1
|
Anthonymuthu S, Sabui S, Manzon KI, Sheikh A, Fleckenstein JM, Said HM. Bacterial lipopolysaccharide inhibits free thiamin uptake along the intestinal tract via interference with membrane expression of thiamin transporters 1 and 2. Am J Physiol Cell Physiol 2024; 327:C1163-C1177. [PMID: 39246143 PMCID: PMC11559647 DOI: 10.1152/ajpcell.00570.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024]
Abstract
This study examined the effect of exposure of small and large intestinal epithelial cells to the bacterial lipopolysaccharide (LPS) on uptake of free form of vitamin B1, i.e., thiamin. The intestinal tract encounters two sources of thiamin: diet and the gut microbiota. Absorption of thiamin in both the small and large intestine occurs via a carrier-mediated process that involves thiamin transporters 1 and 2 (THTR-1 and -2). Complementary in vitro (human duodenal epithelial HuTu-80 cells and human colonic epithelial NCM460 cells), in vivo (mice), and ex vivo (human primary differentiated enteroid and colonoid monolayers) models were used. The results showed that exposure to LPS causes a significant inhibition in carrier-mediated [3H]-thiamin uptake by small and large intestinal epithelia, with no change in the levels of expression of THTR-1 and -2 mRNAs and their total cellular proteins. However, a significant decrease in the fractions of the THTR-1 and -2 proteins that are expressed at the cell membranes of these epithelial cells was observed. These effects of LPS appeared to involve a protein kinase A (PKA) signaling pathway as activating this pathway caused a reversal in the inhibition of thiamin uptake and level of expression of its transporters at the cell membrane. These findings demonstrate that exposure of gut epithelia to LPS (a situation that occurs under different pathological conditions) leads to inhibition in thiamin uptake due to a decrease in level of expression of its transporters at the cell membrane that is likely mediated via a PKA signaling pathway. NEW & NOTEWORTHY This study shows that the exposure of gut epithelial cells to bacterial LPS negatively impact the uptake process of the free form of vitamin B1 (i.e., thiamin). This appears to be mediated via suppression in the level of thiamin transporters 1 and 2 (THTR-1 and -2) expression at the cell membrane and involves a protein kinase A (PKA) signaling pathway.
Collapse
Affiliation(s)
- Selvaraj Anthonymuthu
- Departments of Physiology/Biophysics, School of Medicine, University of California, Irvine, California, United States
| | - Subrata Sabui
- Departments of Physiology/Biophysics, School of Medicine, University of California, Irvine, California, United States
- Department of Medical Research, Tibor Rubin VA Medical Center, Long Beach, California, United States
| | - Kameron Isaiah Manzon
- Departments of Physiology/Biophysics, School of Medicine, University of California, Irvine, California, United States
| | - Alaullah Sheikh
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - James M Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Medicine Service, Infectious Disease Section, Veterans Affairs Health Care System, St. Louis, Missouri, United States
| | - Hamid M Said
- Departments of Physiology/Biophysics, School of Medicine, University of California, Irvine, California, United States
- Department of Medicine, School of Medicine, University of California, Irvine, California, United States
- Department of Medical Research, Tibor Rubin VA Medical Center, Long Beach, California, United States
| |
Collapse
|
2
|
Feldman HH, Luchsinger JA, Léger GC, Taylor C, Jacobs DM, Salmon DP, Edland SD, Messer K, Revta C, Flowers SA, Jones KS, Koulman A, Yarasheski KE, Verghese PB, Venkatesh V, Zetterberg H, Durant J, Lupo JL, Gibson GE. Protocol for a seamless phase 2A-phase 2B randomized double-blind placebo-controlled trial to evaluate the safety and efficacy of benfotiamine in patients with early Alzheimer's disease (BenfoTeam). PLoS One 2024; 19:e0302998. [PMID: 38809849 PMCID: PMC11135745 DOI: 10.1371/journal.pone.0302998] [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: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Benfotiamine provides an important novel therapeutic direction in Alzheimer's disease (AD) with possible additive or synergistic effects to amyloid targeting therapeutic approaches. OBJECTIVE To conduct a seamless phase 2A-2B proof of concept trial investigating tolerability, safety, and efficacy of benfotiamine, a prodrug of thiamine, as a first-in-class small molecule oral treatment for early AD. METHODS This is the protocol for a randomized, double-blind, placebo-controlled 72-week clinical trial of benfotiamine in 406 participants with early AD. Phase 2A determines the highest safe and well-tolerated dose of benfotiamine to be carried forward to phase 2B. During phase 2A, real-time monitoring of pre-defined safety stopping criteria in the first approximately 150 enrollees will help determine which dose (600 mg or 1200 mg) will be carried forward into phase 2B. The phase 2A primary analysis will test whether the rate of tolerability events (TEs) is unacceptably high in the high-dose arm compared to placebo. The primary safety endpoint in phase 2A is the rate of TEs compared between active and placebo arms, at each dose. The completion of phase 2A will seamlessly transition to phase 2B without pausing or stopping the trial. Phase 2B will assess efficacy and longer-term safety of benfotiamine in a larger group of participants through 72 weeks of treatment, at the selected dose. The co-primary efficacy endpoints in phase 2B are CDR-Sum of Boxes and ADAS-Cog13. Secondary endpoints include safety and tolerability measures; pharmacokinetic measures of thiamine and its esters, erythrocyte transketolase activity as blood markers of efficacy of drug delivery; ADCS-ADL-MCI; and MoCA. CONCLUSION The BenfoTeam trial utilizes an innovative seamless phase 2A-2B design to achieve proof of concept. It includes an adaptive dose decision rule, thus optimizing exposure to the highest and best-tolerated dose. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT06223360, registered on January 25, 2024. https://classic.clinicaltrials.gov/ct2/show/NCT06223360.
Collapse
Affiliation(s)
- Howard H. Feldman
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Alzheimer’s Disease Cooperative Study, University of California San Diego, La Jolla, California, United States of America
- Shiley-Marcos Alzheimer’s Disease Research Center, La Jolla, California, United States of America
| | - José A. Luchsinger
- Columbia University Irving Medical Center, New York, New York, United States of America
| | - Gabriel C. Léger
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Alzheimer’s Disease Cooperative Study, University of California San Diego, La Jolla, California, United States of America
| | - Curtis Taylor
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Alzheimer’s Disease Cooperative Study, University of California San Diego, La Jolla, California, United States of America
| | - Diane M. Jacobs
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Alzheimer’s Disease Cooperative Study, University of California San Diego, La Jolla, California, United States of America
- Shiley-Marcos Alzheimer’s Disease Research Center, La Jolla, California, United States of America
| | - David P. Salmon
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Alzheimer’s Disease Cooperative Study, University of California San Diego, La Jolla, California, United States of America
- Shiley-Marcos Alzheimer’s Disease Research Center, La Jolla, California, United States of America
| | - Steven D. Edland
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Alzheimer’s Disease Cooperative Study, University of California San Diego, La Jolla, California, United States of America
- Shiley-Marcos Alzheimer’s Disease Research Center, La Jolla, California, United States of America
| | - Karen Messer
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Alzheimer’s Disease Cooperative Study, University of California San Diego, La Jolla, California, United States of America
| | - Carolyn Revta
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Alzheimer’s Disease Cooperative Study, University of California San Diego, La Jolla, California, United States of America
| | - Sarah A. Flowers
- University of Virginia, Charlottesville, Virginia, United States of America
| | - Kerry S. Jones
- Nutritional Biomarker Laboratory, MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Albert Koulman
- Nutritional Biomarker Laboratory, MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | | | | | - Venky Venkatesh
- C2N Diagnostics, St. Louis, Missouri, United States of America
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States of America
| | - January Durant
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Alzheimer’s Disease Cooperative Study, University of California San Diego, La Jolla, California, United States of America
| | - Jody-Lynn Lupo
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Alzheimer’s Disease Cooperative Study, University of California San Diego, La Jolla, California, United States of America
| | - Gary E. Gibson
- Burke Neurological Institute, Weill Cornell Medicine, White Plains, New York, United States of America
| | | |
Collapse
|
3
|
Ramamoorthy K, Sabui S, Manzon KI, Balamurugan AN, Said HM. miR-122-5p is involved in posttranscriptional regulation of the mitochondrial thiamin pyrophosphate transporter ( SLC25A19) in pancreatic acinar cells. Am J Physiol Gastrointest Liver Physiol 2023; 325:G347-G355. [PMID: 37529835 PMCID: PMC10642993 DOI: 10.1152/ajpgi.00106.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/19/2023] [Accepted: 07/26/2023] [Indexed: 08/03/2023]
Abstract
Thiamin (vitamin B1) plays a vital role in cellular energy metabolism/ATP production. Pancreatic acinar cells (PACs) obtain thiamin from circulation and convert it to thiamin pyrophosphate (TPP) in the cytoplasm. TPP is then taken up by the mitochondria via a carrier-mediated process that involves the mitochondrial TPP transporter (MTPPT; encoded by the gene SLC25A19). We have previously characterized different aspects of the mitochondrial carrier-mediated TPP uptake process, but nothing is known about its possible regulation at the posttranscriptional level. We address this issue in the current investigations focusing on the role of miRNAs in this regulation. First, we subjected the human (and rat) 3'-untranslated region (3'-UTR) of the SLC25A19 to three in-silico programs, and all have identified putative binding sites for miR-122-5p. Transfecting pmirGLO-hSLC25A19 3'-UTR into rat PAC AR42J resulted in a significant reduction in luciferase activity compared with cells transfected with pmirGLO-empty vector. Mutating as well as truncating the putative miR-122-5p binding sites in the hSLC25A19 3'-UTR led to abrogation of inhibition in luciferase activity in PAC AR42J. Furthermore, transfecting/transducing PAC AR42J and human primary PACs with mimic of miR-122-5p led to a significant inhibition in the level of expression of the MTPPT mRNA and protein as well as in mitochondrial carrier-mediated TPP uptake. Conversely, transfecting PAC AR42J with an inhibitor of miR-122-5p increased MTPPT expression and function. These findings show, for the first time, that expression and function of the MTPPT in PACs are subject to posttranscriptional regulation by miR-122-5p.NEW & NOTEWORTHY This study shows that the expression and function of mitochondrial TPP transporter (MTPPT) are subject to posttranscriptional regulation by miRNA-122-5p in pancreatic acinar cells.
Collapse
Affiliation(s)
- Kalidas Ramamoorthy
- Department of Physiology/Biophysics, University of California, Irvine, California, United States
| | - Subrata Sabui
- Department of Physiology/Biophysics, University of California, Irvine, California, United States
- Department of Medical Research, Tibor Rubin Veterans Affairs Medical Center, Long Beach, California, United States
| | - Kameron I Manzon
- Department of Physiology/Biophysics, University of California, Irvine, California, United States
| | - Appakalai N Balamurugan
- Center for Clinical and Translational Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, United States
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Hamid M Said
- Department of Medicine, University of California, Irvine, California, United States
- Department of Physiology/Biophysics, University of California, Irvine, California, United States
- Department of Medical Research, Tibor Rubin Veterans Affairs Medical Center, Long Beach, California, United States
| |
Collapse
|
4
|
Sheikh A, Tumala B, Vickers TJ, Martin JC, Rosa BA, Sabui S, Basu S, Simoes RD, Mitreva M, Storer C, Tyksen E, Head RD, Beatty W, Said HM, Fleckenstein JM. Enterotoxigenic Escherichia coli heat-labile toxin drives enteropathic changes in small intestinal epithelia. Nat Commun 2022; 13:6886. [PMID: 36371425 PMCID: PMC9653437 DOI: 10.1038/s41467-022-34687-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/01/2022] [Indexed: 11/14/2022] Open
Abstract
Enterotoxigenic E. coli (ETEC) produce heat-labile (LT) and/or heat-stable (ST) enterotoxins, and commonly cause diarrhea in resource-poor regions. ETEC have been linked repeatedly to sequelae in children including enteropathy, malnutrition, and growth impairment. Although cellular actions of ETEC enterotoxins leading to diarrhea are well-established, their contributions to sequelae remain unclear. LT increases cellular cAMP to activate protein kinase A (PKA) that phosphorylates ion channels driving intestinal export of salt and water resulting in diarrhea. As PKA also modulates transcription of many genes, we interrogated transcriptional profiles of LT-treated intestinal epithelia. Here we show that LT significantly alters intestinal epithelial gene expression directing biogenesis of the brush border, the major site for nutrient absorption, suppresses transcription factors HNF4 and SMAD4 critical to enterocyte differentiation, and profoundly disrupts microvillus architecture and essential nutrient transport. In addition, ETEC-challenged neonatal mice exhibit substantial brush border derangement that is prevented by maternal vaccination with LT. Finally, mice repeatedly challenged with toxigenic ETEC exhibit impaired growth recapitulating the multiplicative impact of recurring ETEC infections in children. These findings highlight impacts of ETEC enterotoxins beyond acute diarrheal illness and may inform approaches to prevent major sequelae of these common infections including malnutrition that impact millions of children.
Collapse
Affiliation(s)
- Alaullah Sheikh
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Brunda Tumala
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Tim J Vickers
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - John C Martin
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Bruce A Rosa
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Subrata Sabui
- Departments of Medicine and Physiology/Biophysics, School of Medicine, University of California-Irvine, Irvine, CA, 92697, USA
- Department of Research, VA Medical Center, Long Beach, CA, 90822, USA
| | - Supratim Basu
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Rita D Simoes
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Makedonka Mitreva
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Chad Storer
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Erik Tyksen
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Richard D Head
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Wandy Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Hamid M Said
- Departments of Medicine and Physiology/Biophysics, School of Medicine, University of California-Irvine, Irvine, CA, 92697, USA
- Department of Research, VA Medical Center, Long Beach, CA, 90822, USA
| | - James M Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Infectious Diseases, Medicine Service, Veterans Affairs Saint Louis Health Care System, Saint Louis, MO, 63106, USA.
| |
Collapse
|
5
|
Gibson GE, Feldman HH, Zhang S, Flowers SA, Luchsinger JA. Pharmacological thiamine levels as a therapeutic approach in Alzheimer's disease. Front Med (Lausanne) 2022; 9:1033272. [PMID: 36275801 PMCID: PMC9585656 DOI: 10.3389/fmed.2022.1033272] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
of the study.
Collapse
Affiliation(s)
- Gary E. Gibson
- Weill Cornell Medicine, Brain and Mind Research Institute, Burke Neurological Institute, White Plains, NY, United States
| | - Howard H. Feldman
- Alzheimer's Disease Cooperative Study and Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - Sheng Zhang
- Proteomics and Metabolomics Facility, Institute of Biotechnology, Cornell University, Ithaca, NY, United States
| | - Sarah A. Flowers
- Department of Neuroscience, Georgetown University, Washington, DC, United States
| | - José A. Luchsinger
- Departments of Medicine and Epidemiology, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
6
|
Ramamoorthy K, Yoshimura R, Al-Juburi S, Anandam KY, Kapadia R, Alachkar A, Abbott GW, Said HM. Alzheimer's disease is associated with disruption in thiamin transport physiology: A potential role for neuroinflammation. Neurobiol Dis 2022; 171:105799. [PMID: 35750148 PMCID: PMC9744268 DOI: 10.1016/j.nbd.2022.105799] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/30/2022] [Accepted: 06/17/2022] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by Amyloid-β peptide (Aβ) containing plaques and cognitive deficits. The pathophysiology of AD also involves neuroinflammation. Vitamin B1 (thiamin) is indispensable for normal cellular energy metabolism. Thiamin homeostasis is altered in AD, and its deficiency is known to aggravate AD pathology. Little, however, is known about possible alterations in level of expression of thiamin transporters-1 and -2 (THTR-1 and -2) in the brain of AD, and whether pro-inflammatory cytokines affect thiamin uptake by brain cells. We addressed these issues using brain tissue samples [prefrontal cortex (PFC) and hippocampus (HIP)] from AD patients and from 5XFAD mouse model of AD, together with cultured human neuroblastoma SH-SY5Y cells as model. Our results revealed a significantly lower expression of both THTR-1 and THTR-2 in the PFC and HIP of AD patients and 5XFAD mouse model of AD compared to appropriate normal controls. Further, we found that exposure of the SH-SY5Y cells to pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) led to a significant inhibition in thiamin uptake. Focusing on IL-1β, we found the inhibition in thiamin uptake to be time-dependent and reversible; it was also associated with a substantial reduction in expression of THTR-1 (but not THTR-2) protein and mRNA as well as a decrease in promoter activity of the SLC19A2 gene (which encodes THTR-1). Finally, using transcriptomic analysis, we found that thiamin availability in SH-SY5Y cells caused changes in the expression of genes relevant to AD pathways. These studies demonstrate, for the first time, that thiamin transport physiology/molecular biology parameters are negatively impacted in AD brain and that pro-inflammatory cytokines inhibit thiamin uptake by neuroblastoma cells. The results also support a possible role for thiamin in the pathophysiology of AD.
Collapse
Affiliation(s)
- Kalidas Ramamoorthy
- Departments of Physiology & Biophysics, University of California, Irvine, CA 92697, United States of America
| | - Ryan Yoshimura
- Departments of Physiology & Biophysics, University of California, Irvine, CA 92697, United States of America
| | - Saleh Al-Juburi
- Departments of Physiology & Biophysics, University of California, Irvine, CA 92697, United States of America
| | - Kasin Y Anandam
- Departments of Physiology & Biophysics, University of California, Irvine, CA 92697, United States of America
| | - Rubina Kapadia
- Medicine, School of Medicine, University of California, Irvine, CA 92697, United States of America
| | - Amal Alachkar
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, United States of America
| | - Geoffrey W Abbott
- Departments of Physiology & Biophysics, University of California, Irvine, CA 92697, United States of America
| | - Hamid M Said
- Departments of Physiology & Biophysics, University of California, Irvine, CA 92697, United States of America; Medicine, School of Medicine, University of California, Irvine, CA 92697, United States of America; Department of Veteran Affairs, VA Medical Center, Long Beach, CA 90822, United States of America.
| |
Collapse
|
7
|
Sabui S, Ramamoorthy K, Romero JM, Simoes RD, Fleckenstein JM, Said HM. Hypoxia inhibits colonic uptake of the microbiota-generated forms of vitamin B1 via HIF-1α-mediated transcriptional regulation of their transporters. J Biol Chem 2022; 298:101562. [PMID: 34998824 PMCID: PMC8800108 DOI: 10.1016/j.jbc.2022.101562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/29/2021] [Accepted: 01/02/2022] [Indexed: 01/19/2023] Open
Abstract
Hypoxia exerts profound effects on cell physiology, but its effect on colonic uptake of the microbiota-generated forms of vitamin B1 (i.e., thiamin pyrophosphate [TPP] and free thiamine) has not been described. Here, we used human colonic epithelial NCM460 cells and human differentiated colonoid monolayers as in vitro and ex vivo models, respectively, and were subjected to either chamber (1% O2, 5% CO2, and 94% N2) or chemically (desferrioxamine; 250 μM)-induced hypoxia followed by determination of different physiological-molecular parameters. We showed that hypoxia causes significant inhibition in TPP and free thiamin uptake by colonic NCM460 cells and colonoid monolayers; it also caused a significant reduction in the expression of TPP (SLC44A4) and free thiamin (SLC19A2 and SLC19A3) transporters and in activity of their gene promoters. Furthermore, hypoxia caused a significant induction in levels of hypoxia-inducible transcription factor (HIF)-1α but not HIF-2α. Knocking down HIF-1α using gene-specific siRNAs in NCM460 cells maintained under hypoxic conditions, on the other hand, led to a significant reversal in the inhibitory effect of hypoxia on TPP and free thiamin uptake as well as on the expression of their transporters. Finally, a marked reduction in level of expression of the nuclear factors cAMP responsive element-binding protein 1 and gut-enriched Krüppel-like factor 4 (required for activity of SLC44A4 and SLC19A2 promoters, respectively) was observed under hypoxic conditions. In summary, hypoxia causes severe inhibition in colonic TPP and free thiamin uptake that is mediated at least in part via HIF-1α-mediated transcriptional mechanisms affecting their respective transporters.
Collapse
Affiliation(s)
- Subrata Sabui
- Department of Physiology and Biophysics, UCI, Irvine, California, USA; Department of Research, VA Medical Center, Long Beach, California, USA
| | | | - Jose M Romero
- Department of Research, VA Medical Center, Long Beach, California, USA; Department of Medicine, UCI, Irvine, California, USA
| | - Rita D Simoes
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - James M Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA; Department Medicine Service, Veterans Affairs Medical Center, St Louis, Missouri, USA
| | - Hamid M Said
- Department of Physiology and Biophysics, UCI, Irvine, California, USA; Department of Research, VA Medical Center, Long Beach, California, USA; Department of Medicine, UCI, Irvine, California, USA.
| |
Collapse
|
8
|
Inhibition of hypoxia-inducible factor-1α alleviates acinar cell necrosis in a mouse model of acute pancreatitis. Biochem Biophys Res Commun 2021; 572:72-79. [PMID: 34358966 DOI: 10.1016/j.bbrc.2021.07.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 12/20/2022]
Abstract
Hypoxia-inducible factor-1α (Hif1α) is activated in hypoxia and is closely related to oxidative stress, immunity and cell metabolism. Recently, it is reported that Hif1α is involved in atherosclerosis, ischemia-reperfusion (I/R) injury, alcoholic liver disease and pancreatic tumors. In this study, we found that Hif1 signal pathway is significantly changed in pancreas of acute pancreatitis (AP) mice. Meanwhile, we verified that the high expression of Hif1α injured pancreatic tissues of cerulean-induced AP mice, which prompting that Hif1α participated in the progress of histopathology on AP. We applied a Hif1α inhibitor PX478 and observed that it could alleviate histological injury of pancreas as well as the levels of serum amylase, lipase and proinflammatory cytokine in the murine model of AP induced by caerulein. In addition, PX478 could reduce the formation of necrosome (RIP3 and p-MLKL) and the generation of reactive oxygen species (ROS) in AP mice. Correspondingly, we further confirmed the effectiveness of PX478 in vitro and found that inhibiting Hif1α could mitigated the necrosis of pancreatic acinar cells via reducing the RIP3 and p-MLKL expression and the ROS production. In conclusion, inhibiting Hif1α could protect against acinar cells necrosis in AP, which may provide a new target for the prevention and treatment of AP clinically.
Collapse
|