1
|
Wang H, Xu F, Yao C, Dai H, Xu J, Wu B, Tian B, Shi X, Wang C. Engineering bacteria for cancer immunotherapy by inhibiting IDO activity and reprogramming CD8+ T cell response. Proc Natl Acad Sci U S A 2024; 121:e2412070121. [PMID: 39693352 DOI: 10.1073/pnas.2412070121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024] Open
Abstract
Inhibiting indoleamine 2,3 dioxygenase (IDO) for anticancer therapy has garnered significant attention in recent years. However, current IDO inhibitors face significant challenges which limit their clinical application. Here, we genetically engineered a high tryptophan-expressing Clostridium butyricum (L-Trp CB) strain that can colonize tumors strictly following systemic administration. We revealed that butyrate produced by L-Trp CB can inhibit IDO activity, preventing tryptophan catabolism and kynurenine accumulation in tumors. In addition, the large released tryptophan by L-Trp CB can provide discrete signals that support CD8+ T cell activation and energy metabolism within the tumor microenvironment. We observed that L-Trp CB significantly restored the proportion and function of CD8+ T cells, leading to significantly delayed tumor growth in both mouse and rabbit multiple tumor models with limited side effects. We here provide a synthetic biology treatment strategy for enhanced tumor immunotherapy by inhibiting IDO activity and reprogramming CD8+ T cell response in tumors.
Collapse
Affiliation(s)
- Heng Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Fang Xu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Chenlu Yao
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Huaxing Dai
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jialu Xu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Bingbing Wu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| | - Bo Tian
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Xiaolin Shi
- Medical College of Soochow University, Suzhou 215123, China
| | - Chao Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
2
|
Glover HJ, Holliday H, Shparberg RA, Winkler D, Day M, Morris MB. Signalling pathway crosstalk stimulated by L-proline drives mouse embryonic stem cells to primitive-ectoderm-like cells. Development 2023; 150:dev201704. [PMID: 37823343 PMCID: PMC10652046 DOI: 10.1242/dev.201704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 10/02/2023] [Indexed: 10/13/2023]
Abstract
The amino acid L-proline exhibits growth factor-like properties during development - from improving blastocyst development to driving neurogenesis in vitro. Addition of 400 μM L-proline to self-renewal medium drives naïve mouse embryonic stem cells (ESCs) to early primitive ectoderm-like (EPL) cells - a transcriptionally distinct primed or partially primed pluripotent state. EPL cells retain expression of pluripotency genes, upregulate primitive ectoderm markers, undergo a morphological change and have increased cell number. These changes are facilitated by a complex signalling network hinging on the Mapk, Fgfr, Pi3k and mTor pathways. Here, we use a factorial experimental design coupled with statistical modelling to understand which signalling pathways are involved in the transition between ESCs and EPL cells, and how they underpin changes in morphology, cell number, apoptosis, proliferation and gene expression. This approach reveals pathways which work antagonistically or synergistically. Most properties were affected by more than one inhibitor, and each inhibitor blocked specific aspects of the naïve-to-primed transition. These mechanisms underpin progression of stem cells across the in vitro pluripotency continuum and serve as a model for pre-, peri- and post-implantation embryogenesis.
Collapse
Affiliation(s)
- Hannah J. Glover
- School of Medical Sciences, University of Sydney, Sydney 2006, Australia
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Holly Holliday
- School of Medical Sciences, University of Sydney, Sydney 2006, Australia
| | | | - David Winkler
- Department of Biochemistry and Chemistry, Latrobe Institute for Molecular Science, Latrobe University, Bundoora 3083, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
- Advanced Materials and Healthcare Technologies, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Margot Day
- School of Medical Sciences, University of Sydney, Sydney 2006, Australia
| | - Michael B. Morris
- School of Medical Sciences, University of Sydney, Sydney 2006, Australia
| |
Collapse
|
3
|
Calderari S, Archilla C, Jouneau L, Daniel N, Peynot N, Dahirel M, Richard C, Mourier E, Schmaltz-Panneau B, Vitorino Carvalho A, Rousseau-Ralliard D, Lager F, Marchiol C, Renault G, Gatien J, Nadal-Desbarats L, Couturier-Tarrade A, Duranthon V, Chavatte-Palmer P. Alteration of the embryonic microenvironment and sex-specific responses of the preimplantation embryo related to a maternal high-fat diet in the rabbit model. J Dev Orig Health Dis 2023; 14:602-613. [PMID: 37822211 DOI: 10.1017/s2040174423000260] [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: 10/13/2023]
Abstract
The maternal metabolic environment can be detrimental to the health of the offspring. In a previous work, we showed that maternal high-fat (HH) feeding in rabbit induced sex-dependent metabolic adaptation in the fetus and led to metabolic syndrome in adult offspring. As early development representing a critical window of susceptibility, in the present work we aimed to explore the effects of the HH diet on the oocyte, preimplantation embryo and its microenvironment. In oocytes from females on HH diet, transcriptomic analysis revealed a weak modification in the content of transcripts mainly involved in meiosis and translational control. The effect of maternal HH diet on the embryonic microenvironment was investigated by identifying the metabolite composition of uterine and embryonic fluids collected in vivo by biomicroscopy. Metabolomic analysis revealed differences in the HH uterine fluid surrounding the embryo, with increased pyruvate concentration. Within the blastocoelic fluid, metabolomic profiles showed decreased glucose and alanine concentrations. In addition, the blastocyst transcriptome showed under-expression of genes and pathways involved in lipid, glucose and amino acid transport and metabolism, most pronounced in female embryos. This work demonstrates that the maternal HH diet disrupts the in vivo composition of the embryonic microenvironment, where the presence of nutrients is increased. In contrast to this nutrient-rich environment, the embryo presents a decrease in nutrient sensing and metabolism suggesting a potential protective process. In addition, this work identifies a very early sex-specific response to the maternal HH diet, from the blastocyst stage.
Collapse
Affiliation(s)
- Sophie Calderari
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Catherine Archilla
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Luc Jouneau
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Nathalie Daniel
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Nathalie Peynot
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Michele Dahirel
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Christophe Richard
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
- Plateforme MIMA2-CIMA, Jouy en Josas, France
| | - Eve Mourier
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
- Plateforme MIMA2-CIMA, Jouy en Josas, France
| | - Barbara Schmaltz-Panneau
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Anaïs Vitorino Carvalho
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Delphine Rousseau-Ralliard
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Franck Lager
- Université Paris Cité, Institut Cochin, Inserm, CNRS, ParisF-75014, France
| | - Carmen Marchiol
- Université Paris Cité, Institut Cochin, Inserm, CNRS, ParisF-75014, France
| | - Gilles Renault
- Université Paris Cité, Institut Cochin, Inserm, CNRS, ParisF-75014, France
| | - Julie Gatien
- Research and Development Department, Eliance, Nouzilly, France
| | - Lydie Nadal-Desbarats
- UMR 1253, iBrain, University of Tours, Inserm, Tours, France
- PST-ASB, University of Tours, Tours, France
| | - Anne Couturier-Tarrade
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Véronique Duranthon
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| | - Pascale Chavatte-Palmer
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort94700, France
| |
Collapse
|
4
|
Van Winkle LJ. Perspective: Might Maternal Dietary Monosodium Glutamate (MSG) Consumption Impact Pre- and Peri-Implantation Embryos and Their Subsequent Development? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:13611. [PMID: 36294193 PMCID: PMC9602898 DOI: 10.3390/ijerph192013611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 06/16/2023]
Abstract
MSG alters metabolism, especially in the brain, when administered to experimental animals via gavage or similar means. Such administration is, however, not applicable to humans. More recently, though, MSG was shown to have these effects even when added to the food of mammals. Moreover, the levels of MSG in food needed to cause these metabolic changes are the same as those needed for optimum flavor enhancement. Near physiological concentrations of glutamate also cause mammalian blastocysts to develop with fewer cells, especially in their inner cell masses, when these embryos are cultured with this amino acid. We propose that consumption of MSG in food may overwhelm the otherwise well-regulated glutamate signaling needed for optimal development by pre- and peri-implantation mammalian embryos. In addition to immediate changes in cellular proliferation and differentiation as embryos develop, MSG ingestion during early pregnancy might result in undesirable conditions, including metabolic syndrome, in adults. Since these conditions are often the result of epigenetic changes, they could become transgenerational. In light of these possibilities, we suggest several studies to test the merit of our hypothesis.
Collapse
Affiliation(s)
- Lon J. Van Winkle
- Department of Biochemistry, Midwestern University, Downers Grove, IL 60515, USA;
- Department of Medical Humanities, Rocky Vista University, 8401 S. Chambers Road, Parker, CO 80112, USA
| |
Collapse
|
5
|
Suzuki D, Sasaki K, Kumamoto S, Tanaka K, Ogawa H. Dynamic Changes of Gene Expression in Mouse Mural Trophectoderm Regulated by Cdx2 During Implantation. Front Cell Dev Biol 2022; 10:945241. [PMID: 36051443 PMCID: PMC9425295 DOI: 10.3389/fcell.2022.945241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/21/2022] [Indexed: 11/24/2022] Open
Abstract
Implantation of the blastocyst into the uterus is a specific and essential process for mammalian embryonic development. In mice, implantation is initiated from the mural trophectoderm of the blastocyst and the mTE controls implantation progression by acquiring the ability to attach and invade into the endometrium while differentiating into primary trophoblast giant cells. Nevertheless, it remains largely unclear when and how the mTE differentiates and acquires this ability during implantation. Here, by RNA sequencing analysis with the pre- and peri-implantation mTE, we show that the mTE undergoes stage-specific and dynamic changes of gene expression during implantation. We also reveal that the mTE begins down-regulating Cdx2 and up-regulating differentiation marker genes during the peri-implantation stage. In addition, using trophectoderm (TE) -specific lentiviral vector-mediated gene transduction, we demonstrate that TE-specific Cdx2 overexpression represses differentiation of the mTE into the primary trophoblast giant cells. Moreover, we reveal that TE-specific Cdx2 overexpression also represses the up-regulation of cell adhesion- and migration-related genes, including Slc6a14, Slc16a3, Itga7, Itgav and Itgb3, which are known to regulate migration of trophectoderm cells. In particular, the expression of Itgb3, an integrin subunit gene, exhibits high inverse correlation with that of Cdx2 in the TE. Reflecting the down-regulation of the genes for TE migration, TE-specific Cdx2 overexpression causes suppression of the blastocyst outgrowth in vitro and abnormal progression of implantation in vivo. Thus, our results specify the time-course changes of global gene expression in the mTE during implantation and uncover the significance of Cdx2 down-regulation for implantation progression.
Collapse
Affiliation(s)
- Daisuke Suzuki
- Department of Bioscience, Graduate School of Life Science, Tokyo University of Agriculture, Tokyo, Japan
| | - Keisuke Sasaki
- Bioresource Center, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Soichiro Kumamoto
- NODAI Genome Research Center, Tokyo University of Agriculture, Tokyo, Japan
| | - Keisuke Tanaka
- NODAI Genome Research Center, Tokyo University of Agriculture, Tokyo, Japan
| | - Hidehiko Ogawa
- Department of Bioscience, Graduate School of Life Science, Tokyo University of Agriculture, Tokyo, Japan
- *Correspondence: Hidehiko Ogawa,
| |
Collapse
|
6
|
Yu Q, Tu H, Yin X, Peng C, Dou C, Yang W, Wu W, Guan X, Li J, Yan H, Zang Y, Jiang H, Xia Q. Targeting Glutamine Metabolism Ameliorates Autoimmune Hepatitis via Inhibiting T Cell Activation and Differentiation. Front Immunol 2022; 13:880262. [PMID: 35663990 PMCID: PMC9160195 DOI: 10.3389/fimmu.2022.880262] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/21/2022] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Autoimmune hepatitis (AIH) is mediated by a cascade of T cell-mediated events directed at liver cells and persistent inflammation within the liver can eventually result in liver cirrhosis. Targeting glutamine metabolism has an impact on T cell activation and differentiation. However, the effect of glutamine metabolism blocking upon AIH remains unknown. We use glutaminase antagonist 6-diazo-5-oxo-L-norleucine (DON) for in vitro assays and its prodrug 2-(2-amino-4-methylpentanamido)-DON (JHU083) for in vivo assays to investigate the potential therapeutic effect and molecular mechanism of glutamine metabolism blocking in an AIH murine model. METHODS AIH mice were treated with JHU083 or vehicle before concanavalin A (ConA) administration, and disease severity was examined. Then activation and differentiation [including Th1/Th17 cells and cytotoxic T lymphocytes (CTL)] of T cells from Vehicle-WT, JHU083-AIH and Vehicle-AIH mice were tested. Furthermore, in vitro T cell activation and differentiation were measured using separated splenocytes stimulated with ConA with or without DON. The activation and differentiation of T cells were tested using flow cytometry, qRT-PCR and ELISA. Phosphorylation level of mammalian target of rapamycin (mTOR) and 70 kDa ribosomal protein S6 kinase (P70S6K) were examined by western blotting. RESULTS JHU083 and DON significantly suppressed the activation of T cells and inhibited the differentiation of Th1/Th17 cells and CTL in vivo and in vitro. Besides, we demonstrated that glutamine metabolism blocking inhibited T cells activation and differentiation through decreasing the mRNA expression of amino acid transporter solute carrier family 7 member 5 (SLC7A5) and mitigating the activation of mTOR signaling. CONCLUSIONS We proved that targeting glutamine metabolism represents a potential new treatment strategy for patients with AIH and other T cell-mediated disease. Mechanistically, we demonstrated that glutamine metabolism blocking inhibits T cells activation and suppresses the differentiation of Th1/Th17 cells and CTL.
Collapse
Affiliation(s)
- Qiang Yu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Honghu Tu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xueyi Yin
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chang Peng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Chuanyun Dou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Wenhua Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wenbiao Wu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences (UCAS), Hangzhou, China
| | - Xiaotong Guan
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences (UCAS), Hangzhou, China
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Hexin Yan
- Department of Anesthesia, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Zang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences (UCAS), Hangzhou, China
| | - Haowen Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai, China
- Shanghai Institute of Transplantation, Shanghai, China
| |
Collapse
|
7
|
Epigenetic Modifications at the Center of the Barker Hypothesis and Their Transgenerational Implications. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182312728. [PMID: 34886453 PMCID: PMC8656758 DOI: 10.3390/ijerph182312728] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/15/2021] [Accepted: 11/29/2021] [Indexed: 02/08/2023]
Abstract
Embryo/fetal nutrition and the environment in the reproductive tract influence the subsequent risk of developing adult diseases and disorders, as formulated in the Barker hypothesis. Metabolic syndrome, obesity, heart disease, and hypertension in adulthood have all been linked to unwanted epigenetic programing in embryos and fetuses. Multiple studies support the conclusion that environmental challenges, such as a maternal low-protein diet, can change one-carbon amino acid metabolism and, thus, alter histone and DNA epigenetic modifications. Since histones influence gene expression and the program of embryo development, these epigenetic changes likely contribute to the risk of adult disease onset not just in the directly affected offspring, but for multiple generations to come. In this paper, we hypothesize that the effects of parental nutritional status on fetal epigenetic programming are transgenerational and warrant further investigation. Numerous studies supporting this hypothesis are reviewed, and potential research techniques to study these transgenerational epigenetic effects are offered.
Collapse
|
8
|
Amino Acid Transport and Metabolism Regulate Early Embryo Development: Species Differences, Clinical Significance, and Evolutionary Implications. Cells 2021; 10:cells10113154. [PMID: 34831375 PMCID: PMC8618253 DOI: 10.3390/cells10113154] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 12/11/2022] Open
Abstract
In this review we discuss the beneficial effects of amino acid transport and metabolism on pre- and peri-implantation embryo development, and we consider how disturbances in these processes lead to undesirable health outcomes in adults. Proline, glutamine, glycine, and methionine transport each foster cleavage-stage development, whereas leucine uptake by blastocysts via transport system B0,+ promotes the development of trophoblast motility and the penetration of the uterine epithelium in mammalian species exhibiting invasive implantation. (Amino acid transport systems and transporters, such as B0,+, are often oddly named. The reader is urged to focus on the transporters’ functions, not their names.) B0,+ also accumulates leucine and other amino acids in oocytes of species with noninvasive implantation, thus helping them to produce proteins to support later development. This difference in the timing of the expression of system B0,+ is termed heterochrony—a process employed in evolution. Disturbances in leucine uptake via system B0,+ in blastocysts appear to alter the subsequent development of embryos, fetuses, and placentae, with undesirable consequences for offspring. These consequences may include greater adiposity, cardiovascular dysfunction, hypertension, neural abnormalities, and altered bone growth in adults. Similarly, alterations in amino acid transport and metabolism in pluripotent cells in the blastocyst inner cell mass likely lead to epigenetic DNA and histone modifications that produce unwanted transgenerational health outcomes. Such outcomes might be avoided if we learn more about the mechanisms of these effects.
Collapse
|
9
|
Chen PR, Redel BK, Kerns KC, Spate LD, Prather RS. Challenges and Considerations during In Vitro Production of Porcine Embryos. Cells 2021; 10:cells10102770. [PMID: 34685749 PMCID: PMC8535139 DOI: 10.3390/cells10102770] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 02/02/2023] Open
Abstract
Genetically modified pigs have become valuable tools for generating advances in animal agriculture and human medicine. Importantly, in vitro production and manipulation of embryos is an essential step in the process of creating porcine models. As the in vitro environment is still suboptimal, it is imperative to examine the porcine embryo culture system from several angles to identify methods for improvement. Understanding metabolic characteristics of porcine embryos and considering comparisons with other mammalian species is useful for optimizing culture media formulations. Furthermore, stressors arising from the environment and maternal or paternal factors must be taken into consideration to produce healthy embryos in vitro. In this review, we progress stepwise through in vitro oocyte maturation, fertilization, and embryo culture in pigs to assess the status of current culture systems and address points where improvements can be made.
Collapse
Affiliation(s)
- Paula R. Chen
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | | | - Karl C. Kerns
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - Lee D. Spate
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
- National Swine Resource and Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Randall S. Prather
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
- National Swine Resource and Research Center, University of Missouri, Columbia, MO 65211, USA
- Correspondence:
| |
Collapse
|
10
|
Leese HJ, McKeegan PJ, Sturmey RG. Amino Acids and the Early Mammalian Embryo: Origin, Fate, Function and Life-Long Legacy. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:9874. [PMID: 34574797 PMCID: PMC8467587 DOI: 10.3390/ijerph18189874] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 12/11/2022]
Abstract
Amino acids are now recognised as having multiple cellular functions in addition to their traditional role as constituents of proteins. This is well-illustrated in the early mammalian embryo where amino acids are now known to be involved in intermediary metabolism, as energy substrates, in signal transduction, osmoregulation and as intermediaries in numerous pathways which involve nitrogen metabolism, e.g., the biosynthesis of purines, pyrimidines, creatine and glutathione. The amino acid derivative S-adenosylmethionine has emerged as a universal methylating agent with a fundamental role in epigenetic regulation. Amino acids are now added routinely to preimplantation embryo culture media. This review examines the routes by which amino acids are supplied to the early embryo, focusing on the role of the oviduct epithelium, followed by an outline of their general fate and function within the embryo. Functions specific to individual amino acids are then considered. The importance of amino acids during the preimplantation period for maternal health and that of the conceptus long term, which has come from the developmental origins of health and disease concept of David Barker, is discussed and the review concludes by considering the potential utility of amino acid profiles as diagnostic of embryo health.
Collapse
Affiliation(s)
- Henry J. Leese
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull HU6 7RX, UK;
| | - Paul J. McKeegan
- Centre for Anatomical and Human Sciences, Hull York Medical School, University of Hull, Hull HU6 7RX, UK;
| | - Roger G. Sturmey
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull HU6 7RX, UK;
- Division of Developmental Biology and Medicine, The University of Manchester, St Mary’s Hospital, Manchester M13 9WL, UK
| |
Collapse
|
11
|
Sahoo DP, Van Winkle LJ, Díaz de la Garza RI, Dubrovsky JG. Interkingdom Comparison of Threonine Metabolism for Stem Cell Maintenance in Plants and Animals. Front Cell Dev Biol 2021; 9:672545. [PMID: 34557481 PMCID: PMC8454773 DOI: 10.3389/fcell.2021.672545] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 08/11/2021] [Indexed: 01/12/2023] Open
Abstract
In multicellular organisms, tissue generation, maintenance, and homeostasis depend on stem cells. Cellular metabolic status is an essential component of different differentiated states, from stem to fully differentiated cells. Threonine (Thr) metabolism has emerged as a critical factor required to maintain pluripotent/multipotent stem cells in both plants and animals. Thus, both kingdoms conserved or converged upon this fundamental feature of stem cell function. Here, we examine similarities and differences in Thr metabolism-dependent mechanisms supporting stem cell maintenance in these two kingdoms. We then consider common features of Thr metabolism in stem cell maintenance and predict and speculate that some knowledge about Thr metabolism and its role in stem cell function in one kingdom may apply to the other. Finally, we outline future research directions to explore these hypotheses.
Collapse
Affiliation(s)
- Debee Prasad Sahoo
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Lon J. Van Winkle
- Department of Biochemistry, Midwestern University, Downers Grove, IL, United States
- Department of Medical Humanities, Rocky Vista University, Parker, CO, United States
| | | | - Joseph G. Dubrovsky
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| |
Collapse
|
12
|
Fleming TP, Sun C, Denisenko O, Caetano L, Aljahdali A, Gould JM, Khurana P. Environmental Exposures around Conception: Developmental Pathways Leading to Lifetime Disease Risk. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:9380. [PMID: 34501969 PMCID: PMC8431664 DOI: 10.3390/ijerph18179380] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/11/2022]
Abstract
Environment around conception can influence the developmental programme with lasting effects on gestational and postnatal phenotype and with consequences for adult health and disease risk. Peri-conception exposure comprises a crucial part of the 'Developmental Origins of Health and Disease' (DOHaD) concept. In this review, we consider the effects of maternal undernutrition experienced during the peri-conception period in select human models and in a mouse experimental model of protein restriction. Human datasets indicate that macronutrient deprivation around conception affect the epigenome, with enduring effects on cardiometabolic and neurological health. The mouse model, comprising maternal low protein diet exclusively during the peri-conception period, has revealed a stepwise progression in altered developmental programming following induction through maternal metabolite deficiency. This progression includes differential effects in extra-embryonic and embryonic cell lineages and tissues, leading to maladaptation in the growth trajectory and increased chronic disease comorbidities. The timeline embraces an array of mechanisms across nutrient sensing and signalling, cellular, metabolic, epigenetic and physiological processes with a coordinating role for mTORC1 signalling proposed. Early embryos appear active participants in environmental sensing to optimise the developmental programme for survival but with the trade-off of later disease. Similar adverse health outcomes may derive from other peri-conception environmental experiences, including maternal overnutrition, micronutrient availability, pollutant exposure and assisted reproductive treatments (ART) and support the need for preconception health before pregnancy.
Collapse
Affiliation(s)
- Tom P. Fleming
- Biological Sciences, Southampton General Hospital, University of Southampton, Southampton SO16 6YD, UK; (L.C.); (A.A.); (P.K.)
| | - Congshan Sun
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
- Center for Genetic Muscle Disorders, Hugo W. Moser Research Institute at Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Oleg Denisenko
- Department of Medicine, University of Washington, 850 Republican St., Rm 242, Seattle, WA 98109, USA;
| | - Laura Caetano
- Biological Sciences, Southampton General Hospital, University of Southampton, Southampton SO16 6YD, UK; (L.C.); (A.A.); (P.K.)
| | - Anan Aljahdali
- Biological Sciences, Southampton General Hospital, University of Southampton, Southampton SO16 6YD, UK; (L.C.); (A.A.); (P.K.)
- Department of Biological Sciences, Faculty of Science, Alfaisaliah campus, University of Jeddah, Jeddah 23442, Saudi Arabia
| | - Joanna M. Gould
- Clinical Neurosciences and Psychiatry, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK;
| | - Pooja Khurana
- Biological Sciences, Southampton General Hospital, University of Southampton, Southampton SO16 6YD, UK; (L.C.); (A.A.); (P.K.)
- Institute for Biogenesis Research, Research Corporation of the University of Hawaii, Manoa, Honolulu, HI 96822, USA
| |
Collapse
|
13
|
ÇEVİK M, GENÇ MD. Embryonic Diapause. MEHMET AKIF ERSOY ÜNIVERSITESI VETERINER FAKÜLTESI DERGISI 2021. [DOI: 10.24880/maeuvfd.835288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
14
|
Calderari S, Daniel N, Mourier E, Richard C, Dahirel M, Lager F, Marchiol C, Renault G, Gatien J, Nadal-Desbarats L, Chavatte-Palmer P, Duranthon V. Metabolomic differences in blastocoel and uterine fluids collected in vivo by ultrasound biomicroscopy on rabbit embryos†. Biol Reprod 2021; 104:794-805. [PMID: 33459770 DOI: 10.1093/biolre/ioab005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/24/2020] [Accepted: 01/07/2021] [Indexed: 11/12/2022] Open
Abstract
The success of embryo development and implantation depends in part on the environment in which the embryo evolves. However, the composition of the uterine fluid surrounding the embryo in the peri-implantation period remains poorly studied. In this work, we aimed to develop a new strategy to visualize, collect, and analyze both blastocoelic liquid and juxta-embryonic uterine fluid from in vivo peri-implantation rabbit embryos. Using high-resolution ultrasound biomicroscopy, embryos were observed as fluid-filled anechoic vesicles, some of which were surrounded by a thin layer of uterine fluid. Ultrasound-guided puncture and aspiration of both the blastocoelic fluid contained in the embryo and the uterine fluid in the vicinity of the embryo were performed. Using nuclear magnetic resonance spectroscopy, altogether 24 metabolites were identified and quantified, of which 21 were detected in both fluids with a higher concentration in the uterus compared to the blastocoel. In contrast, pyruvate was detected at a higher concentration in blastocoelic compared to uterine fluid. Two acidic amino acids, glutamate and aspartate, were not detected in uterine fluid in contrast to blastocoelic fluid, suggesting a local regulation of uterine fluid composition. To our knowledge, this is the first report of simultaneous analysis of blastocoelic and uterine fluids collected in vivo at the time of implantation in mammals, shedding new insight for understanding the relationship between the embryo and its local environment at this critical period of development.
Collapse
Affiliation(s)
- Sophie Calderari
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| | - Nathalie Daniel
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| | - Eve Mourier
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France.,Plateforme MIMA2-CIMA, Jouy en Josas, France
| | - Christophe Richard
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France.,Plateforme MIMA2-CIMA, Jouy en Josas, France
| | - Michele Dahirel
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France.,Plateforme MIMA2-CIMA, Jouy en Josas, France
| | - Franck Lager
- INSERM U1016, Institut Cochin, Université Paris Descartes, PRES Sorbonne Paris Cité, Paris, France
| | - Carmen Marchiol
- INSERM U1016, Institut Cochin, Université Paris Descartes, PRES Sorbonne Paris Cité, Paris, France
| | - Gilles Renault
- INSERM U1016, Institut Cochin, Université Paris Descartes, PRES Sorbonne Paris Cité, Paris, France
| | - Julie Gatien
- Research and Development Department, Allice, Nouzilly, France
| | - Lydie Nadal-Desbarats
- UMR 1253, iBrain, Inserm, University of Tours, Tours, France.,PST-ASB, University of Tours, Tours, France
| | - Pascale Chavatte-Palmer
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France.,Plateforme MIMA2-CIMA, Jouy en Josas, France
| | - Véronique Duranthon
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| |
Collapse
|
15
|
Nałęcz KA. Amino Acid Transporter SLC6A14 (ATB 0,+) - A Target in Combined Anti-cancer Therapy. Front Cell Dev Biol 2020; 8:594464. [PMID: 33195271 PMCID: PMC7609839 DOI: 10.3389/fcell.2020.594464] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer cells are characterized by quick growth and proliferation, demanding constant supply of various nutrients. Several plasma membrane transporters delivering such compounds are upregulated in cancer. Solute carrier family 6 member 14 (SLC6A14), known as amino acid transporter B0,+ (ATB0,+) transports all amino acids with exception of the acidic ones: aspartate and glutamate. Its malfunctioning is correlated with several pathological states and it is upregulated in solid tumors. The high expression of SLC6A14 is prognostic and unfavorable in pancreatic cancer, while in breast cancer it is expressed in estrogen receptor positive cells. As many plasma membrane transporters it resides in endoplasmic reticulum (ER) membrane after translation before further trafficking through Golgi to the cell surface. Transporter exit from ER is strictly controlled. The proper folding of SLC6A14 was shown to be controlled from the cytoplasmic side by heat shock proteins, further exit from ER and formation of coatomer II (COPII) coated vesicles depends on specific interaction with COPII cargo-recognizing subunit SEC24C, phosphorylated by kinase AKT. Inhibition of heat shock proteins, known to be upregulated in cancer, directs SLC6A14 to degradation. Targeting proteins regulating SLC6A14 trafficking is proposed as an additional pharmacological treatment of cancer.
Collapse
Affiliation(s)
- Katarzyna A Nałęcz
- Laboratory of Transport Through Biomembranes, Nencki Institute of Experimental Biology, Warsaw, Poland
| |
Collapse
|
16
|
Fenelon JC, Murphy BD. New functions for old factors: the role of polyamines during the establishment of pregnancy. Reprod Fertil Dev 2020; 31:1228-1239. [PMID: 30418870 DOI: 10.1071/rd18235] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/01/2018] [Indexed: 12/16/2022] Open
Abstract
Implantation is essential for the establishment of a successful pregnancy, and the preimplantation period plays a significant role in ensuring implantation occurs in a timely and coordinated manner. This requires effective maternal-embryonic signalling, established during the preimplantation period, to synchronise development. Although multiple factors have been identified as present during this time, the exact molecular mechanisms involved are unknown. Polyamines are small cationic molecules that are ubiquitously expressed from prokaryotes to eukaryotes. Despite being first identified over 300 years ago, their essential roles in cell proliferation and growth, including cancer, have only been recently recognised, with new technologies and interest resulting in rapid expansion of the polyamine field. This review provides a summary of our current understanding of polyamine synthesis, regulation and function with a focus on recent developments demonstrating the requirements for polyamines during the establishment of pregnancy up to the implantation stage, in particular the role of polyamines in the control of embryonic diapause and the identification of an alternative pathway for their synthesis in sheep pregnancy. This, along with other novel discoveries, provides new insights into the control of the peri-implantation period in mammals and highlights the complexities that exist in regulating this critical period of pregnancy.
Collapse
Affiliation(s)
- Jane C Fenelon
- School of BioSciences, The University of Melbourne, Parkville, Vic. 3010, Australia
| | - Bruce D Murphy
- Centre de recherché en reproduction et fertilité, Faculté de médicine vétérinaire, Université de Montréal, 3200 Rue Sicotte, Saint-Hyacinthe, Quebec J2S 2M2, Canada
| |
Collapse
|
17
|
Juntanapum W, Bunchasak C, Poeikhampha T, Rakangthong C, Poungpong K. The Effects of Supplementing Lysophosphatidylcholine (LPC) in the Diet on Production Performance, Fat Digestibility, Blood Lipid Profile, and Gene Expression Related to Nutrients Transport in Small Intestine of Laying Hens. JOURNAL OF ANIMAL AND FEED SCIENCES 2020. [DOI: 10.22358/jafs/127689/2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
18
|
To CY, Freeman M, Van Winkle LJ. Consumption of a Branched-Chain Amino Acid (BCAA) during Days 2-10 of Pregnancy Causes Abnormal Fetal and Placental Growth: Implications for BCAA Supplementation in Humans. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17072445. [PMID: 32260232 PMCID: PMC7178017 DOI: 10.3390/ijerph17072445] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 03/29/2020] [Accepted: 03/30/2020] [Indexed: 12/27/2022]
Abstract
A relatively large branched-chain amino acid (BCAA) supplement, consumed for more than 10 days, appears to be especially effective at alleviating muscle damage and soreness during intense human training. However, perturbations in amino acid and protein consumption could have unwanted transgenerational effects on male and female reproduction. This paper hypothesizes that isoleucine consumption by female mice from days 2 to 10 of pregnancy will alter fetal and placental growth later in gestation. Mice that had received 118 mM isoleucine in their drinking water delivered pups on day 19 of pregnancy that were 9% larger than normal, whereas the reverse was true for pups born on day 20. Moreover, the inverse correlation between birth weight and litter size was lost in mice that previously consumed excess isoleucine. Similarly, the normal correlations between fetal and placental weights were lost by day 18 of pregnancy in mice that had consumed excess isoleucine. Mice that consumed excess isoleucine had placentas smaller than, and fetuses larger than normal on day 18 of pregnancy, but the reverse was true on day 15. Other unintended and unexpected effects of BCAA consumption should be studied more thoroughly due to the increasing use of BCAAs to alleviate muscle damage and soreness in athletes.
Collapse
Affiliation(s)
- Chiu Yuen To
- Department of Surgery, Division of Neurosurgery, St Francis Hospital, Memphis, TN 38119, USA;
- Department of Biochemistry, Midwestern University, Downers Grove, IL 60515, USA;
| | - Muriel Freeman
- Department of Biochemistry, Midwestern University, Downers Grove, IL 60515, USA;
- Department of Surgery, Division of Podiatric Medicine and Surgery, Carle Richland Memorial Hospital, Olney, IL 62450, USA
| | - Lon J. Van Winkle
- Department of Biochemistry, Midwestern University, Downers Grove, IL 60515, USA;
- Department of Medical Humanities, Rocky Vista University 8401 S. Chambers Road, Parker, CO 80134, USA
- Correspondence:
| |
Collapse
|
19
|
Hussein AM, Wang Y, Mathieu J, Margaretha L, Song C, Jones DC, Cavanaugh C, Miklas JW, Mahen E, Showalter MR, Ruzzo WL, Fiehn O, Ware CB, Blau CA, Ruohola-Baker H. Metabolic Control over mTOR-Dependent Diapause-like State. Dev Cell 2020; 52:236-250.e7. [PMID: 31991105 DOI: 10.1016/j.devcel.2019.12.018] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 09/13/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022]
Abstract
Regulation of embryonic diapause, dormancy that interrupts the tight connection between developmental stage and time, is still poorly understood. Here, we characterize the transcriptional and metabolite profiles of mouse diapause embryos and identify unique gene expression and metabolic signatures with activated lipolysis, glycolysis, and metabolic pathways regulated by AMPK. Lipolysis is increased due to mTORC2 repression, increasing fatty acids to support cell survival. We further show that starvation in pre-implantation ICM-derived mouse ESCs induces a reversible dormant state, transcriptionally mimicking the in vivo diapause stage. During starvation, Lkb1, an upstream kinase of AMPK, represses mTOR, which induces a reversible glycolytic and epigenetically H4K16Ac-negative, diapause-like state. Diapause furthermore activates expression of glutamine transporters SLC38A1/2. We show by genetic and small molecule inhibitors that glutamine transporters are essential for the H4K16Ac-negative, diapause state. These data suggest that mTORC1/2 inhibition, regulated by amino acid levels, is causal for diapause metabolism and epigenetic state.
Collapse
Affiliation(s)
- Abdiasis M Hussein
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Yuliang Wang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA 98195, USA
| | - Julie Mathieu
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Comparative Medicine, University of Washington, Seattle, WA 98195, USA
| | - Lilyana Margaretha
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Molecular and Cellular Biology, University of Washington, Seattle, WA 98109, USA
| | - Chaozhong Song
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Medicine, Division of Hematology, University of Washington, Seattle, WA 98195, USA
| | - Daniel C Jones
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA 98195, USA
| | - Christopher Cavanaugh
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Comparative Medicine, University of Washington, Seattle, WA 98195, USA
| | - Jason W Miklas
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Elisabeth Mahen
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Medicine, Division of Hematology, University of Washington, Seattle, WA 98195, USA
| | - Megan R Showalter
- West Coast Metabolomics Center, University of California, Davis, Davis, CA 95616, USA
| | - Walter L Ruzzo
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA 98195, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, Davis, CA 95616, USA
| | - Carol B Ware
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Comparative Medicine, University of Washington, Seattle, WA 98195, USA
| | - C Anthony Blau
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Medicine, Division of Hematology, University of Washington, Seattle, WA 98195, USA
| | - Hannele Ruohola-Baker
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
20
|
Poncet N, Halley PA, Lipina C, Gierliński M, Dady A, Singer GA, Febrer M, Shi Y, Yamaguchi TP, Taylor PM, Storey KG. Wnt regulates amino acid transporter Slc7a5 and so constrains the integrated stress response in mouse embryos. EMBO Rep 2020; 21:e48469. [PMID: 31789450 PMCID: PMC6944906 DOI: 10.15252/embr.201948469] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 10/18/2019] [Accepted: 10/25/2019] [Indexed: 12/29/2022] Open
Abstract
Amino acids are essential for cellular metabolism, and it is important to understand how nutrient supply is coordinated with changing energy requirements during embryogenesis. Here, we show that the amino acid transporter Slc7a5/Lat1 is highly expressed in tissues undergoing morphogenesis and that Slc7a5-null mouse embryos have profound neural and limb bud outgrowth defects. Slc7a5-null neural tissue exhibited aberrant mTORC1 activity and cell proliferation; transcriptomics, protein phosphorylation and apoptosis analyses further indicated induction of the integrated stress response as a potential cause of observed defects. The pattern of stress response gene expression induced in Slc7a5-null embryos was also detected at low level in wild-type embryos and identified stress vulnerability specifically in tissues undergoing morphogenesis. The Slc7a5-null phenotype is reminiscent of Wnt pathway mutants, and we show that Wnt/β-catenin loss inhibits Slc7a5 expression and induces this stress response. Wnt signalling therefore normally supports the metabolic demands of morphogenesis and constrains cellular stress. Moreover, operation in the embryo of the integrated stress response, which is triggered by pathogen-mediated as well as metabolic stress, may provide a mechanistic explanation for a range of developmental defects.
Collapse
Affiliation(s)
- Nadège Poncet
- Division of Cell & Developmental BiologySchool of Life SciencesUniversity of DundeeDundeeUK
- Present address:
Institute of PhysiologyUniversity of ZürichZürichSwitzerland
| | - Pamela A Halley
- Division of Cell & Developmental BiologySchool of Life SciencesUniversity of DundeeDundeeUK
| | - Christopher Lipina
- Division of Cell Signalling and ImmunologySchool of Life SciencesUniversity of DundeeDundeeUK
| | - Marek Gierliński
- Division of Computational BiologySchool of Life SciencesUniversity of DundeeDundeeUK
| | - Alwyn Dady
- Division of Cell & Developmental BiologySchool of Life SciencesUniversity of DundeeDundeeUK
| | - Gail A Singer
- Division of Cell & Developmental BiologySchool of Life SciencesUniversity of DundeeDundeeUK
| | - Melanie Febrer
- Sequencing FacilitySchool of Life SciencesUniversity of DundeeDundeeUK
- Present address:
Illumina CanadaVictoriaBCCanada
| | - Yun‐Bo Shi
- Section on Molecular MorphogenesisNICHD, NIHBethesdaMDUSA
| | - Terry P Yamaguchi
- Cancer and Developmental Biology LaboratoryCenter for Cancer ResearchNational Cancer Institute‐Frederick, NIHFrederickMDUSA
| | - Peter M Taylor
- Division of Cell Signalling and ImmunologySchool of Life SciencesUniversity of DundeeDundeeUK
| | - Kate G Storey
- Division of Cell & Developmental BiologySchool of Life SciencesUniversity of DundeeDundeeUK
| |
Collapse
|
21
|
Van Winkle LJ, Ryznar R. One-Carbon Metabolism Regulates Embryonic Stem Cell Fate Through Epigenetic DNA and Histone Modifications: Implications for Transgenerational Metabolic Disorders in Adults. Front Cell Dev Biol 2019; 7:300. [PMID: 31824950 PMCID: PMC6882271 DOI: 10.3389/fcell.2019.00300] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022] Open
Abstract
Human (h) and mouse (m) embryonic stem (ES) cells need specific amino acids to proliferate. mES cells require threonine (Thr) metabolism for epigenetic histone modifications. Thr is converted to glycine and acetyl CoA, and the glycine is metabolized specifically to regulate trimethylation of lysine (Lys) residue 4 in histone H3 (H3K4me3). DNA methylation and methylation of other H3 Lys residues remain unimpaired by Thr deprivation in mES cell culture medium. Similarly, hES cells require methionine (Met) to maintain the Met-SAM (S-adenosyl methionine) cycle of 1-carbon metabolism also for H3K4me3 formation. H3K4me3 is needed specifically to regulate and maintain both mES and hES cell proliferation and their pluripotent states. Better understanding of this regulation is essential since treatment of human diseases and disorders will increasingly involve hES cells. Furthermore, since ES cells are derived from their progenitor cells in preimplantation blastocysts, they serve as models of 1-carbon metabolism in these precursors of all mammalian tissues and organs. One-carbon metabolism challenges, such as a maternal low protein diet (LPD) during preimplantation blastocyst development, contribute to development of metabolic syndrome and related abnormalities in adults. These 1-carbon metabolism challenges result in altered epigenetic DNA and histone modifications in ES progenitor cells and the tissues and organs to which they develop. Moreover, the modified histones could have extracellular as well as intracellular effects, since histones are secreted in uterine fluid and influence early embryo development. Hence, the mechanisms and transgenerational implications of these altered epigenetic DNA and histone modifications warrant concerted further study.
Collapse
Affiliation(s)
- Lon J Van Winkle
- Department of Biochemistry, Midwestern University, Downers Grove, IL, United States.,Department of Medical Humanities, Rocky Vista University, Parker, CO, United States
| | - Rebecca Ryznar
- Molecular Biology, Department of Biomedical Sciences, Rocky Vista University, Parker, CO, United States
| |
Collapse
|
22
|
Abstract
Amino acids perform a variety of functions in cells and organisms, particularly in the synthesis of proteins, as energy metabolites, neurotransmitters, and precursors for many other molecules. Amino acid transport plays a key role in all these functions. Inhibition of amino acid transport is pursued as a therapeutic strategy in several areas, such as diabetes and related metabolic disorders, neurological disorders, cancer, and stem cell biology. The role of amino acid transporters in these disorders and processes is well established, but the implementation of amino acid transporters as drug targets is still in its infancy. This is at least in part due to the underdeveloped pharmacology of this group of membrane proteins. Recent advances in structural biology, membrane protein expression, and inhibitor screening methodology will see an increased number of improved and selective inhibitors of amino acid transporters that can serve as tool compounds for further studies.
Collapse
Affiliation(s)
- Stefan Bröer
- 1 Research School of Biology, College of Science, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
23
|
Van Winkle LJ, Ryznar R. Can uterine secretion of modified histones alter blastocyst implantation, embryo nutrition, and transgenerational phenotype? Biomol Concepts 2018; 9:176-183. [PMID: 30864391 DOI: 10.1515/bmc-2018-0017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/03/2018] [Indexed: 12/19/2022] Open
Abstract
Extracellular histones support rodent and human embryo development in at least two ways. First, these molecules in uterine secretions protect embryos from inflammation caused by pathogens that gain access to the reproductive tract. Also, histones in uterine secretions likely support penetration of the uterine epithelium by blastocysts during embryo implantation. Extracellular histones seem to preserve amino acid transport system B0,+ in blastocysts by inhibiting its activity. Preservation of system B0,+ is needed because, at the time of invasion of the uterine epithelium by motile trophoblasts, system B0,+ is likely reactivated to help remove tryptophan from the implantation chamber. If tryptophan is not removed, T-cells proliferate and reject the implanting blastocyst. Epigenetic modification of histones could alter their promotion of normal implantation through, say, incomplete tryptophan removal and, thus, allow partial T-cell rejection of the conceptus. Such partial rejection could impair placental development, embryonal/fetal nutrition, and weight gain prior to birth. Small-for-gestational-age offspring are predisposed to developing metabolic syndrome, obesity, and associated complications as adults. Shifting expression of these phenotypes might contribute to transgenerational variation and evolution. The spectrum of possible extracellular histone targets in early development warrant new research, especially since the effects of epigenetic histone modifications might be transgenerational.
Collapse
Affiliation(s)
- Lon J Van Winkle
- Emeritus Professor, Department of Biochemistry, Midwestern University, chicago, USA.,Professor, Department of Medical Humanities, Rocky Vista University (RVU), Parker, USA
| | - Rebecca Ryznar
- Assistant Professor of Molecular Biology, Department of Biomedical Sciences, RVU, 8401 S. Chambers Road, Parker, CO 80134, USA
| |
Collapse
|
24
|
Abstract
The small intestine mediates the absorption of amino acids after ingestion of protein and sustains the supply of amino acids to all tissues. The small intestine is an important contributor to plasma amino acid homeostasis, while amino acid transport in the large intestine is more relevant for bacterial metabolites and fluid secretion. A number of rare inherited disorders have contributed to the identification of amino acid transporters in epithelial cells of the small intestine, in particular cystinuria, lysinuric protein intolerance, Hartnup disorder, iminoglycinuria, and dicarboxylic aminoaciduria. These are most readily detected by analysis of urine amino acids, but typically also affect intestinal transport. The genes underlying these disorders have all been identified. The remaining transporters were identified through molecular cloning techniques to the extent that a comprehensive portrait of functional cooperation among transporters of intestinal epithelial cells is now available for both the basolateral and apical membranes. Mouse models of most intestinal transporters illustrate their contribution to amino acid homeostasis and systemic physiology. Intestinal amino acid transport activities can vary between species, but these can now be explained as differences of amino acid transporter distribution along the intestine. © 2019 American Physiological Society. Compr Physiol 9:343-373, 2019.
Collapse
Affiliation(s)
- Stefan Bröer
- Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia
| | - Stephen J Fairweather
- Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia
| |
Collapse
|
25
|
Kovalchuk V, Samluk Ł, Juraszek B, Jurkiewicz-Trząska D, Sucic S, Freissmuth M, Nałęcz KA. Trafficking of the amino acid transporter B 0,+ (SLC6A14) to the plasma membrane involves an exclusive interaction with SEC24C for its exit from the endoplasmic reticulum. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:252-263. [PMID: 30445147 PMCID: PMC6314439 DOI: 10.1016/j.bbamcr.2018.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 11/02/2018] [Accepted: 11/12/2018] [Indexed: 12/16/2022]
Abstract
A plasma membrane amino acid transporter B0,+ (ATB0,+), encoded by the SLC6A14 gene, is specific for neutral and basic amino acids. It is up-regulated in several types of malignant cancers. Neurotransmitter transporters of the SLC6 family interact with specific SEC24 proteins of the COPII complex along their pathway from the endoplasmic reticulum (ER) to Golgi. This study focused on the possible role of SEC24 proteins in ATB0,+ trafficking. Rat ATB0,+ was expressed in HEK293 cells, its localization and trafficking were examined by Western blot, deglycosylation, immunofluorescence (co-localization with ER and trans-Golgi markers) and biotinylation. The expression of ATB0,+ at the plasma membrane was decreased by dominant negative mutants of SAR1, a GTPase, whose activity triggers the formation of the COPII complex. ATB0,+ co-precipitated with SEC24C (but not with the remaining isoforms A, B and D). This interaction was confirmed by immunocytochemistry and the proximity ligation assay. Co-localization of SEC24C with endogenous ATB0,+ was also observed in MCF-7 breast cancer cells. Contrary to the endogenous transporter, part of the overexpressed ATB0,+ is directed to proteolysis, a process significantly reversed by a proteasome inhibitor bortezomib. Co-transfection with a SEC24C dominant negative mutant attenuated ATB0,+ expression at the plasma membrane, due to proteolytic degradation. These results support a hypothesis that lysine at position +2 downstream of the ER export "RI" motif on the cargo protein is crucial for SEC24C binding and for further trafficking to the Golgi. Moreover, there is an equilibrium between ER export and degradation mechanisms in case of overexpressed transporter.
Collapse
Affiliation(s)
- Vasylyna Kovalchuk
- Laboratory of Transport through Biomembranes, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Łukasz Samluk
- Laboratory of Transport through Biomembranes, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Barbara Juraszek
- Laboratory of Transport through Biomembranes, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Dominika Jurkiewicz-Trząska
- Laboratory of Transport through Biomembranes, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Sonja Sucic
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Katarzyna A Nałęcz
- Laboratory of Transport through Biomembranes, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland.
| |
Collapse
|
26
|
Kandasamy P, Gyimesi G, Kanai Y, Hediger MA. Amino acid transporters revisited: New views in health and disease. Trends Biochem Sci 2018; 43:752-789. [PMID: 30177408 DOI: 10.1016/j.tibs.2018.05.003] [Citation(s) in RCA: 302] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 05/23/2018] [Accepted: 05/25/2018] [Indexed: 02/09/2023]
Abstract
Amino acid transporters (AATs) are membrane-bound transport proteins that mediate transfer of amino acids into and out of cells or cellular organelles. AATs have diverse functional roles ranging from neurotransmission to acid-base balance, intracellular energy metabolism, and anabolic and catabolic reactions. In cancer cells and diabetes, dysregulation of AATs leads to metabolic reprogramming, which changes intracellular amino acid levels, contributing to the pathogenesis of cancer, obesity and diabetes. Indeed, the neutral amino acid transporters (NATs) SLC7A5/LAT1 and SLC1A5/ASCT2 are likely involved in several human malignancies. However, a clinical therapy that directly targets AATs has not yet been developed. The purpose of this review is to highlight the structural and functional diversity of AATs, their diverse physiological roles in different tissues and organs, their wide-ranging implications in human diseases and the emerging strategies and tools that will be necessary to target AATs therapeutically.
Collapse
Affiliation(s)
- Palanivel Kandasamy
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland
| | - Gergely Gyimesi
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland
| | - Yoshikatsu Kanai
- Division of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Matthias A Hediger
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland.
| |
Collapse
|
27
|
Velazquez MA, Sheth B, Smith SJ, Eckert JJ, Osmond C, Fleming TP. Insulin and branched-chain amino acid depletion during mouse preimplantation embryo culture programmes body weight gain and raised blood pressure during early postnatal life. Biochim Biophys Acta Mol Basis Dis 2017; 1864:590-600. [PMID: 29196239 PMCID: PMC5764225 DOI: 10.1016/j.bbadis.2017.11.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/09/2017] [Accepted: 11/26/2017] [Indexed: 02/07/2023]
Abstract
Mouse maternal low protein diet exclusively during preimplantation development (Emb-LPD) is sufficient to programme altered growth and cardiovascular dysfunction in offspring. Here, we use an in vitro model comprising preimplantation culture in medium depleted in insulin and branched-chain amino acids (BCAA), two proposed embryo programming inductive factors from Emb-LPD studies, to examine the consequences for blastocyst organisation and, after embryo transfer (ET), postnatal disease origin. Two-cell embryos were cultured to blastocyst stage in defined KSOM medium supplemented with four combinations of insulin and BCAA concentrations. Control medium contained serum insulin and uterine luminal fluid amino acid concentrations (including BCAA) found in control mothers from the maternal diet model (N-insulin + N-bcaa). Experimental medium (three groups) contained 50% reduction in insulin and/or BCAA (L-insulin + N-bcaa, N-insulin + L-bcaa, and L-insulin + N-bcaa). Lineage-specific cell numbers of resultant blastocysts were not affected by treatment. Following ET, a combined depletion of insulin and BCAA during embryo culture induced a non sex-specific increase in birth weight and weight gain during early postnatal life. Furthermore, male offspring displayed relative hypertension and female offspring reduced heart/body weight, both characteristics of Emb-LPD offspring. Combined depletion of metabolites also resulted in a strong positive correlation between body weight and glucose metabolism that was absent in the control group. Our results support the notion that composition of preimplantation culture medium can programme development and associate with disease origin affecting postnatal growth and cardiovascular phenotypes and implicate two important nutritional mediators in the inductive mechanism. Our data also have implications for human assisted reproductive treatment (ART) practice. Chronic disease may derive from maternal undernutrition during pregnancy, including the periconceptional period. Mouse embryos cultured in medium low in insulin and select amino acids gave rise to offspring with disease symptoms. We propose these metabolite deficiencies around conception induce adverse programming of the early embryo leading to increased disease risk in later life.
Collapse
Affiliation(s)
- Miguel A Velazquez
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; School of Natural and Environmental Sciences, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK
| | - Bhavwanti Sheth
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Stephanie J Smith
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Judith J Eckert
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Clive Osmond
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Tom P Fleming
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| |
Collapse
|
28
|
Van Winkle LJ. Uterine Histone Secretion Likely Fosters Early Embryo Development So Efforts to Mitigate Histone Cytotoxicity Should Be Cautious. Front Cell Dev Biol 2017; 5:100. [PMID: 29230391 PMCID: PMC5711778 DOI: 10.3389/fcell.2017.00100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 11/13/2017] [Indexed: 12/27/2022] Open
Affiliation(s)
- Lon J Van Winkle
- Department of Biochemistry, Midwestern University, Downers Grove, IL, United States.,Department of Medical Humanities, Rocky Vista University, Parker, CO, United States
| |
Collapse
|
29
|
Abstract
Embryonic diapause – a period of embryonic suspension at the blastocyst stage – is a fascinating phenomenon that occurs in over 130 species of mammals, ranging from bears and badgers to mice and marsupials. It might even occur in humans. During diapause, there is minimal cell division and greatly reduced metabolism, and development is put on hold. Yet there are no ill effects for the pregnancy when it eventually continues. Multiple factors can induce diapause, including seasonal supplies of food, temperature, photoperiod and lactation. The successful reactivation and continuation of pregnancy then requires a viable embryo, a receptive uterus and effective molecular communication between the two. But how do the blastocysts survive and remain viable during this period of time, which can be up to a year in some cases? And what are the signals that bring it out of suspended animation? Here, we provide an overview of the process of diapause and address these questions, focussing on recent molecular data.
Collapse
Affiliation(s)
- Marilyn B. Renfree
- School of BioSciences, The University of Melbourne, Victoria, Australia 3010
| | - Jane C. Fenelon
- The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H8L6
| |
Collapse
|
30
|
Sferruzzi-Perri AN, Sandovici I, Constancia M, Fowden AL. Placental phenotype and the insulin-like growth factors: resource allocation to fetal growth. J Physiol 2017; 595:5057-5093. [PMID: 28337745 DOI: 10.1113/jp273330] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/27/2017] [Indexed: 12/17/2022] Open
Abstract
The placenta is the main determinant of fetal growth and development in utero. It supplies all the nutrients and oxygen required for fetal growth and secretes hormones that facilitate maternal allocation of nutrients to the fetus. Furthermore, the placenta responds to nutritional and metabolic signals in the mother by altering its structural and functional phenotype, which can lead to changes in maternal resource allocation to the fetus. The molecular mechanisms by which the placenta senses and responds to environmental cues are poorly understood. This review discusses the role of the insulin-like growth factors (IGFs) in controlling placental resource allocation to fetal growth, particularly in response to adverse gestational environments. In particular, it assesses the impact of the IGFs and their signalling machinery on placental morphogenesis, substrate transport and hormone secretion, primarily in the laboratory species, although it draws on data from human and other species where relevant. It also considers the role of the IGFs as environmental signals in linking resource availability to fetal growth through changes in the morphological and functional phenotype of the placenta. As altered fetal growth is associated with increased perinatal morbidity and mortality and a greater risk of developing adult-onset diseases in later life, understanding the role of IGFs during pregnancy in regulating placental resource allocation to fetal growth is important for identifying the mechanisms underlying the developmental programming of offspring phenotype by suboptimal intrauterine growth.
Collapse
Affiliation(s)
- Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Ionel Sandovici
- Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Department of Obstetrics and Gynaecology and NIHR Cambridge Biomedical Research Centre, Robinson Way, Cambridge, CB2 0SW, UK
| | - Miguel Constancia
- Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Department of Obstetrics and Gynaecology and NIHR Cambridge Biomedical Research Centre, Robinson Way, Cambridge, CB2 0SW, UK
| | - Abigail L Fowden
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge, CB2 3EG, UK
| |
Collapse
|
31
|
Regulation of amino acid transporters in pluripotent cell populations in the embryo and in culture; novel roles for sodium-coupled neutral amino acid transporters. Mech Dev 2016; 141:32-39. [DOI: 10.1016/j.mod.2016.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/16/2016] [Accepted: 06/20/2016] [Indexed: 12/17/2022]
|
32
|
Demiral İ, Doğan M, Baştu E, Buyru F. Genomic, proteomic and lipidomic evaluation of endometrial receptivity. Turk J Obstet Gynecol 2015; 12:237-243. [PMID: 28913076 PMCID: PMC5588478 DOI: 10.4274/tjod.98475] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 01/02/2015] [Indexed: 01/11/2023] Open
Abstract
Endometrial receptivity is a complex phenomenon that plays a vital role in infertility. Although quality of embryo can be evaluated for a successful implantation, endometrial receptivity is still an unknown factor. With advances in technology, the microarray approach has provided an 'omic' tool to evaluate endometrial receptivity. In Latin, 'omic' means the whole family. The genomic, proteomic, and lipidomic evaluations of endometrium mean a wholesome evaluation of the genes, lipids and proteins of the endometrium. Evaluation of receptivity with this three-way approach may provide insight to the potential markers of implantation. Genomic analysis has been limited to date because not every gene alteration affects protein expression. Lipidomic analysis has recently gained popularity because lipids are strictly controlled during the implantation period. In summary, with the recent advances in microarray technology, genomic, lipidomic, and proteomic analyses of the endometrium may provide 'optimal' evaluation tools and criteria to assess receptivity in the near future.
Collapse
Affiliation(s)
- İrem Demiral
- İstanbul University Faculty of Medicine, Department of Obstetrics and Gynecology, İstanbul, Turkey
| | - Murat Doğan
- Acıbadem Fulya Hospital, Division of Reproductive Endocrinology and Infertility, İstanbul, Turkey
| | - Ercan Baştu
- İstanbul University Faculty of Medicine, Department of Obstetrics and Gynecology, İstanbul, Turkey
| | - Faruk Buyru
- İstanbul University Faculty of Medicine, Department of Obstetrics and Gynecology, İstanbul, Turkey
| |
Collapse
|
33
|
Land SC, Scott CL, Walker D. mTOR signalling, embryogenesis and the control of lung development. Semin Cell Dev Biol 2014; 36:68-78. [PMID: 25289569 DOI: 10.1016/j.semcdb.2014.09.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 09/07/2014] [Accepted: 09/11/2014] [Indexed: 12/15/2022]
Abstract
The existence of a nutrient sensitive "autocatakinetic" regulator of embryonic tissue growth has been hypothesised since the early 20th century, beginning with pioneering work on the determinants of foetal size by the Australian physiologist, Thorburn Brailsford-Robertson. We now know that the mammalian target of rapamycin complexes (mTORC1 and 2) perform this essential function in all eukaryotic tissues by balancing nutrient and energy supply during the first stages of embryonic cleavage, the formation of embryonic stem cell layers and niches, the highly specified programmes of tissue growth during organogenesis and, at birth, paving the way for the first few breaths of life. This review provides a synopsis of the role of the mTOR complexes in each of these events, culminating in an analysis of lung branching morphogenesis as a way of demonstrating the central role mTOR in defining organ structural complexity. We conclude that the mTOR complexes satisfy the key requirements of a nutrient sensitive growth controller and can therefore be considered as Brailsford-Robertson's autocatakinetic centre that drives tissue growth programmes during foetal development.
Collapse
Affiliation(s)
- Stephen C Land
- Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK.
| | - Claire L Scott
- Prostrakan Pharmaceuticals, Galabank Business Park, Galashiels TD1 1PR, UK
| | - David Walker
- School of Psychology & Neuroscience, Westburn Lane, St Andrews KY16 9JP, UK
| |
Collapse
|
34
|
Abstract
Amino acid (AA) transporters may act as sensors, as well as carriers, of tissue nutrient supplies. This review considers recent advances in our understanding of the AA-sensing functions of AA transporters in both epithelial and nonepithelial cells. These transporters mediate AA exchanges between extracellular and intracellular fluid compartments, delivering substrates to intracellular AA sensors. AA transporters on endosomal (eg, lysosomal) membranes may themselves function as intracellular AA sensors. AA transporters at the cell surface, particularly those for large neutral AAs such as leucine, interact functionally with intracellular nutrient-signaling pathways that regulate metabolism: for example, the mammalian target of rapamycin complex 1 (mTORC1) pathway, which promotes cell growth, and the general control non-derepressible (GCN) pathway, which is activated by AA starvation. Under some circumstances, upregulation of AA transporter expression [notably a leucine transporter, solute carrier 7A5 (SLC7A5)] is required to initiate AA-dependent activation of the mTORC1 pathway. Certain AA transporters may have dual receptor-transporter functions, operating as "transceptors" to sense extracellular (or intracellular) AA availability upstream of intracellular signaling pathways. New opportunities for nutritional therapy may include targeting of AA transporters (or mechanisms that upregulate their expression) to promote protein-anabolic signals for retention or recovery of lean tissue mass.
Collapse
Affiliation(s)
- Peter M Taylor
- Division of Cell Signalling & Immunology, College of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
35
|
Cheong Y, Boomsma C, Heijnen C, Macklon N. Uterine secretomics: a window on the maternal-embryo interface. Fertil Steril 2013; 99:1093-9. [DOI: 10.1016/j.fertnstert.2013.01.144] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 01/16/2013] [Accepted: 01/19/2013] [Indexed: 01/11/2023]
|
36
|
Eckert JJ, Porter R, Watkins AJ, Burt E, Brooks S, Leese HJ, Humpherson PG, Cameron IT, Fleming TP. Metabolic induction and early responses of mouse blastocyst developmental programming following maternal low protein diet affecting life-long health. PLoS One 2012; 7:e52791. [PMID: 23300778 PMCID: PMC3531326 DOI: 10.1371/journal.pone.0052791] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 11/21/2012] [Indexed: 12/21/2022] Open
Abstract
Previously, we have shown that a maternal low protein diet, fed exclusively during the preimplantation period of mouse development (Emb-LPD), is sufficient to induce by the blastocyst stage a compensatory growth phenotype in late gestation and postnatally, correlating with increased risk of adult onset cardiovascular disease and behavioural dysfunction. Here, we examine mechanisms of induction of maternal Emb-LPD programming and early compensatory responses by the embryo. Emb-LPD induced changes in maternal serum metabolites at the time of blastocyst formation (E3.5), notably reduced insulin and increased glucose, together with reduced levels of free amino acids (AAs) including branched chain AAs leucine, isoleucine and valine. Emb-LPD also caused reduction in the branched chain AAs within uterine fluid at the blastocyst stage. These maternal changes coincided with an altered content of blastocyst AAs and reduced mTORC1 signalling within blastocysts evident in reduced phosphorylation of effector S6 ribosomal protein and its ratio to total S6 protein but no change in effector 4E-BP1 phosphorylated and total pools. These changes were accompanied by increased proliferation of blastocyst trophectoderm and total cells and subsequent increased spreading of trophoblast cells in blastocyst outgrowths. We propose that induction of metabolic programming following Emb-LPD is achieved through mTORC1signalling which acts as a sensor for preimplantation embryos to detect maternal nutrient levels via branched chain AAs and/or insulin availability. Moreover, this induction step associates with changes in extra-embryonic trophectoderm behaviour occurring as early compensatory responses leading to later nutrient recovery.
Collapse
Affiliation(s)
- Judith J. Eckert
- Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
- Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Richard Porter
- Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Adam J. Watkins
- Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Elizabeth Burt
- Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Suzanne Brooks
- Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Henry J. Leese
- Centre for Cardiovascular and Metabolic Research, The Hull York Medical School, University of Hull, Hull, United Kingdom
| | | | - Iain T. Cameron
- Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Tom P. Fleming
- Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
- * E-mail:
| |
Collapse
|
37
|
González IM, Martin PM, Burdsal C, Sloan JL, Mager S, Harris T, Sutherland AE. Leucine and arginine regulate trophoblast motility through mTOR-dependent and independent pathways in the preimplantation mouse embryo. Dev Biol 2011; 361:286-300. [PMID: 22056783 DOI: 10.1016/j.ydbio.2011.10.021] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 10/18/2011] [Accepted: 10/19/2011] [Indexed: 01/06/2023]
Abstract
Uterine implantation is a critical element of mammalian reproduction and is a tightly and highly coordinated event. An intricate and reciprocal uterine-embryo dialog exists to synchronize uterine receptivity with the concomitant activation of the blastocyst, maximizing implantation success. While a number of pathways involved in regulating uterine receptivity have been identified in the mouse, less is understood about blastocyst activation, the process by which the trophectoderm (TE) receives extrinsic cues that initiate new characteristics essential for implantation. Amino acids (AA) have been found to regulate blastocyst activation and TE motility in vitro. In particular, we find that arginine and leucine alone are necessary and sufficient to induce TE motility. Both arginine and leucine act individually and additively to propagate signals that are dependent on the activity of the mammalian target of rapamycin complex 1 (mTORC1). The activities of the well-established downstream targets of mTORC1, p70S6K and 4EBP, do not correlate with trophoblast motility, suggesting that an independent-rapamycin-sensitive pathway operates to induce trophoblast motility, or that other, parallel amino acid-dependent pathways are also involved. We find that endogenous uterine factors act to induce mTORC1 activation and trophoblast motility at a specific time during pregnancy, and that this uterine signal is later than the previously defined signal that induces the attachment reaction. In vivo matured blastocysts exhibit competence to respond to an 8-hour AA stimulus by activating mTOR and subsequently undergoing trophoblast outgrowth by the morning of day 4.5 of pregnancy, but not on day 3.5. By the late afternoon of day 4.5, the embryos no longer require any exposure to AA to undergo trophoblast outgrowth in vitro, demonstrating the existence and timing of an equivalent in vivo signal. These results suggest that there are two separate uterine signals regulating implantation, one that primes the embryo for the attachment reaction and another that activates mTOR and initiates invasive behavior.
Collapse
Affiliation(s)
- Isabel M González
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA 22901, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Tomi M, Nishimura T, Nakashima E. Mother-to-fetus transfer of antiviral drugs and the involvement of transporters at the placental barrier. J Pharm Sci 2011; 100:3708-18. [DOI: 10.1002/jps.22642] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 04/21/2011] [Accepted: 05/10/2011] [Indexed: 12/15/2022]
|
39
|
Castro W, Bueno Sánchez JC, Piedrahita-Ochoa C, Cadavid J AP. Modulation of murine blastocyst hatching in vitro by glutamine and tryptophan. Braz J Med Biol Res 2011; 44:748-53. [PMID: 21710103 DOI: 10.1590/s0100-879x2011007500080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 06/10/2011] [Indexed: 11/22/2022] Open
Abstract
Enrichment of culture media with amino acids improves embryo development. However, little is known about the specific action of each amino acid during embryogenesis. The present study was undertaken to examine the effect of L-glutamine (Gln) and tryptophan (Trp) on mouse embryo hatching, expansion and viability in vitro. Blastocysts were collected from 6- to 8-week-old female BALB/c mice (N = 30) and cultured in M2 medium containing either 0.125, 0.25 or 0.5 mM Trp, 1 mM Gln, or M2 alone. Gln significantly increased (100%; P < 0.05) blastocyst hatching at 24 h compared to M2 alone or Trp; moreover, Trp inhibited blastocyst hatching when compared to M2 alone (P < 0.05) at 72 h. In contrast, the percentage of embryos reaching the state of expanded blastocyst at 48 h was significantly higher in medium with 1 mM Gln (66.6%; P < 0.05) or with 0.125 mM Trp (61.1%; P < 0.05). Unexpectedly, Trp increased the percentage of degenerated blastocysts after 48 h (67.7%; P < 0.05), while Gln preserved blastocyst viability. These results suggest that Gln may enhance blastocyst hatching, expansion and viability in vitro.
Collapse
Affiliation(s)
- W Castro
- Grupo Reproducción, Sede de Investigación Universitaria, Universidad de Antioquia, Medellín, Colombia
| | | | | | | |
Collapse
|
40
|
Tan BSN, Lonic A, Morris MB, Rathjen PD, Rathjen J. The amino acid transporter SNAT2 mediates l-proline-induced differentiation of ES cells. Am J Physiol Cell Physiol 2011; 300:C1270-9. [DOI: 10.1152/ajpcell.00235.2010] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
There is an increasing appreciation that amino acids can act as signaling molecules in the regulation of cellular processes through modulation of intracellular cell signaling pathways. In culture, embryonic stem (ES) cells can be differentiated to a second, pluripotent cell population, early primitive ectoderm-like cells in response to biological activities within the conditioned medium MEDII. The amino acid l-proline has been identified as a component of MEDII required for ES cell differentiation. Here, we define the primary l-proline transporter on ES and early primitive ectoderm-like cells as sodium-coupled neutral amino acid transporter 2 (SNAT2). SNAT2 uptake of l-proline can be inhibited by the addition of millimolar concentrations of other substrates. The addition of excess amino acids was used to regulate the uptake of l-proline by ES cells, and the effect on differentiation was analyzed. The ability of SNAT2 substrates, but not other amino acids, to prevent changes in morphology, gene expression, and differentiation kinetics suggested that l-proline uptake through SNAT2 was required for ES cell differentiation. These data reveal an unexpected role for amino acid uptake and the amino acid transporter SNAT2 in regulation of pluripotent cells in culture and provides a number of specific, inexpensive, and nontoxic culture additives with the potential to improve the quality of ES cell culture.
Collapse
Affiliation(s)
| | - Ana Lonic
- School of Molecular and Biomedical Science, University of Adelaide, South Australia; and
- Australian Stem Cell Centre, Monash University, Clayton, Victoria, Australia
| | - Michael B. Morris
- School of Molecular and Biomedical Science, University of Adelaide, South Australia; and
- Australian Stem Cell Centre, Monash University, Clayton, Victoria, Australia
| | - Peter D. Rathjen
- Department of Zoology, University of Melbourne, Melbourne, Victoria
- School of Molecular and Biomedical Science, University of Adelaide, South Australia; and
- Australian Stem Cell Centre, Monash University, Clayton, Victoria, Australia
| | - Joy Rathjen
- Department of Zoology, University of Melbourne, Melbourne, Victoria
| |
Collapse
|
41
|
Abstract
Amino acids are essential building blocks of all mammalian cells. In addition to their role in protein synthesis, amino acids play an important role as energy fuels, precursors for a variety of metabolites and as signalling molecules. Disorders associated with the malfunction of amino acid transporters reflect the variety of roles that they fulfil in human physiology. Mutations of brain amino acid transporters affect neuronal excitability. Mutations of renal and intestinal amino acid transporters affect whole-body homoeostasis, resulting in malabsorption and renal problems. Amino acid transporters that are integral parts of metabolic pathways reduce the function of these pathways. Finally, amino acid uptake is essential for cell growth, thereby explaining their role in tumour progression. The present review summarizes the involvement of amino acid transporters in these roles as illustrated by diseases resulting from transporter malfunction.
Collapse
|
42
|
Lefèvre PLC, Palin MF, Chen G, Turecki G, Murphy BD. Polyamines are implicated in the emergence of the embryo from obligate diapause. Endocrinology 2011; 152:1627-39. [PMID: 21303959 DOI: 10.1210/en.2010-0955] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Embryonic diapause is a poorly understood phenomenon of reversible arrest of embryo development prior to implantation. In many carnivores, such as the mink (Neovison vison), obligate diapause characterizes each gestation. Embryo reactivation is controlled by the uterus by mechanisms that remain elusive. Because polyamines are essential regulators of cell proliferation and growth, it was hypothesized that they trigger embryo reactivation. To test this, mated mink females were treated with α-difluoromethylornithine, an inhibitor of ornithine decarboxylase 1, the rate-limiting enzyme in polyamine biosynthesis, or saline as a control during the first 5 d of reactivation. This treatment induced polyamine deprivation with the consequence of rearrest in embryo cell proliferation. A mink trophoblast cell line in vitro subjected to α-difluoromethylornithine treatment likewise displayed an arrest in cell proliferation, morphological changes, and intracellular translocation of ornithine decarboxylase 1 protein. The arrest in embryo development deferred implantation for a period consistent with the length of treatment. Successful implantation and parturition ensued. We conclude that polyamine deprivation brought about a reversible rearrest of embryo development, which returned the mink embryo to diapause and induced a second delay in embryo implantation. The results are the first demonstration of a factor essential to reactivation of embryos in obligate diapause.
Collapse
Affiliation(s)
- Pavine L C Lefèvre
- Centre de Recherche en Reproduction Animale, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Rue Sicotte, St. Hyacinthe, Québec, Canada J2S 7C6.
| | | | | | | | | |
Collapse
|
43
|
Eckert JJ, Fleming TP. The effect of nutrition and environment on the preimplantation embryo. ACTA ACUST UNITED AC 2011. [DOI: 10.1576/toag.13.1.43.27640] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
44
|
Pelland AMD, Corbett HE, Baltz JM. Amino Acid transport mechanisms in mouse oocytes during growth and meiotic maturation. Biol Reprod 2009; 81:1041-54. [PMID: 19605782 DOI: 10.1095/biolreprod.109.079046] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Amino acids are transported into cells by a number of different transport systems, each with their own specific range of substrates. The amino acid transport systems active in preimplantation embryos and the amino acids required by embryos for optimal development have been extensively investigated. Much less is known about amino acid transport systems active in growing and meiotically maturing oocytes or about developmental changes in their activity. As a first step in determining the array of amino acid transporters active in oocytes, the transport characteristics of nine amino acids were measured in small, medium, and large growing oocytes; in fully grown germinal vesicle (GV)-stage oocytes; in metaphase I oocytes; and in metaphase II eggs. Whether each of 11 classically defined amino acid transport systems was likely active in oocytes at each stage was determined using assays based on measuring the transport of radiolabeled amino acids into oocytes and the effect of a limited set of potential competitive inhibitors. Six amino acid transport systems were found to be active during oocyte growth or maturation. L, b(0,+), and ASC/asc were active throughout oocyte growth and maturation, increasing during growth. In contrast, GLY, beta, and x(c)(-) had little or no activity during growth but became activated during meiotic maturation. Surprisingly, the presence of follicular cells surrounding medium growing oocytes or cumulus cells surrounding GV oocytes did not confer amino acid transport by additional transport systems not present in the oocyte. In some cases, however, follicular cells coupled to the oocyte enhanced uptake of amino acids by the same systems present in the oocyte.
Collapse
|
45
|
Gao H, Wu G, Spencer TE, Johnson GA, Bazer FW. Select Nutrients in the Ovine Uterine Lumen. IV. Expression of Neutral and Acidic Amino Acid Transporters in Ovine Uteri and Peri-Implantation Conceptuses1. Biol Reprod 2009; 80:1196-208. [DOI: 10.1095/biolreprod.108.075440] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
46
|
Sturmey RG, Brison DR, Leese HJ. Symposium: innovative techniques in human embryo viability assessment. Assessing embryo viability by measurement of amino acid turnover. Reprod Biomed Online 2009; 17:486-96. [PMID: 18854101 DOI: 10.1016/s1472-6483(10)60234-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This review assesses the ability of non-invasive 'amino acid profiling' to predict early embryo viability. The history of amino acid supplementation of embryo culture media and the role of amino acids in early embryo development are first considered and these are followed by a review of methods to quantify amino acid depletion and production by single embryos. Data on amino acid profiling of embryos from a number of species are then discussed. It is concluded that this technology has excellent potential to improve the selection of single embryos for transfer in clinical IVF.
Collapse
|
47
|
Bröer S. Amino acid transport across mammalian intestinal and renal epithelia. Physiol Rev 2008; 88:249-86. [PMID: 18195088 DOI: 10.1152/physrev.00018.2006] [Citation(s) in RCA: 647] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The transport of amino acids in kidney and intestine is critical for the supply of amino acids to all tissues and the homeostasis of plasma amino acid levels. This is illustrated by a number of inherited disorders affecting amino acid transport in epithelial cells, such as cystinuria, lysinuric protein intolerance, Hartnup disorder, iminoglycinuria, dicarboxylic aminoaciduria, and some other less well-described disturbances of amino acid transport. The identification of most epithelial amino acid transporters over the past 15 years allows the definition of these disorders at the molecular level and provides a clear picture of the functional cooperation between transporters in the apical and basolateral membranes of mammalian epithelial cells. Transport of amino acids across the apical membrane not only makes use of sodium-dependent symporters, but also uses the proton-motive force and the gradient of other amino acids to efficiently absorb amino acids from the lumen. In the basolateral membrane, antiporters cooperate with facilitators to release amino acids without depleting cells of valuable nutrients. With very few exceptions, individual amino acids are transported by more than one transporter, providing backup capacity for absorption in the case of mutational inactivation of a transport system.
Collapse
Affiliation(s)
- Stefan Bröer
- School of Biochemistry and Molecular Biology, Australian National University, Canberra, Australian Capital Territory, Australia.
| |
Collapse
|
48
|
Grillo MA, Lanza A, Colombatto S. Transport of amino acids through the placenta and their role. Amino Acids 2008; 34:517-23. [PMID: 18172742 DOI: 10.1007/s00726-007-0006-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Accepted: 11/13/2007] [Indexed: 01/01/2023]
Abstract
Amino acids are transported across the human placenta mediated by transporter proteins that differ in structure, mechanism and substrate specificity. Some of them are Na+-dependent systems, whereas others are Na+-independent. Among these there are transporters composed of a heavy chain, a glycoprotein, and a light chain. Moreover, they can be differently distributed in the two membranes forming the syncytiotrophoblast. The transport mechanisms involved and their regulation are only partially known. In the placenta itself, part of the amino acids is metabolized to form other compounds important for the fetus. This occurs for instance for arginine, which gives rise to polyamines and to NO. Interconversion occurs among few other amino acids Transport is altered in pregnancy complications, such as restricted fetal growth.
Collapse
Affiliation(s)
- M A Grillo
- Dipartimento di Medicina e Oncologia Sperimentale, Sezione di Biochimica, Università di Torino, Via Michelangelo 27, 10126 Torino, Italy.
| | | | | |
Collapse
|
49
|
Eckert JJ, Houghton FD, Hawkhead JA, Balen AH, Leese HJ, Picton HM, Cameron IT, Fleming TP. Human embryos developing in vitro are susceptible to impaired epithelial junction biogenesis correlating with abnormal metabolic activity. Hum Reprod 2007; 22:2214-24. [PMID: 17623723 DOI: 10.1093/humrep/dem147] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Blastocyst biogenesis occurs over several cell cycles during the preimplantation period comprising the gradual expression and membrane assembly of junctional protein complexes which distinguish the outer epithelial trophectoderm (TE) cells from the inner cell mass (ICM). In the human, TE integrity and the formation of a junctional seal can often be impaired. Embryos likely to result in a successful pregnancy after transfer are mostly selected according to morphological criteria. Recent data suggest that non-invasive measurement of amino acid turnover may be useful to complement such morphological scores. Whether morphological and metabolic criteria can be linked to poor TE differentiation thereby underpinning developmental predictions mechanistically remains unknown. METHODS We examined TE intercellular junction formation in human embryos by immunofluorescence and confocal microscopy and correlated this process with morphological criteria and amino acid turnover during late cleavage. RESULTS Our results show that TE differentiation may be compromised by failure of membrane assembly of specific junction constituents. This abnormality relates more closely to metabolic profiles than morphological criteria. CONCLUSION Our data identify that amino acid turnover can predict TE differentiation. These findings are the first to link two mechanisms, metabolism and junction membrane assembly, which contribute to early embryo development.
Collapse
Affiliation(s)
- Judith J Eckert
- School of Biological Sciences, University of Southampton, Bassett Crescent East, Southampton, UK.
| | | | | | | | | | | | | | | |
Collapse
|