1
|
Foguet C, Jiang X, Ritchie SC, Persyn E, Xu Y, Ben-Eghan C, Taylor HJ, Di Angelantonio E, Danesh J, Butterworth AS, Lambert SA, Inouye M. Metabolic reaction fluxes as amplifiers and buffers of risk alleles for coronary artery disease. Mol Syst Biol 2025:10.1038/s44320-025-00097-2. [PMID: 40175777 DOI: 10.1038/s44320-025-00097-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 04/04/2025] Open
Abstract
Genome-wide association studies have identified thousands of variants associated with disease risk but the mechanism by which such variants contribute to disease remains largely unknown. Indeed, a major challenge is that variants do not act in isolation but rather in the framework of highly complex biological networks, such as the human metabolic network, which can amplify or buffer the effect of specific risk alleles on disease susceptibility. Here we use genetically predicted reaction fluxes to perform a systematic search for metabolic fluxes acting as buffers or amplifiers of coronary artery disease (CAD) risk alleles. Our analysis identifies 30 risk locus-reaction flux pairs with significant interaction on CAD susceptibility involving 18 individual reaction fluxes and 8 independent risk loci. Notably, many of these reactions are linked to processes with putative roles in the disease such as the metabolism of inflammatory mediators. In summary, this work establishes proof of concept that biochemical reaction fluxes can have non-additive effects with risk alleles and provides novel insights into the interplay between metabolism and genetic variation on disease susceptibility.
Collapse
Affiliation(s)
- Carles Foguet
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
| | - Xilin Jiang
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Scott C Ritchie
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Elodie Persyn
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Yu Xu
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Chief Ben-Eghan
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Henry J Taylor
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Emanuele Di Angelantonio
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Health Data Science Research Centre, Fondazione Human Technopole, Milan, Italy
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Department of Human Genetics, the Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
| | - Samuel A Lambert
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK.
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK.
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
| |
Collapse
|
2
|
Thuy VT, Viet NL, Nghia NT, Cangelosi G, Petrelli F, Nguyen CTT. The impact of the solute carrier gene superfamily polymorphisms on tyrosine kinase inhibitors responses among chronic myeloid leukemia: A meta-analysis. Leuk Res 2025; 151:107673. [PMID: 40048815 DOI: 10.1016/j.leukres.2025.107673] [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: 01/06/2025] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 03/24/2025]
Abstract
BACKGROUND Tyrosine kinase inhibitors (TKIs) are currently the first-line therapy for chronic myeloid leukemia (CML), but proportion of treatment responses may be influenced by genetic polymorphisms, especially, the solute carrier gene superfamily (SLC). This study was conducted to evaluate the relationship of polymorphisms in the SLC genes family and treatment responses to TKIs among CML patients. METHODS A systematic search was conducted from four databases, including PubMed, Cochrane Library, Embase and Web of Science, up to March 2023. The relationship between SLC polymorphisms and TKI efficacy was assessed by pooled odds ratios (ORs) of the complete cytogenetic response (CCyR) and major molecular biological response (MMR) with 95 % confidence intervals (95 %CIs) across five genetic models (dominant, recessive, homozygote, heterozygote, and allele). Meta-analyses, heterogeneity between studies, publication bias, sensitivity, meta-regression and subgroup analysis were all performed. RESULTS A total of 19/983 studies meeting the criteria were included in the meta-analysis, with eight variants belonging to three genes (SLC22A1, SLCO1B3, and SLC22A4). The results showed that there was a statistically significant association between the SLC22A1 rs683369 variant and a lower rate of achieving MMR in all 05 genetic models. Similar results were also recorded in the dominant and homozygote models of the SLC22A1 rs628031 variant (OR= 0.61 (95 %CI= 0.46-0.82); OR= 0.46 (95 %CI= 0.23-0.94), respectively); particularly in Asian patients. No relationship was identified between MMR and other genes, as well as that of CCyR and all variants. CONCLUSION SLC variants can be predictive signals of imatinib responses among CML; Asian patients should be paid attention during the treatment.
Collapse
Affiliation(s)
- Vu Thi Thuy
- Falculty of Pharmacy, Ho Chi Minh City University of Technology, Ho Chi Minh City 70000, Vietnam
| | - Nguyen Linh Viet
- Falculty of Pharmacy, Ho Chi Minh City University of Technology, Ho Chi Minh City 70000, Vietnam
| | - Nguyen Trong Nghia
- Falculty of Pharmacy, Ho Chi Minh City University of Technology, Ho Chi Minh City 70000, Vietnam
| | | | - Fabio Petrelli
- School of Pharmacy, Polo Medicina Sperimentale e Sanità Pubblica, Camerino 62032, Italy
| | - Cuc Thi Thu Nguyen
- Falculty of Pharmaceutical Management and Economics, Hanoi University of Pharmacy, Hanoi 100000, Vietnam.
| |
Collapse
|
3
|
Rönnpagel V, Morof F, Römer S, Meyer-Tönnies MJ, Tzvetkov MV. Substrate-specific inhibition of organic cation transporter 1 revealed using a multisubstrate drug cocktail. Drug Metab Dispos 2025; 53:100074. [PMID: 40319555 DOI: 10.1016/j.dmd.2025.100074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/07/2025] [Accepted: 03/23/2025] [Indexed: 05/07/2025] Open
Abstract
Transporters of the SLC22 family, such as organic cation transporter 1 (OCT1), possess very broad substrate specificity. It is unclear to what extent the inhibitory potencies of OCT1 depend on the substrate used. Here, we describe a multisubstrate drug cocktail that allows for the simultaneous testing of drug-drug interactions using 8 different victim drugs: fenoterol, salbutamol, sumatriptan, zolmitriptan, ipratropium, trospium, methylnaltrexone, and metformin. There were no significant differences in Michaelis constant (KM) and vmax of the OCT1-mediated uptake of the substrates alone or in the cocktail. Depending on the victim drug analyzed, we observed 6.7-fold differences in the inhibitory potency of fenoterol (IC50 of 0.75 μM for metformin and 5.1 μM for sumatriptan). Similarly, the inhibitory potency of verapamil varied 6.7-fold (IC50 of 1.3 μM for zolmitriptan and 8.7 μM for ipratropium). Two groups of inhibitors showed strong correlations in their victim-dependent inhibitory potencies. Group 1 comprised verapamil, quinidine, fenoterol, and ipratropium, and group 2 comprised metformin, sumatriptan, and trimethoprim. By comparing OCT1 paralogs and orthologs, the broadest substrate spectra were observed for OCT1 and multidrug and toxin extrusion 1, followed by OCT2, multidrug and toxin extrusion 2-K, and OCT3. In contrast, organic cation transporters novel 1 and organic cation transporters novel 2 exhibited very narrow substrate specificity, transporting only L-carnitine and L-ergothioneine, respectively. In conclusion, OCT1 demonstrates substantial differences in inhibitory potencies, depending on the victim drug used. We developed a cocktail approach that enables rapid screening for such differences, facilitating the identification of drug-drug interactions at the early stages of drug development. This approach can be extended to other transporters with broad substrate specificity. SIGNIFICANCE STATEMENT: Polyspecific transporters have a broad substrate-binding cavity with no defined single binding position. Consequently, inhibitors may exhibit different inhibitory potencies depending on the victim drug used for testing. Here, we demonstrate this for organic cation transporter 1 (OCT1, SLC22A1) and presents a drug cocktail designed to identify varying inhibitory potencies in vitro and prevent false-negative drug-drug interaction results during early drug development. This approach can be extended to other polyspecific drug transporters.
Collapse
Affiliation(s)
- Vincent Rönnpagel
- Department of General Pharmacology, Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany
| | - Felix Morof
- Department of General Pharmacology, Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany
| | - Sarah Römer
- Department of General Pharmacology, Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany
| | - Marleen J Meyer-Tönnies
- Department of General Pharmacology, Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany
| | - Mladen V Tzvetkov
- Department of General Pharmacology, Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
4
|
Zhang C, Yang X, Xue Y, Li H, Zeng C, Chen M. The Role of Solute Carrier Family Transporters in Hepatic Steatosis and Hepatic Fibrosis. J Clin Transl Hepatol 2025; 13:233-252. [PMID: 40078199 PMCID: PMC11894391 DOI: 10.14218/jcth.2024.00348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 03/14/2025] Open
Abstract
Solute carrier (SLC) family transporters are crucial transmembrane proteins responsible for transporting various molecules, including amino acids, electrolytes, fatty acids, and nucleotides. To date, more than fifty SLC transporter subfamilies have been identified, many of which are linked to the progression of hepatic steatosis and fibrosis. These conditions are often caused by factors such as non-alcoholic fatty liver disease and non-alcoholic steatohepatitis, which are major contributors to the global liver disease burden. The activity of SLC members regulates the transport of substrates across biological membranes, playing key roles in lipid synthesis and metabolism, mitochondrial function, and ferroptosis. These processes, in turn, influence the function of hepatocytes, hepatic stellate cells, and macrophages, thereby contributing to the development of hepatic steatosis and fibrosis. Additionally, some SLC transporters are involved in drug transport, acting as critical regulators of drug-induced hepatic steatosis. Beyond substrate transport, certain SLC members also exhibit additional functions. Given the pivotal role of the SLC family in hepatic steatosis and fibrosis, this review aimed to summarize the molecular mechanisms through which SLC transporters influence these conditions.
Collapse
Affiliation(s)
| | | | - Yi Xue
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Huan Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chuanfei Zeng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Mingkai Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
5
|
Fan J, Xie W, Ke H, Zhang J, Wang J, Wang H, Guo N, Bai Y, Lei X. Structural Basis for Inhibition of Urate Reabsorption in URAT1. JACS AU 2025; 5:1308-1319. [PMID: 40151250 PMCID: PMC11937972 DOI: 10.1021/jacsau.4c01188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/29/2025]
Abstract
The urate transporter 1 (URAT1) is the primary urate transporter in the kidney responsible for urate reabsorption and, therefore, is crucial for urate homeostasis. Hyperuricemia causes the common human disease gout and other pathological consequences. Inhibition of urate reabsorption through URAT1 has been shown as a promising strategy in alleviating hyperuricemia, and clinical and preclinical drug candidates targeting URAT1 are emerging. However, how small molecules inhibit URAT1 remains undefined, and the lack of accurate URAT1 complex structures hinders the development of better therapeutics. Here, we present cryoelectron microscopy structures of a humanized rat URAT1 bound with benzbromarone, lingdolinurad, and verinurad, elucidating the structural basis for drug recognition and inhibition. The three small molecules reside in the URAT1 central cavity with different binding modes, locking URAT1 in an inward-facing conformation. This study provides mechanistic insights into the drug modulation of URAT1 and sheds light on the rational design of potential URAT1-specific therapeutics for treating hyperuricemia.
Collapse
Affiliation(s)
- Junping Fan
- Beijing National
Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry
and Molecular Engineering of Ministry of Education, Institute of Organic
Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Wenjun Xie
- Beijing National
Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry
and Molecular Engineering of Ministry of Education, Institute of Organic
Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua
Center for Life Sciences, Peking University, Beijing 100871, China
| | - Han Ke
- Beijing National
Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry
and Molecular Engineering of Ministry of Education, Institute of Organic
Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jing Zhang
- Jiangsu
JITRI Molecular Engineering Inst. Co., Ltd., Jiangsu 215500, China
| | - Jin Wang
- Beijing National
Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry
and Molecular Engineering of Ministry of Education, Institute of Organic
Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Haijun Wang
- Beijing National
Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry
and Molecular Engineering of Ministry of Education, Institute of Organic
Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Nianxin Guo
- Beijing National
Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry
and Molecular Engineering of Ministry of Education, Institute of Organic
Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yingjie Bai
- Institute
for Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Xiaoguang Lei
- Beijing National
Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry
and Molecular Engineering of Ministry of Education, Institute of Organic
Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua
Center for Life Sciences, Peking University, Beijing 100871, China
- Institute
for Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518107, China
| |
Collapse
|
6
|
Martinez SA, Karel IZ, Silvaroli JA, Ahmed E, Kim JY, Stayton A, Patel PS, Afjal MA, Horton T, Bohmer M, Vanichapol T, Sander V, Andrade GM, Allison CV, Mondal M, Thorson VC, Partey A, Nimkar K, Williams V, Quimby J, Ganesan L, Madhavan SM, Davidson AJ, Peterson BR, Adebiyi A, Rao R, Sweet DH, Singh P, Bennett KM, Zepeda-Orozco D, Mallipattu SK, Eisenmann ED, Sparreboom A, Rovin BH, Bajwa A, Pabla NS. Resazurin dye is an in vivo sensor of kidney tubular function. Kidney Int 2025; 107:508-520. [PMID: 39733791 PMCID: PMC11845305 DOI: 10.1016/j.kint.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 11/01/2024] [Accepted: 12/16/2024] [Indexed: 12/31/2024]
Abstract
Glomerular filtration rate (GFR) is the main functional index of kidney health and disease. Currently, no methods are available to directly measure tubular mass and function. Here, we report a serendipitous finding that the in vitro cell viability dye resazurin can be used in mice as an exogenous sensor of tubular function. Intravenously injected resazurin exhibited significant plasma protein binding and was found to mainly undergo tubular secretion. Mechanistic studies showed that the blue-colored, weakly fluorescent resazurin is taken up by tubular cells through organic anion transporters, followed by conversion to a highly fluorescent, pink-colored resorufin by mitochondrial and cytosolic reductases, converted to an orange-colored β-d-glucuronide with subsequent efflux into the urine. Here we report a simple method in which the intravenous injection of resazurin is followed by the measurement of fluorescent metabolites in the urine, providing a sensitive readout of tubular function. Three mouse models of acute kidney injury (rhabdomyolysis, bilateral ischemia-reperfusion injury, and cisplatin nephrotoxicity) were tested and the resazurin-based method was able to sensitively detect the loss of tubular function much earlier than the increase in serum creatinine levels. Strikingly, in mice with unilateral ischemia-reperfusion injury and genetic mutation-linked kidney hypoplasia (oligosyndactylism, a genetic model for congenital kidney hypoplasia), the resazurin-based method was able to detect loss of tubular mass and function despite normal GFR levels. Collectively, our findings establish the preclinical utility of resazurin as a sensitive exogenous marker of tubular function and support future examination in larger animals for potential clinical translation.
Collapse
Affiliation(s)
- Shirely Acosta Martinez
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Isaac Z Karel
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Josie A Silvaroli
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Eman Ahmed
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Ji Young Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Amanda Stayton
- Transplant Research Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Prisha S Patel
- Transplant Research Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Mohammad Amir Afjal
- Transplant Research Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Thomas Horton
- Transplant Research Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Margaret Bohmer
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Thitinee Vanichapol
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Veronika Sander
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Gabriel Mayoral Andrade
- Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Corynne Vermillion Allison
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Milon Mondal
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Victoria C Thorson
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Alexandra Partey
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Kartik Nimkar
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Victoria Williams
- Division of Clinical Pharmacy Practice and Science, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Jessica Quimby
- Department of Veterinary Clinical Sciences, The Ohio State University Veterinary Medical Center, Columbus, Ohio, USA
| | - Latha Ganesan
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Sethu M Madhavan
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Alan J Davidson
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Blake R Peterson
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Adebowale Adebiyi
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - Reena Rao
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA; Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Douglas H Sweet
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Prabhleen Singh
- Division of Nephrology and Hypertension, University of California San Diego, San Diego, California, USA
| | - Kevin M Bennett
- Washington University in St. Louis, Mallinckrodt Institute of Radiology, St. Louis, Missouri, USA
| | - Diana Zepeda-Orozco
- Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Sandeep K Mallipattu
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, New York, USA; Renal Section, Northport VA Medical Center, Northport, New York, USA
| | - Eric D Eisenmann
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Brad H Rovin
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Amandeep Bajwa
- Transplant Research Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Genetics, Genomics, and Informatics, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Microbiology, Immunology, and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA.
| | - Navjot S Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
7
|
Zuo Y, Li T, Yang S, Chen X, Tao X, Dong D, Liu F, Zhu Y. Contribution and expression of renal drug transporters in renal cell carcinoma. Front Pharmacol 2025; 15:1466877. [PMID: 40034145 PMCID: PMC11873565 DOI: 10.3389/fphar.2024.1466877] [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: 07/18/2024] [Accepted: 12/23/2024] [Indexed: 03/05/2025] Open
Abstract
Renal cell carcinoma (RCC) is a common substantive tumor. According to incomplete statistics, RCC incidence accounts for approximately 90% of renal malignant tumors, and is the second most prevalent major malignant tumor in the genitourinary system, following bladder cancer. Only 10%-15% of chemotherapy regimens for metastatic renal cell carcinoma (mRCC) are effective, and mRCC has a high mortality. Drug transporters are proteins located on the cell membrane that are responsible for the absorption, distribution, and excretion of drugs. Lots of drug transporters are expressed in the kidneys. Changes in carrier function weaken balance, cause disease, or modify the effectiveness of drug treatment. The changes in expression of these transporters during cancer pathology results in multi-drug resistance to cancer chemotherapy. In the treatment of RCC, the study of drug transporters helps to optimize treatment regimens, improve therapeutic effects, and reduce drug side effects. In this review, we summarize advances in the role of renal drug transporters in the genesis, progression, and treatment of RCC.
Collapse
Affiliation(s)
- Yawen Zuo
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tong Li
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shilei Yang
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xuyang Chen
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xufeng Tao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Deshi Dong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fang Liu
- Department of Medical Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yanna Zhu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
8
|
Wu X, Luo Y, Feng S, Ma H, Ke B, Wang K, Su Z, Yang D. Structures and membrane interactions of human OAT1 in complex with clinical used drugs. SCIENCE ADVANCES 2025; 11:eads5405. [PMID: 39951534 PMCID: PMC11827633 DOI: 10.1126/sciadv.ads5405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025]
Abstract
Organic anion transporters (OATs) in mammals mediate the renal excretion of numerous structurally diverse organic anionic compounds. Therapeutically inhibiting OATs has emerged as a strategy to modulate the elimination or retention of these substrates. Among them, OAT1 plays a pivotal role in the pharmacokinetics and drug-drug interactions of a wide range of prescription medications. Despite extensive structural investigations, the molecular structure, and basis of polyspecific anionic drug recognition of human OAT1 (hOAT1) have remained elusive. Here, we present cryogenic electron microscopy structures of hOAT1 and its complexes with the antiviral drug cidofovir and an FDA-approved type II diabetes medication glibenclamide, respectively. Our findings reveal that both cidofovir and glibenclamide bind to a central binding site, capturing the transporter in inward-facing conformations. These structures elucidate how specific residues within the central site orchestrate the binding of chemically diverse inhibitors and provide a structural basis for the drug recognition mechanism of hOAT1.
Collapse
Affiliation(s)
- Xuening Wu
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan 610044, China
| | - Yongbo Luo
- The State Key Laboratory of Biotherapy, Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610044, China
| | - Shijian Feng
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan 610044, China
| | - Haiyun Ma
- The State Key Laboratory of Biotherapy, Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610044, China
| | - Bowen Ke
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Kunjie Wang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan 610044, China
| | - Zhaoming Su
- The State Key Laboratory of Biotherapy, Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610044, China
| | - Dongxue Yang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan 610044, China
| |
Collapse
|
9
|
Tian L, Qiang T, Xia J, Zhang B, Lu Q, Liu Y, Hu J, Kang K, Li J, Zhang J, Yang X, Wang Y, Zhang D, Gao H, Liang C. Kidney Targeting Smart Antibiotic Discovery: Multimechanism Pleuromutilins for Pyelonephritis Therapy. J Med Chem 2025; 68:3335-3355. [PMID: 39813601 DOI: 10.1021/acs.jmedchem.4c02557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Multidrug-resistant (MDR) bacteria pose a global health threat, underscoring the need for new antibiotics. Lefamulin, the first novel-mechanism antibiotic approved by the FDA in decades, showcases pleuromutilins' promise due to low mutation frequency. However, their clinical use is limited by poor pharmacokinetics and organ toxicity. To overcome these limitations, we modified lefamulin's C14 side chain via quaternization and incorporated rigid molecular fragments to enhance pharmacological properties. Introducing a quaternary ammonium group improved liver and kidney targeting via organic cation transporters (OCTs). Candidate 8i, a quaternized imidazo[4,5-c]pyridine pleuromutilin, demonstrated broad-spectrum activity against MDR bacteria, Mycoplasma and Chlamydophila at low doses. 8i targeted transport to infected kidneys, disrupted biofilms, damaged membranes, and inhibited protein synthesis by targeting 50S ribosomal subunit. It cleared rapidly, reducing long-term toxicity. Daily injections were an effective short-course treatment for systemic infections and pyelonephritis. This research presents a novel OCT-mediated, organ-targeted antibiotic design strategy to manage antibiotic-resistant infections.
Collapse
Affiliation(s)
- Lei Tian
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi'an 710021, China
| | - Taotao Qiang
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Juan Xia
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang 524045, China
| | - Boxin Zhang
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi'an 710021, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Qi Lu
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi'an 710021, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Yuting Liu
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi'an 710021, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Jinrong Hu
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi'an 710021, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Kairui Kang
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi'an 710021, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Jialin Li
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi'an 710021, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Jiayun Zhang
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi'an 710021, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Xiuding Yang
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi'an 710021, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Yongbo Wang
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi'an 710021, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Dezhu Zhang
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi'an 710021, China
- Shaanxi Panlong Pharmaceutical Group Co., Ltd., Xi'an 710025, China
| | - Hong Gao
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi'an 710021, China
- Shaanxi Pioneer Biotech Co., Ltd., Xi'an 710021, China
| | - Chengyuan Liang
- Xi'an Key Laboratory for Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi'an 710021, China
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi'an 710021, China
| |
Collapse
|
10
|
Du J, Shen M, Chen J, Yan H, Xu Z, Yang X, Yang B, Luo P, Ding K, Hu Y, He Q. The impact of solute carrier proteins on disrupting substance regulation in metabolic disorders: insights and clinical applications. Front Pharmacol 2025; 15:1510080. [PMID: 39850557 PMCID: PMC11754210 DOI: 10.3389/fphar.2024.1510080] [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: 10/12/2024] [Accepted: 12/20/2024] [Indexed: 01/25/2025] Open
Abstract
Carbohydrates, lipids, bile acids, various inorganic salt ions and organic acids are the main nutrients or indispensable components of the human body. Dysregulation in the processes of absorption, transport, metabolism, and excretion of these metabolites can lead to the onset of severe metabolic disorders, such as type 2 diabetes, non-alcoholic fatty liver disease, gout and hyperbilirubinemia. As the second largest membrane receptor supergroup, several major families in the solute carrier (SLC) supergroup have been found to play key roles in the transport of substances such as carbohydrates, lipids, urate, bile acids, monocarboxylates and zinc ions. Based on common metabolic dysregulation and related metabolic substances, we explored the relationship between several major families of SLC supergroup and metabolic diseases, providing examples of drugs targeting SLC proteins that have been approved or are currently in clinical/preclinical research as well as SLC-related diagnostic techniques that are in clinical use or under investigation. By highlighting these connections, we aim to provide insights that may contribute to the development of improved treatment strategies and targeted therapies for metabolic disorders.
Collapse
Affiliation(s)
- Jiangxia Du
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Minhui Shen
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiajia Chen
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bo Yang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Pharmaceutical and Translational Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| | - Kefeng Ding
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuhuai Hu
- Yuhong Pharmaceutical Technology Co., Ltd., Hangzhou, Zhejiang, China
| | - Qiaojun He
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
- Department of Pharmaceutical and Translational Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
11
|
Zhang J, Chen Y, Luo G, Luo Y. Molecular mechanism of geniposide against ANIT-induced intrahepatic cholestasis by integrative analysis of transcriptomics and metabolomics. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:765-779. [PMID: 39052058 DOI: 10.1007/s00210-024-03320-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
Geniposide (GE), a bioactive compound extracted from the fruit of Gardenia jasminoides Ellis, has attracted significant attention for its hepatoprotective therapeutic applications. Although GE displays a protective effect on treating intrahepatic cholestasis (IC), the underlying mechanism remains elusive. In this study, we aimed to elucidate the pharmacological mechanisms of GE in treating IC by an integrated analysis of transcriptomics and metabolomics. Firstly, we evaluated the hepatoprotective effect of GE in α-naphthylisothiocyanate (ANIT)-induced IC rats by examining biochemical indices, inflammatory factors, and oxidative stress levels. Secondly, by transcriptomics and serum metabolomics, we identified differentially expressed genes and metabolites, revealing phenotype-related metabolic pathways and gene functions. Lastly, we screened the core targets of GE in the treatment of IC by integrating transcriptomic and metabolomic data and validated these targets using western blotting. The results indicated that GE improved serum indexes and alleviated inflammation reactions and oxidative stress in the liver. The transcriptomics analysis revealed 739 differentially expressed genes after GE treatment, mainly enriched in retinol metabolism, steroid hormone synthesis, PPAR signal transduction, bile secretion metabolism, and other pathways. The metabolomics analysis identified 98 differential metabolites and 10 metabolic pathways. By constructing a "genes-targets-pathways-compounds" network, we identified two pathways: the bile secretion pathway and the glutathione pathway. Within these pathways, we discovered nine crucial targets that were subsequently validated through western blotting. The results revealed that the GE group significantly increased the expression of ABCG5, NCEH1, OAT3, and GST, compared with the ANIT group. We speculate that GE has a therapeutic effect on IC by modulating the bile secretion pathway and the glutathione pathway and regulating the expression of ABCG5, NCEH1, OAT3, and GST.
Collapse
Affiliation(s)
- Junyi Zhang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Yunting Chen
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Guangming Luo
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China.
| | - Yangjing Luo
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China.
| |
Collapse
|
12
|
Pan Y, Yang Z, Wei M, Gan Y, Liu M, Zou W. Hypertensive Nephropathy Changes the Expression of Drug-Metabolizing Enzymes and Transporters in Spontaneously Hypertensive Rat Liver and Kidney. Eur J Drug Metab Pharmacokinet 2025; 50:39-51. [PMID: 39522097 DOI: 10.1007/s13318-024-00923-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND OBJECTIVES Hypertensive nephropathy (HN) has become one of the main causes of end-stage renal disease. Drug combination therapy is a common clinical treatment for HN. However, the impact of HN on drug-metabolizing enzymes and transporters, which may lead to drug-drug interactions (DDIs) and even trigger toxic side effects, remains unclear. The aim of this study was to investigate changes in major drug-metabolizing enzymes and transporters in the liver and kidney of HN rats to improve the scientific foundations for the clinical treatment of HN. METHODS Spontaneously hypertensive rats (SHRs) were used as an animal HN model because their hypertension is similar to that of humans. Wistar-Kyoto rats (WKYs) were used as the control group. Body weight, blood pressure, hematoxylin-eosin (HE) staining and biochemical analysis were performed to evaluate whether the HN model was successfully constructed. Quantitative real-time polymerase chain reaction (PCR) and western blotting were used to evaluate the mRNA and protein expression of drug-metabolizing enzymes, transporters and related nuclear transcription factors. RESULTS In HN rats, the mRNA expression of the drug-metabolizing enzymes cytochrome P450 (Cyp) 2b1, Cyp2c11, Cyp3a1 and Cyp7a1 was significantly upregulated. The protein level of CYP3A1 was consistent with its mRNA expression. Interestingly, the mRNA expression of the hepatic transporters organic cation transporter (Oct) 1, Oct2, organic anion transporter (Oat) 1, Oat2, multidrug resistant protein (Mrp) 2, multidrug resistance (Mdr) 1, organic anion transporting polypeptide (Oatp) 1b2 and na+/taurocholate cotransporting polypeptide (Ntcp) was also markedly upregulated. This may be directly influenced by the upregulation of the expression of the nuclear receptors farnesoid X receptor (Fxr), pregnane X receptor (Pxr), liver X-activated receptor (Lxr) and constitutive androstane receptor (Car). In the kidney of HN rats, the mRNA level of the drug-metabolizing enzyme Cyp2b1 significantly increased, while levels of Cyp1a1, Cyp2c11, Cyp3a1 and Cyp3a2 did not significantly change. The mRNA expression of the transporters multidrug and toxin extrusion (Mate) 1 and Mrp2 was obviously increased but was markedly depressed for peptide transporters (Pept) 1 and Pept2. These changes may be related to the cross effects of Pxr, Fxr and Car in kidney. CONCLUSION HN pathological status can alter the expression of drug-metabolizing enzymes and transporters in the liver and kidney to varying degrees, thus affecting the disposition of substrate drugs in vivo. This suggests that to avoid potential risks, caution should be exercised when administering combination therapy for HN treatment.
Collapse
Affiliation(s)
- Yueqing Pan
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 1023 Shatai South Road, Baiyun District, Guangzhou, 510515, Guangdong, China
| | - Zhuan Yang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 1023 Shatai South Road, Baiyun District, Guangzhou, 510515, Guangdong, China
| | - Minlong Wei
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 1023 Shatai South Road, Baiyun District, Guangzhou, 510515, Guangdong, China
| | - Yulin Gan
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 1023 Shatai South Road, Baiyun District, Guangzhou, 510515, Guangdong, China
| | - Menghua Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, 1023 Shatai South Road, Baiyun District, Guangzhou, 510515, Guangdong, China.
| | - Wei Zou
- Changsha Research and Development Center on Obstetric and Gynecologic Traditional Chinese Medicine Preparation, NHC Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, No. 53 Xiangchun Road, Kaifu District, Changsha, 410008, Hunan, China.
| |
Collapse
|
13
|
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
|
14
|
Elbahnsi A, Dudas B, Callebaut I, Hinzpeter A, Miteva MA. ATP-Binding Cassette and Solute Carrier Transporters: Understanding Their Mechanisms and Drug Modulation Through Structural and Modeling Approaches. Pharmaceuticals (Basel) 2024; 17:1602. [PMID: 39770445 PMCID: PMC11676857 DOI: 10.3390/ph17121602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
The ATP-binding cassette (ABC) and solute carrier (SLC) transporters play pivotal roles in cellular transport mechanisms, influencing a wide range of physiological processes and impacting various medical conditions. Recent advancements in structural biology and computational modeling have provided significant insights into their function and regulation. This review provides an overview of the current knowledge of human ABC and SLC transporters, emphasizing their structural and functional relationships, transport mechanisms, and the contribution of computational approaches to their understanding. Current challenges and promising future research and methodological directions are also discussed.
Collapse
Affiliation(s)
- Ahmad Elbahnsi
- Inserm U1268 MCTR, CiTCoM UMR 8038 CNRS, Université Paris Cité, 75006 Paris, France
| | - Balint Dudas
- Inserm U1268 MCTR, CiTCoM UMR 8038 CNRS, Université Paris Cité, 75006 Paris, France
| | - Isabelle Callebaut
- Muséum National d’Histoire Naturelle, UMR CNRS 7590, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie—IMPMC, Sorbonne Université, 75005 Paris, France
| | - Alexandre Hinzpeter
- CNRS, INSERM, Institut Necker Enfants Malades—INEM, Université Paris Cité, 75015 Paris, France
| | - Maria A. Miteva
- Inserm U1268 MCTR, CiTCoM UMR 8038 CNRS, Université Paris Cité, 75006 Paris, France
| |
Collapse
|
15
|
Jones LP, Bergeron HC, Martin DE, Murray J, Sancilio FD, Tripp RA. Probenecid Inhibits Extracellular Signal-Regulated Kinase and c-Jun N-Terminal Kinase Mitogen-Activated Protein Kinase Pathways in Regulating Respiratory Syncytial Virus Response. Int J Mol Sci 2024; 25:12452. [PMID: 39596517 PMCID: PMC11594929 DOI: 10.3390/ijms252212452] [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: 10/09/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
We examined the effect of probenecid in regulating the ERK and JNK downstream MAPK pathways affecting respiratory syncytial virus replication. BACKGROUND We have previously shown that probenecid inhibits RSV, influenza virus, and SARS-CoV-2 replication in vitro in preclinical animal models and in humans. In a Phase two randomized, placebo-controlled, single-blind, dose range-finding study using probenecid to treat non-hospitalized patients with symptomatic, mild-to-moderate COVID-19, we previously showed that a 1000 mg twice daily treatment for 5 days reduced the median time to viral clearance from 11 to 7 days, and a 500 mg twice daily treatment for 5 days reduced the time to viral clearance from 11 to 9 days more than the placebo. METHODS In this study, we sought to determine the mechanism of action of the probenecid inhibition of RSV replication in human respiratory epithelial (A549) cells. RESULTS We show that probenecid inhibits the RSV-induced phosphorylation of JNKs and ERKs and the downstream phosphorylation of c-jun, a component of the AP-1 transcription complex needed for virus replication. The inhibition of JNKs by probenecid reversed the repression of transcription factor HNF-4. CONCLUSION The probenecid inhibition of JNK and ERK phosphorylation involves the MAPK pathway that precludes virus replication.
Collapse
Affiliation(s)
- Les P. Jones
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA; (L.P.J.); (H.C.B.); (J.M.)
| | - Harrison C. Bergeron
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA; (L.P.J.); (H.C.B.); (J.M.)
| | | | - Jackelyn Murray
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA; (L.P.J.); (H.C.B.); (J.M.)
| | - Fred D. Sancilio
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, FL 33431, USA;
| | - Ralph A. Tripp
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA; (L.P.J.); (H.C.B.); (J.M.)
- TrippBio, Inc., Jacksonville, FL 32256, USA;
| |
Collapse
|
16
|
Granados JC, Nigam SK. Organic anion transporters in remote sensing and organ crosstalk. Pharmacol Ther 2024; 263:108723. [PMID: 39284369 DOI: 10.1016/j.pharmthera.2024.108723] [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: 05/01/2024] [Revised: 08/29/2024] [Accepted: 09/13/2024] [Indexed: 11/05/2024]
Abstract
The organic anion transporters, OAT1 and OAT3, regulate the movement of drugs, toxins, and endogenous metabolites. In 2007, we proposed that OATs and other SLC22 transporters are involved in "remote sensing" and organ crosstalk. This is now known as the Remote Sensing and Signaling Theory (RSST). In the proximal tubule of the kidney, OATs regulate signaling molecules such as fatty acids, bile acids, indoxyl sulfate, kynurenine, alpha-ketoglutarate, urate, flavonoids, and antioxidants. OAT1 and OAT3 function as key hubs in a large homeostatic network involving multi-, oligo- and monospecific transporters, enzymes, and nuclear receptors. The Remote Sensing and Signaling Theory emphasizes the functioning of OATs and other "drug" transporters in the network at multiple biological scales (inter-organismal, organism, organ, cell, organelle). This network plays an essential role in the homeostasis of urate, bile acids, prostaglandins, sex steroids, odorants, thyroxine, gut microbiome metabolites, and uremic toxins. The transported metabolites have targets in the kidney and other organs, including nuclear receptors (e.g., HNF4a, AHR), G protein-coupled receptors (GPCRs), and protein kinases. Feed-forward and feedback loops allow OAT1 and OAT3 to mediate organ crosstalk as well as modulate energy metabolism, redox state, and remote sensing. Furthermore, there is intimate inter-organismal communication between renal OATs and the gut microbiome. Extracellular vesicles containing microRNAs and proteins (exosomes) play a key role in the Remote Sensing and Signaling System as does the interplay with the neuroendocrine, hormonal, and immune systems. Perturbation of function with OAT-interacting drugs (e.g., probenecid, diuretics, antivirals, antibiotics, NSAIDs) can lead to drug-metabolite interactions. The RSST has general applicability to other multi-specific SLC and ABC "drug" transporters (e.g., OCT1, OCT2, SLCO1B1, SLCO1B3, ABCG2, P-gp, ABCC2, ABCC3, ABCC4). Recent high-resolution structures of SLC22 and other transporters, together with chemoinformatic and artificial intelligence methods, will aid drug development and also lead to a deeper mechanistic understanding of polymorphisms.
Collapse
Affiliation(s)
- Jeffry C Granados
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Sanjay K Nigam
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine (Nephrology), University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
17
|
Dai Y, Lee CH. Transport mechanism and structural pharmacology of human urate transporter URAT1. Cell Res 2024; 34:776-787. [PMID: 39245778 PMCID: PMC11528023 DOI: 10.1038/s41422-024-01023-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/20/2024] [Indexed: 09/10/2024] Open
Abstract
Urate is an endogenous product of purine metabolism in the liver. High urate levels in the blood lead to gout, a very common and painful inflammatory arthritis. Excreted urate is reabsorbed in the kidney mainly by URAT1 antiporter, a key target for anti-gout drugs. To uncover the mechanisms of urate transport and drug inhibition, we determined cryo-EM structures of human URAT1 with urate, counter anion pyrazinoate, or anti-gout drugs of different chemotypes - lesinurad, verinurad, and dotinurad. We captured the outward-to-inward transition of URAT1 during urate uptake, revealing that urate binds in a phenylalanine-rich pocket and engages with key gating residues to drive the transport cycle. In contrast to the single binding site for urate, pyrazinoate interacts with three distinct, functionally relevant sites within URAT1, a mechanism that has not yet been observed in other anion antiporters. In addition, we found that while all three drugs compete with substrates and halt the transport cycle, verinurad and dotinurad further hijack gating residues to achieve high potency. These insights advance our understanding of organic anion transport and provide a foundation for designing improved gout therapeutics.
Collapse
Affiliation(s)
- Yaxin Dai
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Chia-Hsueh Lee
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
18
|
Yu Z, You G. Recent Advances on the Regulations of Organic Anion Transporters. Pharmaceutics 2024; 16:1355. [PMID: 39598479 PMCID: PMC11597148 DOI: 10.3390/pharmaceutics16111355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
The organic anion transporter (OAT) family of over 10 members within the solute carrier (SLC) superfamily of membrane proteins plays critical roles in facilitating the flux of negatively charged molecules in and out of cell membranes. These anionic molecules include various endogenous and exogenous compounds such as signaling molecules, nutrients, metabolites, toxins, and drugs. Therefore, OATs actively contribute to the systemic homeostasis and efficacy of therapeutics. This article provides a brief overview on recent advances in the understanding of the regulatory mechanisms that control the expression and activity of OATs in both health and diseases.
Collapse
Affiliation(s)
| | - Guofeng You
- Department of Pharmaceutics, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA;
| |
Collapse
|
19
|
Wang C, Vidal B, Sural S, Loer C, Aguilar GR, Merritt DM, Toker IA, Vogt MC, Cros CC, Hobert O. A neurotransmitter atlas of C. elegans males and hermaphrodites. eLife 2024; 13:RP95402. [PMID: 39422452 PMCID: PMC11488851 DOI: 10.7554/elife.95402] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Mapping neurotransmitter identities to neurons is key to understanding information flow in a nervous system. It also provides valuable entry points for studying the development and plasticity of neuronal identity features. In the Caenorhabditis elegans nervous system, neurotransmitter identities have been largely assigned by expression pattern analysis of neurotransmitter pathway genes that encode neurotransmitter biosynthetic enzymes or transporters. However, many of these assignments have relied on multicopy reporter transgenes that may lack relevant cis-regulatory information and therefore may not provide an accurate picture of neurotransmitter usage. We analyzed the expression patterns of 16 CRISPR/Cas9-engineered knock-in reporter strains for all main types of neurotransmitters in C. elegans (glutamate, acetylcholine, GABA, serotonin, dopamine, tyramine, and octopamine) in both the hermaphrodite and the male. Our analysis reveals novel sites of expression of these neurotransmitter systems within both neurons and glia, as well as non-neural cells, most notably in gonadal cells. The resulting expression atlas defines neurons that may be exclusively neuropeptidergic, substantially expands the repertoire of neurons capable of co-transmitting multiple neurotransmitters, and identifies novel sites of monoaminergic neurotransmitter uptake. Furthermore, we also observed unusual co-expression patterns of monoaminergic synthesis pathway genes, suggesting the existence of novel monoaminergic transmitters. Our analysis results in what constitutes the most extensive whole-animal-wide map of neurotransmitter usage to date, paving the way for a better understanding of neuronal communication and neuronal identity specification in C. elegans.
Collapse
Affiliation(s)
- Chen Wang
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Berta Vidal
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Surojit Sural
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Curtis Loer
- Department of Biology, University of San DiegoSan DiegoUnited States
| | - G Robert Aguilar
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Daniel M Merritt
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Itai Antoine Toker
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Merly C Vogt
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Cyril C Cros
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Oliver Hobert
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| |
Collapse
|
20
|
Cui J, Wen Z, Huang H, Qin S, Luo Y, Zhang W, Wu W. The Pharmacokinetics and Liver-Targeting Evaluation of Silybin Liposomes Mediated by the NTCP/OCTN2 Dual Receptors. Mol Pharm 2024; 21:4912-4923. [PMID: 39370820 DOI: 10.1021/acs.molpharmaceut.3c01245] [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] [Indexed: 10/08/2024]
Abstract
The disadvantage of a traditional dosage regimen is the inability to deliver a sufficient drug concentration to the lesion site, which can result in adverse side effects due to nonspecific drug delivery. Actively targeting hepatic cells is a promising therapeutic strategy for liver disease. In this study, l-carnitine and a targeting peptide derived from the hepatitis B virus large envelope protein were used to modify liposomes for drug delivery to the liver through the sodium taurocholate cotransporting polypeptide (NTCP) and the organic cation/carnitine transporter 2 (OCTN2) receptors. Silybin was selected as the model drug. The solubility of silybin can reach 0.3 mg/mL after encapsulation in liposomes. The NTCP-specific and OCTN2-accelerated Myrcludex B and l-carnitine dual-modified liposomes were validated in vitro. The uptake of coumarin-6 in dual ligand-modified liposomes by hepatocytes was up to 2.36 μg/mg compared with unmodified liposomes (1.05 μg/mg). The pharmacokinetics and targeting abilities of various liposome formulations were evaluated in Kunming mice. Targeted liposomes increased the concentration of silybin and prolonged the drug's retention time in the liver. The area under the liver's pharmacokinetic curve of targeted liposomes was twice that of silybin injection, suggesting the promising application potential of silybin-loaded hepatotropic nanovesicles.
Collapse
Affiliation(s)
- Jian Cui
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Zhiwei Wen
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Huajie Huang
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Shuilin Qin
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Yanjie Luo
- Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, Guangxi 541002, China
| | - Wei Zhang
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Wei Wu
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
| |
Collapse
|
21
|
Xu Y, Liu C, Meng Q. ZNF217 Mediates Transcriptional Activation of GRHL3 to Regulate SLC22A31 and Promote Malignant Progression in Thyroid Cancer. Mol Biotechnol 2024:10.1007/s12033-024-01292-6. [PMID: 39354204 DOI: 10.1007/s12033-024-01292-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/17/2024] [Indexed: 10/03/2024]
Abstract
The incidence of thyroid cancer (THCA) has increased worldwide during the past 40 years. However, an understanding of the mechanisms and major transcription factors involved in THCA is insufficient to identify therapeutic targets against THCA. To reveal such mechanisms, we conducted bioinformatics analyses to assess the differential expression in human THCA sample and normal tissue sample, leading us to focus on the function of the ZNF217/GRHL3/ SLC22A31 axis in mediating biological activity in THCA. The genes of interest were interfered with lentiviral vectors, and transfection efficiency was verified using RT-qPCR. ZNF217, GRHL3, and SLC22A31 were abundantly expressed in THCA tissues or cells. Knockdown of GRHL3, ZNF217, or SLC22A31 all significantly curtailed the malignant biological behavior of THCA cells. ZNF217 promoted GRHL3 expression through transcriptional activation, thereby increasing the transcription of SLC22A31. Ectopic expression of GRHL3 or SLC22A31 abated the suppressing impact of ZNF217 or GRHL3 knockdown on the biological activity of THCA cells. Collectively, our results demonstrated that ZNF217 acted as an activator of GRHL3, thereby promoting the expression of SLC22A31 and the malignant activity of THCA cells.
Collapse
Affiliation(s)
- Ying Xu
- Department of Ultrasound Medicine, Daqing Oilfield General Hospital, No.9, Zhongkang Street, Daqing, 163000, Heilongjiang, People's Republic of China
| | - Chunxu Liu
- Department of Ultrasound Medicine, Daqing Oilfield General Hospital, No.9, Zhongkang Street, Daqing, 163000, Heilongjiang, People's Republic of China
| | - Qingrui Meng
- Department of Ultrasound Medicine, Daqing Oilfield General Hospital, No.9, Zhongkang Street, Daqing, 163000, Heilongjiang, People's Republic of China.
| |
Collapse
|
22
|
Pernecker M, Ciarimboli G. Regulation of renal organic cation transporters. FEBS Lett 2024; 598:2328-2347. [PMID: 38831380 DOI: 10.1002/1873-3468.14943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/30/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024]
Abstract
Transporters for organic cations (OCs) facilitate exchange of positively charged molecules through the plasma membrane. Substrates for these transporters encompass neurotransmitters, metabolic byproducts, drugs, and xenobiotics. Consequently, these transporters actively contribute to the regulation of neurotransmission, cellular penetration and elimination process for metabolic products, drugs, and xenobiotics. Therefore, these transporters have significant physiological, pharmacological, and toxicological implications. In cells of renal proximal tubules, the vectorial secretion pathways for OCs involve expression of organic cation transporters (OCTs) and multidrug and toxin extrusion proteins (MATEs) on basolateral and apical membrane domains, respectively. This review provides an overview of documented regulatory mechanisms governing OCTs and MATEs. Additionally, regulation of these transporters under various pathological conditions is summarized. The expression and functionality of OCTs and MATEs are subject to diverse pre- and post-translational modifications, providing insights into their regulation in various pathological conditions. Typically, in diseases, downregulation of transporter expression is observed, probably as a protective mechanism to prevent additional damage to kidney tissue. This regulation may be attributed to the intricate network of modifications these transporters undergo, shedding light on their dynamic responses in pathological contexts.
Collapse
Affiliation(s)
- Moritz Pernecker
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, Germany
| | - Giuliano Ciarimboli
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, Germany
| |
Collapse
|
23
|
Uchimura Y, Hino K, Hattori K, Kubo Y, Owada A, Kimura T, Sugawara L, Kume S, Bellier JP, Yanagisawa D, Shiino A, Nakayama T, Daigo Y, Mashimo T, Udagawa J. Knockout of the orphan membrane transporter Slc22a23 leads to a lean and hyperactive phenotype with a small hippocampal volume. PLoS One 2024; 19:e0309461. [PMID: 39197039 PMCID: PMC11356391 DOI: 10.1371/journal.pone.0309461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/12/2024] [Indexed: 08/30/2024] Open
Abstract
Epidemiological studies suggest that poor nutrition during pregnancy predisposes offspring to the development of lifestyle-related noncommunicable diseases and psychiatric disorders later in life. However, the molecular mechanisms underlying this predisposition are not well understood. In our previous study, using rats as model animals, we showed that behavioral impairments are induced by prenatal undernutrition. In this study, we identified solute carrier 22 family member 23 (Slc22a23) as a gene that is irreversibly upregulated in the rat brain by undernutrition during fetal development. Because the substrate of the SLC22A23 transporter has not yet been identified and the biological role of the Slc22a23 gene in vivo is not fully understood, we generated pan-Slc22a23 knockout rats and examined their phenotype in detail. The Slc22a23 knockout rats showed a lean phenotype, an increase in spontaneous locomotion, and improved endurance, indicating that they are not overweight and are even healthier in an ad libitum feeding environment. However, the knockout rats had reduced hippocampal volume, and the behavioral analysis suggested that they may have impaired cognitive function regarding novel objects.
Collapse
Affiliation(s)
- Yasuhiro Uchimura
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Kodai Hino
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Kosuke Hattori
- Division of Animal Genetics, Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoshinori Kubo
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Airi Owada
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Tomoko Kimura
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Lucia Sugawara
- Department of Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Shinji Kume
- Department of Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Jean-Pierre Bellier
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Daijiro Yanagisawa
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Akihiko Shiino
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Takahisa Nakayama
- Division of Human Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Yataro Daigo
- Department of Medical Oncology, Cancer Center and Center for Advanced Medicine against Cancer, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Tomoji Mashimo
- Division of Animal Genetics, Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Jun Udagawa
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Shiga, Japan
| |
Collapse
|
24
|
Alves LDF, Moore JB, Kell DB. The Biology and Biochemistry of Kynurenic Acid, a Potential Nutraceutical with Multiple Biological Effects. Int J Mol Sci 2024; 25:9082. [PMID: 39201768 PMCID: PMC11354673 DOI: 10.3390/ijms25169082] [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: 07/19/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Kynurenic acid (KYNA) is an antioxidant degradation product of tryptophan that has been shown to have a variety of cytoprotective, neuroprotective and neuronal signalling properties. However, mammalian transporters and receptors display micromolar binding constants; these are consistent with its typically micromolar tissue concentrations but far above its serum/plasma concentration (normally tens of nanomolar), suggesting large gaps in our knowledge of its transport and mechanisms of action, in that the main influx transporters characterized to date are equilibrative, not concentrative. In addition, it is a substrate of a known anion efflux pump (ABCC4), whose in vivo activity is largely unknown. Exogeneous addition of L-tryptophan or L-kynurenine leads to the production of KYNA but also to that of many other co-metabolites (including some such as 3-hydroxy-L-kynurenine and quinolinic acid that may be toxic). With the exception of chestnut honey, KYNA exists at relatively low levels in natural foodstuffs. However, its bioavailability is reasonable, and as the terminal element of an irreversible reaction of most tryptophan degradation pathways, it might be added exogenously without disturbing upstream metabolism significantly. Many examples, which we review, show that it has valuable bioactivity. Given the above, we review its potential utility as a nutraceutical, finding it significantly worthy of further study and development.
Collapse
Affiliation(s)
- Luana de Fátima Alves
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - J. Bernadette Moore
- School of Food Science & Nutrition, University of Leeds, Leeds LS2 9JT, UK;
- Department of Biochemistry, Cell & Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
| | - Douglas B. Kell
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Søltofts Plads, 2800 Kongens Lyngby, Denmark
- Department of Biochemistry, Cell & Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
| |
Collapse
|
25
|
Hu H, Li W, Hao Y, Peng Z, Zou Z, Wei J, Zhou Y, Liang W, Cao Y. The SGLT2 inhibitor dapagliflozin ameliorates renal fibrosis in hyperuricemic nephropathy. Cell Rep Med 2024; 5:101690. [PMID: 39168099 PMCID: PMC11384938 DOI: 10.1016/j.xcrm.2024.101690] [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: 01/03/2024] [Revised: 07/01/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024]
Abstract
Hyperuricemic nephropathy (HN) is a global metabolic disorder characterized by uric acid (UA) metabolism dysfunction, resulting in hyperuricemia (HUA) and tubulointerstitial fibrosis (TIF). Sodium-dependent glucose transporter 2 inhibitor, dapagliflozin, has shown potential in reducing serum UA levels in patients with chronic kidney disease (CKD), though its protective effects against HN remain uncertain. This study investigates the functional, pathological, and molecular changes in HN through histological, biochemical, and transcriptomic analyses in patients, HN mice, and UA-stimulated HK-2 cells. Findings indicate UA-induced tubular dysfunction and fibrotic activation, which dapagliflozin significantly mitigates. Transcriptomic analysis identifies estrogen-related receptor α (ERRα), a downregulated transcription factor in HN. ERRα knockin mice and ERRα-overexpressed HK-2 cells demonstrate UA resistance, while ERRα inhibition exacerbates UA effects. Dapagliflozin targets ERRα, activating the ERRα-organic anion transporter 1 (OAT1) axis to enhance UA excretion and reduce TIF. Furthermore, dapagliflozin ameliorates renal fibrosis in non-HN CKD models, underscoring the therapeutic significance of the ERRα-OAT1 axis in HN and CKD.
Collapse
Affiliation(s)
- Hongtu Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Key Clinical Research Center of Kidney Disease in Hubei, 238 Jiefang Road, Wuhan, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weiwei Li
- Division of Nephrology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, No. 158 Wuyang Avenue, Enshi, China
| | - Yiqun Hao
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhuan Peng
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhengping Zou
- Division of Nephrology, Qianjiang Hospital Affiliated to Renmin Hospital of Wuhan University, Wuhan, China; Qianjiang Clinical Medical College, Health Science Center, Yangtze University, Jingzhou, China
| | - Jiali Wei
- Department of Nephrology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Ying Zhou
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, No. 45 Changchun St, Xicheng District, Beijing 100053, China.
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Key Clinical Research Center of Kidney Disease in Hubei, 238 Jiefang Road, Wuhan, China.
| | - Yun Cao
- Department of Nephrology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China.
| |
Collapse
|
26
|
Han C, He C, Ding X, Li Z, Peng T, Zhang C, Chen H, Zuo Z, Huang J, Hu W. WWC1 upregulation accelerates hyperuricemia by reduction in renal uric acid excretion through Hippo signaling pathway. J Biol Chem 2024; 300:107485. [PMID: 38906255 PMCID: PMC11301351 DOI: 10.1016/j.jbc.2024.107485] [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: 01/24/2024] [Revised: 05/24/2024] [Accepted: 06/05/2024] [Indexed: 06/23/2024] Open
Abstract
Hyperuricemia (HUA) is a metabolic disorder characterized by elevated serum uric acid (UA), primarily attributed to the hepatic overproduction and renal underexcretion of UA. Despite the elucidation of molecular pathways associated with this underexcretion, the etiology of HUA remains largely unknown. In our study, using by Uox knockout rats, HUA mouse, and cell line models, we discovered that the increased WWC1 levels were associated with decreased renal UA excretion. Additionally, using knockdown and overexpression approaches, we found that WWC1 inhibited UA excretion in renal tubular epithelial cells. Mechanistically, WWC1 activated the Hippo pathway, leading to phosphorylation and subsequent degradation of the downstream transcription factor YAP1, thereby impairing the ABCG2 and OAT3 expression through transcriptional regulation. Consequently, this reduction led to a decrease in UA excretion in renal tubular epithelial cells. In conclusion, our study has elucidated the role of upregulated WWC1 in renal tubular epithelial cells inhibiting the excretion of UA in the kidneys and causing HUA.
Collapse
Affiliation(s)
- Changshun Han
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Chengyong He
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xiaoyan Ding
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zixuan Li
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Tianyun Peng
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Chensong Zhang
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Haibing Chen
- Department of Endocrinology and Metabolism, Shanghai 10th People's Hospital, Tongji University, Shanghai, China
| | - Zhenghong Zuo
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jiyi Huang
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China.
| | - Weiping Hu
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
27
|
Marin JJG, Serrano MA, Herraez E, Lozano E, Ortiz-Rivero S, Perez-Silva L, Reviejo M, Briz O. Impact of genetic variants in the solute carrier ( SLC) genes encoding drug uptake transporters on the response to anticancer chemotherapy. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:27. [PMID: 39143954 PMCID: PMC11322974 DOI: 10.20517/cdr.2024.42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 08/16/2024]
Abstract
Cancer drug resistance constitutes a severe limitation for the satisfactory outcome of these patients. This is a complex problem due to the co-existence in cancer cells of multiple and synergistic mechanisms of chemoresistance (MOC). These mechanisms are accounted for by the expression of a set of genes included in the so-called resistome, whose effectiveness often leads to a lack of response to pharmacological treatment. Additionally, genetic variants affecting these genes further increase the complexity of the question. This review focuses on a set of genes encoding members of the transportome involved in drug uptake, which have been classified into the MOC-1A subgroup of the resistome. These proteins belong to the solute carrier (SLC) superfamily. More precisely, we have considered here several members of families SLC2, SLC7, SLC19, SLC22, SLCO, SLC28, SLC29, SLC31, SLC46, and SLC47 due to the impact of their expression and genetic variants in anticancer drug uptake by tumor cells or, in some cases, general bioavailability. Changes in their expression levels and the appearance of genetic variants can contribute to the Darwinian selection of more resistant clones and, hence, to the development of a more malignant phenotype. Accordingly, to address this issue in future personalized medicine, it is necessary to characterize both changes in resistome genes that can affect their function. It is also essential to consider the time-dependent dimension of these features, as the genetic expression and the appearance of genetic variants can change during tumor progression and in response to treatment.
Collapse
Affiliation(s)
- Jose J. G. Marin
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Maria A. Serrano
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Elisa Herraez
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Elisa Lozano
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Sara Ortiz-Rivero
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Laura Perez-Silva
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
| | - Maria Reviejo
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Oscar Briz
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| |
Collapse
|
28
|
Rox K, Kühne A, Herrmann J, Jansen R, Hüttel S, Bernecker S, Hagos Y, Brönstrup M, Stadler M, Hesterkamp T, Müller R. Interaction of the Atypical Tetracyclines Chelocardin and Amidochelocardin with Renal Drug Transporters. ACS Pharmacol Transl Sci 2024; 7:2093-2109. [PMID: 39022358 PMCID: PMC11249637 DOI: 10.1021/acsptsci.4c00183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Antimicrobial resistance is expected to increase mortality rates by up to several million deaths per year by 2050 without new treatment options at hand. Recently, we characterized the pharmacokinetic (PK) and pharmacodynamic properties of two atypical tetracyclines, chelocardin (CHD) and amidochelocardin (CDCHD) that exhibit no cross-resistance with clinically used antibacterials. Both compounds were preferentially renally cleared and demonstrated pronounced effects in an ascending urinary tract infection model against E. coli. Renal drug transporters are known to influence clearance into the urine. In particular, inhibition of apical transporters in renal tubular epithelial cells can lead to intracellular accumulation and potential cell toxicity, whereas inhibition of basolateral transporters can cause a higher systemic exposure. Here, selected murine and human organic cation (Oct), organic anion (Oat), and efflux transporters were studied to elucidate interactions with CHD and CDCHD underlying their PK behavior. CHD exhibited stronger inhibitory effects on mOat1 and mOat3 and their human homologues hOAT1 and hOAT3 compared to CDCHD. While CHD was a substrate of mOat3 and mOct1, CDCHD was not. By contrast, no inhibitory effect was observed on Octs. CDCHD rather appeared to foster enhanced substrate transport on mOct1. CHD and CDCHD inhibited the efflux transporter hMRP2 on the apical side. In summary, the substrate nature of CHD in conjunction with its autoinhibition toward mOat3 rationalizes the distinct urine concentration profile compared to CDCHD that was previously observed in vivo. Further studies are needed to investigate the accumulation in renal tubular cells and the nephrotoxicity risk.
Collapse
Affiliation(s)
- Katharina Rox
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research (HZI), 38124 Braunschweig, Germany
- German
Center for Infection Research (DZIF), partner site Braunschweig-Hannover, 38124 Braunschweig, Germany
| | - Annett Kühne
- PortaCellTec
Biosciences GmbH, 37079 Göttingen, Germany
| | - Jennifer Herrmann
- German
Center for Infection Research (DZIF), partner site Braunschweig-Hannover, 38124 Braunschweig, Germany
- Department
of Microbial Natural Products, Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research (HZI) and Department of Pharmacy, Saarland
University, 66123 Saarbrücken, Germany
| | - Rolf Jansen
- Department
of Microbial Drugs, Helmholtz Centre for
Infection Research (HZI), 38124 Braunschweig, Germany
| | - Stephan Hüttel
- German
Center for Infection Research (DZIF), partner site Braunschweig-Hannover, 38124 Braunschweig, Germany
- Department
of Microbial Drugs, Helmholtz Centre for
Infection Research (HZI), 38124 Braunschweig, Germany
| | - Steffen Bernecker
- Department
of Microbial Drugs, Helmholtz Centre for
Infection Research (HZI), 38124 Braunschweig, Germany
| | | | - Mark Brönstrup
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research (HZI), 38124 Braunschweig, Germany
- German
Center for Infection Research (DZIF), partner site Braunschweig-Hannover, 38124 Braunschweig, Germany
| | - Marc Stadler
- German
Center for Infection Research (DZIF), partner site Braunschweig-Hannover, 38124 Braunschweig, Germany
- Department
of Microbial Drugs, Helmholtz Centre for
Infection Research (HZI), 38124 Braunschweig, Germany
| | - Thomas Hesterkamp
- German
Center for Infection Research (DZIF), partner site Braunschweig-Hannover, 38124 Braunschweig, Germany
- Translational
Product Management Office, German Center
for Infection Research (DZIF), partner site Braunschweig-Hannover, 38124 Braunschweig, Germany
| | - Rolf Müller
- German
Center for Infection Research (DZIF), partner site Braunschweig-Hannover, 38124 Braunschweig, Germany
- Department
of Microbial Natural Products, Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research (HZI) and Department of Pharmacy, Saarland
University, 66123 Saarbrücken, Germany
| |
Collapse
|
29
|
Kanbay M, Copur S, Guldan M, Ozbek L, Hatipoglu A, Covic A, Mallamaci F, Zoccali C. Proximal tubule hypertrophy and hyperfunction: a novel pathophysiological feature in disease states. Clin Kidney J 2024; 17:sfae195. [PMID: 39050867 PMCID: PMC11267238 DOI: 10.1093/ckj/sfae195] [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: 05/12/2024] [Indexed: 07/27/2024] Open
Abstract
The role of proximal tubules (PTs), a major component of the renal tubular structure in the renal cortex, has been examined extensively. Along with its physiological role in the reabsorption of various molecules, including electrolytes, amino acids and monosaccharides, transcellular transport of different hormones and regulation of homeostasis, pathological events affecting PTs may underlie multiple disease states. PT hypertrophy or a hyperfunctioning state, despite being a compensatory mechanism at first in response to various stimuli or alterations at tubular transport proteins, have been shown to be critical pathophysiological events leading to multiple disorders, including diabetes mellitus, obesity, metabolic syndrome and congestive heart failure. Moreover, pharmacotherapeutic agents have primarily targeted PTs, including sodium-glucose cotransporter 2, urate transporters and carbonic anhydrase enzymes. In this narrative review, we focus on the physiological role of PTs in healthy states and the current understanding of the PT pathologies leading to disease states and potential therapeutic targets.
Collapse
Affiliation(s)
- Mehmet Kanbay
- Department of Internal Medicine, Division of Nephrology, Koç University School of Medicine, Istanbul, Turkey
| | - Sidar Copur
- Department of Medicine, Koç University School of Medicine, Istanbul, Turkey
| | - Mustafa Guldan
- Department of Internal Medicine, Division of Internal Medicine, Koç University School of Medicine, Istanbul, Turkey
| | - Lasin Ozbek
- Department of Internal Medicine, Division of Internal Medicine, Koç University School of Medicine, Istanbul, Turkey
| | - Alper Hatipoglu
- Department of Internal Medicine, Division of Internal Medicine, Koç University School of Medicine, Istanbul, Turkey
| | - Adrian Covic
- Nephrology, Dialysis and Transplantation, University Grigore T Popa, Iasi, Romania
| | - Francesca Mallamaci
- Nephrology, Dialysis and Transplantation Unit, Grande Ospedale Metropolitano, Reggio Calabria, Italy
- CNR-IFC, Research Unit of Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Institute of Clinical Physiology, Reggio Calabria, Italy
| | - Carmine Zoccali
- Renal Research Institute, New York, NY, USA
- Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale, Grande Ospedale Metropolitano, c/o Nefrologia, Reggio Calabria, Italy
| |
Collapse
|
30
|
Jing F, Zhu L, Bai J, Zhou X, Sun L, Zhang H, Li T. A prognostic model built on amino acid metabolism patterns in HPV-associated head and neck squamous cell carcinoma. Arch Oral Biol 2024; 163:105975. [PMID: 38626700 DOI: 10.1016/j.archoralbio.2024.105975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/08/2024] [Accepted: 04/12/2024] [Indexed: 04/18/2024]
Abstract
OBJECTIVES To compare amino acid metabolism patterns between HPV-positive and HPV-negative head and neck squamous cell carcinoma (HNSCC) patients and identify key genes for a prognostic model. DESIGN Utilizing the Cancer Genome Atlas dataset, we analyzed amino acid metabolism genes, differentiated genes between HPV statuses, and selected key genes via LASSO regression for the prognostic model. The model's gene expression was verified through immunohistochemistry in clinical samples. Functional enrichment and CIBERSORTx analyses explored biological functions, molecular mechanisms, and immune cell correlations. The model's prognostic capability was assessed using nomograms, calibration, and decision curve analysis. RESULTS We identified 1157 key genes associated with amino acid metabolism in HNSCC and HPV status. The prognostic model, featuring genes like IQCN, SLC22A1, SYT12, and TLX3, highlighted functions in development, metabolism, and pathways related to receptors and enzymes. It significantly correlated with immune cell infiltration and outperformed traditional staging in prognosis prediction, despite immunohistochemistry results showing limited clinical identification of HPV-related HNSCC. CONCLUSIONS Distinct amino acid metabolism patterns differentiate HPV-positive from negative HNSCC patients, underscoring the prognostic model's utility in predicting outcomes and guiding therapeutic strategies.
Collapse
Affiliation(s)
- Fengyang Jing
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, China; Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences (2019RU034), Beijing 100081, China
| | - Lijing Zhu
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, China; Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences (2019RU034), Beijing 100081, China
| | - Jiaying Bai
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Xuan Zhou
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, China; Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences (2019RU034), Beijing 100081, China
| | - Lisha Sun
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing 100081, China; Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences (2019RU034), Beijing 100081, China.
| | - Heyu Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing 100081, China; Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences (2019RU034), Beijing 100081, China.
| | - Tiejun Li
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, China; Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences (2019RU034), Beijing 100081, China.
| |
Collapse
|
31
|
Wang C, Vidal B, Sural S, Loer C, Aguilar GR, Merritt DM, Toker IA, Vogt MC, Cros C, Hobert O. A neurotransmitter atlas of C. elegans males and hermaphrodites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.24.573258. [PMID: 38895397 PMCID: PMC11185579 DOI: 10.1101/2023.12.24.573258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Mapping neurotransmitter identities to neurons is key to understanding information flow in a nervous system. It also provides valuable entry points for studying the development and plasticity of neuronal identity features. In the C. elegans nervous system, neurotransmitter identities have been largely assigned by expression pattern analysis of neurotransmitter pathway genes that encode neurotransmitter biosynthetic enzymes or transporters. However, many of these assignments have relied on multicopy reporter transgenes that may lack relevant cis-regulatory information and therefore may not provide an accurate picture of neurotransmitter usage. We analyzed the expression patterns of 16 CRISPR/Cas9-engineered knock-in reporter strains for all main types of neurotransmitters in C. elegans (glutamate, acetylcholine, GABA, serotonin, dopamine, tyramine, and octopamine) in both the hermaphrodite and the male. Our analysis reveals novel sites of expression of these neurotransmitter systems within both neurons and glia, as well as non-neural cells. The resulting expression atlas defines neurons that may be exclusively neuropeptidergic, substantially expands the repertoire of neurons capable of co-transmitting multiple neurotransmitters, and identifies novel neurons that uptake monoaminergic neurotransmitters. Furthermore, we also observed unusual co-expression patterns of monoaminergic synthesis pathway genes, suggesting the existence of novel monoaminergic transmitters. Our analysis results in what constitutes the most extensive whole-animal-wide map of neurotransmitter usage to date, paving the way for a better understanding of neuronal communication and neuronal identity specification in C. elegans.
Collapse
Affiliation(s)
- Chen Wang
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| | - Berta Vidal
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| | - Surojit Sural
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| | - Curtis Loer
- Department of Biology, University of San Diego, San Diego, California, USA
| | - G. Robert Aguilar
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| | - Daniel M. Merritt
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| | - Itai Antoine Toker
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| | - Merly C. Vogt
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| | - Cyril Cros
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| | - Oliver Hobert
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, USA
| |
Collapse
|
32
|
Thévenod F, Lee WK. Cadmium transport by mammalian ATP-binding cassette transporters. Biometals 2024; 37:697-719. [PMID: 38319451 PMCID: PMC11101381 DOI: 10.1007/s10534-024-00582-5] [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: 09/15/2023] [Accepted: 01/04/2024] [Indexed: 02/07/2024]
Abstract
Cellular responses to toxic metals depend on metal accessibility to intracellular targets, reaching interaction sites, and the intracellular metal concentration, which is mainly determined by uptake pathways, binding/sequestration and efflux pathways. ATP-binding cassette (ABC) transporters are ubiquitous in the human body-usually in epithelia-and are responsible for the transfer of indispensable physiological substrates (e.g. lipids and heme), protection against potentially toxic substances, maintenance of fluid composition, and excretion of metabolic waste products. Derailed regulation and gene variants of ABC transporters culminate in a wide array of pathophysiological disease states, such as oncogenic multidrug resistance or cystic fibrosis. Cadmium (Cd) has no known physiological role in mammalians and poses a health risk due to its release into the environment as a result of industrial activities, and eventually passes into the food chain. Epithelial cells, especially within the liver, lungs, gastrointestinal tract and kidneys, are particularly susceptible to the multifaceted effects of Cd because of the plethora of uptake pathways available. Pertinent to their broad substrate spectra, ABC transporters represent a major cellular efflux pathway for Cd and Cd complexes. In this review, we summarize current knowledge concerning transport of Cd and its complexes (mainly Cd bound to glutathione) by the ABC transporters ABCB1 (P-glycoprotein, MDR1), ABCB6, ABCC1 (multidrug resistance related protein 1, MRP1), ABCC7 (cystic fibrosis transmembrane regulator, CFTR), and ABCG2 (breast cancer related protein, BCRP). Potential detoxification strategies underlying ABC transporter-mediated efflux of Cd and Cd complexes are discussed.
Collapse
Affiliation(s)
- Frank Thévenod
- Institute for Physiology, Pathophysiology and Toxicology & ZBAF, Witten/Herdecke University, 58453, Witten, Germany
- Physiology and Pathophysiology of Cells and Membranes, Medical School OWL, Bielefeld University, Morgenbreede 1, 33615, Bielefeld, Germany
| | - Wing-Kee Lee
- Physiology and Pathophysiology of Cells and Membranes, Medical School OWL, Bielefeld University, Morgenbreede 1, 33615, Bielefeld, Germany.
| |
Collapse
|
33
|
Meijers B, Zadora W, Lowenstein J. A Historical Perspective on Uremia and Uremic Toxins. Toxins (Basel) 2024; 16:227. [PMID: 38787079 PMCID: PMC11126090 DOI: 10.3390/toxins16050227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/12/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
Uremia, also known as uremic syndrome, refers to the clinical symptoms in the final stage of renal failure. The definition of the term has changed over time due to an improved comprehension of the kidney's function and the advancement of dialysis technology. Here, we aim to present an overview of the various concepts that have developed regarding uremia throughout the years. We provide a comprehensive review of the historical progression starting from the early days of Kolff and his predecessors, continuing with the initial research conducted by Niwa et al., and culminating in the remote sensing hypothesis of Nigam. Additionally, we explore the subsequent investigation into the function of these toxins as signaling molecules in various somatic cells.
Collapse
Affiliation(s)
- Björn Meijers
- Nephrology and Transplantation Unit, University Hospitals Leuven, 30000 Leuven, Belgium; (B.M.); (W.Z.)
- Laboratory of Nephrology, Katholieke Universiteit Leuven, 30000 Leuven, Belgium
| | - Ward Zadora
- Nephrology and Transplantation Unit, University Hospitals Leuven, 30000 Leuven, Belgium; (B.M.); (W.Z.)
- Laboratory of Nephrology, Katholieke Universiteit Leuven, 30000 Leuven, Belgium
| | - Jerome Lowenstein
- Nephrology Division, NYU Langone Medical Center, New York, NY 10016, USA
| |
Collapse
|
34
|
Shi X, Zhao T, Wang S, Xu S, Liao H, Gao S, Gao Z, Zhang J, Qi D, Zhang Z, Zheng F, Wang Y, Wang Z, Yang M, Yang Q, Yi F, Pang J, Liu X, Zhan P. Discovery of a Novel Thienopyrimidine Compound as a Urate Transporter 1 and Glucose Transporter 9 Dual Inhibitor with Improved Efficacy and Favorable Druggability. J Med Chem 2024; 67:5032-5052. [PMID: 38482820 DOI: 10.1021/acs.jmedchem.4c00136] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Gout and hyperuricemia are metabolic diseases characterized with high serum uric acid (SUA) levels that significantly impact human health. Lesinurad, a uricosuric agent, is limited to concurrent use with xanthine oxidase inhibitors (XOIs) in clinical practice due to its restricted efficacy and potential nephrotoxicity. Herein, extensive structural modifications of lesinurad were conducted through scaffold hopping and substituent modification strategies, affording 54 novel derivatives containing pyrimidine-fused cyclic structures. Notably, the thienopyrimidine compound 29 demonstrated a remarkable 2-fold increase in SUA-lowering in vivo activity compared to lesinurad, while exhibiting potent inhibitory activity against the urate transporter 1 (URAT1, IC50 = 2.01 μM) and glucose transporter 9 (GLUT9, IC50 = 18.21 μM). Furthermore, it possessed a lower effective dosage of 0.5 mg/kg, favorable safety profile without any apparent acute toxicity at doses of 1000 mg/kg, and improved pharmacokinetic properties. Overall, we have discovered an efficacious URAT1/GLUT9 dual inhibitor for inhibiting urate reabsorption with favorable pharmacokinetic profiles.
Collapse
Affiliation(s)
- Xiaoyu Shi
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, 250012 Shandong, PR China
| | - Tong Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, 250012 Shandong, PR China
| | - Shuo Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, 250012 Shandong, PR China
| | - Shujing Xu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, 250012 Shandong, PR China
| | - Hui Liao
- School of Pharmaceutical Sciences, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou 510515, PR China
| | - Shenghua Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, 250012 Shandong, PR China
| | - Zhen Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, 250012 Shandong, PR China
| | - Jian Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, 250012 Shandong, PR China
| | - Danhui Qi
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, 250012 Shandong, PR China
| | - Zhijiao Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, 250012 Shandong, PR China
| | - Fengxin Zheng
- School of Pharmaceutical Sciences, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou 510515, PR China
| | - Youzhao Wang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Zhenqian Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, 250012 Shandong, PR China
| | - Mingyu Yang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, 250012 Shandong, PR China
| | - Qian Yang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, 250012 Shandong, PR China
| | - Fan Yi
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Jianxin Pang
- School of Pharmaceutical Sciences, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou 510515, PR China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, 250012 Shandong, PR China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, 250012 Shandong, PR China
| |
Collapse
|
35
|
Pochini L, Galluccio M, Console L, Scalise M, Eberini I, Indiveri C. Inflammation and Organic Cation Transporters Novel (OCTNs). Biomolecules 2024; 14:392. [PMID: 38672410 PMCID: PMC11048549 DOI: 10.3390/biom14040392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Inflammation is a physiological condition characterized by a complex interplay between different cells handled by metabolites and specific inflammatory-related molecules. In some pathological situations, inflammation persists underlying and worsening the pathological state. Over the years, two membrane transporters namely OCTN1 (SLC22A4) and OCTN2 (SLC22A5) have been shown to play specific roles in inflammation. These transporters form the OCTN subfamily within the larger SLC22 family. The link between these proteins and inflammation has been proposed based on their link to some chronic inflammatory diseases such as asthma, Crohn's disease (CD), and rheumatoid arthritis (RA). Moreover, the two transporters show the ability to mediate the transport of several compounds including carnitine, carnitine derivatives, acetylcholine, ergothioneine, and gut microbiota by-products, which have been specifically associated with inflammation for their anti- or proinflammatory action. Therefore, the absorption and distribution of these molecules rely on the presence of OCTN1 and OCTN2, whose expression is modulated by inflammatory cytokines and transcription factors typically activated by inflammation. In the present review, we wish to provide a state of the art on OCTN1 and OCTN2 transport function and regulation in relationships with inflammation and inflammatory diseases focusing on the metabolic signature collected in different body districts and gene polymorphisms related to inflammatory diseases.
Collapse
Affiliation(s)
- Lorena Pochini
- Laboratory of Biochemistry, Molecular Biotechnology and Molecular Biology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (M.G.); (L.C.); (M.S.)
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via Amendola 122/O, 70126 Bari, Italy
| | - Michele Galluccio
- Laboratory of Biochemistry, Molecular Biotechnology and Molecular Biology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (M.G.); (L.C.); (M.S.)
| | - Lara Console
- Laboratory of Biochemistry, Molecular Biotechnology and Molecular Biology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (M.G.); (L.C.); (M.S.)
| | - Mariafrancesca Scalise
- Laboratory of Biochemistry, Molecular Biotechnology and Molecular Biology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (M.G.); (L.C.); (M.S.)
| | - Ivano Eberini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Cesare Indiveri
- Laboratory of Biochemistry, Molecular Biotechnology and Molecular Biology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (M.G.); (L.C.); (M.S.)
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via Amendola 122/O, 70126 Bari, Italy
| |
Collapse
|
36
|
Laddha AP, Dzielak L, Lewis C, Xue R, Manautou JE. Impact of non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) on the expression and function of hepatobiliary transporters: A comprehensive mechanistic review. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167037. [PMID: 38295624 DOI: 10.1016/j.bbadis.2024.167037] [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: 09/07/2023] [Revised: 01/11/2024] [Accepted: 01/20/2024] [Indexed: 02/02/2024]
Abstract
The liver plays a central role in the biotransformation and disposition of endogenous molecules and xenobiotics. In addition to drug-metabolizing enzymes, transporter proteins are key determinants of drug hepatic clearance. Hepatic transporters are transmembrane proteins that facilitate the movement of chemicals between sinusoidal blood and hepatocytes. Other drug transporters translocate molecules from hepatocytes into bile canaliculi for biliary excretion. The formers are known as basolateral, while the latter are known as canalicular transporters. Also, these transporters are classified into two super-families, the solute carrier transporter (SLC) and the adenosine triphosphate (ATP)-binding cassette (ABC) transporter. The expression and function of transporters involve complex regulatory mechanisms, which are contributing factors to interindividual variability in drug pharmacokinetics and disposition. A considerable number of liver diseases are known to alter the expression and function of drug transporters. Among them, non-alcoholic fatty liver disease (NAFLD) is a chronic condition with a rapidly increasing incidence worldwide. NAFLD, recently reclassified as metabolic dysfunction-associated steatotic liver disease (MASLD), is a disease continuum that includes steatosis with or without mild inflammation (NASH), and potentially neuroinflammatory pathology. NASH is additionally characterized by the presence of hepatocellular injury. During NAFLD and NASH, drug transporters exhibit altered expression and function, leading to altered drug pharmacokinetics and pharmacodynamics, thus increasing the risk of adverse drug reactions. The purpose of the present review is to provide comprehensive mechanistic information on the expression and function of hepatic transporters under fatty liver conditions and hence, the impact on the pharmacokinetic profiles of certain drugs from the available pre-clinical and clinical literature.
Collapse
Affiliation(s)
- Ankit P Laddha
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Lindsey Dzielak
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA; Non-Clinical Drug Safety (NDS) Department, Boehringer Ingelheim Pharmaceutical Co., Ridgefield, CT, USA
| | - Cedric Lewis
- Non-Clinical Drug Safety (NDS) Department, Boehringer Ingelheim Pharmaceutical Co., Ridgefield, CT, USA
| | - Raymond Xue
- Charles River Laboratories, Inc., Shrewsbury, MA, USA
| | - José E Manautou
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
37
|
Madison J, Wilhelm K, Meehan DT, Gratton MA, Vosik D, Samuelson G, Ott M, Fascianella J, Nelson N, Cosgrove D. Ramipril therapy in integrin α1-null, autosomal recessive Alport mice triples lifespan: mechanistic clues from RNA-seq analysis. J Pathol 2024; 262:296-309. [PMID: 38129319 PMCID: PMC10872630 DOI: 10.1002/path.6231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 10/10/2023] [Accepted: 10/29/2023] [Indexed: 12/23/2023]
Abstract
The standard of care for patients with Alport syndrome (AS) is angiotensin-converting enzyme (ACE) inhibitors. In autosomal recessive Alport (ARAS) mice, ACE inhibitors double lifespan. We previously showed that deletion of Itga1 in Alport mice [double-knockout (DKO) mice] increased lifespan by 50%. This effect seemed dependent on the prevention of laminin 211-mediated podocyte injury. Here, we treated DKO mice with vehicle or ramipril starting at 4 weeks of age. Proteinuria and glomerular filtration rates were measured at 5-week intervals. Glomeruli were analyzed for laminin 211 deposition in the glomerular basement membrane (GBM) and GBM ultrastructure was analyzed using transmission electron microscopy (TEM). RNA sequencing (RNA-seq) was performed on isolated glomeruli at all time points and the results were compared with cultured podocytes overlaid (or not) with recombinant laminin 211. Glomerular filtration rate declined in ramipril-treated DKO mice between 30 and 35 weeks. Proteinuria followed these same patterns with normalization of foot process architecture in ramipril-treated DKO mice. RNA-seq revealed a decline in the expression of Foxc2, nephrin (Nphs1), and podocin (Nphs2) mRNAs, which was delayed in the ramipril-treated DKO mice. GBM accumulation of laminin 211 was delayed in ramipril-treated DKO mice, likely due to a role for α1β1 integrin in CDC42 activation in Alport mesangial cells, which is required for mesangial filopodial invasion of the subendothelial spaces of the glomerular capillary loops. Ramipril synergized with Itga1 knockout, tripling lifespan compared with untreated ARAS mice. © 2023 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jacob Madison
- Boys Town National Research Hospital, Omaha, NE, USA
| | - Kevin Wilhelm
- Boys Town National Research Hospital, Omaha, NE, USA
| | | | | | - Denise Vosik
- Boys Town National Research Hospital, Omaha, NE, USA
| | | | - Megan Ott
- Boys Town National Research Hospital, Omaha, NE, USA
| | | | - Noa Nelson
- Boys Town National Research Hospital, Omaha, NE, USA
| | | |
Collapse
|
38
|
Chrysopoulou M, Rinschen MM. Metabolic Rewiring and Communication: An Integrative View of Kidney Proximal Tubule Function. Annu Rev Physiol 2024; 86:405-427. [PMID: 38012048 DOI: 10.1146/annurev-physiol-042222-024724] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The kidney proximal tubule is a key organ for human metabolism. The kidney responds to stress with altered metabolite transformation and perturbed metabolic pathways, an ultimate cause for kidney disease. Here, we review the proximal tubule's metabolic function through an integrative view of transport, metabolism, and function, and embed it in the context of metabolome-wide data-driven research. Function (filtration, transport, secretion, and reabsorption), metabolite transformation, and metabolite signaling determine kidney metabolic rewiring in disease. Energy metabolism and substrates for key metabolic pathways are orchestrated by metabolite sensors. Given the importance of renal function for the inner milieu, we also review metabolic communication routes with other organs. Exciting research opportunities exist to understand metabolic perturbation of kidney and proximal tubule function, for example, in hypertension-associated kidney disease. We argue that, based on the integrative view outlined here, kidney diseases without genetic cause should be approached scientifically as metabolic diseases.
Collapse
Affiliation(s)
| | - Markus M Rinschen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark;
- III. Department of Medicine and Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark
| |
Collapse
|
39
|
Yan Y, Liu H, Abedini A, Sheng X, Palmer M, Li H, Susztak K. Unraveling the epigenetic code: human kidney DNA methylation and chromatin dynamics in renal disease development. Nat Commun 2024; 15:873. [PMID: 38287030 PMCID: PMC10824731 DOI: 10.1038/s41467-024-45295-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 01/19/2024] [Indexed: 01/31/2024] Open
Abstract
Epigenetic changes may fill a critical gap in our understanding of kidney disease development, as they not only reflect metabolic changes but are also preserved and transmitted during cell division. We conducted a genome-wide cytosine methylation analysis of 399 human kidney samples, along with single-nuclear open chromatin analysis on over 60,000 cells from 14 subjects, including controls, and diabetes and hypertension attributed chronic kidney disease (CKD) patients. We identified and validated differentially methylated positions associated with disease states, and discovered that nearly 30% of these alterations were influenced by underlying genetic variations, including variants known to be associated with kidney disease in genome-wide association studies. We also identified regions showing both methylation and open chromatin changes. These changes in methylation and open chromatin significantly associated gene expression changes, most notably those playing role in metabolism and expressed in proximal tubules. Our study further demonstrated that methylation risk scores (MRS) can improve disease state annotation and prediction of kidney disease development. Collectively, our results suggest a causal relationship between epigenetic changes and kidney disease pathogenesis, thereby providing potential pathways for the development of novel risk stratification methods.
Collapse
Affiliation(s)
- Yu Yan
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
- Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
| | - Hongbo Liu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
- Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
| | - Amin Abedini
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
- Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
| | - Xin Sheng
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
- Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
| | - Matthew Palmer
- Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
- Department of Epidemiology and Biostatistics, Perelman School of Medicine, Philadelphia, PA, 19014, USA
| | - Hongzhe Li
- Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA
- Department of Pathology, Perelman School of Medicine, Philadelphia, PA, 19014, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA.
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA.
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA.
- Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19014, USA.
| |
Collapse
|
40
|
Williams LA, Hamilton MC, Edin ML, Lih FB, Eccles-Miller JA, Tharayil N, Leonard E, Baldwin WS. Increased Perfluorooctanesulfonate (PFOS) Toxicity and Accumulation Is Associated with Perturbed Prostaglandin Metabolism and Increased Organic Anion Transport Protein (OATP) Expression. TOXICS 2024; 12:106. [PMID: 38393201 PMCID: PMC10893382 DOI: 10.3390/toxics12020106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/12/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024]
Abstract
Perfluorooctanesulfonate (PFOS) is a widespread environmental pollutant with a long half-life and clearly negative outcomes on metabolic diseases such as fatty liver disease and diabetes. Male and female Cyp2b-null and humanized CYP2B6-transgenic (hCYP2B6-Tg) mice were treated with 0, 1, or 10 mg/kg/day PFOS for 21 days, and surprisingly it was found that PFOS was retained at greater concentrations in the serum and liver of hCYP2B6-Tg mice than those of Cyp2b-null mice, with greater differences in the females. Thus, Cyp2b-null and hCYP2B6-Tg mice provide new models for investigating individual mechanisms for PFOS bioaccumulation and toxicity. Overt toxicity was greater in hCYP2B6-Tg mice (especially females) as measured by mortality; however, steatosis occurred more readily in Cyp2b-null mice despite the lower PFOS liver concentrations. Targeted lipidomics and transcriptomics from PFOS-treated Cyp2b-null and hCYP2B6-Tg mouse livers were performed and compared to PFOS retention and serum markers of toxicity using PCA. Several oxylipins, including prostaglandins, thromboxanes, and docosahexaenoic acid metabolites, are associated or inversely associated with PFOS toxicity. Both lipidomics and transcriptomics indicate PFOS toxicity is associated with PPAR activity in all models. GO terms associated with reduced steatosis were sexually dimorphic with lipid metabolism and transport increased in females and circadian rhythm associated genes increased in males. However, we cannot rule out that steatosis was initially protective from PFOS toxicity. Moreover, several transporters are associated with increased retention, probably due to increased uptake. The strongest associations are the organic anion transport proteins (Oatp1a4-6) genes and a long-chain fatty acid transport protein (fatp1), enriched in female hCYP2B6-Tg mice. PFOS uptake was also reduced in cultured murine hepatocytes by OATP inhibitors. The role of OATP1A6 and FATP1 in PFOS transport has not been tested. In summary, Cyp2b-null and hCYP2B6-Tg mice provided unique models for estimating the importance of novel mechanisms in PFOS retention and toxicity.
Collapse
Affiliation(s)
- Lanie A. Williams
- Biological Sciences, Clemson University, Clemson, SC 29634, USA; (L.A.W.); (M.C.H.); (J.A.E.-M.)
| | - Matthew C. Hamilton
- Biological Sciences, Clemson University, Clemson, SC 29634, USA; (L.A.W.); (M.C.H.); (J.A.E.-M.)
| | - Matthew L. Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, Washington, NC 27709, USA; (M.L.E.); (F.B.L.)
| | - Fred B. Lih
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, Washington, NC 27709, USA; (M.L.E.); (F.B.L.)
| | - Jazmine A. Eccles-Miller
- Biological Sciences, Clemson University, Clemson, SC 29634, USA; (L.A.W.); (M.C.H.); (J.A.E.-M.)
| | - Nishanth Tharayil
- Plant and Environmental Sciences, Clemson University, Clemson, SC 29634, USA; (N.T.); (E.L.)
| | - Elizabeth Leonard
- Plant and Environmental Sciences, Clemson University, Clemson, SC 29634, USA; (N.T.); (E.L.)
| | - William S. Baldwin
- Biological Sciences, Clemson University, Clemson, SC 29634, USA; (L.A.W.); (M.C.H.); (J.A.E.-M.)
| |
Collapse
|
41
|
Gong C, Bertagnolli LN, Boulton DW, Coppola P. A Literature Review of Changes in Phase II Drug-Metabolizing Enzyme and Drug Transporter Expression during Pregnancy. Pharmaceutics 2023; 15:2624. [PMID: 38004602 PMCID: PMC10674389 DOI: 10.3390/pharmaceutics15112624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
The purpose of this literature review is to comprehensively summarize changes in the expression of phase II drug-metabolizing enzymes and drug transporters in both the pregnant woman and the placenta. Using PubMed®, a systematic search was conducted to identify literature relevant to drug metabolism and transport in pregnancy. PubMed was searched with pre-specified terms during the period of 26 May 2023 to 10 July 2023. The final dataset of 142 manuscripts was evaluated for evidence regarding the effect of gestational age and hormonal regulation on the expression of phase II enzymes (n = 16) and drug transporters (n = 38) in the pregnant woman and in the placenta. This comprehensive review exposes gaps in current knowledge of phase II enzyme and drug transporter localization, expression, and regulation during pregnancy, which emphasizes the need for further research. Moreover, the information collected in this review regarding phase II drug-metabolizing enzyme and drug transporter changes will aid in optimizing pregnancy physiologically based pharmacokinetic (PBPK) models to inform dose selection in the pregnant population.
Collapse
Affiliation(s)
- Christine Gong
- School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Lynn N. Bertagnolli
- AstraZeneca LP, Biopharmaceuticals R&D, Clinical Pharmacology & Safety Sciences, Clinical Pharmacology & Quantitative Pharmacology, Gaithersburg, MD 20878, USA
| | - David W. Boulton
- AstraZeneca LP, Biopharmaceuticals R&D, Clinical Pharmacology & Safety Sciences, Clinical Pharmacology & Quantitative Pharmacology, Gaithersburg, MD 20878, USA
| | - Paola Coppola
- AstraZeneca LP, Biopharmaceuticals R&D, Clinical Pharmacology & Safety Sciences, Clinical Pharmacology & Quantitative Pharmacology, Cambridge CB2 0AA, UK
| |
Collapse
|
42
|
Falconi CA, Fogaça-Ruiz F, da Silva JV, Neres-Santos RS, Sanz CL, Nakao LS, Stinghen AEM, Junho CVC, Carneiro-Ramos MS. Renocardiac Effects of p-Cresyl Sulfate Administration in Acute Kidney Injury Induced by Unilateral Ischemia and Reperfusion Injury In Vivo. Toxins (Basel) 2023; 15:649. [PMID: 37999512 PMCID: PMC10674368 DOI: 10.3390/toxins15110649] [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: 10/16/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
The precise mechanisms underlying the cardiovascular complications due to acute kidney injury (AKI) and the retention of uremic toxins like p-cresyl sulfate (PCS) remain incompletely understood. The objective of this study was to evaluate the renocardiac effects of PCS administration in animals subjected to AKI induced by ischemia and reperfusion (IR) injury. C57BL6 mice were subjected to distinct protocols: (i) administration with PCS (20, 40, or 60 mg/L/day) for 15 days and (ii) AKI due to unilateral IR injury associated with PCS administration for 15 days. The 20 mg/L dose of PCS led to a decrease in renal mass, an increase in the gene expression of Cystatin C and kidney injury molecule 1 (KIM-1), and a decrease in the α-actin in the heart. During AKI, PCS increased the renal injury biomarkers compared to control; however, it did not exacerbate these markers. Furthermore, PCS did not enhance the cardiac hypertrophy observed after 15 days of IR. An increase, but not potentialized, in the cardiac levels of interleukin (IL)-1β and IL-6 in the IR group treated with PCS, as well as in the injured kidney, was also noticed. In short, PCS administration did not intensify kidney injury, inflammation, and cardiac outcomes after AKI.
Collapse
Affiliation(s)
- Carlos Alexandre Falconi
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-170, SP, Brazil; (C.A.F.); (F.F.-R.); (J.V.d.S.); (R.S.N.-S.)
| | - Fernanda Fogaça-Ruiz
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-170, SP, Brazil; (C.A.F.); (F.F.-R.); (J.V.d.S.); (R.S.N.-S.)
| | - Jéssica Verônica da Silva
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-170, SP, Brazil; (C.A.F.); (F.F.-R.); (J.V.d.S.); (R.S.N.-S.)
| | - Raquel Silva Neres-Santos
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-170, SP, Brazil; (C.A.F.); (F.F.-R.); (J.V.d.S.); (R.S.N.-S.)
| | - Carmen Lucía Sanz
- Department of Basic Pathology, Universidade Federal do Paraná, Curitiba 81530-000, PR, Brazil; (C.L.S.); (L.S.N.)
| | - Lia Sumie Nakao
- Department of Basic Pathology, Universidade Federal do Paraná, Curitiba 81530-000, PR, Brazil; (C.L.S.); (L.S.N.)
| | - Andréa Emília Marques Stinghen
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, PR, Brazil;
| | - Carolina Victoria Cruz Junho
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-170, SP, Brazil; (C.A.F.); (F.F.-R.); (J.V.d.S.); (R.S.N.-S.)
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Marcela Sorelli Carneiro-Ramos
- Laboratory of Cardiovascular Immunology, Center of Natural and Human Sciences (CCNH), Federal University of ABC, Santo André 09210-170, SP, Brazil; (C.A.F.); (F.F.-R.); (J.V.d.S.); (R.S.N.-S.)
| |
Collapse
|
43
|
Hou X, Wang Y, Yang Y, Xiao Z. Discovery of Novel Biphenyl Carboxylic Acid Derivatives as Potent URAT1 Inhibitors. Molecules 2023; 28:7415. [PMID: 37959834 PMCID: PMC10648204 DOI: 10.3390/molecules28217415] [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: 10/14/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Urate transporter 1 (URAT1) is a clinically validated target for the treatment of hyperuricemia and gout. Due to the absence of protein structures, the molecular design of new URAT1 inhibitors generally resorts to ligand-based approaches. Two series of biphenyl carboxylic acids were designed based on the structures of URAT1 inhibitors Epaminurad and Telmisartan via a strategy of pharmacophore fusion. Fifty-one novel compounds were synthesized and most of them showed obvious inhibition against human URAT1. A1 and B21 were identified as the most potent URAT1 inhibitors in series A and B, respectively. They exhibited IC50 values of 0.93 μM and 0.17 μM, which were comparable or superior to the clinical uricosuric drug benzbromarone. The results confirmed the effectiveness of ligand-based approaches in identifying novel and potent URAT1 inhibitors.
Collapse
Affiliation(s)
| | | | | | - Zhiyan Xiao
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
44
|
Lai Q, Zhu X, Zhang L, Kou J, Liu F, Yu B, Li F. Inhibition of OAT1/3 and CMPF uptake attenuates myocardial ischemia-induced chronic heart failure via decreasing fatty acid oxidation and the therapeutic effects of ruscogenin. Transl Res 2023; 261:1-15. [PMID: 37315712 DOI: 10.1016/j.trsl.2023.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/18/2023] [Accepted: 06/08/2023] [Indexed: 06/16/2023]
Abstract
Chronic heart failure (CHF) as a long-term disease is highly prevalent in elder people worldwide. Early diagnosis and treatments are crucial for preventing the development of CHF. Herein, we aimed to identify novel diagnostic biomarker, therapeutic target and drug for CHF. Untargeted metabolomic analysis has been used to characterize the different metabolomic profile between CHF patients and healthy people. Meanwhile, the targeted metabolomic study demonstrated the elevation of 3-carboxy-4-methyl-5-propyl-2-furanpropanoic acid (CMPF) in the serum of CHF patients and coronary artery ligation-induced CHF mice. Subsequently, we firstly observed that elevation of CMPF impaired cardiac function and aggravated myocardial injury by enhancing fatty acid oxidation (FAO). Interestingly, inhibition of responsible transporters organic anion transporter 1/3 (OAT1/3) has been found to decrease the CMPF level, and suppress FAO-related key protein expressions including peroxisome proliferator-activated receptor alpha, peroxisome proliferative activated receptor-α, carnitine palmitoyl transferase 1, and malonyl CoA decarboxylase in coronary artery ligation-induced CHF mice. Meanwhile, the inhibitor of OAT1/3 presented an excellent improvement in cardiac function and histological injury. Based on the above findings, molecular docking was adopted to screen the potential therapeutic drug targeting OAT1/3, and ruscogenin (RUS) exhibited a great binding affinity with OAT1 and OAT3. Next, it was verified that RUS could remarkedly decrease the expression of OAT1/3 and CMPF levels in heart tissue of CHF mice, as well as suppress the expression of FAO-related proteins. What's more, RUS can effectively improve cardiac function, myocardial fibrosis and morphological damage. Collectively, this study provided a potential metabolic marker CMPF and novel target OAT1/3 for CHF, which were demonstrated to be involved in FAO. And RUS was identified as a potential anti-FAO drug for CHF by regulating OAT1/3.
Collapse
Affiliation(s)
- Qiong Lai
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xiaozhou Zhu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Lu Zhang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Junping Kou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Fuming Liu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Fang Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.
| |
Collapse
|
45
|
Thévenod F, Herbrechter R, Schlabs C, Pethe A, Lee WK, Wolff NA, Roussa E. Role of the SLC22A17/lipocalin-2 receptor in renal endocytosis of proteins/metalloproteins: a focus on iron- and cadmium-binding proteins. Am J Physiol Renal Physiol 2023; 325:F564-F577. [PMID: 37589051 DOI: 10.1152/ajprenal.00020.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 08/18/2023] Open
Abstract
The transmembrane protein SLC22A17 [or the neutrophil gelatinase-associated lipocalin/lipocalin-2 (LCN2)/24p3 receptor] is an atypical member of the SLC22 family of organic anion and cation transporters: it does not carry typical substrates of SLC22 transporters but mediates receptor-mediated endocytosis (RME) of LCN2. One important task of the kidney is the prevention of urinary loss of proteins filtered by the glomerulus by bulk reabsorption of multiple ligands via megalin:cubilin:amnionless-mediated endocytosis in the proximal tubule (PT). Accordingly, overflow, glomerular, or PT damage, as in Fanconi syndrome, results in proteinuria. Strikingly, up to 20% of filtered proteins escape the PT under physiological conditions and are reabsorbed by the distal nephron. The renal distal tubule and collecting duct express SLC22A17, which mediates RME of filtered proteins that evade the PT but with limited capacity to prevent proteinuria under pathological conditions. The kidney also prevents excretion of filtered essential and nonessential transition metals, such as iron or cadmium, respectively, that are largely bound to proteins with high affinity, e.g., LCN2, transferrin, or metallothionein, or low affinity, e.g., microglobulins or albumin. Hence, increased uptake of transition metals may cause nephrotoxicity. Here, we assess the literature on SLC22A17 structure, topology, tissue distribution, regulation, and assumed functions, emphasizing renal SLC22A17, which has relevance for physiology, pathology, and nephrotoxicity due to the accumulation of proteins complexed with transition metals, e.g., cadmium or iron. Other putative renal functions of SLC22A17, such as its contribution to osmotic stress adaptation, protection against urinary tract infection, or renal carcinogenesis, are discussed.
Collapse
Affiliation(s)
- Frank Thévenod
- Institute for Physiology, Pathophysiology and Toxicology, Center for Biomedical Education and Research, Witten/Herdecke University, Witten, Germany
- Physiology and Pathophysiology of Cells and Membranes, Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Robin Herbrechter
- Institute for Physiology, Pathophysiology and Toxicology, Center for Biomedical Education and Research, Witten/Herdecke University, Witten, Germany
| | - Carolin Schlabs
- Institute for Physiology, Pathophysiology and Toxicology, Center for Biomedical Education and Research, Witten/Herdecke University, Witten, Germany
| | - Abhishek Pethe
- Department of Molecular Embryology, Faculty of Medicine, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Wing-Kee Lee
- Physiology and Pathophysiology of Cells and Membranes, Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Natascha A Wolff
- Institute for Physiology, Pathophysiology and Toxicology, Center for Biomedical Education and Research, Witten/Herdecke University, Witten, Germany
| | - Eleni Roussa
- Department of Molecular Embryology, Faculty of Medicine, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
46
|
Parker JL, Kato T, Kuteyi G, Sitsel O, Newstead S. Molecular basis for selective uptake and elimination of organic anions in the kidney by OAT1. Nat Struct Mol Biol 2023; 30:1786-1793. [PMID: 37482561 PMCID: PMC10643130 DOI: 10.1038/s41594-023-01039-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/20/2023] [Indexed: 07/25/2023]
Abstract
In mammals, the kidney plays an essential role in maintaining blood homeostasis through the selective uptake, retention or elimination of toxins, drugs and metabolites. Organic anion transporters (OATs) are responsible for the recognition of metabolites and toxins in the nephron and their eventual urinary excretion. Inhibition of OATs is used therapeutically to improve drug efficacy and reduce nephrotoxicity. The founding member of the renal organic anion transporter family, OAT1 (also known as SLC22A6), uses the export of α-ketoglutarate (α-KG), a key intermediate in the Krebs cycle, to drive selective transport and is allosterically regulated by intracellular chloride. However, the mechanisms linking metabolite cycling, drug transport and intracellular chloride remain obscure. Here, we present cryogenic-electron microscopy structures of OAT1 bound to α-KG, the antiviral tenofovir and clinical inhibitor probenecid, used in the treatment of Gout. Complementary in vivo cellular assays explain the molecular basis for α-KG driven drug elimination and the allosteric regulation of organic anion transport in the kidney by chloride.
Collapse
Affiliation(s)
- Joanne L Parker
- Department of Biochemistry, University of Oxford, Oxford, UK.
- The Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK.
| | - Takafumi Kato
- Department of Biochemistry, University of Oxford, Oxford, UK.
- The Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK.
| | - Gabriel Kuteyi
- Department of Biochemistry, University of Oxford, Oxford, UK
- The Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Oleg Sitsel
- Department of Biochemistry, University of Oxford, Oxford, UK
- Max Planck Institute of Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Simon Newstead
- Department of Biochemistry, University of Oxford, Oxford, UK.
- The Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK.
| |
Collapse
|
47
|
Chang L. OAT1 structures reveal insights into drug transport in the kidney. Nat Struct Mol Biol 2023; 30:1615-1617. [PMID: 37957303 DOI: 10.1038/s41594-023-01144-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Affiliation(s)
- Leifu Chang
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
48
|
Dou T, Lian T, Shu S, He Y, Jiang J. The substrate and inhibitor binding mechanism of polyspecific transporter OAT1 revealed by high-resolution cryo-EM. Nat Struct Mol Biol 2023; 30:1794-1805. [PMID: 37845412 PMCID: PMC11406556 DOI: 10.1038/s41594-023-01123-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 09/12/2023] [Indexed: 10/18/2023]
Abstract
Organic anion transporters (OATs) of the SLC22 family have crucial roles in the transport of organic anions, including metabolites and therapeutic drugs, and in transporter-mediated drug-drug interactions. In the kidneys, OATs facilitate the elimination of metabolic waste products and xenobiotics. However, their transport activities can lead to the accumulation of certain toxic compounds within cells, causing kidney damage. Moreover, OATs are important drug targets, because their inhibition modulates the elimination or retention of substrates linked to diseases. Despite extensive research on OATs, the molecular basis of their substrate and inhibitor binding remains poorly understood. Here we report the cryo-EM structures of rat OAT1 (also known as SLC22A6) and its complexes with para-aminohippuric acid and probenecid at 2.1, 2.8 and 2.9 Å resolution, respectively. Our findings reveal a highly conserved substrate binding mechanism for SLC22 transporters, wherein four aromatic residues form a cage to accommodate the polyspecific binding of diverse compounds.
Collapse
Affiliation(s)
- Tongyi Dou
- Laboratory of Membrane Proteins and Structural Biology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Tengfei Lian
- Laboratory of Membrane Proteins and Structural Biology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Shi Shu
- Laboratory of Membrane Proteins and Structural Biology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Yi He
- Fermentation Facility, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Jiansen Jiang
- Laboratory of Membrane Proteins and Structural Biology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA.
| |
Collapse
|
49
|
Zeng YC, Sobti M, Quinn A, Smith NJ, Brown SHJ, Vandenberg JI, Ryan RM, O'Mara ML, Stewart AG. Structural basis of promiscuous substrate transport by Organic Cation Transporter 1. Nat Commun 2023; 14:6374. [PMID: 37821493 PMCID: PMC10567722 DOI: 10.1038/s41467-023-42086-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 09/29/2023] [Indexed: 10/13/2023] Open
Abstract
Organic Cation Transporter 1 (OCT1) plays a crucial role in hepatic metabolism by mediating the uptake of a range of metabolites and drugs. Genetic variations can alter the efficacy and safety of compounds transported by OCT1, such as those used for cardiovascular, oncological, and psychological indications. Despite its importance in drug pharmacokinetics, the substrate selectivity and underlying structural mechanisms of OCT1 remain poorly understood. Here, we present cryo-EM structures of full-length human OCT1 in the inward-open conformation, both ligand-free and drug-bound, indicating the basis for its broad substrate recognition. Comparison of our structures with those of outward-open OCTs provides molecular insight into the alternating access mechanism of OCTs. We observe that hydrophobic gates stabilize the inward-facing conformation, whereas charge neutralization in the binding pocket facilitates the release of cationic substrates. These findings provide a framework for understanding the structural basis of the promiscuity of drug binding and substrate translocation in OCT1.
Collapse
Affiliation(s)
- Yi C Zeng
- Molecular, Structural and Computational Biology Division, The Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia.
| | - Meghna Sobti
- Molecular, Structural and Computational Biology Division, The Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Ada Quinn
- Australian Institute of Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Nicola J Smith
- School of Biomedical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
| | - Simon H J Brown
- School of Chemistry and Molecular Bioscience, Molecular Horizons, and Australian Research Council Centre for Cryo-electron Microscopy of Membrane Proteins, University of Wollongong, Wollongong, NSW, Australia
| | - Jamie I Vandenberg
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
- Molecular Cardiology and Biophysics Division, The Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Renae M Ryan
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Megan L O'Mara
- Australian Institute of Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Alastair G Stewart
- Molecular, Structural and Computational Biology Division, The Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
50
|
Gillard BT, Amor N, Iraizoz FA, Pauža AG, Campbell C, Greenwood MP, Alagaili AN, Murphy D. Mobilisation of jerboa kidney gene networks during dehydration and opportunistic rehydration. iScience 2023; 26:107574. [PMID: 37664605 PMCID: PMC10470305 DOI: 10.1016/j.isci.2023.107574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/30/2023] [Accepted: 08/04/2023] [Indexed: 09/05/2023] Open
Abstract
Desert animals have evolved systems that enable them to thrive under dry conditions. Focusing on the kidney, we have investigated the transcriptomic adaptations that enable a desert rodent, the Lesser Egyptian Jerboa (Jaculus jaculus), to withstand water deprivation and opportunistic rehydration. Analysis of the whole kidney transcriptome showed many differentially expressed genes in the Jerboa kidney, 6.4% of genes following dehydration and an even greater number (36.2%) following rehydration compared to control. Genes correlated with the rehydration condition included many ribosomal protein coding genes suggesting a concerted effort to accelerate protein synthesis when water is made available. We identify an increase in TGF-beta signaling antagonists in dehydration (e.g., GREM2). We also describe expression of multiple aquaporin and solute carrier transporters mapped to specific nephron segments. The desert adapted renal transcriptome presented here is a valuable resource to expand our understanding of osmoregulation beyond that derived from model organisms.
Collapse
Affiliation(s)
- Benjamin T. Gillard
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, England
| | - Nabil Amor
- LR18ES05, Laboratory of Biodiversity, Parasitology and Ecology of Aquatic Ecosystems, Department of Biology - Faculty of Sciences of Tunis, University of Tunis El Manar, Tunisia
| | - Fernando Alvira Iraizoz
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, England
| | - Audrys G. Pauža
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, England
| | - Colin Campbell
- Department of Engineering Mathematics, Ada Lovelace Building, University of Bristol, Bristol, England
| | - Michael P. Greenwood
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, England
| | | | - David Murphy
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, England
| |
Collapse
|