Role of Vitamin A/Retinoic Acid in Regulation of Embryonic and Adult Hematopoiesis
Tóm tắt
Vitamin A is an essential micronutrient throughout life. Its physiologically active metabolite retinoic acid (RA), acting through nuclear retinoic acid receptors (RARs), is a potent regulator of patterning during embryonic development, as well as being necessary for adult tissue homeostasis. Vitamin A deficiency during pregnancy increases risk of maternal night blindness and anemia and may be a cause of congenital malformations. Childhood Vitamin A deficiency can cause xerophthalmia, lower resistance to infection and increased risk of mortality. RA signaling appears to be essential for expression of genes involved in developmental hematopoiesis, regulating the endothelial/blood cells balance in the yolk sac, promoting the hemogenic program in the aorta-gonad-mesonephros area and stimulating eryrthropoiesis in fetal liver by activating the expression of erythropoietin. In adults, RA signaling regulates differentiation of granulocytes and enhances erythropoiesis. Vitamin A may facilitate iron absorption and metabolism to prevent anemia and plays a key role in mucosal immune responses, modulating the function of regulatory T cells. Furthermore, defective RA/RARα signaling is involved in the pathogenesis of acute promyelocytic leukemia due to a failure in differentiation of promyelocytes. This review focuses on the different roles played by vitamin A/RA signaling in physiological and pathological mouse hematopoiesis duddurring both, embryonic and adult life, and the consequences of vitamin A deficiency for the blood system.
Từ khóa
Tài liệu tham khảo
Semba, 2012, The discovery of the vitamins, Int. J. Vitam. Nutr. Res., 823, 10
Adamo, 1979, Retinoid-induced adhesion in cultured, transformed mouse fibroblasts, J. Natl. Cancer Inst., 62, 1473
Kato, 1987, Retinoic acid modulates attachment of mouse fibroblasts to laminin substrates, Exp. Cell Res., 173, 450, 10.1016/0014-4827(87)90285-0
Gerster, 1997, Vitamin A-functions, dietary requirements and safety in humans, Int. J. Vitam. Nutr. Res., 67, 71
Tanumihardjo, 2011, Vitamin A: Biomarkers of nutrition for development, Am. J. Clin. Nutr., 94, 658S, 10.3945/ajcn.110.005777
Theodosiou, 2011, Vitamin A: A multifunctional tool for development, Semin. Cell Dev. Biol., 22, 603, 10.1016/j.semcdb.2011.06.001
McLaren, 2012, Manual on vitamin A deficiency disorders (VADD), World Rev. Nutr. Diet., 103, 1, 10.1159/000342734
Sanchez, 2016, Retinoic acid induces nuclear FAK translocation and reduces breast cancer cell adhesion through Moesin, FAK, and Paxillin, Mol. Cell Endocrinol., 15, 1, 10.1016/j.mce.2016.04.021
Liu, 2015, All-trans retinoic acid arrests cell cycle in leukemic bone marrow stromal cells by increasing intercellular communication through connexin 43-mediated gap junction, J. Hematol. Oncol., 7, 110, 10.1186/s13045-015-0212-7
Labrecque, 1995, A novel isoenzyme of aldehyde dehydrogenase specifically involved in the biosynthesis of 9-cis and all-trans retinoic acid, Biochem. J., 15, 681, 10.1042/bj3050681
Zhao, 1996, Molecular identification of a major retinoic-acid-synthesizing enzyme, a retinaldehyde-specific dehydrogenase, Eur. J. Biochem., 15, 15, 10.1111/j.1432-1033.1996.0015h.x
Mic, 2000, RALDH3, a retinaldehyde dehydrogenase that generates retinoic acid, is expressed in the ventral retina, otic vesicle and olfactory pit during mouse development, Mech. Dev., 97, 227, 10.1016/S0925-4773(00)00434-2
Duester, 2000, Families of retinoid dehydrogenases regulating vitamin A function: Production of visual pigment and retinoic acid, Eur. J. Biochem., 267, 4315, 10.1046/j.1432-1327.2000.01497.x
Sima, 2009, Kinetic characterization of recombinant mouse retinal dehydrogenase types 3 and 4 for retinal substrates, Biochim. Biophys. Acta, 1790, 1660, 10.1016/j.bbagen.2009.09.004
McKenna, N.J. (2012). EMBO Retinoids 2011: Mechanisms, biology and pathology of signaling by retinoic acid and retinoic acid receptors. Nucl. Recept. Signal, 10.
2014, RARA fusion genes in acute promyelocytic leukemia: A review, Expert Rev. Hematol., 7, 347, 10.1586/17474086.2014.903794
Ablain, 2014, Retinoic acid signaling in cancer: The parable of acute promyelocytic leukemia, Int. J. Cancer, 15, 2262, 10.1002/ijc.29081
Cano, 2014, Signaling by Retinoic Acid in Embryonic and Adult Hematopoiesis, J. Dev. Biol., 2, 18, 10.3390/jdb2010018
North, 1999, Cbfa2 is required for the formation of intra-aortic hematopoietic clusters, Development, 126, 2563, 10.1242/dev.126.11.2563
Jung, 2016, Hematopoiesis during development, aging, and disease, Exp. Hematol., 44, 689, 10.1016/j.exphem.2016.05.007
Bollerot, 2005, The embryonic origins of hematopoietic stem cells: A tale of hemangioblast and hemogenic endothelium, APMIS, 113, 790, 10.1111/j.1600-0463.2005.apm_317.x
Pouget, 2006, Are intra-aortic hemopoietic cells derived from endothelial cells during ontogeny?, Trends Cardiovasc. Med., 16, 128, 10.1016/j.tcm.2006.02.005
Dzierzak, 2008, Of lineage and legacy-the development of mammalian hemopoetic stem cells, Nat. Immunol., 9, 129, 10.1038/ni1560
Hirschi, 2012, Hemogenic endothelium during development and beyond, Blood, 24, 4823, 10.1182/blood-2011-12-353466
Gritz, 2016, Specification and function of hemogenic endothelium during embryogenesis, Cell Mol. Life Sci., 73, 1547, 10.1007/s00018-016-2134-0
Goldie, 2008, Cell signaling directing the formation and function of hemogenic endothelium during murine embryogenesis, Blood, 15, 3194, 10.1182/blood-2008-02-139055
Niederreither, 1999, Embryonic retinoic acid synthesis is essential for early mouse post-implantation development, Nat. Genet., 21, 444, 10.1038/7788
Lai, 2003, Retinoic acid regulates endothelial cell proliferation during vasculogenesis, Development, 130, 6465, 10.1242/dev.00887
Bohnsack, 2004, Signaling hierarchy downstream of retinoic acid that independently regulates vascular remodeling and endothelial cell proliferation, Genes Dev., 1, 1345, 10.1101/gad.1184904
Chanda, 2013, Retinoic acid signaling is essential for embryonic hematopoietic stem cell development, Cell, 155, 215, 10.1016/j.cell.2013.08.055
Li, 1993, Normal development and growth of mice carrying a targeted disruption of the alpha 1 retinoic acid receptor gene, Proc. Natl. Acad. Sci. USA, 15, 1590, 10.1073/pnas.90.4.1590
Lufkin, 1993, High postnatal lethality and testis degeneration in retinoic acid receptor alpha mutant mice, Proc. Natl. Acad. Sci. USA, 90, 7225, 10.1073/pnas.90.15.7225
Marcelo, 2013, Hemogenic endothelial cell specification requires c-Kit, Notch signaling, and p27-mediated cell-cycle control, Dev. Cell, 9, 504, 10.1016/j.devcel.2013.11.004
Dou, 2016, Medial HOXA genes demarcate haematopoietic stem cell fate during human development, Nat. Cell Biol., 18, 595, 10.1038/ncb3354
Fournier, 2016, Hoxa cluster genes determine the proliferative activity of adult mouse hematopoietic stem and progenitor cells, Blood, 7, 87
Makita, 2005, Retinoic acid, hypoxia, and GATA factors cooperatively control the onset of fetal liver erythropoietin expression and erythropoietic differentiation, Dev. Biol., 1, 59, 10.1016/j.ydbio.2005.01.001
Ottersbach, 2005, The murine placenta contains hematopoietic stem cells within the vascular labyrinth region, Dev. Cell, 8, 377, 10.1016/j.devcel.2005.02.001
Mikkola, 2005, Placenta as a site for hematopoietic stem cell development, Exp. Hematol., 33, 1048, 10.1016/j.exphem.2005.06.011
Zovein, 2008, Fate tracing reveals the endothelial origin of hematopoietic stem cells, Cell Stem Cell, 4, 625, 10.1016/j.stem.2008.09.018
Ottersbach, 2009, Analysis of the mouse placenta as a hematopoietic stem cell niche, Methods Mol. Biol., 538, 335, 10.1007/978-1-59745-418-6_17
Robin, 2009, Human placenta is a potent hematopoietic niche containing hematopoietic stem and progenitor cells throughout development, Cell Stem Cell, 5, 385, 10.1016/j.stem.2009.08.020
Ottersbach, 2012, The placenta as a haematopoietic organ, Int. J. Dev. Biol., 54, 1099, 10.1387/ijdb.093057ko
Niu, 2012, Characterization of hemangioblast in umbilical arteries of mid-gestation mouse embryos, Int. J. Hematol., 95, 632, 10.1007/s12185-012-1068-z
Sapin, 1997, Defects of the chorioallantoic placenta in mouse RXRα null fetuses, Dev. Biol., 19, 29, 10.1006/dbio.1997.8687
Wendling, 1999, Retinoid X receptors are essential for early mouse development and placentogenesis, Proc. Natl. Acad. Sci. USA, 19, 547, 10.1073/pnas.96.2.547
Pereira, 2016, Hematopoietic reprogramming in vitro informs in vivo identification of hemogenic precursors to definitive hematopoietic stem cells, Dev. Cell, 36, 525, 10.1016/j.devcel.2016.02.011
Pillay, L.M., Mackowetzky, K.J., Widen, S.A., and Waskiewicz, A.J. (2016). Somite-Derived Retinoic Acid Regulates Zebrafish Hematopoietic Stem Cell Formation. PLoS ONE, 18.
Davidson, 2010, Interaction of retinoic acid and scl controls primitive blood development, Blood, 116, 201, 10.1182/blood-2009-10-249557
Samarut, 1849, ZebRA: An overview of retinoic acid signaling during zebrafish development, Biochim. Biophys. Acta, 1849, 73, 10.1016/j.bbagrm.2014.05.030
Liang, D., Jia, W., Li, J., Li, K., and Zhao, Q. (2012). Retinoic acid signaling plays a restrictive role in zebrafish primitive myelopoiesis. PLoS ONE, 7.
Purton, 2006, RAR gamma is critical for maintaining a balance between hematopoietic stem cell self-renewal and differentiation, J. Exp. Med., 15, 1283, 10.1084/jem.20052105
Kastner, 2001, Function of RAR alpha during the maturation of neutrophils, Oncogene, 29, 7178, 10.1038/sj.onc.1204757
Dewamitta, 2014, Erythroid-extrinsic regulation of normal erythropoiesis by retinoic acid receptors, Br. J. Haematol., 164, 280, 10.1111/bjh.12578
Walkley, 2007, A microenvironment-induced myeloproliferative syndrome caused by retinoic acid receptor gamma deficiency, Cell, 15, 1097, 10.1016/j.cell.2007.05.014
Xu, 2000, Pluripotent, cytokine-dependent, hematopoietic stem cells are immortalized by constitutive Notch1 signaling, Nat. Med., 6, 1278, 10.1038/81390
Antonchuk, 2001, HOXB4 overexpression mediates very rapid stem cell regeneration and competitive hematopoietic repopulation, Exp. Hematol., 29, 1125, 10.1016/S0301-472X(01)00681-6
Dzhagalov, 2007, Regulation of CD8+ T lymphocyte effector function and macrophage inflammatory cytokine production by retinoic acid receptor gamma, J. Immunol., 15, 2113, 10.4049/jimmunol.178.4.2113
Joseph, 2016, Retinoic Acid Receptor γ Regulates B and T Lymphopoiesis via Nestin-Expressing Cells in the Bone Marrow and Thymic Microenvironments, J. Immunol., 1, 2132, 10.4049/jimmunol.1501246
Ghiaur, 2013, Regulation of human hematopoietic stem cell self-renewal by the microenvironment’s control of retinoic acid signaling, Proc. Natl. Acad. Sci. USA, 1, 16121, 10.1073/pnas.1305937110
Chute, 2006, Inhibition of aldehyde dehydrogenase and retinoid signaling induces the expansion of human hematopoietic stem cells, Proc. Natl. Acad. Sci. USA, 1, 11707, 10.1073/pnas.0603806103
Brown, G., Marchwicka, A., Cunningham, A., Toellner, K.M., and Marcinkowska, E. (2016). Antagonizing Retinoic Acid Receptors Increases Myeloid Cell Production by Cultured Human Hematopoietic Stem Cells. Arch. Immunol. Ther. Exp. (Warsz.), 13.
Hall, 2011, Essential role for retinoic acid in the promotion of CD4(+) T cell effector responses via retinoic acid receptor alpha, Immunity, 25, 435, 10.1016/j.immuni.2011.03.003
Ross, 2012, Vitamin A and retinoic acid in T cell-related immunity, Am. J. Clin. Nutr., 96, 1166S, 10.3945/ajcn.112.034637
Raverdeau, 2014, Modulation of T cell and innate immune responses by retinoic acid, J. Immunol., 1, 2953, 10.4049/jimmunol.1303245
Cassani, 2012, Vitamin A and immune regulation: Role of retinoic acid in gut-associated dendritic cell education, immune protection and tolerance, Mol. Asp. Med., 33, 63, 10.1016/j.mam.2011.11.001
Kim, 2015, Retinoic Acid Differentially Regulates the Migration of Innate Lymphoid Cell Subsets to the Gut, Immunity, 21, 107, 10.1016/j.immuni.2015.06.009
Bakdash, 2015, Retinoic acid primes human dendritic cells to induce gut-homing, IL-10-producing regulatory T cells, Mucosal Immunol., 8, 265, 10.1038/mi.2014.64
Bono, M.R., Tejon, G., Flores-Santibañez, F., Fernandez, D., Rosemblatt, M., and Sauma, D. (2016). Retinoic Acid as a Modulator of T Cell Immunity. Nutrients, 8.
Naderi, 2009, Regulation of B cell proliferation and differentiation by retinoic acid, Semin. Immunol., 21, 36, 10.1016/j.smim.2008.06.005
Ross, 2011, Vitamin A and retinoic acid in the regulation of B-cell development and antibody production, Vitam. Horm., 86, 103, 10.1016/B978-0-12-386960-9.00005-8
Holm, 2016, Myeloid cell leukemia 1 has a vital role in retinoic acid-mediated protection of Toll-like receptor 9-stimulated B cells from spontaneous and DNA damage-induced apoptosis, Immunology, 149, 62, 10.1111/imm.12629
Testa, 2015, Targeting of leukemia-initiating cells in acute promyelocytic leukemia, Stem Cell Investig., 29, 2
Cicconi, 2016, PML-RARα kinetics and impact of FLT3-ITD mutations in newly diagnosed acute promyelocytic leukemia treated with ATRA and ATO or ATRA and chemotherapy, Leukemia, 30, 1987, 10.1038/leu.2016.122
Cicconi, 2016, Current management of newly diagnosed acute promyelocytic leukemia, Ann. Oncol., 27, 1474, 10.1093/annonc/mdw171
Ma, Y., Liu, L., Jin, J., and Lou, Y. (2016). All-Trans Retinoic Acid Plus Arsenic Trioxide versus All-Trans Retinoic Acid plus Chemotherapy for Newly Diagnosed Acute Promyelocytic Leukemia: A Meta-Analysis. PLoS ONE, 8.
Tallman, 2013, National Comprehensive Cancer Network. Acute myeloid leukemia, J. Natl. Compr. Cancer Netw., 1, 1047
Orlando, 2013, Treatment of acute promyelocytic leukemia, N. Engl. J. Med., 10, 1472
Zhu, 2016, Long-term survival of acute promyelocytic leukemia patients treated with arsenic and retinoic acid, Br. J. Haematol., 174, 820, 10.1111/bjh.13809
Takahashi, 2016, High event-free survival rate with minimum-dose-anthracycline treatment in childhood acute promyelocytic leukemia: A nationwide prospective study by the Japanese Paediatric Leukemia/Lymphoma Study Group, Br. J. Haematol., 174, 437, 10.1111/bjh.14068
Platzbecker, 2017, Improved outcomes with Retinoic Acid and Arsenic Trioxide compared with Retinoic Acid and chemotherapy in Non-High-Risk Acute Promyelocytic Leukemia: Final results of the Randomized Italian-German APL0406 Trial, J. Clin. Oncol., 35, 605, 10.1200/JCO.2016.67.1982
Norsworthy, 2016, Optimal treatment strategies for high-risk acute promyelocytic leukemia, Curr. Opin. Hematol., 23, 127, 10.1097/MOH.0000000000000215
Podoltsev, N.A., Stahl, M., Zeidan, A.M., and Gore, S.D. (2016). Selecting initial treatment of acute myeloid leukemia in older adults. Blood Rev., 8.
Wang, 2016, LG-362B targets PML-RARα and blocks ATRA resistance of acute promyelocytic leukemia, Leukemia, 30, 1465, 10.1038/leu.2016.50
Wallace, A.S., Supnick, H.T., Bunaciu, R.P., and Yen, A. (2016). RRD-251 enhances all-trans retinoic acid (RA)-induced differentiation of HL-60 myeloblastic leukemia cells. Oncotarget, 17.
Atashrazm, F., Lowenthal, R.M., Dickinson, J.L., Holloway, A.F., and Woods, G.M. (2016). Fucoidan enhances the therapeutic potential of arsenic trioxide and all-trans retinoic acid in acute promyelocytic leukemia, in vitro and in vivo. Oncotarget, 14.
Shao, X., Liu, Y., Li, Y., Xian, M., Zhou, Q., Yang, B., Ying, M., and He, Q. (2016). The HER2 inhibitor TAK165 Sensitizes Human Acute Myeloid Leukemia Cells to Retinoic Acid-Induced Myeloid Differentiation by activating MEK/ERK mediated RARα/STAT1 axis. Sci. Rep., 14.
Montesinos, 2011, The differentiation syndrome in patients with acute promyelocytic leukemia: Experience of the pethema group and review of the literature, Mediterr. J. Hematol. Infect. Dis., 3, e2011059, 10.4084/mjhid.2011.059
Sanz, 2014, How we prevent and treat differentiation syndrome in patients with acute promyelocytic leukemia, Blood, 1, 2777, 10.1182/blood-2013-10-512640
Schwager, 2015, Ascorbic acid modulates cell migration in differentiated HL-60 cells and peripheral blood leukocytes, Mol. Nutr. Food Res., 59, 1513, 10.1002/mnfr.201400893
Sumi, 2016, Arsenic trioxide augments all-trans retinoic acid-induced differentiation of HL-60 cells, Life Sci., 15, 42, 10.1016/j.lfs.2016.02.054
Weng, 2016, RAF-1/MEK/ERK pathway regulates ATRA-induced differentiation in acute promyelocytic leukemia cells through C/EBPβ, C/EBPε and PU.1, Leuk. Res., 45, 68, 10.1016/j.leukres.2016.03.008
Guo, 2016, All-trans retinoic acid inhibits HOXA7 expression in leukemia cell NB4, Cell Mol. Biol., 22, 63
Liu, 2016, Effect of ATRA on the expression of HOXA5 gene in K562 cells and its relationship with cell cycle and apoptosis, Mol. Med. Rep., 13, 4221, 10.3892/mmr.2016.5086
Li, 2015, Knockdown of homeobox A5 by small hairpin RNA inhibits proliferation and enhances cytarabine chemosensitivity of acute myeloid leukemia cells, Mol. Med. Rep., 12, 6861, 10.3892/mmr.2015.4331
Mihara, 2016, All-trans retinoic acid and interferon-α increase CD38 expression on adult T-cell leukemia cells and sensitize them to T cells bearing anti-CD38 chimeric antigen receptors, Blood Cancer J., 13, 421, 10.1038/bcj.2016.30
Sashida, 2012, Epigenetic regulation of hematopoiesis, Int. J. Hematol., 96, 405, 10.1007/s12185-012-1183-x
2005, Chromatin modifying activity of leukemia associated fusion proteins, Hum. Mol. Genet., 15, 77
Rice, 2007, Epigenetic regulation of normal and malignant hematopoiesis, Oncogene, 15, 6697, 10.1038/sj.onc.1210755
Fazi, 2007, Heterochromatic gene repression of the retinoic acid pathway in acute myeloid leukemia, Blood, 15, 4432, 10.1182/blood-2006-09-045781
Przespolewski, 2016, Inhibitors of LSD1 as a potential therapy for acute myeloid leukemia, Expert Opin. Investig. Drugs, 25, 771, 10.1080/13543784.2016.1175432
Guidez, 2007, RARα-PLZF overcomes PLZF-mediated repression of CRABPI, contributing to retinoid resistance in t(11;17) acute promyelocytic leukemia, Proc. Natl. Acad. Sci. USA, 20, 18694, 10.1073/pnas.0704433104
Choi, 2014, Role of promyelocytic leukemia zinc finger (PLZF) in cell proliferation and cyclin-dependent kinase inhibitor 1A (p21WAF/CDKN1A) gene repression, J. Biol. Chem., 4, 18625, 10.1074/jbc.M113.538751
Choi, 2014, Promyelocytic leukemia zinc finger-retinoic acid receptor α (PLZF-RARα), an oncogenic transcriptional repressor of cyclin-dependent kinase inhibitor 1A (p21WAF/CDKN1A) and tumor protein p53 (TP53) genes, J. Biol. Chem., 4, 18641, 10.1074/jbc.M113.538777
Jones, 2004, RARbeta2 is a candidate tumor suppressor gene in myelofibrosis with myeloid metaplasia, Oncogene, 14, 7846, 10.1038/sj.onc.1207510
Nervi, 2007, Emerging role for microRNAs in acute promyelocytic leukemia, Curr. Top. Microbiol. Immunol., 313, 73
Haussler, 1997, The vitamin D hormone and its nuclear receptor: Molecular actions and disease states, J. Endocrinol., 154, S57
Thompson, 1998, Heterodimeric DNA binding by the vitamin D receptor and retinoid X receptors is enhanced by 1,25-dihydroxyvitamin D3 and inhibited by 9-cis-retinoic acid. Evidence for allosteric receptor interactions, J. Biol. Chem., 273, 8483, 10.1074/jbc.273.14.8483
Luong, 2005, Vitamin D compounds in leukemia, J. Steroid Biochem. Mol. Biol., 97, 195, 10.1016/j.jsbmb.2005.06.017
Kelly, 2010, A case-control study of ultraviolet radiation exposure, vitamin D, and lymphoma risk in adults, Cancer Causes Control, 21, 1265, 10.1007/s10552-010-9554-1
Jeanson, 2010, Vitamin D receptor deletion leads to increased hematopoietic stem and progenitor cells residing in the spleen, Blood, 18, 4126, 10.1182/blood-2010-04-280552
Bunce, 1997, Vitamin D and hematopoiesis, Trends Endocrinol. Metab., 8, 245, 10.1016/S1043-2760(97)00066-0
Hall, 2013, The role of vitamin D in hematologic disease and stem cell transplantation, Nutrients, 18, 2206, 10.3390/nu5062206
Yu, 2008, Failure of T cell homing, reduced CD4/CD8αα intraepithelial lymphocytes, and inflammation in the gut of vitamin D receptor KO mice, Proc. Natl. Acad. Sci. USA, 30, 20834, 10.1073/pnas.0808700106
Yu, 2011, Epigenetic reduction in invariant NKT cells following in utero vitamin D deficiency in mice, J. Immunol., 1, 1384, 10.4049/jimmunol.1002545
Yu, 2011, Invariant NKT cell defects in vitamin D receptor knockout mice prevents experimental lung inflammation, J. Immunol., 1, 4907, 10.4049/jimmunol.1101519
Grande, 2002, Physiological levels of 1alpha, 25 dihydroxyvitamin D3 induce the monocytic commitment of CD34+ hematopoietic progenitors, J. Leuk. Biol., 71, 641, 10.1189/jlb.71.4.641
Kim, 2012, Application of vitamin D and derivatives in hematological malignancies, Cancer Lett., 1, 8, 10.1016/j.canlet.2011.10.026
Marchwicka, 2014, Perspectives of differentiation therapies of acute myeloid leukemia: The search for the molecular basis of patients’ variable responses to 1,25-dihydroxyvitamin d and vitamin d analogs, Front. Oncol., 27, 125
Gocek, 2012, Opposite regulation of vitamin D receptor by ATRA in AML cells susceptible and resistant to vitamin D-induced differentiation, J. Steroid Biochem. Mol. Biol., 132, 220, 10.1016/j.jsbmb.2012.07.001
Marchwicka, 2016, Regulation of vitamin D receptor expression by retinoic acid receptor alpha in acute myeloid leukemia cells, J. Steroid Biochem. Mol. Biol., 159, 121, 10.1016/j.jsbmb.2016.03.013
Kuwata, 2000, Vitamin A deficiency in mice causes a systemic expansion of myeloid cells, Blood, 1, 3349, 10.1182/blood.V95.11.3349
Walkley, 2007, Rb regulates interactions between hematopoietic stem cells and their bone marrow microenvironment, Cell, 15, 1081, 10.1016/j.cell.2007.03.055
Siqueira, 2014, Vitamin A deficiency modulates iron metabolism via ineffective erythropoiesis, J. Nutr. Biochem., 25, 1035, 10.1016/j.jnutbio.2014.05.005
Jiang, 2012, Vitamin a deficiency aggravates iron deficiency by upregulating the expression of iron regulatory protein-2, Nutrition, 28, 281, 10.1016/j.nut.2011.08.015
Citelli, 2012, Vitamin a modulates the expression of genes involved in iron bioavailability, Biol. Trace Element Res., 149, 64, 10.1007/s12011-012-9397-6
Wiseman, E.M., Bar-El Dadon, S., and Reifen, R. (2016). The vicious cycle of vitamin A deficiency: A review. Crit. Rev. Food Sci. Nutr., 29.
Michelazzo, 2013, The influence of vitamin A supplementation on iron status, Nutrients, 7, 4399, 10.3390/nu5114399
Akhtar, 2013, Prevalence of vitamin A deficiency in South Asia: Causes, outcomes, and possible remedies, J. Health Popul. Nutr., 31, 413
Hall, 2011, The role of retinoic acid in tolerance and immunity, Immunity, 22, 13, 10.1016/j.immuni.2011.07.002
Sommer, 2012, A global clinical view on vitamin A and carotenoids, Am. J. Clin. Nutr., 96, 1204S, 10.3945/ajcn.112.034868
Micronutrient Deficiencies: Vitamin A Deficiency. Available online: http://www.who.int/nutrition/topics/vad/en/.
WHO Global Database on Vitamin A Deficiency. Available online: http://www.who.int/vmnis/database/vitamina/en/.
Bailey, 2015, The epidemiology of global micronutrient deficiencies, Ann. Nutr. Metab., 66, 22, 10.1159/000371618
Mayo-Wilson, E., Imdad, A., Herzer, K., Yakoob, M.Y., and Bhutta, Z.A. (2011). Vitamin A supplements for preventing mortality, illness, and blindness in children aged under 5: Systematic review and meta-analysis. BMJ, 25.
Kraemer, K., and Zimmermann, M.B. (2007). Nutritional Anemia, Sight and Life Press.
Zhang, 2010, Effect of biscuits fortified with different doses of vitamin A on indices of vitamin A status, haemoglobin and physical growth levels of pre-school children in Chongqing, Public Health Nutr., 13, 1462, 10.1017/S1368980010000820
Palmer, 2015, Maternal vitamin A supplementation increases natural antibody concentrations of preadolescent offspring in rural Nepal, Nutrition, 31, 813, 10.1016/j.nut.2014.11.016
Arguello, 2015, Circulating IGF-1 may mediate improvements in haemoglobin associated with vitamin A status during pregnancy in rural Nepalese women, Asia Pac. J. Clin. Nutr., 24, 128
Gebremedhin, 2014, Prevalence and correlates of maternal anemia in rural Sidama, Southern Ethiopia, Afr. J. Reprod. Health, 18, 44
Cardoso, M.A., Augusto, R.A., Bortolini, G.A., Oliveira, C.S., Tietzman, D.C., Sequeira, L.A., Hadler, M.C., Peixoto- Mdo, R., Muniz, P.T., and Vitolo, M.R. (2016). Effect of Providing Multiple Micronutrients in Powder through Primary Healthcare on Anemia in Young Brazilian Children: A Multicentre Pragmatic Controlled Trial. PLoS ONE, 14.
Thompson, B., and Amoroso, L. (2011). Combating Micronutrient Deficiencies: Food-based Approaches, CAB International and FAO.
Nair, 2016, Food-Based Interventions to Modify Diet Quality and Diversity to Address Multiple Micronutrient Deficiency, Front. Public Health, 5, 277
Awasthi, 2013, Population deworming every 6 months with albendazole in 1 million pre-school children in North India: DEVTA, a cluster-randomised trial, Lancet, 27, 1478, 10.1016/S0140-6736(12)62126-6
Sommer, 2013, Vitamin A supplementation in Indian children, Lancet, 17, 591, 10.1016/S0140-6736(13)60645-5
Benn, 2015, An enigma: Why vitamin A supplementation does not always reduce mortality even though vitamin A deficiency is associated with increased mortality, Int. J. Epidemiol., 44, 906, 10.1093/ije/dyv117
Dary, 2002, International Vitamin A Consultative Group. Food fortification to reduce vitamin A deficiency: International Vitamin A Consultative Group recommendations, J. Nutr., 132, 2927S, 10.1093/jn/132.9.2927S
Gannon, 2014, Biofortified orange maize is as efficacious as a vitamin A supplement in Zambian children even in the presence of high liver reserves of vitamin A: A community-based, randomized placebo-controlled trial, Am. J. Clin. Nutr., 100, 1541, 10.3945/ajcn.114.087379
Palmer, 2016, Provitamin A Carotenoid-Biofortified Maize Consumption Increases Pupillary Responsiveness among Zambian Children in a Randomized Controlled Trial, J. Nutr., 146, 2551, 10.3945/jn.116.239202
Talsma, 2016, Biofortified yellow cassava and vitamin A status of Kenyan children: A randomized controlled trial, Am. J. Clin. Nutr., 103, 258, 10.3945/ajcn.114.100164
Paine, 2005, Improving the nutritional value of Golden Rice through increased pro-vitamin A content, Nat. Biotechnol., 23, 482, 10.1038/nbt1082
Moghissi, 2016, Golden rice: scientific, regulatory and public information processes of a genetically modified organism, Crit. Rev. Biotechnol., 36, 535