1
|
Alam J, Yaman E, de Paiva CS, Li DQ, Villalba Silva GC, Zuo Z, Pflugfelder SC. Changes in conjunctival mononuclear phagocytes and suppressive activity of regulatory macrophages in desiccation induced dry eye. Ocul Surf 2024; 34:348-362. [PMID: 39306240 DOI: 10.1016/j.jtos.2024.09.003] [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: 01/12/2024] [Revised: 09/07/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024]
Abstract
PURPOSE To evaluate the effects of dry eye on conjunctival immune cell number and transcriptional profiles with attention to mononuclear phagocytes. METHODS Expression profiling was performed by single-cell RNA sequencing on sorted conjunctival immune cells from non-stressed and C57BL/6 mice subjected to desiccating stress (DS). Monocle 3 modeled cell trajectory, scATAC-seq assessed chromatin accessibility and IPA identified canonical pathways. Inflammation and goblet cells were measured after depletion of MRC1+ MΦs with mannosylated clodronate liposomes. RESULTS Mononuclear phagocytes (monocytes, MΦs, DCs) comprised 72 % of immune cells and showed the greatest changes with DS. Distinct DS induced gene expression patterns were seen in phagocytes classified by expression of Ccr2 and [Timd4, Lyve1, Folr2 (TLR)]. Expression of phagocytosis/efferocytosis genes increased in TLF+CCR2- MΦs. Monocytes showed the highest expression of Ace, Cx3cr1, Vegfa, Ifngr1,2, and Stat1 and TLF-CCR2+ cells expressed higher levels of inflammatory mediators (Il1a, Il1b, Il1rn, Nfkb1, Ccl5, MHCII, Cd80, Cxcl10, Icam1). A trajectory from monocyte precursors branched to terminate in regulatory MΦs or in mDCs via transitional MΦ and cDC clusters. Activated pathways in TLF+ cells include phagocytosis, PPAR/RXRα activation, IL-10 signaling, alternate MΦ activation, while inflammatory pathways were suppressed. Depletion of MRC1+ MΦs increased IL-17 and IFN-γ expression and cytokine-expressing T cells, reduced IL-10 and worsened goblet loss. CONCLUSIONS Dryness stimulates distinct gene expression patterns in conjunctival phagocytes, increasing expression of regulatory genes in TLF+ cells regulated in part by RXRα, and inflammatory genes in CCR2+ cells. Regulatory MΦs depletion worsens DS induced inflammation and goblet cell loss.
Collapse
Affiliation(s)
- Jehan Alam
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Ebru Yaman
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Cintia S de Paiva
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - De-Quan Li
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Gerda Cristal Villalba Silva
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Zhen Zuo
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Stephen C Pflugfelder
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
2
|
Kawczak P, Feszak I, Brzeziński P, Bączek T. Structure-Activity Relationships and Therapeutic Applications of Retinoids in View of Potential Benefits from Drug Repurposing Process. Biomedicines 2024; 12:1059. [PMID: 38791021 PMCID: PMC11117600 DOI: 10.3390/biomedicines12051059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Vitamin A, an essential micronutrient, is integral to various biological processes crucial for organismal development and maintenance. Dietary sources of vitamin A encompass preformed retinol, retinyl esters, and provitamin A carotenoids. Retinoic acid (RA), a key component, plays pivotal roles in vision, cell proliferation, apoptosis, immune function, and gene regulation. Drug repurposing, an effective strategy for identifying new therapeutic applications for existing drugs, has gained prominence in recent years. This review seeks to provide a comprehensive overview of the current research landscape surrounding retinoids and drug repurposing. The scope of this review encompasses a comprehensive examination of retinoids and their potential for repurposing in various therapeutic contexts. Despite their efficacy in treating dermatological conditions, concerns about toxicity persist, driving the search for safer and more potent retinoids. The molecular mechanisms underlying retinoid activity involve binding to retinoic acid receptors (RARs) and retinoid X receptors (RXRs), leading to transcriptional regulation of target genes. This review seeks to shed light on the possibilities for repurposing retinoids to cover a wider spectrum of therapeutic uses by exploring recent scientific progress. It also aims to offer a more comprehensive understanding of the therapeutic prospects of retinoids and the broader impact of drug repositioning in contemporary medicine.
Collapse
Affiliation(s)
- Piotr Kawczak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, 80-416 Gdańsk, Poland;
| | - Igor Feszak
- Department of Nursing, Faculty of Health Sciences, Pomeranian University in Słupsk, 76-200 Słupsk, Poland;
| | - Piotr Brzeziński
- Department of Physiotherapy and Medical Emergency, Institute of Health Sciences, Pomeranian University in Słupsk, 76-200 Słupsk, Poland;
- Department of Dermatology, Voivodeship Specialist Hospital, 76-200 Słupsk, Poland
| | - Tomasz Bączek
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, 80-416 Gdańsk, Poland;
- Department of Nursing, Faculty of Health Sciences, Pomeranian University in Słupsk, 76-200 Słupsk, Poland;
| |
Collapse
|
3
|
Yan J, Xiao L, Feng D, Chen B, Yang T, Tong B, Luo R, Wang Y, Chen J. Vitamin A deficiency suppresses CEACAM1 to impair colonic epithelial barrier function via downregulating microbial-derived short-chain fatty acids. Genes Dis 2024; 11:1066-1081. [PMID: 37692511 PMCID: PMC10491915 DOI: 10.1016/j.gendis.2023.03.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/08/2023] [Accepted: 03/29/2023] [Indexed: 09/12/2023] Open
Abstract
Vitamin A (VA) plays an essential role in modulating both the gut microbiota and gut barrier function. Short-chain fatty acids (SCFAs), as metabolites of the gut microbiota, protect the physiological intestinal barrier; however, they are compromised when VA is deficient. Thus, there is an urgent need to understand how and which SCFAs modulate colonic epithelial barrier integrity in VA deficiency (VAD). Herein, compared with normal VA rats (VAN), at the beginning of pregnancy, we confirmed that the colonic desmosome junction was impaired in the VAD group, and the amounts of acetate, propionate, and butyrate declined because of the decreased abundance of SCFA-producing bacteria (Romboutsia, Collinsella, and Allobaculum). The differentially expressed genes correlated with the gut barrier and the histone deacetylase complex between the VAD and VAN groups were enriched by RNA sequencing. In the VAD group, the expression levels of colonic CEA cell adhesion molecule 1 (CEACAM1) were down-regulated, and the levels of histone deacetylase 1 (HDAC1) and HDAC3 were up-regulated. Intriguingly, the above changes in the VAD groups were rescued by VA supplementation in the early postnatal period. Further study indicated that in Caco-2 cells, butyrate treatment significantly repressed the enrichment of HDAC3 on the promoter of the CEACAM1 gene to induce its expression. Our findings support that butyrate intervention can alleviate the impairment of colonic barrier function caused by VAD, and timely postnatal VA intervention may reverse the damage caused by VAD on gut barrier integrity during pregnancy.
Collapse
Affiliation(s)
- Junyan Yan
- Children Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 401122, China
| | - Lu Xiao
- Children Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 401122, China
- Department of Gastroenterology, Children's Hospital of Chongqing Medical University, Chongqing 401122, China
| | - Di Feng
- Children Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 401122, China
| | - Baolin Chen
- Children Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 401122, China
| | - Ting Yang
- Children Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 401122, China
| | - Bei Tong
- Children Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 401122, China
| | - Ruifang Luo
- Children Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 401122, China
| | - Yuting Wang
- Department of Gastroenterology, Children's Hospital of Chongqing Medical University, Chongqing 401122, China
| | - Jie Chen
- Children Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Child Nutrition and Health, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 401122, China
| |
Collapse
|
4
|
Calderon RM, Golczak M, Paik J, Blaner WS. Dietary Vitamin A Affects the Function of Incretin-Producing Enteroendocrine Cells in Male Mice Fed a High-Fat Diet. J Nutr 2023; 153:2901-2914. [PMID: 37648113 PMCID: PMC10613727 DOI: 10.1016/j.tjnut.2023.08.030] [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: 07/01/2023] [Revised: 08/12/2023] [Accepted: 08/24/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Retinol-binding protein 2 (RBP2) is an intracellular carrier for vitamin A in the absorptive enterocytes. Mice lacking RBP2 (Rbp2-/-) display an unexpected phenotype of obesity, glucose intolerance, and elevated glucose-dependent insulinotropic polypeptide (GIP) levels. GIP and glucagon-like peptide 1 (GLP-1) are incretin hormones secreted by enteroendocrine cells (EECs). We recently demonstrated the presence of RBP2 and other retinoid-related proteins in EECs. OBJECTIVES Given RBP2's role in intracellular retinoid trafficking, we aimed to evaluate whether dietary vitamin A affects incretin-secreting cell function and gene expression. METHODS Male Rbp2-/- mice and sex- and age-matched controls (n = 6-9) were fed a high-fat diet (HFD) for 18 wk containing normal (VAN, 4000 IU/kg of diet) or low (VAL, 25% of normal) vitamin A concentrations. Body weight was recorded biweekly. Plasma GIP and GLP-1 levels were obtained fasting and 30 min after an oral fat gavage at week 16. Glucose tolerance tests were also performed. Mice were killed at week 18, and blood and tissue samples were obtained. RESULTS Rbp2-/- mice displayed greater weight gain on the VAN compared with the VAL diet from week 7 of the intervention (P ≤ 0.01). Stimulated GIP levels were elevated in Rbp2-/- mice compared with their controls fed the VAN diet (P = 0.02), whereas their GIP response was lower when fed the VAL diet (P = 0.03). Although no differences in GLP-1 levels were observed in the VAN diet group, a lower GLP-1 response was seen in Rbp2-/- mice fed the VAL diet (P = 0.02). Changes in incretin gene expression and that of other genes associated with EEC lineage and function were consistent with these observations. Circulating and hepatic retinoid levels revealed no systemic vitamin A deficiency across dietary groups. CONCLUSIONS Our data support a role for RBP2 and dietary vitamin A in incretin secretion and gene expression in mice fed a HFD.
Collapse
Affiliation(s)
- Rossana M Calderon
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, United States.
| | - Marcin Golczak
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, United States; Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Jisun Paik
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - William S Blaner
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, United States
| |
Collapse
|
5
|
Jackson C, Kolba N, Tako E. Assessing the Interactions between Zinc and Vitamin A on Intestinal Functionality, Morphology, and the Microbiome In Vivo ( Gallus gallus). Nutrients 2023; 15:2754. [PMID: 37375657 DOI: 10.3390/nu15122754] [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: 05/23/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Dietary deficiencies in zinc (Zn) and vitamin A (VA) are among the leading micronutrient deficiencies globally and previous research has proposed a notable interaction between Zn and VA physiological status. This study aimed to assess the effects of zinc and vitamin A (isolated and combined) on intestinal functionality and morphology, and the gut microbiome (Gallus gallus). The study included nine treatment groups (n~11)-no-injection (NI); H2O; 0.5% oil; normal zinc (40 mg/kg ZnSO4) (ZN); low zinc (20 mg/kg) (ZL); normal retinoid (1500 IU/kg retinyl palmitate) (RN); low retinoid (100 IU/kg) (RL); normal zinc and retinoid (40 mg/kg; 1500 IU/kg) (ZNRN); low zinc and retinoid (ZLRL) (20 mg/kg; 100 IU/kg). Samples were injected into the amniotic fluid of the fertile broiler eggs. Tissue samples were collected upon hatch to target biomarkers. ZLRL reduced ZIP4 gene expression and upregulated ZnT1 gene expression (p < 0.05). Duodenal surface area increased the greatest in RL compared to RN (p < 0.01), and ZLRL compared to ZNRN (p < 0.05). All nutrient treatments yielded shorter crypt depths (p < 0.01). Compared to the oil control, ZLRL and ZNRN reduced (p < 0.05) the cecal abundance of Bifidobacterium and Clostridium genera (p < 0.05). These results suggest a potentially improved intestinal epithelium proceeding with Zn and VA intra-amniotic administration. Intestinal functionality and gut bacteria were modulated. Further research should characterize long-term responses and the microbiome profile.
Collapse
Affiliation(s)
- Cydney Jackson
- Department of Food Science, Cornell University, Ithaca, NY 14850, USA
| | - Nikolai Kolba
- Department of Food Science, Cornell University, Ithaca, NY 14850, USA
| | - Elad Tako
- Department of Food Science, Cornell University, Ithaca, NY 14850, USA
| |
Collapse
|
6
|
Tsai CY, Saito T, Sarangdhar M, Abu-El-Haija M, Wen L, Lee B, Yu M, Lipata DA, Manohar M, Barakat MT, Contrepois K, Tran TH, Theoret Y, Bo N, Ding Y, Stevenson K, Ladas EJ, Silverman LB, Quadro L, Anthony TG, Jegga AG, Husain SZ. A systems approach points to a therapeutic role for retinoids in asparaginase-associated pancreatitis. Sci Transl Med 2023; 15:eabn2110. [PMID: 36921036 PMCID: PMC10205044 DOI: 10.1126/scitranslmed.abn2110] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/22/2023] [Indexed: 03/17/2023]
Abstract
Among drug-induced adverse events, pancreatitis is life-threatening and results in substantial morbidity. A prototype example is the pancreatitis caused by asparaginase, a crucial drug used to treat acute lymphoblastic leukemia (ALL). Here, we used a systems approach to identify the factors affecting asparaginase-associated pancreatitis (AAP). Connectivity Map analysis of the transcriptomic data showed that asparaginase-induced gene signatures were potentially reversed by retinoids (vitamin A and its analogs). Analysis of a large electronic health record database (TriNetX) and the U.S. Federal Drug Administration Adverse Events Reporting System demonstrated a reduction in AAP risk with concomitant exposure to vitamin A. Furthermore, we performed a global metabolomic screening of plasma samples from 24 individuals with ALL who developed pancreatitis (cases) and 26 individuals with ALL who did not develop pancreatitis (controls), before and after a single exposure to asparaginase. Screening from this discovery cohort revealed that plasma carotenoids were lower in the cases than in controls. This finding was validated in a larger external cohort. A 30-day dietary recall showed that the cases received less dietary vitamin A than the controls did. In mice, asparaginase administration alone was sufficient to reduce circulating and hepatic retinol. Based on these data, we propose that circulating retinoids protect against pancreatic inflammation and that asparaginase reduces circulating retinoids. Moreover, we show that AAP is more likely to develop with reduced dietary vitamin A intake. The systems approach taken for AAP provides an impetus to examine the role of dietary vitamin A supplementation in preventing or treating AAP.
Collapse
Affiliation(s)
- Cheng-Yu Tsai
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, 94304, USA
| | - Toshie Saito
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, 94304, USA
| | - Mayur Sarangdhar
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
- Division of Oncology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Maisam Abu-El-Haija
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Division of Pediatric Gastroenterology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Li Wen
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100006, China
| | - Bomi Lee
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, 94304, USA
| | - Mang Yu
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, 94304, USA
| | - Den A. Lipata
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, 94304, USA
| | - Murli Manohar
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, 94304, USA
| | - Monique T. Barakat
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, 94304, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Kévin Contrepois
- Department of Genetics, School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Thai Hoa Tran
- Division of Pediatric Hematology Oncology, Charles-Bruneau Cancer Center, CHU Sainte-Justine, Montreal, QC, H3T 1C5, Canada
| | - Yves Theoret
- Département Clinique de Médecine de Laboratoire, Secteur Pharmacologie Clinique, Optilab Montréal - CHU Sainte-Justine, Montreal, H3T 1C5, Canada
| | - Na Bo
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Ying Ding
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Kristen Stevenson
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Elena J. Ladas
- Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Institute of Human Nutrition, Columbia University, New York, NY, 10032, USA
| | - Lewis B. Silverman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Division of Pediatric Hematology-Oncology, Boston, Children’s Hospital, Boston, MA, 02115, USA
| | - Loredana Quadro
- Department of Food Science, Rutgers Center for Lipid Research and the New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, 08901, USA
| | - Tracy G. Anthony
- Department of Nutritional Sciences and the New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, 08901, USA
| | - Anil G. Jegga
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Sohail Z. Husain
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, CA, 94304, USA
| |
Collapse
|
7
|
Weinert-Nelson JR, Biddle AS, Sampath H, Williams CA. Fecal Microbiota, Forage Nutrients, and Metabolic Responses of Horses Grazing Warm- and Cool-Season Grass Pastures. Animals (Basel) 2023; 13:ani13050790. [PMID: 36899650 PMCID: PMC10000167 DOI: 10.3390/ani13050790] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023] Open
Abstract
Integrating warm-season grasses into cool-season equine grazing systems can increase pasture availability during summer months. The objective of this study was to evaluate effects of this management strategy on the fecal microbiome and relationships between fecal microbiota, forage nutrients, and metabolic responses of grazing horses. Fecal samples were collected from 8 mares after grazing cool-season pasture in spring, warm-season pasture in summer, and cool-season pasture in fall as well as after adaptation to standardized hay diets prior to spring grazing and at the end of the grazing season. Random forest classification was able to predict forage type based on microbial composition (accuracy: 0.90 ± 0.09); regression predicted forage crude protein (CP) and non-structural carbohydrate (NSC) concentrations (p < 0.0001). Akkermansia and Clostridium butyricum were enriched in horses grazing warm-season pasture and were positively correlated with CP and negatively with NSC; Clostridum butyricum was negatively correlated with peak plasma glucose concentrations following oral sugar tests (p ≤ 0.05). These results indicate that distinct shifts in the equine fecal microbiota occur in response different forages. Based on relationships identified between the microbiota, forage nutrients, and metabolic responses, further research should focus on the roles of Akkermansia spp. and Clostridium butyricum within the equine hindgut.
Collapse
Affiliation(s)
- Jennifer R. Weinert-Nelson
- Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Amy S. Biddle
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19711, USA
| | - Harini Sampath
- Department of Nutritional Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Carey A. Williams
- Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Correspondence:
| |
Collapse
|
8
|
Reboul E. Proteins involved in fat-soluble vitamin and carotenoid transport across the intestinal cells: New insights from the past decade. Prog Lipid Res 2023; 89:101208. [PMID: 36493998 DOI: 10.1016/j.plipres.2022.101208] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
It is now well established that vitamins D, E, and K and carotenoids are not absorbed solely through passive diffusion. Broad-specificity membrane transporters such as SR-BI (scavenger receptor class B type I), CD36 (CD36 molecule), NPC1L1 (Niemann Pick C1-like 1) or ABCA1 (ATP-binding cassette A1) are involved in the uptake of these micronutrients from the lumen to the enterocyte cytosol and in their secretion into the bloodstream. Recently, the existence of efflux pathways from the enterocyte back to the lumen or from the bloodstream to the lumen, involving ABCB1 (P-glycoprotein/MDR1) or the ABCG5/ABCG8 complex, has also been evidenced for vitamins D and K. Surprisingly, no membrane proteins have been involved in dietary vitamin A uptake so far. After an overview of the metabolism of fat-soluble vitamins and carotenoids along the gastrointestinal tract (from the mouth to the colon where interactions with microbiota may occur), a focus is placed on the identified and candidate proteins participating in the apical uptake, intracellular transport, basolateral secretion and efflux back to the lumen of fat-soluble vitamins and carotenoids in enterocytes. This review also highlights the mechanisms that remain to be identified to fully unravel the pathways involved in fat-soluble vitamin and carotenoid intestinal absorption.
Collapse
|
9
|
Holloway C, Zhong G, Kim YK, Ye H, Sampath H, Hammerling U, Isoherranen N, Quadro L. Retinoic acid regulates pyruvate dehydrogenase kinase 4 (Pdk4) to modulate fuel utilization in the adult heart: Insights from wild-type and β-carotene 9',10' oxygenase knockout mice. FASEB J 2022; 36:e22513. [PMID: 36004605 PMCID: PMC9544431 DOI: 10.1096/fj.202101910rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 08/05/2022] [Accepted: 08/11/2022] [Indexed: 11/11/2022]
Abstract
Regulation of the pyruvate dehydrogenase (PDH) complex by the pyruvate dehydrogenase kinase PDK4 enables the heart to respond to fluctuations in energy demands and substrate availability. Retinoic acid, the transcriptionally active form of vitamin A, is known to be involved in the regulation of cardiac function and growth during embryogenesis as well as under pathological conditions. Whether retinoic acid also maintains cardiac health under physiological conditions is unknown. However, vitamin A status and intake of its carotenoid precursor β-carotene have been linked to the prevention of heart diseases. Here, we provide in vitro and in vivo evidence that retinoic acid regulates cardiac Pdk4 expression and thus PDH activity. Furthermore, we show that mice lacking β-carotene 9',10'-oxygenase (BCO2), the only enzyme of the adult heart that cleaves β-carotene to generate retinoids (vitamin A and its derivatives), displayed cardiac retinoic acid insufficiency and impaired metabolic flexibility linked to a compromised PDK4/PDH pathway. These findings provide novel insights into the functions of retinoic acid in regulating energy metabolism in adult tissues, especially the heart.
Collapse
Affiliation(s)
- Chelsee Holloway
- Graduate Program in Endocrinology and Animal Bioscience, Rutgers University, New Brunswick, New Jersey, USA.,Department of Food Science, Rutgers University, New Brunswick, New Jersey, USA.,Rutgers Center for Lipid Research and Institute of Food Nutrition and Health, Rutgers University, New Brunswick, New Jersey, USA
| | - Guo Zhong
- Department of Pharmaceutics Health Sciences, University of Washington, Seattle, Washington, USA
| | - Youn-Kyung Kim
- Department of Food Science, Rutgers University, New Brunswick, New Jersey, USA.,Rutgers Center for Lipid Research and Institute of Food Nutrition and Health, Rutgers University, New Brunswick, New Jersey, USA
| | - Hong Ye
- Department of Food Science, Rutgers University, New Brunswick, New Jersey, USA.,Rutgers Center for Lipid Research and Institute of Food Nutrition and Health, Rutgers University, New Brunswick, New Jersey, USA.,Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey, USA
| | - Harini Sampath
- Rutgers Center for Lipid Research and Institute of Food Nutrition and Health, Rutgers University, New Brunswick, New Jersey, USA.,Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey, USA
| | - Ulrich Hammerling
- Department of Food Science, Rutgers University, New Brunswick, New Jersey, USA.,Rutgers Center for Lipid Research and Institute of Food Nutrition and Health, Rutgers University, New Brunswick, New Jersey, USA
| | - Nina Isoherranen
- Department of Pharmaceutics Health Sciences, University of Washington, Seattle, Washington, USA
| | - Loredana Quadro
- Department of Food Science, Rutgers University, New Brunswick, New Jersey, USA.,Rutgers Center for Lipid Research and Institute of Food Nutrition and Health, Rutgers University, New Brunswick, New Jersey, USA
| |
Collapse
|
10
|
β-carotene improves fecal dysbiosis and intestinal dysfunctions in a mouse model of vitamin A deficiency. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159122. [PMID: 35158041 PMCID: PMC9940628 DOI: 10.1016/j.bbalip.2022.159122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/20/2022] [Accepted: 02/01/2022] [Indexed: 02/03/2023]
Abstract
Vitamin A deficiency (VAD) results in intestinal inflammation, increased redox stress and reactive oxygen species (ROS) levels, imbalanced inflammatory and immunomodulatory cytokines, compromised barrier function, and perturbations of the gut microbiome. To combat VAD dietary interventions with β-carotene, the most abundant precursor of vitamin A, are recommended. However, the impact of β-carotene on intestinal health during VAD has not been fully clarified, especially regarding the VAD-associated intestinal dysbiosis. Here we addressed this question by using Lrat-/-Rbp-/- (vitamin A deficient) mice deprived of dietary preformed vitamin A and supplemented with β-carotene as the sole source of the vitamin, alongside with WT (vitamin A sufficient) mice. We found that dietary β-carotene impacted intestinal vitamin A status, barrier integrity and inflammation in both WT and Lrat-/-Rbp-/- (vitamin A deficient) mice on the vitamin A-free diet. However, it did so to a greater extent under overt VAD. Dietary β-carotene also modified the taxonomic profile of the fecal microbiome, but only under VAD. Given the similarity of the VAD-associated intestinal phenotypes with those of several other disorders of the gut, collectively known as Inflammatory Bowel Disease (IBD) Syndrome, these findings are broadly relevant to the effort of developing diet-based intervention strategies to ameliorate intestinal pathological conditions.
Collapse
|
11
|
Pang B, Jin H, Liao N, Li J, Jiang C, Shi J. Vitamin A supplementation ameliorates ulcerative colitis in gut microbiota-dependent manner. Food Res Int 2021; 148:110568. [PMID: 34507723 DOI: 10.1016/j.foodres.2021.110568] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 06/24/2021] [Accepted: 06/27/2021] [Indexed: 12/27/2022]
Abstract
Ulcerative colitis (UC), is a chronic relapsing inflammatory condition of the gastrointestinal track. The purpose of this study is to explore whether Vitamin A (VA) can treat UC and its mechanisms. A mouse model of UC was established using 3.0% (w/v) dextran sodium sulfate (DSS). VA was used to treat UC by intragastric administration of 5000 international unit (IU) retinyl acetate. Fecal microbiota transplantation (FMT) was also used to treat the UC model mice to verify the effect of influenced gut microbiota. The content of short-chain fatty acids (SCFAs) in cecal contents was quantitatively detected by gas chromatography and mass spectrometry. VA supplementation significantly ameliorated UC. 16S rRNA sequencing indicated that VA-treated mice exhibited much more abundant gut microbial diversity and flora composition. Targeted metabolomics analysis manifested the increased production of SCFAs in VA-treated mice. Gut microbiota depletion and FMT results confirmed the gut microbiota-dependent mechanism as that VA relieved UC via regulating gut microbiota: increase in SCFA-producing genera and decrease in UC-related genera. The restore of intestinal barrier and the inhibition of inflammation were also found to contribute to the amelioration of UC by VA. It was concluded that a VA supplement was enough to cause a significant change in gut microbiota and amelioration of UC.
Collapse
Affiliation(s)
- Bing Pang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China
| | - Han Jin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Ning Liao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China
| | - Junjun Li
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China
| | - Chunmei Jiang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Junling Shi
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| |
Collapse
|