrate = 2:1). We then utilized an optimization algorithm to find the set of numeric values for the microscopic rate constants, which provided the largest value for the substrate uptake rate: The same optimized rate was achieved by different sets of numerical values for the microscopic rate constants. An in-depth analysis of these sets provided the following insights: (i) In the presence of a low extracellular substrate concentration, a transporter can only cycle at a high rate, if it has low values for both, the Michaelis-Menten constant (KM) for substrate and the maximal substrate uptake rate (Vmax). (ii) The opposite is true for a transporter operating at high extracellular substrate concentrations. (iii) Random order of substrate and co-substrate binding is superior to sequential order, if a transporter is to maintain a high rate of substrate uptake in the presence of accumulating intracellular substrate. Our kinetic models provide a framework to understand how and why the transport cycles of closely related transporters differ.
Collapse
Affiliation(s)
| | | | | | | | - Walter Sandtner
- Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
212
|
Bazzone A, Körner A, Meincke M, Bhatt M, Dondapati S, Barthmes M, Kubick S, Fertig N. SSM-based electrophysiology, a label-free real-time method reveals sugar binding & transport events in SGLT1. Biosens Bioelectron 2022; 197:113763. [PMID: 34768066 DOI: 10.1016/j.bios.2021.113763] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/26/2021] [Accepted: 10/30/2021] [Indexed: 01/26/2023]
Abstract
Here, we present a solid-supported membrane (SSM)-based electrophysiological approach to study sugar binding and Na+/glucose cotransport by SGLT1 in membrane vesicles. SSM-based electrophysiology delivers a cumulative real-time current readout from numerous SGLT1 proteins simultaneously using a gold-coated sensor chip. In contrast to conventional techniques, which mainly operate with voltage steps, currents are triggered by sugar or sodium addition. Sugar concentration jumps in the presence of sodium lead to transport currents between 5 and 10 nA. Remarkably, in the absence of sodium (i.e. no transport), we observed fast pre-steady-state (PSS) currents with time constants between 3 and 10 ms. These PSS currents mainly originate from sugar binding. Sodium binding does not induce PSS currents. Due to high time resolution, PSS currents were distinguished from transport and eventually correlated with conformational transitions within the sugar translocation pathway. In addition, we analyzed the impact of driving forces on transport and binding currents, showing that membrane voltage and sodium concentration gradients lead to an increased transport rate without affecting sugar binding kinetics. We also compared Na+/sugar efflux with physiologically relevant influx and found similar transport rates, but lower affinity in efflux mode. SSM-based electrophysiology is a powerful technique, which overcomes bottlenecks for transport measurements observed in other techniques such as the requirement of labels or the lack of real-time data. Rapid solution exchange enables the observation of substrate-induced electrogenic events like conformational transitions, opening novel perspectives for in-depth functional studies of SGLT1 and other transporters.
Collapse
Affiliation(s)
- Andre Bazzone
- Nanion Technologies GmbH, Ganghoferstr. 70a, 80339, Munich, Germany.
| | - Alexander Körner
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Am Mühlenberg 13, 14476, Potsdam, Germany; Institute of Biotechnology, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355, Berlin, Germany
| | - Melanie Meincke
- Nanion Technologies GmbH, Ganghoferstr. 70a, 80339, Munich, Germany
| | - Manan Bhatt
- Nanion Technologies GmbH, Ganghoferstr. 70a, 80339, Munich, Germany
| | - Srujan Dondapati
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Am Mühlenberg 13, 14476, Potsdam, Germany
| | - Maria Barthmes
- Nanion Technologies GmbH, Ganghoferstr. 70a, 80339, Munich, Germany
| | - Stefan Kubick
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Am Mühlenberg 13, 14476, Potsdam, Germany; Freie Universität Berlin, Institute of Chemistry and Biochemistry - Biochemistry, 14195, Berlin, Germany; Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane and the University of Potsdam, Germany
| | - Niels Fertig
- Nanion Technologies GmbH, Ganghoferstr. 70a, 80339, Munich, Germany
| |
Collapse
|
213
|
Kim M, Eleftheriou A, Ravotto L, Weber B, Rivlin M, Navon G, Capozza M, Anemone A, Longo DL, Aime S, Zaiss M, Herz K, Deshmane A, Lindig T, Bender B, Golay X. What do we know about dynamic glucose-enhanced (DGE) MRI and how close is it to the clinics? Horizon 2020 GLINT consortium report. MAGMA (NEW YORK, N.Y.) 2022; 35:87-104. [PMID: 35032288 PMCID: PMC8901523 DOI: 10.1007/s10334-021-00994-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/27/2022]
Abstract
Cancer is one of the most devastating diseases that the world is currently facing, accounting for 10 million deaths in 2020 (WHO). In the last two decades, advanced medical imaging has played an ever more important role in the early detection of the disease, as it increases the chances of survival and the potential for full recovery. To date, dynamic glucose-enhanced (DGE) MRI using glucose-based chemical exchange saturation transfer (glucoCEST) has demonstrated the sensitivity to detect both d-glucose and glucose analogs, such as 3-oxy-methyl-d-glucose (3OMG) uptake in tumors. As one of the recent international efforts aiming at pushing the boundaries of translation of the DGE MRI technique into clinical practice, a multidisciplinary team of eight partners came together to form the “glucoCEST Imaging of Neoplastic Tumors (GLINT)” consortium, funded by the Horizon 2020 European Commission. This paper summarizes the progress made to date both by these groups and others in increasing our knowledge of the underlying mechanisms related to this technique as well as translating it into clinical practice.
Collapse
Affiliation(s)
- Mina Kim
- Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK.,Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | - Afroditi Eleftheriou
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Luca Ravotto
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, Zurich, Switzerland
| | - Michal Rivlin
- School of Chemistry, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Gil Navon
- School of Chemistry, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Martina Capozza
- Molecular Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Annasofia Anemone
- Molecular Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Torino, Italy
| | - Silvio Aime
- Molecular Imaging Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Moritz Zaiss
- Magnetic Resonance Center, Max Planck Institute for Biological Cybernetics, Tübingen, Germany.,Neuroradiology, University Clinic Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Kai Herz
- Magnetic Resonance Center, Max Planck Institute for Biological Cybernetics, Tübingen, Germany.,Department of Biomedical Magnetic Resonance, University of Tübingen, Tübingen, Germany
| | - Anagha Deshmane
- Magnetic Resonance Center, Max Planck Institute for Biological Cybernetics, Tübingen, Germany.,Department of Biomedical Magnetic Resonance, University of Tübingen, Tübingen, Germany
| | - Tobias Lindig
- Magnetic Resonance Center, Max Planck Institute for Biological Cybernetics, Tübingen, Germany.,Department of Diagnostic and Interventional Neuroradiology, University Hospital Tübingen, Tübingen, Germany
| | - Benjamin Bender
- Department of Diagnostic and Interventional Neuroradiology, University Hospital Tübingen, Tübingen, Germany
| | - Xavier Golay
- Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK.
| | | |
Collapse
|
214
|
Cariou B. The metabolic triad of non-alcoholic fatty liver disease, visceral adiposity and type 2 diabetes: Implications for treatment. Diabetes Obes Metab 2022; 24 Suppl 2:15-27. [PMID: 35014161 DOI: 10.1111/dom.14651] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/06/2022] [Accepted: 01/06/2022] [Indexed: 12/11/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is associated with visceral obesity, insulin resistance, type 2 diabetes (T2D) and has been often considered as the hepatic expression of the metabolic syndrome (MetS). Epidemiological studies highlight a bidirectional relationship of NAFLD with T2D in which NAFLD increases the risk of incident T2D and T2D increases the risk of severe non-alcoholic steatohepatitis (NASH) and liver fibrosis. Regarding the molecular determinants of NAFLD, we specifically focused in this review on adipocyte dysfunction as a key molecular link between visceral adipose tissue, MetS and NAFLD. Notably, the subcutaneous white adipose tissue expandability appears a critical adaptive buffering mechanism to prevent lipotoxicity and its related metabolic complications, such as NAFLD and T2D. There is a clinical challenge to consider therapeutic strategies targeting the metabolic dysfunction common to NASH and T2D pathogenesis. Strategies that promote significant and sustained weight loss (~10% of total body weight) such as metabolic and bariatric surgery or incretin-based therapies (GLP-1 receptor agonists or dual GLP-1/GIP or GLP-1/glucagon receptor co-agonists) are among the most efficient ones. In addition, insulin sensitizers such as PPARγ (pioglitazone) and pan-PPARs agonists (lanifibranor) have shown some beneficial effects on both NASH and liver fibrosis. Since NASH is a complex and multifactorial disease, it is conceivable that targeting different pathways, not only insulin resistance but also inflammation and fibrotic processes, is required to achieve NASH resolution.
Collapse
Affiliation(s)
- Bertrand Cariou
- Université de Nantes, Inserm, CNRS, CHU Nantes, l'institut du thorax, Nantes, France
| |
Collapse
|
215
|
Bamba R, Okamura T, Hashimoto Y, Majima S, Senmaru T, Ushigome E, Nakanishi N, Asano M, Yamazaki M, Takakuwa H, Hamaguchi M, Fukui M. Extracellular lipidome change by an SGLT2 inhibitor, luseogliflozin, contributes to prevent skeletal muscle atrophy in db/db mice. J Cachexia Sarcopenia Muscle 2022; 13:574-588. [PMID: 34854254 PMCID: PMC8818690 DOI: 10.1002/jcsm.12814] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 08/18/2021] [Accepted: 09/04/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Diabetes mellitus increases the excretion of urinary glucose from the renal glomeruli due to elevated blood glucose levels. In the renal tubules, SGLT2 is expressed and reabsorbs the excreted urinary glucose. In the pathogenesis of diabetes mellitus, glucose reabsorption by SGLT2 is increased, and SGLT2 inhibitors improve hyperglycaemia by inhibiting this reabsorption. When urinary glucose excretion is enhanced, glucose supply to skeletal muscle may be insufficient and muscle protein catabolism may be accelerated. On the other hand, SGLT2 inhibitors not only ameliorate hyperglycaemia but also improve fatty acid metabolism in muscle, which may prevent muscle atrophy. METHODS Eight-week-old male db/m mice or db/db mice were fed a standard diet with or without the SGLT2i luseogliflozin (0.01% w/w in chow) for 8 weeks. Mice were sacrificed at 16 weeks of age, and skeletal muscle and serum lipidomes, as well as skeletal muscle transcriptome, were analysed. RESULTS Administration of SGLT2i led to not only decreased visceral fat accumulation (P = 0.004) but also increased soleus muscle weight (P = 0.010) and grip strength (P = 0.0001). The levels of saturated fatty acids, especially palmitic acid, decreased in both muscles (P = 0.017) and sera (P = 0.041) upon administration of SGLT2i, while the content of monosaturated fatty acids, especially oleic acid, increased in both muscle (P < 0.0001) and sera (P = 0.009). Finally, the accumulation of transcripts associated with fatty acid metabolism, such as Scd1, Fasn, and Elovl6, and of muscle atrophy-associated transcripts, such as Foxo1, Mstn, Trim63, and Fbxo32, decreased following SGLT2i administration. CONCLUSIONS Intramuscular fatty acid metabolism and gene expression were influenced by the extracellular lipidome, which was modified by SGLT2i. Hence, secondary effects, other than the hypoglycaemic effects of SGLT2i, might lead to the alleviation of sarcopenia.
Collapse
Affiliation(s)
- Ryo Bamba
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takuro Okamura
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshitaka Hashimoto
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Saori Majima
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takafumi Senmaru
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Emi Ushigome
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Naoko Nakanishi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mai Asano
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masahiro Yamazaki
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroshi Takakuwa
- Agilent Technologies, Chromatography Mass Spectrometry Sales Department, Life Science and Applied Markets Group, Tokyo, Japan
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
216
|
Du J, Gu J, Deng J, Kong L, Guo Y, Jin C, Bao Y, Fu D, Li J. The expression and survival significance of sodium glucose transporters in pancreatic cancer. BMC Cancer 2022; 22:116. [PMID: 35090421 PMCID: PMC8796473 DOI: 10.1186/s12885-021-09060-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 11/25/2021] [Indexed: 12/31/2022] Open
Abstract
Background Sodium glucose transporters (SGLTs) play vital roles in glucose uptake in many solid cancers, including pancreatic cancer (PC). However, their expression profile in pancreatic cancer and correlation with prognosis are not clear. Thus, we aimed to analyse the expression profile and prognostic significance of SGLT-1 and SGLT-2 in PC. Methods Eighty-eight patients with pancreatic ductal adenocarcinoma (PDAC) undergoing surgery in Huashan Hospital, Fudan University, from July 2017 to June 2020 were enrolled in the study. Specimens for immunohistochemistry were obtained through surgical resection. Bioinformatics analysis was performed based on the Gene Expression Omnibus (GEO), Oncomine and The Cancer Genome Atlas (TCGA) databases. The statistics were calculated using IBM SPSS Statistics, version 20 and R 4.1.1. P values lower than 0.05 were considered to indicate statistical significance. Results SGLT-1 but not SGLT-2 was significantly overexpressed in PDAC. Survival analysis showed that the median overall survival (OS) and progression-free survival (PFS) of patients with high SGLT-1 expression were significantly longer than that of patients with low SGLT-1 expression. Cox regression indicated that high SGLT-1 expression was an independent predictor for a better prognosis, while residual tumour status (R1 and R2) was an independent risk factor for a poor prognosis. Finally, PDZK1-interacting protein 1 (PDZK1IP1), a protein participating in the generation of reactive oxygen species, was overexpressed in PDAC and its expression was significantly correlated with SGLT-1. Conclusions SGLT-1 but not SGLT-2 was overexpressed in PDAC, and the overexpression of SGLT-1 could be a predictor of a better prognosis. Residual tumour status (R1 and R2) was a risk factor for poor prognosis and disease progression. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-09060-4.
Collapse
|
217
|
Rizzo MR, Di Meo I, Polito R, Auriemma MC, Gambardella A, di Mauro G, Capuano A, Paolisso G. Cognitive impairment and type 2 diabetes mellitus: Focus of SGLT2 inhibitors treatment. Pharmacol Res 2022; 176:106062. [PMID: 35017046 DOI: 10.1016/j.phrs.2022.106062] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 02/09/2023]
Abstract
Gliflozins are a novel class of oral anti-diabetic drugs, acting as inhibitors of sodium-glucose co-transporters (SGLTs) through the proximal convoluted tubules (PCT) and intestinal epithelium. The sodium-glucose co-transporters 2 (SGLT2) are mainly expressed in S1 and S2 segments of the proximal convoluted tubule in the kidneys. Clinical guidelines recommend their use especially in Type 2 Diabetes mellitus (T2DM) patients with vascular complications and/or heart failure highlighting the importance of sodium-glucose co-transporter 2 inhibitors (SGLT2i) pleiotropic effects. Interestingly, cognitive decline is a widely recognized complication of T2DM and, in addition, to clarify its pathophysiology, there is an urgent need to understand how and if diabetes therapies can control diabetes-related cognitive dysfunction. At the time, although SGLT2 proteins are present in the Central Nervous System (CNS), the SGLT2i effects on cognitive impairments remain partly unknown. In pre-clinical studies, SGLT2i ameliorates cognitive dysfunction in obese and T2DM mice, reducing oxidative stress, neuroinflammation and improving neuronal plasticity and mitochondrial brain pathway. In addition, SGLT2i could bring back mTOR to a physiological state of activation, stopping neurodegenerative diseases' onset or progression. Instead, clinical studies on T2DM-related cognitive dysfunction treated by SGLT2i are much more limited. For these reasons, further studies are needed to better elucidate if SGLT2i therapy can affect T2DM-related cognitive decline. In this scenario, this review aims to summarize the state of knowledge on the role of SGLT2i in T2DM-related cognitive dysfunction and stimulate new clinical trials.
Collapse
Affiliation(s)
- Maria Rosaria Rizzo
- Department of Advanced Medical and Surgical Sciences - University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Irene Di Meo
- Department of Advanced Medical and Surgical Sciences - University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Rita Polito
- Department of Advanced Medical and Surgical Sciences - University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Maria Chiara Auriemma
- Department of Advanced Medical and Surgical Sciences - University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Antonio Gambardella
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Gabriella di Mauro
- Department of Experimental Medicine - Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Annalisa Capuano
- Department of Experimental Medicine - Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences - University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| |
Collapse
|
218
|
Günther I, Rimbach G, Nevermann S, Neuhauser C, Stadlbauer V, Schwarzinger B, Schwarzinger C, Ipharraguerre IR, Weghuber J, Lüersen K. Avens Root ( Geum Urbanum L.) Extract Discovered by Target-Based Screening Exhibits Antidiabetic Activity in the Hen's Egg Test Model and Drosophila melanogaster. Front Pharmacol 2022; 12:794404. [PMID: 34975489 PMCID: PMC8715001 DOI: 10.3389/fphar.2021.794404] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/29/2021] [Indexed: 01/23/2023] Open
Abstract
Medicinal plant extracts are becoming increasingly important as an alternative for traditional drugs against diabetes mellitus (DM). For this reason, we initialized a target-based screening of 111 root extracts from an open access plant extract library (PECKISH) by ascertaining their in-vitro inhibitory efficacy on α-glucosidase. The two most active extracts Geum urbanum L. (roseroot) and Rhodiola rosea L. (avens root) were further tested for their antidiabetic activities in terms of their impact on different regulatory key points of glucose homeostasis. To this end, various enzyme- and cell culture-based in-vitro assays were employed including the determination of sodium-dependent glucose transporter 1 (SGLT1) activity in Caco-2 monolayers by Ussing chambers and of glucose transporter 4 (GLUT4) translocation in a GFP-reporter cell line. Subsequently, the antidiabetic potential of the root extracts were further evaluated in in-vivo models, namely hen’s eggs test and the fruit fly Drosophila melanogaster. Avens root extract was found to be a more potent inhibitor of the enzymes α-glucosidase and dipeptidyl peptidase-4 (DPP4) than roseroot extract. Most importantly, only avens root extract exhibited antidiabetic activity in the two in-vivo models eliciting a reduced blood glucose level in the in-ovo model and a decline of the triglyceride level in a dietary starch-induced D. melanogaster obesity model. Analyses of the polyphenolic composition of the avens root extract by HPLC revealed a high content of ellagic acid and its derivatives as well as ellagitannins such as pedunculagin, stenophyllanin, stachyurin, casuarinin and gemin A. In conclusion, avens root extract represents a promising medicinal plant that should be considered in further in-vivo studies on hyperglycemia in laboratory rodents and humans.
Collapse
Affiliation(s)
- Ilka Günther
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Gerald Rimbach
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Sandra Nevermann
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Cathrina Neuhauser
- School of Engineering, University of Applied Sciences Upper Austria, Wels, Austria
| | - Verena Stadlbauer
- School of Engineering, University of Applied Sciences Upper Austria, Wels, Austria.,FFoQSI - Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
| | - Bettina Schwarzinger
- School of Engineering, University of Applied Sciences Upper Austria, Wels, Austria
| | - Clemens Schwarzinger
- Institute for Chemical Technology of Organic Materials, Johannes Kepler University, Linz, Austria
| | | | - Julian Weghuber
- School of Engineering, University of Applied Sciences Upper Austria, Wels, Austria.,FFoQSI - Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
| | - Kai Lüersen
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| |
Collapse
|
219
|
Abstract
PURPOSE OF REVIEW This review offers a critical narrative evaluation of emerging evidence that sodium-glucose co-transporter-2 (SGLT2) inhibitors exert nephroprotective effects in people with type 2 diabetes. RECENT FINDINGS The SGLT2 inhibitor class of glucose-lowering agents has recently shown beneficial effects to reduce the onset and progression of renal complications in people with and without diabetes. Randomised clinical trials and 'real world' observational studies, mostly involving type 2 diabetes patients, have noted that use of an SGLT2 inhibitor can slow the decline in glomerular filtration rate (GFR), reduce the onset of microalbuminuria and slow or reverse the progression of proteinuria. The nephroprotective effects of SGLT2 inhibitors are class effects observed with each of the approved agents in people with a normal or impaired GFR. These effects are also observed in non-diabetic, lean and normotensive individuals suggesting that the mechanisms extend beyond the glucose-lowering, weight-lowering and blood pressure-lowering effects that accompany their glucosuric action in diabetes patients. A key mechanism is tubuloglomerular feedback in which SGLT2 inhibitors cause more sodium to pass along the nephron: the sodium is sensed by macula cells which act via adenosine to constrict afferent glomerular arterioles, thereby protecting glomeruli by reducing intraglomerular pressure. Other effects of SGLT2 inhibitors improve tubular oxygenation and metabolism and reduce renal inflammation and fibrosis. SGLT2 inhibitors have not increased the risk of urinary tract infections or the risk of acute kidney injury. However, introduction of an SGLT2 inhibitor in patients with a very low GFR is not encouraged due to an initial dip in GFR, and it is prudent to discontinue therapy if there is an acute renal event, hypovolaemia or hypotension.
Collapse
Affiliation(s)
| | - Caroline Day
- Life and Health Sciences, Aston University, Birmingham, B4 7ET, UK
| | - Srikanth Bellary
- Life and Health Sciences, Aston University, Birmingham, B4 7ET, UK
| |
Collapse
|
220
|
Niu Y, Liu R, Guan C, Zhang Y, Chen Z, Hoerer S, Nar H, Chen L. Structural basis of inhibition of the human SGLT2-MAP17 glucose transporter. Nature 2022; 601:280-284. [PMID: 34880493 DOI: 10.1038/s41586-021-04212-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/03/2021] [Indexed: 01/01/2023]
Abstract
Human sodium-glucose cotransporter 2 (hSGLT2) mediates the reabsorption of the majority of filtrated glucose in the kidney1. Pharmacological inhibition of hSGLT2 by oral small-molecule inhibitors, such as empagliflozin, leads to enhanced excretion of glucose and is widely used in the clinic to manage blood glucose levels for the treatment of type 2 diabetes1. Here we determined the cryogenic electron microscopy structure of the hSGLT2-MAP17 complex in the empagliflozin-bound state to an overall resolution of 2.95 Å. Our structure shows eukaryotic SGLT-specific structural features. MAP17 interacts with transmembrane helix 13 of hSGLT2. Empagliflozin occupies both the sugar-substrate-binding site and the external vestibule to lock hSGLT2 in an outward-open conformation, thus inhibiting the transport cycle. Our work provides a framework for understanding the mechanism of SLC5A family glucose transporters and also develops a foundation for the future rational design and optimization of new inhibitors targeting these transporters.
Collapse
Affiliation(s)
- Yange Niu
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China
| | - Rui Liu
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China
| | - Chengcheng Guan
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China
| | - Yuan Zhang
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China
| | - Zhixing Chen
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Stefan Hoerer
- Boehringer-Ingelheim Pharma, GmbH & Co KG, Biberach, Germany
| | - Herbert Nar
- Boehringer-Ingelheim Pharma, GmbH & Co KG, Biberach, Germany
| | - Lei Chen
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China. .,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
221
|
Xi Y, Zhang D, Liang Y, Shan Z, Teng X, Teng W. Proteomic Analysis of the Intestinal Resistance to Thyroid Hormone Mouse Model With Thyroid Hormone Receptor Alpha Mutations. Front Endocrinol (Lausanne) 2022; 13:773516. [PMID: 35574030 PMCID: PMC9095823 DOI: 10.3389/fendo.2022.773516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 03/24/2022] [Indexed: 11/23/2022] Open
Abstract
Thyroid hormone is critical during the development of vertebrates and affects the function of many organs and tissues, especially the intestine. Triiodothyronine (T3) is the active form and can bind to thyroid hormone nuclear receptors (TRs) to play a vital role in the development of vertebrates. The resistance to thyroid hormone α, as seen in patients, has been mimicked by the ThraE403X mutation. To investigate the mechanisms underlying the effect of TRα1 on intestinal development, the present study employed proteomic analysis to identify differentially expressed proteins (DEPs) in the distal ileum between homozygous ThraE403X/E403X and wild-type Thra+/+ mice. A total of 1,189 DEPs were identified, including 603 upregulated and 586 downregulated proteins. Proteomic analysis revealed that the DEPs were highly enriched in the metabolic process, the developmental process, the transporter of the nutrients, and the intestinal immune system-related pathway. Of these DEPs, 20 proteins were validated by parallel reaction monitoring analysis. Our intestinal proteomic results provide promising candidates for future studies, as they suggest novel mechanisms by which TRα1 may influence intestinal development, such as the transport of intestinal nutrients and the establishment of innate and adaptive immune barriers of the intestine.
Collapse
Affiliation(s)
- Yue Xi
- Department of Endocrinology and Metabolism, Endocrine Institute, and Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, China
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Dan Zhang
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yue Liang
- Department of Endocrinology and Metabolism, Endocrine Institute, and Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, China
| | - Zhongyan Shan
- Department of Endocrinology and Metabolism, Endocrine Institute, and Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, China
| | - Xiaochun Teng
- Department of Endocrinology and Metabolism, Endocrine Institute, and Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, China
- *Correspondence: Xiaochun Teng, ; Weiping Teng,
| | - Weiping Teng
- Department of Endocrinology and Metabolism, Endocrine Institute, and Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Hospital of China Medical University, Shenyang, China
- *Correspondence: Xiaochun Teng, ; Weiping Teng,
| |
Collapse
|
222
|
van Ruiten CC, Veltman DJ, Nieuwdorp M, IJzerman RG. Brain Activation in Response to Low-Calorie Food Pictures: An Explorative Analysis of a Randomized Trial With Dapagliflozin and Exenatide. Front Endocrinol (Lausanne) 2022; 13:863592. [PMID: 35600575 PMCID: PMC9114766 DOI: 10.3389/fendo.2022.863592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/31/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND AND AIM Sodium-glucose cotransporter-2 inhibitors (SGLT2i) induce less weight loss than expected. This may be explained by SGLT2i-induced alterations in central reward and satiety circuits, contributing to increased appetite and food intake. This hyperphagia may be specific to high-calorie foods. Glucagon-like peptide-1 receptor agonists (GLP-1RA) are associated with lower preferences for high-calorie foods, and with decreased activation in areas regulating satiety and reward in response to high-calorie food pictures, which may reflect this lower preference for energy-dense foods. To optimize treatment, we need a better understanding of how intake is controlled, and how [(un)healthy] food choices are made. The aim of the study was to investigate the effects of dapagliflozin, exenatide, and their combination on brain activation in response to low-calorie food pictures. METHODS We performed an exploratory analysis of a larger, 16-week, double-blind, randomized, placebo-controlled trial. Sixty-eight subjects with obesity and type 2 diabetes were randomized to dapagliflozin, exenatide, dapagliflozin plus exenatide, or double placebo. Using functional MRI, the effects of treatments on brain responses to low-calorie food pictures were assessed after 10 days and 16 weeks. RESULTS Dapagliflozin versus placebo decreased activity in response to low-calorie food pictures, in the caudate nucleus, insula, and amygdala after 10 days, and in the insula after 16 weeks. Exenatide versus placebo increased activation in the putamen in response to low-calorie food pictures after 10 days, but not after 16 weeks. Dapagliflozin plus exenatide versus placebo had no effect on brain responses, but after 10 days dapagliflozin plus exenatide versus dapagliflozin increased activity in the insula and amygdala in response to low-calorie food pictures. CONCLUSION Dapagliflozin decreased activation in response to low-calorie food pictures, which may reflect a specific decreased preference for low-calorie foods, in combination with the previously found increased activation in response to high-calorie foods, which may reflect a specific preference for high-calorie foods, and may hamper SGLT2i-induced weight loss. Exenatide treatment increased activation in response to low-calorie foods. Combination treatment may lead to more favorable brain responses to low-calorie food cues, as we observed that the dapagliflozin-induced decreased response to low-calorie food pictures had disappeared.
Collapse
Affiliation(s)
- Charlotte C. van Ruiten
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, Vrije University Medical Center (VUmc), Amsterdam, Netherlands
- *Correspondence: Charlotte C. van Ruiten,
| | - Dick J. Veltman
- Department of Psychiatry, Amsterdam University Medical Center, Vrije University Medical Center (VUmc), Amsterdam, Netherlands
| | - Max Nieuwdorp
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, Vrije University Medical Center (VUmc), Amsterdam, Netherlands
- Department of Vascular Medicine, Amsterdam University Medical Center (AMC), Amsterdam, Netherlands
| | - Richard G. IJzerman
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Center, Vrije University Medical Center (VUmc), Amsterdam, Netherlands
| |
Collapse
|
223
|
Yang Y, Xu G. Update on Pathogenesis of Glomerular Hyperfiltration in Early Diabetic Kidney Disease. Front Endocrinol (Lausanne) 2022; 13:872918. [PMID: 35663316 PMCID: PMC9161673 DOI: 10.3389/fendo.2022.872918] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
In the existing stages of diabetic kidney disease (DKD), the first stage of DKD is called the preclinical stage, characterized by glomerular hyperfiltration, an abnormally elevated glomerular filtration rate. Glomerular hyperfiltration is an independent risk factor for accelerated deterioration of renal function and progression of nephropathy, which is associated with a high risk for metabolic and cardiovascular disease. It is imperative to understand hyperfiltration and identify potential treatments to delay DKD progress. This paper summarizes the current mechanisms of hyperfiltration in early DKD. We pay close attention to the effect of glucose reabsorption mediated by sodium-glucose cotransporters and renal growth on hyperfiltration in DKD patients, as well as the mechanisms of nitric oxide and adenosine actions on renal afferent arterioles via tubuloglomerular feedback. Furthermore, we also focus on the contribution of the atrial natriuretic peptide, cyclooxygenase, renin-angiotensin-aldosterone system, and endothelin on hyperfiltration. Proposing potential treatments based on these mechanisms may offer new therapeutic opportunities to reduce the renal burden in this population.
Collapse
|
224
|
Ota T, Ishikawa T, Sakakida T, Endo Y, Matsumura S, Yoshida J, Hirai Y, Mizushima K, Oka K, Doi T, Okayama T, Inoue K, Kamada K, Uchiyama K, Takagi T, Konishi H, Naito Y, Itoh Y. Treatment with broad-spectrum antibiotics upregulates Sglt1 and induces small intestinal villous hyperplasia in mice. J Clin Biochem Nutr 2022; 70:21-27. [PMID: 35068677 PMCID: PMC8764108 DOI: 10.3164/jcbn.21-42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/02/2021] [Indexed: 11/22/2022] Open
Abstract
Although extensive evidence indicates that the gut microbiota plays a crucial role in regulating glucose homeostasis, the exact regulatory mechanism remains unclear. This study aimed to investigate the effect of broad-spectrum antibiotics on the expression of glucose transporters, histomorphology of the small intestine, and glucose metabolism in mice. C57BL/6 mice were administered drinking water with or without a broad-spectrum antibiotic combination for 4 weeks. Thereafter, an oral glucose tolerance test was performed. Body weight, small intestine histopathology, mRNA levels of glucose transporters (SGLT1 and GLUT2) and intestinal transcription factors (CDX1 and CDX2) were evaluated. SGLT1 and CDX1 were upregulated in the small intestine upon antibiotic administration compared with that in the control group. The height and surface area of the jejunal villi were significantly higher upon antibiotic administration than in the control group. Fasting glucose levels were significantly higher upon antibiotic administration than in the control group. The present results indicate that treatment with broad-spectrum antibiotics upregulates SGLT1 and CDX1 and induces hyperplasia in the small intestine, thus increasing fasting blood glucose levels. Our results further the current understanding of the effects of broad-spectrum antibiotics on the gut microbiota and glucose homeostasis that may have future clinical implications.
Collapse
Affiliation(s)
- Takayuki Ota
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Takeshi Ishikawa
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Tomoki Sakakida
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Yuki Endo
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Shinya Matsumura
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Juichirou Yoshida
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Yasuko Hirai
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Katsura Mizushima
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Kaname Oka
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Toshifumi Doi
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Tetsuya Okayama
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Ken Inoue
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Kazuhiro Kamada
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Kazuhiko Uchiyama
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Tomohisa Takagi
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Hideyuki Konishi
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Yuji Naito
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine
| |
Collapse
|
225
|
Structural basis of the selective sugar transport in sodium-glucose cotransporters. J Mol Biol 2022; 434:167464. [DOI: 10.1016/j.jmb.2022.167464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/23/2022]
|
226
|
Chen H, Zhang X, Liao N, Ji Y, Mi L, Gan Y, Su Y, Wen F. Decreased expression of Glucagon-like peptide-1 receptor and Sodium-glucose co-transporter 2 in patients with proliferative diabetic retinopathy. Front Endocrinol (Lausanne) 2022; 13:1020252. [PMID: 36465606 PMCID: PMC9712753 DOI: 10.3389/fendo.2022.1020252] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
PURPOSE To investigate the expression of Glucagon-like peptide-1 receptor (GLP-1R), sodium-glucose co-transporter (SGLT) 1, SGLT2, Glucose transporter type 1 (GLUT1) and GLUT2 in patients with diabetic retinopathy (DR). METHODS We obtained peripheral blood mononuclear cells (PBMCs) and vitreous samples from 26 proliferative DR (PDR) patients, 25 non-proliferative DR (NPDR) patients, 25 non-DR (NDR) patients, and 26 nondiabetic patients with idiopathic epiretinal membranes (ERMs, control). The protein level and mRNA expression level of GLP-1R were quantified by immunoblot and qRT-PCR and the levels of SGLT1, SGLT2, GLUT1, and GLUT2 expression were determined by PCR. Their association with clinical parameters and PBMCs/vitreous cytokine was analyzed. Furthermore, immunofluorescence staining of GLP-1R and SGLT2 was carried out on samples of fibrovascular membranes (FVMs) retrieved from 26 patients with PDR and 26 patients with ERMs. RESULTS The transcriptional levels of GLP-1R and SGLT2 in PBMCs were significantly more decreased in PDR patients than in patients without DR and controls, which was simultaneously associated with an increased level of expression of tumor necrosis factor (TNF)-α and interferon (IFN)-γ. The expression levels of GLUT1 and GLUT2 were tightly correlated with their SGLT partners, respectively. Further, Immunofluorescence staining showed no positive staining of GLP-1R and SGLT2 was detected in the FVMs from PDR. CONCLUSIONS GLP-1R and SGLT2 were significantly decreased in PDR patients which was associated with an increased level of expression of TNF-α and IFN-γ. These findings implicate that defective GLP-1R and SGLT2 signaling may potentially correlate with immune response cytokines in patients with PDR.
Collapse
|
227
|
Negi CK, Babica P, Bajard L, Bienertova-Vasku J, Tarantino G. Insights into the molecular targets and emerging pharmacotherapeutic interventions for nonalcoholic fatty liver disease. Metabolism 2022; 126:154925. [PMID: 34740573 DOI: 10.1016/j.metabol.2021.154925] [Citation(s) in RCA: 138] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/14/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common form of chronic liver disease worldwide. With no Food and Drug Administration approved drugs, current treatment options include dietary restrictions and lifestyle modification. NAFLD is closely associated with metabolic disorders such as obesity, type 2 diabetes, and dyslipidemia. Hence, clinically various pharmacological approaches using existing drugs such as antidiabetic, anti-obesity, antioxidants, and cytoprotective agents have been considered in the management of NAFLD and nonalcoholic steatohepatitis (NASH). However, several pharmacological therapies aiming to alleviate NAFLD-NASH are currently being examined at various phases of clinical trials. Emerging data from these studies with drugs targeting diverse molecular mechanisms show promising outcomes. This review summarizes the current understanding of the pathogenic mechanisms of NAFLD and provides an insight into the pharmacological targets and emerging therapeutics with specific interventional mechanisms. In addition, we also discuss the importance and utility of new approach methodologies and regulatory perspectives for NAFLD-NASH drug development.
Collapse
Affiliation(s)
- Chander K Negi
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic
| | - Pavel Babica
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic.
| | - Lola Bajard
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic
| | - Julie Bienertova-Vasku
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno, Czech Republic; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Giovanni Tarantino
- Department of Clinical Medicine and Surgery, Federico II University Medical School of Naples, Naples, Italy
| |
Collapse
|
228
|
Nguyen YTK, Ha HTT, Nguyen TH, Nguyen LN. The role of SLC transporters for brain health and disease. Cell Mol Life Sci 2021; 79:20. [PMID: 34971415 PMCID: PMC11071821 DOI: 10.1007/s00018-021-04074-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 09/05/2021] [Accepted: 10/21/2021] [Indexed: 12/19/2022]
Abstract
The brain exchanges nutrients and small molecules with blood via the blood-brain barrier (BBB). Approximately 20% energy intake for the body is consumed by the brain. Glucose is known for its critical roles for energy production and provides substrates for biogenesis in neurons. The brain takes up glucose via glucose transporters GLUT1 and 3, which are expressed in several neural cell types. The brain is also equipped with various transport systems for acquiring amino acids, lactate, ketone bodies, lipids, and cofactors for neuronal functions. Unraveling the mechanisms by which the brain takes up and metabolizes these nutrients will be key in understanding the nutritional requirements in the brain. This could also offer opportunities for therapeutic interventions in several neurological disorders. For instance, emerging evidence suggests a critical role of lactate as an alternative energy source for neurons. Neuronal cells express monocarboxylic transporters to acquire lactate. As such, treatment of GLUT1-deficient patients with ketogenic diets to provide the brain with alternative sources of energy has been shown to improve the health of the patients. Many transporters are present in the brain, but only a small number has been characterized. In this review, we will discuss about the roles of solute carrier (SLC) transporters at the blood brain barrier (BBB) and neural cells, in transport of nutrients and metabolites in the brain.
Collapse
Affiliation(s)
- Yen T K Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Hoa T T Ha
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Tra H Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Long N Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore.
- SLING/Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore.
- Immunology Translational and Cardiovascular Disease Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore.
| |
Collapse
|
229
|
Wright EM. SGLT2 Inhibitors: Physiology and Pharmacology. KIDNEY360 2021; 2:2027-2037. [PMID: 35419546 PMCID: PMC8986039 DOI: 10.34067/kid.0002772021] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 09/17/2021] [Indexed: 02/07/2023]
Abstract
SGLTs are sodium glucose transporters found on the luminal membrane of the proximal tubule, where they reabsorb some 180 g (1 mol) of glucose from the glomerular filtrate each day. The natural glucoside phlorizin completely blocks glucose reabsorption. Oral SGLT2 inhibitors are rapidly absorbed into the blood stream, where theyremain in the circulation for hours. On glomerular filtration, they bind specifically to SGLT2 in the luminal membrane of the early proximal tubule to reduce glucose reabsorption by 50%-60%. Because of glucose excretion, these drugs lower plasma glucose and glycosylated hemoglobin levels in patients with type 2 diabetes mellitus. The drugs also protect against heart and renal failure. The aim of this review is to summarize what is known about the physiology of renal SGLTs and the pharmacology of SGLT drugs.
Collapse
Affiliation(s)
- Ernest M Wright
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
230
|
Vallon V, Nakagawa T. Renal Tubular Handling of Glucose and Fructose in Health and Disease. Compr Physiol 2021; 12:2995-3044. [PMID: 34964123 PMCID: PMC9832976 DOI: 10.1002/cphy.c210030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The proximal tubule of the kidney is programmed to reabsorb all filtered glucose and fructose. Glucose is taken up by apical sodium-glucose cotransporters SGLT2 and SGLT1 whereas SGLT5 and potentially SGLT4 and GLUT5 have been implicated in apical fructose uptake. The glucose taken up by the proximal tubule is typically not metabolized but leaves via the basolateral facilitative glucose transporter GLUT2 and is returned to the systemic circulation or used as an energy source by distal tubular segments after basolateral uptake via GLUT1. The proximal tubule generates new glucose in metabolic acidosis and the postabsorptive phase, and fructose serves as an important substrate. In fact, under physiological conditions and intake, fructose taken up by proximal tubules is primarily utilized for gluconeogenesis. In the diabetic kidney, glucose is retained and gluconeogenesis enhanced, the latter in part driven by fructose. This is maladaptive as it sustains hyperglycemia. Moreover, renal glucose retention is coupled to sodium retention through SGLT2 and SGLT1, which induces secondary deleterious effects. SGLT2 inhibitors are new anti-hyperglycemic drugs that can protect the kidneys and heart from failing independent of kidney function and diabetes. Dietary excess of fructose also induces tubular injury. This can be magnified by kidney formation of fructose under pathological conditions. Fructose metabolism is linked to urate formation, which partially accounts for fructose-induced tubular injury, inflammation, and hemodynamic alterations. Fructose metabolism favors glycolysis over mitochondrial respiration as urate suppresses aconitase in the tricarboxylic acid cycle, and has been linked to potentially detrimental aerobic glycolysis (Warburg effect). © 2022 American Physiological Society. Compr Physiol 12:2995-3044, 2022.
Collapse
Affiliation(s)
- Volker Vallon
- Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, La Jolla, California, USA,Department of Pharmacology, University of California San Diego, La Jolla, California, USA,VA San Diego Healthcare System, San Diego, California, USA,Correspondence to and
| | - Takahiko Nakagawa
- Division of Nephrology, Rakuwakai-Otowa Hospital, Kyoto, Japan,Correspondence to and
| |
Collapse
|
231
|
Sodium Glucose Cotransporter 1 (SGLT1) Inhibitors in Cardiovascular Protection: Mechanism Progresses and Challenges. Pharmacol Res 2021; 176:106049. [PMID: 34971725 DOI: 10.1016/j.phrs.2021.106049] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/15/2021] [Accepted: 12/26/2021] [Indexed: 12/20/2022]
Abstract
In recent years, multiple clinical trials have shown that sodium glucose cotransporter 1 (SGLT1) inhibitors have significant beneficial cardiovascular effects. This includes reducing the incidence of cardiovascular deaths and heart failure hospitalizations in people with and without diabetes, as well as those with and without generalized heart failure. The exact mechanism responsible for these beneficial effects is not completely understood. To explain the cardiovascular protective effects of SGLT1 inhibitors, several potential arguments have been proposed, including decreasing oxidative stress, regulating cardiac glucose uptake, preventing ischemia/reperfusion injury, alleviating the activation of cardiac fibroblasts, attenuating apoptosis, reducing intermittent high glucose-induced pyroptosis, ameliorating cardiac hypertrophy, attenuating arrhythmic vulnerabilities, and improving left ventricular systolic disorder. This article reviews the advantages and disadvantages of these mechanisms, and attempts to synthesize and prioritize mechanisms related to the reduction of clinical events.
Collapse
|
232
|
Brindangnanam P, Sawant AR, Prashanth K, Coumar MS. Bacterial effluxome as a barrier against antimicrobial agents: structural biology aspects and drug targeting. Tissue Barriers 2021; 10:2013695. [PMID: 34957912 DOI: 10.1080/21688370.2021.2013695] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Antimicrobial resistance (AMR) is fast becoming a medical crisis affecting the entire global population. The bacterial membrane is the first layer of defense for the bacteria against antimicrobial agents (AMA), specifically transporters in the membrane efflux these AMA out of the bacteria and plays a significant role in the AMR development. Understanding the structure and the functions of these efflux transporters is essential to overcome AMR. This review discusses efflux transporters (primary, secondary, and tripartite), their domain architectures, substrate specificities, and efflux pump inhibitors (EPI). Special emphasis on nosocomial ESKAPEE (Enterococcus faecium., Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter spp. and Escherichia coli) pathogens, their multidrug efflux targets and inhibitors are discussed. Deep knowledge about the functioning of efflux pumps and their structural aspects will open up opportunities for developing new EPI, which could be used along with AMA as combination therapy to overcome the emerging AMR crisis.
Collapse
Affiliation(s)
- Pownraj Brindangnanam
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Pondicherry, India
| | - Ajit Ramesh Sawant
- Department of Biotechnology, School of Life Sciences, Pondicherry University, Pondicherry, India
| | - K Prashanth
- Department of Biotechnology, School of Life Sciences, Pondicherry University, Pondicherry, India
| | - Mohane Selvaraj Coumar
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Pondicherry, India
| |
Collapse
|
233
|
Afshar N, Safaei S, Nickerson DP, Hunter PJ, Suresh V. Computational Modelling of Glucose Uptake by SGLT1 and Apical GLUT2 in the Enterocyte. Front Physiol 2021; 12:699152. [PMID: 34950044 PMCID: PMC8688934 DOI: 10.3389/fphys.2021.699152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 11/04/2021] [Indexed: 11/18/2022] Open
Abstract
It has been suggested that glucose absorption in the small intestine depends on both constitutively expressed SGLT1 and translocated GLUT2 in the brush border membrane, especially in the presence of high levels of luminal glucose. Here, we present a computational model of non-isotonic glucose uptake by small intestinal epithelial cells. The model incorporates apical uptake via SGLT1 and GLUT2, basolateral efflux into the blood via GLUT2, and cellular volume changes in response to non-isotonic conditions. The dependence of glucose absorption on luminal glucose, blood flow rate, and inlet blood glucose concentration is studied. Uptake via apical GLUT2 is found to be sensitive to all these factors. Under a range of conditions, the maximum apical GLUT2 flux is about half of the SGLT1 flux and is achieved at high luminal glucose (> 50 mM), high blood flow rates, and low inlet blood concentrations. In contrast, SGLT1 flux is less sensitive to these factors. When luminal glucose concentration is less than 10 mM, apical GLUT2 serves as an efflux pathway for glucose to move from the blood to the lumen. The model results indicate that translocation of GLUT2 from the basolateral to the apical membrane increases glucose uptake into the cell; however, the reduction of efflux capacity results in a decrease in net absorption. Recruitment of GLUT2 from a cytosolic pool elicits a 10–20% increase in absorption for luminal glucose levels in the a 20–100 mM range. Increased SGLT1 activity also leads to a roughly 20% increase in absorption. A concomitant increase in blood supply results in a larger increase in absorption. Increases in apical glucose transporter activity help to minimise cell volume changes by reducing the osmotic gradient between the cell and the lumen.
Collapse
Affiliation(s)
- Nima Afshar
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Soroush Safaei
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - David P Nickerson
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Peter J Hunter
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Vinod Suresh
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand.,Department of Engineering Science, University of Auckland, Auckland, New Zealand
| |
Collapse
|
234
|
Sędzikowska A, Szablewski L. Human Glucose Transporters in Renal Glucose Homeostasis. Int J Mol Sci 2021; 22:13522. [PMID: 34948317 PMCID: PMC8708129 DOI: 10.3390/ijms222413522] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/26/2022] Open
Abstract
The kidney plays an important role in glucose homeostasis by releasing glucose into the blood stream to prevent hypoglycemia. It is also responsible for the filtration and subsequent reabsorption or excretion of glucose. As glucose is hydrophilic and soluble in water, it is unable to pass through the lipid bilayer on its own; therefore, transport takes place using carrier proteins localized to the plasma membrane. Both sodium-independent glucose transporters (GLUT proteins) and sodium-dependent glucose transporters (SGLT proteins) are expressed in kidney tissue, and mutations of the genes coding for these glucose transporters lead to renal disorders and diseases, including renal cancers. In addition, several diseases may disturb the expression and/or function of renal glucose transporters. The aim of this review is to describe the role of the kidney in glucose homeostasis and the contribution of glucose transporters in renal physiology and renal diseases.
Collapse
Affiliation(s)
| | - Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland;
| |
Collapse
|
235
|
Jamalizadeh M, Hasanzad M, Sarhangi N, Sharifi F, Nasli-Esfahani E, Larijani B. Pilot study in pharmacogenomic management of empagliflozin in type 2 diabetes mellitus patients. J Diabetes Metab Disord 2021; 20:1407-1413. [PMID: 34900792 DOI: 10.1007/s40200-021-00874-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/03/2021] [Indexed: 11/26/2022]
Abstract
Background Type 2 diabetes mellitus (T2DM) is a metabolic disorder in which the patients with high blood sugar develop insufficient insulin secretion or insulin resistance. The solute carrier family, 5 member 2 (SLC5A2) gene is a member of sodium/glucose transporter family which can reduce heart and kidney problems. The current study aims to look into any association between rs11646054 variant in SLC5A2 gene and the anti-diabetic efficacy and safety of empagliflozin. Methods 14 T2DM who failed to respond to previous treatments, empagliflozin 10 mg was added for 6 months. Genotyping of the rs11646054 variant of SLC5A2 gene was performed by polymerase chain reaction (PCR) followed by Sanger sequencing. Results Although hemoglobin A1c (HbA1c) and low-density lipoprotein (LDL) were not significantly different, but the mean fasting blood sugar (FBS), 2-h post prandial (2hpp), albumin-to-creatinine ratio (ACR), and total cholesterol (TC) were significantly decreased after 6 months empagliflozin treatment. There was a significant difference in the mean final reductions in FBS level among genotypes. It's important to mention that those who were GG homozygotes had a tendency to have more decrements. Conclusions The study results indicate that effects of variation in SLC5A2 (rs11646054) on the clinical efficacy of empagliflozin were negligible.
Collapse
Affiliation(s)
- Mahdieh Jamalizadeh
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mandana Hasanzad
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Negar Sarhangi
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshad Sharifi
- Elderly Health Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ensieh Nasli-Esfahani
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, No.10-Jalal-e-Ale-Ahmad Street, Chamran Highway, 1411713119 Tehran, Iran
| |
Collapse
|
236
|
Yildirim K, Winkler B, Pogodalla N, Mackensen S, Baldenius M, Garcia L, Naffin E, Rodrigues S, Klämbt C. Redundant functions of the SLC5A transporters Rumpel, Bumpel, and Kumpel in ensheathing glial cells. Biol Open 2021; 11:274028. [PMID: 34897385 PMCID: PMC8790523 DOI: 10.1242/bio.059128] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 11/26/2021] [Indexed: 11/20/2022] Open
Abstract
Neuronal processing is energy demanding, and relies on sugar metabolism. To nurture the Drosophila nervous system, the blood-brain barrier forming glial cells take up trehalose from the hemolymph and then distribute the metabolic products further to all neurons. This function is provided by glucose and lactate transporters of the solute carrier (SLC) 5A family. Here we identified three SLC5A genes that are specifically expressed in overlapping sets of CNS glial cells, rumpel, bumpel and kumpel. We generated mutants in all genes and all mutants are viable and fertile, lacking discernible phenotypes. Loss of rumpel causes subtle locomotor phenotypes and flies display increased daytime sleep. In addition, in bumpel kumpel double mutants, and to an even greater extent in rumpel bumpel kumpel triple mutants, oogenesis is disrupted at the onset of the vitollegenic phase. This indicates a partially redundant functions between these genes. Rescue experiments exploring this effect indicate that oogenesis can be affected by CNS glial cells. Moreover, expression of heterologous mammalian SLC5A transporters, with known transport properties, suggest that Bumpel and/or Kumpel transport glucose or lactate. Overall, our results imply a redundancy in SLC5A nutrient sensing functions in Drosophila glial cells, affecting ovarian development and behavior.
Collapse
Affiliation(s)
- Kerem Yildirim
- Institute for Neuro- and Behavioral Biology, University of Münster, Badestr. 9, 48149 Münster, Germany.,Centre for Organismal Studies (COS) Heidelberg, University of Heidelberg, Im Neuenheimer Feld 230, 9120 Heidelberg, Germany
| | - Bente Winkler
- Institute for Neuro- and Behavioral Biology, University of Münster, Badestr. 9, 48149 Münster, Germany
| | - Nicole Pogodalla
- Institute for Neuro- and Behavioral Biology, University of Münster, Badestr. 9, 48149 Münster, Germany
| | - Steffi Mackensen
- Institute for Neuro- and Behavioral Biology, University of Münster, Badestr. 9, 48149 Münster, Germany
| | - Marie Baldenius
- Institute for Neuro- and Behavioral Biology, University of Münster, Badestr. 9, 48149 Münster, Germany
| | - Luis Garcia
- Institute for Neuro- and Behavioral Biology, University of Münster, Badestr. 9, 48149 Münster, Germany
| | - Elke Naffin
- Institute for Neuro- and Behavioral Biology, University of Münster, Badestr. 9, 48149 Münster, Germany
| | - Silke Rodrigues
- Institute for Neuro- and Behavioral Biology, University of Münster, Badestr. 9, 48149 Münster, Germany
| | - Christian Klämbt
- Institute for Neuro- and Behavioral Biology, University of Münster, Badestr. 9, 48149 Münster, Germany
| |
Collapse
|
237
|
Moinul M, Amin SA, Kumar P, Patil UK, Gajbhiye A, Jha T, Gayen S. Exploring sodium glucose cotransporter (SGLT2) inhibitors with machine learning approach: A novel hope in anti-diabetes drug discovery. J Mol Graph Model 2021; 111:108106. [PMID: 34923429 DOI: 10.1016/j.jmgm.2021.108106] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/15/2021] [Accepted: 12/06/2021] [Indexed: 12/29/2022]
Abstract
Conventional anti-diabetes agents exhibit some undesirable side effects. Recently, lactic acidosis and/or bladder cancer were also reported with the use of these agents. Hence, there is an urgent need for alternative anti-diabetes in order to reduce/avoid the unwanted effects. In this scenario sodium glucose cotransporter 2 (SGLT2) inhibitors has already been established as an important class of anti-diabetic drug. The search for new generation SGLT2 inhibitors with high affinity is still an ongoing process. Here, we aim to develop computational models to predict the SGLT2 inhibitory activity of small molecules based on chemical structures. This work provides in-silico analysis to propose possible fragment/fingerprint identification recommended for SGLT2 inhibitors. Up-to-our knowledge, this study is an initiative to propose fingerprints responsible for SGLT2 inhibition. Furthermore, we used nine different algorithms to build machine learning (ML) models that could be used to prioritize compounds as SGLT2 inhibitors from large libraries. The best performing ML models were applied to virtually screen a large collection of FDA approved drugs. The best predicted compounds have been recommended to be biologically investigated in future in order to identify next generation SGLT2 inhibitors with different chemical structure.
Collapse
Affiliation(s)
- Md Moinul
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar, 470003, India
| | - Sk Abdul Amin
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Prabhat Kumar
- Department of Computer Science, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Umesh Kumar Patil
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar, 470003, India
| | - Asmita Gajbhiye
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar, 470003, India
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| | - Shovanlal Gayen
- Laboratory of Drug Design and Discovery, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| |
Collapse
|
238
|
Real-Time Monitoring of Metabolism during Exercise by Exhaled Breath. Metabolites 2021; 11:metabo11120856. [PMID: 34940614 PMCID: PMC8709070 DOI: 10.3390/metabo11120856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/29/2021] [Accepted: 12/04/2021] [Indexed: 01/24/2023] Open
Abstract
Continuous monitoring of metabolites in exhaled breath has recently been introduced as an advanced method to allow non-invasive real-time monitoring of metabolite shifts during rest and acute exercise bouts. The purpose of this study was to continuously measure metabolites in exhaled breath samples during a graded cycle ergometry cardiopulmonary exercise test (CPET), using secondary electrospray high resolution mass spectrometry (SESI-HRMS). We also sought to advance the research area of exercise metabolomics by comparing metabolite shifts in exhaled breath samples with recently published data on plasma metabolite shifts during CPET. We measured exhaled metabolites using SESI-HRMS during spiroergometry (ramp protocol) on a bicycle ergometer. Real-time monitoring through gas analysis enabled us to collect high-resolution data on metabolite shifts from rest to voluntary exhaustion. Thirteen subjects participated in this study (7 female). Median age was 30 years and median peak oxygen uptake (VO2max) was 50 mL·/min/kg. Significant changes in metabolites (n = 33) from several metabolic pathways occurred during the incremental exercise bout. Decreases in exhaled breath metabolites were measured in glyoxylate and dicarboxylate, tricarboxylic acid cycle (TCA), and tryptophan metabolic pathways during graded exercise. This exploratory study showed that selected metabolite shifts could be monitored continuously and non-invasively through exhaled breath, using SESI-HRMS. Future studies should focus on the best types of metabolites to monitor from exhaled breath during exercise and related sources and underlying mechanisms.
Collapse
|
239
|
Han L, Qu Q, Aydin D, Panova O, Robertson MJ, Xu Y, Dror RO, Skiniotis G, Feng L. Structure and mechanism of the SGLT family of glucose transporters. Nature 2021; 601:274-279. [PMID: 34880492 DOI: 10.1038/s41586-021-04211-w] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 11/04/2021] [Indexed: 12/24/2022]
Abstract
Glucose is a primary energy source in living cells. The discovery in 1960s that a sodium gradient powers the active uptake of glucose in the intestine1 heralded the concept of a secondary active transporter that can catalyse the movement of a substrate against an electrochemical gradient by harnessing energy from another coupled substrate. Subsequently, coupled Na+/glucose transport was found to be mediated by sodium-glucose cotransporters2,3 (SGLTs). SGLTs are responsible for active glucose and galactose absorption in the intestine and for glucose reabsorption in the kidney4, and are targeted by multiple drugs to treat diabetes5. Several members within the SGLT family transport key metabolites other than glucose2. Here we report cryo-electron microscopy structures of the prototypic human SGLT1 and a related monocarboxylate transporter SMCT1 from the same family. The structures, together with molecular dynamics simulations and functional studies, define the architecture of SGLTs, uncover the mechanism of substrate binding and selectivity, and shed light on water permeability of SGLT1. These results provide insights into the multifaceted functions of SGLTs.
Collapse
Affiliation(s)
- Lei Han
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Qianhui Qu
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.,Shanghai Stomatological Hospital, Institutes of Biomedical Science, Department of Systems Biology for Medicine, Fudan University, Shanghai, China
| | - Deniz Aydin
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Computer Science, Stanford University, Stanford, CA, USA.,Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Ouliana Panova
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael J Robertson
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yan Xu
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ron O Dror
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Computer Science, Stanford University, Stanford, CA, USA.,Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Georgios Skiniotis
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Liang Feng
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
240
|
Abstract
Sodium glucose cotransporter 2 (SGLT-2) inhibitors are the latest class of antidiabetic medications. They prevent glucose reabsorption in the proximal convoluted tubule to decrease blood sugar. Several animal studies revealed that SGLT-2 is profoundly involved in the inflammatory response, fibrogenesis, and regulation of numerous intracellular signaling pathways. Likewise, SGLT-2 inhibitors markedly attenuated inflammation and fibrogenesis and improved the function of damaged organ in animal studies, observational studies, and clinical trials. SGLT-2 inhibitors can decrease blood pressure and ameliorate hypertriglyceridemia and obesity. Likewise, they improve the outcome of cardiovascular diseases such as heart failure, arrhythmias, and ischemic heart disease. SGLT-2 inhibitors are associated with lower cardiovascular and all-cause mortality as well. Meanwhile, they protect against nonalcoholic fatty liver disease (NAFLD), chronic kidney disease, acute kidney injury, and improve micro- and macroalbuminuria. SGLT-2 inhibitors can reprogram numerous signaling pathways to improve NAFLD, cardiovascular diseases, and renal diseases. For instance, they enhance lipolysis, ketogenesis, mitochondrial biogenesis, and autophagy while they attenuate the renin-angiotensin-aldosterone system, lipogenesis, endoplasmic reticulum stress, oxidative stress, apoptosis, and fibrogenesis. This review explains the beneficial effects of SGLT-2 inhibitors on NAFLD and cardiovascular and renal diseases and dissects the underlying molecular mechanisms in detail. This narrative review explains the beneficial effects of SGLT-2 inhibitors on NAFLD and cardiovascular and renal diseases using the results of latest observational studies, clinical trials, and meta-analyses. Thereafter, it dissects the underlying molecular mechanisms involved in the clinical effects of SGLT-2 inhibitors on these diseases.
Collapse
Affiliation(s)
- Moein Ala
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
241
|
Hu Z, Liao Y, Wang J, Wen X, Shu L. Potential impacts of diabetes mellitus and anti-diabetes agents on expressions of sodium-glucose transporters (SGLTs) in mice. Endocrine 2021; 74:571-581. [PMID: 34255273 DOI: 10.1007/s12020-021-02818-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 07/01/2021] [Indexed: 01/22/2023]
Abstract
PURPOSE Sodium-glucose transporters (SGLTs) are important targets for therapeutic intervention of type 2 diabetes. This study aims to evaluate the physiological influences of diabetes mellitus and the potential impacts of metformin and fluoxetine on SGLTs expressions. METHODS Alterations of SGLT1 and SGLT2 were measured in organs involved in glucose homeostasis (kidney, intestine, liver and pancreas) of streptozotocin (STZ) and high-fat diet (HFD) induced diabetic mice by western blotting and real-time PCR (RT-PCR) respectively. RESULTS In kidney, duodenal segments of intestine, liver, and pancreas of HFD diabetic mice, expressions of SGLT2 were all elevated compared to control mice. The level of SGLT1 was significantly increased in intestine, but was decreased in pancreas. SGLT1 expression in kidney was unaffected, and SGLT1 was undetectable in hepatocytes. Similar results were obtained in STZ diabetic mice. More importantly, here we noticed metformin decreased levels of SGLT2 in kidney, intestine, and pancreas of HFD mice markedly. Expressions of SGLT1 in intestine and pancreas were reduced by metformin as well. In contrast, fluoxetine increased abundances of SGLT2 and SGLT1 in kidney of HFD mice, but decreased SGLT1 expression in intestine. CONCLUSIONS The present study provided evidence that expressions of SGLT1 and SGLT2 were significantly modulated by diabetes mellitus as well as by metformin and fluoxetine, which indicated the efficacy of SGLT2 inhibitors might be impacted by these factors.
Collapse
Affiliation(s)
- Ziqi Hu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanjun Liao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Xiaohua Wen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Luan Shu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
- Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China.
| |
Collapse
|
242
|
Dykstra H, LaRose C, Fisk C, Waldhart A, Meng X, Zhao G, Wu N. TXNIP interaction with GLUT1 depends on PI(4,5)P 2. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2021; 1863:183757. [PMID: 34478732 DOI: 10.1016/j.bbamem.2021.183757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/12/2021] [Accepted: 08/24/2021] [Indexed: 12/31/2022]
Abstract
GLUT1 is a major glucose facilitator expressed ubiquitously among tissues. Upregulation of its expression plays an important role in the development of many types of cancer and metabolic diseases. Thioredoxin-interacting protein (TXNIP) is an α-arrestin that acts as an adaptor for GLUT1 in clathrin-mediated endocytosis. It regulates cellular glucose uptake in response to both intracellular and extracellular signals via its control on GLUT1-4. In order to understand the interaction between GLUT1 and TXNIP, we generated GLUT1 lipid nanodiscs and carried out isothermal titration calorimetry and single-particle electron microscopy experiments. We found that GLUT1 lipid nanodiscs and TXNIP interact in a 1:1 ratio and that this interaction requires phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2 or PIP2).
Collapse
Affiliation(s)
| | - Cassi LaRose
- Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Chelsea Fisk
- Van Andel Institute, Grand Rapids, MI 49503, USA
| | | | - Xing Meng
- Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Gongpu Zhao
- Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Ning Wu
- Van Andel Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
243
|
|
244
|
Abstract
Background: Iodide transport defect is an uncommon cause of dyshormonogenic congenital hypothyroidism due to homozygous or compound heterozygous pathogenic variants in the SLC5A5 gene, which encodes the sodium/iodide symporter (NIS), causing deficient iodide accumulation in thyroid follicular cells, thus impairing thyroid hormonogenesis. Methods:SLC5A5 gene variants were compiled from public databases and research articles exploring the molecular bases of congenital hypothyroidism. Using a dataset of 198 missense NIS variants classified as either benign or pathogenic, we developed and validated a machine learning-based NIS-specific variant classifier to predict the impact of missense NIS variants. Results: We generated a manually curated dataset containing 7793 unique SLC5A5 variants. As most databases compiled exome sequencing data, variant mapping revealed an increased density of variants in SLC5A5 coding exons. Based on allele frequency (AF) analysis, we established an AF threshold of 1:10,000 above which a variant should be considered benign. Most pathogenic NIS variants were located in the protein-coding region, as most patients were genetically diagnosed by using a candidate gene strategy limited to this region. Significantly, we evidenced that 94.5% of missense NIS variants were classified as of uncertain significance. Therefore, we developed an NIS-specific variant classifier to improve the prediction of pathogenicity of missense variants. Our classifier predicted the clinical outcome of missense variants with high accuracy (90%), outperforming state-of-the-art pathogenicity predictors, such as REVEL, PolyPhen-2, and SIFT. Based on the excellent performance of our classifier, we predicted the mutational landscape of NIS. The analysis of the mutational landscape revealed that most missense variants located in transmembrane segments are frequently pathogenic. Moreover, we predicted that ∼28% of all single-nucleotide variants that could cause missense NIS variants are pathogenic, thus putatively leading to congenital hypothyroidism if present in homozygous or compound heterozygous state. Conclusions: We reported the first NIS-specific variant classifier aiming at improving the interpretation of missense NIS variants in clinical practice. Deciphering the mutational landscape for every protein involved in thyroid hormonogenesis is a relevant task for a deep understanding of the molecular mechanisms causing dyshormonogenic congenital hypothyroidism.
Collapse
Affiliation(s)
- Mariano Martín
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Juan Pablo Nicola
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| |
Collapse
|
245
|
Agarwal S, Sudhini YR, Polat OK, Reiser J, Altintas MM. Renal cell markers: lighthouses for managing renal diseases. Am J Physiol Renal Physiol 2021; 321:F715-F739. [PMID: 34632812 DOI: 10.1152/ajprenal.00182.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Kidneys, one of the vital organs in our body, are responsible for maintaining whole body homeostasis. The complexity of renal function (e.g., filtration, reabsorption, fluid and electrolyte regulation, and urine production) demands diversity not only at the level of cell types but also in their overall distribution and structural framework within the kidney. To gain an in depth molecular-level understanding of the renal system, it is imperative to discern the components of kidney and the types of cells residing in each of the subregions. Recent developments in labeling, tracing, and imaging techniques have enabled us to mark, monitor, and identify these cells in vivo with high efficiency in a minimally invasive manner. In this review, we summarize different cell types, specific markers that are uniquely associated with those cell types, and their distribution in the kidney, which altogether make kidneys so special and different. Cellular sorting based on the presence of certain proteins on the cell surface allowed for the assignment of multiple markers for each cell type. However, different studies using different techniques have found contradictions in cell type-specific markers. Thus, the term "cell marker" might be imprecise and suboptimal, leading to uncertainty when interpreting the data. Therefore, we strongly believe that there is an unmet need to define the best cell markers for a cell type. Although the compendium of renal-selective marker proteins presented in this review is a resource that may be useful to researchers, we acknowledge that the list may not be necessarily exhaustive.
Collapse
Affiliation(s)
- Shivangi Agarwal
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | | | - Onur K Polat
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | - Jochen Reiser
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | | |
Collapse
|
246
|
Ho H, Kikuchi K, Oikawa D, Watanabe S, Kanemitsu Y, Saigusa D, Kujirai R, Ikeda‐Ohtsubo W, Ichijo M, Akiyama Y, Aoki Y, Mishima E, Ogata Y, Oikawa Y, Matsuhashi T, Toyohara T, Suzuki C, Suzuki T, Mano N, Kagawa Y, Owada Y, Katayama T, Nakayama T, Tomioka Y, Abe T. SGLT-1-specific inhibition ameliorates renal failure and alters the gut microbial community in mice with adenine-induced renal failure. Physiol Rep 2021; 9:e15092. [PMID: 34921520 PMCID: PMC8683788 DOI: 10.14814/phy2.15092] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/20/2021] [Accepted: 10/13/2021] [Indexed: 12/25/2022] Open
Abstract
Sodium-dependent glucose cotransporters (SGLTs) have attracted considerable attention as new targets for type 2 diabetes mellitus. In the kidney, SGLT2 is the major glucose uptake transporter in the proximal tubules, and inhibition of SGLT2 in the proximal tubules shows renoprotective effects. On the other hand, SGLT1 plays a role in glucose absorption from the gastrointestinal tract, and the relationship between SGLT1 inhibition in the gut and renal function remains unclear. Here, we examined the effect of SGL5213, a novel and potent intestinal SGLT1 inhibitor, in a renal failure (RF) model. SGL5213 improved renal function and reduced gut-derived uremic toxins (phenyl sulfate and trimethylamine-N-oxide) in an adenine-induced RF model. Histological analysis revealed that SGL5213 ameliorated renal fibrosis and inflammation. SGL5213 also reduced gut inflammation and fibrosis in the ileum, which is a primary target of SGL5213. Examination of the gut microbiota community revealed that the Firmicutes/Bacteroidetes ratio, which suggests gut dysbiosis, was increased in RF and SGL5213 rebalanced the ratio by increasing Bacteroidetes and reducing Firmicutes. At the genus level, Allobaculum (a major component of Erysipelotrichaceae) was significantly increased in the RF group, and this increase was canceled by SGL5213. We also measured the effect of SGL5213 on bacterial phenol-producing enzymes that catalyze tyrosine into phenol, following the reduction of phenyl sulfate, which is a novel marker and a therapeutic target for diabetic kidney disease DKD. We found that the enzyme inhibition was less potent, suggesting that the change in the microbial community and the reduction of uremic toxins may be related to the renoprotective effect of SGL5213. Because SGL5213 is a low-absorbable SGLT1 inhibitor, these data suggest that the gastrointestinal inhibition of SGLT1 is also a target for chronic kidney diseases.
Collapse
Affiliation(s)
- Hsin‐Jung Ho
- Department of Medical ScienceTohoku University Graduate School of Biomedical EngineeringSendaiJapan
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Koichi Kikuchi
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
- Department of Medical MegabankTohoku UniversitySendaiJapan
| | - Daiki Oikawa
- Department of Biomolecular Engineering Applied Life ChemistryTohoku University Graduate School of EngineeringSendaiJapan
| | - Shun Watanabe
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
- Department of Clinical Biology and Hormonal RegulationTohoku University Graduate School of MedicineSendaiJapan
| | | | - Daisuke Saigusa
- Department of Integrative Genomics, Tohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Ryota Kujirai
- Laboratory of OncologyPharmacy Practice and SciencesTohoku University Graduate School of Pharmaceutical SciencesSendaiJapan
| | - Wakako Ikeda‐Ohtsubo
- Laboratory of Animal Products ChemistryGraduate School of Agricultural ScienceTohoku UniversitySendaiJapan
| | - Mariko Ichijo
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Yukako Akiyama
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Yuichi Aoki
- Department of Integrative Genomics, Tohoku Medical Megabank OrganizationTohoku UniversitySendaiJapan
| | - Eikan Mishima
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Yoshiaki Ogata
- Department of Clinical Biology and Hormonal RegulationTohoku University Graduate School of MedicineSendaiJapan
| | - Yoshitsugu Oikawa
- Department of PediatricsTohoku University Graduate School of MedicineSendaiJapan
| | - Tetsuro Matsuhashi
- Department of PediatricsTohoku University Graduate School of MedicineSendaiJapan
| | - Takafumi Toyohara
- Department of Medical ScienceTohoku University Graduate School of Biomedical EngineeringSendaiJapan
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Chitose Suzuki
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Takehiro Suzuki
- Department of Medical ScienceTohoku University Graduate School of Biomedical EngineeringSendaiJapan
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Nariyasu Mano
- Department of Pharmaceutical SciencesTohoku University HospitalSendaiJapan
| | - Yoshiteru Kagawa
- Department of Organ AnatomyTohoku University Graduate School of MedicineSendaiJapan
| | - Yuji Owada
- Department of Organ AnatomyTohoku University Graduate School of MedicineSendaiJapan
| | - Takane Katayama
- Laboratory of Molecular Biology of BioresponseGraduate School of BiostudiesKyoto UniversityKyotoJapan
| | - Toru Nakayama
- Department of Biomolecular Engineering Applied Life ChemistryTohoku University Graduate School of EngineeringSendaiJapan
| | - Yoshihisa Tomioka
- Laboratory of Molecular Biology of BioresponseGraduate School of BiostudiesKyoto UniversityKyotoJapan
| | - Takaaki Abe
- Department of Medical ScienceTohoku University Graduate School of Biomedical EngineeringSendaiJapan
- Division of Nephrology, Endocrinology and Vascular MedicineTohoku University Graduate School of MedicineSendaiJapan
- Department of Clinical Biology and Hormonal RegulationTohoku University Graduate School of MedicineSendaiJapan
| |
Collapse
|
247
|
Pawlos A, Broncel M, Woźniak E, Gorzelak-Pabiś P. Neuroprotective Effect of SGLT2 Inhibitors. Molecules 2021; 26:7213. [PMID: 34885795 PMCID: PMC8659196 DOI: 10.3390/molecules26237213] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 12/17/2022] Open
Abstract
Patients with diabetes are at higher risk of cardiovascular diseases and cognitive impairment. SGLT2 inhibitors (Empagliflozin, Canagliflozin, Dapagliflozin, Ertugliflozin, Sotagliflozin) are newer hypoglycemic agents with many pleiotropic effects. In this review, we discuss their neuroprotective potential. SGLT2 inhibitors (SGLT2i) are lipid-soluble and reach the brain/serum ratio from 0.3 to 0.5. SGLT receptors are present in the central nervous system (CNS). Flozins are not fully SGLT2-selective and have an affinity for the SGLT1 receptor, which is associated with protection against ischemia/reperfusion brain damage. SGLT2i show an anti-inflammatory and anti-atherosclerotic effect, including reduction of proinflammatory cytokines, M2 macrophage polarization, JAK2/STAT1 and NLRP3 inflammasome inhibition, as well as cIMT regression. They also mitigate oxidative stress. SGLT2i improve endothelial function, prevent remodeling and exert a protective effect on the neurovascular unit, blood-brain barrier, pericytes, astrocytes, microglia, and oligodendrocytes. Flozins are also able to inhibit AChE, which contributes to cognitive improvement. Empagliflozin significantly increases the level of cerebral BDNF, which modulates neurotransmission and ensures growth, survival, and plasticity of neurons. Moreover, they may be able to restore the circadian rhythm of mTOR activation, which is quite a novel finding in the field of research on metabolic diseases and cognitive impairment. SGLT2i have a great potential to protect against atherosclerosis and cognitive impairment in patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
| | - Marlena Broncel
- Laboratory of Tissue Immunopharmacology, Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, Kniaziewicza 1/5, 91-347 Lodz, Poland; (A.P.); (E.W.); (P.G.-P.)
| | | | | |
Collapse
|
248
|
The SGLT-2 Inhibitors in Personalized Therapy of Diabetes Mellitus Patients. J Pers Med 2021; 11:jpm11121249. [PMID: 34945721 PMCID: PMC8708213 DOI: 10.3390/jpm11121249] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus (DM) represents a major public health problem, with yearly increasing prevalence. DM is considered a progressive vascular disease that develops macro and microvascular complications, with a great impact on the quality of life of diabetic patients. Over time, DM has become one of the most studied diseases; indeed, finding new pharmacological ways to control it is the main purpose of the research involved in this issue. Sodium–glucose cotransporter 2 inhibitors (SGLT-2i) are a modern drug class of glucose-lowering agents, whose use in DM patients has increased in the past few years. Besides the positive outcomes regarding glycemic control and cardiovascular protection in DM patients, SGLT-2i have also been associated with metabolic benefits, blood pressure reduction, and improved kidney function. The recent perception and understanding of SGLT-2i pathophysiological pathways place this class of drugs towards a particularized patient-centered approach, moving away from the well-known glycemic control strategy. SGLT-2i have been shown not only to reduce death from cardiovascular causes, but also to reduce the risk of stroke and heart failure hospitalization. This article aims to review and highlight the existing literature on the effects of SGLT-2i, emphasizing their role as oral antihyperglycemic agents in type 2 DM, with important cardiovascular and metabolic benefits.
Collapse
|
249
|
Al-Shamasi AA, Elkaffash R, Mohamed M, Rayan M, Al-Khater D, Gadeau AP, Ahmed R, Hasan A, Eldassouki H, Yalcin HC, Abdul-Ghani M, Mraiche F. Crosstalk between Sodium-Glucose Cotransporter Inhibitors and Sodium-Hydrogen Exchanger 1 and 3 in Cardiometabolic Diseases. Int J Mol Sci 2021; 22:12677. [PMID: 34884494 PMCID: PMC8657861 DOI: 10.3390/ijms222312677] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/12/2021] [Indexed: 12/14/2022] Open
Abstract
Abnormality in glucose homeostasis due to hyperglycemia or insulin resistance is the hallmark of type 2 diabetes mellitus (T2DM). These metabolic abnormalities in T2DM lead to cellular dysfunction and the development of diabetic cardiomyopathy leading to heart failure. New antihyperglycemic agents including glucagon-like peptide-1 receptor agonists and the sodium-glucose cotransporter-2 inhibitors (SGLT2i) have been shown to attenuate endothelial dysfunction at the cellular level. In addition, they improved cardiovascular safety by exhibiting cardioprotective effects. The mechanism by which these drugs exert their cardioprotective effects is unknown, although recent studies have shown that cardiovascular homeostasis occurs through the interplay of the sodium-hydrogen exchangers (NHE), specifically NHE1 and NHE3, with SGLT2i. Another theoretical explanation for the cardioprotective effects of SGLT2i is through natriuresis by the kidney. This theory highlights the possible involvement of renal NHE transporters in the management of heart failure. This review outlines the possible mechanisms responsible for causing diabetic cardiomyopathy and discusses the interaction between NHE and SGLT2i in cardiovascular diseases.
Collapse
Affiliation(s)
- Al-Anood Al-Shamasi
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Rozina Elkaffash
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Meram Mohamed
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Menatallah Rayan
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Dhabya Al-Khater
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Alain-Pierre Gadeau
- INSERM, Biology of Cardiovascular Disease, University of Bordeaux, U1034 Pessac, France;
| | - Rashid Ahmed
- Department of Mechanical and Chemical Engineering, College of Engineering, Qatar University, Doha P.O. Box 2713, Qatar; (R.A.); (A.H.)
- Biomedical Research Centre (BRC), Qatar University, Doha P.O. Box 2713, Qatar;
| | - Anwarul Hasan
- Department of Mechanical and Chemical Engineering, College of Engineering, Qatar University, Doha P.O. Box 2713, Qatar; (R.A.); (A.H.)
- Biomedical Research Centre (BRC), Qatar University, Doha P.O. Box 2713, Qatar;
| | - Hussein Eldassouki
- College of Kinesiology, University of Saskatchewan, Saskatoon, SK S7N 5B5, Canada;
| | | | - Muhammad Abdul-Ghani
- Division of Diabetes, University of Texas Health Science Center at San Antonio, Floyd Curl Drive, San Antonio, TX 7703, USA;
| | - Fatima Mraiche
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
250
|
Abstract
Patients with type 2 diabetes mellitus (T2D) are at increased risk of cardiovascular (CV) disease. Sodium glucose cotransporter 2 (SGLT2) inhibitors, also known as gliflozins, are a class of medications used to treat T2D by preventing the reabsorption of glucose filtered through the kidney and thereby facilitating glucose excretion in the urine. Over the past 5 years, many cardiovascular outcome trials (CVOTs) have evaluated the safety and efficacy of SGLT2 inhibitors in preventing CV events. The results of 7 CVOTs have provided solid evidence that the use of SGLT2 in patients with T2D and at high CV risk significantly reduced the risk of death from CV causes. Moreover, in patient with heart failure with reduced ejection fraction, regardless of the presence or absence of T2D, SGLT2 inhibitors use significantly reduced the risk of worsening heart failure and death from CV causes. Although the exact mechanism of the cardiorenal benefit of SGLT2 inhibitors is still unknown, studies have shown that the beneficial effect of these drugs cannot be exclusively explained by their glucose lowering effect, and several possible mechanisms have been proposed. This review will explore the changing role of SGLT2 inhibitors from a diabetes drug to clinical practice guideline-supported therapy for the prevention and treatment of CV diseases, including heart failure.
Collapse
Affiliation(s)
- Reza Mohebi
- Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - James L Januzzi
- Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Baim Institute for Clinical Research, Boston, MA, USA
| |
Collapse
|