1
|
Sun L, Hong X, Zhu Q, Xiang Y, Xu C, Weng L, Cai J, Liang N, Xue M, Ge H. Dietary intervention enhances fertility in obese male mice by regulating SLC2As. J Mol Histol 2025; 56:174. [PMID: 40425964 DOI: 10.1007/s10735-025-10470-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025]
Abstract
Research has demonstrated that obesity can affect male fertility and reproductive potential, but the underlying mechanism remains unclear. This study aimed to investigate the effects of a high-fat diet (HFD) and dietary intervention on reproductive function, glucose metabolism, and related SLC2As in male mice. Forty 4-week-old male ICR mice were randomly divided into the normal diet (ND) group (group N, n = 15) and the HFD group (group F, n = 25). After 12 weeks, the mice were further divided into the following groups: ND maintenance group (NN group, n = 10), HFD maintenance group (FF group, n = 10), and transition to ND group (FN group, n = 10) through dietary intervention for 8 weeks. Intraperitoneal glucose tolerance test (IPGTT) was performed, and parameters including fasting blood glucose, body weight, sperm count, sperm motility, and testis and epididymis measurements were recorded. Testicular morphology was observed through hematoxylin-eosin staining. Western blot and immunofluorescence were used to detect the protein expression and localisation of SLC2As in the testis. Long-term HFD consumption resulted in increased body and testicular weights, decreased testicular and epididymal organ coefficients, reduced sperm motility rate, and increased area under the curve in the IPGTT test. After dietary intervention, compared to the NN group, the FF and FN groups exhibited increased testis weight, decreased testicular and epididymal organ coefficients, decreased sperm motility rate, reduced SLC2A3 and SLC2A8 protein expression levels in the FF group, and decreased SLC2A8 protein expression in the FN group. Obesity induced by HFD caused damage to the reproductive system of male mice and affected testicular glucose metabolism and the expression of sugar transporter SLC2As. Transitioning from HFD to ND can improve reproductive dysfunction caused by dietary obesity and its impact on sugar transporter protein expression to a certain extent.
Collapse
Affiliation(s)
- Liujia Sun
- Reproduction Medicine Centre, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
- Shaoxing People's Hospital, Shaoxing, China
| | - Xiaoqi Hong
- Reproduction Medicine Centre, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
- Graduate School, Nanjing Medical University, Nanjing, China
| | - Qi Zhu
- Reproduction Medicine Centre, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
- Graduate School, Nanjing Medical University, Nanjing, China
| | - Yu Xiang
- Reproduction Medicine Centre, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Cen Xu
- Reproduction Medicine Centre, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Lingling Weng
- Reproduction Medicine Centre, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jieyu Cai
- Reproduction Medicine Centre, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Na Liang
- Reproduction Medicine Centre, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
- Graduate School, Dalian Medical University, Dalian, China
| | - Mingrui Xue
- Reproduction Medicine Centre, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
- Graduate School, Dalian Medical University, Dalian, China
| | - Hongshan Ge
- Reproduction Medicine Centre, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China.
- Graduate School, Nanjing Medical University, Nanjing, China.
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, China.
- Graduate School, Dalian Medical University, Dalian, China.
| |
Collapse
|
2
|
Ida-Yonemochi H. Role of glucose metabolism in amelogenesis. J Oral Biosci 2025; 67:100667. [PMID: 40306383 DOI: 10.1016/j.job.2025.100667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 04/27/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Cell energy metabolism plays a pivotal role in organ development and function by regulating cell behavior in pathophysiological conditions. Glucose metabolism is the central cascade for obtaining energy in mammalian cells, and cells alter the glucose metabolic pathway depending on intra- and extracellular environments. Therefore, glucose metabolism is closely associated with cell differentiation stages, and cell energy metabolism plays a vital role not only in energy production but also in cell fate regulation in organogenesis. HIGHLIGHT During enamel formation, the timing of the expression of passive and active glucose transporters, glycogen synthesis, and glycogen degradation is strictly regulated according to the energy demand of ameloblast-lineage cells. These glucose metabolic reactions are particularly active in the maturation stage of ameloblasts. Furthermore, autophagy, a key regulator of cellular energy homeostasis that modulates glucose metabolism, occurs during both the secretory and maturation stages of ameloblasts. Disruption of glucose metabolism cascade and autophagy induces enamel hypoplasia, as demonstrated in both in vitro and in vivo models. CONCLUSION Adequate energy supply via glucose metabolism is essential for enamel matrix secretion and maturation. A thorough understanding of the precise regulation of energy metabolism in amelogenesis facilitates comprehension of the normal enamel formation process and pathological conditions affecting it. This review summarizes glucose metabolic processes during amelogenesis, focusing on glucose uptake, glycogenesis, and glycogenolysis.
Collapse
Affiliation(s)
- Hiroko Ida-Yonemochi
- Division of Anatomy and Cell Biology of the Hard Tissue, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, 2-5274 Gakkocho-dori, Chuo-ku, Niigata 951-8514, Japan.
| |
Collapse
|
3
|
Jin SH, Shui F, He TT, Jia FM, Wang X, Liu X, Liu XL, Ling ZQ, Geng ZY. Transcriptome analysis reveals key genes and signalling pathways related to residual feed intake in meat-type ducks. Animal 2025; 19:101521. [PMID: 40367888 DOI: 10.1016/j.animal.2025.101521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 05/16/2025] Open
Abstract
The feed utilisation rate is a key factor that affects the economic benefits of meat-type duck breeding. In recent years, residual feed intake (RFI) has been routinely used in poultry breeding as an index for evaluating feed utilisation. However, the genetic mechanism underlying RFI in meat-type ducks remains poorly understood. In the present study, 1 000 meat-type ducks with similar BW were randomly selected to measure BW gain and feed intake from 21 to 42 d of age to assess RFI. Six high- and six low-RFI meat-type ducks were randomly selected for a transcriptome survey of livers. Protein-protein interaction (PPI) network and gene set enrichment (GSEA) analyses were used to elucidate the molecular basis of RFI. We identified 1 297 differentially expressed genes (DEGs) in the LRFI group, of which 686 and 611 were markedly up- and downregulated, respectively. Functional annotation showed that DEGs were mainly enriched in gene ontology terms related to the regulation of biosynthetic and metabolic processes. Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis showed significant enrichment of insulin signalling and multiple pathways related to energy metabolism. Furthermore, the PPI network analysis revealed that PRKACB, PRKAR2A, FYN, PTK2, ITGB1, ESR1, and PIK3CB were primarily related to the biological processes of lipid metabolism, glucose transport, and immunological stress and may affect RFI. Moreover, the GSEA suggested that upregulated genes in the LRFI group were associated with immune, lipid transport, and insulin signalling. ACLY and SLC50A1 were the most markedly upregulated and downregulated DEGs, respectively. In the PPI network, PRKACB, PRKAR2A, and FYN were identified as pivot genes. The aforementioned DEGs were mainly involved in lipid and glucose metabolism and inflammatory response, consistent with the KEGG and GSEA results. Therefore, these results revealed PRKACB, PRKAR2A, and FYN as potentially key genes for improving feed efficiency traits in meat-type ducks. Our results provide insights into the biological basis of RFI in meat-type ducks and will be useful for the selection of meat-type ducks with a greater feed efficiency phenotype in future breeding programmes.
Collapse
Affiliation(s)
- S H Jin
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - F Shui
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - T T He
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - F M Jia
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - X Wang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - X Liu
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - X L Liu
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Z Q Ling
- Huangshan Qiangying Duck Breeding, Co. Ltd., Huangshan 245461, China
| | - Z Y Geng
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.
| |
Collapse
|
4
|
Pourkarim F, Entezari-Maleki T, Rezaee H. Current Evidence on SGLT-2 Inhibitors in Prediabetes: A Review of Preclinical and Clinical Data. J Clin Pharmacol 2025. [PMID: 40207728 DOI: 10.1002/jcph.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025]
Abstract
Individuals with prediabetes have a higher risk of cardiovascular events and diabetes mellitus. Therefore, the prevention or delay of prediabetes progression to diabetes via lifestyle modification and medications is an important measure to reduce morbidity and mortality in this population. Based on the American Diabetes Association (ADA) guidelines, metformin is the only recommended drug for prediabetes. A growing body of evidence has shown the beneficial effects of sodium-glucose transporter 2 (SGLT-2) inhibitors in prediabetes. These drugs offer cardiovascular mortality benefits over metformin. This review aimed to summarize current evidence about the clinical effects of SGLT-2 inhibitors in prediabetes.
Collapse
Affiliation(s)
- Fariba Pourkarim
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Taher Entezari-Maleki
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Haleh Rezaee
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Infectious Diseases and Tropical Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
5
|
Meng Y, Lv L, Yao H, Lin Z, Dong Y. Genome-Wide Analysis Reveals Expansion and Positive Selection of Monocarboxylate Transporter Genes Linked to Enhanced Salinity and Ammonia Tolerance in Sinonovacula constricta. Animals (Basel) 2025; 15:795. [PMID: 40150324 PMCID: PMC11939762 DOI: 10.3390/ani15060795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Our previous genome analysis of Sinonovacula constricta revealed an expansion of the monocarboxylate transporter gene family, which is crucial for metabolic dynamic balance and intracellular pH regulation. To further elucidate the role of these expanded MCT genes in response to variable environmental conditions, we conducted a comprehensive genome-wide identification, phylogenetic evolution and expression analysis. In this study, 16 sodium-coupled monocarboxylate transporter genes (designated as ScSMCTs) and 54 proton-coupled monocarboxylate transporter genes (designated as ScMCTs) were identified from the S. constricta genome. The results of gene number comparison indicated significant expansion of ScSMCTs and ScMCTs in mollusks compared to vertebrates, likely due to tandem repeats and dispersed duplications in S. constricta. The syntenic analysis demonstrated that the razor-clam MCT genes had the highest number of homologous gene pairs with Meretrix meretrix. The phylogenetic tree showed that MCT and SMCT proteins were distinctly clustered in two large branches. Moreover, positive selection analysis revealed three positive selection sites in the MCT amino acid sequences sites. Multi-transcriptome analyses and the temporal expression patterns displayed that ScSMCTs and ScMCTs play distinct roles in response to salinity and ammonia stressors. It is worth noting that the majority of these genes involved in abiotic stresses belong to MCTs. Overall, our findings revealed the important roles of ScSMCTs and ScMCTs under abiotic stress, and provided valuable information for the evolution of this family in mollusks, as well as a theoretical basis for the further study of the mechanism and function of this gene family in S. constricta.
Collapse
Affiliation(s)
- Yiping Meng
- School of Marine Sciences, Ningbo University, Ningbo 315010, China;
- Ninghai Institute of Mariculture Breeding and Seed Industry, Zhejiang Wanli University, Ninghai 315604, China
| | - Liyuan Lv
- Ninghai Institute of Mariculture Breeding and Seed Industry, Zhejiang Wanli University, Ninghai 315604, China
| | - Hanhan Yao
- Key Laboratory of Aquatic Germplasm Resources of Zhejiang, Zhejiang Wanli University, Ningbo 315100, China; (H.Y.); (Z.L.)
| | - Zhihua Lin
- Key Laboratory of Aquatic Germplasm Resources of Zhejiang, Zhejiang Wanli University, Ningbo 315100, China; (H.Y.); (Z.L.)
| | - Yinghui Dong
- College of Advanced Agricultural Sciences, Zhejiang Wanli University, Ningbo 315101, China
| |
Collapse
|
6
|
Huang Y, Li L, Hong Y, Cheng L, Gu Z. The effect of carbohydrates with different levels of digestibility on energy metabolism in vivo under hypobaric hypoxic conditions. Carbohydr Polym 2025; 351:123114. [PMID: 39779022 DOI: 10.1016/j.carbpol.2024.123114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025]
Abstract
Current strategies for improving energy supply in hypobaric hypoxic environments are limited. Therefore, this study investigates the effects of four carbohydrates with different levels of digestibility on energy metabolism in vivo in hypobaric hypoxic environments. First, we characterized the four types of carbohydrates. Subsequently, reverse transcription quantitative polymerase chain reaction (RT-qPCR) was used to characterize the expression of GLUT1, GLUT2, and SGLT1 in the glucose transport pathway in vivo. In addition, the effects of different levels of carbohydrate digestibility on energy expenditure were evaluated in vivo. The results showed that pre-gelatinized corn starch significantly increased GLUT1 gene expression in the hypobaric hypoxic conditions (1.58 times, compared to normobaric normoxic). In addition, pre-gelatinized corn starch increased energy expenditure in the hypobaric hypoxic conditions and performed better in terms of glycogen accumulation and glucose transport. Therefore, pre-gelatinized corn starch administration may be a promising strategy for long-term energy supplementation in hypobaric hypoxic.
Collapse
Affiliation(s)
- Yali Huang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, China
| | - Lingjin Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, China
| | - Yan Hong
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, China.
| | - Li Cheng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, China
| | - Zhengbiao Gu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
7
|
Quan C, Jiang X. The molecular mechanism underlying the human glucose facilitators inhibition. VITAMINS AND HORMONES 2025; 128:49-92. [PMID: 40097253 DOI: 10.1016/bs.vh.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Glucose is the primary energy substrate and an essential precursor for cellular metabolism. Maintaining glucose homeostasis necessitates the presence of glucose transporters, as the hydrophilic nature of glucose prevents its passage across the cell membrane. The GLUT family is a crucial group of glucose transporters that facilitate glucose diffusion along the transmembrane glucose concentration gradient. Dysfunction in GLUTs is associated with diseases, such as GLUT1 deficiency syndrome, Fanconi-Bickel syndrome, and type 2 diabetes. Furthermore, elevated expression of GLUTs fuels aerobic glycolysis, known as the Warburg effect, in various types of cancers, making GLUT isoforms possible targets for antineoplastic therapies. To date, 30 GLUT and homolog structures have been released on the Protein Data Bank (PDB), showcasing multiple conformational and ligand-binding states. These structures elucidate the molecular mechanisms underlying substrate recognition, the alternating access cycle, and transport inhibition. Here, we summarize the current knowledge of human GLUTs and their role in cancer, highlighting recent advances in the structural characterization of GLUTs. We also compare the inhibition mechanisms of exofacial and endofacial GLUT inhibitors, providing insights into the design and optimization of GLUT inhibitors for therapeutic applications.
Collapse
Affiliation(s)
- Cantao Quan
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, The Department of Medical Genetics, The Department of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, P.R. China
| | - Xin Jiang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, The Department of Medical Genetics, The Department of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, P.R. China.
| |
Collapse
|
8
|
Corral P, Nardelli N, Elbert A, Aranguren F, Schreier L. Impact of SGLT2 Inhibitors on Lipoproteins in Type 2 Diabetes. Curr Diab Rep 2025; 25:16. [PMID: 39762665 DOI: 10.1007/s11892-024-01572-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE OF REVIEW This article explores the cardiovascular effects of sodium-glucose cotransporter 2 inhibitors (SGLT2i) in patients with type 2 diabetes mellitus (T2DM), with a particular focus on their impact on lipid profiles. As evidence grows of the cardiovascular benefits of SGLT2i beyond glucose control, it is essential to better understand their effects on lipoproteins and their impact on cardiovascular disease. RECENT FINDINGS SGLT2i have shown significant cardiovascular benefits in patients with type 2 diabetes mellitus, beyond their role in lowering blood glucose. Studies indicate that SGLT2i reduce major adverse cardiovascular events by impacting factors such as blood pressure, body weight, and arterial stiffness. However, their effects on lipid profile remain complex and somewhat inconsistent. Some research points to modest increases in LDL cholesterol, while others report shifts toward less atherogenic lipid profile, including reductions in triglycerides and small, dense LDL particles, and increases in HDL-C. SGLT2i represent a significant advancement in managing diabetes and associated cardiovascular risks, with benefits such as triglyceride reduction and HDL-C increase. While their impact on LDL-C remains controversial and varies across studies, the reduction of small, dense LDL particles may mitigate negative effects. This article highlights the need for future research to better understand the specific mechanisms behind lipid modulation.
Collapse
Affiliation(s)
- Pablo Corral
- Facultad de Medicina, Departamento de Farmacología e Investigación, Universidad FASTA, Instituto de Investigaciones Clínicas (IIC), Mar del Plata, Argentina.
| | - Natalia Nardelli
- Centro de Nutrición y Diabetes (CENUDIAB), Ciudad Autónoma de Buenos Aires, Argentina
| | - Alicia Elbert
- Centro de Enfermedades Renales e Hipertensión Arterial (CEREHA S.A.), Ciudad Autónoma de Buenos Aires, Argentina
| | | | - Laura Schreier
- Facultad de Farmacia y Bioquímica, Laboratorio de Lípidos y Aterosclerosis, Universidad de Buenos Aires, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC-UBA), Buenos Aires, Argentina
| |
Collapse
|
9
|
Shafi S, Khan MA, Ahmad J, Rabbani SA, Singh S, Najmi AK. Envisioning Glucose Transporters (GLUTs and SGLTs) as Novel Intervention against Cancer: Drug Discovery Perspective and Targeting Approach. Curr Drug Targets 2025; 26:109-131. [PMID: 39377414 DOI: 10.2174/0113894501335877240926101134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 10/09/2024]
Abstract
Metabolic reprogramming and altered cellular energetics have been recently established as an important cancer hallmark. The modulation of glucose metabolism is one of the important characteristic features of metabolic reprogramming in cancer. It contributes to oncogenic progression by supporting the increased biosynthetic and bio-energetic demands of tumor cells. This oncogenic transformation consequently results in elevated expression of glucose transporters in these cells. Moreover, various cancers exhibit abnormal transporter expression patterns compared to normal tissues. Recent investigations have underlined the significance of glucose transporters in regulating cancer cell survival, proliferation, and metastasis. Abnormal regulation of these transporters, which exhibit varying affinities for hexoses, could enable cancer cells to efficiently manage their energy supply, offering a crucial edge for proliferation. Exploiting the upregulated expression of glucose transporters, GLUTs, and Sodium Linked Glucose Transporters (SGLTs), could serve as a novel therapeutic intervention for anti-cancer drug discovery as well as provide a unique targeting approach for drug delivery to specific tumor tissues. This review aims to discussthe previous and emerging research on the expression of various types of glucose transporters in tumor tissues, the role of glucose transport inhibitors as a cancer therapy intervention as well as emerging GLUT/SGLT-mediated drug delivery strategies that can be therapeutically employed to target various cancers.
Collapse
Affiliation(s)
- Sadat Shafi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Mohammad Ahmed Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Kingdom of Saudi Arabia (KSA)
| | - Syed Arman Rabbani
- Department of Clinical Pharmacy and Pharmacology, Ras Al Khaimah College of Pharmacy, Ras Al Khaimah Medical and Health Science University, Ras Al Khaimah, United Arab Emirates
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| |
Collapse
|
10
|
Wang G, Jin W, Zhang L, Dong M, Zhang X, Zhou Z, Wang X. SLC50A1 inhibits the doxorubicin sensitivity in hepatocellular carcinoma cells through regulating the tumor glycolysis. Cell Death Discov 2024; 10:495. [PMID: 39695152 DOI: 10.1038/s41420-024-02261-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/20/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
Metabolic reprogramming has been found to be closely associated with the occurrence and development of hepatocellular carcinoma (HCC). The relationship between SLC50A1, a member of the SLC family involved in glucose transmembrane transport, and HCC remains unclear. This study aims to investigate the function and underlying mechanisms of SLC50A1 in the occurrence and progression of HCC. Based on bioinformatics analysis and clinical sample testing, we observed a significant upregulation of SLC50A1 in HCC, which is correlated with unfavorable prognosis in HCC patients. Additionally, there is a noticeable correlation between the expressions of SLC50A1 and METTL3. Further in vitro and in vivo experiments confirmed that SLC50A1 can regulate cellular glycolysis and the cell cycle, thereby promoting the proliferation of HCC cells while reducing apoptosis. Moreover, our findings indicate that SLC50A1 enhances resistance of HCC cells to DOX and 2-DG. Furthermore, we discovered that the m6A methyltransferase METTL3 mediates the methylation modification of SLC50A1. The recognition and binding of the modified SLC50A1 by IGF2BP2 subsequently promote its stability and translational expression. Consequently, our research identifies the METTL3/SLC50A1 axis as a novel therapeutic target in the context of HCC.
Collapse
Affiliation(s)
- Ganggang Wang
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Wenzhi Jin
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Lianmei Zhang
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Meiyuan Dong
- Graduate School of Hebei Medical University, Shijiazhuang, People's Republic of China
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xin Zhang
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Zhijie Zhou
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xiaoliang Wang
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.
| |
Collapse
|
11
|
Zhang Y, Liao X, Xu J, Yin J, Li S, Li M, Shi X, Zhang S, Li C, Xu W, Yu X, Yang Y. The Promising Potency of Sodium-Glucose Cotransporter 2 Inhibitors in the Prevention of and as Treatment for Cognitive Impairment Among Type 2 Diabetes Patients. Biomedicines 2024; 12:2783. [PMID: 39767690 PMCID: PMC11673520 DOI: 10.3390/biomedicines12122783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 11/28/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM), accounting for the majority of diabetes mellitus prevalence, is associated with an increased risk of cognition decline and deterioration of cognition function in diabetic patients. The sodium-glucose cotransporter 2 (SGLT2), located in the renal proximal tubule, plays a role in urine glucose reabsorption. SGLT2 inhibitors (SGLT2i), have shown potential benefits beyond cardiac and renal improvement in preventing and treating cognitive impairment (CI), including mild cognitive impairment, Alzheimer's disease and vascular dementia in T2DM patients. Studies suggest that SGLT2i may ameliorate diabetic CI through metabolism pathways, inflammation, oxidative stress, neurotrophic factors and AChE inhibition. Clinical trials and meta-analyses have reported significant and insignificant results. Given their vascular effects, SGLT2i may offer unique protection against vascular CI. This review compiles mechanisms and clinical evidence, emphasizing the need for future analysis, evaluation, trials and meta-analyses to verify and recommend optimal SGLT2i selection and dosage for specific patients.
Collapse
Affiliation(s)
- Yibin Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Second Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaobin Liao
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Second Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jialu Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Jiaxin Yin
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Shan Li
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Mengni Li
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Xiaoli Shi
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Shujun Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Chunyu Li
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Weijie Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Xuefeng Yu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Yan Yang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Z.); (X.L.); (J.X.); (J.Y.); (S.L.); (M.L.); (X.S.); (S.Z.); (C.L.); (W.X.); (X.Y.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| |
Collapse
|
12
|
Peng C, Ghanbari M, May A, Abeel T. Effects of antibiotic growth promoter and its natural alternative on poultry cecum ecosystem: an integrated analysis of gut microbiota and host expression. Front Microbiol 2024; 15:1492270. [PMID: 39687871 PMCID: PMC11646981 DOI: 10.3389/fmicb.2024.1492270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 11/05/2024] [Indexed: 12/18/2024] Open
Abstract
Background In-feed antibiotic growth promoters (AGPs) have been a cornerstone in the livestock industry due to their role in enhancing growth and feed efficiency. However, concerns over antibiotic resistance have driven a shift away from AGPs toward natural alternatives. Despite the widespread use, the exact mechanisms of AGPs and alternatives are not fully understood. This necessitates holistic studies that investigate microbiota dynamics, host responses, and the interactions between these elements in the context of AGPs and alternative feed additives. Methods In this study, we conducted a multifaceted investigation of how Bacitracin, a common AGP, and a natural alternative impact both cecum microbiota and host expression in chickens. In addition to univariate and static differential abundance and expression analyses, we employed multivariate and time-course analyses to study this problem. To reveal host-microbe interactions, we assessed their overall correspondence and identified treatment-specific pairs of species and host expressed genes that showed significant correlations over time. Results Our analysis revealed that factors such as developmental age substantially impacted the cecum ecosystem more than feed additives. While feed additives significantly altered microbial compositions in the later stages, they did not significantly affect overall host gene expression. The differential expression indicated that with AGP administration, host transmembrane transporters and metallopeptidase activities were upregulated around day 21. Together with the modulated kininogen binding and phenylpyruvate tautomerase activity over time, this likely contributes to the growth-promoting effects of AGPs. The difference in responses between AGP and PFA supplementation suggests that these additives operate through distinct mechanisms. Conclusion We investigated the impact of a common AGP and its natural alternative on poultry cecum ecosystem through an integrated analysis of both the microbiota and host responses. We found that AGP appears to enhance host nutrient utilization and modulate immune responses. The insights we gained are critical for identifying and developing effective AGP alternatives to advance sustainable livestock farming practices.
Collapse
Affiliation(s)
- Chengyao Peng
- Delft Bioinformatics Lab, Delft University of Technology, Delft, Netherlands
| | - Mahdi Ghanbari
- dsm-firmenich, Animal Nutrition and Health R&D Center, Tulln, Austria
| | - Ali May
- dsm-firmenich, Science and Research, Delft, Netherlands
| | - Thomas Abeel
- Delft Bioinformatics Lab, Delft University of Technology, Delft, Netherlands
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
13
|
Spellman MJ, Assaf T, Nangia S, Fernandez J, Nicholson KC, Shepard BD. Handling the sugar rush: the role of the renal proximal tubule. Am J Physiol Renal Physiol 2024; 327:F1013-F1025. [PMID: 39447117 PMCID: PMC11687834 DOI: 10.1152/ajprenal.00265.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 10/26/2024] Open
Abstract
Blood glucose homeostasis is critical to ensure the proper functioning of the human body. Through the processes of filtration, reabsorption, secretion, and metabolism, much of this task falls to the kidneys. With a rise in glucose and other added sugars, there is an increased burden on this organ, mainly the proximal tubule, which is responsible for all glucose reabsorption. In this review, we focus on the current physiological and cell biological functions of the renal proximal tubule as it works to reabsorb and metabolize glucose and fructose. We also highlight the physiological adaptations that occur within the proximal tubule as sugar levels rise under pathophysiological conditions including diabetes. This includes the detrimental impacts of an excess glucose load that leads to glucotoxicity. Finally, we explore some of the emerging therapeutics that modulate renal glucose handling and the systemic protection that can be realized by targeting the reabsorptive properties of the kidney.
Collapse
Affiliation(s)
- Michael J Spellman
- Department of Human Science, Georgetown University, Washington, District of Columbia, United States
| | - Tala Assaf
- Department of Human Science, Georgetown University, Washington, District of Columbia, United States
| | - Shivani Nangia
- Department of Human Science, Georgetown University, Washington, District of Columbia, United States
| | - Joel Fernandez
- Department of Human Science, Georgetown University, Washington, District of Columbia, United States
| | - Kyle C Nicholson
- Department of Human Science, Georgetown University, Washington, District of Columbia, United States
| | - Blythe D Shepard
- Department of Human Science, Georgetown University, Washington, District of Columbia, United States
| |
Collapse
|
14
|
Pham LT, Mangmool S, Parichatikanond W. Sodium-Glucose Cotransporter 2 (SGLT2) Inhibitors: Guardians against Mitochondrial Dysfunction and Endoplasmic Reticulum Stress in Heart Diseases. ACS Pharmacol Transl Sci 2024; 7:3279-3298. [PMID: 39539254 PMCID: PMC11555527 DOI: 10.1021/acsptsci.4c00240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/11/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors are an innovative class of antidiabetic drugs that provide cardiovascular benefits to both diabetic and nondiabetic patients, surpassing those of other antidiabetic drugs. Although the roles of mitochondria and endoplasmic reticulum (ER) in cardiovascular research are increasingly recognized as promising therapeutic targets, the exact molecular mechanisms by which SGLT2 inhibitors influence mitochondrial and ER homeostasis in the heart remain incompletely elucidated. This review comprehensively summarizes and discusses the impacts of SGLT2 inhibitors on mitochondrial dysfunction and ER stress in heart diseases including heart failure, ischemic heart disease/myocardial infarction, and arrhythmia from preclinical and clinical studies. Based on the existing evidence, the effects of SGLT2 inhibitors may potentially involve the restoration of mitochondrial biogenesis and alleviation of ER stress. Such consequences are achieved by enhancing adenosine triphosphate (ATP) production, preserving mitochondrial membrane potential, improving the activity of electron transport chain complexes, maintaining mitochondrial dynamics, mitigating oxidative stress and apoptosis, influencing cellular calcium and sodium handling, and targeting the unfolded protein response (UPR) through three signaling pathways including inositol requiring enzyme 1α (IRE1α), protein kinase R like endoplasmic reticulum kinase (PERK), and activating transcription factor 6 (ATF6). Therefore, SGLT2 inhibitors have emerged as a promising target for treating heart diseases due to their potential to improve mitochondrial functions and ER stress.
Collapse
Affiliation(s)
- Linh Thi
Truc Pham
- Biopharmaceutical
Sciences Program, Faculty of Pharmacy, Mahidol
University, Bangkok, 10400 Thailand
- Department
of Pharmacology, Faculty of Pharmacy, Mahidol
University, Bangkok, 10400 Thailand
| | - Supachoke Mangmool
- Department
of Pharmaceutical Care, Faculty of Pharmacy, Chiang Mai University, Chiang
Mai, 50200 Thailand
| | | |
Collapse
|
15
|
Mishra S, Stany B, Das A, Kanagavel D, Vijayan M. A Comprehensive Review of Membrane Transporters and MicroRNA Regulation in Alzheimer's Disease. Mol Neurobiol 2024; 61:8739-8758. [PMID: 38558361 DOI: 10.1007/s12035-024-04135-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024]
Abstract
Alzheimer's disease (AD) is a distressing neurodegenerative condition characterized by the accumulation of amyloid-beta (Aβ) plaques and tau tangles within the brain. The interconnectedness between membrane transporters (SLCs) and microRNAs (miRNAs) in AD pathogenesis has gained increasing attention. This review explores the localization, substrates, and functions of SLC transporters in the brain, emphasizing the roles of transporters for glutamate, glucose, nucleosides, and other essential compounds. The examination delves into the significance of SLCs in AD, their potential for drug development, and the intricate realm of miRNAs, encompassing their transcription, processing, functions, and regulation. MiRNAs have emerged as significant players in AD, including those associated with mitochondria and synapses. Furthermore, this review discusses the intriguing nexus of miRNAs targeting SLC transporters and their potential as therapeutic targets in AD. Finally, the review underscores the interaction between SLC transporters and miRNA regulation within the context of Alzheimer's disease, underscoring the need for further research in this area. This comprehensive review aims to shed light on the complex mechanisms underlying the causation of AD and provides insights into potential therapeutic approaches.
Collapse
Affiliation(s)
- Shatakshi Mishra
- School of Biosciences and Technology, Department of Biotechnology, VIT University, Vellore, Tamil Nadu, 632014, India
| | - B Stany
- School of Biosciences and Technology, Department of Biotechnology, VIT University, Vellore, Tamil Nadu, 632014, India
| | - Anushka Das
- School of Biosciences and Technology, Department of Biotechnology, VIT University, Vellore, Tamil Nadu, 632014, India
| | - Deepankumar Kanagavel
- School of Biosciences and Technology, Department of Biotechnology, VIT University, Vellore, Tamil Nadu, 632014, India.
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX, 79430, USA.
| |
Collapse
|
16
|
Zhang L, Wu M, Zhang J, Liu T, Fu S, Wang Y, Xu Z. The pivotal role of glucose transporter 1 in diabetic kidney disease. Life Sci 2024; 353:122932. [PMID: 39067659 DOI: 10.1016/j.lfs.2024.122932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/12/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
Diabetes mellitus (DM) is a significant public health problem. Diabetic kidney disease (DKD) is the most common complication of DM, and its incidence has been increasing with the increasing prevalence of DM. Given the association between DKD and mortality in patients with DM, DKD is a significant burden on public health resources. Despite its significance in DM progression, the pathogenesis of DKD remains unclear. Aberrant glucose uptake by cells is an important pathophysiological mechanism underlying DKD renal injury. Glucose is transported across the bilayer cell membrane by a glucose transporter (GLUT) located on the cell membrane. Multiple GLUT proteins have been identified in the kidney, and GLUT1 is one of the most abundantly expressed isoforms. GLUT1 is a crucial regulator of intracellular glucose metabolism and plays a key pathological role in the phenotypic changes in DKD mesangial cells. In an attempt to understand the pathogenesis of DKD better, we here present a review of studies on the role of GLUT1 in the development and progression of DKD.
Collapse
Affiliation(s)
- Li Zhang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Meiyan Wu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jizhou Zhang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Tingting Liu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Shaojie Fu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yue Wang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Zhonggao Xu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
17
|
Baghel R, Chhikara N, Kumar P, Tamrakar AK. SGLT2 inhibitors for the treatment of diabetes: a patent review (2019-23). Expert Opin Ther Pat 2024; 34:807-823. [PMID: 39078140 DOI: 10.1080/13543776.2024.2379929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/27/2024] [Accepted: 07/04/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION The sodium-glucose co-transporter 2 (SGLT2) inhibitors are FDA-approved class of drugs for diabetes management. They improve glycemic control by inducing glucosuria. Notwithstanding with potent anti-hyperglycemic activity, SGLT2 inhibitors are emerging as drugs with multifaceted therapeutic potential, evidenced for cardioprotective, renoprotective, antihypertensive, and neuroprotective activities. Continuous attempts are being accomplished through structural modification, development of new formulation, or combination with other drugs, to enhance the bioactivity spectrum of SGLT2 inhibitors for better management of diabetes and related complications. AREAS COVERED This review comprises a summary of patent applications, acquired using the Espacenet Patent Search database, concerning SGLT2 inhibitors from 2019 to 2023, with focus on improving therapeutic potentials in management of diabetes and metabolic complications. EXPERT OPINION SGLT2 inhibitors have provided an exciting treatment option for diabetes. Originally developed as anti-hyperglycemic agents, SGLT2 inhibitors exert pleiotropic metabolic responses and have emerged as promising antidiabetic agents with cardio-protective and reno-protective activities. Given their distinct therapeutic profile, SGLT2 inhibitors have revolutionized the management of diabetes and associated complications. Emerging evidences on their therapeutic potential against cancer, male reproductive dysfunctions, and neurodegenerative diseases indicate that further research in this field may unfold novel prospective on their plausible use in the management of other chronic conditions.
Collapse
Affiliation(s)
- Rahul Baghel
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad, Ghaziabad, India
| | - Nikita Chhikara
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad, Ghaziabad, India
| | - Pawan Kumar
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad, Ghaziabad, India
| | - Akhilesh Kumar Tamrakar
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad, Ghaziabad, India
| |
Collapse
|
18
|
U N, R C T, R KR, Mahalingam G. Glucose transporters and their energy homeostasis function in various organs. VITAMINS AND HORMONES 2024; 128:1-47. [PMID: 40097247 DOI: 10.1016/bs.vh.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Glucose transporters (GLUTs) belong to a membrane-protein family that essentially participates in easing the transportation and absorption of glucose molecules throughout the cellular membranes. From the brain to the eyes, each section delves into the intricate mechanisms of glucose uptake and utilization, shedding light on the unique adaptations and regulatory pathways in different anatomical structures. Beginning with the brain, known for its high energy demands, the chapter explicates the specialized GLUT expression patterns crucial for neuronal function and synaptic transmission. Moving to metabolic powerhouses like the liver, muscles, and adipose tissue, it elucidates the dynamic interplay of GLUT isoforms in energy storage, mobilization, and insulin responsiveness. Furthermore, the chapter navigates through the kidneys, lungs, skin, and reproductive organs, unveiling the diverse roles of GLUTs in renal glucose reabsorption, pulmonary-epithelial transportation, skin barrier associated functions, and gonadal development. It also explores the significance of placental GLUTs in fatal nutrient supply and the implications of cardiac GLUTs in myocardial energy metabolism. Additionally, it examines the intricate regulation of GLUTs at key barriers like the BBB (Blood-Brain Barrier) and placenta, as well as in endocrine glands such as the pancreas, adrenal medulla and thyroid. Moreover, it further elucidates the less-explored territories of GLUT expression in the bones, gastrointestinal tract, and ocular tissues like the retina, unraveling their implications in immune function, bone metabolism, intestinal glucose-sensing, and retinal physiology.
Collapse
Affiliation(s)
- Nithya U
- Department of Bio-Medical Sciences, School of Bio, sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Theijeswini R C
- Department of Bio-Medical Sciences, School of Bio, sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Karthick Raja R
- Department of Bio-Medical Sciences, School of Bio, sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Gayathri Mahalingam
- Department of Bio-Medical Sciences, School of Bio, sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| |
Collapse
|
19
|
Golovina E, Heizer T, Daumova L, Bajecny M, Fontana S, Griggio V, Jones R, Coscia M, Riganti C, Savvulidi Vargova K. MiR-155 deficiency and hypoxia results in metabolism switch in the leukemic B-cells. Cancer Cell Int 2024; 24:251. [PMID: 39020347 PMCID: PMC11256420 DOI: 10.1186/s12935-024-03437-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 07/08/2024] [Indexed: 07/19/2024] Open
Abstract
Hypoxia represents one of the key factors that stimulates the growth of leukemic cells in their niche. Leukemic cells in hypoxic conditions are forced to reprogram their original transcriptome, miRNome, and metabolome. How the coupling of microRNAs (miRNAs)/mRNAs helps to maintain or progress the leukemic status is still not fully described. MiRNAs regulate practically all biological processes within cells and play a crucial role in the development/progression of leukemia. In the present study, we aimed to uncover the impact of hsa-miR-155-5p (miR-155, MIR155HG) on the metabolism, proliferation, and mRNA/miRNA network of human chronic lymphocytic leukemia cells (CLL) in hypoxic conditions. As a model of CLL, we used the human MEC-1 cell line where we deleted mature miR-155 with CRISPR/Cas9. We determined that miR-155 deficiency in leukemic MEC-1 cells results in lower proliferation even in hypoxic conditions in comparison to MEC-1 control cells. Additionally, in MEC-1 miR-155 deficient cells we observed decreased number of populations of cells in S phase. The miR-155 deficiency under hypoxic conditions was accompanied by an increased apoptosis. We detected a stimulatory effect of miR-155 deficiency and hypoxia at the level of gene expression, seen in significant overexpression of EGLN1, GLUT1, GLUT3 in MEC-1 miR-155 deficient cells. MiR-155 deficiency and hypoxia resulted in increase of glucose and lactate uptake. Pyruvate, ETC and ATP were reduced. To conclude, miR-155 deficiency and hypoxia affects glucose and lactate metabolism by stimulating the expression of glucose transporters as GLUT1, GLUT3, and EGLN1 [Hypoxia-inducible factor prolyl hydroxylase 2 (HIF-PH2)] genes in the MEC-1 cells.
Collapse
Affiliation(s)
- Elena Golovina
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tomas Heizer
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Center for Advanced Preclinical Imaging (CAPI), First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lenka Daumova
- Institute Biocev, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Martin Bajecny
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Simona Fontana
- Oncological Pharmacology Laboratory, Department of Oncology, University of Torino, Torino, Italy
| | - Valentina Griggio
- Department of Molecular Biotechnology and Health Sciences, University of Torino and Division of Hematology, A.O.U. Città Della Salute E Della Scienza Di Torino, Torino, Italy
| | - Rebecca Jones
- Department of Molecular Biotechnology and Health Sciences, University of Torino and Division of Hematology, A.O.U. Città Della Salute E Della Scienza Di Torino, Torino, Italy
| | - Marta Coscia
- Department of Molecular Biotechnology and Health Sciences, University of Torino and Division of Hematology, A.O.U. Città Della Salute E Della Scienza Di Torino, Torino, Italy
| | - Chiara Riganti
- Oncological Pharmacology Laboratory, Department of Oncology, University of Torino, Torino, Italy
| | - Karina Savvulidi Vargova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
20
|
Salazar OR, Chen K, Melino VJ, Reddy MP, Hřibová E, Čížková J, Beránková D, Arciniegas Vega JP, Cáceres Leal LM, Aranda M, Jaremko L, Jaremko M, Fedoroff NV, Tester M, Schmöckel SM. SOS1 tonoplast neo-localization and the RGG protein SALTY are important in the extreme salinity tolerance of Salicornia bigelovii. Nat Commun 2024; 15:4279. [PMID: 38769297 PMCID: PMC11106269 DOI: 10.1038/s41467-024-48595-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 05/07/2024] [Indexed: 05/22/2024] Open
Abstract
The identification of genes involved in salinity tolerance has primarily focused on model plants and crops. However, plants naturally adapted to highly saline environments offer valuable insights into tolerance to extreme salinity. Salicornia plants grow in coastal salt marshes, stimulated by NaCl. To understand this tolerance, we generated genome sequences of two Salicornia species and analyzed the transcriptomic and proteomic responses of Salicornia bigelovii to NaCl. Subcellular membrane proteomes reveal that SbiSOS1, a homolog of the well-known SALT-OVERLY-SENSITIVE 1 (SOS1) protein, appears to localize to the tonoplast, consistent with subcellular localization assays in tobacco. This neo-localized protein can pump Na+ into the vacuole, preventing toxicity in the cytosol. We further identify 11 proteins of interest, of which SbiSALTY, substantially improves yeast growth on saline media. Structural characterization using NMR identified it as an intrinsically disordered protein, localizing to the endoplasmic reticulum in planta, where it can interact with ribosomes and RNA, stabilizing or protecting them during salt stress.
Collapse
Affiliation(s)
- Octavio R Salazar
- Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
- Center for Desert Agriculture, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
- Red Sea Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Ke Chen
- Rice Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Vanessa J Melino
- Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
- Center for Desert Agriculture, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Muppala P Reddy
- Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
- Center for Desert Agriculture, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Eva Hřibová
- Institute of Experimental Botany of the Czech Academy of Sciences, Centre of Plant Structural and Functional Genomics, Šlechtitelů 31, 77900, Olomouc, Czech Republic
| | - Jana Čížková
- Institute of Experimental Botany of the Czech Academy of Sciences, Centre of Plant Structural and Functional Genomics, Šlechtitelů 31, 77900, Olomouc, Czech Republic
| | - Denisa Beránková
- Institute of Experimental Botany of the Czech Academy of Sciences, Centre of Plant Structural and Functional Genomics, Šlechtitelů 31, 77900, Olomouc, Czech Republic
| | - Juan Pablo Arciniegas Vega
- Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
- Center for Desert Agriculture, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Lina María Cáceres Leal
- Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
- Center for Desert Agriculture, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Manuel Aranda
- Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
- Red Sea Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Lukasz Jaremko
- Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Mariusz Jaremko
- Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Nina V Fedoroff
- Department of Biology, Penn State University, University Park, PA, 16801, US
| | - Mark Tester
- Biological and Environmental Sciences & Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia.
- Center for Desert Agriculture, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia.
| | - Sandra M Schmöckel
- Department Physiology of Yield Stability, Institute of Crop Science, University of Hohenheim, Fruwirthstr. 21, 70599, Stuttgart, Germany
| |
Collapse
|
21
|
Geist BK, Ramirez JC, Binder P, Einspieler H, Ibeschitz H, Langsteger W, Nics L, Rausch I, Diemling M, Sohlberg A, Hacker M, Rasul S. In vivo assessment of safety, biodistribution, and radiation dosimetry of the [ 18F]Me4FDG PET-radiotracer in adults. EJNMMI Res 2024; 14:46. [PMID: 38750398 PMCID: PMC11096136 DOI: 10.1186/s13550-024-01098-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/22/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Approaches targeting the sodium-glucose cotransporter (SGLT) could represent a promising future therapeutic strategy for numerous oncological and metabolic diseases. In this study, we evaluated the safety, biodistribution and radiation dosimetry of the glucose analogue positron emission tomography (PET) agent [18F] labeled alpha-methyl-4-deoxy-4-[18F]fluoro-D-glucopyranoside ([18F]Me4FDG) with high sodium-glucose cotransporter and low glucose transporter (GLUT) affinity. For this purpose, five healthy volunteers (1 man, 4 women) underwent multiple whole-body PET/computed tomography (CT) examinations starting with injection and up to 4 h after injection of averaged (2.4 ± 0.1) MBq/kg (range: 2.3-2.5 MBq/kg) administered activity. The PET/CT scans were conducted in 5 separate sessions, blood pressure and temperature were measured, and blood and urine samples were collected before the scans and one hour after injection to assess toxicity. Measurements of [18F]Me4FDG radioactivity in organs of interest were determined from the PET/CT scans at 5 time points. Internal dosimetry was performed on voxel level using a fast Monte Carlo approach. RESULTS All studied volunteers could well tolerate the [18F]Me4FDG and no adverse event was reported. The calculated effective dose was (0.013 ± 0.003) mSv/MBq. The organs with the highest absorbed dose were the kidneys with 0.05 mSv/MBq per kidney. The brain showed almost no uptake. After 60 min, (12 ± 15) % of the administered dose was excreted into the bladder. CONCLUSION Featuring an effective dose of only 0.013 ± 0.003 mSv/MBq and no occurrence of side effects, the glucose analogue [18F]Me4FDG seems to be a safe radio-tracer with a favorable biodistribution for PET imaging and also within several consecutive scans. TRIAL REGISTRATION NUMBER NCT03557138, Registered 22 February 2017, https://ichgcp.net/clinical-trials-registry/NCT03557138 .
Collapse
Affiliation(s)
| | | | - Patrick Binder
- Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Holger Einspieler
- Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Harald Ibeschitz
- Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Werner Langsteger
- Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Lukas Nics
- Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Ivo Rausch
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | | | - Antti Sohlberg
- Hermes Medical Solutions, Vienna, Austria
- Department of Nuclear Medicine, Päijät-Häme Central Hospital, Lahti, Finland
| | - Marcus Hacker
- Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria.
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Floor 5L, Vienna, 1090, Austria.
| | - Sazan Rasul
- Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
22
|
Morace C, Lorello G, Bellone F, Quartarone C, Ruggeri D, Giandalia A, Mandraffino G, Minutoli L, Squadrito G, Russo GT, Marini HR. Ketoacidosis and SGLT2 Inhibitors: A Narrative Review. Metabolites 2024; 14:264. [PMID: 38786741 PMCID: PMC11122992 DOI: 10.3390/metabo14050264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
An acute metabolic complication of diabetes mellitus, especially type 1, is diabetic ketoacidosis (DKA), which is due to an increase in blood ketone concentrations. Sodium/glucose co-transporter-2 inhibitor (SGLT2-i) drugs have been associated with the occurrence of a particular type of DKA defined as euglycemic (euDKA), characterized by glycemic levels below 300 mg/dL. A fair number of euDKA cases in SGLT2-i-treated patients have been described, especially in the last few years when there has been a significant increased use of these drugs. This form of euDKA is particularly insidious because of its latent onset, associated with unspecific symptomatology, until it evolves (progressing) to severe systemic forms. In addition, its atypical presentation can delay diagnosis and treatment. However, the risk of euDKA associated with SGLT2-i drugs remains relatively low, but it is essential to promptly diagnose and manage it to prevent its serious life-threatening complications. In this narrative review, we intended to gather current research evidence on SGLT2i-associated euDKA from randomized controlled trials and real-world evidence studies, its diagnostic criteria and precipitating factors.
Collapse
Affiliation(s)
- Carmela Morace
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.M.); (F.B.); (G.M.); (L.M.); (G.S.); (G.T.R.)
- Lipid Clinic and Cardiometabolic Disease Center, University Hospital of Messina, 98124 Messina, Italy
| | - Giuseppe Lorello
- Internal Medicine and Diabetology Unit, University Hospital of Messina, 98124 Messina, Italy; (G.L.); (C.Q.); (D.R.); (A.G.)
| | - Federica Bellone
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.M.); (F.B.); (G.M.); (L.M.); (G.S.); (G.T.R.)
- Lipid Clinic and Cardiometabolic Disease Center, University Hospital of Messina, 98124 Messina, Italy
| | - Cristina Quartarone
- Internal Medicine and Diabetology Unit, University Hospital of Messina, 98124 Messina, Italy; (G.L.); (C.Q.); (D.R.); (A.G.)
| | - Domenica Ruggeri
- Internal Medicine and Diabetology Unit, University Hospital of Messina, 98124 Messina, Italy; (G.L.); (C.Q.); (D.R.); (A.G.)
| | - Annalisa Giandalia
- Internal Medicine and Diabetology Unit, University Hospital of Messina, 98124 Messina, Italy; (G.L.); (C.Q.); (D.R.); (A.G.)
- Department of Human Pathology of Adulthood and Childhood “G. Barresi”, University of Messina, 98125 Messina, Italy
| | - Giuseppe Mandraffino
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.M.); (F.B.); (G.M.); (L.M.); (G.S.); (G.T.R.)
- Lipid Clinic and Cardiometabolic Disease Center, University Hospital of Messina, 98124 Messina, Italy
| | - Letteria Minutoli
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.M.); (F.B.); (G.M.); (L.M.); (G.S.); (G.T.R.)
| | - Giovanni Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.M.); (F.B.); (G.M.); (L.M.); (G.S.); (G.T.R.)
- Internal Medicine and Diabetology Unit, University Hospital of Messina, 98124 Messina, Italy; (G.L.); (C.Q.); (D.R.); (A.G.)
| | - Giuseppina T. Russo
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.M.); (F.B.); (G.M.); (L.M.); (G.S.); (G.T.R.)
- Internal Medicine and Diabetology Unit, University Hospital of Messina, 98124 Messina, Italy; (G.L.); (C.Q.); (D.R.); (A.G.)
| | - Herbert Ryan Marini
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.M.); (F.B.); (G.M.); (L.M.); (G.S.); (G.T.R.)
- Internal Medicine and Diabetology Unit, University Hospital of Messina, 98124 Messina, Italy; (G.L.); (C.Q.); (D.R.); (A.G.)
| |
Collapse
|
23
|
Weinrauch AM, Bouyoucos IA, Conlon JM, Anderson WG. The chondrichthyan glucagon-like peptide 3 regulates hepatic ketone metabolism in the Pacific spiny dogfish Squalus suckleyi. Gen Comp Endocrinol 2024; 350:114470. [PMID: 38346454 DOI: 10.1016/j.ygcen.2024.114470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/07/2023] [Accepted: 02/03/2024] [Indexed: 02/17/2024]
Abstract
Chondrichthyans have a novel proglucagon-derived peptide, glucagon-like peptide (GLP)-3, in addition to GLP-1 and GLP-2 that occur in other vertebrates. Given that the GLPs are important regulators of metabolic homeostasis across vertebrates, we sought to investigate whether GLP-3 displays functional actions on metabolism within a representative chondrichthyan, the Pacific spiny dogfish Squalus suckleyi. There were no observed effects of GLP-3 perfusion (10 nM for 15 min) on the rate of glucose or oleic acid acquisition at the level of the spiral valve nor were there any measured effects on intermediary metabolism within this tissue. Despite no effects on apparent glucose transport or glycolysis in the liver, a significant alteration to ketone metabolism occurred. Firstly, ketone flux through the perfused liver switched from a net endogenous production to consumption following hormone application. Accompanying this change, significant increases in mRNA transcript abundance of putative ketone transporters and in the activity of β-hydroxybutyrate dehydrogenase (a key enzyme regulating ketone flux in the liver) were observed. Overall, while these results show effects on hepatic metabolism, the physiological actions of GLP are distinct between this chondrichthyan and those of GLP-1 on teleost fishes. Whether this is the result of the particular metabolic dependency on ketone bodies in chondrichthyans or a differential function of a novel GLP remains to be fully elucidated.
Collapse
Affiliation(s)
- Alyssa M Weinrauch
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; Bamfield Marine Sciences Centre, Bamfield, BC V0R 1B0, Canada.
| | - Ian A Bouyoucos
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; Bamfield Marine Sciences Centre, Bamfield, BC V0R 1B0, Canada
| | - J Michael Conlon
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, Northern Ireland, UK
| | - W Gary Anderson
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; Bamfield Marine Sciences Centre, Bamfield, BC V0R 1B0, Canada
| |
Collapse
|
24
|
Song W, Fu J, Wu J, Ren J, Xiang R, Kong C, Fu T. CircFBXW4 Suppresses Colorectal Cancer Progression by Regulating the MiR-338-5p/SLC5A7 Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2300129. [PMID: 38461489 PMCID: PMC11095154 DOI: 10.1002/advs.202300129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/04/2024] [Indexed: 03/12/2024]
Abstract
Dysregulated circular RNAs (circRNAs) contribute to tumourigenesis and cancer progression. However, the expression patterns and biological functions of circRNAs in colorectal cancer (CRC) remain elusive. Here, RNA sequencing and bioinformatics analyses are applied to screen for aberrantly expressed circRNAs. The expression of circFBXW4 in CRC tissues and cell lines is determined by quantitative real-time PCR. A series of in vitro and in vivo biological function assays are implemented to assess the functions of circFBXW4. The regulatory mechanisms linking circFBXW4, miR-338-5p, and SLC5A7 are explored by western blotting, dual luciferase reporter assays, and RNA pull-down assays. CircFBXW4 is dramatically downregulated in CRC tissues and cell lines. circFBXW4 downregulation is clearly correlated with malignant features and patient overall survival in CRC. Functionally, ectopic expression of circFBXW4 strikingly impairs the proliferation, migration, and invasion capacities of CRC cells in vitro and in vivo, whereas circFBXW4 knockdown has the opposite effects. Mechanistically, circFBXW4 competitively binds to miR-338-5p and prevents it from interacting with and repressing its target SLC5A7, thus suppressing the progression of CRC. This study reveals the specific critical role of circFBXW4 in inhibiting CRC progression via the miR-338-5p/SLC5A7 axis and provides an additional target for eradicating CRC.
Collapse
Affiliation(s)
- Wei Song
- Department of Gastrointestinal Surgery IIRenmin Hospital of Wuhan UniversityWuhan430060P. R. China
| | - Jincheng Fu
- Department of Gastrointestinal Surgery IIRenmin Hospital of Wuhan UniversityWuhan430060P. R. China
- Department of General SurgeryQingdao Municipal HospitalQingdao266071P. R. China
| | - Jing Wu
- Department of Gastrointestinal Surgery IIRenmin Hospital of Wuhan UniversityWuhan430060P. R. China
| | - Jun Ren
- Department of Gastrointestinal Surgery IIRenmin Hospital of Wuhan UniversityWuhan430060P. R. China
| | - Rensheng Xiang
- Department of Gastrointestinal Surgery IIRenmin Hospital of Wuhan UniversityWuhan430060P. R. China
| | - Can Kong
- Department of Gastrointestinal Surgery IIRenmin Hospital of Wuhan UniversityWuhan430060P. R. China
| | - Tao Fu
- Department of General SurgeryQingdao Municipal HospitalQingdao266071P. R. China
| |
Collapse
|
25
|
Mizutani K, Yoshida Y, Nakanishi E, Miyata Y, Tokumoto S, Fuse H, Gusev O, Kikuta S, Kikawada T. A sodium-dependent trehalose transporter contributes to anhydrobiosis in insect cell line, Pv11. Proc Natl Acad Sci U S A 2024; 121:e2317254121. [PMID: 38551840 PMCID: PMC10998604 DOI: 10.1073/pnas.2317254121] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 02/13/2024] [Indexed: 04/02/2024] Open
Abstract
Pv11 is the only animal cell line that, when preconditioned with a high concentration of trehalose, can be preserved in the dry state at room temperature for more than one year while retaining the ability to resume proliferation. This extreme desiccation tolerance is referred to as anhydrobiosis. Here, we identified a transporter that contributes to the recovery of Pv11 cells from anhydrobiosis. In general, the solute carrier 5 (SLC5)-type secondary active transporters cotransport Na+ and carbohydrates including glucose. The heterologous expression systems showed that the transporter belonging to the SLC5 family, whose expression increases upon rehydration, exhibits Na+-dependent trehalose transport activity. Therefore, we named it STRT1 (sodium-ion trehalose transporter 1). We report an SLC5 family member that transports a naturally occurring disaccharide, such as trehalose. Knockout of the Strt1 gene significantly reduced the viability of Pv11 cells upon rehydration after desiccation. During rehydration, when intracellular trehalose is no longer needed, Strt1-knockout cells released the disaccharide more slowly than the parental cell line. During rehydration, Pv11 cells became roughly spherical due to osmotic pressure changes, but then returned to their original spindle shape after about 30 min. Strt1-knockout cells, however, required about 50 min to adopt their normal morphology. STRT1 probably regulates intracellular osmolality by releasing unwanted intracellular trehalose with Na+, thereby facilitating the recovery of normal cell morphology during rehydration. STRT1 likely improves the viability of dried Pv11 cells by rapidly alleviating the significant physical stresses that arise during rehydration.
Collapse
Affiliation(s)
- Kosuke Mizutani
- Department of Integrated Biosciences, Graduate School of Frontier Science, The University of Tokyo, Kashiwa, Chiba277-8562, Japan
| | - Yuki Yoshida
- Division of Biomaterial Sciences, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Ibaraki305-8634, Japan
| | - Eita Nakanishi
- Department of Integrated Biosciences, Graduate School of Frontier Science, The University of Tokyo, Kashiwa, Chiba277-8562, Japan
| | - Yugo Miyata
- Department of Medical Chemistry, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo113-8510, Japan
| | - Shoko Tokumoto
- Division of Biomaterial Sciences, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Ibaraki305-8634, Japan
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo113-8421, Japan
| | - Hiroto Fuse
- Department of Integrated Biosciences, Graduate School of Frontier Science, The University of Tokyo, Kashiwa, Chiba277-8562, Japan
| | - Oleg Gusev
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo113-8421, Japan
| | - Shingo Kikuta
- Department of Regional and Comprehensive Agriculture, College of Agriculture, Ibaraki University, Ami, Ibaraki300-0393, Japan
| | - Takahiro Kikawada
- Department of Integrated Biosciences, Graduate School of Frontier Science, The University of Tokyo, Kashiwa, Chiba277-8562, Japan
- Division of Biomaterial Sciences, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Ibaraki305-8634, Japan
| |
Collapse
|
26
|
Sun M, Sun J, Sun W, Li X, Wang Z, Sun L, Wang Y. Unveiling the anticancer effects of SGLT-2i: mechanisms and therapeutic potential. Front Pharmacol 2024; 15:1369352. [PMID: 38595915 PMCID: PMC11002155 DOI: 10.3389/fphar.2024.1369352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/14/2024] [Indexed: 04/11/2024] Open
Abstract
Cancer and diabetes are significant diseases that pose a threat to human health. Their interconnection is complex, particularly when they coexist, often necessitating multiple therapeutic approaches to attain remission. Sodium-glucose cotransporter protein two inhibitors (SGLT-2i) emerged as a treatment for hyperglycemia, but subsequently exhibited noteworthy extra-glycemic properties, such as being registered for the treatment of heart failure and chronic kidney disease, especially with co-existing albuminuria, prompting its assessment as a potential treatment for various non-metabolic diseases. Considering its overall tolerability and established use in diabetes management, SGLT-2i may be a promising candidate for cancer therapy and as a supplementary component to conventional treatments. This narrative review aimed to examine the potential roles and mechanisms of SGLT-2i in the management of diverse types of cancer. Future investigations should focus on elucidating the antitumor efficacy of individual SGLT-2i in different cancer types and exploring the underlying mechanisms. Additionally, clinical trials to evaluate the safety and feasibility of incorporating SGLT-2i into the treatment regimen of specific cancer patients and determining appropriate dosage combinations with established antitumor agents would be of significant interest.
Collapse
Affiliation(s)
- Min Sun
- Department of Geriatrics, First Hospital, Jilin University, Changchun, China
| | - Jilei Sun
- Changchun Traditional Chinese Medicine Hospital, Changchun, China
| | - Wei Sun
- First Affiliated Hospital of Jilin University, Changchun, China
| | - Xiaonan Li
- Department of Geriatrics, First Hospital, Jilin University, Changchun, China
| | - Zhe Wang
- Department of Geriatrics, First Hospital, Jilin University, Changchun, China
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Yuehui Wang
- Department of Geriatrics, First Hospital, Jilin University, Changchun, China
| |
Collapse
|
27
|
Xu X, Khunsriraksakul C, Eales JM, Rubin S, Scannali D, Saluja S, Talavera D, Markus H, Wang L, Drzal M, Maan A, Lay AC, Prestes PR, Regan J, Diwadkar AR, Denniff M, Rempega G, Ryszawy J, Król R, Dormer JP, Szulinska M, Walczak M, Antczak A, Matías-García PR, Waldenberger M, Woolf AS, Keavney B, Zukowska-Szczechowska E, Wystrychowski W, Zywiec J, Bogdanski P, Danser AHJ, Samani NJ, Guzik TJ, Morris AP, Liu DJ, Charchar FJ, Tomaszewski M. Genetic imputation of kidney transcriptome, proteome and multi-omics illuminates new blood pressure and hypertension targets. Nat Commun 2024; 15:2359. [PMID: 38504097 PMCID: PMC10950894 DOI: 10.1038/s41467-024-46132-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
Genetic mechanisms of blood pressure (BP) regulation remain poorly defined. Using kidney-specific epigenomic annotations and 3D genome information we generated and validated gene expression prediction models for the purpose of transcriptome-wide association studies in 700 human kidneys. We identified 889 kidney genes associated with BP of which 399 were prioritised as contributors to BP regulation. Imputation of kidney proteome and microRNAome uncovered 97 renal proteins and 11 miRNAs associated with BP. Integration with plasma proteomics and metabolomics illuminated circulating levels of myo-inositol, 4-guanidinobutanoate and angiotensinogen as downstream effectors of several kidney BP genes (SLC5A11, AGMAT, AGT, respectively). We showed that genetically determined reduction in renal expression may mimic the effects of rare loss-of-function variants on kidney mRNA/protein and lead to an increase in BP (e.g., ENPEP). We demonstrated a strong correlation (r = 0.81) in expression of protein-coding genes between cells harvested from urine and the kidney highlighting a diagnostic potential of urinary cell transcriptomics. We uncovered adenylyl cyclase activators as a repurposing opportunity for hypertension and illustrated examples of BP-elevating effects of anticancer drugs (e.g. tubulin polymerisation inhibitors). Collectively, our studies provide new biological insights into genetic regulation of BP with potential to drive clinical translation in hypertension.
Collapse
Affiliation(s)
- Xiaoguang Xu
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | | | - James M Eales
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Sebastien Rubin
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - David Scannali
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Sushant Saluja
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - David Talavera
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Havell Markus
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Lida Wang
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Maciej Drzal
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Akhlaq Maan
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Abigail C Lay
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Priscilla R Prestes
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Australia
| | - Jeniece Regan
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Avantika R Diwadkar
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Matthew Denniff
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Grzegorz Rempega
- Department of Urology, Medical University of Silesia, Katowice, Poland
| | - Jakub Ryszawy
- Department of Urology, Medical University of Silesia, Katowice, Poland
| | - Robert Król
- Department of General, Vascular and Transplant Surgery, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - John P Dormer
- Department of Cellular Pathology, University Hospitals of Leicester, Leicester, UK
| | - Monika Szulinska
- Department of Obesity, Metabolic Disorders Treatment and Clinical Dietetics, Karol Marcinkowski University of Medical Sciences, Poznan, Poland
| | - Marta Walczak
- Department of Internal Diseases, Metabolic Disorders and Arterial Hypertension, Poznan University of Medical Sciences, Poznan, Poland
| | - Andrzej Antczak
- Department of Urology and Uro-oncology, Karol Marcinkowski University of Medical Sciences, Poznan, Poland
| | - Pamela R Matías-García
- Institute of Epidemiology, Helmholtz Center Munich, Neuherberg, Germany
- Research Unit Molecular Epidemiology, Helmholtz Center Munich, Neuherberg, Germany
- German Research Center for Cardiovascular Disease (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Melanie Waldenberger
- Institute of Epidemiology, Helmholtz Center Munich, Neuherberg, Germany
- Research Unit Molecular Epidemiology, Helmholtz Center Munich, Neuherberg, Germany
- German Research Center for Cardiovascular Disease (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Royal Manchester Children's Hospital and Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Bernard Keavney
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust Manchester, Manchester Royal Infirmary, Manchester, UK
| | | | - Wojciech Wystrychowski
- Department of General, Vascular and Transplant Surgery, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Joanna Zywiec
- Department of Internal Medicine, Diabetology and Nephrology, Zabrze, Medical University of Silesia, Katowice, Poland
| | - Pawel Bogdanski
- Department of Obesity, Metabolic Disorders Treatment and Clinical Dietetics, Karol Marcinkowski University of Medical Sciences, Poznan, Poland
| | - A H Jan Danser
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Tomasz J Guzik
- Department of Internal Medicine, Jagiellonian University Medical College, Kraków, Poland
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Kraków, Poland
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal & Dermatological Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Dajiang J Liu
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Fadi J Charchar
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Australia
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- Department of Physiology, University of Melbourne, Melbourne, Australia
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK.
- Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust Manchester, Manchester Royal Infirmary, Manchester, UK.
| |
Collapse
|
28
|
Zhao T, Zhao Y, Chen H, Sun W, Guan Y. A GC-MS-based untargeted metabolomics approach for comprehensive metabolic profiling of mycophenolate mofetil-induced toxicity in mice. Front Mol Biosci 2024; 11:1332090. [PMID: 38516185 PMCID: PMC10955473 DOI: 10.3389/fmolb.2024.1332090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/08/2024] [Indexed: 03/23/2024] Open
Abstract
Background: Mycophenolate mofetil (MMF), the morpholinoethyl ester of mycophenolic acid, is widely used for maintenance immunosuppression in transplantation. The gastrointestinal toxicity of MMF has been widely uncovered. However, the comprehensive metabolic analysis of MMF-induced toxicity is lacking. This study is aimed to ascertain the metabolic changes after MMF administration in mice. Methods: A total of 700 mg MMF was dissolved in 7 mL dimethyl sulfoxide (DMSO), and then 0.5 mL of mixture was diluted with 4.5 mL of saline (100 mg/kg). Mice in the treatment group (n = 9) were given MMF (0.1 mL/10 g) each day via intraperitoneal injection lasting for 2 weeks, while those in the control group (n = 9) received the same amount of blank solvent (DMSO: saline = 1:9). Gas chromatography-mass spectrometry was utilized to identify the metabolic profiling in serum samples and multiple organ tissues of mice. The potential metabolites were identified using orthogonal partial least squares discrimination analysis. Meanwhile, we used the MetaboAnalyst 5.0 (http://www.metaboanalyst.ca) and Kyoto Encyclopedia of Genes and Genomes database (http://www.kegg.jp) to depict the metabolic pathways. The percentages of lymphocytes in spleens were assessed by multiparameter flow cytometry analysis. Results: Compared to the control group, we observed that MMF treatment induced differential expression of metabolites in the intestine, hippocampus, lung, liver, kidney, heart, serum, and cortex tissues. Subsequently, we demonstrated that multiple amino acids metabolism and fatty acids biosynthesis were disrupted following MMF treatment. Additionally, MMF challenge dramatically increased CD4+ T cell percentages but had no significant influences on other types of lymphocytes. Conclusion: MMF can affect the metabolism in various organs and serum in mice. These data may provide preliminary judgement for MMF-induced toxicity and understand the metabolic mechanism of MMF more comprehensively.
Collapse
Affiliation(s)
- Tongfeng Zhao
- Department of Hematology, Jining No.1 People’s Hospital, Jining, China
| | - Yaxin Zhao
- Department of Pharmacy, Jining No.1 People’s Hospital, Jining, China
| | - Haotian Chen
- Department of Hematology, Jining No.1 People’s Hospital, Jining, China
| | - Wenxue Sun
- Translational Pharmaceutical Laboratory, Jining No.1 People’s Hospital, Jining, China
- Postdoctoral of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yun Guan
- Department of Hematology, Jining No.1 People’s Hospital, Jining, China
| |
Collapse
|
29
|
Zhang C, Gu L, Xie H, Liu Y, Huang P, Zhang J, Luo D, Zhang J. Glucose transport, transporters and metabolism in diabetic retinopathy. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166995. [PMID: 38142757 DOI: 10.1016/j.bbadis.2023.166995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/02/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
Diabetic retinopathy (DR) is the most common reason for blindness in working-age individuals globally. Prolonged high blood glucose is a main causative factor for DR development, and glucose transport is prerequisite for the disturbances in DR caused by hyperglycemia. Glucose transport is mediated by its transporters, including the facilitated transporters (glucose transporter, GLUTs), the "active" glucose transporters (sodium-dependent glucose transporters, SGLTs), and the SLC50 family of uniporters (sugars will eventually be exported transporters, SWEETs). Glucose transport across the blood-retinal barrier (BRB) is crucial for nourishing the neuronal retina in the context of retinal physiology. This physiological process primarily relies on GLUTs and SGLTs, which mediate the glucose transportation across both the cell membrane of retinal capillary endothelial cells and the retinal pigment epithelium (RPE). Under diabetic conditions, increased accumulation of extracellular glucose enhances the retinal cellular glucose uptake and metabolism via both glycolysis and glycolytic side branches, which activates several biochemical pathways, including the protein kinase C (PKC), advanced glycation end-products (AGEs), polyol pathway and hexosamine biosynthetic pathway (HBP). These activated biochemical pathways further increase the production of reactive oxygen species (ROS), leading to oxidative stress and activation of Poly (ADP-ribose) polymerase (PARP). The activated PARP further affects all the cellular components in the retina, and finally resulting in microangiopathy, neurodegeneration and low-to-moderate grade inflammation in DR. This review aims to discuss the changes of glucose transport, glucose transporters, as well as its metabolism in DR, which influences the retinal neurovascular unit (NVU) and implies the possible therapeutic strategies for treating DR.
Collapse
Affiliation(s)
- Chaoyang Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Limin Gu
- Department of Ophthalmology, Shanghai Aier Eye Hospital, Shanghai, China.
| | - Hai Xie
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Yan Liu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Peirong Huang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Jingting Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Dawei Luo
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| |
Collapse
|
30
|
Sun J, Xie F, Wang J, Luo J, Chen T, Jiang Q, Xi Q, Liu GE, Zhang Y. Integrated meta-omics reveals the regulatory landscape involved in lipid metabolism between pig breeds. MICROBIOME 2024; 12:33. [PMID: 38374121 PMCID: PMC10877772 DOI: 10.1186/s40168-023-01743-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 12/19/2023] [Indexed: 02/21/2024]
Abstract
BACKGROUND Domesticated pigs serve as an ideal animal model for biomedical research and also provide the majority of meat for human consumption in China. Porcine intramuscular fat content associates with human health and diseases and is essential in pork quality. The molecular mechanisms controlling lipid metabolism and intramuscular fat accretion across tissues in pigs, and how these changes in response to pig breeds, remain largely unknown. RESULTS We surveyed the tissue-resident cell types of the porcine jejunum, colon, liver, and longissimus dorsi muscle between Lantang and Landrace breeds by single-cell RNA sequencing. Combining lipidomics and metagenomics approaches, we also characterized gene signatures and determined key discriminating markers of lipid digestibility, absorption, conversion, and deposition across tissues in two pig breeds. In Landrace, lean-meat swine mainly exhibited breed-specific advantages in lipid absorption and oxidation for energy supply in small and large intestinal epitheliums, nascent high-density lipoprotein synthesis for reverse cholesterol transport in enterocytes and hepatocytes, bile acid formation, and secretion for fat emulsification in hepatocytes, as well as intestinal-microbiota gene expression involved in lipid accumulation product. In Lantang, obese-meat swine showed a higher synthesis capacity of chylomicrons responsible for high serum triacylglycerol levels in small intestinal epitheliums, the predominant characteristics of lipid absorption in muscle tissue, and greater intramuscular adipcytogenesis potentials from muscular fibro-adipogenic progenitor subpopulation. CONCLUSIONS The findings enhanced our understanding of the cellular biology of lipid metabolism and opened new avenues to improve animal production and human diseases. Video Abstract.
Collapse
Affiliation(s)
- Jiajie Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Fang Xie
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Jing Wang
- Institute of Animal Husbandry and Veterinary Medicine, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Junyi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| | - George E Liu
- Animal Genomics and Improvement Laboratory, USDA-ARS, BARC-East, Beltsville, MD, 20705, USA.
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| |
Collapse
|
31
|
Kusunoki M, Hisano F, Matsuda SI, Kusunoki A, Abe T, Tsutsumi K, Miyata T. Effects of SGLT2 Inhibitors and DPP-4 Inhibitors on Advanced Glycation End Products. Drug Res (Stuttg) 2024; 74:77-80. [PMID: 38286421 DOI: 10.1055/a-2234-1797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2024]
Abstract
Clinical trials have revealed that sodium glucose cotransporter 2 (SGLT2) inhibitors suppress the onset of heart failure and cardiovascular death in diabetic patients. On the other hand, few reports have been published concerning such effects of dipeptidyl peptidase-4 (DPP-4) inhibitors. We undertook the present study to evaluate the effects of SGLT2 inhibitors and DPP-4 inhibitors on the advanced glycation end products (AGEs), well known as a risk factor for the development of cardiovascular disorders.Type 2 diabetes mellitus were divided into two groups and treated with either SGLT2 inhibitors or DPP-4 inhibitors for 3 months. Before and after the 3-month treatment period with each drug, the AGEs and diabetes-related parameters were measured. Methylglyoxal-derived hydroimidazolone-1 (MG-H1) was measured as one of the AGEs.In the SGLT2 inhibitor group, both the blood HbA1c and MG-H1 levels decreased significantly after the 3-month treatment period. In the DPP-4 inhibitor group, only the blood HbA1c level decreased significantly, with no significant change of the blood MG-H1 level.SGLT2 inhibitor reduced both the blood levels of HbA1c and AGEs (MG-H1). Considering that the blood levels of AGEs are associated with the risk of heart failure and cardiovascular disorders, the results of the present study suggest that the effect of SGLT2 inhibitors in suppressing cardiovascular death might be mediated by the reduction in the blood levels of AGEs induced by this class of drugs. DPP-4 inhibitors showed no significant effects on the blood levels of AGEs.
Collapse
Affiliation(s)
- Masataka Kusunoki
- Department of Diabetes, Motor Function and Metabolism, Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya City, Aichi, Japan
| | - Fumiya Hisano
- Graduate School of Medicine, Department of Integrated Health Sciences, Nagoya University, Nagoya City, Aichi, Japan
| | - Shin-Ichi Matsuda
- Department of Data Science, Faculty of Science and Technology, Nanzan University, Nagoya City, Aichi, Japan
| | | | - Tomokazu Abe
- Department of Diabetes, Motor Function and Metabolism, Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya City, Aichi, Japan
| | | | - Tetsuro Miyata
- Office of Medical Education, School of Medicine, International University of Health and Welfare, Narita City, Chiba, Japan
| |
Collapse
|
32
|
Iwai S, Motono N, Oyama T, Shioya A, Yamada S, Uramoto H. The Clinical Relevance of the Expression of SGLT2 in Lung Adenocarcinoma. Oncology 2024; 102:710-719. [PMID: 38232717 DOI: 10.1159/000536060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 12/29/2023] [Indexed: 01/19/2024]
Abstract
PURPOSE We aimed to elucidate the functions and clinical relevance of sodium-glucose cotransporter 2 (SGLT2) in resected lung adenocarcinoma. METHODS The protein expression of SGLT2 in tumor samples from 199 patients with lung adenocarcinoma was analyzed by immunohistochemistry, and the protein expression, clinical variables, and survival outcomes were compared. RESULTS The median SGLT2 expression was significantly higher in advanced-stage and more aggressive adenocarcinomas. Age ≥70 (p < 0.01), BI ≥600 (p < 0.01), PRDX4 <25 (p < 0.01), and SGLT2 ≥12% (p = 0.03) were significant factors for RFS in multivariate analysis. Significant differences were observed in the RFS rates of the groups divided using the cutoff value of SGLT2 ≥12% (5-year RFS: 72.6% vs. 90%) (p < 0.01). CONCLUSION The expression of SGLT2 was more frequently detected in advanced-stage and more aggressive adenocarcinomas with aggressive biological behavior than in their counterparts. The survival analysis revealed that the strong expression of SGLT2 was associated with poorer RFS. The SGLT2 expression predicts postoperative recurrence in lung adenocarcinoma patients.
Collapse
Affiliation(s)
- Shun Iwai
- Department of Thoracic Surgery, Kanazawa Medical University, Uchinada-machi, Japan
| | - Nozomu Motono
- Department of Thoracic Surgery, Kanazawa Medical University, Uchinada-machi, Japan
| | - Tsunehiro Oyama
- Department of Thoracic Surgery, Kanazawa Medical University, Uchinada-machi, Japan
- Imamitsu Home Care Clinic, Kitakyushu, Japan
| | - Akihiro Shioya
- Department of Pathology and Laboratory Medicine, Kanazawa Medical University, Uchinada-machi, Japan
| | - Sohsuke Yamada
- Department of Pathology and Laboratory Medicine, Kanazawa Medical University, Uchinada-machi, Japan
| | - Hidetaka Uramoto
- Department of Thoracic Surgery, Kanazawa Medical University, Uchinada-machi, Japan
| |
Collapse
|
33
|
Shahid Malik M, Ou YY. Integrating Pre-Trained protein language model and multiple window scanning deep learning networks for accurate identification of secondary active transporters in membrane proteins. Methods 2023; 220:11-20. [PMID: 37871661 DOI: 10.1016/j.ymeth.2023.10.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/04/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023] Open
Abstract
Secondary active transporters play pivotal roles in regulating ion and molecule transport across cell membranes, with implications in diseases like cancer. However, studying transporters via biochemical experiments poses challenges. We propose an effective computational approach to identify secondary active transporters from membrane protein sequences using pre-trained language models and deep learning neural networks. Our dataset comprised 290 secondary active transporters and 5,420 other membrane proteins from UniProt. Three types of features were extracted - one-hot encodings, position-specific scoring matrix profiles, and contextual embeddings from the ProtTrans language model. A multi-window convolutional neural network architecture scanned the ProtTrans embeddings using varying window sizes to capture multi-scale sequence patterns. The proposed model combining ProtTrans embeddings and multi-window convolutional neural networks achieved 86% sensitivity, 99% specificity and 98% overall accuracy in identifying secondary active transporters, outperforming conventional machine learning approaches. This work demonstrates the promise of integrating pre-trained language models like ProtTrans with multi-scale deep neural networks to effectively interpret transporter sequences for functional analysis. Our approach enables more accurate computational identification of secondary active transporters, advancing membrane protein research.
Collapse
Affiliation(s)
- Muhammad Shahid Malik
- Department of Computer Science and Engineering, Yuan Ze University, Chung-Li 32003, Taiwan; Department of Computer Sciences, Karakoram International University, Gilgit-Baltistan, Pakistan
| | - Yu-Yen Ou
- Department of Computer Science and Engineering, Yuan Ze University, Chung-Li 32003, Taiwan; Graduate Program in Biomedical Informatics, Yuan Ze University, Chung-Li 32003, Taiwan.
| |
Collapse
|
34
|
Fos-Codoner FS, Bouwman LMS, Keijer J, van Schothorst EM. Dietary Galactose Increases the Expression of Mitochondrial OXPHOS Genes and Modulates the Carbohydrate Oxidation Pathways in Mouse Intestinal Mucosa. J Nutr 2023; 153:3448-3457. [PMID: 37858726 DOI: 10.1016/j.tjnut.2023.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Prolonged lactation provides substantial health benefits, possibly because of galactose as part of milk sugar lactose. Isocaloric replacement of dietary glucose [16 energy%(en%)] with galactose within a normal diet (64en% carbohydrates) during a 3-wk postweaning period provided substantial benefits on short- and long-term physiologic and metabolic parameters at the whole-body level and liver in female mice, which might be attributable to intestinal function. OBJECTIVES This study aimed to investigate if partial dietary replacement of glucose with galactose alters intestinal metabolism underlying hepatic health effects. METHODS Proximal intestinal mucosa gene profiles in female mice were analyzed using RNAseq technology, validated, and correlated with hepatic health parameters. RESULTS Transcriptome analysis revealed that the presence of galactose primarily affected the pathways involved in energy metabolism. A consistently higher expression was observed in the subset of mitochondrial transcripts (78 of 80, all P.adj < 0.1). Oxidative phosphorylation (OXPHOS) represented the most upregulated process (all top 10 pathways) independent of the total mitochondrial mass (P = 0.75). Moreover, galactose consistently upregulated carbohydrate metabolism pathways, specifically glycolysis till acetyl-CoA production and fructose metabolism. Also, the expression of transcripts involved in these pathways was negatively correlated with circulating serum amyloid A3 protein, a marker of hepatic inflammation [R (-0.61, -0.5), P (0.002, 0.01)]. Accordingly, CD163+ cells were decreased in the liver. Additionally, the expression of key fructolytic enzymes in the small intestinal mucosa was negatively correlated with triglyceride accumulation in the liver [R (-0.45, -0.4), P (0.03, 0.05)]. CONCLUSIONS To our knowledge, our results show for the first time the role of galactose as an OXPHOS activator in vivo. Moreover, the concept of intestinal cells acting as the body's metabolic gatekeeper is strongly supported, as they alter substrate availability and thereby contribute to the maintenance of metabolic homeostasis, protecting other organs, as evidenced by their potential ability to shield the liver from the potentially detrimental effects of fructose.
Collapse
Affiliation(s)
| | - Lianne M S Bouwman
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | | |
Collapse
|
35
|
Piccirillo S, Preziuso A, Cerqueni G, Serfilippi T, Terenzi V, Vinciguerra A, Amoroso S, Lariccia V, Magi S. A strategic tool to improve the study of molecular determinants of Alzheimer's disease: The role of glyceraldehyde. Biochem Pharmacol 2023; 218:115869. [PMID: 37871878 DOI: 10.1016/j.bcp.2023.115869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/14/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia and is characterized by progressive neurodegeneration leading to severe cognitive, memory, and behavioral impairments. The onset of AD involves a complex interplay among various factors, including age, genetics, chronic inflammation, and impaired energy metabolism. Despite significant efforts, there are currently no effective therapies capable of modifying the course of AD, likely owing to an excessive focus on the amyloid hypothesis and a limited consideration of other intracellular pathways. In the present review, we emphasize the emerging concept of AD as a metabolic disease, where alterations in energy metabolism play a critical role in its development and progression. Notably, glucose metabolism impairment is associated with mitochondrial dysfunction, oxidative stress, Ca2+ dyshomeostasis, and protein misfolding, forming interconnected processes that perpetuate a detrimental self-feeding loop sustaining AD progression. Advanced glycation end products (AGEs), neurotoxic compounds that accumulate in AD, are considered an important consequence of glucose metabolism disruption, and glyceraldehyde (GA), a glycolytic intermediate, is a key contributor to AGEs formation in both neurons and astrocytes. Exploring the impact of GA-induced glucose metabolism impairment opens up exciting possibilities for creating an easy-to-handle in vitro model that recapitulates the early stage of the disease. This model holds great potential for advancing the development of novel therapeutics targeting various intracellular pathways implicated in AD pathogenesis. In conclusion, looking beyond the conventional amyloid hypothesis could lead researchers to discover promising targets for intervention, offering the possibility of addressing the existing medical gaps in AD treatment.
Collapse
Affiliation(s)
- Silvia Piccirillo
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy.
| | - Alessandra Preziuso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy.
| | - Giorgia Cerqueni
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy.
| | - Tiziano Serfilippi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy.
| | - Valentina Terenzi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy.
| | - Antonio Vinciguerra
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy.
| | - Salvatore Amoroso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy.
| | - Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy.
| | - Simona Magi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy.
| |
Collapse
|
36
|
Subramaniam M, Loewen ME. Review: A species comparison of the kinetic homogeneous and heterogeneous organization of sodium-dependent glucose transport systems along the intestine. Comp Biochem Physiol A Mol Integr Physiol 2023; 285:111492. [PMID: 37536429 DOI: 10.1016/j.cbpa.2023.111492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/21/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
The targeted use of carbohydrates by feed and food industries to create balanced and cost-effective diets has generated a tremendous amount of research in carbohydrate digestion and absorption in different species. Specifically, this research has led us to a larger observation that identified different organizations of intestinal sodium-dependent glucose absorption across species, which has not been previously collated and reviewed. Thus, this review will compare the kinetic segregation of sodium-dependent glucose transport across the intestine of different species, which we have termed either homogeneous or heterogeneous systems. For instance, the pig follows a heterogeneous system of sodium-dependent glucose transport with a high-affinity, super-low-capacity (Ha/sLc) in the jejunum, and a high-affinity, super-high-capacity (Ha/sHc) in the ileum. This is achieved by multiple sodium-dependent glucose transporters contributing to each segment. In contrast, tilapia have a homogenous system characterized by high-affinity, high-capacity (Ha/Hc) throughout the intestine. Additionally, we are the first to report glucose transporter patterns across species presented from vertebrates to invertebrates. Finally, other kinetic transport systems are briefly covered to illustrate possible contributions/modulations to sodium-dependent glucose transporter organization. Overall, we present a new perspective on the organization of glucose absorption along the intestinal tract.
Collapse
Affiliation(s)
- Marina Subramaniam
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan S7N 5B4, Canada
| | - Matthew E Loewen
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan S7N 5B4, Canada.
| |
Collapse
|
37
|
Nicola MS, Kalb AL, Barbosa AA, Velasquez BES, Rincon JAA, Feijó JO, Dellagostin EN, Martins AWS, Blödorn EB, Domingues WB, Lopes F, Quinteiro-Filho WM, Mondadori RG, Campos VF, Rabassa VR, Komninou ER, Delpino FAB, Corrêa MN. Butyrate supplementation in the liquid diet of dairy calves leads to a rapid recovery from diarrhea and reduces its occurrence and relapses in the preweaning period. J Dairy Sci 2023; 106:7908-7923. [PMID: 37164858 DOI: 10.3168/jds.2022-22670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 04/11/2023] [Indexed: 05/12/2023]
Abstract
The present study aimed to evaluate the effect of continuous butyrate administration in dairy calves' liquid diet considering diarrhea, metabolic profile, gastrointestinal development, and corporal growth. Immediately after birth, calves were randomly allocated into 2 groups of 62 calves (50 females and 12 males), with access to water and a solid feed ad libitum. The butyrate group (BG) received 4 g/d of sodium butyrate (Admix Easy, Adisseo) diluted in the whole milk, and the control group (CG) received whole milk with no supplementation. Sodium butyrate was administered from d 1 of life until the weaning at 90 d. Feces consistency was assessed daily for the first 30 d of life and characterized by scores from 0 to 4 (0 and 1 for normal, and 2, 3, and 4 for abnormal feces). Diarrhea was diagnosed when the animals had abnormal feces and fever. Morbidity, recurrence, mortality, and lethality data were recorded and compared between the groups. Average daily gain (ADG) and corporal growth (body weight, thoracic perimeter, height at the withers, and croup width) were evaluated weekly, from the first day to d 30, and later at 45, 60, and 90 d of life. Blood samples were taken weekly for up to 30 d to determine the circulating levels of total calcium, phosphorus, chloride, bicarbonate, glucose, β-hydroxybutyrate, and nonesterified fatty acids. The males were euthanized at 15 (n = 6 per group) and 30 d (n = 6 per group) for morphometric, histological, and gene expression analysis of the gastrointestinal tract. The results showed that the BG had a lower rate of morbidity (BG = 30% vs. CG = 50%) and recurrence (BG = 26.7% vs. CG = 60%) of diarrhea than the CG. In addition, the BG had abnormal feces for a shorter period (BG = 4.64 ± 0.47 d vs. CG = 8.6 ± 0.65 d). The ADG tended to be higher in BG than CG up to 30 and 60 d. Metabolic evaluations showed the lowest levels of glucose and highest levels of nonesterified fatty acids in BG. On d 30 of life, rumen papillae length, papilla area, duodenum villus length, and crypt depth were higher in BG than in CG. The duodenal gene expression at 30 d showed that animals with diarrhea episodes that did not receive butyrate had the highest levels of transcripts for the LCT and GLP2 genes. In addition, in different ways, both butyrate and neonatal diarrhea affected the gene expression of IGF1, SLC5A1, and AQP3. These results allow us to conclude that continuous supplementation with sodium butyrate improves gastrointestinal development, reduces the occurrence of diarrhea, and makes clinical conditions milder with faster recovery, favoring a higher ADG in the first 30 and 60 d of life. Based on these results, we conclude that sodium butyrate can be indicated for liquid diet supplementation to accelerate gastrointestinal tract development and prevent severe cases of neonatal diarrhea, tending to improve average daily gain until weaning.
Collapse
Affiliation(s)
- M S Nicola
- NUPEEC (Núcleo de Pesquisa, Ensino e Extensão em Pecuária), Programa de Pós-Graduação em Veterinária (PPGV), Faculdade de Veterinária, Universidade Federal de Pelotas, 96010-610, Pelotas, RS, Brazil.
| | - A L Kalb
- NUPEEC (Núcleo de Pesquisa, Ensino e Extensão em Pecuária), Programa de Pós-Graduação em Veterinária (PPGV), Faculdade de Veterinária, Universidade Federal de Pelotas, 96010-610, Pelotas, RS, Brazil
| | - A A Barbosa
- NUPEEC (Núcleo de Pesquisa, Ensino e Extensão em Pecuária), Programa de Pós-Graduação em Veterinária (PPGV), Faculdade de Veterinária, Universidade Federal de Pelotas, 96010-610, Pelotas, RS, Brazil
| | - B E S Velasquez
- NUPEEC (Núcleo de Pesquisa, Ensino e Extensão em Pecuária), Programa de Pós-Graduação em Veterinária (PPGV), Faculdade de Veterinária, Universidade Federal de Pelotas, 96010-610, Pelotas, RS, Brazil
| | - J A A Rincon
- Facultad de Ciencias Agropecuarias, Universidad de La Salle, Yopal, Casanare 850008, Colombia
| | - J O Feijó
- NUPEEC (Núcleo de Pesquisa, Ensino e Extensão em Pecuária), Programa de Pós-Graduação em Veterinária (PPGV), Faculdade de Veterinária, Universidade Federal de Pelotas, 96010-610, Pelotas, RS, Brazil
| | - E N Dellagostin
- Laboratório de Genômica Estrutural, Programa de Pós-Graduação em Biotecnologia (PPGBio), Centro de Desenvolvimento Tecnológico (CDTec), Universidade Federal de Pelotas, 96010-610, Pelotas, RS, Brazil
| | - A W S Martins
- Laboratório de Genômica Estrutural, Programa de Pós-Graduação em Biotecnologia (PPGBio), Centro de Desenvolvimento Tecnológico (CDTec), Universidade Federal de Pelotas, 96010-610, Pelotas, RS, Brazil
| | - E B Blödorn
- Laboratório de Genômica Estrutural, Programa de Pós-Graduação em Biotecnologia (PPGBio), Centro de Desenvolvimento Tecnológico (CDTec), Universidade Federal de Pelotas, 96010-610, Pelotas, RS, Brazil
| | - W B Domingues
- Laboratório de Genômica Estrutural, Programa de Pós-Graduação em Biotecnologia (PPGBio), Centro de Desenvolvimento Tecnológico (CDTec), Universidade Federal de Pelotas, 96010-610, Pelotas, RS, Brazil
| | - F Lopes
- Adisseo Brasil Nutrição Animal, São Paulo, 05804-900, Brazil
| | | | - R G Mondadori
- NUPEEC (Núcleo de Pesquisa, Ensino e Extensão em Pecuária), Programa de Pós-Graduação em Veterinária (PPGV), Faculdade de Veterinária, Universidade Federal de Pelotas, 96010-610, Pelotas, RS, Brazil
| | - V F Campos
- Laboratório de Genômica Estrutural, Programa de Pós-Graduação em Biotecnologia (PPGBio), Centro de Desenvolvimento Tecnológico (CDTec), Universidade Federal de Pelotas, 96010-610, Pelotas, RS, Brazil
| | - V R Rabassa
- NUPEEC (Núcleo de Pesquisa, Ensino e Extensão em Pecuária), Programa de Pós-Graduação em Veterinária (PPGV), Faculdade de Veterinária, Universidade Federal de Pelotas, 96010-610, Pelotas, RS, Brazil
| | - E R Komninou
- NUPEEC (Núcleo de Pesquisa, Ensino e Extensão em Pecuária), Programa de Pós-Graduação em Veterinária (PPGV), Faculdade de Veterinária, Universidade Federal de Pelotas, 96010-610, Pelotas, RS, Brazil; Laboratório de Genômica Estrutural, Programa de Pós-Graduação em Biotecnologia (PPGBio), Centro de Desenvolvimento Tecnológico (CDTec), Universidade Federal de Pelotas, 96010-610, Pelotas, RS, Brazil
| | - F A B Delpino
- NUPEEC (Núcleo de Pesquisa, Ensino e Extensão em Pecuária), Programa de Pós-Graduação em Veterinária (PPGV), Faculdade de Veterinária, Universidade Federal de Pelotas, 96010-610, Pelotas, RS, Brazil
| | - M N Corrêa
- NUPEEC (Núcleo de Pesquisa, Ensino e Extensão em Pecuária), Programa de Pós-Graduação em Veterinária (PPGV), Faculdade de Veterinária, Universidade Federal de Pelotas, 96010-610, Pelotas, RS, Brazil
| |
Collapse
|
38
|
Kusunoki M, Hisano F, Matsuda SI, Kusunoki A, Wakazono N, Tsutsumi K, Miyata T. Effects of SGLT2 inhibitors on the intestinal bacterial flora in Japanese patients with type 2 diabetes mellitus. Drug Res (Stuttg) 2023; 73:412-416. [PMID: 37236230 DOI: 10.1055/a-2037-5250] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Selective inhibitors of sodium glucose co-transporter-2 (SGLT2) suppress renal glucose reabsorption and promote urinary glucose excretion, thereby lowering blood glucose. SGLT2 inhibitors have been reported to reduce body weight. However, the mechanism underlying the reduction in the body weight induced by SGLT2 inhibitor treatment remains to be elucidated. In this study, we investigated the effects of SGLT2 inhibitors on the intestinal bacterial flora. A total of 36 Japanese patients with type 2 diabetes mellitus received a SGLT2 inhibitor (luseogliflozin or dapagliflozin) for 3 months, and the prevalences of balance-regulating bacteria and balance-disturbing bacteria in the feces of the patients before and after SGLT2 inhibitor treatment were determined. SGLT2 inhibitor treatment was associated with a significant increase of the overall prevalence of the 12 types of balance-regulating bacteria. In addition, significant increases in the prevalences of the short-chain fatty acid (SCFAs)-producing bacteria among the balance-regulating bacteria were also observed. Individual analyses of the balance-regulating bacteria revealed that the SGLT2 inhibitor treatment was associated with a significant increase in the prevalence of Ruminococci, which are balance-regulating bacteria classified as SCFAs-producing bacteria. However, SGLT2 inhibitor had no effect on the balance-disturbing bacteria. These results suggested that SGLT2 inhibitor treatment was associated with an overall increase in the prevalence of balance-regulating bacteria. Among the balance-regulating bacteria, the prevalences of SCFAs-producing bacteria increased. SCFAs have been reported to prevent obesity. The results of the present study suggest that SGLT2 inhibitors might induce body weight reduction via their actions on the intestinal bacterial flora.
Collapse
Affiliation(s)
- Masataka Kusunoki
- Department of Diabetes, Motor Function and Metabolism, Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya City, Aichi, Japan
| | - Fumiya Hisano
- Graduate School of Medicine, Department of Integrated Health Sciences, Nagoya University, Nagoya City, Aichi, Japan
| | - Shin-Ichi Matsuda
- Department of Data Science, Faculty of Science and Technology, Nanzan University, Nagoya City, Aichi, Japan
| | | | - Naomi Wakazono
- Department of Diabetes, Motor Function and Metabolism, Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya City, Aichi, Japan
| | | | - Tetsuro Miyata
- Office of Medical Education, School of Medicine, International University of Health and Welfare, Narita City, Chiba, Japan
| |
Collapse
|
39
|
Zhao M, Li N, Zhou H. SGLT1: A Potential Drug Target for Cardiovascular Disease. Drug Des Devel Ther 2023; 17:2011-2023. [PMID: 37435096 PMCID: PMC10332373 DOI: 10.2147/dddt.s418321] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 06/29/2023] [Indexed: 07/13/2023] Open
Abstract
SGLT1 and SGLT2 are the two main members of the sodium-glucose cotransporters (SGLTs), which are mainly responsible for glucose reabsorption in the body. In recent years, many large clinical trials have shown that SGLT2 inhibitors have cardiovascular protection for diabetic and non-diabetic patients independent of lowering blood glucose. However, SGLT2 was barely detected in the hearts of humans and animals, while SGLT1 was highly expressed in myocardium. As SGLT2 inhibitors also have a moderate inhibitory effect on SGLT1, the cardiovascular protection of SGLT2 inhibitors may be due to SGLT1 inhibition. SGLT1 expression is associated with pathological processes such as cardiac oxidative stress, inflammation, fibrosis, and cell apoptosis, as well as mitochondrial dysfunction. The purpose of this review is to summarize the protective effects of SGLT1 inhibition on hearts in various cell types, including cardiomyocytes, endothelial cells, and fibroblasts in preclinical studies, and to highlight the underlying molecular mechanisms of protection against cardiovascular diseases. Selective SGLT1 inhibitors could be considered a class of drugs for cardiac-specific therapy in the future.
Collapse
Affiliation(s)
- Mengnan Zhao
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People’s Republic of China
| | - Na Li
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People’s Republic of China
| | - Hong Zhou
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People’s Republic of China
| |
Collapse
|
40
|
Şahin E, Orhan C, Erten F, Şahin F, Şahin N, Şahin K. The effect of different boron compounds on nutrient digestibility, intestinal nutrient transporters, and liver lipid metabolism. Turk J Med Sci 2023; 53:619-629. [PMID: 37476906 PMCID: PMC10387887 DOI: 10.55730/1300-0144.5624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 04/15/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Gastrointestinal health is essential for maintaining a healthy lifestyle. Improving nutrient absorption and energy metabolism are the critical targets for intestinal health. This study aimed to determine the effects of different boron (B) derivatives on nutrient digestibility, intestinal nutrient transporters, and lipid metabolism in rats. METHODS Twenty-one rats were allocated to three groups (n = 7) as follows: (i) Control, (ii) Sodium pentaborate pentahydrate (SPP), and (iii) boric acid (BA). The rats were fed a chow diet (AIN-93M) and supplemented with 8 mg/kg elemental B from SPP (45.2 mg/kg BW) and BA (42.7 mg/kg BW) via oral gavage every other day for 12 weeks. The nutrient digestibility of rats in each group was measured using the indigestible indicator (chromium oxide, Cr2 O3, 0.20%). At the end of the experiment, animals were decapitated by cervical dislocation and jejunum, and liver samples were taken from each animal. The nutrient transporters and lipid-regulated transcription factors were determined by RT-PCR. RESULTS The nutrient digestibility (except for ash) was increased by SPP and BA supplementation (p < 0.05). SPP and BA-supplemented rats had higher jejunal glucose transporter 1 (GLUT1), GLUT2, GLUT5, sodium-dependent glucose transporter 1 (SGLT1), fatty acid transport protein-1 (FATP1), and FATP4 mRNA expression levels compared to nonsupplemented rats (p < 0.0001). BA-supplemented rats had remarkably higher peroxisome proliferator-activated receptor gamma (PPARγ) levels than nonsupplemented rats (p < 0.0001). In contrast, sterol regulatory element-binding protein 1c (SREBP-1c), liver X receptor alpha (LxR-α), and fatty acid synthase (FAS) levels decreased by SPP supplementation compared to other groups (p < 0.05). DISCUSSION SPP and BA administration enhanced nutrient digestibility, intestinal nutrient transporters, and liver lipid metabolism in rats.
Collapse
Affiliation(s)
- Emre Şahin
- Department of Animal Nutrition, Faculty of Veterinary Medicine, Bingöl University, Bingöl, Turkey
| | - Cemal Orhan
- Department of Animal Nutrition, Faculty of Veterinary Medicine, Fırat University, Elazığ, Turkey
| | - Füsun Erten
- Department of Veterinary Science, Pertek Sakine Genç Vocational School, Munzur University, Tunceli, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, İstanbul, Turkey
| | - Nurhan Şahin
- Department of Animal Nutrition, Faculty of Veterinary Medicine, Fırat University, Elazığ, Turkey
| | - Kazim Şahin
- Department of Animal Nutrition, Faculty of Veterinary Medicine, Fırat University, Elazığ, Turkey
| |
Collapse
|
41
|
Carbó R, Rodríguez E. Relevance of Sugar Transport across the Cell Membrane. Int J Mol Sci 2023; 24:ijms24076085. [PMID: 37047055 PMCID: PMC10094530 DOI: 10.3390/ijms24076085] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Sugar transport through the plasma membrane is one of the most critical events in the cellular transport of nutrients; for example, glucose has a central role in cellular metabolism and homeostasis. The way sugars enter the cell involves complex systems. Diverse protein systems participate in the membrane traffic of the sugars from the extracellular side to the cytoplasmic side. This diversity makes the phenomenon highly regulated and modulated to satisfy the different needs of each cell line. The beautiful thing about this process is how evolutionary processes have diversified a single function: to move glucose into the cell. The deregulation of these entrance systems causes some diseases. Hence, it is necessary to study them and search for a way to correct the alterations and utilize these mechanisms to promote health. This review will highlight the various mechanisms for importing the valuable sugars needed to create cellular homeostasis and survival in all kinds of cells.
Collapse
Affiliation(s)
- Roxana Carbó
- Cardiovascular Biomedicine Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano #1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
- Correspondence: ; Tel.: +52-55557-32911 (ext. 25704)
| | - Emma Rodríguez
- Cardiology Laboratory at Translational Research Unit UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano #1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico;
| |
Collapse
|
42
|
Clostridium butyricum Prevents Dysbiosis and the Rise in Blood Pressure in Spontaneously Hypertensive Rats. Int J Mol Sci 2023; 24:ijms24054955. [PMID: 36902386 PMCID: PMC10002514 DOI: 10.3390/ijms24054955] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
Hypertension is accompanied by dysbiosis and a decrease in the relative abundance of short-chain fatty acid (SCFA)-producing bacteria. However, there is no report to examine the role of C. butyricum in blood pressure regulation. We hypothesized that a decrease in the relative abundance of SCFA-producing bacteria in the gut was the cause of spontaneously hypertensive rats (SHR)-induced hypertension. C. butyricum and captopril were used to treat adult SHR for six weeks. C. butyricum modulated SHR-induced dysbiosis and significantly reduced systolic blood pressure (SBP) in SHR (p < 0.01). A 16S rRNA analysis determined changes in the relative abundance of the mainly SCFA-producing bacteria Akkermansia muciniphila, Lactobacillus amylovorus, and Agthobacter rectalis, which increased significantly. Total SCFAs, and particularly butyrate concentrations, in the SHR cecum and plasma were reduced (p < 0.05), while C. butyricum prevented this effect. Likewise, we supplemented SHR with butyrate for six weeks. We analyzed the flora composition, cecum SCFA concentration, and inflammatory response. The results showed that butyrate prevented SHR-induced hypertension and inflammation, and the decline of cecum SCFA concentrations (p < 0.05). This research revealed that increasing cecum butyrate concentrations by probiotics, or direct butyrate supplementation, prevented the adverse effects of SHR on intestinal flora, vascular, and blood pressure.
Collapse
|
43
|
Neutral effect of SGLT2 inhibitors on lipoprotein metabolism: From clinical evidence to molecular mechanisms. Pharmacol Res 2023; 188:106667. [PMID: 36657502 DOI: 10.1016/j.phrs.2023.106667] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/19/2022] [Accepted: 01/15/2023] [Indexed: 01/18/2023]
Abstract
Sodium-glucose cotransporter-2 inhibitors (SGLT2i) are effective, well-tolerated, and safe glucose-lowering compounds for patients with type 2 diabetes mellitus (T2DM). SGLT2i benefit encompasses protection from heart and kidney failure, independently of the presence of diabetes. In addition, SGLT2i consistently reduce the risk of hospitalization for heart failure and, although with some heterogeneity between specific members of the class, favourably affect the risk of cardiovascular outcomes. The molecular mechanisms underlying the cardiovascular favourable effect are not fully clarified. Studies testing the efficacy of SGLT2i in human cohorts and experimental models of atherosclerotic cardiovascular disease (ASCVD) have reported significant differences in circulating levels and composition of lipoprotein classes. In randomized clinical trials, small but significant increases in low-density lipoprotein cholesterol (LDL-C) levels have been observed, with a still undefined clinical significance; on the other hand, favourable (although modest) effects on high-density lipoprotein cholesterol (HDL-C) and triglycerides have been reported. At the molecular level, glycosuria may promote a starving-like state that ultimately leads to a metabolic improvement through the mobilization of fatty acids from the adipose tissue and their oxidation for the production of ketone bodies. This, however, may also fuel hepatic cholesterol synthesis, thus inhibiting atherogenic lipoprotein uptake from the liver. Long-term studies collecting detailed information on lipid-lowering therapies at baseline and during the trials with SGLT2i, as well as regularly monitoring lipid profiles are warranted to disentangle the potential implications of SGLT2i in modulating lipoprotein-mediated atherosclerotic cardiovascular risk.
Collapse
|
44
|
England A, Gharib-Naseri K, Kheravii SK, Wu SB. Influence of sex and rearing method on performance and flock uniformity in broilers-implications for research settings. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2022; 12:276-283. [PMID: 36712408 PMCID: PMC9869427 DOI: 10.1016/j.aninu.2022.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 11/02/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
Male and female broiler chickens differ in their growth performance, carcass part weights and nutrient requirements. The potential reasons for these differences have been explored by looking at differences in nutrient digestibility, nutrient transporter gene expression as well as gut microbiota populations between male and female birds. Studies have shown that male broilers have higher crude protein requirements compared to female broilers. The expression of monosaccharide and amino acid transporters show conflicting results as expression depends on the interactions between sex and bird age and breed as well as which tissue is sampled. Differences in microbiota populations between the genders were reported which may contribute towards performance differences, however research in this area is limited. The differences observed between the sexes contribute to increased variation in nutrition trials, and the potential to rear birds as equally mixed-sex becomes an option to reduce the variation introduced by the sex effect. Difference in rearing options obviously would only be feasible provided a quick, practical and cost-effective method of sexing birds is available, a topic that is also discussed in this review.
Collapse
|
45
|
SGLT-2 Inhibitors in Cancer Treatment-Mechanisms of Action and Emerging New Perspectives. Cancers (Basel) 2022; 14:cancers14235811. [PMID: 36497303 PMCID: PMC9738342 DOI: 10.3390/cancers14235811] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/11/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
A new group of antidiabetic drugs, sodium-glucose cotransporter 2 inhibitors (SGLT-2 inhibitors), have recently been shown to have anticancer effects and their expression has been confirmed in many cancer cell lines. Given the metabolic reprogramming of these cells in a glucose-based model, the ability of SGLT-2 inhibitors to block the glucose uptake by cancer cells appears to be an attractive therapeutic approach. In addition to tumour cells, SGLT-2s are only found in the proximal tubules in the kidneys. Furthermore, as numerous clinical trials have shown, the use of SGLT-2 inhibitors is well-tolerated and safe in patients with diabetes and/or heart failure. In vitro cell culture studies and preclinical in vivo studies have confirmed that SGLT-2 inhibitors exhibit antiproliferative effects on certain types of cancer. However, the mechanisms of this action remain unclear. Even in those tumour cell types in which SGLT-2 is present, there is sometimes an SGLT-2-independent mechanism of anticancer action of this group of drugs. This article presents the current state of knowledge of the potential mechanisms of the anticancer action of SGLT-2 inhibitors and their possible future application in clinical oncology.
Collapse
|
46
|
Brosch PK, Korsa T, Taban D, Eiring P, Kreisz P, Hildebrand S, Neubauer J, Zimmermann H, Sauer M, Shirakashi R, Djuzenova CS, Sisario D, Sukhorukov VL. Glucose and Inositol Transporters, SLC5A1 and SLC5A3, in Glioblastoma Cell Migration. Cancers (Basel) 2022; 14:5794. [PMID: 36497276 PMCID: PMC9738886 DOI: 10.3390/cancers14235794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/04/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022] Open
Abstract
(1) Background: The recurrence of glioblastoma multiforme (GBM) is mainly due to invasion of the surrounding brain tissue, where organic solutes, including glucose and inositol, are abundant. Invasive cell migration has been linked to the aberrant expression of transmembrane solute-linked carriers (SLC). Here, we explore the role of glucose (SLC5A1) and inositol transporters (SLC5A3) in GBM cell migration. (2) Methods: Using immunofluorescence microscopy, we visualized the subcellular localization of SLC5A1 and SLC5A3 in two highly motile human GBM cell lines. We also employed wound-healing assays to examine the effect of SLC inhibition on GBM cell migration and examined the chemotactic potential of inositol. (3) Results: While GBM cell migration was significantly increased by extracellular inositol and glucose, it was strongly impaired by SLC transporter inhibition. In the GBM cell monolayers, both SLCs were exclusively detected in the migrating cells at the monolayer edge. In single GBM cells, both transporters were primarily localized at the leading edge of the lamellipodium. Interestingly, in GBM cells migrating via blebbing, SLC5A1 and SLC5A3 were predominantly detected in nascent and mature blebs, respectively. (4) Conclusion: We provide several lines of evidence for the involvement of SLC5A1 and SLC5A3 in GBM cell migration, thereby complementing the migration-associated transportome. Our findings suggest that SLC inhibition is a promising approach to GBM treatment.
Collapse
Affiliation(s)
- Philippa K. Brosch
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Tessa Korsa
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany; (J.N.); (H.Z.)
| | - Danush Taban
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Patrick Eiring
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Philipp Kreisz
- Julius-von-Sachs Institute, University of Würzburg, 97082 Würzburg, Germany;
| | - Sascha Hildebrand
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Julia Neubauer
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany; (J.N.); (H.Z.)
| | - Heiko Zimmermann
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany; (J.N.); (H.Z.)
- Department of Molecular and Cellular Biotechnology, Saarland University, 66123 Saarbrücken, Germany
- Faculty of Marine Science, Universidad Católica del Norte, Coquimbo 1281, Chile
| | - Markus Sauer
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Ryo Shirakashi
- Institute of Industrial Science, The University of Tokyo, Tokyo 153-8505, Japan;
| | - Cholpon S. Djuzenova
- Department of Radiation Oncology, University Hospital of Würzburg, 97080 Würzburg, Germany;
| | - Dmitri Sisario
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Vladimir L. Sukhorukov
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| |
Collapse
|
47
|
Zhang Q, Fang Y, She C, Zheng R, Hong C, Chen C, Wu J. Diagnostic and prognostic significance of SLC50A1 expression in patients with primary early breast cancer. Exp Ther Med 2022; 24:616. [PMID: 36160901 PMCID: PMC9468843 DOI: 10.3892/etm.2022.11553] [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: 04/26/2022] [Accepted: 07/22/2022] [Indexed: 02/05/2023] Open
Abstract
There is a lack of validated biomarkers for the diagnosis of early breast cancer (EBC). The current study aimed to determine the diagnostic and prognostic value of solute carrier family 50 member 1 (SLC50A1) in patients with EBC. Therefore, 123 patients with EBC, 30 patients with benign breast disease (BBD) and 26 healthy controls (HCs) were recruited. The serum levels of SLC50A1 in paired sera of 40 postoperative patients were assessed by ELISA. Immunohistochemical staining for SLC50A1 was performed in surgical tissue derived from 83 patients with EBC and 30 patients with BBD. mRNA expression of SLC50A1 and its diagnostic and prognostic value in patients with EBC was evaluated using an RNA-sequencing database. The results showed that serum levels of SLC50A1 in patients with EBC were significantly higher compared with those in patients with BBD and HCs (both P<0.001). Additionally, receiver operating characteristic curve analysis revealed that the serum levels of SLC50A1 distinguished patients with EBC from patients with BBD and HCs with a sensitivity of 76.42% and specificity of 76.79% [area under the curve (AUC)=0.783; P<0.001]. The diagnostic value of SLC50A1 was significantly greater than that of carcinoembryonic (P<0.005) and carbohydrate antigen 15-3 (P<0.029). Furthermore, the number of SLC50A1 positive cells significantly increased in tissue of patients with EBC compared with patients with BBD (P<0.001). A positive association between serum levels of SLC50A1 and its expression in tissue samples was observed in patients with EBC (ρ=0.700; P<0.001). Additionally, bioinformatics analysis verified the diagnostic value of SLC50A1, with an AUC of 0.983 (P<0.001). Multivariate analysis demonstrated that SLC50A1 was an independent prognostic factor in patients with EBC with a hazard ratio of 1.917 (P=0.013). These findings indicated that SLC50A1 may be a potential diagnostic biomarker for primary EBC and that SLC50A1 upregulation may be associated with unfavorable prognosis in patients with EBC.
Collapse
Affiliation(s)
- Qunchen Zhang
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Yutong Fang
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Chuanghong She
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Rongji Zheng
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Chaoqun Hong
- Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Chunfa Chen
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Correspondence to: Dr Chunfa Chen or Dr Jundong Wu, The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Rao Ping Road, Shantou, Guangdong 515041, P.R. China
| | - Jundong Wu
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Correspondence to: Dr Chunfa Chen or Dr Jundong Wu, The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Rao Ping Road, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
48
|
Gyimesi G, Hediger MA. Systematic in silico discovery of novel solute carrier-like proteins from proteomes. PLoS One 2022; 17:e0271062. [PMID: 35901096 PMCID: PMC9333335 DOI: 10.1371/journal.pone.0271062] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 06/22/2022] [Indexed: 12/26/2022] Open
Abstract
Solute carrier (SLC) proteins represent the largest superfamily of transmembrane transporters. While many of them play key biological roles, their systematic analysis has been hampered by their functional and structural heterogeneity. Based on available nomenclature systems, we hypothesized that many as yet unidentified SLC transporters exist in the human genome, which await further systematic analysis. Here, we present criteria for defining "SLC-likeness" to curate a set of "SLC-like" protein families from the Transporter Classification Database (TCDB) and Protein families (Pfam) databases. Computational sequence similarity searches surprisingly identified ~120 more proteins in human with potential SLC-like properties compared to previous annotations. Interestingly, several of these have documented transport activity in the scientific literature. To complete the overview of the "SLC-ome", we present an algorithm to classify SLC-like proteins into protein families, investigating their known functions and evolutionary relationships to similar proteins from 6 other clinically relevant experimental organisms, and pinpoint structural orphans. We envision that our work will serve as a stepping stone for future studies of the biological function and the identification of the natural substrates of the many under-explored SLC transporters, as well as for the development of new therapeutic applications, including strategies for personalized medicine and drug delivery.
Collapse
Affiliation(s)
- Gergely Gyimesi
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department for BioMedical Research, Inselspital, University of Bern, Bern, Switzerland
- * E-mail: (GG); (MAH)
| | - Matthias A. Hediger
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department for BioMedical Research, Inselspital, University of Bern, Bern, Switzerland
- * E-mail: (GG); (MAH)
| |
Collapse
|
49
|
Shah N, Perkovic V, Kotwal S. Impact of SGLT2 inhibitors on the kidney in people with type 2 diabetes and severely increased albuminuria. Expert Rev Clin Pharmacol 2022; 15:827-842. [PMID: 35912871 DOI: 10.1080/17512433.2022.2108402] [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: 11/04/2022]
Abstract
INTRODUCTION Diabetes is the most common cause of end stage kidney disease. Therapies such as sodium-glucose co-transporter-2 inhibitors have been identified over the last decade as effective oral hypoglycemic agents that also confer additional cardio and kidney protection. Knowledge of their mechanism of action and impact on patients with diabetes and albuminuria is vital in galvanizing prescriber confidence and increasing clinical uptake. AREAS COVERED This manuscript discusses the pathophysiology of diabetic kidney disease, patho-physiological mechanisms for sodium-glucose co-transporter-2 inhibitors, and their impact on patients with Type 2 diabetes mellitus and albuminuric kidney disease. EXPERT OPINION Sodium-glucose co-transporter-2 inhibitors reduce albuminuria with consequent benefits on cardiovascular and kidney outcomes in patients with diabetes and severe albuminuria. Whilst they have been incorporated into guidelines, the uptake of these agents into clinical practice has been slow. Increasing the uptake of these agents into clinical practice is necessary to improve outcomes for the large number of patients with diabetic kidney disease globally.
Collapse
Affiliation(s)
- Nasir Shah
- Faculty of Medicine, UNSW, Kensington, Sydney Australia 2052
| | - Vlado Perkovic
- Faculty of Medicine, UNSW, Kensington, Sydney Australia 2052.,The George Institute for Global Health, UNSW, 1 King Street, Newtown, Sydney, Australia 2042
| | - Sradha Kotwal
- The George Institute for Global Health, UNSW, 1 King Street, Newtown, Sydney, Australia 2042.,Prince of Wales Hospital, High Street, Sydney, Australia, 2031
| |
Collapse
|
50
|
Li Y, Liu B, Yin X, Jiang Z, Fang C, Chen N, Zhang B, Dai L, Yin Y. Targeted demethylation of the SLC5A7 promotor inhibits colorectal cancer progression. Clin Epigenetics 2022; 14:92. [PMID: 35858918 PMCID: PMC9301853 DOI: 10.1186/s13148-022-01308-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 07/04/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND SLC5A7 (solute carrier family 5 member 7), also known as choline transporter 1 (CHT1), is downregulated in colorectal cancer (CRC) and functions as a tumor suppressor. However, the mechanisms underlying the inactivation of SLC5A7 in CRC remain to be elucidated. RESULTS In the present study, two broad-spectrum demethylation agents (azacitidine and decitabine) employed to treat CRC cells significantly upregulated SLC5A7 expression. Further results based on the CRC cohort and TCGA database indicated that SLC5A7 promoter methylation inversely correlated with SLC5A7 expression, and the status of SLC5A7 promotor methylation showed a promising prognostic value for patients with CRC. Next, the dCas9-multiGCN4/scFv-TET1CD-based precision demethylation system was constructed, which could significantly and specifically promote SLC5A7 expression in CRC cells through sgRNA targeting the SLC5A7 promoter. Both in vitro and in vivo experiments demonstrated that targeted demethylation of SLC5A7 by dCas9-multiGCN4/scFv-TET1CD-sgSLC5A7 inhibited tumor growth by stabilizing p53 and regulating downstream targets. CONCLUSIONS Collectively, DNA promoter methylation caused inactivation of SLC5A7 in CRC, and targeted demethylation of SLC5A7 might be a therapeutic target for CRC and other cancers.
Collapse
Affiliation(s)
- Yang Li
- Department of Gastrointestinal Surgery, Guang'an People's Hospital, Guang'an, 638500, Sichuan, People's Republic of China
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Baike Liu
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiaonan Yin
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zhiyuan Jiang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Chao Fang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Na Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, People's Republic of China
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People's Republic of China
| | - Bo Zhang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Yuan Yin
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|