1
|
Xiang F, Zhang Z, Xie J, Xiong S, Yang C, Liao D, Xia B, Lin L. Comprehensive review of the expanding roles of the carnitine pool in metabolic physiology: beyond fatty acid oxidation. J Transl Med 2025; 23:324. [PMID: 40087749 PMCID: PMC11907856 DOI: 10.1186/s12967-025-06341-5] [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: 11/09/2024] [Accepted: 03/01/2025] [Indexed: 03/17/2025] Open
Abstract
Traditionally, the carnitine pool is closely related to fatty acid metabolism. However, with increasing research, the pleiotropic effects of the carnitine pool have gradually emerged. The purpose of this review is to comprehensively investigate of the emerging understanding of the pleiotropic role of the carnitine pool, carnitine/acylcarnitines are not only auxiliaries or metabolites of fatty acid oxidation, but also play more complex and diverse roles, including energy metabolism, mitochondrial homeostasis, epigenetic regulation, regulation of inflammation and the immune system, tumor biology, signal transduction, and neuroprotection. This review provides an overview of the complex network of carnitine synthesis, transport, shuttle, and regulation, carnitine/acylcarnitines have the potential to be used as communication molecules, biomarkers and therapeutic targets for multiple diseases, with profound effects on intercellular communication, metabolic interactions between organs and overall metabolic health. The purpose of this review is to comprehensively summarize the multidimensional biological effects of the carnitine pool beyond its traditional role in fatty acid oxidation and to summarize the systemic effects mediated by carnitine/acylcarnitine to provide new perspectives for pharmacological research and treatment innovation and new strategies for the prevention and treatment of a variety of diseases.
Collapse
Affiliation(s)
- Feng Xiang
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zhimin Zhang
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Jingchen Xie
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Suhui Xiong
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Chen Yang
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Duanfang Liao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Bohou Xia
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Limei Lin
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
2
|
An F, Jia X, Shi Y, Xiao X, Yang F, Su J, Peng X, Geng G, Yan C. The ultimate microbial composition for correcting Th17/Treg cell imbalance and lipid metabolism disorders in osteoporosis. Int Immunopharmacol 2025; 144:113613. [PMID: 39571271 DOI: 10.1016/j.intimp.2024.113613] [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: 09/10/2024] [Revised: 10/28/2024] [Accepted: 11/07/2024] [Indexed: 12/15/2024]
Abstract
Osteoporosis is a systemic bone disease characterised by decreased bone mass and a deteriorated bone microstructure, leading to increased bone fragility and fracture risk. Disorders of the intestinal microbiota may be key inducers of osteoporosis. Furthermore, such disorders may contribute to osteoporosis by influencing immune function and lipid metabolism. Therefore, in this review, we aimed to summarise the molecular mechanisms through which the intestinal microbiota affect the onset and development of osteoporosis by regulating Th17/Treg imbalance and lipid metabolism disorders. We also discussed the regulatory mechanisms underlying the effect of intestinal microbiota-related modulators on Th17/Treg imbalance and lipid metabolism disorders in osteoporosis, to explore new molecular targets for its treatment and provide a theoretical basis for clinical management.
Collapse
Affiliation(s)
- Fangyu An
- Teaching Experiment Training Center, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China.
| | - Xueru Jia
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Yangyang Shi
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Xiaolong Xiao
- School of Tradional Chinese and Werstern Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Fan Yang
- School of Tradional Chinese and Werstern Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Junchang Su
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Xia Peng
- School of Tradional Chinese and Werstern Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Guangqin Geng
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Chunlu Yan
- School of Tradional Chinese and Werstern Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China.
| |
Collapse
|
3
|
Mazza T, Scalise M, Console L, Galluccio M, Giangregorio N, Tonazzi A, Pochini L, Indiveri C. Carnitine traffic and human fertility. Biochem Pharmacol 2024; 230:116565. [PMID: 39368751 DOI: 10.1016/j.bcp.2024.116565] [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: 07/31/2024] [Revised: 09/18/2024] [Accepted: 10/01/2024] [Indexed: 10/07/2024]
Abstract
Carnitine is a vital molecule in human metabolism, prominently involved in fatty acid β-oxidation within mitochondria. Predominantly sourced from dietary intake, carnitine also derives from endogenous synthesis. This review delves into the complex network of carnitine transport and distribution, emphasizing its pivotal role in human fertility. Together with its role in fatty acid oxidation, carnitine modulates the acety-CoA/CoA ratio, influencing carbohydrate metabolism, lipid biosynthesis, and gene expression. The intricate regulation of carnitine homeostasis involves a network of membrane transporters, notably OCTN2, which is central in its absorption, reabsorption, and distribution. OCTN2 dysfunction, results in Primary Carnitine Deficiency (PCD), characterized by systemic carnitine depletion and severe clinical manifestations, including fertility issues. In the male reproductive system, carnitine is crucial for sperm maturation and motility. In the female reproductive system, carnitine supports mitochondrial function necessary for oocyte quality, folliculogenesis, and embryonic development. Indeed, deficiencies in carnitine or its transporters have been linked to asthenozoospermia, reduced sperm quality, and suboptimal fertility outcomes in couples. Moreover, the antioxidant properties of carnitine protect spermatozoa from oxidative stress and help in managing conditions like polycystic ovary syndrome (PCOS) and endometriosis, enhancing sperm viability and fertilization potential of oocytes. This review summarizes the key role of membrane transporters in guaranteeing carnitine homeostasis with a special focus on the implications in fertility and possible treatments of infertility and other related disorders.
Collapse
Affiliation(s)
- Tiziano Mazza
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, Arcavacata di Rende 87036, Italy
| | - Mariafrancesca Scalise
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, Arcavacata di Rende 87036, Italy
| | - Lara Console
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, Arcavacata di Rende 87036, Italy
| | - Michele Galluccio
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, Arcavacata di Rende 87036, Italy
| | - Nicola Giangregorio
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), via Amendola 122/O, Bari 70126, Italy
| | - Annamaria Tonazzi
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), via Amendola 122/O, Bari 70126, Italy
| | - Lorena Pochini
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, Arcavacata di Rende 87036, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), via Amendola 122/O, Bari 70126, Italy.
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, Arcavacata di Rende 87036, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), via Amendola 122/O, Bari 70126, Italy.
| |
Collapse
|
4
|
Dai X, Liang M, Dai Y, Ding S, Sun X, Xu L. Causality of genetically determined blood metabolites on irritable bowel syndrome: A Mendelian randomization study. PLoS One 2024; 19:e0298963. [PMID: 38568932 PMCID: PMC10990233 DOI: 10.1371/journal.pone.0298963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/01/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is one of the most common functional bowel disorders and dysmetabolism plays an important role in the pathogenesis of disease. Nevertheless, there remains a lack of information regarding the causal relationship between circulating metabolites and IBS. A two-sample Mendelian randomization (MR) analysis was conducted in order to evaluate the causal relationship between genetically proxied 486 blood metabolites and IBS. METHODS A two-sample MR analysis was implemented to assess the causality of blood metabolites on IBS. The study utilized a genome-wide association study (GWAS) to examine 486 metabolites as the exposure variable while employing a GWAS study with 486,601 individuals of European descent as the outcome variable. The inverse-variance weighted (IVW) method was used to estimate the causal relationship of metabolites on IBS, while several methods were performed to eliminate the pleiotropy and heterogeneity. Another GWAS data was used for replication and meta-analysis. In addition, reverse MR and linkage disequilibrium score regression (LDSC) were employed for additional assessment. Multivariable MR analysis was conducted in order to evaluate the direct impact of metabolites on IBS. RESULTS Three known and two unknown metabolites were identified as being associated with the development of IBS. Higher levels of butyryl carnitine (OR(95%CI):1.10(1.02-1.18),p = 0.009) and tetradecanedioate (OR(95%CI):1.13(1.04-1.23),p = 0.003)increased susceptibility of IBS and higher levels of stearate(18:0)(OR(95%CI):0.72(0.58-0.89),p = 0.003) decreased susceptibility of IBS. CONCLUSION The metabolites implicated in the pathogenesis of IBS possess potential as biomarkers and hold promise for elucidating the underlying biological mechanisms of this condition.
Collapse
Affiliation(s)
- Xinyi Dai
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Min Liang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Yanna Dai
- Department of Traditional Chinese Medicine, Wuxi Xinwu District Rehabilitation Hospital, Wuxi, China
| | - Shaohua Ding
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaohe Sun
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Luzhou Xu
- Nanjing University of Chinese Medicine, Nanjing, China
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| |
Collapse
|
5
|
Wang Z, Jiang D, Wang X, Jiang Y, Sun Q, Ling W, An X, Ji C, Li S, Qi Y, Kang B. Spermidine improves the antioxidant capacity and morphology of intestinal tissues and regulates intestinal microorganisms in Sichuan white geese. Front Microbiol 2024; 14:1292984. [PMID: 38293560 PMCID: PMC10824853 DOI: 10.3389/fmicb.2023.1292984] [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: 10/04/2023] [Accepted: 12/29/2023] [Indexed: 02/01/2024] Open
Abstract
Introduction Intestinal health is very important to the health of livestock and poultry, and is even a major determining factor in the performance of livestock and poultry production. Spermidine is a type of polyamine that is commonly found in a variety of foods, and can resist oxidative stress, promote cell proliferation and regulate intestinal flora. Methods In this study, we explored the effects of spermidine on intestinal health under physiological states or oxidative stress conditions by irrigation with spermidine and intraperitoneal injection of 3-Nitropropionic acid (3-NPA) in Sichuan white goose. Results and discussion Our results showed that spermidine could increase the ratio of intestinal villus to crypt and improve intestinal morphology. In addition, spermidine can also reduce malondialdehyde (MDA) accumulation caused by 3-NPA by increasing superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPX) enzyme activity, thus alleviating intestinal damage. Furthermore, spermidine can regulate intestinal digestive enzyme activities and affect intestinal digestion and absorption ability. Spermidine can also promote an increase in intestinal microbial diversity and abundance and alleviate the change of microflora structure caused by 3-NPA. In conclusion, spermidine promotes the production of beneficial intestinal metabolites such as Wikstromol, Alpha-bisabolol and AS 1-5, thus improving the level of intestinal health. Taken together, these results indicate that spermidine can improve intestinal health by improving intestinal morphology, increasing antioxidant capacity and regulating intestinal flora structure.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Bo Kang
- State Key Laboratory of Swine and Poultry Breeding Industry, Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
6
|
Zhang HY, Xiao HL, Wang GX, Lu ZQ, Xie MR, Li CS. Predictive value of presepsin and acylcarnitines for severity and biliary drainage in acute cholangitis. World J Gastroenterol 2023; 29:2502-2514. [PMID: 37179587 PMCID: PMC10167903 DOI: 10.3748/wjg.v29.i16.2502] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/21/2023] [Accepted: 03/31/2023] [Indexed: 04/24/2023] Open
Abstract
BACKGROUND Bacteremia, which is a major cause of mortality in patients with acute cholangitis, induces hyperactive immune response and mitochondrial dysfunction. Presepsin is responsible for pathogen recognition by innate immunity. Acylcarnitines are established mitochondrial biomarkers. AIM To clarify the early predictive value of presepsin and acylcarnitines as biomarkers of severity of acute cholangitis and the need for biliary drainage. METHODS Of 280 patients with acute cholangitis were included and the severity was stratified according to the Tokyo Guidelines 2018. Blood presepsin and plasma acylcarnitines were tested at enrollment by chemiluminescent enzyme immunoassay and ultra-high-performance liquid chromatography-mass spectrometry, respectively. RESULTS The concentrations of presepsin, procalcitonin, short- and medium-chain acylcarnitines increased, while long-chain acylcarnitines decreased with the severity of acute cholangitis. The areas under the receiver operating characteristic curves (AUC) of presepsin for diagnosing moderate/severe and severe cholangitis (0.823 and 0.801, respectively) were greater than those of conventional markers. The combination of presepsin, direct bilirubin, alanine aminotransferase, temperature, and butyryl-L-carnitine showed good predictive ability for biliary drainage (AUC: 0.723). Presepsin, procalcitonin, acetyl-L-carnitine, hydroxydodecenoyl-L-carnitine, and temperature were independent predictors of bloodstream infection. After adjusting for severity classification, acetyl-L-carnitine was the only acylcarnitine independently associated with 28-d mortality (hazard ratio 14.396; P < 0.001) (AUC: 0.880). Presepsin concentration showed positive correlation with direct bilirubin or acetyl-L-carnitine. CONCLUSION Presepsin could serve as a specific biomarker to predict the severity of acute cholangitis and need for biliary drainage. Acetyl-L-carnitine is a potential prognostic factor for patients with acute cholangitis. Innate immune response was associated with mitochondrial metabolic dysfunction in acute cholangitis.
Collapse
Affiliation(s)
- Han-Yu Zhang
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Hong-Li Xiao
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Guo-Xing Wang
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Zhao-Qing Lu
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Miao-Rong Xie
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Chun-Sheng Li
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
7
|
Ennab W, Ye N, Wu H, Ullah S, Hadi T, Bassey AP, Mustafa S, Jiang J, Wei Q, Shi F. The Synergistic Effects of the Combination of L-Carnitine and Lycopene on the Lycopene Bioavailability and Duodenal Health of Roosters. Animals (Basel) 2023; 13:ani13081274. [PMID: 37106837 PMCID: PMC10134981 DOI: 10.3390/ani13081274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/26/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
The objective of this study was to investigate the impact of Lycopene and L-Carnitine, individually or in combination, on various physiological and molecular factors related to intestinal health and absorption ability in Roosters, such as intestinal morphology, serum biochemical parameters, genes involved in Lycopene uptake, nutritional transport genes, and tight junction genes. The findings of the study revealed that the combination of L-Carnitine and Lycopene supplementation had been found to increase the serum concentration levels of TP and ALB. Interestingly, the relative mRNA expression of genes responsible for Lycopene uptakes, such as SR-BI and BCO2, was higher in the LC group compared to other groups. Additionally, the expression of specific nutritional transport genes in the duodenum was significantly affected by both CAR and LC supplementation groups. The tight junction gene OCLN showed a significant increase in expression in the combination group compared to using either Lycopene or L-Carnitine alone. This study concludes that using Lycopene and L-carnitine in combination in poultry feed can potentially improve intestinal morphology and serum biochemical parameters, increase Lycopene bioavailability, improve nutrients uptake, and enhance the integrity of duodenal tight junctions in Roosters.
Collapse
Affiliation(s)
- Wael Ennab
- National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Nanwei Ye
- National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Haoze Wu
- National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Saif Ullah
- National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Tavakolikazerooni Hadi
- National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Anthony Pius Bassey
- National Center of Meat Quality and Safety Control, Synergistic Innovation Center of Food Safety and Nutrition, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Sheeraz Mustafa
- Faculty of Veterinary Animal Sciences, Ziauddin University (ZUFVAS), Karachi 75600, Pakistan
| | - Jingle Jiang
- Shanghai Endangered Species Conservation and Research Centre, Shanghai Zoo, Shanghai 200335, China
| | - Quanwei Wei
- National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Fangxiong Shi
- National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
8
|
Gyimesi G, Hediger MA. Transporter-Mediated Drug Delivery. Molecules 2023; 28:molecules28031151. [PMID: 36770817 PMCID: PMC9919865 DOI: 10.3390/molecules28031151] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 01/27/2023] Open
Abstract
Transmembrane transport of small organic and inorganic molecules is one of the cornerstones of cellular metabolism. Among transmembrane transporters, solute carrier (SLC) proteins form the largest, albeit very diverse, superfamily with over 400 members. It was recognized early on that xenobiotics can directly interact with SLCs and that this interaction can fundamentally determine their efficacy, including bioavailability and intertissue distribution. Apart from the well-established prodrug strategy, the chemical ligation of transporter substrates to nanoparticles of various chemical compositions has recently been used as a means to enhance their targeting and absorption. In this review, we summarize efforts in drug design exploiting interactions with specific SLC transporters to optimize their therapeutic effects. Furthermore, we describe current and future challenges as well as new directions for the advanced development of therapeutics that target SLC transporters.
Collapse
|
9
|
One-year supplementation with Lactobacillus reuteri ATCC PTA 6475 counteracts a degradation of gut microbiota in older women with low bone mineral density. NPJ Biofilms Microbiomes 2022; 8:84. [PMID: 36261538 PMCID: PMC9581899 DOI: 10.1038/s41522-022-00348-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/06/2022] [Indexed: 11/10/2022] Open
Abstract
Recent studies have shown that probiotic supplementation has beneficial effects on bone metabolism. In a randomized controlled trial (RCT) we demonstrated that supplementation of Lactobacillus reuteri ATCC PTA 6475 reduced bone loss in older women with low bone mineral density. To investigate the mechanisms underlying the effect of L. reuteri ATCC PTA 6475 on bone metabolism, 20 women with the highest changes (good responders) and the lowest changes (poor responders) in tibia total volumetric BMD after one-year supplementation were selected from our previous RCT. In the current study we characterized the gut microbiome composition and function as well as serum metabolome in good responders and poor responders to the probiotic treatment as a secondary analysis. Although there were no significant differences in the microbial composition at high taxonomic levels, gene richness of the gut microbiota was significantly higher (P < 0.01 by the Wilcoxon rank-sum test) and inflammatory state was improved (P < 0.05 by the Wilcoxon signed-rank test) in the good responders at the end of the 12-month daily supplementation. Moreover, detrimental changes including the enrichment of E. coli (adjusted P < 0.05 by DESeq2) and its biofilm formation (P < 0.05 by GSA) observed in the poor responders were alleviated in the good responders by the treatment. Our results indicate that L. reuteri ATCC PTA 6475 supplementation has the potential to prevent a deterioration of the gut microbiota and inflammatory status in elderly women with low bone mineral density, which might have beneficial effects on bone metabolism.
Collapse
|
10
|
Liu T, Wang D, Zhou X, Song J, Yang Z, Shi C, Li R, Zhang Y, Zhang J, Yan J, Zhu X, Li Y, Gong M, Wang C, Yuan C, Cui Y, Wu X. Study on the mechanism of American ginseng extract for treating type 2 diabetes mellitus based on metabolomics. Front Pharmacol 2022; 13:960050. [PMID: 36120310 PMCID: PMC9479495 DOI: 10.3389/fphar.2022.960050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/01/2022] [Indexed: 11/29/2022] Open
Abstract
American ginseng extract (AGE) is an efficient and low-toxic adjuvant for type 2 diabetes mellitus (T2DM). However, the metabolic mechanisms of AGE against T2DM remain unknown. In this study, a rat model of T2DM was created and administered for 28 days. Their biological (body weight and serum biochemical indicators) and pathological (pancreatic sections stained with HE) information were collected for further pharmacodynamic evaluation. Moreover, an ultra-performance liquid chromatography–mass spectrometry–based (UHPLC–MS/MS–based) untargeted metabolomics method was used to identify potential biomarkers of serum samples from all rats and related metabolic pathways. The results indicated that body weight, fasting blood glucose (FBG), fasting blood insulin (FINS), blood triglyceride concentration (TG), high-density lipoprotein cholesterol (HDL-C), insulin resistance index (HOMA-IR) and insulin sensitivity index (ISI), and impaired islet cells were significantly improved after the high dose of AGE (H_AGE) and metformin treatment. Metabolomics analysis identified 101 potential biomarkers among which 94 metabolites had an obvious callback. These potential biomarkers were mainly enriched in nine metabolic pathways linked to amino acid metabolism and lipid metabolism. Tryptophan metabolism and glutathione metabolism, as differential metabolic pathways between AGE and metformin for treating T2DM, were further explored. Further analysis of the aforementioned results suggested that the anti-T2DM effect of AGE was closely associated with inflammation, oxidative stress, endothelial dysfunction, dyslipidemia, immune response, insulin resistance, insulin secretion, and T2DM-related complications. This study can provide powerful support for the systematic exploration of the mechanism of AGE against T2DM and a basis for the clinical diagnosis of T2DM.
Collapse
Affiliation(s)
- Tiantian Liu
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Dan Wang
- School of Pharmacy, Tianjin Medical University, Tianjin, China
- Department of Pharmacy, Chu Hisen-I Memorial Hospital, Tianjin Medical University, Tianjin, China
| | - Xinfeng Zhou
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Jiayin Song
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Zijun Yang
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Chang Shi
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Rongshan Li
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Yanwen Zhang
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Jun Zhang
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Jiuxing Yan
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xuehui Zhu
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Ying Li
- Tianjin Neurological Institute, Tianjin Medical University, Tianjin, China
| | - Min Gong
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Chongzhi Wang
- Tang Center for Herbal Medicine Research, University of Chicago, Chicago, IL, United States
| | - Chunsu Yuan
- Tang Center for Herbal Medicine Research, University of Chicago, Chicago, IL, United States
| | - Yan Cui
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- *Correspondence: Yan Cui, ; Xiaohui Wu,
| | - Xiaohui Wu
- School of Pharmacy, Tianjin Medical University, Tianjin, China
- *Correspondence: Yan Cui, ; Xiaohui Wu,
| |
Collapse
|
11
|
Trafficking to the Cell Surface of Amino Acid Transporter SLC6A14 Upregulated in Cancer Is Controlled by Phosphorylation of SEC24C Protein by AKT Kinase. Cells 2021; 10:cells10071800. [PMID: 34359969 PMCID: PMC8307180 DOI: 10.3390/cells10071800] [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: 06/17/2021] [Revised: 07/07/2021] [Accepted: 07/14/2021] [Indexed: 01/24/2023] Open
Abstract
Cancer cells need a constant supply of nutrients. SLC6A14, an amino acid transporter B0,+ (ATB0,+) that is upregulated in many cancers, transports all but acidic amino acids. In its exit from the endoplasmic reticulum (ER), it is recognized by the SEC24C subunit of coatomer II (COPII) for further vesicular trafficking to the plasma membrane. SEC24C has previously been shown to be phosphorylated by protein kinase B/AKT, which is hyper-activated in cancer; therefore, we analyzed the influence of AKT on SLC6A14 trafficking to the cell surface. Studies on overexpressed and endogenous transporters in the breast cancer cell line MCF-7 showed that AKT inhibition with MK-2206 correlated with a transient increase of the transporter in the plasma membrane, not resulting from the inhibition of ER-associated protein degradation. Two-dimensional electrophoresis demonstrated the decreased phosphorylation of SLC6A14 and SEC24C upon AKT inhibition. A proximity ligation assay confirmed this conclusion: AKT inhibition is correlated with decreased SLC6A14 phosphothreonine and SEC24C phosphoserine. Augmented levels of SLC6A14 in plasma membrane led to increased leucine transport. These results show that the inactivation of AKT can rescue amino acid delivery through SLC6A14 trafficking to the cell surface, supporting cancer cell survival. The regulation of the ER export of the amino acid transporter seems to be a novel function of AKT.
Collapse
|
12
|
Alanazi WA, Al-Harbi NO, Imam F, Ansari MA, Alhoshani A, Alasmari AF, Alasmari F, Alanazi MM, Ali N. Role of carnitine in regulation of blood pressure (MAP/SBP) and gene expression of cardiac hypertrophy markers (α/β-MHC) during insulin-induced hypoglycaemia: Role of oxidative stress. Clin Exp Pharmacol Physiol 2021; 48:478-489. [PMID: 33368625 DOI: 10.1111/1440-1681.13455] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 12/09/2020] [Indexed: 11/28/2022]
Abstract
Cardiovascular disease is a leading cause of death in diabetic patients. Hyperglycaemia and iatrogenic hypoglycaemia exacerbate several pathogenic mechanisms underlying hypertension and heart diseases. Carnitine is a potent endogenous antioxidant and cellular fatty acid transporter for antioxidative stress and energy production in the cardiovascular system. The current study aimed to find the role of carnitine in the regulation of hypoglycaemia-induced hypertension and cardiac hypertrophy. Male rats received insulin glargine (InG) to induce hypoglycaemia followed by D-carnitine or acetyl-L-carnitine for carnitine depletion or carnitine supplementation, respectively. The obtained results showed that carnitine deficiency provoked hypoglycaemia-induced hypertension. Mean arterial pressure was elevated from 78.16 ± 11.4 to 100 ± 5.11 mm Hg in InG treated group, and from 78.2 ± 8.5 to 123.4 ± 28.2 mm Hg in InG + D-carnitine treated group. Acetyl-L-carnitine resisted the elevation in blood pressure in all hypoglycaemic animals and kept it within the normal values (68.33 ± 6.7 mm Hg). Acetyl-L-carnitine increased myocardial carnitine content leading to the attenuation of hypoglycaemia-induced oxidative stress, which was evaluated through measurement of the oxidative stress biomarkers such as inducible nitric oxide synthase, NAD(P)H quinone dehydrogenase-1, heme oxygenase-I, and glutathione S-transferase. Moreover, acetyl-L-carnitine prevented induction of gene expression of cardiac hypertrophy markers during hypoglycaemic conditions, which was assessed via the evaluation of mRNA expression of α-myosin heavy chain and β-myosin heavy chain. These findings demonstrate that carnitine might play an essential role in prevention of hypoglycaemia-induced hypertension and cardiac hypertrophy through providing energy and antioxidants to the cardiovascular system.
Collapse
Affiliation(s)
- Wael A Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Naif O Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Faisal Imam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ali Alhoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed M Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
13
|
Li P, Sundh D, Ji B, Lappa D, Ye L, Nielsen J, Lorentzon M. Metabolic Alterations in Older Women With Low Bone Mineral Density Supplemented With Lactobacillus reuteri. JBMR Plus 2021; 5:e10478. [PMID: 33869994 PMCID: PMC8046097 DOI: 10.1002/jbm4.10478] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 02/03/2021] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis and its associated fractures are highly prevalent in older women. Recent studies have shown that gut microbiota play important roles in regulating bone metabolism. A previous randomized controlled trial (RCT) found that supplementation with Lactobacillus reuteri ATCC PTA 6475 (L.reuteri) led to substantially reduced bone loss in older women with low BMD. However, the total metabolic effects of L. reuteri supplementation on older women are still not clear. In this study, a post hoc analysis (not predefined) of serum metabolomic profiles of older women from the previous RCT was performed to investigate the metabolic dynamics over 1 year and to evaluate the effects of L. reuteri supplementation on human metabolism. Distinct segregation of the L. reuteri and placebo groups in response to the treatment was revealed by partial least squares‐discriminant analysis. Although no individual metabolite was differentially and significantly associated with treatment after correction for multiple testing, 97 metabolites responded differentially at any one time point between L. reuteri and placebo groups (variable importance in projection score >1 and p value <0.05). These metabolites were involved in multiple processes, including amino acid, peptide, and lipid metabolism. Butyrylcarnitine was particularly increased at all investigated time points in the L. reuteri group compared with placebo, indicating that the effects of L. reuteri on bone loss are mediated through butyrate signaling. Furthermore, the metabolomic profiles in a case (low BMD) and control population (high BMD) of elderly women were analyzed to confirm the associations between BMD and the identified metabolites regulated by L. reuteri supplementation. The amino acids, especially branched‐chain amino acids, showed association with L. reuteri treatment and with low BMD in older women, and may serve as potential therapeutic targets. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Peishun Li
- Department of Biology and Biological Engineering Chalmers University of Technology Gothenburg Sweden
| | - Daniel Sundh
- Geriatric Medicine, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Boyang Ji
- Department of Biology and Biological Engineering Chalmers University of Technology Gothenburg Sweden
| | - Dimitra Lappa
- Department of Biology and Biological Engineering Chalmers University of Technology Gothenburg Sweden
| | - Lingqun Ye
- Department of Biology and Biological Engineering Chalmers University of Technology Gothenburg Sweden
| | - Jens Nielsen
- Department of Biology and Biological Engineering Chalmers University of Technology Gothenburg Sweden.,Novo Nordisk Foundation Center for Biosustainability Technical University of Denmark Kgs. Lyngby Denmark.,BioInnovation Institute Copenhagen Denmark
| | - Mattias Lorentzon
- Geriatric Medicine, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden.,Region Västra Götaland, Geriatric Medicine Clinic Sahlgrenska University Hospital Mölndal Sweden.,Mary MacKillop Institute for Health Research Australian Catholic University Melbourne Victoria Australia
| |
Collapse
|
14
|
Nałęcz KA. Amino Acid Transporter SLC6A14 (ATB 0,+) - A Target in Combined Anti-cancer Therapy. Front Cell Dev Biol 2020; 8:594464. [PMID: 33195271 PMCID: PMC7609839 DOI: 10.3389/fcell.2020.594464] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer cells are characterized by quick growth and proliferation, demanding constant supply of various nutrients. Several plasma membrane transporters delivering such compounds are upregulated in cancer. Solute carrier family 6 member 14 (SLC6A14), known as amino acid transporter B0,+ (ATB0,+) transports all amino acids with exception of the acidic ones: aspartate and glutamate. Its malfunctioning is correlated with several pathological states and it is upregulated in solid tumors. The high expression of SLC6A14 is prognostic and unfavorable in pancreatic cancer, while in breast cancer it is expressed in estrogen receptor positive cells. As many plasma membrane transporters it resides in endoplasmic reticulum (ER) membrane after translation before further trafficking through Golgi to the cell surface. Transporter exit from ER is strictly controlled. The proper folding of SLC6A14 was shown to be controlled from the cytoplasmic side by heat shock proteins, further exit from ER and formation of coatomer II (COPII) coated vesicles depends on specific interaction with COPII cargo-recognizing subunit SEC24C, phosphorylated by kinase AKT. Inhibition of heat shock proteins, known to be upregulated in cancer, directs SLC6A14 to degradation. Targeting proteins regulating SLC6A14 trafficking is proposed as an additional pharmacological treatment of cancer.
Collapse
Affiliation(s)
- Katarzyna A Nałęcz
- Laboratory of Transport Through Biomembranes, Nencki Institute of Experimental Biology, Warsaw, Poland
| |
Collapse
|
15
|
Couto MR, Gonçalves P, Magro F, Martel F. Microbiota-derived butyrate regulates intestinal inflammation: Focus on inflammatory bowel disease. Pharmacol Res 2020; 159:104947. [DOI: 10.1016/j.phrs.2020.104947] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/04/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022]
|
16
|
Overcoming the intestinal barrier: A look into targeting approaches for improved oral drug delivery systems. J Control Release 2020; 322:486-508. [DOI: 10.1016/j.jconrel.2020.04.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 12/17/2022]
|
17
|
Singh S, Arthur S, Sundaram U. Mechanisms of Regulation of Transporters of Amino Acid Absorption in Inflammatory Bowel Diseases. Compr Physiol 2020; 10:673-686. [PMID: 32163200 DOI: 10.1002/cphy.c190016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Intestinal absorption of dietary amino acids/peptides is essential for protein homeostasis, which in turn is crucial for maintaining health as well as restoration of health from significant diseases. Dietary amino acids/peptides are absorbed by unique transporter processes present in the brush border membrane of absorptive villus cells, which line the entire length of the intestine. To date, the only nutrient absorptive system described in the secretory crypt cells in the mammalian intestine is the one that absorbs the amino acid glutamine. Majority of the amino acid transporters are sodium dependent and therefore require basolateral membrane Na-K-ATPase to maintain an efficient transcellular Na gradient for their activity. These transport processes are tightly regulated by various cellular and molecular mechanisms that facilitate their optimal activity during normal physiological processes. Malabsorption of amino acids, recently described in pathophysiological states such as in inflammatory bowel disease (IBD), is undoubtedly responsible for the debilitating symptoms of IBD such as malnutrition, weight loss and ultimately a failure to thrive. Also recently, in vivo models of IBD and in vitro studies have demonstrated that specific immune-inflammatory mediators/pathways regulate specific amino acid transporters. This provides possibilities to derive novel nutrition and immune-based treatment options for conditions such as IBD. © 2020 American Physiological Society. Compr Physiol 10:673-686, 2020.
Collapse
Affiliation(s)
- Soudamani Singh
- Department of Clinical and Translational Sciences and Appalachian Clinical and Translational Science Institute, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, USA
| | - Subha Arthur
- Department of Clinical and Translational Sciences and Appalachian Clinical and Translational Science Institute, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, USA
| | - Uma Sundaram
- Department of Clinical and Translational Sciences and Appalachian Clinical and Translational Science Institute, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, USA
| |
Collapse
|
18
|
Scalise M, Pochini L, Galluccio M, Console L, Indiveri C. Glutamine transporters as pharmacological targets: From function to drug design. Asian J Pharm Sci 2020; 15:207-219. [PMID: 32373200 PMCID: PMC7193454 DOI: 10.1016/j.ajps.2020.02.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/18/2020] [Accepted: 02/29/2020] [Indexed: 12/17/2022] Open
Abstract
Among the different targets of administered drugs, there are membrane transporters that play also a role in drug delivery and disposition. Moreover, drug-transporter interactions are responsible for off-target effects of drugs underlying their toxicity. The improvement of the drug design process is subjected to the identification of those membrane transporters mostly relevant for drug absorption, delivery and side effect production. A peculiar group of proteins with great relevance to pharmacology is constituted by the membrane transporters responsible for managing glutamine traffic in different body districts. The interest around glutamine metabolism lies in its physio-pathological role; glutamine is considered a conditionally essential amino acid because highly proliferative cells have an increased request of glutamine that cannot be satisfied only by endogenous synthesis. Then, glutamine transporters provide cells with this special nutrient. Among the glutamine transporters, SLC1A5, SLC6A14, SLC6A19, SLC7A5, SLC7A8 and some members of SLC38 family are the best characterized, so far, in both physiological and pathological conditions. Few 3D structures have been solved by CryoEM; other structural data on these transporters have been obtained by computational analysis. Interactions with drugs have been described for several transporters of this group. For some of them, the studies are at an advanced stage, for others, the studies are still in nuce and novel biochemical findings open intriguing perspectives.
Collapse
Affiliation(s)
- Mariafrancesca Scalise
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | - Lorena Pochini
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | - Michele Galluccio
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | - Lara Console
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | - Cesare Indiveri
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| |
Collapse
|
19
|
Wang G, Zhao L, Jiang Q, Sun Y, Zhao D, Sun M, He Z, Sun J, Wang Y. Intestinal OCTN2- and MCT1-targeted drug delivery to improve oral bioavailability. Asian J Pharm Sci 2020; 15:158-173. [PMID: 32256846 PMCID: PMC7118283 DOI: 10.1016/j.ajps.2020.02.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 12/08/2019] [Accepted: 02/12/2020] [Indexed: 12/18/2022] Open
Abstract
Various drug transporters are widely expressed throughout the intestine and play important roles in absorbing nutrients and drugs, thus providing high quality targets for the design of prodrugs or nanoparticles to facilitate oral drug delivery. In particular, intestinal carnitine/organic cation transporter 2 (OCTN2) and mono-carboxylate transporter protein 1 (MCT1) possess high transport capacities and complementary distributions. Therefore, we outline recent developments in transporter-targeted oral drug delivery with regard to the OCTN2 and MCT1 proteins in this review. First, basic information of the two transporters is reviewed, including their topological structures, characteristics and functions, expression and key features of their substrates. Furthermore, progress in transporter-targeting prodrugs and nanoparticles to increase oral drug delivery is discussed, including improvements in the oral absorption of anti-inflammatory drugs, antiepileptic drugs and anticancer drugs. Finally, the potential of a dual transporter-targeting strategy is discussed.
Collapse
Affiliation(s)
- Gang Wang
- Zhuang Yao Medicine Center of Engineering and Technology, Guang Xi University of Chinese Medicine, Nanning 530200, China
| | - Lichun Zhao
- Zhuang Yao Medicine Center of Engineering and Technology, Guang Xi University of Chinese Medicine, Nanning 530200, China.,School of Pharmacy, Guang Xi University of Chinese Medicine, Nanning 530200, China
| | - Qikun Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yixin Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dongyang Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Mengchi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhonggui He
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yang Wang
- School of Pharmacy, Guang Xi University of Chinese Medicine, Nanning 530200, China
| |
Collapse
|
20
|
Zhang L, Sui C, Yang W, Luo Q. Amino acid transporters: Emerging roles in drug delivery for tumor-targeting therapy. Asian J Pharm Sci 2020; 15:192-206. [PMID: 32373199 PMCID: PMC7193455 DOI: 10.1016/j.ajps.2019.12.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/22/2019] [Accepted: 12/22/2019] [Indexed: 12/16/2022] Open
Abstract
Amino acid transporters, which play a vital role in transporting amino acids for the biosynthesis of mammalian cells, are highly expressed in types of tumors. Increasing studies have shown the feasibility of amino acid transporters as a component of tumor-targeting therapy. In this review, we focus on tumor-related amino acid transporters and their potential use in tumor-targeting therapy. Firstly, the expression characteristics of amino acid transporters in cancer and their relationship with tumor growth are reviewed. Secondly, the recognition requirements are discussed, focusing on the "acid-base" properties, conformational isomerism and structural analogues. Finally, recent developments in amino acid transporter-targeting drug delivery strategies are highlighted, including prodrugs and nanocarriers, with special attention to the latest findings of molecular mechanisms and targeting efficiency of transporter-mediated endocytosis. We aim to offer related clues that might lead to valuable tumor-targeting strategies by the utilization of amino acid transporters.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Biotherapy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Chengguang Sui
- Department of Biotherapy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Wenhan Yang
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
- Department of Pharmacy, China Medical University, Shenyang 110001, China
| | - Qiuhua Luo
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
- Department of Pharmacy, China Medical University, Shenyang 110001, China
| |
Collapse
|
21
|
Kang S, You HJ, Lee YG, Jeong Y, Johnston TV, Baek NI, Ku S, Ji GE. Production, Structural Characterization, and In Vitro Assessment of the Prebiotic Potential of Butyl-Fructooligosaccharides. Int J Mol Sci 2020; 21:ijms21020445. [PMID: 31936703 PMCID: PMC7013684 DOI: 10.3390/ijms21020445] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/06/2020] [Accepted: 01/08/2020] [Indexed: 12/11/2022] Open
Abstract
Short-chain fatty acids (SCFAs), especially butyrate, produced in mammalian intestinal tracts via fermentation of dietary fiber, are known biofunctional compounds in humans. However, the variability of fermentable fiber consumed on a daily basis and the diversity of gut microbiota within individuals often limits the production of short-chain fatty acids in the human gut. In this study, we attempted to enhance the butyrate levels in human fecal samples by utilizing butyl-fructooligosaccharides (B-FOS) as a novel prebiotic substance. Two major types of B-FOS (GF3-1B and GF3-2B), composed of short-chain fructooligosaccharides (FOS) bound to one or two butyric groups by ester bonds, were synthesized. Qualitative analysis of these B-FOS using Fourier transform infrared (FT-IR) spectroscopy, matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS), nuclear magnetic resonance (NMR) and low-resolution fast-atom bombardment mass spectra (LR-FAB-MS), showed that the chemical structure of GF3-1B and GF3-2B were [O-(1-buty-β-D-fru-(2→1)-O-β-D-fru-(2→1)-O-β-D-fru-O-α-D-glu] and [O-(1-buty)-β-D-fru-(2→1)-O-β-D-fru-(2→1)-O-(4-buty)-β-D-fru-O-α-D-glu], respectively. The ratio of these two compounds was approximately 5:3. To verify their biofunctionality as prebiotic oligosaccharides, proliferation and survival patterns of human fecal microbiota were examined in vitro via 16S rRNA metagenomics analysis compared to a positive FOS control and a negative control without a carbon source. B-FOS treatment showed different enrichment patterns on the fecal microbiota community during fermentation, and especially stimulated the growth of major butyrate producing bacterial consortia and modulated specific butyrate producing pathways with significantly enhanced butyrate levels. Furthermore, the relative abundance of Fusobacterium and ammonia production with related metabolic genes were greatly reduced with B-FOS and FOS treatment compared to the control group. These findings indicate that B-FOS differentially promotes butyrate production through the enhancement of butyrate-producing bacteria and their metabolic genes, and can be applied as a novel prebiotic compound in vivo.
Collapse
Affiliation(s)
- Sini Kang
- Department of Food and Nutrition, Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea; (S.K.); (Y.J.)
| | - Hyun Ju You
- Institute of Health and Environment, Graduate School of Public Health, Seoul National University, Seoul 08826, Korea;
| | - Yeong-Geun Lee
- Graduate School of Biotechnology and Department of Oriental Medicinal Biotechnology, Kyung Hee University, Yongin 17104, Korea; (Y.-G.L.); (N.-I.B.)
| | - Yunju Jeong
- Department of Food and Nutrition, Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea; (S.K.); (Y.J.)
| | - Tony V. Johnston
- Fermentation Science Program, School of Agriculture, College of Basic and Applied Sciences, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
| | - Nam-In Baek
- Graduate School of Biotechnology and Department of Oriental Medicinal Biotechnology, Kyung Hee University, Yongin 17104, Korea; (Y.-G.L.); (N.-I.B.)
| | - Seockmo Ku
- Fermentation Science Program, School of Agriculture, College of Basic and Applied Sciences, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
- Correspondence: (S.K.); (G.E.J.); Tel.: +1-615-904-8290 (S.K.); +82-2-880-6282 (G.E.J.)
| | - Geun Eog Ji
- Department of Food and Nutrition, Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea; (S.K.); (Y.J.)
- Research Center, BIFIDO Co., Ltd., Hongcheon 25117, Korea
- Correspondence: (S.K.); (G.E.J.); Tel.: +1-615-904-8290 (S.K.); +82-2-880-6282 (G.E.J.)
| |
Collapse
|
22
|
Solute carrier transporters: the metabolic gatekeepers of immune cells. Acta Pharm Sin B 2020; 10:61-78. [PMID: 31993307 PMCID: PMC6977534 DOI: 10.1016/j.apsb.2019.12.006] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/29/2019] [Accepted: 10/31/2019] [Indexed: 02/06/2023] Open
Abstract
Solute carrier (SLC) transporters meditate many essential physiological functions, including nutrient uptake, ion influx/efflux, and waste disposal. In its protective role against tumors and infections, the mammalian immune system coordinates complex signals to support the proliferation, differentiation, and effector function of individual cell subsets. Recent research in this area has yielded surprising findings on the roles of solute carrier transporters, which were discovered to regulate lymphocyte signaling and control their differentiation, function, and fate by modulating diverse metabolic pathways and balanced levels of different metabolites. In this review, we present current information mainly on glucose transporters, amino-acid transporters, and metal ion transporters, which are critically important for mediating immune cell homeostasis in many different pathological conditions.
Collapse
Key Words
- 3-PG, 3-phosphoglyceric acid
- ABC, ATP-binding cassette
- AIF, apoptosis-inducing factor
- AP-1, activator protein 1
- ASCT2, alanine serine and cysteine transporter system 2
- ATP, adenosine triphosphate
- BCR, B cell receptor
- BMDMs, bone marrow-derived macrophages
- CD45R, a receptor-type protein tyrosine phosphatase
- CTL, cytotoxic T lymphocytes
- DC, dendritic cells
- EAATs, excitatory amino acid transporters
- ER, endoplasmic reticulum
- ERRα, estrogen related receptor alpha
- FFA, free fatty acids
- G-6-P, glucose 6-phosphate
- GLUT, glucose transporters
- GSH, glutathione
- Glucose
- Glutamine
- HIF-1α, hypoxia-inducible factor 1-alpha
- HIV-1, human immunodeficiency virus type 1
- Hk1, hexokinase-1
- IFNβ, interferon beta
- IFNγ, interferon gamma
- IKK, IκB kinase
- IKKβ, IκB kinase beta subunit
- IL, interleukin
- LDHA, lactate dehydrogenase A
- LPS, lipopolysaccharide
- Lymphocytes
- Lyn, tyrosine-protein kinase
- MAPK, mitogen-activated protein kinase
- MCT, monocarboxylate transporters
- MS, multiple sclerosis
- Metal ion
- NADPH, nicotinamide adenine dinucleotide phosphate
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NO, nitric oxide
- NOD2, nucleotide-binding oligomerization domain containing 2
- PEG2, prostaglandin E2
- PI-3K/AKT, phosphatidylinositol-3-OH kinase/serine–threonine kinase
- PPP, pentose phosphate pathway
- Pfk, phosphofructokinase
- RA, rheumatoid arthritis
- RLR, RIG-I-like receptor
- ROS, reactive oxygen species
- SLC, solute carrier
- SLE, systemic lupus erythematosus
- SNAT, sodium-coupled neutral amino acid transporters
- STAT, signal transducers and activators of transcription
- Solute carrier
- TAMs, tumor-associated macrophages
- TCA, tricarboxylic acid
- TCR, T cell receptor
- TLR, toll-like receptor
- TNF, tumor necrosis factor
- TRPM7, transient receptor potential cation channel subfamily M member 7
- Teffs, effector T cells
- Th1/2/17, type 1/2/17 helper T cells
- Tregs, regulatory T cells
- VEGF, vascular endothelial growth factor
- ZIP, zrt/irt-like proteins
- iNOS, inducible nitric oxide synthase
- iTregs, induced regulatory T cells
- mTORC1, mammalian target of rapamycin complex 1
- α-KG, α-ketoglutaric acid
Collapse
|
23
|
Hsu WH, Lee CH, Chao YM, Kuo CH, Ku WC, Chen CC, Lin YL. ASIC3-dependent metabolomics profiling of serum and urine in a mouse model of fibromyalgia. Sci Rep 2019; 9:12123. [PMID: 31431652 PMCID: PMC6702159 DOI: 10.1038/s41598-019-48315-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 07/30/2019] [Indexed: 12/27/2022] Open
Abstract
Fibromyalgia (FM) is characterized by chronic widespread pain. The pathogenesis of FM remains unclear. No specific biomarkers are available. Animal models of FM may provide an opportunity to explore potential biomarkers in a relative homogenous disease condition. Here, we probed the metabolomics profiles of serum and urine in a mouse model of FM induced by intermittent cold stress (ICS). We focused on the role of acid-sensing ion channel 3 (ASIC3) in the metabolomics profiling because ICS treatment induced chronic widespread muscle pain lasting for 1 month in wild-type (Asic3+/+) but not Asic3-knockout (Asic3−/−) mice. Serum and urine samples were collected from both genotypes at different ICS stages, including before ICS (basal level) and post-ICS at days 10 (middle phase, P10) and 40 (recovery phase, P40). Control naïve mice and ICS-induced FM mice differed in 1H-NMR- and LC-MS-based metabolomics profiling. On pathway analysis, the leading regulated pathways in Asic3+/+ mice were taurine and hypotaurine, cysteine and methionine, glycerophospholipid, and ascorbate and aldarate metabolisms, and the major pathways in Asic3−/− mice involved amino acid-related metabolism. Finally, we developed an algorithm for the impactful metabolites in the FM model including cis-aconitate, kynurenate, taurine, pyroglutamic acid, pyrrolidonecarboxylic acid, and 4-methoxyphenylacetic acid in urine as well as carnitine, deoxycholic acid, lysoPC(16:0), lysoPC(20:3), oleoyl-L-carnitine, and trimethylamine N-oxide in serum. Asic3−/− mice were impaired in only muscle allodynia development but not other pain symptoms in the ICS model, so the ASIC3-dependent metabolomics changes could be useful for developing diagnostic biomarkers specific to chronic widespread muscle pain, the core symptom of FM. Further pharmacological validations are needed to validate these metabolomics changes as potential biomarkers for FM diagnosis and/or treatment responses.
Collapse
Affiliation(s)
- Wei-Hsiang Hsu
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, 40402, Taiwan
| | - Cheng-Han Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Yen-Ming Chao
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, 40402, Taiwan
| | - Ching-Hua Kuo
- Department of Pharmacy, National Taiwan University, Taipei, 100, Taiwan
| | - Wei-Chi Ku
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, 24205, Taiwan
| | - Chih-Cheng Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan. .,Taiwan Mouse Clinic - National Comprehensive Mouse Phenotyping and Drug Testing Center, Academia Sinica, Taipei, 115, Taiwan.
| | - Yun-Lian Lin
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
24
|
Hausner EA, Elmore SA, Yang X. Overview of the Components of Cardiac Metabolism. Drug Metab Dispos 2019; 47:673-688. [PMID: 30967471 PMCID: PMC7333657 DOI: 10.1124/dmd.119.086611] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/26/2019] [Indexed: 12/20/2022] Open
Abstract
Metabolism in organs other than the liver and kidneys may play a significant role in how a specific organ responds to chemicals. The heart has metabolic capability for energy production and homeostasis. This homeostatic machinery can also process xenobiotics. Cardiac metabolism includes the expression of numerous organic anion transporters, organic cation transporters, organic carnitine (zwitterion) transporters, and ATP-binding cassette transporters. Expression and distribution of the transporters within the heart may vary, depending on the patient's age, disease, endocrine status, and various other factors. Several cytochrome P450 (P450) enzyme classes have been identified within the heart. The P450 hydroxylases and epoxygenases within the heart produce hydroxyeicosatetraneoic acids and epoxyeicosatrienoic acids, metabolites of arachidonic acid, which are critical in regulating homeostatic processes of the heart. The susceptibility of the cardiac P450 system to induction and inhibition from exogenous materials is an area of expanding knowledge, as are the metabolic processes of glucuronidation and sulfation in the heart. The susceptibility of various transcription factors and signaling pathways of the heart to disruption by xenobiotics is not fully characterized but is an area with implications for disruption of normal postnatal development, as well as modulation of adult cardiac health. There are knowledge gaps in the timelines of physiologic maturation and deterioration of cardiac metabolism. Cross-species characterization of cardiac-specific metabolism is needed for nonclinical work of optimum translational value to predict possible adverse effects, identify sensitive developmental windows for the design and conduct of informative nonclinical and clinical studies, and explore the possibilities of organ-specific therapeutics.
Collapse
Affiliation(s)
- Elizabeth A Hausner
- United States Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, Maryland (E.A.H., X.Y.); and National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (S.A.E.)
| | - Susan A Elmore
- United States Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, Maryland (E.A.H., X.Y.); and National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (S.A.E.)
| | - Xi Yang
- United States Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, Maryland (E.A.H., X.Y.); and National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (S.A.E.)
| |
Collapse
|
25
|
Irizarry KA, Mager DR, Triador L, Muehlbauer MJ, Haqq AM, Freemark M. Hormonal and metabolic effects of carbohydrate restriction in children with Prader-Willi syndrome. Clin Endocrinol (Oxf) 2019; 90:553-561. [PMID: 30614551 PMCID: PMC6805129 DOI: 10.1111/cen.13933] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 11/23/2018] [Accepted: 01/01/2019] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Macronutrient regulation of hyperphagia and adiposity in Prader-Willi syndrome (PWS) is poorly understood. We compared fasting and postprandial concentrations of hormones and metabolites in eight PWS children (age 9-18 years) fed, in random order, low carbohydrate, high-fat (LC, 15% carb; 65% fat; 20% protein) and low-fat, high carbohydrate (LF, 65% carb, 15% fat, 20% protein) diets matched for calories and protein. METHODS Participants were randomized to consume either the LC or LF diet during a first hospital admission and the second diet during a subsequent admission. Blood samples were obtained after overnight fasting and 1 hour after a mixed meal. RESULTS Relative to subjects consuming the LF diet, subjects consuming the LC diet had: lower postprandial insulin concentrations (P = 0.02); higher fasting GLP-1 AND GIP concentrations and increased postprandial GLP-1 (P < 0.02); reduced ratio of fasting ghrelin to GLP-1 (P = 0.0078); increased FFA and fatty acid oxidation, as assessed by concentrations of even-chain acylcarnitines (P < 0.001); lower fasting TG and TG/HDL ratio (P < 0.01); and higher concentrations of branch chain amino acids (P < 0.01). There were no changes in glucose, PYY, or adiponectin. CRP, AST and ALT were all higher (P < 0.01) on the LC diet. CONCLUSIONS Increases in GLP-1 with low carbohydrate feeding and reductions in the ratio of ghrelin to GLP-1 might limit food intake and improve glycaemic control in PWS. Other potential benefits of carbohydrate restriction may include fat mobilization and oxidation and reductions in the TG/HDL ratio, a marker of insulin resistance. However, increases in CRP, AST and ALT necessitate longer-term studies of low carbohydrate efficacy and safety.
Collapse
Affiliation(s)
- Krystal A. Irizarry
- Division of Pediatric Endocrinology and Diabetes, Duke University Medical Center, Durham NC
| | - Diana R. Mager
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Canada
| | - Lucila Triador
- Division of Pediatric Endocrinology, University of Alberta, Edmonton, Canada
| | | | - Andrea M. Haqq
- Division of Pediatric Endocrinology, University of Alberta, Edmonton, Canada
| | - Michael Freemark
- Division of Pediatric Endocrinology and Diabetes, Duke University Medical Center, Durham NC
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham NC
| |
Collapse
|
26
|
Pochini L, Galluccio M, Scalise M, Console L, Indiveri C. OCTN: A Small Transporter Subfamily with Great Relevance to Human Pathophysiology, Drug Discovery, and Diagnostics. SLAS DISCOVERY 2018; 24:89-110. [PMID: 30523710 DOI: 10.1177/2472555218812821] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OCTN is a small subfamily of membrane transport proteins that belongs to the larger SLC22 family. Two of the three members of the subfamily, namely, OCTN2 and OCTN1, are present in humans. OCTN2 plays a crucial role in the absorption of carnitine from diet and in its distribution to tissues, as demonstrated by the occurrence of severe pathologies caused by malfunctioning or altered expression of this transporter. These findings suggest avoiding a strict vegetarian diet during pregnancy and in childhood. Other roles of OCTN2 are related to the traffic of carnitine derivatives in many tissues. The role of OCTN1 is still unclear, despite the identification of some substrates such as ergothioneine, acetylcholine, and choline. Plausibly, the transporter acts on the control of inflammation and oxidative stress, even though knockout mice do not display phenotypes. A clear role of both transporters has been revealed in drug interaction and delivery. The polyspecificity of the OCTNs is at the base of the interactions with drugs. Interestingly, OCTN2 has been recently exploited in the prodrug approach and in diagnostics. A promising application derives from the localization of OCTN2 in exosomes that represent a noninvasive diagnostic tool.
Collapse
Affiliation(s)
- Lorena Pochini
- 1 Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Michele Galluccio
- 1 Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Mariafrancesca Scalise
- 1 Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Lara Console
- 1 Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Cesare Indiveri
- 1 Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy.,2 CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, Bari, Italy
| |
Collapse
|
27
|
Kou L, Sun R, Ganapathy V, Yao Q, Chen R. Recent advances in drug delivery via the organic cation/carnitine transporter 2 (OCTN2/SLC22A5). Expert Opin Ther Targets 2018; 22:715-726. [PMID: 30016594 DOI: 10.1080/14728222.2018.1502273] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Transporters in the plasma membrane have been exploited successfully for the delivery of drugs in the form of prodrugs and nanoparticles. Organic cation/carnitine transporter 2 (OCTN2, SLC22A5) has emerged as a viable target for drug delivery. OCTN2 is a Na+-dependent high-affinity transporter for L-carnitine and a Na+-independent transporter for organic cations. OCTN2 is expressed in the blood-brain barrier, heart, liver, kidney, intestinal tract and placenta and plays an essential role in L-carnitine homeostasis in the body. Areas covered: In recent years, several studies have been reported in the literature describing the utility of OCTN2 to enhance the delivery of drugs, prodrugs and nanoparticles. Here we summarize the salient features of OCTN2 in terms of its role in the cellular uptake of its physiological substrate L-carnitine in physiological and pathological context; the structural requirements for recognition and the recent advances in OCTN2-targeted drug delivery systems, including prodrugs and nanoparticles, are discussed. Expert opinion: This transporter has great potential to be utilized as a target for drug delivery to improve oral absorption of drugs in the intestinal tract. It also has potential to facilitate the transfer of drugs across the biological barriers such as the blood-brain barrier, blood-retinal barrier, and maternal-fetal barrier.
Collapse
Affiliation(s)
- Longfa Kou
- a Department of Pharmacy , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| | - Rui Sun
- a Department of Pharmacy , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| | - Vadivel Ganapathy
- a Department of Pharmacy , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China.,b Department of Cell Biology and Biochemistry , School of Medicine, Texas Tech University Health Sciences Center , Lubbock , TX , USA
| | - Qing Yao
- c School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , China
| | - Ruijie Chen
- a Department of Pharmacy , The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou , China
| |
Collapse
|
28
|
Sikder MOF, Yang S, Ganapathy V, Bhutia YD. The Na+/Cl−-Coupled, Broad-Specific, Amino Acid Transporter SLC6A14 (ATB0,+): Emerging Roles in Multiple Diseases and Therapeutic Potential for Treatment and Diagnosis. AAPS JOURNAL 2017; 20:12. [DOI: 10.1208/s12248-017-0164-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/13/2017] [Indexed: 12/21/2022]
|
29
|
Nealon NJ, Yuan L, Yang X, Ryan EP. Rice Bran and Probiotics Alter the Porcine Large Intestine and Serum Metabolomes for Protection against Human Rotavirus Diarrhea. Front Microbiol 2017; 8:653. [PMID: 28484432 PMCID: PMC5399067 DOI: 10.3389/fmicb.2017.00653] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 03/30/2017] [Indexed: 01/20/2023] Open
Abstract
Human rotavirus (HRV) is a leading cause of severe childhood diarrhea, and there is limited vaccine efficacy in the developing world. Neonatal gnotobiotic pigs consuming a prophylactic synbiotic combination of probiotics and rice bran (Pro+RB) did not exhibit HRV diarrhea after challenge. Multiple immune, gut barrier protective, and anti-diarrheal mechanisms contributed to the prophylactic efficacy of Pro+RB when compared to probiotics (Pro) alone. In order to understand the molecular signature associated with diarrheal protection by Pro+RB, a global non-targeted metabolomics approach was applied to investigate the large intestinal contents and serum of neonatal gnotobiotic pigs. The ultra-high performance liquid chromatography-tandem mass spectrometry platform revealed significantly different metabolites (293 in LIC and 84 in serum) in the pigs fed Pro+RB compared to Pro, and many of these metabolites were lipids and amino acid/peptides. Lipid metabolites included 2-oleoylglycerol (increased 293.40-fold in LIC of Pro+RB, p = 3.04E-10), which can modulate gastric emptying, andhyodeoxycholate (decreased 0.054-fold in the LIC of Pro+RB, p = 0.0040) that can increase colonic mucus production to improve intestinal barrier function. Amino acid metabolites included cysteine (decreased 0.40-fold in LIC, p = 0.033, and 0.62-fold in serum, p = 0.014 of Pro+RB), which has been found to reduce inflammation, lower oxidative stress and modulate mucosal immunity, and histamine (decreased 0.18-fold in LIC, p = 0.00030, of Pro+RB and 1.57-fold in serum, p = 0.043), which modulates local and systemic inflammatory responses as well as influences the enteric nervous system. Alterations to entire LIC and serum metabolic pathways further contributed to the anti-diarrheal and anti-viral activities of Pro+RB such as sphingolipid, mono/diacylglycerol, fatty acid, secondary bile acid, and polyamine metabolism. Sphingolipid and long chain fatty acid profiles influenced the ability of HRV to both infect and replicate within cells, suggesting that Pro+RB created a protective lipid profile that interferes with HRV activity. Polyamines act on enterocyte calcium-sensing receptors to modulate intracellular calcium levels, and may directly interfere with rotavirus replication. These results support that multiple host and probiotic metabolic networks, notably those involving lipid and amino acid/peptide metabolism, are important mechanisms through which Pro+RB protected against HRV diarrhea in neonatal gnotobiotic pigs.
Collapse
Affiliation(s)
- Nora Jean Nealon
- Nutrition and Toxicology Laboratory, Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort CollinsCO, USA
| | - Lijuan Yuan
- Yuan Laboratory, Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, BlacksburgVA, USA
| | - Xingdong Yang
- Laboratory of Infectious Diseases, Viral Pathogenesis and Evolution Section, National Institute of Allergy and Infectious Diseases, National Institute of Health, BethesdaMD, USA
| | - Elizabeth P Ryan
- Nutrition and Toxicology Laboratory, Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort CollinsCO, USA
| |
Collapse
|
30
|
Murakami T. A Minireview: Usefulness of Transporter-Targeted Prodrugs in Enhancing Membrane Permeability. J Pharm Sci 2016; 105:2515-2526. [DOI: 10.1016/j.xphs.2016.05.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/28/2016] [Accepted: 05/03/2016] [Indexed: 11/26/2022]
|
31
|
Manal M, Chandrasekar M, Gomathi Priya J, Nanjan M. Inhibitors of histone deacetylase as antitumor agents: A critical review. Bioorg Chem 2016; 67:18-42. [DOI: 10.1016/j.bioorg.2016.05.005] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/29/2016] [Accepted: 05/15/2016] [Indexed: 12/11/2022]
|
32
|
Irwin MH, Moos WH, Faller DV, Steliou K, Pinkert CA. Epigenetic Treatment of Neurodegenerative Disorders: Alzheimer and Parkinson Diseases. Drug Dev Res 2016; 77:109-23. [PMID: 26899010 DOI: 10.1002/ddr.21294] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Preclinical Research In this review, we discuss epigenetic-driven methods for treating neurodegenerative disorders associated with mitochondrial dysfunction, focusing on carnitinoid antioxidant-histone deacetylase inhibitors that show an ability to reinvigorate synaptic plasticity and protect against neuromotor decline in vivo. Aging remains a major risk factor in patients who progress to dementia, a clinical syndrome typified by decreased mental capacity, including impairments in memory, language skills, and executive function. Energy metabolism and mitochondrial dysfunction are viewed as determinants in the aging process that may afford therapeutic targets for a host of disease conditions, the brain being primary in such thinking. Mitochondrial dysfunction is a core feature in the pathophysiology of both Alzheimer and Parkinson diseases and rare mitochondrial diseases. The potential of new therapies in this area extends to glaucoma and other ophthalmic disorders, migraine, Creutzfeldt-Jakob disease, post-traumatic stress disorder, systemic exertion intolerance disease, and chemotherapy-induced cognitive impairment. An emerging and hopefully more promising approach to addressing these hard-to-treat diseases leverages their sensitivity to activation of master regulators of antioxidant and cytoprotective genes, antioxidant response elements, and mitophagy. Drug Dev Res 77 : 109-123, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael H Irwin
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Walter H Moos
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, CA, USA.,SRI Biosciences, A Division of SRI International, Menlo Park, CA, USA
| | - Douglas V Faller
- Cancer Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Kosta Steliou
- Cancer Research Center, Boston University School of Medicine, Boston, MA, USA.,PhenoMatriX, Inc., Boston, MA, USA
| | - Carl A Pinkert
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA.,Department of Biological Sciences, College of Arts and Sciences, The University of Alabama, Tuscaloosa, AL, USA
| |
Collapse
|
33
|
Bioprotective Carnitinoids: Lipoic Acid, Butyrate, and Mitochondria-Targeting to Treat Radiation Injury: Mitochondrial Drugs Come of Age. Drug Dev Res 2015; 76:167-75. [DOI: 10.1002/ddr.21258] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 05/28/2015] [Indexed: 12/16/2022]
|
34
|
Wojtal KA, Cee A, Lang S, Götze O, Frühauf H, Geier A, Pastor-Anglada M, Torres-Torronteras J, Martí R, Fried M, Lutz TA, Maggiorini M, Gassmann M, Rogler G, Vavricka SR. Downregulation of duodenal SLC transporters and activation of proinflammatory signaling constitute the early response to high altitude in humans. Am J Physiol Gastrointest Liver Physiol 2014; 307:G673-88. [PMID: 24970780 DOI: 10.1152/ajpgi.00353.2013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Solute carrier (SLC) transporters mediate the uptake of biologically active compounds in the intestine. Reduced oxygenation (hypoxia) is an important factor influencing intestinal homeostasis. The aim of this study was to investigate the pathophysiological consequences of hypoxia on the expression and function of SLCs in human intestine. Hypoxia was induced in human intestinal epithelial cells (IECs) in vitro (0.2; 1% O2 or CoCl2). For human in vivo studies, duodenal biopsies and serum samples were obtained from individuals (n = 16) acutely exposed to 4,554 meters above sea levels. Expression of relevant targets was analyzed by quantitative PCR, Western blotting, or immunofluorescence. Serum levels of inflammatory mediators and nucleosides were determined by ELISA and LC/MS-MS, respectively. In the duodenum of volunteers exposed to high altitude we observed decreased mRNA levels of apical sodium-dependent bile acid transporter (ASBT), concentrative nucleoside transporters 1/2 (CNT1/2), organic anion transporting polypeptide 2B1 (OATP2B1), organic cation transporter 2 (OCTN2), peptide transporter 1 (PEPT1), serotonin transporter (SERT), and higher levels of IFN-γ, IL-6, and IL-17A. Serum levels of IL-10, IFN-γ, matrix metalloproteinase-2 (MMP-2), and serotonin were elevated, whereas the levels of uridine decreased upon exposure to hypoxia. Hypoxic IECs showed reduced levels of equilibrative nucleoside transporter 2 (ENT2), OCTN2, and SERT mRNAs in vitro, which was confirmed on the protein level and was accompanied by activation of ERK1/2, increase of hypoxia-inducible factor (HIF) proteins, and production of IL-8 mRNA. Costimulation with IFN-γ and IL-6 during hypoxia further decreased the expression of SERT, ENT2, and CNT2 in vitro. Reduced oxygen supply affects the expression pattern of duodenal SLCs that is accompanied by changes in serum levels of proinflammatory cytokines and biologically active compounds demonstrating that intestinal transport is affected during systemic exposure to hypoxia in humans.
Collapse
Affiliation(s)
- Kacper A Wojtal
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University Hospital Zurich, Zurich, Switzerland;
| | - Alexandra Cee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Silvia Lang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Oliver Götze
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University Hospital Zurich, Zurich, Switzerland; Department of Gastroenterology and Hepatology, University Clinic Würzburg, Würzburg, Germany
| | - Heiko Frühauf
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University Hospital Zurich, Zurich, Switzerland; Division of Gastroenterology and Hepatology, Hospital Triemli, Zurich, Switzerland
| | - Andreas Geier
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University Hospital Zurich, Zurich, Switzerland; Department of Gastroenterology and Hepatology, University Clinic Würzburg, Würzburg, Germany
| | - Marçal Pastor-Anglada
- Pharmacology and Experimental Therapeutics, Department of Biochemistry and Molecular Biology, Institute of Biomedicine and Oncology Programme, National Biomedical Research Institute of Liver and Gastrointestinal Disease (CIBER EHD), University of Barcelona, Barcelona, Spain
| | - Javier Torres-Torronteras
- Neuromuscular and Mitochondrial Disorders Unit, and Biomedical Network Research Centre on Rare Diseases (CIBERER), Vall d'Hebron Institut de Recerca, Autonomous University of Barcelona, Barcelona, Spain
| | - Ramon Martí
- Neuromuscular and Mitochondrial Disorders Unit, and Biomedical Network Research Centre on Rare Diseases (CIBERER), Vall d'Hebron Institut de Recerca, Autonomous University of Barcelona, Barcelona, Spain
| | - Michael Fried
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), Zurich, Switzerland
| | - Marco Maggiorini
- Intensive Care Unit, Department of Internal Medicine, University of Zurich, Zurich, Switzerland
| | - Max Gassmann
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Cayetano Heredia University (UPCH), Lima, Peru; and Zurich Center for Integrative Human Physiology (ZIHP), Zurich, Switzerland
| | - Gerhard Rogler
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University Hospital Zurich, Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), Zurich, Switzerland
| | - Stephan R Vavricka
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University Hospital Zurich, Zurich, Switzerland; Division of Gastroenterology and Hepatology, Hospital Triemli, Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), Zurich, Switzerland
| |
Collapse
|
35
|
Tousson E, Hafez E, Zaki S, Gad A. P53, Bcl-2 and CD68 expression in response to amethopterin-induced lung injury and ameliorating role of L-carnitine. Biomed Pharmacother 2014; 68:631-9. [PMID: 24986327 DOI: 10.1016/j.biopha.2014.05.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 05/31/2014] [Indexed: 11/25/2022] Open
Abstract
Amethopterin (methotrexate, MTX) is an antimetabolite and antifolate drug with antiflammatory properities and is used to treat autoimmune diseases, such as psoriasis, rheumatoid arthritis and certain types of cancer, such as breast, lymphoma and lung. The present study aimed to study the changes in P53, Bcl-2 and CD68 expression in response to amethopterin-induced lung injury and ameliorating the role of l-carnitine. A total of 36 male albino rats were equally divided into six groups: the first and second groups were the control and l-carnitine groups respectively while the 3rd group was amethopterin rat group; the 4th and 5th groups were co- and post-treated amethopterin rat with l-carnitine respectively and the 6th group was self treated amethopterin rat group. Our results shows that lung in amethopterin-treated rats showed many of histopathological alterations as severe to strong alveolar damage in the form of collapsed alveoli and strong thickened interalveolar septa with heavy infiltration of inflammatory cells. This damage was increased or remaining in self-amethopterin-treated group. Treatment (co- and post) with l-carnitine were improved in the lung structure that was treated with amethopterin. A significant increase in p53 and CD68 and decrease in Bc1-2 immunoreactivity in the lung in amethopterin group is observed when compared with the control group. However, treatment of rats with l-carnitine decreased the intensity of P53-ir and CD68-ir and increased the intensity of Bcl-2 in lung when compared with amethopterin rat group. Co-treatment with l-carnitine improved lung damage induced with amethopterin.
Collapse
Affiliation(s)
- Ehab Tousson
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt; Biology Department, Faculty of Science, Tabuk University, Tabuk 71491, Saudi Arabia.
| | - Ezar Hafez
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Somia Zaki
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Amani Gad
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
36
|
Sahoo S, Aurich MK, Jonsson JJ, Thiele I. Membrane transporters in a human genome-scale metabolic knowledgebase and their implications for disease. Front Physiol 2014; 5:91. [PMID: 24653705 PMCID: PMC3949408 DOI: 10.3389/fphys.2014.00091] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 02/17/2014] [Indexed: 01/18/2023] Open
Abstract
Membrane transporters enable efficient cellular metabolism, aid in nutrient sensing, and have been associated with various diseases, such as obesity and cancer. Genome-scale metabolic network reconstructions capture genomic, physiological, and biochemical knowledge of a target organism, along with a detailed representation of the cellular metabolite transport mechanisms. Since the first reconstruction of human metabolism, Recon 1, published in 2007, progress has been made in the field of metabolite transport. Recently, we published an updated reconstruction, Recon 2, which significantly improved the metabolic coverage and functionality. Human metabolic reconstructions have been used to investigate the role of metabolism in disease and to predict biomarkers and drug targets. Given the importance of cellular transport systems in understanding human metabolism in health and disease, we analyzed the coverage of transport systems for various metabolite classes in Recon 2. We will review the current knowledge on transporters (i.e., their preferred substrates, transport mechanisms, metabolic relevance, and disease association for each metabolite class). We will assess missing coverage and propose modifications and additions through a transport module that is functional when combined with Recon 2. This information will be valuable for further refinements. These data will also provide starting points for further experiments by highlighting areas of incomplete knowledge. This review represents the first comprehensive overview of the transporters involved in central metabolism and their transport mechanisms, thus serving as a compendium of metabolite transporters specific for human metabolic reconstructions.
Collapse
Affiliation(s)
- Swagatika Sahoo
- Center for Systems Biology, University of Iceland Reykjavik, Iceland ; Molecular Systems Physiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg Belval, Luxembourg
| | - Maike K Aurich
- Center for Systems Biology, University of Iceland Reykjavik, Iceland ; Molecular Systems Physiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg Belval, Luxembourg
| | - Jon J Jonsson
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Iceland Reykjavik, Iceland ; Department of Genetics and Molecular Medicine, Landspitali, National University Hospital of Iceland Reykjavik, Iceland
| | - Ines Thiele
- Center for Systems Biology, University of Iceland Reykjavik, Iceland ; Molecular Systems Physiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg Belval, Luxembourg
| |
Collapse
|
37
|
Salsoso R, Guzmán-Gutiérrez E, Arroyo P, Salomón C, Zambrano S, Ruiz-Armenta MV, Blanca AJ, Pardo F, Leiva A, Mate A, Sobrevia L, Vázquez CM. Reduced L-carnitine transport in aortic endothelial cells from spontaneously hypertensive rats. PLoS One 2014; 9:e90339. [PMID: 24587332 PMCID: PMC3938671 DOI: 10.1371/journal.pone.0090339] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 01/30/2014] [Indexed: 12/28/2022] Open
Abstract
Impaired L-carnitine uptake correlates with higher blood pressure in adult men, and L-carnitine restores endothelial function in aortic rings from spontaneously hypertensive rat (SHR). Thus, endothelial dysfunction in hypertension could result from lower L-carnitine transport in this cell type. L-Carnitine transport is mainly mediated by novel organic cation transporters 1 (Octn1, Na+-independent) and 2 (Octn2, Na+-dependent); however, their kinetic properties and potential consequences in hypertension are unknown. We hypothesize that L-carnitine transport kinetic properties will be altered in aortic endothelium from spontaneously hypertensive rats (SHR). L-Carnitine transport was measured at different extracellular pH (pHo 5.5–8.5) in the absence or presence of sodium in rat aortic endothelial cells (RAECs) from non-hypertensive Wistar-Kyoto (WKY) rats and SHR. Octn1 and Octn2 mRNA relative expression was also determined. Dilation of endothelium-intact or denuded aortic rings in response to calcitonine gene related peptide (CGRP, 0.1–100 nmol/L) was measured (myography) in the absence or presence of L-carnitine. Total L-carnitine transport was lower in cells from SHR compared with WKY rats, an effect due to reduced Na+-dependent (Na+dep) compared with Na+-independent (Na+indep) transport components. Saturable L-carnitine transport kinetics show maximal velocity (Vmax), without changes in apparent Km for Na+indep transport in SHR compared with WKY rats. Total and Na+dep component of transport were increased, but Na+indep transport was reduced by extracellular alkalization in WKY rats. However, alkalization reduced total and Na+indep transport in cells from SHR. Octn2 mRNA was higher than Octn-1 mRNA expression in cells from both conditions. Dilation of artery rings in response to CGRP was reduced in vessels from SHR compared with WKY rats. CGRP effect was endothelium-dependent and restored by L-carnitine. All together these results suggest that reduced L-carnitine transport (likely via Na+-dependent Octn2) could limit this compound's potential beneficial effects in RAECs from SHR.
Collapse
Affiliation(s)
- Rocío Salsoso
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Enrique Guzmán-Gutiérrez
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Arroyo
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos Salomón
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sonia Zambrano
- Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Sevilla, Spain
| | | | - Antonio Jesús Blanca
- Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Sevilla, Spain
| | - Fabián Pardo
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Andrea Leiva
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alfonso Mate
- Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Sevilla, Spain
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, Queensland, Australia
- * E-mail: (CMV); (LS)
| | - Carmen María Vázquez
- Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Sevilla, Spain
- * E-mail: (CMV); (LS)
| |
Collapse
|
38
|
Koepsell H. The SLC22 family with transporters of organic cations, anions and zwitterions. Mol Aspects Med 2013; 34:413-35. [PMID: 23506881 DOI: 10.1016/j.mam.2012.10.010] [Citation(s) in RCA: 275] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Accepted: 08/18/2012] [Indexed: 12/14/2022]
Abstract
The SLC22 family contains 13 functionally characterized human plasma membrane proteins each with 12 predicted α-helical transmembrane domains. The family comprises organic cation transporters (OCTs), organic zwitterion/cation transporters (OCTNs), and organic anion transporters (OATs). The transporters operate as (1) uniporters which mediate facilitated diffusion (OCTs, OCTNs), (2) anion exchangers (OATs), and (3) Na(+)/zwitterion cotransporters (OCTNs). They participate in small intestinal absorption and hepatic and renal excretion of drugs, xenobiotics and endogenous compounds and perform homeostatic functions in brain and heart. Important endogeneous substrates include monoamine neurotransmitters, l-carnitine, α-ketoglutarate, cAMP, cGMP, prostaglandins, and urate. It has been shown that mutations of the SLC22 genes encoding these transporters cause specific diseases like primary systemic carnitine deficiency and idiopathic renal hypouricemia and are correlated with diseases such as Crohn's disease and gout. Drug-drug interactions at individual transporters may change pharmacokinetics and toxicities of drugs.
Collapse
Affiliation(s)
- Hermann Koepsell
- University of Würzburg, Institute of Anatomy and Cell Biology, Koellikerstr. 6, 97070 Würzburg, Germany.
| |
Collapse
|
39
|
Llorach R, Urpi-Sarda M, Tulipani S, Garcia-Aloy M, Monagas M, Andres-Lacueva C. Metabolomic fingerprint in patients at high risk of cardiovascular disease by cocoa intervention. Mol Nutr Food Res 2013; 57:962-73. [PMID: 23637065 DOI: 10.1002/mnfr.201200736] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 03/01/2013] [Accepted: 03/15/2013] [Indexed: 02/05/2023]
Abstract
SCOPE Metabolomics approach is focused on identifying all metabolites present in a biological sample (metabolome). Consumption of cocoa products has been related to health benefits including positive effect on cardiovascular health. METHODS AND RESULTS Twenty volunteers were included in this randomized, crossover, and controlled clinical trial. After a 2-wk washout period, subjects received 40 g/day of cocoa powder with 500 mL skimmed milk (cocoa with skimmed milk intervention) or 500 mL/day of skimmed milk (skimmed milk intervention) for 4-wk. Urine (24 h) samples were collected at baseline and after each intervention and were analyzed by HPLC-hybrid quadrupole TOF in negative and positive ionization modes followed by multivariate analysis. This analysis revealed a marked separation between the cocoa with skimmed milk intervention and skimmed milk intervention and baseline periods. Thirty-nine compounds linked with cocoa intake, including alkaloid metabolites, polyphenol host and gut microbial metabolites (hydroxyphenylvalerolactones and hydroxyphenylvaleric acids), diketopiperazines and N-phenylpropenoyl-l-amino acids were identified. In the case of endogenous metabolites, putative identifications suggested that metabolites linked with carnitine metabolism and sulfation of tyrosine were decreased by the consumption of cocoa. CONCLUSION LC-MS metabolomics strategy allows the defining of a complex metabolic profile derived from cocoa phytochemicals. Likewise, the identification of endogenous markers could lead to new hypotheses to unravel the relationship between cocoa intake and cardiovascular diseases.
Collapse
Affiliation(s)
- Rafael Llorach
- Department of Nutrition and Food Science, XaRTA, INSA, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
40
|
Steliou K, Boosalis MS, Perrine SP, Sangerman J, Faller DV. Butyrate histone deacetylase inhibitors. Biores Open Access 2013; 1:192-8. [PMID: 23514803 PMCID: PMC3559235 DOI: 10.1089/biores.2012.0223] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In addition to being a part of the metabolic fatty acid fuel cycle, butyrate is also capable of inducing growth arrest in a variety of normal cell types and senescence-like phenotypes in gynecological cancer cells, inhibiting DNA synthesis and cell growth in colonic tumor cell lines, suppressing hTERT mRNA expression and telomerase activity in human prostate cancer cells, and inducing stem cell differentiation and apoptosis by DNA fragmentation. It regulates gene expression by inhibiting histone deacetylases (HDACs), enhances memory recovery and formation in mice, stimulates neurogenesis in the ischemic brain, promotes osteoblast formation, selectively blocks cell replication in transformed cells (compared to healthy cells), and can prevent and treat diet-induced obesity and insulin resistance in mouse models of obesity, as well as stimulate fetal hemoglobin expression in individuals with hematologic diseases such as the thalassemias and sickle-cell disease, in addition to a multitude of other biochemical effects in vivo. However, efforts to exploit the potential of butyrate in the clinical treatment of cancer and other medical disorders are thwarted by its poor pharmacological properties (short half-life and first-pass hepatic clearance) and the multigram doses needed to achieve therapeutic concentrations in vivo. Herein, we review some of the methods used to overcome these difficulties with an emphasis on HDAC inhibition.
Collapse
Affiliation(s)
- Kosta Steliou
- PhenoMatriX, Inc. , Boston, Massachusetts. ; Cancer Research Center, Boston University School of Medicine , Boston, Massachusetts
| | | | | | | | | |
Collapse
|
41
|
Girardin M, Dionne S, Goyette P, Rioux J, Bitton A, Elimrani I, Charlebois P, Qureshi I, Levy E, Seidman EG. Expression and functional analysis of intestinal organic cation/L-carnitine transporter (OCTN) in Crohn's disease. J Crohns Colitis 2012; 6:189-97. [PMID: 22325173 DOI: 10.1016/j.crohns.2011.08.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 07/27/2011] [Accepted: 08/11/2011] [Indexed: 02/06/2023]
Abstract
BACKGROUND The IBD5 locus is a genetic risk factor for IBD, particularly Crohn's Disease, coding for the organic cation/carnitine transporters (OCTN1 and 2). Two variants of OCTN are associated with susceptibility to Crohn's Disease. Modified transport of carnitine in vitro has been reported for a polymorphism of OCTN1. The aim was to investigate the function of intestinal OCTNs in IBD in relation to genetic polymorphisms. METHODS Intestinal tissue was obtained from endoscopic biopsies and surgical resections from IBD patients (n=33 and 14, resp.) and controls (n=22 and 14, resp.). OCTN protein levels were measured in intestinal biopsies and carnitine transport was quantified in intestinal resections. RESULTS OCTN1 protein levels were significantly higher in ileal versus colonic tissue (2.95% ± 0.4 vs 0.66% ± 0.2, resp.; p<0.0002). OCTN1 expression was higher in Crohn's disease patients with mutant homozygous or heterozygous genotypes (0.6% ± 0.1 vs 3% ± 0.8, resp., p<0.02). Carnitine transport was very rapid and Na+ dependent (10s). It was not different comparing Crohn's Disease and control groups (0.45 ± 0.12 vs 0.51 ± 0.12 nM carnitine/mg prot/min, resp.). Carnitine transport tended to be higher in subjects with mutant homozygous and heterozygous OCTN1 and OCTN2 genotypes (0.19 vs 0.59 and 0.25 vs 0.6, respectively). CONCLUSIONS The present data reveal that OCTN protein levels appear to be similar in intestinal tissue from Crohn's Disease patients and controls. Overall, ileal carnitine transport appears to as well equal in Crohn's Disease and control groups. However, there was a trend towards higher carnitine transport in subjects with OCTN1 and OCTN2 mutations.
Collapse
Affiliation(s)
- Marc Girardin
- DigestiveLab, Research Institute, McGill University Health Centre, Montreal, QC, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Xia Y, Li Q, Zhong W, Dong J, Wang Z, Wang C. L-carnitine ameliorated fatty liver in high-calorie diet/STZ-induced type 2 diabetic mice by improving mitochondrial function. Diabetol Metab Syndr 2011; 3:31. [PMID: 22082204 PMCID: PMC3226540 DOI: 10.1186/1758-5996-3-31] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 11/15/2011] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND There are an increasing number of patients suffering from fatty liver caused by type 2 diabetes. We intended to study the preventive and therapeutic effect of L-carnitine (LC) on nonalcoholic fatty liver disease (NAFLD) in streptozotocin (STZ)-induced type 2 diabetic mice and to explore its possible mechanism. METHODS Thirty male Kungming mice were randomly divided into five groups: control group, diabetic group, pre-treatment group (125 mg/kg BW), low-dose (125 mg/kg BW) therapeutic group and high-dose (250 mg/kg BW) therapeutic group. The morphology of hepatocytes was observed by light and electron microscopy. LC and ALC (acetyl L-carnitine) concentrations in the liver were determined by high-performance liquid chromatography (HPLC). Moreover, liver weight, insulin levels and free fatty acid (FFA) and triglyceride (TG) levels in the liver and plasma were measured. RESULTS Average liver LC and ALC levels were 33.7% and 20% lower, respectively, in diabetic mice compared to control mice (P < 0.05). After preventive and therapeutic treatment with LC, less hepatocyte steatosis, clearer crista and fewer glycogen granules in the mitochondria were observed. Decreased liver weight, TG levels, and FFA concentrations (P < 0.05) in the liver were also observed after treatment with LC in diabetic mice. Moreover, liver LC and ALC levels increased upon treatment with LC, whereas the ratio of LC and ALC decreased significantly (P < 0.01). CONCLUSION LC supplements ameliorated fatty liver in type 2 diabetic mice by increasing fatty acid oxidation and decreasing the LC/ALC ratio in the liver. Therefore, oral administration of LC protected mitochondrial function in liver.
Collapse
Affiliation(s)
- Yunqiu Xia
- Center for Human Functional Experiment, Medical College, Qingdao University, Ningxia Road, Qingdao, China
| | - Qing Li
- Class 5, Grade 2007, Medical College, Qingdao University, Ningxia Road, Qingdao, China
| | - Weizhen Zhong
- Center for Human Functional Experiment, Medical College, Qingdao University, Ningxia Road, Qingdao, China
| | - Jing Dong
- Physiology Department of the Medical College, Qingdao University, Ningxia Road, Qingdao, China
| | - Zhulin Wang
- Class 5, Grade 2007, Medical College, Qingdao University, Ningxia Road, Qingdao, China
| | - Chunbo Wang
- Center for Human Functional Experiment, Medical College, Qingdao University, Ningxia Road, Qingdao, China
| |
Collapse
|
43
|
Diao L, Polli JE. Synthesis and in vitro characterization of drug conjugates of l-carnitine as potential prodrugs that target human Octn2. J Pharm Sci 2011; 100:3802-16. [DOI: 10.1002/jps.22557] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 01/21/2011] [Accepted: 03/02/2011] [Indexed: 01/11/2023]
|
44
|
Flanagan JL, Simmons PA, Vehige J, Willcox MD, Garrett Q. Role of carnitine in disease. Nutr Metab (Lond) 2010; 7:30. [PMID: 20398344 PMCID: PMC2861661 DOI: 10.1186/1743-7075-7-30] [Citation(s) in RCA: 388] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Accepted: 04/16/2010] [Indexed: 02/06/2023] Open
Abstract
Carnitine is a conditionally essential nutrient that plays a vital role in energy production and fatty acid metabolism. Vegetarians possess a greater bioavailability than meat eaters. Distinct deficiencies arise either from genetic mutation of carnitine transporters or in association with other disorders such as liver or kidney disease. Carnitine deficiency occurs in aberrations of carnitine regulation in disorders such as diabetes, sepsis, cardiomyopathy, malnutrition, cirrhosis, endocrine disorders and with aging. Nutritional supplementation of L-carnitine, the biologically active form of carnitine, is ameliorative for uremic patients, and can improve nerve conduction, neuropathic pain and immune function in diabetes patients while it is life-saving for patients suffering primary carnitine deficiency. Clinical application of carnitine holds much promise in a range of neural disorders such as Alzheimer's disease, hepatic encephalopathy and other painful neuropathies. Topical application in dry eye offers osmoprotection and modulates immune and inflammatory responses. Carnitine has been recognized as a nutritional supplement in cardiovascular disease and there is increasing evidence that carnitine supplementation may be beneficial in treating obesity, improving glucose intolerance and total energy expenditure.
Collapse
|
45
|
Talián G, Lakner L, Bene J, Komlósi K, Horváth K, Gasztonyi B, Miheller P, Figler M, Mózsik G, Tulassay Z, Melegh B. Plasma carnitine ester profiles in Crohn's disease and ulcerative colitis patients with different IGR2230a_1 genotypes. Int J Immunogenet 2009; 36:329-335. [PMID: 19735486 DOI: 10.1111/j.1744-313x.2009.00834.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
An association has been repeatedly demonstrated between inflammatory bowel disease (IBD) and the IBD5 locus in the 5q31 chromosomal region. The aim of the present study was to examine the prevalence of the IGR2230a_1 intronic nucleotide polymorphism of the slc22a5 gene (coding for the OCTN2 carnitine transporter protein) lying within this region, and its possible relationship with the carnitine metabolism in Hungarian IBD patients and controls. We genotyped by restriction fragment length polymorphism 200 Crohn's disease (CD) and 246 ulcerative colitis (UC) patients, as well as 187 healthy controls. From plasma samples we determined detailed carnitine ester profiles of 76 CD, 43 UC patients and 45 control persons using electrospray ionization triple quadruple tandem mass spectrometry. The distribution of the genotypes was not significantly different in the CD or the UC group compared with the controls. We found no significant alterations of the carnitine profile in the carrier/non-carrier or the homozygote/non-homozygote comparisons in both the CD and the UC groups, stratified by IGR2230a_1 genotype. Our data suggest that this polymorphism alone is not associated with CD and UC in the Hungarian population, and has no effect on the carnitine metabolism.
Collapse
Affiliation(s)
- G Talián
- Department of Medical Genetics and Child Development, University of Pécs, H-7624 Pécs, Hungary
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Van Immerseel F, Ducatelle R, De Vos M, Boon N, Van De Wiele T, Verbeke K, Rutgeerts P, Sas B, Louis P, Flint HJ. Butyric acid-producing anaerobic bacteria as a novel probiotic treatment approach for inflammatory bowel disease. J Med Microbiol 2009; 59:141-143. [PMID: 19942690 DOI: 10.1099/jmm.0.017541-0] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Filip Van Immerseel
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Richard Ducatelle
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Martine De Vos
- Department of Gastroenterology, Ghent University Hospital, Ghent, Belgium
| | - Nico Boon
- Laboratory of Microbial Ecology and Technology (LabMET), Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Tom Van De Wiele
- Laboratory of Microbial Ecology and Technology (LabMET), Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Kristin Verbeke
- Department of Gastroenterology and Leuven Food Science and Nutrition Research Centre (LFoRCe), University Hospital, Leuven, Belgium
| | - Paul Rutgeerts
- Department of Gastroenterology and Leuven Food Science and Nutrition Research Centre (LFoRCe), University Hospital, Leuven, Belgium
| | - Benedikt Sas
- Centre of Excellence Food2Know, Ghent University, Ghent, Belgium
| | - Petra Louis
- Microbial Ecology Group, Gut Health Programme, Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, UK
| | - Harry J Flint
- Microbial Ecology Group, Gut Health Programme, Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
47
|
Gong XW, Li JP, Wu JF, Xun L, Zhai HM, Feng B, Xu WF, Tang W. Synthesis of Carnitine Benzyl Esters as Prodrugs. JOURNAL OF CHEMICAL RESEARCH 2008. [DOI: 10.3184/030823408x324689] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The introduction of C-2 acyl groups and a benzyl ester onto L-carnitine generated a series of carnitine benzyl esters as prodrug with improved liposolubility, stability and bioavailability.
Collapse
Affiliation(s)
- Xiao-wei Gong
- College of Pharmacy, Shandong University, Ji'nan 250012, P.R. China
| | - Jin-pei Li
- Liaocheng People's Hospital, 252000, P.R. China
| | - Ji-feng Wu
- Ji'nan Public Security, Shandong, 250000, P.R. China
| | - Li Xun
- College of Pharmacy, Shandong University, Ji'nan 250012, P.R. China
| | - Hai-min Zhai
- Qidu Pharmaceutical Co. LTD, Zibo, 255400, P.R. China
| | - Bo Feng
- Qidu Pharmaceutical Co. LTD, Zibo, 255400, P.R. China
| | - Wen-fang Xu
- College of Pharmacy, Shandong University, Ji'nan 250012, P.R. China
| | - Wei Tang
- College of Pharmacy, Shandong University, Ji'nan 250012, P.R. China
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, the University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
48
|
Oz HS, Ebersole JL. Application of prodrugs to inflammatory diseases of the gut. Molecules 2008; 13:452-74. [PMID: 18305431 PMCID: PMC6244946 DOI: 10.3390/molecules13020452] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2008] [Revised: 02/20/2008] [Accepted: 02/21/2008] [Indexed: 12/11/2022] Open
Abstract
Oral delivery is the most common and preferred route of drug administration although the digestive tract exhibits several obstacles to drug delivery including motility and intraluminal pH profiles. The gut milieu represents the largest mucosal surface exposed to microorganisms with 1010-12 colony forming bacteria/g of colonic content. Approximately, one third of fecal dry matter is made of bacteria/ bacterial components. Indeed, the normal gut microbiota is responsible for healthy digestion of dietary fibers (polysaccharides) and fermentation of short chain fatty acids such as acetate and butyrate that provide carbon sources (fuel) for these bacteria. Inflammatory bowel disease (IBD) results in breakage of the mucosal barrier, an altered microbiota and dysregulated gut immunity. Prodrugs that are chemically constructed to target colonic release or are degraded specifically by colonic bacteria, can be useful in the treatment of IBD. This review describes the progress in digestive tract prodrug design and delivery in light of gut metabolic activities.
Collapse
Affiliation(s)
- Helieh S Oz
- Center for Oral Health Research, College of Dentistry and Department of Internal Medicine, University of Kentucky, Medical Center, 800 Rose Street, Lexington, KY 40536, USA.
| | | |
Collapse
|