Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-37315736

ABSTRACT

In the maternal circulation, apoB-containing low-density lipoproteins (LDL) and apoA1-containing high-density lipoproteins (HDL) transport lipids. The production of lipoproteins in the placenta has been suggested, but the directionality of release has not been resolved. We compared apolipoprotein concentrations and size-exclusion chromatography elution profiles of lipoproteins in maternal/fetal circulations, and in umbilical arteries/veins; identified placental lipoprotein-producing cells; and studied temporal induction of lipoprotein-synthesizing machinery during pregnancy. We observed that maternal and fetal lipoproteins are different with respect to concentrations and elution profiles. Surprisingly, concentrations and elution profiles of lipoproteins in umbilical arteries and veins were similar indicating their homeostatic control. Human placental cultures synthesized apoB100-containing LDL-sized and apoA1-containing HDL-sized particles. Immunolocalization techniques revealed that ApoA1 was present mainly in syncytiotrophoblasts. MTP, a critical protein for lipoprotein assembly, was in these trophoblasts. ApoB was in the placental stroma indicating that trophoblasts secrete apoB-containing lipoproteins into the stroma. ApoB and MTP expressions increased in placentas from the 2nd trimester to term, whereas apoA1 expression was unchanged. Thus, our studies provide new information regarding the timing of lipoprotein gene induction during gestation, the cells involved in lipoprotein assembly and the gel filtration profiles of human placental lipoproteins. Next, we observed that mouse placenta produces MTP, apoB100, apoB48 and apoA1. The expression of genes gradually increased and peaked in late gestation. This information may be useful in identifying transcription factors regulating the induction of these genes in gestation and the importance of placental lipoprotein assembly in fetal development.


Subject(s)
Carrier Proteins , Placenta , Mice , Animals , Humans , Female , Pregnancy , Placenta/metabolism , Carrier Proteins/metabolism , Lipoproteins/metabolism , Apolipoproteins B/metabolism , Lipoproteins, LDL/metabolism
2.
Sci Transl Med ; 15(687): eabn2110, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36921036

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.


Subject(s)
Antineoplastic Agents , Pancreatitis , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Animals , Mice , Asparaginase/adverse effects , Retinoids/adverse effects , Vitamin A/therapeutic use , Pancreatitis/chemically induced , Pancreatitis/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Systems Analysis , Antineoplastic Agents/adverse effects
3.
Commun Biol ; 6(1): 227, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36854887

ABSTRACT

This Perspective discusses how retinol catalyzes resonance energy transfer (RET) reactions pivotally important for mitochondrial energy homeostasis by protein kinase C δ (PKCδ). PKCδ signals to the pyruvate dehydrogenase complex, controlling oxidative phosphorylation. The PKCδ-retinol complex reversibly responds to the redox potential of cytochrome c, that changes with the electron transfer chain workload. In contrast, the natural retinoid anhydroretinol irreversibly activates PKCδ. Its elongated conjugated-double-bond system limits the energy quantum absorbed by RET. Consequently, while capable of triggering the exergonic activating pathway, anhydroretinol fails to activate the endergonic silencing path, trapping PKCδ in the ON position and causing harmful levels of reactive oxygen species. However, physiological retinol levels displace anhydroretinol, buffer cyotoxicity and potentially render anhydroretinol useful for rapid energy generation. Intriguingly, apocarotenoids, the primary products of the mitochondrial ß-carotene,9'-10'-oxygenase, have all the anhydroretinol-like features, including modulation of energy homeostasis. We predict significant conceptual advances to stem from further understanding of the retinoid-catalyzed RET.


Subject(s)
Retinoids , Vitamin A , beta Carotene , Biology
4.
Methods Enzymol ; 674: 343-362, 2022.
Article in English | MEDLINE | ID: mdl-36008012

ABSTRACT

Dietary ß-carotene is the most abundant vitamin A precursor. Once absorbed by the enterocytes, the provitamin A carotenoid can either be cleaved into retinoids (vitamin A and its derivatives) or incorporated in its intact form within chylomicrons to be distributed throughout the body for utilization and/or storage by other tissues. From the liver, together with endogenous lipids, intact ß-carotene can also be incorporated within very low-density lipoprotein/low-density lipoprotein (VLDL/LDL) for transport to other tissues and organs. Microsomal triglyceride transfer protein (MTP) is a key regulator of lipoprotein biosynthesis in intestine and liver as it facilitates the incorporation of dietary and endogenous lipids into nascent lipoproteins. MTP is also critical for transferring ß-carotene into lipoprotein particles for secretion. Here, we present an in vitro method to assess the transfer of ß-carotene by MTP from donor to acceptor vesicles. This transfer can be assessed by precipitating donor vesicles and measuring amounts of ß-carotene transferred to acceptor vesicles. The levels of transferred ß-carotene are quantified by HPLC analysis and intrinsic fluorescence of ß-carotene. This chapter demonstrates the feasibility of this method which is also useful to study the role of MTP for incorporation of other carotenoids that are known to be carried within VLDL/LDL and chylomicrons for organ distribution.


Subject(s)
Vitamin A , beta Carotene , Carotenoids , Carrier Proteins , Chylomicrons , Lipoproteins , Lipoproteins, LDL , Lipoproteins, VLDL/metabolism , beta Carotene/metabolism
5.
FASEB J ; 36(9): e22513, 2022 09.
Article in English | MEDLINE | ID: mdl-36004605

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 ß-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 ß-carotene 9',10'-oxygenase (BCO2), the only enzyme of the adult heart that cleaves ß-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.


Subject(s)
Dioxygenases , beta Carotene , Animals , Dioxygenases/metabolism , Mice , Mice, Knockout , Oxygenases , Protein Kinases , Pyruvate Dehydrogenase Complex/metabolism , Tretinoin , Vitamin A
6.
Article in English | MEDLINE | ID: mdl-35158041

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 ß-carotene, the most abundant precursor of vitamin A, are recommended. However, the impact of ß-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 ß-carotene as the sole source of the vitamin, alongside with WT (vitamin A sufficient) mice. We found that dietary ß-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 ß-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.


Subject(s)
Intestinal Diseases , Vitamin A Deficiency , Animals , Disease Models, Animal , Dysbiosis/complications , Dysbiosis/drug therapy , Inflammation/complications , Inflammation/drug therapy , Mice , Vitamin A/therapeutic use , Vitamin A Deficiency/complications , Vitamin A Deficiency/drug therapy , Vitamin A Deficiency/pathology , beta Carotene/pharmacology , beta Carotene/therapeutic use
7.
Nutrients ; 13(6)2021 May 21.
Article in English | MEDLINE | ID: mdl-34063790

ABSTRACT

Background: While the current national prevalence rate of vitamin A deficiency (VAD) is estimated to be less than 1%, it is suggested that it varies between different ethnic groups and races within the U.S. We assessed the prevalence of VAD in pregnant women of different ethnic groups and tested these prevalence rates for associations with the vitamin A-related single nucleotide polymorphism (SNP) allele frequencies in each ethnic group. Methods: We analyzed two independent datasets of serum retinol levels with self-reported ethnicities and the differences of allele frequencies of the SNPs associated with vitamin A metabolism between groups in publicly available datasets. Results: Non-Hispanic Black and Hispanic pregnant women showed high VAD prevalence in both datasets. Interestingly, the VAD prevalence for Hispanic pregnant women significantly differed between datasets (p = 1.973 × 10-10, 95%CI 0.04-0.22). Alleles known to confer the risk of low serum retinol (rs10882272 C and rs738409 G) showed higher frequencies in the race/ethnicity groups with more VAD. Moreover, minor allele frequencies of a set of 39 previously reported SNPs associated with vitamin A metabolism were significantly different between the populations of different ancestries than those of randomly selected SNPs (p = 0.030). Conclusions: Our analysis confirmed that VAD prevalence varies between different ethnic groups/races and may be causally associated with genetic variants conferring risk for low retinol levels. Assessing genetic variant information prior to performing an effective nutrient supplementation program will help us plan more effective food-based interventions.


Subject(s)
Ethnicity/genetics , Polymorphism, Single Nucleotide , Pregnancy Complications/ethnology , Vitamin A Deficiency/ethnology , Vitamin A/genetics , Adult , Black or African American/genetics , Alleles , Female , Gene Frequency , Hispanic or Latino/genetics , Humans , Nutrition Surveys , Nutritional Status , Pregnancy , Pregnancy Complications/epidemiology , Pregnancy Complications/genetics , Prevalence , Racial Groups/genetics , Risk Factors , United States/epidemiology , Vitamin A/blood , Vitamin A Deficiency/epidemiology , Vitamin A Deficiency/genetics , Young Adult
8.
J Lipid Res ; 62: 100046, 2021.
Article in English | MEDLINE | ID: mdl-33587919

ABSTRACT

Lecithin:retinol acyltransferase and retinol-binding protein enable vitamin A (VA) storage and transport, respectively, maintaining tissue homeostasis of retinoids (VA derivatives). The precarious VA status of the lecithin:retinol acyltransferase-deficient (Lrat-/-) retinol-binding protein-deficient (Rbp-/-) mice rapidly deteriorates upon dietary VA restriction, leading to signs of severe vitamin A deficiency (VAD). As retinoids impact gut morphology and functions, VAD is often linked to intestinal pathological conditions and microbial dysbiosis. Thus, we investigated the contribution of VA storage and transport to intestinal retinoid homeostasis and functionalities. We showed the occurrence of intestinal VAD in Lrat-/-Rbp-/- mice, demonstrating the critical role of both pathways in preserving gut retinoid homeostasis. Moreover, in the mutant colon, VAD resulted in a compromised intestinal barrier as manifested by reduced mucins and antimicrobial defense, leaky gut, increased inflammation and oxidative stress, and altered mucosal immunocytokine profiles. These perturbations were accompanied by fecal dysbiosis, revealing that the VA status (sufficient vs. deficient), rather than the amount of dietary VA per se, is likely a major initial discriminant of the intestinal microbiome. Our data also pointed to a specific fecal taxonomic profile and distinct microbial functionalities associated with VAD. Overall, our findings revealed the suitability of the Lrat-/-Rbp-/- mice as a model to study intestinal dysfunctions and dysbiosis promoted by changes in tissue retinoid homeostasis induced by the host VA status and/or intake.


Subject(s)
Vitamin A
9.
Mol Metab ; 42: 101088, 2020 12.
Article in English | MEDLINE | ID: mdl-32992038

ABSTRACT

OBJECTIVE: Transformation of white into brown fat ("browning") reduces obesity in many preclinical models and holds great promise as a therapeutic concept in metabolic disease. Vitamin A metabolites (retinoids) have been linked to thermogenic programming of adipose tissue; however, the physiologic importance of systemic retinoid transport for adipose tissue browning and adaptive thermogenesis is unknown. METHODS: We performed cold exposure studies in mice and humans and used a genetic model of defective vitamin A transport, the retinol binding protein deficient (Rbp-/-) mouse, to study the effects of cooling on systemic vitamin A and the relevance of intact retinoid transport on cold-induced adipose tissue browning. RESULTS: We show that cold stimulation in mice and humans leads to an increase in circulating retinol and its plasma transporter, Rbp. In Rbp-/- mice, thermogenic programming of adipocytes and oxidative mitochondrial function are dramatically impaired in subcutaneous white fat, which renders Rbp-/- mice more cold-sensitive. In contrast, retinol stimulation in primary human adipocytes promotes thermogenic gene expression and mitochondrial respiration. In humans, cold-mediated retinol increase is associated with a shift in oxidative substrate metabolism suggestive of higher lipid utilisation. CONCLUSIONS: Systemic vitamin A levels are regulated by cold exposure in mice and humans, and intact retinoid transport is essential for cold-induced adipose tissue browning and adaptive thermogenesis.


Subject(s)
Adipose Tissue/metabolism , Retinol-Binding Proteins/metabolism , Thermogenesis/physiology , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adult , Animals , Body Temperature Regulation/genetics , Body Temperature Regulation/physiology , Energy Metabolism , Female , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Retinol-Binding Proteins/genetics , Thermogenesis/genetics , Vitamin A/metabolism , Vitamin A/physiology
11.
Methods Enzymol ; 637: 95-117, 2020.
Article in English | MEDLINE | ID: mdl-32359662

ABSTRACT

STRA6 (stimulated by retinoic acid 6) is a 75kDa polytopic transmembrane protein that facilitates cellular retinol uptake from retinol-binding protein (RBP). Structural characterization of STRA6 from Danio rerio purified in detergent and reconstituted in amphipol A8-35 was achieved by single-particle cryo-electron microscopy (cryo-EM). This provided the first high-resolution snapshot of this protein, showing a novel topology of a tightly assembled homodimer, and an unexpected physiological association with calmodulin in addition to insights into its potential mechanism of function. Specifically, a large hydrophobic cavity in the center of STRA6 linked to the known site of interaction with RBP suggested a route of retinol entry into the cell by diffusion into the membrane through a lateral opening of the cavity directly into the bilayer. Moreover, the capability to produce pure and homogeneous protein has allowed previously unattainable functional characterization of STRA6 in a reconstituted system. Here, we describe detailed methods for Danio rerio STRA6 expression in insect cells, purification in detergent and reconstitution in amphipol for structural characterization by cryo-EM. Furthermore, we show reconstitution of the protein in liposomes for an in vitro proteoliposome-based assay of STRA6-mediated retinol uptake. Finally, we present methods and preliminary cryo-EM data for STRA6 incorporated in lipid-filled nanodiscs, a close to native milieu to study membrane protein structure and function.


Subject(s)
Membrane Proteins , Retinol-Binding Proteins , Calmodulin , Cryoelectron Microscopy , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Binding , Retinol-Binding Proteins/metabolism
12.
Subcell Biochem ; 95: 27-55, 2020.
Article in English | MEDLINE | ID: mdl-32297295

ABSTRACT

The placenta, a hallmark of mammalian embryogenesis, allows nutrients to be exchanged between the mother and the fetus. Vitamin A (VA), an essential nutrient, cannot be synthesized by the embryo, and must be acquired from the maternal circulation through the placenta. Our understanding of how this transfer is accomplished is still in its infancy. In this chapter, we recapitulate the early studies about the relationship between maternal dietary/supplemental VA intake and fetal VA levels. We then describe how the discovery of retinol-binding protein (RBP or RBP4), the development of labeling and detection techniques, and the advent of knockout mice shifted this field from a macroscopic to a molecular level. The most recent data indicate that VA and its derivatives (retinoids) and the pro-VA carotenoid, ß-carotene, are transferred across the placenta by distinct proteins, some of which overlap with proteins involved in lipoprotein uptake. The VA status and dietary intake of the mother influence the expression of these proteins, creating feedback signals that control the uptake of retinoids and that may also regulate the uptake of lipids, raising the intriguing possibility of crosstalk between micronutrient and macronutrient metabolism. Many questions remain about the temporal and spatial patterns by which these proteins are expressed and transferred throughout gestation. The answers to these questions are highly relevant to human health, considering that those with either limited or excessive intake of retinoids/carotenoids during pregnancy may be at risk of obtaining improper amounts of VA that ultimately impact the development and health of their offspring.


Subject(s)
Embryonic Development , Vitamin A/metabolism , Animals , Female , Humans , Pregnancy , Pregnancy Complications/metabolism , Retinol-Binding Proteins/metabolism , Vitamin A Deficiency/metabolism , beta Carotene/metabolism
13.
PLoS One ; 15(2): e0228436, 2020.
Article in English | MEDLINE | ID: mdl-32027669

ABSTRACT

BACKGROUND: Cytochrome P450 1b1 (Cyp1b1) deletion and dietary retinol deficiency during pregnancy (GVAD) affect perinatal liver functions regulated by Srebp. Cyp1b1 is not expressed in perinatal liver but appears in the E9.5 embryo, close to sites of retinoic acid (RA) signaling. HYPOTHESIS: Parallel effects of Cyp1b1 and retinol on postnatal Srebp derive from effects in the developing liver or systemic signaling. APPROACH: Cluster postnatal increases in hepatic genes in relation to effects of GVAD or Cyp1b1 deletion. Sort expression changes in relation to genes regulated by Srebp1 and Srebp2.Test these treatments on embryos at E9.5, examining changes at the site of liver initiation. Use in situ hybridization to resolve effects on mRNA distributions of Aldh1a2 and Cyp26a1 (RA homeostasis); Hoxb1 and Pax6 (RA targets). Assess mice lacking Lrat and Rbp4 (DKO mice) that severely limits retinol supply to embryos. RESULTS: At birth, GVAD and Cyp1b1 deletion stimulate gene markers of hepatic stellate cell (HSC) activation but also suppress Hamp. These treatments then selectively prevent the postnatal onset of genes that synthesize cholesterol (Hmgcr, Sqle) and fatty acids (Fasn, Scd1), but also direct cholesterol transport (Ldlr, Pcsk9, Stard4) and retinoid synthesis (Aldh1a1, Rdh11). Extensive support by Cyp1b1 is implicated, but with distinct GVAD interventions for Srebp1 and Srebp2. At E9.5, Cyp1b1 is expressed in the septum transversum mesenchyme (STM) with ß-carotene oxygenase (Bco1) that generates retinaldehyde. STM provides progenitors for the HSC and supports liver expansion. GVAD and Cyp1b1-/- do not affect RA-dependent Hoxb1 and Pax6. In DKO embryos, RA-dependent Cyp26a1 is lost but Hoxb1 is sustained with Cyp1b1 at multiple sites. CONCLUSION: Cyp1b1-/- suppresses genes supported by Srebp. GVAD effects distinguish Srebp1 and Srebp2 mediation. Srebp regulation overlaps appreciably in cholesterol and retinoid homeostasis. Bco1/Cyp1b1 partnership in the STM may contribute to this later liver regulation.


Subject(s)
Cholesterol/biosynthesis , Cytochrome P-450 CYP1B1/physiology , Fetal Development , Liver/metabolism , Sterol Regulatory Element Binding Proteins/physiology , Tretinoin/metabolism , Animals , Animals, Newborn , Cytochrome P-450 CYP1B1/genetics , Embryo, Mammalian , Female , Fetal Development/drug effects , Fetal Development/genetics , Liver/drug effects , Liver/embryology , Liver/growth & development , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Retinol-Binding Proteins, Plasma/genetics , Retinol-Binding Proteins, Plasma/metabolism , Tretinoin/pharmacology
14.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1865(11): 158591, 2020 11.
Article in English | MEDLINE | ID: mdl-31863969

ABSTRACT

Vitamin A is an essential nutrient, critical for proper embryonic development in mammals. Both embryonic vitamin A-deficiency or -excess lead to congenital malformations or lethality in mammals, including humans. This is due to the defective transcriptional action of retinoic acid, the active form of vitamin A, that regulates in a spatial- and temporal-dependent manner the expression of genes essential for organogenesis. Thus, an adequate supply of vitamin A from the maternal circulation is vital for normal mammalian fetal development. Provitamin A carotenoids circulate in the maternal bloodstream and are available to the embryo. Of all the dietary carotenoids, ß-carotene is the main vitamin A precursor, contributing at least 30% of the vitamin A intake in the industrialized countries and often constituting the sole source of retinoids (vitamin A and its derivatives) in the developing world. In humans, up to 40% of the absorbed dietary ß-carotene is incorporated in its intact form in chylomicrons for distribution to other organs within the body, including the developing tissues. Here, it can serve as a source of vitamin A upon conversion into apocarotenoids by its cleavage enzymes. Given that ß-carotene is carried in the bloodstream by lipoproteins, and that the placenta acquires, assembles and secretes lipoproteins, it is becoming evident that the maternal-fetal transfer of ß-carotene relies on lipoprotein metabolism. Here, we will explore the current knowledge about this important biological process, the cross-talk between carotenoid and lipid metabolism in the context of the maternal-fetal transfer of this provitamin A precursor, and the mechanisms whereby ß-carotene is metabolized by the developing tissues. This article is part of a Special Issue entitled Carotenoids recent advances in cell and molecular biology edited by Johannes von Lintig and Loredana Quadro.


Subject(s)
Lipoproteins/metabolism , Vitamin A Deficiency/metabolism , Vitamin A/metabolism , beta Carotene/metabolism , Animals , Carotenoids/metabolism , Embryonic Development/drug effects , Female , Humans , Maternal-Fetal Relations/drug effects , Placenta/drug effects , Placenta/metabolism , Pregnancy , Vitamin A Deficiency/drug therapy , Vitamin A Deficiency/genetics , beta Carotene/therapeutic use
15.
J Perinat Med ; 47(2): 183-189, 2019 Feb 25.
Article in English | MEDLINE | ID: mdl-30231012

ABSTRACT

Background As breastfeeding awareness and social acceptance are increased, maternal nutritional deficiency requires more investigation. Methods A prospective cohort study was conducted to determine if vitamin A deficiency is more common in pregnant, lactating post-bariatric surgery women in an inner city population. Antepartum, women after bariatric surgery and controls with no history of malabsorption were recruited. Third trimester, postpartum maternal blood and cord blood were collected as well as three breast milk samples: colostrum, transitional and mature milk. A nutritional survey of diet was completed. Each serum sample was analyzed for total retinol and ß-carotene; breast milk samples were analyzed for retinol and retinyl esters, total retinol and ß-carotene. Results Fifty-three women after bariatric surgery and 66 controls were recruited. Postpartum serum retinol was significantly higher in women after bariatric surgery in the univariate analysis (P<0.0001) and confirmed in the multiple linear mixed model (P=0.0001). Breast milk colostrum retinol and transitional milk total retinol were significantly greater in the bariatric surgery group in the univariate analysis (P=0.03 and P=0.02, respectively), but not after adjusting for confounders. Serum ß-carotene in the third trimester and postpartum were lower (P<0.0001 and P=0.003, respectively) in the bariatric surgery group but not after adjusting for confounders. Vitamin A deficiency was high in both groups in serum and breast milk samples. Conclusion Nutritional deficiencies in breastfeeding women after bariatric surgeries may in fact be less common than in control women in an inner city.


Subject(s)
Bariatric Surgery/adverse effects , Breast Feeding/statistics & numerical data , Milk, Human/chemistry , Vitamin A Deficiency , Vitamin A , beta Carotene , Adult , Bariatric Surgery/methods , Female , Humans , Lactation/physiology , Nutrition Assessment , Nutrition Disorders/diagnosis , Nutrition Disorders/epidemiology , Nutrition Disorders/etiology , Obesity/surgery , Perinatal Care/methods , Perinatal Care/statistics & numerical data , Pregnancy , Pregnancy Trimester, Third/blood , United States/epidemiology , Urban Population/statistics & numerical data , Vitamin A/analysis , Vitamin A/blood , Vitamin A Deficiency/diagnosis , Vitamin A Deficiency/epidemiology , Vitamin A Deficiency/etiology , beta Carotene/analysis , beta Carotene/blood
16.
Sci Rep ; 8(1): 8834, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29892071

ABSTRACT

Vitamin A deficiency is still a public health concern affecting millions of pregnant women and children. Retinoic acid, the active form of vitamin A, is critical for proper mammalian embryonic development. Embryos can generate retinoic acid from maternal circulating ß-carotene upon oxidation of retinaldehyde produced via the symmetric cleavage enzyme ß-carotene 15,15'-oxygenase (BCO1). Another cleavage enzyme, ß-carotene 9',10'-oxygenase (BCO2), asymmetrically cleaves ß-carotene in adult tissues to prevent its mitochondrial toxicity, generating ß-apo-10'-carotenal, which can be converted to retinoids (vitamin A and its metabolites) by BCO1. However, the role of BCO2 during mammalian embryogenesis is unknown. We found that mice lacking BCO2 on a vitamin A deficiency-susceptible genetic background (Rbp4-/-) generated severely malformed vitamin A-deficient embryos. Maternal ß-carotene supplementation impaired fertility and did not restore normal embryonic development in the Bco2-/-Rbp4-/- mice, despite the expression of BCO1. These data demonstrate that BCO2 prevents ß-carotene toxicity during embryogenesis under severe vitamin A deficiency. In contrast, ß-apo-10'-carotenal dose-dependently restored normal embryonic development in Bco2-/-Rbp4-/- but not Bco1-/-Bco2-/-Rbp4-/- mice, suggesting that ß-apo-10'-carotenal facilitates embryogenesis as a substrate for BCO1-catalyzed retinoid formation. These findings provide a proof of principle for the important role of BCO2 in embryonic development and invite consideration of ß-apo-10'-carotenal as a nutritional supplement to sustain normal embryonic development in vitamin A-deprived pregnant women.


Subject(s)
Carotenoids/metabolism , Embryonic Development , Retinoids/metabolism , Vitamin A Deficiency/complications , Vitamin A Deficiency/physiopathology , Animals , Dioxygenases/deficiency , Dioxygenases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Retinol-Binding Proteins, Plasma/deficiency , Retinol-Binding Proteins, Plasma/metabolism , beta-Carotene 15,15'-Monooxygenase/deficiency , beta-Carotene 15,15'-Monooxygenase/metabolism
17.
Annu Rev Nutr ; 38: 153-172, 2018 08 21.
Article in English | MEDLINE | ID: mdl-29751734

ABSTRACT

Apocarotenoids are cleavage products of C40 isoprenoid pigments, named carotenoids, synthesized exclusively by plants and microorganisms. The colors of flowers and fruits and the photosynthetic process are examples of the biological properties conferred by carotenoids to these organisms. Mammals do not synthesize carotenoids but obtain them from foods of plant origin. Apocarotenoids are generated upon enzymatic and nonenzymatic cleavage of the parent compounds both in plants and in the tissues of mammals that have ingested carotenoid-containing foods. The best-characterized apocarotenoids are retinoids (vitamin A and its derivatives), generated upon central oxidative cleavage of provitamin A carotenoids, mainly ß-carotene. In addition to the well-known biological actions of vitamin A, it is becoming apparent that nonretinoid apocarotenoids also have the potential to regulate a broad spectrum of critical cellular functions, thus influencing mammalian health. This review discusses the current knowledge about the generation and biological activities of nonretinoid apocarotenoids in mammals.


Subject(s)
Carotenoids/chemistry , Carotenoids/metabolism , Animals , Diet , Food Analysis , Humans , Intestinal Absorption
18.
Arch Biochem Biophys ; 647: 33-40, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29654731

ABSTRACT

It is now widely accepted that nutrition during critical periods in early development, both pre- and postnatal, may have lifetime consequences in determining health or onset of major diseases in the adult life. Dietary carotenoids have shown beneficial health effects throughout the life cycle due to their potential antioxidant properties, their ability to serves as precursors of vitamin A and to the emerging signaling functions of their metabolites. The non-provitamin A carotenoids lutein and zeaxanthin are emerging as important modulators of infant and child visual and cognitive development, as well as critical effectors in the prevention and treatment of morbidity associated with premature births. This review provides a general overview of lutein and zeaxanthin metabolism in mammalian tissues and highlights the major advancements and remaining gaps in knowledge in regards to their metabolism and health effects during pre- and early post-natal development. Furthering our knowledge in this area of research will impact dietary recommendation and supplementation strategies aimed at sustaining proper fetal and infant growth.


Subject(s)
Lutein/metabolism , Zeaxanthins/metabolism , Animals , Breast Feeding , Diet , Dietary Supplements/analysis , Female , Fetus/metabolism , Humans , Infant , Intestinal Absorption , Lactation , Lutein/analysis , Maternal-Fetal Exchange , Milk/chemistry , Milk/metabolism , Nutritional Status , Pregnancy , Zeaxanthins/analysis
19.
Nutrients ; 8(12)2016 Nov 29.
Article in English | MEDLINE | ID: mdl-27916814

ABSTRACT

Vitamin A regulates many essential mammalian biological processes, including embryonic development. ß-carotene is the main source of vitamin A in the human diet. Once ingested, it is packaged into lipoproteins, predominantly low-density lipoproteins (LDL), and transported to different sites within the body, including the liver and developing tissues, where it can either be stored or metabolized to retinoids (vitamin A and its derivatives). The molecular mechanisms of ß-carotene uptake by the liver or developing tissues remain elusive. Here, we investigated the role of the LDL receptor (LDLr) in ß-carotene uptake by maternal liver, placenta and embryo. We administered a single dose of ß-carotene to Ldlr+/- and Ldlr-/- pregnant mice via intraperitoneal injection at mid-gestation and monitored the changes in ß-carotene content among maternal lipoproteins and the liver, as well as the accumulation of ß-carotene in the placental-fetal unit. We showed an abnormal ß-carotene distribution among serum lipoproteins and reduced hepatic ß-carotene uptake in Ldlr-/- dams. These data strongly imply that LDLr significantly contributes to ß-carotene uptake in the adult mouse liver. In contrast, LDLr does not seem to mediate acquisition of ß-carotene by the placental-fetal unit.


Subject(s)
Liver/metabolism , Receptors, LDL/metabolism , beta Carotene/metabolism , Animals , Female , Gene Expression Regulation , Genotype , Lipoproteins/chemistry , Maternal-Fetal Exchange , Mice , Mice, Knockout , Placental Circulation , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, LDL/genetics , Retinoids/chemistry , Retinoids/metabolism , beta Carotene/blood
20.
Science ; 353(6302)2016 08 26.
Article in English | MEDLINE | ID: mdl-27563101

ABSTRACT

Vitamin A homeostasis is critical to normal cellular function. Retinol-binding protein (RBP) is the sole specific carrier in the bloodstream for hydrophobic retinol, the main form in which vitamin A is transported. The integral membrane receptor STRA6 mediates cellular uptake of vitamin A by recognizing RBP-retinol to trigger release and internalization of retinol. We present the structure of zebrafish STRA6 determined to 3.9-angstrom resolution by single-particle cryo-electron microscopy. STRA6 has one intramembrane and nine transmembrane helices in an intricate dimeric assembly. Unexpectedly, calmodulin is bound tightly to STRA6 in a noncanonical arrangement. Residues involved with RBP binding map to an archlike structure that covers a deep lipophilic cleft. This cleft is open to the membrane, suggesting a possible mode for internalization of retinol through direct diffusion into the lipid bilayer.


Subject(s)
Membrane Proteins/chemistry , Membrane Transport Proteins/chemistry , Retinol-Binding Proteins/chemistry , Vitamin A/metabolism , Zebrafish Proteins/chemistry , Animals , Biological Transport , Calcium/chemistry , Calmodulin/chemistry , Cryoelectron Microscopy , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Protein Binding , Protein Conformation, alpha-Helical , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Retinol-Binding Proteins/genetics , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...