1
|
Fainsod A, Vadigepalli R. Rethinking retinoic acid self-regulation: A signaling robustness network approach. Curr Top Dev Biol 2024; 161:113-141. [PMID: 39870431 DOI: 10.1016/bs.ctdb.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
All-trans retinoic acid (ATRA) signaling is a major pathway regulating numerous differentiation, proliferation, and patterning processes throughout life. ATRA biosynthesis depends on the nutritional availability of vitamin A and other retinoids and carotenoids, while it is sensitive to dietary and environmental toxicants. This nutritional and environmental influence requires a robustness response that constantly fine-tunes the ATRA metabolism to maintain a context-specific, physiological range of signaling levels. The ATRA metabolic and signaling network is characterized by the existence of multiple enzymes, transcription factors, and binding proteins capable of performing the same activity. The partial spatiotemporal expression overlap of these enzymes and proteins yields different network compositions in the cells and tissues where this pathway is active. Genetic polymorphisms affecting the activity of individual network components further impact the network composition variability and the self-regulatory feedback response to ATRA fluctuations. Experiments directly challenging the robustness response uncovered a Pareto optimality in the ATRA network, such that some genetic backgrounds efficiently deal with excess ATRA but are very limited in their robustness response to reduced ATRA and vice versa. We discuss a network-focused framework to describe the robustness response and the Pareto optimality of the ATRA metabolic and signaling network.
Collapse
Affiliation(s)
- Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
2
|
Nakamura M, Sandell LL. Multiple roles for retinoid signaling in craniofacial development. Curr Top Dev Biol 2024; 161:33-57. [PMID: 39870438 DOI: 10.1016/bs.ctdb.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Retinoic acid (RA) signaling plays multiple essential roles in development of the head and face. Animal models with mutations in genes involved in RA signaling have enabled understanding of craniofacial morphogenic processes that are regulated by the retinoid pathway. During craniofacial morphogenesis RA signaling is active in spatially restricted domains defined by the expression of genes involved in RA production and RA breakdown. The spatial distribution of RA signaling changes with progressive development, corresponding to a multiplicity of craniofacial developmental processes that are regulated by RA. One important role of RA signaling occurs in the hindbrain. There RA contributes to specification of the anterior-posterior (AP) axis of the developing CNS and to the neural crest cells (NCC) which form the bones and nerves of the face and pharyngeal region. In the optic vesicles and frontonasal process RA orchestrates development of the midface, eyes, and nasal airway. Additional roles for RA in craniofacial development include regulation of submandibular salivary gland development and maintaining patency in the sutures of the cranial vault.
Collapse
Affiliation(s)
- Masahiro Nakamura
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, United States
| | - Lisa L Sandell
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, United States.
| |
Collapse
|
3
|
Yu M, Ju H, Ye N, Chen J, Sun L, Wu X, Xu H, Shen Q. Vitamin A Deficiency Disturbs Ret Expression and Induces Urinary Tract Developmental Abnormalities in Mice. Am J Nephrol 2024:1-11. [PMID: 39397601 DOI: 10.1159/000541289] [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: 07/22/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024]
Abstract
INTRODUCTION Moderate vitamin A levels during pregnancy are strongly related to normal embryonic development in both animal models and population studies. Abnormal development of urinary tract system is linked to either an excess or a shortage of vitamin A. The relationships among maternal vitamin A deficiency prior to conception, moderate vitamin A supplementation during pregnancy, and abnormal urinary system development in offspring are unclear. METHODS By creating preconception and preconception + pregnancy vitamin A insufficiency mouse models, we investigated whether moderate vitamin A treatment during pregnancy may reduce the prevalence of CAKUT and increase distant vitamin A levels in offspring, as well as any potential pathways involved. RESULTS We effectively established a prepregnancy vitamin A-deficient mouse model by providing a particular diet with or without vitamin A for 4 weeks. The offspring of the hypovitaminosis A model group presented a greater proportion of neonatal urinary tract developmental malformations. Abnormalities in ureteral bud emergence and key molecules during renal development, such as p-Plcγ and Ret, may be the primary causes of offspring development of CAKUT as a result of mothers' hypovitaminosis A. Normal vitamin A diets, on the other hand, may help mitigate the teratogenic consequences of prepregnancy hypovitaminosis A, as well as defects produced by ureteral budding and major molecular changes. CONCLUSION In contrast, the administration of normal vitamin A feeds during pregnancy may ameliorate the teratogenic effects of prepregnancy hypovitaminosis A to a certain extent and may also ameliorate the abnormalities associated with ureteral budding and key molecular changes.
Collapse
Affiliation(s)
- Minghui Yu
- Department of Nephrology, Children's Hospital of Fudan University, Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
| | - Haixin Ju
- Department of Nephrology, Children's Hospital of Fudan University, Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
| | - Ningli Ye
- Department of Nephrology, Children's Hospital of Fudan University, Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
| | - Jing Chen
- Department of Nephrology, Children's Hospital of Fudan University, Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
| | - Lei Sun
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaohui Wu
- Department of Nephrology, Children's Hospital of Fudan University, Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Hong Xu
- Department of Nephrology, Children's Hospital of Fudan University, Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
- National Key Laboratory of Kidney Diseases, Beijing, China
| | - Qian Shen
- Department of Nephrology, Children's Hospital of Fudan University, Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, China
- National Key Laboratory of Kidney Diseases, Beijing, China
| |
Collapse
|
4
|
Carrazana R, Espinoza F, Ávila A. Mechanistic perspective on the actions of vitamin a in autism spectrum disorder etiology. Neuroscience 2024; 554:72-82. [PMID: 39002756 DOI: 10.1016/j.neuroscience.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 05/07/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Vitamin A (VA) has many functions in the body, some of which are key for the development and functioning of the nervous system, while some others might indirectly influence neural function. Both hypovitaminosis and hypervitaminosis A can lead to clinical manifestations of concern for individuals and for general global health. Scientific evidence on the link between VA and autism spectrum disorder (ASD) is growing, with some clinical studies and accumulating results obtained from basic research using cellular and animal models. Remarkably, it has been shown that VA deficiency can exacerbate autistic symptomatology. In turn, VA supplementation has been shown to be able to improve autistic symptomatology in selected groups of individuals with ASD. However, it is important to recognize that ASD is a highly heterogeneous condition. Therefore, it is important to clarify how and when VA supplementation can be of benefit for affected individuals. Here we delve into the relationship between VA and ASD, discussing clinical observations and mechanistic insights obtained from research on selected autistic syndromes and laboratory models to advance in defining how the VA signaling pathway can be exploited for treatment of ASD.
Collapse
Affiliation(s)
- Ramón Carrazana
- Neurodevelopmental Biology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Francisca Espinoza
- Neurodevelopmental Biology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Ariel Ávila
- Neurodevelopmental Biology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile.
| |
Collapse
|
5
|
Stunes AK, Mosti MP, Gustafsson MK, Børsting T, Thorsby PM, Stafne SN, Syversen U. Maternal vitamin A and D status in second and third trimester of pregnancy and bone mineral content in offspring at nine years of age. Front Endocrinol (Lausanne) 2024; 15:1417656. [PMID: 39006361 PMCID: PMC11239386 DOI: 10.3389/fendo.2024.1417656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/17/2024] [Indexed: 07/16/2024] Open
Abstract
Introduction Maternal nutritional and vitamin status during pregnancy may have long-term effects on offspring health and disease. The aim of this study was to examine the associations between maternal vitamin A and D status in pregnancy and offspring bone mineral content (BMC) at nine years of age. Methods This is a post-hoc study of a randomized control trial including 855 pregnant women from two Norwegian cities; Trondheim and Stavanger. The women were randomized into an exercise intervention or standard antenatal care. Mother and child pairs for the present study were recruited from those still living in Trondheim after 8-10 years. Serum vitamin A (retinol) and vitamin D (25(OH)D) were measured in the 2nd and 3rd trimesters of pregnancy, and active vitamin D (1,25(OH)2D) in serum was measured in a subgroup. Spine BMC and trabecular bone score were measured in the children at nine years of age. Associations were analyzed with linear regression models. Results A total of 119 mother and child pairs were included in the analyses. Vitamin A insufficiency (retinol< 1.05 µmol/L) and vitamin D deficiency (25(OH)D< 50 mmol/L) increased from ~7% to ~43% and from ~28% to ~33%, respectively, from the 2nd to the 3rd trimester. An increase in serum 1,25(OH)2D from the 2nd to the 3rd trimester was observed in the subgroup. There was a negative association between serum retinol in the 2nd trimester and spine BMC in the boys, but not in the girls, when adjusted for maternal and child confounders. No other associations between maternal serum vitamin A or D and BMC in the children were found. Conclusion We observed a high prevalence of vitamin A insufficiency and vitamin D deficiency during pregnancy. A negative association between mid-pregnancy vitamin A status and spine BMC was observed in boys, but not girls, while no associations were found between maternal vitamin D status and child BMC. The implications of optimal vitamin A and D status in pregnancy for offspring bone health, remains a subject for further investigations.
Collapse
Affiliation(s)
- Astrid Kamilla Stunes
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Center for Oral Health Services and Research, Mid-Norway (TkMidt), Trondheim, Norway
| | - Mats Peder Mosti
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Research and Development, Clinic of Substance Use and Addiction Medicine, St. Olavs University Hospital, Trondheim, Norway
| | - Miriam Katarina Gustafsson
- Department of Public Health and Nursing, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Torunn Børsting
- Center for Oral Health Services and Research, Mid-Norway (TkMidt), Trondheim, Norway
- Department of Public Health and Nursing, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Per Medbøe Thorsby
- Hormone Laboratory, Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Signe Nilssen Stafne
- Department of Public Health and Nursing, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinic of Rehabilitation, St. Olavs University Hospital, Trondheim, Norway
| | - Unni Syversen
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Endocrinology, Clinic of Medicine, St. Olavs University Hospital, Trondheim, Norway
| |
Collapse
|
6
|
do Amaral CC, Nedel F, Ferrúa CP, Garcia TF, Corrêa GP, Giorgi R, Longoni dos Santos A, de Assis AM, de Avila Quevedo L, Ghisleni GC, de Matos MB, Pinheiro KAT, Trettim JP, Pinheiro RT. Maternal hsa-miR-423-5p associated with the cognitive development of babies in pregnant women without mental disorders. Front Hum Neurosci 2024; 18:1322820. [PMID: 38487105 PMCID: PMC10937415 DOI: 10.3389/fnhum.2024.1322820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/13/2024] [Indexed: 03/17/2024] Open
Abstract
Background MicroRNAs (miRNAs) are small non-coding RNAs capable of regulating gene expression post-transcriptionally. MiRNAs are recognized as key regulators of diverse biological and developmental processes. During the pregnancy-puerperal cycle, numerous changes occur in the female body for the formation, growth, and development of the baby. After birth, there is a critical period in child development, as rapid gains in the physical, cognitive, and socio-emotional domains constitute the "building blocks" of children's later growth. Objective The aim of this study was to investigate the association between maternal expression of hsa-miR-423-5p during the first and second trimesters of pregnancy and neurocognitive development at 90 days of life in infants. Methods: This is a longitudinal study included in a population-based cohort study, carried out in a city in southern Brazil. The Bayley III was used to assess the babies' cognitive development. Blood samples from mothers were obtained for RNA extraction from serum and analysis of miRNA expression by qRT-PCR. Results In total, 87 dyads (mother-baby) were included. The average gestational age was 15.86 weeks (SD ± 5.55). An association of maternal miRNA with infant cognitive development was found; as maternal miR-423-5p increases, infants' cognitive development increases by 2.40 (95% CI 0.37; 4.43, p = 0.021) points at 3 months of age. Conclusion In this context, it is suggested to use this miRNA as a biomarker of child neurocognitive development detectable in the prenatal period, thus allowing the planning of early interventions.
Collapse
Affiliation(s)
- Cainá Corrêa do Amaral
- Post-Graduate Program in Health and Behavior, Catholic University of Pelotas, Pelotas, Brazil
| | - Fernanda Nedel
- Anatomy Department, Federal University of Pelotas, Pelotas, Brazil
| | - Camila Perelló Ferrúa
- Post-Graduate Program in Health and Behavior, Catholic University of Pelotas, Pelotas, Brazil
| | - Tiago Fernandez Garcia
- Post-Graduate Program in Health and Behavior, Catholic University of Pelotas, Pelotas, Brazil
| | | | - Roberta Giorgi
- Post-Graduate Program in Health and Behavior, Catholic University of Pelotas, Pelotas, Brazil
| | | | | | | | | | - Mariana Bonati de Matos
- Post-Graduate Program in Health and Behavior, Catholic University of Pelotas, Pelotas, Brazil
| | | | | | | |
Collapse
|
7
|
Gonzalez-Ramiro H, Gil MA, Cuello C, Cambra JM, Gonzalez-Plaza A, Vazquez JM, Vazquez JL, Rodriguez-Martinez H, Lucas-Sanchez A, Parrilla I, Martinez CA, Martinez EA. The Use of a Brief Synchronization Treatment after Weaning, Combined with Superovulation, Has Moderate Effects on the Gene Expression of Surviving Pig Blastocysts. Animals (Basel) 2023; 13:ani13091568. [PMID: 37174605 PMCID: PMC10177444 DOI: 10.3390/ani13091568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/21/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
The combination of estrus synchronization and superovulation (SS) treatments causes alterations in ovarian and endometrial gene expression patterns, resulting in abnormal follicle and oocyte growth, fertilization, and embryo development. However, the impact of combined SS treatments on the transcriptome of the surviving embryos remains unidentified. In this study, we examined gene expression changes in day 6 blastocysts that survived a brief regimen of synchronization treatment combined with superovulation. The sows were included in one of three groups: SS7 group (n = 6), sows were administered Altrenogest (ALT) 7 days from the day of weaning and superovulated with eCG 24 h after the end of ALT treatment and hCG at the onset of estrus; SO group (n = 6), ALT nontreated sows were superovulated with eCG 24 h postweaning and hCG at the onset of estrus; control group (n = 6), weaned sows displaying natural estrus. Six days after insemination, the sows underwent a surgical intervention for embryo collection. Transcriptome analysis was performed on blastocyst-stage embryos with good morphology. Differentially expressed genes (DEGs) between groups were detected using one-way ANOVA with an un-adjusted p-value < 0.05 and a fold change </> 1.5. The effect of SO treatment on the number of altered pathways and DEGs within each pathway was minimal. Only four pathways were disrupted comprising only a total of four altered transcripts, which were not related to reproductive functions or embryonic development. On the other hand, the surviving blastocysts subjected to SS7 treatments exhibited moderate gene expression changes in terms of DEGs and fold changes, with seven pathways disrupted containing a total of 10 transcripts affected. In this case, the up-regulation of certain pathways, such as the metabolic pathway, with two up-regulated genes associated with reproductive functions, namely RDH10 and SPTLC2, may suggest suboptimal embryo quality, while the down-regulation of others, such as the glutathione metabolism pathway, with down-regulated genes related to cellular detoxification of reactive oxygen species, namely GSTK1 and GSTO1, could depress the embryos' response to oxidative stress, thereby impairing subsequent embryo development. The gene expression changes observed in the present study in SS7 embryos, along with previous reports indicating SS7 can negatively affect fertilization, embryo production, and reproductive tract gene expression, make its use in embryo transfer programs unrecommendable.
Collapse
Affiliation(s)
- Henar Gonzalez-Ramiro
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
- Department of Research and Development, Grupo Agropor I+D+I, AIE, 30565 Murcia, Spain
| | - Maria A Gil
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
| | - Cristina Cuello
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
| | - Josep M Cambra
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
| | - Alejandro Gonzalez-Plaza
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
| | - Juan M Vazquez
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
| | - Jose L Vazquez
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
| | - Heriberto Rodriguez-Martinez
- Department of Biomedical & Clinical Sciences (BKV), BKH/Obstetrics & Gynecology, Faculty of Medicine and Health Sciences, Linköping University, 58185 Linköping, Sweden
| | | | - Inmaculada Parrilla
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
| | - Cristina A Martinez
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA-CSIC), 28040 Madrid, Spain
| | - Emilio A Martinez
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
| |
Collapse
|
8
|
Wang F, Tang Y, Cai Y, Yang R, Wang Z, Wang X, Yang Q, Wang W, Tian J, An L. Intrafollicular Retinoic Acid Signaling Is Important for Luteinizing Hormone-Induced Oocyte Meiotic Resumption. Genes (Basel) 2023; 14:genes14040946. [PMID: 37107703 PMCID: PMC10137601 DOI: 10.3390/genes14040946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/15/2023] [Accepted: 04/16/2023] [Indexed: 04/29/2023] Open
Abstract
It has been clear that retinoic acid (RA), the most active vitamin A (VA) derivative, plays a central role in governing oocyte meiosis initiation. However, it has not been functionally determined if RA participates in luteinizing hormone (LH)-induced resumption from long-lasting oocyte meiotic arrest, which is essential for haploid oocyte formation. In the present study, using well-established in vivo and in vitro models, we identified that intrafollicular RA signaling is important for normal oocyte meiotic resumption. A mechanistic study indicated that mural granulosa cells (MGCs) are the indispensable follicular compartment for RA-prompted meiotic resumption. Moreover, retinoic acid receptor (RAR) is essential for mediating RA signaling to regulate meiotic resumption. Furthermore, we found zinc finger protein 36 (ZFP36) is the transcriptional target of RAR. Both RA signaling and epidermal growth factor (EGF) signaling were activated in MGCs in response to LH surge, and two intrafollicular signalings cooperate to induce rapid Zfp36 upregulation and Nppc mRNA decrease, which is critical to LH-induced meiotic resumption. These findings extend our understanding of the role of RA in oocyte meiosis: RA not only governs meiotic initiation but also regulates LH-induced meiotic resumption. We also emphasize the importance of LH-induced metabolic changes in MGCs in this process.
Collapse
Affiliation(s)
- Fupeng Wang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Yawen Tang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Yijie Cai
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Ran Yang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Zongyu Wang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Xiaodong Wang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Qianying Yang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Wenjing Wang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Jianhui Tian
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Lei An
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| |
Collapse
|
9
|
Vitamin A- and D-Deficient Diets Disrupt Intestinal Antimicrobial Peptide Defense Involving Wnt and STAT5 Signaling Pathways in Mice. Nutrients 2023; 15:nu15020376. [PMID: 36678247 PMCID: PMC9863741 DOI: 10.3390/nu15020376] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/14/2022] [Accepted: 01/04/2023] [Indexed: 01/15/2023] Open
Abstract
Vitamin A and D deficiencies are associated with immune modulatory effects and intestinal barrier impairment. However, the underlying mechanisms remain unclear. C57BL/6J mice were fed either a diet lacking in vitamin A (VAd), vitamin D (VDd) or a control diet (CD) for 12 weeks. Gut barrier function, antimicrobial peptide (AMP) defense and regulatory pathways were assessed. VAd mice compared to CD mice showed a reduced villus length in the ileum (p < 0.01) and decreased crypt depth in the colon (p < 0.05). In both VAd- and VDd-fed mice, ileal α-defensin 5 (p < 0.05/p < 0.0001 for VAd/VDd) and lysozyme protein levels (p < 0.001/p < 0.0001) were decreased. Moreover, mRNA expression of lysozyme (p < 0.05/p < 0.05) and total cryptdins (p < 0.001/p < 0.01) were reduced compared to controls. Furthermore, matrix metalloproteinase-7 (Mmp7) mRNA (p < 0.0001/p < 0.001) as well as components of the Wnt signaling pathway were decreased. VAd- and VDd-fed mice, compared to control mice, exhibited increased expression of pro-inflammatory markers and β-defensins in the colon. Organoid cell culture confirmed that vitamins A and D regulate AMP expression, likely through the Jak/STAT5 signaling pathway. In conclusion, our data show that vitamin A and D regulate intestinal antimicrobial peptide defense through Wnt and STAT5 signaling pathways.
Collapse
|
10
|
Burns NG, Kardon G. The role of genes and environment in the etiology of congenital diaphragmatic hernias. Curr Top Dev Biol 2022; 152:115-138. [PMID: 36707209 PMCID: PMC10923182 DOI: 10.1016/bs.ctdb.2022.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Structural birth defects are a common cause of abnormalities in newborns. While there are cases of structural birth defects arising due to monogenic defects or environmental exposures, many birth defects are likely caused by a complex interaction between genes and the environment. A structural birth defect with complex etiology is congenital diaphragmatic hernias (CDH), a common and often lethal disruption in diaphragm development. Mutations in more than 150 genes have been implicated in CDH pathogenesis. Although there is generally less evidence for a role for environmental factors in the etiology of CDH, deficiencies in maternal vitamin A and its derivative embryonic retinoic acid are strongly associated with CDH. However, the incomplete penetrance of CDH-implicated genes and environmental factors such as vitamin A deficiency suggest that interactions between genes and environment may be necessary to cause CDH. In this review, we examine the genetic and environmental factors implicated in diaphragm and CDH development. In addition, we evaluate the potential for gene-environment interactions in CDH etiology, focusing on the potential interactions between the CDH-implicated gene, Gata4, and maternal vitamin A deficiency.
Collapse
Affiliation(s)
- Nathan G Burns
- Department of Human Genetics, University of Utah, Salt Lake City, UT, United States
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT, United States.
| |
Collapse
|
11
|
Pierro JD, Ahir BK, Baker NC, Kleinstreuer NC, Xia M, Knudsen TB. Computational model for fetal skeletal defects potentially linked to disruption of retinoic acid signaling. Front Pharmacol 2022; 13:971296. [PMID: 36172177 PMCID: PMC9511990 DOI: 10.3389/fphar.2022.971296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
All-trans retinoic acid (ATRA) gradients determine skeletal patterning morphogenesis and can be disrupted by diverse genetic or environmental factors during pregnancy, leading to fetal skeleton defects. Adverse Outcome Pathway (AOP) frameworks for ATRA metabolism, signaling, and homeostasis allow for the development of new approach methods (NAMs) for predictive toxicology with less reliance on animal testing. Here, a data-driven model was constructed to identify chemicals associated with both ATRA pathway bioactivity and prenatal skeletal defects. The phenotype data was culled from ToxRefDB prenatal developmental toxicity studies and produced a list of 363 ToxRefDB chemicals with altered skeletal observations. Defects were classified regionally as cranial, post-cranial axial, appendicular, and other (unspecified) features based on ToxRefDB descriptors. To build a multivariate statistical model, high-throughput screening bioactivity data from >8,070 chemicals in ToxCast/Tox21 across 10 in vitro assays relevant to the retinoid signaling system were evaluated and compared to literature-based candidate reference chemicals in the dataset. There were 48 chemicals identified for effects on both in vivo skeletal defects and in vitro ATRA pathway targets for computational modeling. The list included 28 chemicals with prior evidence of skeletal defects linked to retinoid toxicity and 20 chemicals without prior evidence. The combination of thoracic cage defects and DR5 (direct repeats of 5 nucleotides for RAR/RXR transactivation) disruption was the most frequently occurring phenotypic and target disturbance, respectively. This data model provides valuable AOP elucidation and validates current mechanistic understanding. These findings also shed light on potential avenues for new mechanistic discoveries related to ATRA pathway disruption and associated skeletal dysmorphogenesis due to environmental exposures.
Collapse
Affiliation(s)
- Jocylin D. Pierro
- Center for Computational Toxicology and Exposure (CCTE), Computational Toxicology and Bioinformatics Branch (CTBB), Office of Research and Development (ORD), U.S. Environmental Protection Agency (USEPA), Research Triangle Park, NC, United States
| | - Bhavesh K. Ahir
- Eurofins Medical Device Testing, Lancaster, PA, United States
| | - Nancy C. Baker
- Scientific Computing and Data Curation Division (SCDCD), Leidos Contractor, Center for Computational Toxicology and Exposure (CCTE), USEPA/ORD, Research Triangle Park, NC, United States
| | - Nicole C. Kleinstreuer
- Interagency Center for the Evaluation of Alternative Toxicological Methods (NICEATM), National Toxicology Program, National Institutes of Health, Research Triangle Park, NC, United States
| | - Menghang Xia
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Thomas B. Knudsen
- Center for Computational Toxicology and Exposure (CCTE), Computational Toxicology and Bioinformatics Branch (CTBB), Office of Research and Development (ORD), U.S. Environmental Protection Agency (USEPA), Research Triangle Park, NC, United States
| |
Collapse
|
12
|
Gur M, Bendelac-Kapon L, Shabtai Y, Pillemer G, Fainsod A. Reduced Retinoic Acid Signaling During Gastrulation Induces Developmental Microcephaly. Front Cell Dev Biol 2022; 10:844619. [PMID: 35372345 PMCID: PMC8967241 DOI: 10.3389/fcell.2022.844619] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/24/2022] [Indexed: 12/21/2022] Open
Abstract
Retinoic acid (RA) is a central signaling molecule regulating multiple developmental decisions during embryogenesis. Excess RA induces head malformations, primarily by expansion of posterior brain structures at the expense of anterior head regions, i.e., hindbrain expansion. Despite this extensively studied RA teratogenic effect, a number of syndromes exhibiting microcephaly, such as DiGeorge, Vitamin A Deficiency, Fetal Alcohol Syndrome, and others, have been attributed to reduced RA signaling. This causative link suggests a requirement for RA signaling during normal head development in all these syndromes. To characterize this novel RA function, we studied the involvement of RA in the early events leading to head formation in Xenopus embryos. This effect was mapped to the earliest RA biosynthesis in the embryo within the gastrula Spemann-Mangold organizer. Head malformations were observed when reduced RA signaling was induced in the endogenous Spemann-Mangold organizer and in the ectopic organizer of twinned embryos. Two embryonic retinaldehyde dehydrogenases, ALDH1A2 (RALDH2) and ALDH1A3 (RALDH3) are initially expressed in the organizer and subsequently mark the trunk and the migrating leading edge mesendoderm, respectively. Gene-specific knockdowns and CRISPR/Cas9 targeting show that RALDH3 is a key enzyme involved in RA production required for head formation. These observations indicate that in addition to the teratogenic effect of excess RA on head development, RA signaling also has a positive and required regulatory role in the early formation of the head during gastrula stages. These results identify a novel RA activity that concurs with its proposed reduction in syndromes exhibiting microcephaly.
Collapse
|
13
|
Engelbrecht E, Metzler MA, Sandell LL. Retinoid signaling regulates angiogenesis and blood-retinal barrier integrity in neonatal mouse retina. Microcirculation 2022; 29:e12752. [PMID: 35203102 DOI: 10.1111/micc.12752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/24/2022] [Accepted: 02/21/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The neonatal mouse retina is a well-characterized experimental model for investigating factors impacting retinal angiogenesis and inner blood-retinal barrier (BRB) integrity. Retinoic acid (RA) is an essential signaling molecule. RA is needed for vasculogenic development in embryos and endothelial barrier integrity in zebrafish retina and adult mouse brain, however the function of this signaling molecule in developing mammalian retinal vasculature remains unknown. This study aims to investigate the role of RA signaling in angiogenesis and inner BRB integrity in mouse neonatal retina. METHODS RA distribution in the developing neurovascular retina was assessed in mice carrying an RA-responsive transgene. RA function in retinal angiogenesis was determined by treating C57BL/6 neonatal pups with a pharmacological inhibitor of RA signaling BMS493 or control vehicle. BRB integrity assessed by monitoring leakage of injected tracer into extravascular retinal tissue. RESULTS RA signaling activity is present in peripheral astrocytes in domains corresponding to RA activity of the underlying neural retina. RA inhibition impaired retinal angiogenesis and reduced endothelial cell proliferation. RA inhibition also compromised BRB integrity. Vascular leakage was not associated with altered expression of CLDN5, PLVAP, LEF1 or VEcad. CONCLUSIONS RA signaling is needed for angiogenesis and integrity of the BRB in the neonatal mouse retina.
Collapse
Affiliation(s)
- Eric Engelbrecht
- University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Melissa A Metzler
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, 40202, USA
| | - Lisa L Sandell
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, 40202, USA
| |
Collapse
|
14
|
Parihar M, Bendelac-Kapon L, Gur M, Abbou T, Belorkar A, Achanta S, Kinberg K, Vadigepalli R, Fainsod A. Retinoic Acid Fluctuation Activates an Uneven, Direction-Dependent Network-Wide Robustness Response in Early Embryogenesis. Front Cell Dev Biol 2021; 9:747969. [PMID: 34746144 PMCID: PMC8564372 DOI: 10.3389/fcell.2021.747969] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/23/2021] [Indexed: 01/15/2023] Open
Abstract
Robustness is a feature of regulatory pathways to ensure signal consistency in light of environmental changes or genetic polymorphisms. The retinoic acid (RA) pathway, is a central developmental and tissue homeostasis regulatory signal, strongly dependent on nutritional sources of retinoids and affected by environmental chemicals. This pathway is characterized by multiple proteins or enzymes capable of performing each step and their integration into a self-regulating network. We studied RA network robustness by transient physiological RA signaling disturbances followed by kinetic transcriptomic analysis of the recovery during embryogenesis. The RA metabolic network was identified as the main regulated module to achieve signaling robustness using an unbiased pattern analysis. We describe the network-wide responses to RA signal manipulation and found the feedback autoregulation to be sensitive to the direction of the RA perturbation: RA knockdown exhibited an upper response limit, whereas RA addition had a minimal feedback-activation threshold. Surprisingly, our robustness response analysis suggests that the RA metabolic network regulation exhibits a multi-objective optimization, known as Pareto optimization, characterized by trade-offs between competing functionalities. We observe that efficient robustness to increasing RA is accompanied by worsening robustness to reduced RA levels and vice versa. This direction-dependent trade-off in the network-wide feedback response, results in an uneven robustness capacity of the RA network during early embryogenesis, likely a significant contributor to the manifestation of developmental defects.
Collapse
Affiliation(s)
- Madhur Parihar
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Liat Bendelac-Kapon
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Jerusalem, Israel
| | - Michal Gur
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Jerusalem, Israel
| | - Tali Abbou
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Jerusalem, Israel
| | - Abha Belorkar
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Sirisha Achanta
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Keren Kinberg
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Jerusalem, Israel
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Jerusalem, Israel
| |
Collapse
|
15
|
Comai GE, Tesařová M, Dupé V, Rhinn M, Vallecillo-García P, da Silva F, Feret B, Exelby K, Dollé P, Carlsson L, Pryce B, Spitz F, Stricker S, Zikmund T, Kaiser J, Briscoe J, Schedl A, Ghyselinck NB, Schweitzer R, Tajbakhsh S. Local retinoic acid signaling directs emergence of the extraocular muscle functional unit. PLoS Biol 2020; 18:e3000902. [PMID: 33201874 PMCID: PMC7707851 DOI: 10.1371/journal.pbio.3000902] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 12/01/2020] [Accepted: 10/01/2020] [Indexed: 12/20/2022] Open
Abstract
Coordinated development of muscles, tendons, and their attachment sites ensures emergence of functional musculoskeletal units that are adapted to diverse anatomical demands among different species. How these different tissues are patterned and functionally assembled during embryogenesis is poorly understood. Here, we investigated the morphogenesis of extraocular muscles (EOMs), an evolutionary conserved cranial muscle group that is crucial for the coordinated movement of the eyeballs and for visual acuity. By means of lineage analysis, we redefined the cellular origins of periocular connective tissues interacting with the EOMs, which do not arise exclusively from neural crest mesenchyme as previously thought. Using 3D imaging approaches, we established an integrative blueprint for the EOM functional unit. By doing so, we identified a developmental time window in which individual EOMs emerge from a unique muscle anlage and establish insertions in the sclera, which sets these muscles apart from classical muscle-to-bone type of insertions. Further, we demonstrate that the eyeballs are a source of diffusible all-trans retinoic acid (ATRA) that allow their targeting by the EOMs in a temporal and dose-dependent manner. Using genetically modified mice and inhibitor treatments, we find that endogenous local variations in the concentration of retinoids contribute to the establishment of tendon condensations and attachment sites that precede the initiation of muscle patterning. Collectively, our results highlight how global and site-specific programs are deployed for the assembly of muscle functional units with precise definition of muscle shapes and topographical wiring of their tendon attachments.
Collapse
Affiliation(s)
- Glenda Evangelina Comai
- Stem Cells & Development Unit, Institut Pasteur, Paris, France
- CNRS UMR 3738, Institut Pasteur, Paris, France
- * E-mail: (GEC); (ST)
| | - Markéta Tesařová
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Valérie Dupé
- Université de Rennes, CNRS, IGDR, Rennes, France
| | - Muriel Rhinn
- IGBMC-Institut de Génétique et de Biologie Moleculaire et Cellulaire, Illkirch, France
| | | | - Fabio da Silva
- Université Côte d'Azur, INSERM, CNRS, iBV, Nice, France
- Division of Molecular Embryology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Betty Feret
- IGBMC-Institut de Génétique et de Biologie Moleculaire et Cellulaire, Illkirch, France
| | | | - Pascal Dollé
- IGBMC-Institut de Génétique et de Biologie Moleculaire et Cellulaire, Illkirch, France
| | - Leif Carlsson
- Umeå Center for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Brian Pryce
- Research Division, Shriners Hospital for Children, Portland, United States of America
| | - François Spitz
- Genomics of Animal Development Unit, Institut Pasteur, Paris, France
| | - Sigmar Stricker
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Tomáš Zikmund
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Jozef Kaiser
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | | | | | - Norbert B. Ghyselinck
- IGBMC-Institut de Génétique et de Biologie Moleculaire et Cellulaire, Illkirch, France
| | - Ronen Schweitzer
- Research Division, Shriners Hospital for Children, Portland, United States of America
| | - Shahragim Tajbakhsh
- Stem Cells & Development Unit, Institut Pasteur, Paris, France
- CNRS UMR 3738, Institut Pasteur, Paris, France
- * E-mail: (GEC); (ST)
| |
Collapse
|
16
|
Bogenschutz EL, Sefton EM, Kardon G. Cell culture system to assay candidate genes and molecular pathways implicated in congenital diaphragmatic hernias. Dev Biol 2020; 467:30-38. [PMID: 32827499 DOI: 10.1016/j.ydbio.2020.07.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 10/23/2022]
Abstract
The mammalian muscularized diaphragm is essential for respiration and defects in the developing diaphragm cause a common and frequently lethal birth defect, congenital diaphragmatic hernia (CDH). Human genetic studies have implicated more than 150 genes and multiple molecular pathways in CDH, but few of these have been validated because of the expense and time to generate mouse mutants. The pleuroperitoneal folds (PPFs) are transient embryonic structures in diaphragm development and defects in PPFs lead to CDH. We have developed a system to culture PPF fibroblasts from E12.5 mouse embryos and show that these fibroblasts, in contrast to the commonly used NIH 3T3 fibroblasts, maintain expression of key genes in normal diaphragm development. Using pharmacological and genetic manipulations that result in CDH in vivo, we also demonstrate that differences in proliferation provide a rapid means of distinguishing healthy and impaired PPF fibroblasts. Thus, the PPF fibroblast cell culture system is an efficient tool for assaying the functional significance of CDH candidate genes and molecular pathways and will be an important resource for elucidating the complex etiology of CDH.
Collapse
Affiliation(s)
- Eric L Bogenschutz
- Department of Human Genetics, University of Utah, Salt Lake City, UT, 84112, United States
| | - Elizabeth M Sefton
- Department of Human Genetics, University of Utah, Salt Lake City, UT, 84112, United States
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT, 84112, United States.
| |
Collapse
|
17
|
Hu J, Xia M, Wang Y, Tian F, Sun B, Yang M, Yang W, Ding X, Xu H, Li W. Paternal exposure to di-n-butyl-phthalate induced developmental toxicity in zebrafish (Danio rerio). Birth Defects Res 2020; 113:14-21. [PMID: 33009721 DOI: 10.1002/bdr2.1812] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 08/21/2020] [Accepted: 09/17/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Dibutyl phthalate (DBP) is an environmental endocrine disruptor detected in water, soil, and other environmental media frequently. Growing concerns regarding DBP exposure focus on toxicity to male reproduction. Reports about the developmental toxicity of paternal DBP exposure are rare. In this study, we investigated the developmental toxicity of paternal exposure to DBP on offspring in zebrafish. METHODS Adult male zebrafish with normal reproductive function were exposed to 0.2, 0.6, 1.8 mg/L of DBP or acetone solvent control for 30 days, and then mated with females. Thirty embryos per group were randomly selected to be observed, and malformations were recorded and photographed. The mating and observations were repeated three times, for a total of 90 embryos per group. RESULTS The results showed that the percentage of malformations, such as edema and a bent trunk, was increased in the 0.6 and 1.8 mg/L DBP exposure groups, the heart rate and spontaneous contraction decreased in the 0.6 and 1.8 mg/L DBP exposure groups and migration of primordial germ cells was disrupted in some F1 embryos in all DBP exposure group after paternal exposure. The axial skeleton was affected in some F1 adults in the 1.8 mg/L DBP exposure group. CONCLUSIONS Our findings demonstrate the developmental toxicity of paternal DBP exposure in zebrafish.
Collapse
Affiliation(s)
- Jingying Hu
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, P.R. China
| | - Minjie Xia
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, P.R. China
| | - Yuzhu Wang
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, P.R. China
| | - Fang Tian
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, P.R. China
| | - Bing Sun
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, P.R. China
| | - Mingjun Yang
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, P.R. China
| | - Wei Yang
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, P.R. China
| | - Xuncheng Ding
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, P.R. China
| | - Huihui Xu
- Division of Health Risk Factors Surveillance and Control, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, P.R. China
| | - Weihua Li
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, P.R. China
| |
Collapse
|
18
|
How Dietary Deficiency Studies Have Illuminated the Many Roles of Vitamin A During Development and Postnatal Life. Subcell Biochem 2020; 95:1-26. [PMID: 32297294 DOI: 10.1007/978-3-030-42282-0_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Vitamin A deficiency studies have been carried out since the early 1900s. Initially, these studies led to the identification of fat soluble A as a unique and essential component of the diet of rodents, birds, and humans. Continuing work established that vitamin A deficiency produces biochemical and physiological dysfunction in almost every vertebrate organ system from conception to death. This chapter begins with a review of representative historical and current studies that used the nutritional vitamin A deficiency research model to gain an understanding of the many roles vitamin A plays in prenatal and postnatal development and well-being. This is followed by a discussion of recent studies that show specific effects of vitamin A deficiency on prenatal development and postnatal maintenance of the olfactory epithelium, brain, and heart. Vitamin A deficiency studies have helped define the necessity of vitamin A for the health of all vertebrates, including farm animals, but the breadth of deficient states and their individual effects on health have not been fully determined. Future work is needed to develop tools to assess the complete vitamin A status of an organism and to define the levels of vitamin A that optimally support molecular and systems level processes during all ages and stages of life.
Collapse
|
19
|
Fainsod A, Bendelac-Kapon L, Shabtai Y. Fetal Alcohol Spectrum Disorder: Embryogenesis Under Reduced Retinoic Acid Signaling Conditions. Subcell Biochem 2020; 95:197-225. [PMID: 32297301 DOI: 10.1007/978-3-030-42282-0_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Fetal Alcohol Spectrum Disorder (FASD) is a complex set of developmental malformations, neurobehavioral anomalies and mental disabilities induced by exposing human embryos to alcohol during fetal development. Several experimental models and a series of developmental and biochemical approaches have established a strong link between FASD and reduced retinoic acid (RA) signaling. RA signaling is involved in the regulation of numerous developmental decisions from patterning of the anterior-posterior axis, starting at gastrulation, to the differentiation of specific cell types within developing organs, to adult tissue homeostasis. Being such an important regulatory signal during embryonic development, mutations or environmental perturbations that affect the level, timing or location of the RA signal can induce multiple and severe developmental malformations. The evidence connecting human syndromes to reduced RA signaling is presented here and the resulting phenotypes are compared to FASD. Available data suggest that competition between ethanol clearance and RA biosynthesis is a major etiological component in FASD.
Collapse
Affiliation(s)
- Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Kerem, POB 12271, 9112102, Jerusalem, Israel.
| | - Liat Bendelac-Kapon
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Kerem, POB 12271, 9112102, Jerusalem, Israel
| | - Yehuda Shabtai
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Kerem, POB 12271, 9112102, Jerusalem, Israel
| |
Collapse
|
20
|
Maternal serum retinol, 25(OH)D and 1,25(OH)2D concentrations during pregnancy and peak bone mass and trabecular bone score in adult offspring at 26-year follow-up. PLoS One 2019; 14:e0222712. [PMID: 31557195 PMCID: PMC6762137 DOI: 10.1371/journal.pone.0222712] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 09/05/2019] [Indexed: 12/19/2022] Open
Abstract
Background Vitamin A and D deficiency is prevalent in pregnant women worldwide. Both vitamins are involved in fetal skeletal development. A positive association between maternal vitamin D levels and offspring bone mineral density (BMD) at adulthood has been observed. The impact of maternal vitamin A status in pregnancy on offspring peak bone mass remains unclear. Method and findings Forty-one mother-child pairs were recruited from a population-based prospective cohort study in Trondheim, Norway, where pregnant women were followed from gestational week 17. Their term-born infants were followed from birth (1986–88). Regression analyses were performed for vitamin A (retinol), 25-hydroxyvitamin D [25(OH)D] and 1,25-dihydroxyvitamin D [1,25(OH)2D] in maternal serum (gestational weeks 17, 33, 37) and cord blood. Offspring BMD and spine trabecular bone score (TBS), a measure of bone quality, were analyzed by dual x-ray absorptiometry at 26 years. Average levels during pregnancy of retinol, 25(OH)D and 1,25(OH)2D were 1.66 (0.32) μmol/L, 59.0 (20.6) nmol/L, and 251.3 (62.4) pmol/L, respectively. 1,25(OH)2D levels were similar in those with 25(OH)D levels <30 and >75 nmol/L. After adjustment for maternal age, BMI, smoking, and education, and offspring birth weight, maternal serum retinol was positively associated with offspring spine BMD [mean change 30.8 (CI 7.6, 54.0) mg/cm2 per 0.2 μmol/L retinol], and with offspring TBS, although non-significant (p = 0.08). No associations were found between maternal 25(OH)D and 1,25(OH)2D levels and offspring bone parameters. Vitamin levels in cord blood were not associated with offspring BMD or TBS. Conclusions This is the first study to show an association between maternal vitamin A status and offspring peak bone mass. Our findings may imply increase future risk for osteoporotic fracture in offspring of mothers with suboptimal vitamin A level. No associations were observed between 25(OH)D and 1,25(OH)2D and offspring BMD.
Collapse
|
21
|
Friedl RM, Raja S, Metzler MA, Patel ND, Brittian KR, Jones SP, Sandell LL. RDH10 function is necessary for spontaneous fetal mouth movement that facilitates palate shelf elevation. Dis Model Mech 2019; 12:12/7/dmm039073. [PMID: 31300413 PMCID: PMC6679383 DOI: 10.1242/dmm.039073] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 06/06/2019] [Indexed: 12/15/2022] Open
Abstract
Cleft palate is a common birth defect, occurring in approximately 1 in 1000 live births worldwide. Known etiological mechanisms of cleft palate include defects within developing palate shelf tissues, defects in mandibular growth and defects in spontaneous fetal mouth movement. Until now, experimental studies directly documenting fetal mouth immobility as an underlying cause of cleft palate have been limited to models lacking neurotransmission. This study extends the range of anomalies directly demonstrated to have fetal mouth movement defects correlated with cleft palate. Here, we show that mouse embryos deficient in retinoic acid (RA) have mispatterned pharyngeal nerves and skeletal elements that block spontaneous fetal mouth movement in utero. Using X-ray microtomography, in utero ultrasound video, ex vivo culture and tissue staining, we demonstrate that proper retinoid signaling and pharyngeal patterning are crucial for the fetal mouth movement needed for palate formation. Embryos with deficient retinoid signaling were generated by stage-specific inactivation of retinol dehydrogenase 10 (Rdh10), a gene crucial for the production of RA during embryogenesis. The finding that cleft palate in retinoid deficiency results from a lack of fetal mouth movement might help elucidate cleft palate etiology and improve early diagnosis in human disorders involving defects of pharyngeal development. Summary: Fetal mouth immobility and defects in pharyngeal patterning underlie cleft palate in retinoid-deficient Rdh10 mutant mouse embryos.
Collapse
Affiliation(s)
- Regina M Friedl
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY 40202, USA
| | - Swetha Raja
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY 40202, USA
| | - Melissa A Metzler
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY 40202, USA
| | - Niti D Patel
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY 40202, USA
| | - Kenneth R Brittian
- Department of Medicine, Diabetes and Obesity Center, University of Louisville, Louisville, KY 40202, USA
| | - Steven P Jones
- Department of Medicine, Diabetes and Obesity Center, University of Louisville, Louisville, KY 40202, USA
| | - Lisa L Sandell
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY 40202, USA
| |
Collapse
|
22
|
Sonoda H, Iizuka H, Ishiwata S, Tsunoda D, Abe M, Takagishi K, Chikuda H, Koibuchi N, Shimokawa N. The retinol‐retinoic acid metabolic pathway is impaired in the lumbar spine of a rat model of congenital kyphoscoliosis. J Cell Biochem 2019; 120:15007-15017. [DOI: 10.1002/jcb.28762] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 12/16/2018] [Accepted: 01/10/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Hiroyuki Sonoda
- Department of Orthopedic Surgery Gunma University Graduate School of Medicine Maebashi Gunma Japan
- Department of Integrative Physiology Gunma University Graduate School of Medicine Maebashi Gunma Japan
| | - Haku Iizuka
- Department of Orthopedic Surgery Gunma University Graduate School of Medicine Maebashi Gunma Japan
| | - Sho Ishiwata
- Department of Orthopedic Surgery Gunma University Graduate School of Medicine Maebashi Gunma Japan
| | - Daisuke Tsunoda
- Department of Orthopedic Surgery Gunma University Graduate School of Medicine Maebashi Gunma Japan
| | - Masako Abe
- Department of Nutrition Takasaki University of Health and Welfare Takasaki Gunma Japan
| | - Kenji Takagishi
- Department of Orthopedic Surgery Gunma University Graduate School of Medicine Maebashi Gunma Japan
| | - Hirotaka Chikuda
- Department of Orthopedic Surgery Gunma University Graduate School of Medicine Maebashi Gunma Japan
| | - Noriyuki Koibuchi
- Department of Integrative Physiology Gunma University Graduate School of Medicine Maebashi Gunma Japan
| | - Noriaki Shimokawa
- Department of Integrative Physiology Gunma University Graduate School of Medicine Maebashi Gunma Japan
- Department of Nutrition Takasaki University of Health and Welfare Takasaki Gunma Japan
| |
Collapse
|
23
|
Hsu JSJ, So M, Tang CSM, Karim A, Porsch RM, Wong C, Yu M, Yeung F, Xia H, Zhang R, Cherny SS, Chung PHY, Wong KKY, Sham PC, Ngo ND, Li M, Tam PKH, Lui VCH, Garcia-Barcelo MM. De novo mutations in Caudal Type Homeo Box transcription Factor 2 (CDX2) in patients with persistent cloaca. Hum Mol Genet 2019; 27:351-358. [PMID: 29177441 DOI: 10.1093/hmg/ddx406] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 10/27/2017] [Indexed: 12/24/2022] Open
Abstract
The cloaca is an embryonic cavity that is divided into the urogenital sinus and rectum upon differentiation of the cloacal epithelium triggered by tissue-specific transcription factors including CDX2. Defective differentiation leads to persistent cloaca in humans (PC), a phenotype recapitulated in Cdx2 mutant mice. PC is linked to hypo/hyper-vitaminosis A. Although no gene has ever been identified, there is a strong evidence for a genetic contribution to PC. We applied whole-exome sequencing and copy-number-variants analyses to 21 PC patients and their unaffected parents. The damaging p.Cys132* and p.Arg237His de novo CDX2 variants were identified in two patients. These variants altered the expression of CYP26A1, a direct CDX2 target encoding the major retinoic acid (RA)-degrading enzyme. Other RA genes, including the RA-receptor alpha, were also mutated. Genes governing the development of cloaca-derived structures were recurrently mutated and over-represented in the basement-membrane components set (q-value < 1.65 × 10-6). Joint analysis of the patients' profile highlighted the extracellular matrix-receptor interaction pathway (MsigDBID: M7098, FDR: q-value < 7.16 × 10-9). This is the first evidence that PC is genetic, with genes involved in the RA metabolism at the lead. Given the CDX2 de novo variants and the role of RA, our observations could potentiate preventive measures. For the first time, a gene recapitulating PC in mouse models is found mutated in humans.
Collapse
Affiliation(s)
- Jacob S J Hsu
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Manting So
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Clara S M Tang
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Anwarul Karim
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Robert M Porsch
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Carol Wong
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Michelle Yu
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Fanny Yeung
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, Guandong, China
| | - Ruizhong Zhang
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, Guandong, China
| | - Stacey S Cherny
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Patrick H Y Chung
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kenneth K Y Wong
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Pak C Sham
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ngoc Diem Ngo
- Department of Human Genetics, National Hospital of Pediatrics, Hà N?i, Vietnam
| | - Miaoxin Li
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Paul K H Tam
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Vincent C H Lui
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
24
|
Berenguer M, Darnaudery M, Claverol S, Bonneu M, Lacombe D, Rooryck C. Prenatal retinoic acid exposure reveals candidate genes for craniofacial disorders. Sci Rep 2018; 8:17492. [PMID: 30504818 PMCID: PMC6269437 DOI: 10.1038/s41598-018-35681-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/09/2018] [Indexed: 12/31/2022] Open
Abstract
Syndromes that display craniofacial anomalies comprise a major class of birth defects. Both genetic and environmental factors, including prenatal retinoic acid (RA) exposure, have been associated with these syndromes. While next generation sequencing has allowed the discovery of new genes implicated in these syndromes, some are still poorly characterized such as Oculo-Auriculo-Vertebral Spectrum (OAVS). Due to the lack of clear diagnosis for patients, developing new strategies to identify novel genes involved in these syndromes is warranted. Thus, our study aimed to explore the link between genetic and environmental factors. Owing to a similar phenotype of OAVS reported after gestational RA exposures in humans and animals, we explored RA targets in a craniofacial developmental context to reveal new candidate genes for these related disorders. Using a proteomics approach, we detected 553 dysregulated proteins in the head region of mouse embryos following their exposure to prenatal RA treatment. This novel proteomic approach implicates changes in proteins that are critical for cell survival/apoptosis and cellular metabolism which could ultimately lead to the observed phenotype. We also identified potential molecular links between three major environmental factors known to contribute to craniofacial defects including maternal diabetes, prenatal hypoxia and RA exposure. Understanding these links could help reveal common key pathogenic mechanisms leading to craniofacial disorders. Using both in vitro and in vivo approaches, this work identified two new RA targets, Gnai3 and Eftud2, proteins known to be involved in craniofacial disorders, highlighting the power of this proteomic approach to uncover new genes whose dysregulation leads to craniofacial defects.
Collapse
Affiliation(s)
- Marie Berenguer
- University Bordeaux, Maladies Rares: Génétique et Métabolisme (MRGM), U 1211 INSERM, F-33000, Bordeaux, France
| | - Muriel Darnaudery
- Université de Bordeaux, Nutrition et neurobiologie intégrée (NUTRINEURO), UMR 1286, 146, rue Léo Saignat, 33076 Bordeaux Cedex, France - Inra, Nutrition et neurobiologie intégrée (NUTRINEURO), UMR 1286, F-33076, Bordeaux, France
| | - Stéphane Claverol
- Center of Functional Genomics, Bordeaux University, Bordeaux, France
| | - Marc Bonneu
- Center of Functional Genomics, Bordeaux University, Bordeaux, France
| | - Didier Lacombe
- University Bordeaux, Maladies Rares: Génétique et Métabolisme (MRGM), U 1211 INSERM, F-33000, Bordeaux, France
- CHU de Bordeaux, Service de Génétique Médicale, Centre de Référence Anomalies du Développement et Syndromes Malformatifs, F-33000, Bordeaux, France
| | - Caroline Rooryck
- University Bordeaux, Maladies Rares: Génétique et Métabolisme (MRGM), U 1211 INSERM, F-33000, Bordeaux, France.
- CHU de Bordeaux, Service de Génétique Médicale, Centre de Référence Anomalies du Développement et Syndromes Malformatifs, F-33000, Bordeaux, France.
| |
Collapse
|
25
|
Delabaere A, Blanchon L, Coste K, Clairefond G, Belville C, Blanc P, Marceau G, Sapin V, Gallot D. Retinoic acid and tracheal occlusion for diaphragmatic hernia treatment in rabbit fetuses. Prenat Diagn 2018; 38:482-492. [DOI: 10.1002/pd.5256] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 02/06/2018] [Accepted: 03/14/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Amélie Delabaere
- “Translational approach to epithelial injury and repair” team, Université Clermont Auvergne, CNRS, Inserm, GReD; 63000 Clermont-Ferrand France
- Fetal Maternal Medicine Unit, Obstetrics and Gynecology Department; Clermont-Ferrand University Hospital; 63000 Clermont-Ferrand France
| | - Loïc Blanchon
- “Translational approach to epithelial injury and repair” team, Université Clermont Auvergne, CNRS, Inserm, GReD; 63000 Clermont-Ferrand France
| | - Karen Coste
- “Translational approach to epithelial injury and repair” team, Université Clermont Auvergne, CNRS, Inserm, GReD; 63000 Clermont-Ferrand France
- Department of Pediatrics; Clermont-Ferrand University Hospital; 63000 Clermont-Ferrand France
| | - Gael Clairefond
- “Translational approach to epithelial injury and repair” team, Université Clermont Auvergne, CNRS, Inserm, GReD; 63000 Clermont-Ferrand France
| | - Corinne Belville
- “Translational approach to epithelial injury and repair” team, Université Clermont Auvergne, CNRS, Inserm, GReD; 63000 Clermont-Ferrand France
| | - Pierre Blanc
- “Translational approach to epithelial injury and repair” team, Université Clermont Auvergne, CNRS, Inserm, GReD; 63000 Clermont-Ferrand France
- EA7281-“Translational approach to epithelial injury and repair”; Auvergne University; 63000 Clermont-Ferrand France
| | - Geoffroy Marceau
- “Translational approach to epithelial injury and repair” team, Université Clermont Auvergne, CNRS, Inserm, GReD; 63000 Clermont-Ferrand France
- Department of Biochemistry and Molecular Biology; Clermont-Ferrand University Hospital; 63000 Clermont-Ferrand France
| | - Vincent Sapin
- “Translational approach to epithelial injury and repair” team, Université Clermont Auvergne, CNRS, Inserm, GReD; 63000 Clermont-Ferrand France
- Department of Biochemistry and Molecular Biology; Clermont-Ferrand University Hospital; 63000 Clermont-Ferrand France
| | - Denis Gallot
- “Translational approach to epithelial injury and repair” team, Université Clermont Auvergne, CNRS, Inserm, GReD; 63000 Clermont-Ferrand France
- Fetal Maternal Medicine Unit, Obstetrics and Gynecology Department; Clermont-Ferrand University Hospital; 63000 Clermont-Ferrand France
| |
Collapse
|
26
|
Acetaldehyde inhibits retinoic acid biosynthesis to mediate alcohol teratogenicity. Sci Rep 2018; 8:347. [PMID: 29321611 PMCID: PMC5762763 DOI: 10.1038/s41598-017-18719-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 12/15/2017] [Indexed: 12/20/2022] Open
Abstract
Alcohol consumption during pregnancy induces Fetal Alcohol Spectrum Disorder (FASD), which has been proposed to arise from competitive inhibition of retinoic acid (RA) biosynthesis. We provide biochemical and developmental evidence identifying acetaldehyde as responsible for this inhibition. In the embryo, RA production by RALDH2 (ALDH1A2), the main retinaldehyde dehydrogenase expressed at that stage, is inhibited by ethanol exposure. Pharmacological inhibition of the embryonic alcohol dehydrogenase activity, prevents the oxidation of ethanol to acetaldehyde that in turn functions as a RALDH2 inhibitor. Acetaldehyde-mediated reduction of RA can be rescued by RALDH2 or retinaldehyde supplementation. Enzymatic kinetic analysis of human RALDH2 shows a preference for acetaldehyde as a substrate over retinaldehyde. RA production by hRALDH2 is efficiently inhibited by acetaldehyde but not by ethanol itself. We conclude that acetaldehyde is the teratogenic derivative of ethanol responsible for the reduction in RA signaling and induction of the developmental malformations characteristic of FASD. This competitive mechanism will affect tissues requiring RA signaling when exposed to ethanol throughout life.
Collapse
|
27
|
Maguire M, Larsen MC, Foong YH, Tanumihardjo S, Jefcoate CR. Cyp1b1 deletion and retinol deficiency coordinately suppress mouse liver lipogenic genes and hepcidin expression during post-natal development. Mol Cell Endocrinol 2017; 454:50-68. [PMID: 28583802 PMCID: PMC5985816 DOI: 10.1016/j.mce.2017.05.037] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 05/30/2017] [Accepted: 05/30/2017] [Indexed: 02/06/2023]
Abstract
UNLABELLED Cyp1b1 deletion and gestational vitamin A deficiency (GVAD) redirect adult liver gene expression. A matched sufficient pre- and post-natal diet, which has high carbohydrate and normal iron content (LF12), increased inflammatory gene expression markers in adult livers that were suppressed by GVAD and Cyp1b1 deletion. At birth on the LF12 diet, Cyp1b1 deletion and GVAD each suppress liver expression of the iron suppressor, hepcidin (Hepc), while increasing stellate cell activation markers and suppressing post-natal increases in lipogenesis. Hepc was less suppressed in Cyp1b1-/- pups with a standard breeder diet, but was restored by iron supplementation of the LF12 diet. CONCLUSIONS The LF12 diet delivered low post-natal iron and attenuated Hepc. Hepc decreases in Cyp1b1-/- and GVAD mice resulted in stellate activation and lipogenesis suppression. Endothelial BMP6, a Hepc stimulant, is a potential coordinator and Cyp1b1 target. These neonatal changes in Cyp1b1-/- mice link to diminished adult obesity and liver inflammation.
Collapse
Affiliation(s)
- Meghan Maguire
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI 53705, United States; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Michele Campaigne Larsen
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Yee Hoon Foong
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Sherry Tanumihardjo
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Colin R Jefcoate
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI 53705, United States; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, United States.
| |
Collapse
|
28
|
DeSantis KA, Stabell AR, Spitzer DC, O'Keefe KJ, Nelson DA, Larsen M. RARα and RARγ reciprocally control K5 + progenitor cell expansion in developing salivary glands. Organogenesis 2017; 13:125-140. [PMID: 28933645 PMCID: PMC5669212 DOI: 10.1080/15476278.2017.1358336] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/25/2017] [Accepted: 07/15/2017] [Indexed: 12/15/2022] Open
Abstract
Understanding the mechanisms of controlled expansion and differentiation of basal progenitor cell populations during organogenesis is essential for developing targeted regenerative therapies. Since the cytokeratin 5-positive (K5+) basal epithelial cell population in the salivary gland is regulated by retinoic acid signaling, we interrogated how isoform-specific retinoic acid receptor (RAR) signaling impacts the K5+ cell population during salivary gland organogenesis to identify RAR isoform-specific mechanisms that could be exploited in future regenerative therapies. In this study, we utilized RAR isoform-specific inhibitors and agonists with murine submandibular salivary gland organ explants. We determined that RARα and RARγ have opposing effects on K5+ cell cycle progression and cell distribution. RARα negatively regulates K5+ cells in both whole organ explants and in isolated epithelial rudiments. In contrast, RARγ is necessary but not sufficient to positively maintain K5+ cells, as agonism of RARγ alone failed to significantly expand the population. Although retinoids are known to stimulate differentiation, K5 levels were not inversely correlated with differentiated ductal cytokeratins. Instead, RARα agonism and RARγ inhibition, corresponding with reduced K5, resulted in premature lumenization, as marked by prominin-1. With lineage tracing, we demonstrated that K5+ cells have the capacity to become prominin-1+ cells. We conclude that RARα and RARγ reciprocally control K5+ progenitor cells endogenously in the developing submandibular salivary epithelium, in a cell cycle-dependent manner, controlling lumenization independently of keratinizing differentiation. Based on these data, isoform-specific targeting RARα may be more effective than pan-RAR inhibitors for regenerative therapies that seek to expand the K5+ progenitor cell pool. SUMMARY STATEMENT RARα and RARγ reciprocally control K5+ progenitor cell proliferation and distribution in the developing submandibular salivary epithelium in a cell cycle-dependent manner while regulating lumenization independently of keratinizing differentiation.
Collapse
Affiliation(s)
- Kara A. DeSantis
- Graduate program in Molecular, Cellular, Developmental, and Neural Biology, University at Albany, SUNY, Albany, NY, USA
- Department of Biological Science, University at Albany, SUNY, Albany, NY, USA
| | - Adam R. Stabell
- Department of Biological Science, University at Albany, SUNY, Albany, NY, USA
| | - Danielle C. Spitzer
- Graduate program in Molecular, Cellular, Developmental, and Neural Biology, University at Albany, SUNY, Albany, NY, USA
- Department of Pathology & Laboratory Medicine and Department of Biology, Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kevin J. O'Keefe
- Graduate program in Molecular, Cellular, Developmental, and Neural Biology, University at Albany, SUNY, Albany, NY, USA
- Department of Biological Science, University at Albany, SUNY, Albany, NY, USA
| | - Deirdre A. Nelson
- Department of Biological Science, University at Albany, SUNY, Albany, NY, USA
| | - Melinda Larsen
- Graduate program in Molecular, Cellular, Developmental, and Neural Biology, University at Albany, SUNY, Albany, NY, USA
- The RNA Institute, University at Albany, SUNY, Albany, NY, USA
| |
Collapse
|
29
|
Wiseman EM, Bar-El Dadon S, Reifen R. The vicious cycle of vitamin a deficiency: A review. Crit Rev Food Sci Nutr 2017; 57:3703-3714. [DOI: 10.1080/10408398.2016.1160362] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Elina Manusevich Wiseman
- The Center of Nutrigenomics, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Shimrit Bar-El Dadon
- The Center of Nutrigenomics, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Ram Reifen
- The Center of Nutrigenomics, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
30
|
Delabaere A, Marceau G, Coste K, Blanchon L, Déchelotte PJ, Blanc P, Sapin V, Gallot D. Effects of tracheal occlusion with retinoic acid administration on normal lung development. Prenat Diagn 2017; 37:427-434. [DOI: 10.1002/pd.5012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 01/09/2017] [Accepted: 01/29/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Amélie Delabaere
- EA7281 - Retinoids, Reproduction Developmental Diseases; Auvergne University; Clermont-Ferrand France
- Fetal Maternal Medicine Unit, Obstetrics and Gynecology Department; Clermont-Ferrand University Hospital; Clermont-Ferrand France
| | - Geoffroy Marceau
- EA7281 - Retinoids, Reproduction Developmental Diseases; Auvergne University; Clermont-Ferrand France
- Biochemistry and Molecular Biology Department; Clermont-Ferrand University Hospital; Clermont-Ferrand France
| | - Karen Coste
- EA7281 - Retinoids, Reproduction Developmental Diseases; Auvergne University; Clermont-Ferrand France
- Department of Paediatrics; Clermont-Ferrand University Hospital; Clermont-Ferrand France
| | - Loïc Blanchon
- EA7281 - Retinoids, Reproduction Developmental Diseases; Auvergne University; Clermont-Ferrand France
| | | | - Pierre Blanc
- EA7281 - Retinoids, Reproduction Developmental Diseases; Auvergne University; Clermont-Ferrand France
| | - Vincent Sapin
- EA7281 - Retinoids, Reproduction Developmental Diseases; Auvergne University; Clermont-Ferrand France
- Biochemistry and Molecular Biology Department; Clermont-Ferrand University Hospital; Clermont-Ferrand France
| | - Denis Gallot
- EA7281 - Retinoids, Reproduction Developmental Diseases; Auvergne University; Clermont-Ferrand France
- Fetal Maternal Medicine Unit, Obstetrics and Gynecology Department; Clermont-Ferrand University Hospital; Clermont-Ferrand France
| |
Collapse
|
31
|
Metzler MA, Sandell LL. Enzymatic Metabolism of Vitamin A in Developing Vertebrate Embryos. Nutrients 2016; 8:E812. [PMID: 27983671 PMCID: PMC5188467 DOI: 10.3390/nu8120812] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/09/2016] [Accepted: 12/13/2016] [Indexed: 12/16/2022] Open
Abstract
Embryonic development is orchestrated by a small number of signaling pathways, one of which is the retinoic acid (RA) signaling pathway. Vitamin A is essential for vertebrate embryonic development because it is the molecular precursor of the essential signaling molecule RA. The level and distribution of RA signaling within a developing embryo must be tightly regulated; too much, or too little, or abnormal distribution, all disrupt embryonic development. Precise regulation of RA signaling during embryogenesis is achieved by proteins involved in vitamin A metabolism, retinoid transport, nuclear signaling, and RA catabolism. The reversible first step in conversion of the precursor vitamin A to the active retinoid RA is mediated by retinol dehydrogenase 10 (RDH10) and dehydrogenase/reductase (SDR family) member 3 (DHRS3), two related membrane-bound proteins that functionally activate each other to mediate the interconversion of retinol and retinal. Alcohol dehydrogenase (ADH) enzymes do not contribute to RA production under normal conditions during embryogenesis. Genes involved in vitamin A metabolism and RA catabolism are expressed in tissue-specific patterns and are subject to feedback regulation. Mutations in genes encoding these proteins disrupt morphogenesis of many systems in a developing embryo. Together these observations demonstrate the importance of vitamin A metabolism in regulating RA signaling during embryonic development in vertebrates.
Collapse
Affiliation(s)
- Melissa A Metzler
- Department of Molecular, Cellular and Craniofacial Biology, University of Louisville, Louisville, KY 40201, USA.
| | - Lisa L Sandell
- Department of Molecular, Cellular and Craniofacial Biology, University of Louisville, Louisville, KY 40201, USA.
| |
Collapse
|
32
|
Chen Y, Reese DH. Disruption of Retinol (Vitamin A) Signaling by Phthalate Esters: SAR and Mechanism Studies. PLoS One 2016; 11:e0161167. [PMID: 27532513 PMCID: PMC4988654 DOI: 10.1371/journal.pone.0161167] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 08/01/2016] [Indexed: 01/03/2023] Open
Abstract
A spectrum of reproductive system anomalies (cryptorchidism, hypospadias, dysgenesis of Wolffian duct-derived tissues and prostate, and reduced sperm production) in male rats exposed in utero to phthalate esters (PEs) are thought to be caused by PE inhibition of fetal testosterone production. Recently, dibutyl and dipentyl phthalate (DBuP, DPnP) were shown to disrupt the retinol signaling pathway (RSP) in mouse pluripotent P19 embryonal carcinoma cells in vitro. The RSP regulates the synthesis and cellular levels of retinoic acid (RA), the active metabolite of retinol (vitamin A). In this new study, a total of 26 di- and mono-esters were screened to identify additional phthalate structures that disrupt the RSP and explore their mechanisms of action. The most potent PEs, those causing > 50% inhibition, contained aryl and cycloalkane groups or C4-C6 alkyl ester chains and were the same PEs reported to cause malformations in utero. They shared similar lipid solubility; logP values were between 4 and 6 and, except for PEs with butyl and phenyl groups, were stable for prolonged periods in culture. Mono- and cognate di-esters varied in ability to disrupt the RSP; e.g., DEHP was inactive but its monoester was active while DBuP was active yet its monoester was inactive. DBuP and dibenzyl phthalate both disrupted the synthesis of RA from retinol but not the ability of RA to activate gene transcription. Both PEs also disrupted the RSP in C3H10T1/2 multipotent mesenchymal stem cells. Based on this in vitro study showing that some PEs disrupt retinol signaling and previous in vivo studies that vitamin A/RA deficiency and PEs both cause strikingly similar anomalies in the male rat reproductive system, we propose that PE-mediated inhibition of testosterone and RA synthesis in utero are both causes of malformations in male rat offspring.
Collapse
Affiliation(s)
- Yanling Chen
- Division of Molecular Biology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. FDA, 8301 Muirkirk Rd., Laurel, MD, 20708, United States of America
| | - David H. Reese
- Division of Molecular Biology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. FDA, 8301 Muirkirk Rd., Laurel, MD, 20708, United States of America
| |
Collapse
|
33
|
Kinetic characterization and regulation of the human retinaldehyde dehydrogenase 2 enzyme during production of retinoic acid. Biochem J 2016; 473:1423-31. [PMID: 27001866 DOI: 10.1042/bcj20160101] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 03/21/2016] [Indexed: 11/17/2022]
Abstract
Retinoic acid (RA) is an important regulator of embryogenesis and tissue homoeostasis. Perturbation of RA signalling causes developmental disorders, osteoarthritis, schizophrenia and several types of tumours. RA is produced by oxidation of retinaldehyde from vitamin A. The main enzyme producing RA in the early embryo is retinaldehyde dehydrogenase 2 (RALDH2, ALDH1A2). In the present study we describe in depth the kinetic properties and regulation of the human RALDH2 (hRALDH2) enzyme. We show that this enzyme produces RA using in vivo and in vitro assays. We studied the naturally occurring all-trans-, 9-cis- and 13-cis-retinaldehyde isomers as substrates of hRALDH2. Based on the values measured for the Michaelis-Menten constant Km and the maximal rate Vmax, in vitro hRALDH2 displays the same catalytic efficiency for their oxidation. We characterized two known inhibitors of the vertebrate RALDH2 and determined their kinetic parameters on hRALDH2. In addition, RA was studied as a possible inhibitor of hRALDH2 and a regulator of its activity. We show that hRALDH2 is not inhibited by its oxidation product, all-trans-RA, suggesting the absence of a negative feedback regulatory loop. Expression of the Raldh2 gene is known to be regulated by RA itself, suggesting that the main regulation of the hRALDH2 activity level is transcriptional.
Collapse
|
34
|
Chen F, Jiang Z, Jiang S, Li L, Lin X, Gou Z, Fan Q. Dietary vitamin A supplementation improved reproductive performance by regulating ovarian expression of hormone receptors, caspase-3 and Fas in broiler breeders. Poult Sci 2016; 95:30-40. [DOI: 10.3382/ps/pev305] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2015] [Indexed: 12/20/2022] Open
|
35
|
The Trk family of neurotrophin receptors is downregulated in the lumbar spines of rats with congenital kyphoscoliosis. Mol Cell Biochem 2015; 412:11-8. [DOI: 10.1007/s11010-015-2603-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/29/2015] [Indexed: 02/07/2023]
|
36
|
Wright DM, Buenger DE, Abashev TM, Lindeman RP, Ding J, Sandell LL. Retinoic acid regulates embryonic development of mammalian submandibular salivary glands. Dev Biol 2015; 407:57-67. [PMID: 26278034 DOI: 10.1016/j.ydbio.2015.08.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 08/03/2015] [Accepted: 08/11/2015] [Indexed: 01/09/2023]
Abstract
Organogenesis is orchestrated by cell and tissue interactions mediated by molecular signals. Identification of relevant signals, and the tissues that generate and receive them, are important goals of developmental research. Here, we demonstrate that Retinoic Acid (RA) is a critical signaling molecule important for morphogenesis of mammalian submandibular salivary glands (SMG). By examining late stage RA deficient embryos of Rdh10 mutant mice we show that SMG development requires RA in a dose-dependent manner. Additionally, we find that active RA signaling occurs in SMG tissues, arising earlier than any other known marker of SMG development and persisting throughout gland morphogenesis. At the initial bud stage of development, we find RA production occurs in SMG mesenchyme, while RA signaling occurs in epithelium. We also demonstrate active RA signaling occurs in glands cultured ex vivo, and treatment with an inhibitor of RA signaling blocks growth and branching. Together these data identify RA signaling as a direct regulator of SMG organogenesis.
Collapse
Affiliation(s)
- Diana M Wright
- University of Louisville School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, KY 40201, USA
| | - Deanna E Buenger
- University of Louisville School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, KY 40201, USA
| | - Timur M Abashev
- University of Louisville School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, KY 40201, USA
| | - Robert P Lindeman
- University of Louisville School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, KY 40201, USA
| | - Jixiang Ding
- University of Louisville School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, KY 40201, USA
| | - Lisa L Sandell
- University of Louisville School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, KY 40201, USA.
| |
Collapse
|
37
|
Coste K, Beurskens LWJE, Blanc P, Gallot D, Delabaere A, Blanchon L, Tibboel D, Labbé A, Rottier RJ, Sapin V. Metabolic disturbances of the vitamin A pathway in human diaphragmatic hernia. Am J Physiol Lung Cell Mol Physiol 2015; 308:L147-57. [PMID: 25416379 DOI: 10.1152/ajplung.00108.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Congenital diaphragmatic hernia (CDH) is a common life-threatening congenital anomaly resulting in high rates of perinatal death and neonatal respiratory distress. Some of the nonisolated forms are related to single-gene mutations or genomic rearrangements, but the genetics of the isolated forms (60% of cases) still remains a challenging issue. Retinoid signaling (RA) is critical for both diaphragm and lung development, and it has been hypothesized that subtle disruptions of this pathway could contribute to isolated CDH etiology. Here we used time series of normal and CDH lungs in humans, in nitrofen-exposed rats, and in surgically induced hernia in rabbits to perform a systematic transcriptional analysis of the RA pathway key components. The results point to CRPBP2, CY26B1, and ALDH1A2 as deregulated RA signaling genes in human CDH. Furthermore, the expression profile comparisons suggest that ALDH1A2 overexpression is not a primary event, but rather a consequence of the CDH-induced lung injury. Taken together, these data show that RA signaling disruption is part of CDH pathogenesis, and also that dysregulation of this pathway should be considered organ specifically.
Collapse
|
38
|
Li Z, Yu X, Shen J. Environmental aspects of congenital scoliosis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2015; 22:5751-5755. [PMID: 25628116 DOI: 10.1007/s11356-015-4144-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 01/15/2015] [Indexed: 06/04/2023]
Abstract
Growing evidence has proved that many aspects of our lifestyle and the environment contribute to the development of congenital disease. Congenital spinal deformities are due to anomalous development of the vertebrae including failure of formation and segmentation during embryogenesis. The causes of congenital scoliosis have not been fully identified. A variety of factors are implicated in the development of vertebral abnormalities. Previous studies have demonstrated that both genetics and environmental factors are implicated in the development of vertebral abnormalities. However, no specific cause for congenital scoliosis has been identified. In our review, we focus on the environmental factors for the development of congenital scoliosis. Various maternal exposures during pregnancy including hypoxia, alcohol use, vitamin deficiency, valproic acid, boric acid, and hyperthermia have been observed to be associated with the occurrence of congenital scoliosis. This review describes the major environmental contributors of congenital scoliosis with an emphasis on treatment aspects associated with environmental disposition in congenital scoliosis.
Collapse
Affiliation(s)
- Zheng Li
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Peking Union Medical College, 100730, Beijing, China
| | | | | |
Collapse
|
39
|
Heying EK, Hovel E, Tanumihardjo SA. Healthy birth weight results in higher vitamin A storage in neonate piglets administered high-dose supplements. Exp Biol Med (Maywood) 2015; 240:1378-85. [PMID: 25681469 DOI: 10.1177/1535370215570185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 12/11/2014] [Indexed: 11/15/2022] Open
Abstract
A proposed intervention for newborn infants in countries with suspected vitamin A (VA) deficiency is to administer 50,000 IU retinyl palmitate at birth to reduce mortality risk. However, no studies have investigated birth weight effects. In this study, low birth weight (LBW; <1 kg, n = 18) and healthy birth weight (HBW) piglets (>1.5 kg, n = 18) from VA-depleted sows were dosed with 25,000 or 50,000 IU retinyl palmitate (26.2 or 52.4 µmol retinol equivalents) at birth to compare VA reserves. Blood was collected at varying times (n = 3-5/time/dose), and piglets were killed at 12 or 24 h for blood, liver, kidneys, spleen, lungs, adrenal gland, and intestinal contents. HBW piglets had significantly higher birth, death, and organ weights than LBW (P < 0.0001 for all). HBW and LBW piglets, which received VA, had higher liver and kidney VA concentrations (0.18 ± 0.09, 0.24 ± 0.10 µmol/g liver and 13.4 ± 4.1, 14.2 ± 4.5 nmol/g kidney, respectively) than controls (n = 10) (0.051 ± 0.01 µmol/g liver and 1.01 ± 0.43 nmol/g kidney) (P = 0.0061 and < 0.0001, respectively). Total liver (9.75 ± 5.16 µmol) and kidney retinol (204 ± 79.1 nmol) were higher in HBW than LBW piglets (P < 0.0001). Extrahepatic tissues, except lung, had higher VA concentration than controls (P < 0.0001). Serum retinol and ester concentrations were higher in treated than control piglets (P = 0.0028, P < 0.0001, respectively), and significantly changed during the times sampled (P = 0.022, P = 0.011, respectively). Peak serum retinyl ester concentrations, which occurred at 3 h, were higher in piglets that received 50,000 IU (4.2 ± 4.4 µmol/L) than 25,000 IU (2.7 ± 2.3 µmol/L) (P = 0.031). Regardless of dose amount, HBW piglets stored more supplemental VA than LBW piglets when administered at birth.
Collapse
Affiliation(s)
- Emily K Heying
- Interdepartmental Graduate Program in Nutritional Sciences, Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Elizabeth Hovel
- Interdepartmental Graduate Program in Nutritional Sciences, Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sherry A Tanumihardjo
- Interdepartmental Graduate Program in Nutritional Sciences, Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
40
|
Khillan JS. Vitamin A/retinol and maintenance of pluripotency of stem cells. Nutrients 2014; 6:1209-22. [PMID: 24662164 PMCID: PMC3967188 DOI: 10.3390/nu6031209] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 02/21/2014] [Accepted: 02/24/2014] [Indexed: 12/22/2022] Open
Abstract
Retinol, the alcohol form of vitamin A is a key dietary component that plays a critical role in vertebrate development, cell differentiation, reproduction, vision and immune system. Natural and synthetic analogs of retinol, called retinoids, have generally been associated with the cell differentiation via retinoic acid which is the most potent metabolite of retinol. However, a direct function of retinol has not been fully investigated. New evidence has now emerged that retinol supports the self-renewal of stem cells including embryonic stem cells (ESCs), germ line stem cells (GSCs) and cancer stem cells (CSCs) by activating the endogenous machinery for self-renewal by a retinoic acid independent mechanism. The studies have also revealed that stem cells do not contain enzymes that are responsible for metabolizing retinol into retinoic acid. This new function of retinol may have important implications for stem cell biology which can be exploited for quantitative production of pure population of pluripotent stem cells for regenerative medicine as well as clinical applications for cancer therapeutics.
Collapse
Affiliation(s)
- Jaspal S Khillan
- Department of Immunology, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA 15261, USA.
| |
Collapse
|
41
|
Abstract
The role of vitamin A status as it relates to bone health is historical yet controversial. Population-based studies have linked high dietary intake of preformed vitamin A, which is obtained from animal-source foods, fortified foods, and some supplements, to greater risk of osteoporosis and hip fracture. In contrast, carotenoids, some of which are vitamin A precursors from plants, are associated with improved bone health. Carotenoids may be a biomarker that reflects a generally healthy lifestyle, which includes fruit and vegetable consumption. Current dietary recommendations to increase fruit and vegetable intake in the Dietary Guidelines for Americans will result in greater intakes of provitamin A carotenoids if consumers comply. This could lead to artificially high intakes of vitamin A in dietary analyses. However, multiple factors affect the bioconversion of provitamin A carotenoids to the active form of vitamin A. The human body will strive to maintain vitamin A balance by down-regulating provitamin A carotenoid bioconversion. If high preformed vitamin A intake is associated with poor bone health and provitamin A carotenoids are protective, future studies are needed to clarify the associations between total body stores of vitamin A, dietary intake of the pre- and pro-forms, and bone health throughout the life cycle.
Collapse
Affiliation(s)
- Sherry A Tanumihardjo
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
42
|
Beurskens LWJE, Schrijver LH, Tibboel D, Wildhagen MF, Knapen MFCM, Lindemans J, de Vries J, Steegers-Theunissen RPM. Dietary vitamin A intake below the recommended daily intake during pregnancy and the risk of congenital diaphragmatic hernia in the offspring. ACTA ACUST UNITED AC 2013; 97:60-6. [PMID: 23283831 DOI: 10.1002/bdra.23093] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 09/17/2012] [Accepted: 09/25/2012] [Indexed: 01/26/2023]
Abstract
BACKGROUND Vitamin A has been related to the etiology of congenital diaphragmatic hernia (CDH). We performed a case-control study to investigate whether maternal dietary vitamin A intake is related to CDH in the offspring. METHODS Thirty-one pregnancies diagnosed with CDH and 46 control pregnancies were included during the study. After CDH diagnosis and inclusion of controls by risk set sampling, maternal vitamin A intake was investigated with a food frequency questionnaire. Serum retinol and retinol-binding protein were determined. Univariable and multivariable logistic regression models were used to calculate risk estimates with adjustment for potential confounders. RESULTS We found no significant differences in the overall nutrient and vitamin A intake between case and control mothers. After stratification in body mass index (BMI) categories, case mothers with normal weight showed a lower energy adjusted vitamin A intake (685 vs. 843 μg retinol activity equivalents [RAEs] / day; p = 0.04) and a slightly lower serum retinol (1.58 vs. 1.67 μmol/L; p = 0.08) than control mothers. Vitamin A intake <800 μg retinol activity equivalents (recommended daily intake) in normal weight mothers was associated with a significantly increased CDH risk (odds ratio [OR], 7.2; 95% confidence interval [CI], 1.5-34.4; p = 0.01). Associations were not significantly different in underweight and overweight mothers. CONCLUSIONS In normal-weight mothers, dietary vitamin A intake during pregnancy below the recommended daily intake is significantly associated with an increased risk of a child with CDH. This finding supports the retinoid hypothesis in human CDH, but warrants further investigation in larger study populations. Birth Defects Research (Part A), 2013. © 2013 Wiley Periodicals, Inc.
Collapse
|
43
|
He X, Li Y, Li M, Jia G, Dong H, Zhang Y, He C, Wang C, Deng L, Yang Y. Hypovitaminosis A coupled to secondary bacterial infection in beef cattle. BMC Vet Res 2012; 8:222. [PMID: 23151297 PMCID: PMC3534487 DOI: 10.1186/1746-6148-8-222] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 08/22/2012] [Indexed: 11/26/2022] Open
Abstract
Background Vitamin A is essential for normal growth, development, reproduction, cell proliferation, cell differentiation, immune function and vision. Hypovitaminosis A can lead to a series of pathological damage in animals. This report describes the case of hypovitaminosis A associated with secondary complications in calves. Case presentation From February to March in 2011, 2-and 3-month old beef calves presented with decreased eyesight, apparent blindness and persistent diarrhea occurred in a cattle farm of Hubei province, China. Based on history inspection and clinical observation, we made a tentative diagnosis of hypovitaminosis A. The disease was confirmed as a congenital vitamin A deficiency by determination of the concentrations of vitamin A in serum and feed samples. Furthermore, pathological and microbiological examination showed that the disease was associated with pathogenic Escherichia coli (E. coli) infection and mucosal barriers damage in intestines. The corresponding treatments were taken immediately, and the disease was finally under control for a month. Conclusions To our knowledge, this is the first report of hypovitaminosis A coupled to secondary infection of E. coli in beef cattle, advancing our knowledge of how vitamin A affects infection and immunity in animals. This study could also be contributed to scientific diagnosis and treatments of complex hypovitaminosis A in cattle.
Collapse
Affiliation(s)
- Xiuyuan He
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Wenhua Road 95#, Zhengzhou, Henan, 450002, PR China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Li Z, Shen J, Wu WKK, Wang X, Liang J, Qiu G, Liu J. Vitamin A deficiency induces congenital spinal deformities in rats. PLoS One 2012; 7:e46565. [PMID: 23071590 PMCID: PMC3465343 DOI: 10.1371/journal.pone.0046565] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 08/31/2012] [Indexed: 11/17/2022] Open
Abstract
Most cases of congenital spinal deformities were sporadic and without strong evidence of heritability. The etiology of congenital spinal deformities is still elusive and assumed to be multi-factorial. The current study seeks to elucidate the effect of maternal vitamin A deficiency and the production of congenital spinal deformities in the offsping. Thirty two female rats were randomized into two groups: control group, which was fed a normal diet; vitamin A deficient group, which were given vitamin A-deficient diet from at least 2 weeks before mating till delivery. Three random neonatal rats from each group were killed the next day of parturition. Female rats were fed an AIN-93G diet sufficient in vitamin A to feed the rest of neonates for two weeks until euthanasia. Serum levels of vitamin A were assessed in the adult and filial rats. Anteroposterior (AP) spine radiographs were obtained at week 2 after delivery to evaluate the presence of the skeletal abnormalities especially of spinal deformities. Liver and vertebral body expression of retinaldehyde dehydrogenase (RALDHs) and RARs mRNA was assessed by reverse transcription-real time PCR. VAD neonates displayed many skeletal malformations in the cervical, thoracic, the pelvic and sacral and limbs regions. The incidence of congenital scoliosis was 13.79% (8/58) in the filial rats of vitamin A deficiency group and 0% in the control group. Furthermore, vitamin A deficiency negatively regulate the liver and verterbral body mRNA levels of RALDH1, RALDH2, RALDH3, RAR-α, RAR-β and RAR-γ. Vitamin A deficiency in pregnancy may induce congenital spinal deformities in the postnatal rats. The decreases of RALDHs and RARs mRNA expression induced by vitamin A deprivation suggest that vertebral birth defects may be caused by a defect in RA signaling pathway during somitogenesis.
Collapse
Affiliation(s)
- Zheng Li
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
45
|
Sandell LL, Lynn ML, Inman KE, McDowell W, Trainor PA. RDH10 oxidation of Vitamin A is a critical control step in synthesis of retinoic acid during mouse embryogenesis. PLoS One 2012; 7:e30698. [PMID: 22319578 PMCID: PMC3271098 DOI: 10.1371/journal.pone.0030698] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Accepted: 12/23/2011] [Indexed: 12/19/2022] Open
Abstract
Retinoic Acid (RA) is a small lipophilic signaling molecule essential for embryonic development and adult tissue maintenance. Both an excess of RA and a deficiency of RA can cause pathogenic anomalies, hence it is critical to understand the mechanisms controlling the spatial and temporal distribution of RA. However, our current understanding of these processes remains incomplete. Vitamin A is metabolized to RA via two sequential enzymatic reactions. The first requires retinol dehydrogenase (RDH) activity to oxidize Vitamin A (retinol) to retinal, and the second requires retinaldehyde activity (RALDH) to oxidize retinal into RA. The first reaction has previously been attributed to the alcohol dehydrogenase (ADH) family, whose genes are ubiquitously or redundantly expressed. Consequently, the specificity of RA synthesis was thought to reside exclusively at the level of the second reaction. To better understand the metabolism of Vitamin A into RA during embryogenesis, we generated new mouse models that disrupt this process. Here we describe a new targeted knockout of Rdh10 in which RA synthesis is severely impaired, particularly at critical early embryonic stages. We also introduce a new mutant allele of Aldh1a2. Both mutations produce similar developmental defects resulting in lethality around embryonic day 10.5 (E10.5). The severity of the Rdh10 null phenotype demonstrates that embryonic oxidation of retinol is carried out primarily by RDH10 and that neither ADHs nor other enzymes contribute significantly to this reaction. We also show that reduced RA production results in upregulation of Rdh10. These data demonstrate that RDH10 plays a critical role in mediating the rate limiting RDH step of Vitamin A metabolism and functions as a nodal point in feedback regulation of RA synthesis. Moreover, RDH10-mediated oxidation of retinol plays as important a role in the control and regulation of RA production during embryogenesis as does the subsequent RALDH-mediated reaction.
Collapse
Affiliation(s)
- Lisa L. Sandell
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- Department of Molecular, Cellular and Craniofacial Biology, University of Louisville, Louisville, Kentucky, United States of America
| | - Megan L. Lynn
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Kimberly E. Inman
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - William McDowell
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Paul A. Trainor
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| |
Collapse
|
46
|
Ross AC, Zolfaghari R. Cytochrome P450s in the regulation of cellular retinoic acid metabolism. Annu Rev Nutr 2011; 31:65-87. [PMID: 21529158 DOI: 10.1146/annurev-nutr-072610-145127] [Citation(s) in RCA: 202] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The active metabolite of vitamin A, retinoic acid (RA), is a powerful regulator of gene transcription. RA is also a therapeutic drug. The oxidative metabolism of RA by certain members of the cytochrome P450 (CYP) superfamily helps to maintain tissue RA concentrations within appropriate bounds. The CYP26 family--CYP26A1, CYP26B1, and CYP26C1--is distinguished by being both regulated by and active toward all-trans-RA (at-RA) while being expressed in different tissue-specific patterns. The CYP26A1 gene is regulated by multiple RA response elements. CYP26A1 is essential for embryonic development, whereas CYP26B1 is essential for postnatal survival as well as germ cell development. Enzyme kinetic studies have demonstrated that several CYP proteins are capable of metabolizing at-RA; however, it is likely that CYP26A1 plays a major role in RA clearance. Thus, pharmacological approaches to limiting the activity of CYP26 enzymes may extend the half-life of RA and could be useful clinically in the future.
Collapse
Affiliation(s)
- A Catharine Ross
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| | | |
Collapse
|
47
|
Disease embryo development network reveals the relationship between disease genes and embryo development genes. J Theor Biol 2011; 287:100-8. [PMID: 21824480 PMCID: PMC7094120 DOI: 10.1016/j.jtbi.2011.07.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 06/15/2011] [Accepted: 07/22/2011] [Indexed: 11/20/2022]
Abstract
A basic problem for contemporary biology and medicine is exploring the correlation between human disease and underlying cellular mechanisms. For a long time, several efforts were made to reveal the similarity between embryo development and disease process, but few from the system level. In this article, we used the human protein-protein interactions (PPIs), disease genes with their classifications and embryo development genes and reconstructed a human disease-embryo development network to investigate the relationship between disease genes and embryo development genes. We found that disease genes and embryo development genes are prone to connect with each other. Furthermore, diseases can be categorized into three groups according to the closeness with embryo development in gene overlapping, interacting pattern in PPI network and co-regulated by microRNAs or transcription factors. Embryo development high-related disease genes show their closeness with embryo development at least in three biological levels. But it is not for embryo development medium-related disease genes and embryo development low-related disease genes. We also found that embryo development high-related disease genes are more central than other disease genes in the human PPI network. In addition, the results show that embryo development high-related disease genes tend to be essential genes compared with other diseases' genes. This network-based approach could provide evidence for the intricate correlation between disease process and embryo development, and help to uncover potential mechanisms of human complex diseases.
Collapse
|
48
|
Clagett-Dame M, Knutson D. Vitamin A in reproduction and development. Nutrients 2011; 3:385-428. [PMID: 22254103 PMCID: PMC3257687 DOI: 10.3390/nu3040385] [Citation(s) in RCA: 268] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Revised: 02/28/2011] [Accepted: 03/22/2011] [Indexed: 12/12/2022] Open
Abstract
The requirement for vitamin A in reproduction was first recognized in the early 1900's, and its importance in the eyes of developing embryos was realized shortly after. A greater understanding of the large number of developmental processes that require vitamin A emerged first from nutritional deficiency studies in rat embryos, and later from genetic studies in mice. It is now generally believed that all-trans retinoic acid (RA) is the form of vitamin A that supports both male and female reproduction as well as embryonic development. This conclusion is based on the ability to reverse most reproductive and developmental blocks found in vitamin A deficiency induced either by nutritional or genetic means with RA, and the ability to recapitulate the majority of embryonic defects in retinoic acid receptor compound null mutants. The activity of the catabolic CYP26 enzymes in determining what tissues have access to RA has emerged as a key regulatory mechanism, and helps to explain why exogenous RA can rescue many vitamin A deficiency defects. In severely vitamin A-deficient (VAD) female rats, reproduction fails prior to implantation, whereas in VAD pregnant rats given small amounts of carotene or supported on limiting quantities of RA early in organogenesis, embryos form but show a collection of defects called the vitamin A deficiency syndrome or late vitamin A deficiency. Vitamin A is also essential for the maintenance of the male genital tract and spermatogenesis. Recent studies show that vitamin A participates in a signaling mechanism to initiate meiosis in the female gonad during embryogenesis, and in the male gonad postnatally. Both nutritional and genetic approaches are being used to elucidate the vitamin A-dependent pathways upon which these processes depend.
Collapse
Affiliation(s)
- Margaret Clagett-Dame
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA;
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, 777 Highland Ave., Madison, WI 53705, USA
| | - Danielle Knutson
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA;
| |
Collapse
|
49
|
Pennimpede T, Cameron DA, MacLean GA, Li H, Abu-Abed S, Petkovich M. The role of CYP26 enzymes in defining appropriate retinoic acid exposure during embryogenesis. ACTA ACUST UNITED AC 2011; 88:883-94. [PMID: 20842651 DOI: 10.1002/bdra.20709] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Retinoic acid (RA) is a pleiotropic derivative of vitamin A, or retinol, which is responsible for all of the bioactivity associated with this vitamin. The teratogenic influences of vitamin A deficiency and excess RA in rodents were first observed more than 50 years ago. Efforts over the last 15-20 years have refined these observations by defining the molecular mechanisms that control RA availability and signaling during murine embryonic development. This review will discuss our current understanding of the role of RA in teratogenesis, with specific emphasis on the essential function of the RA catabolic CYP26 enzymes in preventing teratogenic consequences caused by uncontrolled distribution of RA. Particular focus will be paid to the RA-sensitive tissues of the caudal and cranial regions, the limb, and the testis, and how genetic mutation of factors controlling RA distribution have revealed important roles for RA during embryogenesis.
Collapse
Affiliation(s)
- Tracie Pennimpede
- Department of Pathology and Molecular Medicine, Cancer Research Institute, Division of Cancer Biology and Genetics, Queen's University, Kingston, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
50
|
Ghatpande SK, Zhou HR, Cakstina I, Carlson C, Rondini EA, Romeih M, Zile MH. Transforming growth factor beta2 is negatively regulated by endogenous retinoic acid during early heart morphogenesis. Dev Growth Differ 2010; 52:433-55. [PMID: 20507358 DOI: 10.1111/j.1440-169x.2010.01183.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Vitamin A-deficient (VAD) quail embryos lack the vitamin A-active form, retinoic acid (RA) and are characterized by a phenotype that includes a grossly abnormal cardiovascular system that can be rescued by RA. Here we report that the transforming growth factor, TGFbeta2 is involved in RA-regulated cardiovascular development. In VAD embryos TGFbeta2 mRNA and protein expression are greatly elevated. The expression of TGFbeta receptor II is also elevated in VAD embryos but is normalized by treatment with TGFbeta2-specific antisense oligonucleotides (AS). Administration of this AS or an antibody specific for TGFbeta2 to VAD embryos normalizes posterior heart development and vascularization, while the administration of exogenous active TGFbeta2 protein to normal quail embryos mimics the excessive TGFbeta2 status of VAD embryos and induces VAD cardiovascular phenotype. In VAD embryos pSmad2/3 and pErk1 are not activated, while pErk2 and pcRaf are elevated and pSmad1/5/8 is diminished. We conclude that in the early avian embryo TGFbeta2 has a major role in the retinoic acid-regulated posterior heart morphogenesis for which it does not use Smad2/3 pathways, but may use other signaling pathways. Importantly, we conclude that retinoic acid is a critical negative physiological regulator of the magnitude of TGFbeta2 signals during vertebrate heart formation.
Collapse
|