1
|
Stapornwongkul KS, Hahn E, Poliński P, Salamó Palau L, Arató K, Yao L, Williamson K, Gritti N, Anlas K, Osuna Lopez M, Patil KR, Heemskerk I, Ebisuya M, Trivedi V. Glycolytic activity instructs germ layer proportions through regulation of Nodal and Wnt signaling. Cell Stem Cell 2025; 32:744-758.e7. [PMID: 40245870 PMCID: PMC12048219 DOI: 10.1016/j.stem.2025.03.011] [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: 12/20/2023] [Revised: 10/29/2024] [Accepted: 03/20/2025] [Indexed: 04/19/2025]
Abstract
Metabolic pathways can influence cell fate decisions, yet their regulative role during embryonic development remains poorly understood. Here, we demonstrate an instructive role of glycolytic activity in regulating signaling pathways involved in mesoderm and endoderm specification. Using a mouse embryonic stem cell (mESC)-based in vitro model for gastrulation, we found that glycolysis inhibition increases ectodermal cell fates at the expense of mesodermal and endodermal lineages. We demonstrate that this relationship is dose dependent, enabling metabolic control of germ layer proportions through exogenous glucose levels. We further show that glycolysis acts as an upstream regulator of Nodal and Wnt signaling and that its influence on cell fate specification can be decoupled from its effects on growth. Finally, we confirm the generality of our findings using a human gastrulation model. Our work underscores the dependence of signaling pathways on metabolic conditions and provides mechanistic insight into the nutritional regulation of cell fate decision-making.
Collapse
Affiliation(s)
- Kristina S Stapornwongkul
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain.
| | - Elisa Hahn
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | - Patryk Poliński
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | - Laura Salamó Palau
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | - Krisztina Arató
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | - LiAng Yao
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kate Williamson
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge CB2 1QR, UK
| | - Nicola Gritti
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | - Kerim Anlas
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | | | - Kiran R Patil
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge CB2 1QR, UK
| | - Idse Heemskerk
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Physics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Miki Ebisuya
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain; Cluster of Excellence Physics of Life, TU Dresden, 01307 Dresden, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany.
| | - Vikas Trivedi
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain; Developmental Biology, EMBL Heidelberg, Heidelberg 69117, Germany.
| |
Collapse
|
2
|
Ren H, Tang Y, Zhang D. The emerging role of protein L-lactylation in metabolic regulation and cell signalling. Nat Metab 2025; 7:647-664. [PMID: 40175761 DOI: 10.1038/s42255-025-01259-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 03/03/2025] [Indexed: 04/04/2025]
Abstract
L-Lactate has emerged as a crucial metabolic intermediate, moving beyond its traditional view as a mere waste product. The recent discovery of L-lactate-driven protein lactylation as a post-translational modification has unveiled a pathway that highlights the role of lactate in cellular signalling. In this Perspective, we explore the enzymatic and metabolic mechanisms underlying protein lactylation and its impacts on both histone and non-histone proteins in the contexts of physiology and diseases. We discuss growing evidence suggesting that this modification regulates a wide range of cellular functions and is involved in various physiological and pathological processes, such as cell-fate determination, development, cardiovascular diseases, cancer and autoimmune disorders. We propose that protein lactylation acts as a pivotal mechanism, integrating metabolic and signalling pathways to enable cellular adaptation, and highlight its potential as a therapeutic target in various diseases.
Collapse
Affiliation(s)
- Haowen Ren
- State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
| | - Yuwei Tang
- State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
- Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Di Zhang
- State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| |
Collapse
|
3
|
Ye Z, Sabatier P, van der Hoeven L, Lechner MY, Phlairaharn T, Guzman UH, Liu Z, Huang H, Huang M, Li X, Hartlmayr D, Izaguirre F, Seth A, Joshi HJ, Rodin S, Grinnemo KH, Hørning OB, Bekker-Jensen DB, Bache N, Olsen JV. Enhanced sensitivity and scalability with a Chip-Tip workflow enables deep single-cell proteomics. Nat Methods 2025; 22:499-509. [PMID: 39820750 PMCID: PMC11903336 DOI: 10.1038/s41592-024-02558-2] [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: 02/08/2024] [Accepted: 11/04/2024] [Indexed: 01/19/2025]
Abstract
Single-cell proteomics (SCP) promises to revolutionize biomedicine by providing an unparalleled view of the proteome in individual cells. Here, we present a high-sensitivity SCP workflow named Chip-Tip, identifying >5,000 proteins in individual HeLa cells. It also facilitated direct detection of post-translational modifications in single cells, making the need for specific post-translational modification-enrichment unnecessary. Our study demonstrates the feasibility of processing up to 120 label-free SCP samples per day. An optimized tissue dissociation buffer enabled effective single-cell disaggregation of drug-treated cancer cell spheroids, refining overall SCP analysis. Analyzing nondirected human-induced pluripotent stem cell differentiation, we consistently quantified stem cell markers OCT4 and SOX2 in human-induced pluripotent stem cells and lineage markers such as GATA4 (endoderm), HAND1 (mesoderm) and MAP2 (ectoderm) in different embryoid body cells. Our workflow sets a benchmark in SCP for sensitivity and throughput, with broad applications in basic biology and biomedicine for identification of cell type-specific markers and therapeutic targets.
Collapse
Affiliation(s)
- Zilu Ye
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark.
| | - Pierre Sabatier
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- Cardio-Thoracic Translational Medicine Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Leander van der Hoeven
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Maico Y Lechner
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Teeradon Phlairaharn
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Ulises H Guzman
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Zhen Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Haoran Huang
- Thermo Fisher Scientific (China) Co. Ltd, Shanghai, China
| | - Min Huang
- Thermo Fisher Scientific (China) Co. Ltd, Shanghai, China
| | - Xiangjun Li
- Thermo Fisher Scientific (China) Co. Ltd, Shanghai, China
| | | | | | | | - Hiren J Joshi
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Sergey Rodin
- Cardio-Thoracic Translational Medicine Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Karl-Henrik Grinnemo
- Cardio-Thoracic Translational Medicine Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | | | | | | | - Jesper V Olsen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
4
|
Kim KT, Kim SM, Cha HJ. Crosstalk between Signaling Pathways and Energy Metabolism in Pluripotency. Int J Stem Cells 2025; 18:12-20. [PMID: 38494425 PMCID: PMC11867904 DOI: 10.15283/ijsc23173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/08/2024] [Accepted: 02/14/2024] [Indexed: 03/19/2024] Open
Abstract
The sequential change from totipotency to multipotency occurs during early mammalian embryo development. However, due to the lack of cellular models to recapitulate the distinct potency of stem cells at each stage, their molecular and cellular characteristics remain ambiguous. The establishment of isogenic naïve and primed pluripotent stem cells to represent the pluripotency in the inner cell mass of the pre-implantation blastocyst and in the epiblast from the post-implantation embryo allows the understanding of the distinctive characteristics of two different states of pluripotent stem cells. This review discusses the prominent disparities between naïve and primed pluripotency, including signaling pathways, metabolism, and epigenetic status, ultimately facilitating a comprehensive understanding of their significance during early mammalian embryonic development.
Collapse
Affiliation(s)
- Keun-Tae Kim
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Seong-Min Kim
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Hyuk-Jin Cha
- College of Pharmacy, Seoul National University, Seoul, Korea
| |
Collapse
|
5
|
Finnerty RM, Carulli DJ, Hedge A, Wang Y, Boadu F, Winuthayanon S, Jack Cheng J, Winuthayanon W. Multi-omics analyses and machine learning prediction of oviductal responses in the presence of gametes and embryos. eLife 2025; 13:RP100705. [PMID: 40009070 PMCID: PMC11864756 DOI: 10.7554/elife.100705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025] Open
Abstract
The oviduct is the site of fertilization and preimplantation embryo development in mammals. Evidence suggests that gametes alter oviductal gene expression. To delineate the adaptive interactions between the oviduct and gamete/embryo, we performed a multi-omics characterization of oviductal tissues utilizing bulk RNA-sequencing (RNA-seq), single-cell RNA-sequencing (scRNA-seq), and proteomics collected from distal and proximal at various stages after mating in mice. We observed robust region-specific transcriptional signatures. Specifically, the presence of sperm induces genes involved in pro-inflammatory responses in the proximal region at 0.5 days post-coitus (dpc). Genes involved in inflammatory responses were produced specifically by secretory epithelial cells in the oviduct. At 1.5 and 2.5 dpc, genes involved in pyruvate and glycolysis were enriched in the proximal region, potentially providing metabolic support for developing embryos. Abundant proteins in the oviductal fluid were differentially observed between naturally fertilized and superovulated samples. RNA-seq data were used to identify transcription factors predicted to influence protein abundance in the proteomic data via a novel machine learning model based on transformers of integrating transcriptomics and proteomics data. The transformers identified influential transcription factors and correlated predictive protein expressions in alignment with the in vivo-derived data. Lastly, we found some differences between inflammatory responses in sperm-exposed mouse oviducts compared to hydrosalpinx Fallopian tubes from patients. In conclusion, our multi-omics characterization and subsequent in vivo confirmation of proteins/RNAs indicate that the oviduct is adaptive and responsive to the presence of sperm and embryos in a spatiotemporal manner.
Collapse
Affiliation(s)
- Ryan M Finnerty
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri-ColumbiaColumbiaUnited States
| | - Daniel J Carulli
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri-ColumbiaColumbiaUnited States
| | - Akshata Hedge
- Department of Electrical Engineering and Computer Science, College of Engineering, University of MissouriColumbiaUnited States
| | - Yanli Wang
- Department of Electrical Engineering and Computer Science, College of Engineering, University of MissouriColumbiaUnited States
| | - Frimpong Boadu
- Department of Electrical Engineering and Computer Science, College of Engineering, University of MissouriColumbiaUnited States
| | - Sarayut Winuthayanon
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri-ColumbiaColumbiaUnited States
| | - Jianlin Jack Cheng
- Department of Electrical Engineering and Computer Science, College of Engineering, University of MissouriColumbiaUnited States
| | - Wipawee Winuthayanon
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri-ColumbiaColumbiaUnited States
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri-ColumbiaColumbiaUnited States
| |
Collapse
|
6
|
Finnerty RM, Carulli DJ, Hegde A, Wang Y, Baodu F, Winuthayanon S, Cheng J, Winuthayanon W. Multi-omics analyses and machine learning prediction of oviductal responses in the presence of gametes and embryos. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.13.598905. [PMID: 38915688 PMCID: PMC11195261 DOI: 10.1101/2024.06.13.598905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
The oviduct is the site of fertilization and preimplantation embryo development in mammals. Evidence suggests that gametes alter oviductal gene expression. To delineate the adaptive interactions between the oviduct and gamete/embryo, we performed a multi-omics characterization of oviductal tissues utilizing bulk RNA-sequencing (RNA-seq), single-cell RNA-sequencing (scRNA-seq), and proteomics collected from distal and proximal at various stages after mating in mice. We observed robust region-specific transcriptional signatures. Specifically, the presence of sperm induces genes involved in pro-inflammatory responses in the proximal region at 0.5 days post-coitus (dpc). Genes involved in inflammatory responses were produced specifically by secretory epithelial cells in the oviduct. At 1.5 and 2.5 dpc, genes involved in pyruvate and glycolysis were enriched in the proximal region, potentially providing metabolic support for developing embryos. Abundant proteins in the oviductal fluid were differentially observed between naturally fertilized and superovulated samples. RNA-seq data were used to identify transcription factors predicted to influence protein abundance in the proteomic data via a novel machine learning model based on transformers of integrating transcriptomics and proteomics data. The transformers identified influential transcription factors and correlated predictive protein expressions in alignment with the in vivo-derived data. Lastly, we found some differences between inflammatory responses in sperm-exposed mouse oviducts compared to hydrosalpinx fallopian tubes from patients. In conclusion, our multi-omics characterization and subsequent in vivo confirmation of proteins/RNAs indicate that the oviduct is adaptive and responsive to the presence of sperm and embryos in a spatiotemporal manner.
Collapse
Affiliation(s)
- Ryan M. Finnerty
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri-Columbia, Columbia, Missouri, 65211 USA
| | - Daniel J. Carulli
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri-Columbia, Columbia, Missouri, 65211 USA
| | - Akshata Hegde
- Department of Electrical Engineering and Computer Science, College of Engineering
| | - Yanli Wang
- Department of Electrical Engineering and Computer Science, College of Engineering
| | - Frimpong Baodu
- Department of Electrical Engineering and Computer Science, College of Engineering
| | - Sarayut Winuthayanon
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri-Columbia, Columbia, Missouri, 65211 USA
| | - Jianlin Cheng
- Department of Electrical Engineering and Computer Science, College of Engineering
| | - Wipawee Winuthayanon
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri-Columbia, Columbia, Missouri, 65211 USA
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri-Columbia, Columbia, Missouri, 65211 USA
| |
Collapse
|
7
|
Al Balawi AN, Alblwi NAN, Soliman R, El-Far AH, Hassan MG, El-Sewedy T, Ameen F, Ismail NF, Elmetwalli A. Impact of Vitamin D deficiency on immunological and metabolic responses in women with recurrent pregnancy loss: focus on VDBP/HLA-G1/CTLA-4/ENTPD1/adenosine-fetal-maternal conflict crosstalk. BMC Pregnancy Childbirth 2024; 24:709. [PMID: 39472874 PMCID: PMC11523824 DOI: 10.1186/s12884-024-06914-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND AND AIM Recurrent pregnancy loss (RPL), also known as recurrent implantation failure (RIF), is a distressing condition affecting women characterized by two or more consecutive miscarriages or the inability to carry a pregnancy beyond 20 weeks. Immunological factors and genetic variations, particularly in Vit D Binding Protein (VDBP), have gained attention as potential contributors to RPL. This study aimed to provide insight into the immunological, genetic, and metabolic networks underlying RPL, placing a particular emphasis on the interactions between VDBP, HLA-G1, CTLA-4, ENTPD1, and adenosine-fetal-maternal conflict crosstalk. METHODS A retrospective study included 198 women with three or more consecutive spontaneous abortions. Exclusion criteria comprised uterine abnormalities, endocrine disorders, parental chromosomal abnormalities, infectious factors, autoimmune diseases, or connective tissue diseases. Immunological interplay was investigated in 162 female participants, divided into two groups based on their Vit D levels: normal Vit D-RPL and low Vit D-RPL. Various laboratory techniques were employed, including LC/MS/MS for Vit D measurement, ELISA for protein detection, flow cytometry for immune function analysis, and molecular docking for protein-ligand interaction assessment. RESULTS General characteristics between groups were significant regarding Vit D and glucose levels. Low Vit D levels were associated with decreased NK cell activity and downregulation of HLA-G1 and HLA-G5 proteins, while CTLA-4 revealed upregulation. VDBP was significantly downregulated in the low Vit D group. Our findings highlight the intricate relationship between Vit D status and adenosine metabolism by the downregulation of SGLT1, and NT5E, key components of adenosine metabolism, suggests that Vit D deficiency may disrupt the regulation of adenosine levels, leading to an impaired reproductive outcome. HNF1β, a negative regulator of VDBP, was upregulated, while HNF1α, a positive regulator, was downregulated in low Vit D women after RPL. Molecular docking analysis revealed crucial residues involved in the interaction between Vit D and HNF1β. CONCLUSION Collectively, these findings underscore the importance of Vit D in modulating immune function and molecular pathways relevant to pregnancy maintenance, highlighting the need for further research to elucidate the mechanisms and potential therapeutic interventions for improving pregnancy outcomes in individuals with Vit D deficiency and RPL.
Collapse
Affiliation(s)
- Aisha Nawaf Al Balawi
- Biology Department, University College of Haql, University of Tabuk, Tabuk, Saudi Arabia.
| | | | - Riham Soliman
- Tropical Medicine Department, Faculty of Medicine, Port Said University, Port Said, Egypt
- Egyptian Liver Research Institute and Hospital (ELRIAH), Sherbin, Mansoura, Egypt
| | - Ali H El-Far
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, China
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt
| | - Mervat G Hassan
- Department of Botany and Microbiology, Faculty of Science, Benha University, Benha, 13511, Egypt
| | - Tarek El-Sewedy
- Department of Applied Medical Chemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Fuad Ameen
- Department of Botany and Microbiology, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Nadia F Ismail
- Health Information Management Program, Biochemistry, Faculty of Health Science Technology, Borg El Arab Technological University, Alexandria, Egypt
| | - Alaa Elmetwalli
- Department of Clinical Trial Research Unit and Drug Discovery, Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt.
- Microbiology Division, Higher Technological Institute of Applied Health Sciences, Egypt Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt.
| |
Collapse
|
8
|
Sarnyai Z, Ben-Shachar D. Schizophrenia, a disease of impaired dynamic metabolic flexibility: A new mechanistic framework. Psychiatry Res 2024; 342:116220. [PMID: 39369460 DOI: 10.1016/j.psychres.2024.116220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/21/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Schizophrenia is a chronic, neurodevelopmental disorder with unknown aetiology and pathophysiology that emphasises the role of neurotransmitter imbalance and abnormalities in synaptic plasticity. The currently used pharmacological approach, the antipsychotic drugs, which have limited efficacy and an array of side-effects, have been developed based on the neurotransmitter hypothesis. Recent research has uncovered systemic and brain abnormalities in glucose and energy metabolism, focusing on altered glycolysis and mitochondrial oxidative phosphorylation. These findings call for a re-conceptualisation of schizophrenia pathophysiology as a progressing bioenergetics failure. In this review, we provide an overview of the fundamentals of brain bioenergetics and the changes identified in schizophrenia. We then propose a new explanatory framework positing that schizophrenia is a disease of impaired dynamic metabolic flexibility, which also reconciles findings of abnormal glucose and energy metabolism in the periphery and in the brain along the course of the disease. This evidence-based framework and testable hypothesis has the potential to transform the way we conceptualise this debilitating condition and to develop novel treatment approaches.
Collapse
Affiliation(s)
- Zoltán Sarnyai
- Laboratory of Psychobiology, Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Department of Psychiatry, Rambam Health Campus, Haifa, Israel; Laboratory of Psychiatric Neuroscience, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia.
| | - Dorit Ben-Shachar
- Laboratory of Psychobiology, Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Department of Psychiatry, Rambam Health Campus, Haifa, Israel.
| |
Collapse
|
9
|
Doria-Borrell P, Pérez-García V. Understanding the intersection between placental development and cancer: Lessons from the tumor suppressor BAP1. Commun Biol 2024; 7:1053. [PMID: 39191942 PMCID: PMC11349880 DOI: 10.1038/s42003-024-06689-2] [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: 04/12/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
The placenta, a pivotal organ in mammalian reproduction, allows nutrient exchange and hormonal signaling between the mother and the developing fetus. Understanding its molecular intricacies is essential for deciphering normal embryonic development and pathological conditions such as tumorigenesis. Here, we explore the multifaceted role of the tumor suppressor BRCA1-associated protein 1 (BAP1) in cancer and placentation. Initially recognized for its tumor-suppressive properties, BAP1 has emerged as a key regulator at the intersection of tumorigenesis and placental development. BAP1 influences crucial cellular processes such as cell death, proliferation, metabolism, and response to hypoxic conditions. By integrating insights from tumor and developmental biology, we illuminate the complex molecular pathways orchestrated by BAP1. This perspective highlights BAP1's significant impact on both cancer and placental development, and suggests novel therapeutic strategies that could improve outcomes for pregnancy disorders and cancer.
Collapse
Affiliation(s)
| | - Vicente Pérez-García
- Centro de Investigación Príncipe Felipe, Valencia, Spain.
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.
| |
Collapse
|
10
|
Correia D, Bellot M, Goyenechea J, Prats E, Moro H, Gómez-Canela C, Bedrossiantz J, Tagkalidou N, Ferreira CSS, Raldúa D, Domingues I, Faria M, Oliveira M. Parental exposure to antidepressants has lasting effects on offspring? A case study with zebrafish. CHEMOSPHERE 2024; 355:141851. [PMID: 38579950 DOI: 10.1016/j.chemosphere.2024.141851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/07/2024]
Abstract
Fish have common neurotransmitter pathways with humans, exhibiting a significant degree of conservation and homology. Thus, exposure to fluoxetine makes fish potentially susceptible to biochemical and physiological changes, similarly to what is observed in humans. Over the years, several studies demonstrated the potential effects of fluoxetine on different fish species and at different levels of biological organization. However, the effects of parental exposure to unexposed offspring remain largely unknown. The consequences of 15-day parental exposure to relevant concentrations of fluoxetine (100 and 1000 ng/L) were assessed on offspring using zebrafish as a model organism. Parental exposure resulted in offspring early hatching, non-inflation of the swimming bladder, increased malformation frequency, decreased heart rate and blood flow, and reduced growth. Additionally, a significant behavioral impairment was also found (reduced startle response, basal locomotor activity, and altered non-associative learning during early stages and a negative geotaxis and scototaxis, reduced thigmotaxis, and anti-social behavior at later life stages). These behavior alterations are consistent with decreased anxiety, a significant increase in the expression of the monoaminergic genes slc6a4a (sert), slc6a3 (dat), slc18a2 (vmat2), mao, tph1a, and th2, and altered levels of monoaminergic neurotransmitters. Alterations in behavior, expression of monoaminergic genes, and neurotransmitter levels persisted until offspring adulthood. Given the high conservation of neuronal pathways between fish and humans, data show the possibility of potential transgenerational and multigenerational effects of pharmaceuticals' exposure. These results reinforce the need for transgenerational and multigenerational studies in fish, under realistic scenarios, to provide realistic insights into the impact of these pharmaceuticals.
Collapse
Affiliation(s)
- Daniela Correia
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - Marina Bellot
- Department of Analytical Chemistry and Applied (Chromatography Section), School of Engineering, Institut Químic de Sarrià-Universitat Ramon Llull, Via Augusta 390, 08017, Barcelona, Spain.
| | - Júlia Goyenechea
- Department of Analytical Chemistry and Applied (Chromatography Section), School of Engineering, Institut Químic de Sarrià-Universitat Ramon Llull, Via Augusta 390, 08017, Barcelona, Spain.
| | - Eva Prats
- Center for Research and Development, Spanish National Research Council (CSIC), Spain.
| | - Hugo Moro
- Institute of Environmental Assessment and Water Research, Spanish National Research Council (CSIC), Spain.
| | - Cristian Gómez-Canela
- Department of Analytical Chemistry and Applied (Chromatography Section), School of Engineering, Institut Químic de Sarrià-Universitat Ramon Llull, Via Augusta 390, 08017, Barcelona, Spain.
| | - Juliette Bedrossiantz
- Institute of Environmental Assessment and Water Research, Spanish National Research Council (CSIC), Spain.
| | - Niki Tagkalidou
- Department of Biochemistry and Biotechnology, University of Thessaly, Greece.
| | - Carla S S Ferreira
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - Demetrio Raldúa
- Institute of Environmental Assessment and Water Research, Spanish National Research Council (CSIC), Spain.
| | - Inês Domingues
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - Melissa Faria
- Institute of Environmental Assessment and Water Research, Spanish National Research Council (CSIC), Spain.
| | - Miguel Oliveira
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| |
Collapse
|
11
|
Abstract
Sirtuins (SIRTs) are putative regulators of lifespan in model organisms. Since the initial discovery that SIRTs could promote longevity in nematodes and flies, the identification of additional properties of these proteins has led to understanding of their roles as exquisite sensors that link metabolic activity to oxidative states. SIRTs have major roles in biological processes that are important in kidney development and physiological functions, including mitochondrial metabolism, oxidative stress, autophagy, DNA repair and inflammation. Furthermore, altered SIRT activity has been implicated in the pathophysiology and progression of acute and chronic kidney diseases, including acute kidney injury, diabetic kidney disease, chronic kidney disease, polycystic kidney disease, autoimmune diseases and renal ageing. The renoprotective roles of SIRTs in these diseases make them attractive therapeutic targets. A number of SIRT-activating compounds have shown beneficial effects in kidney disease models; however, further research is needed to identify novel SIRT-targeting strategies with the potential to treat and/or prevent the progression of kidney diseases and increase the average human healthspan.
Collapse
Affiliation(s)
- Luca Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy.
| |
Collapse
|
12
|
Hussain T, Metwally E, Murtaza G, Kalhoro DH, Chughtai MI, Tan B, Omur AD, Tunio SA, Akbar MS, Kalhoro MS. Redox mechanisms of environmental toxicants on male reproductive function. Front Cell Dev Biol 2024; 12:1333845. [PMID: 38469179 PMCID: PMC10925774 DOI: 10.3389/fcell.2024.1333845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/25/2024] [Indexed: 03/13/2024] Open
Abstract
Humans and wildlife, including domesticated animals, are exposed to a myriad of environmental contaminants that are derived from various human activities, including agricultural, household, cosmetic, pharmaceutical, and industrial products. Excessive exposure to pesticides, heavy metals, and phthalates consequently causes the overproduction of reactive oxygen species. The equilibrium between reactive oxygen species and the antioxidant system is preserved to maintain cellular redox homeostasis. Mitochondria play a key role in cellular function and cell survival. Mitochondria are vulnerable to damage that can be provoked by environmental exposures. Once the mitochondrial metabolism is damaged, it interferes with energy metabolism and eventually causes the overproduction of free radicals. Furthermore, it also perceives inflammation signals to generate an inflammatory response, which is involved in pathophysiological mechanisms. A depleted antioxidant system provokes oxidative stress that triggers inflammation and regulates epigenetic function and apoptotic events. Apart from that, these chemicals influence steroidogenesis, deteriorate sperm quality, and damage male reproductive organs. It is strongly believed that redox signaling molecules are the key regulators that mediate reproductive toxicity. This review article aims to spotlight the redox toxicology of environmental chemicals on male reproduction function and its fertility prognosis. Furthermore, we shed light on the influence of redox signaling and metabolism in modulating the response of environmental toxins to reproductive function. Additionally, we emphasize the supporting evidence from diverse cellular and animal studies.
Collapse
Affiliation(s)
- Tarique Hussain
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
- Animal Science Division, Nuclear Institute for Agriculture and Biology College, Pakistan Institute of Engineering and Applied Sciences (NIAB-C, PIEAS), Faisalabad, Pakistan
| | - Elsayed Metwally
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Ghulam Murtaza
- Department of Livestock and Fisheries, Government of Sindh, Karachi, Pakistan
| | - Dildar Hussain Kalhoro
- Department of Veterinary Microbiology, Faculty of Animal Husbandry and Veterinary Sciences, Sindh Agriculture University, Tandojam, Sindh, Pakistan
| | - Muhammad Ismail Chughtai
- Animal Science Division, Nuclear Institute for Agriculture and Biology College, Pakistan Institute of Engineering and Applied Sciences (NIAB-C, PIEAS), Faisalabad, Pakistan
| | - Bie Tan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Ali Dogan Omur
- Department of Artificial Insemination, Faculty, Veterinary Medicine, Ataturk University, Erzurum, Türkiye
| | - Shakeel Ahmed Tunio
- Department of Livestock Management, Faculty of Animal Husbandry and Veterinary Sciences, Sindh Agriculture University, Tandojam, Sindh, Pakistan
| | - Muhammad Shahzad Akbar
- Faculty of Animal Husbandry and Veterinary Sciences, University of Poonch, Rawalakot, Pakistan
| | - Muhammad Saleem Kalhoro
- Department of Agro-Industrial, Food, and Environmental Technology, Faculty of Applied Science, Food and Agro-Industrial Research Centre, King Mongkut’s University of Technology North Bangkok, Bangkok, Thailand
| |
Collapse
|
13
|
Dingare C, Steventon B. Gastruloids - a minimalistic model to study complex developmental metabolism. Emerg Top Life Sci 2023; 7:455-464. [PMID: 38108463 PMCID: PMC10754324 DOI: 10.1042/etls20230082] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/19/2023]
Abstract
Metabolic networks are well placed to orchestrate the coordination of multiple cellular processes associated with embryonic development such as cell growth, proliferation, differentiation and cell movement. Here, we discuss the advantages that gastruloids, aggregates of mammalian embryonic stem cells that self-assemble a rudimentary body plan, have for uncovering the instructive role of metabolic pathways play in directing developmental processes. We emphasise the importance of using such reductionist systems to link specific pathways to defined events of early mammalian development and their utility for obtaining enough material for metabolomic studies. Finally, we review the ways in which the basic gastruloid protocol can be adapted to obtain specific models of embryonic cell types, tissues and regions. Together, we propose that gastruloids are an ideal system to rapidly uncover new mechanistic links between developmental signalling pathways and metabolic networks, which can then inform precise in vivo studies to confirm their function in the embryo.
Collapse
Affiliation(s)
- Chaitanya Dingare
- Department of Genetics, University of Cambridge, Downing Site, Cambridge CB2 3EH, U.K
| | - Ben Steventon
- Department of Genetics, University of Cambridge, Downing Site, Cambridge CB2 3EH, U.K
| |
Collapse
|
14
|
Shastak Y, Pelletier W. From Metabolism to Vitality: Uncovering Riboflavin's Importance in Poultry Nutrition. Animals (Basel) 2023; 13:3554. [PMID: 38003171 PMCID: PMC10668813 DOI: 10.3390/ani13223554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Riboflavin, or vitamin B2, is indispensable for poultry, profoundly impacting their metabolic equilibrium, growth, and overall health. In a climate of increasing demand for poultry products and heightened production intensity, grasping the multifaceted roles of riboflavin in domestic fowl nutrition becomes paramount. This essential vitamin serves as a precursor to two vital coenzymes, flavin mononucleotide and flavin adenine dinucleotide, integral players in pivotal redox reactions and energy metabolism. Inadequate riboflavin levels translate into stunted growth, skeletal deformities, and compromised feed conversion efficiency, thereby adversely affecting poultry performance and bottom-line profitability. Riboflavin goes beyond its fundamental role, ameliorating nutrient utilization, facilitating protein synthesis, and augmenting enzyme activity, rightfully earning its epithet as the "growth-promoting vitamin". Poultry's reproductive success intricately hinges on riboflavin levels, dictating egg production and hatchability. It is imperative to note that riboflavin requirements exhibit variations among poultry species and distinct production phases, emphasizing the importance of judicious and balanced supplementation strategies. Aligning dietary recommendations with genetic advancements holds the promise of fostering sustainable growth within the poultry sector. Exploring the multifaceted aspects of riboflavin empowers researchers, nutritionists, and producers to elevate poultry nutrition and overall well-being, harmonizing with the industry's evolving demands.
Collapse
Affiliation(s)
- Yauheni Shastak
- Nutrition & Health Division, BASF SE, 67063 Ludwigshafen am Rhein, Germany
| | | |
Collapse
|
15
|
Bobori SN, Zhu Y, Saarinen A, Liuzzo AJ, Folmes CDL. Metabolic Remodeling during Early Cardiac Lineage Specification of Pluripotent Stem Cells. Metabolites 2023; 13:1086. [PMID: 37887411 PMCID: PMC10608731 DOI: 10.3390/metabo13101086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/28/2023] Open
Abstract
Growing evidence indicates that metabolites and energy metabolism play an active rather than consequential role in regulating cellular fate. Cardiac development requires dramatic metabolic remodeling from relying primarily on glycolysis in pluripotent stem cells (PSCs) to oxidizing a wide array of energy substrates to match the high bioenergetic demands of continuous contraction in the developed heart. However, a detailed analysis of how remodeling of energy metabolism contributes to human cardiac development is lacking. Using dynamic multiple reaction monitoring metabolomics of central carbon metabolism, we evaluated temporal changes in energy metabolism during human PSC 3D cardiac lineage specification. Significant metabolic remodeling occurs during the complete differentiation, yet temporal analysis revealed that most changes occur during transitions from pluripotency to mesoderm (day 1) and mesoderm to early cardiac (day 5), with limited maturation of cardiac metabolism beyond day 5. Real-time metabolic analysis demonstrated that while hPSC cardiomyocytes (hPSC-CM) showed elevated rates of oxidative metabolism compared to PSCs, they still retained high glycolytic rates, confirming an immature metabolic phenotype. These observations support the opportunity to metabolically optimize the differentiation process to support lineage specification and maturation of hPSC-CMs.
Collapse
Affiliation(s)
| | | | | | | | - Clifford D. L. Folmes
- Departments of Biochemistry and Molecular Biology and Cardiovascular Medicine, Center for Regenerative Biotherapeutics, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA; (S.N.B.)
| |
Collapse
|
16
|
Abstract
Metabolic switches are a crucial hallmark of cellular development and regeneration. In response to changes in their environment or physiological state, cells undergo coordinated metabolic switching that is necessary to execute biosynthetic demands of growth and repair. In this Review, we discuss how metabolic switches represent an evolutionarily conserved mechanism that orchestrates tissue development and regeneration, allowing cells to adapt rapidly to changing conditions during development and postnatally. We further explore the dynamic interplay between metabolism and how it is not only an output, but also a driver of cellular functions, such as cell proliferation and maturation. Finally, we underscore the epigenetic and cellular mechanisms by which metabolic switches mediate biosynthetic needs during development and regeneration, and how understanding these mechanisms is important for advancing our knowledge of tissue development and devising new strategies to promote tissue regeneration.
Collapse
Affiliation(s)
- Ahmed I. Mahmoud
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
17
|
Olivar-Villanueva M, Ren M, Schlame M, Phoon CK. The critical role of cardiolipin in metazoan differentiation, development, and maturation. Dev Dyn 2023; 252:691-712. [PMID: 36692477 PMCID: PMC10238668 DOI: 10.1002/dvdy.567] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 12/27/2022] [Accepted: 01/13/2023] [Indexed: 01/25/2023] Open
Abstract
Cardiolipins are phospholipids that are central to proper mitochondrial functioning. Because mitochondria play crucial roles in differentiation, development, and maturation, we would also expect cardiolipin to play major roles in these processes. Indeed, cardiolipin has been implicated in the mechanism of three human diseases that affect young infants, implying developmental abnormalities. In this review, we will: (1) Review the biology of cardiolipin; (2) Outline the evidence for essential roles of cardiolipin during organismal development, including embryogenesis and cell maturation in vertebrate organisms; (3) Place the role(s) of cardiolipin during embryogenesis within the larger context of the roles of mitochondria in development; and (4) Suggest avenues for future research.
Collapse
Affiliation(s)
| | - Mindong Ren
- Department of Anesthesiology, New York University Grossman School of Medicine, New York, New York, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
| | - Michael Schlame
- Department of Anesthesiology, New York University Grossman School of Medicine, New York, New York, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
| | - Colin K.L. Phoon
- Department of Pediatrics, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
18
|
Gyllenhammer LE, Rasmussen JM, Bertele N, Halbing A, Entringer S, Wadhwa PD, Buss C. Maternal Inflammation During Pregnancy and Offspring Brain Development: The Role of Mitochondria. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:498-509. [PMID: 34800727 PMCID: PMC9086015 DOI: 10.1016/j.bpsc.2021.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/20/2021] [Accepted: 11/04/2021] [Indexed: 01/06/2023]
Abstract
The association between maternal immune activation (MIA) during pregnancy and risk for offspring neuropsychiatric disorders has been increasingly recognized over the past several years. Among the mechanistic pathways that have been described through which maternal inflammation during pregnancy may affect fetal brain development, the role of mitochondria has received little attention. In this review, the role of mitochondria as a potential mediator of the association between MIA during pregnancy and offspring brain development and risk for psychiatric disorders will be proposed. As a basis for this postulation, convergent evidence is presented supporting the obligatory role of mitochondria in brain development, the role of mitochondria as mediators and initiators of inflammatory processes, and evidence of mitochondrial dysfunction in preclinical MIA exposure models and human neurodevelopmental disorders. Elucidating the role of mitochondria as a potential mediator of MIA-induced alterations in brain development and neurodevelopmental disease risk may not only provide new insight into the pathophysiology of mental health disorders that have their origins in exposure to infection/immune activation during pregnancy but also offer new therapeutic targets.
Collapse
Affiliation(s)
- Lauren E Gyllenhammer
- Development, Health and Disease Research Program, University of California, Irvine, School of Medicine, Irvine, California; Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, California
| | - Jerod M Rasmussen
- Development, Health and Disease Research Program, University of California, Irvine, School of Medicine, Irvine, California; Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, California
| | - Nina Bertele
- Department of Medical Psychology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Amy Halbing
- Department of Medical Psychology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; Einstein Center for Neurosciences Berlin, Department of Medical Psychology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sonja Entringer
- Development, Health and Disease Research Program, University of California, Irvine, School of Medicine, Irvine, California; Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, California; Department of Medical Psychology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Pathik D Wadhwa
- Development, Health and Disease Research Program, University of California, Irvine, School of Medicine, Irvine, California; Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, California; Department of Psychiatry and Human Behavior, University of California, Irvine, School of Medicine, Irvine, California; Department of Obstetrics and Gynecology, University of California, Irvine, School of Medicine, Irvine, California; Department of Epidemiology, University of California, Irvine, School of Medicine, Irvine, California
| | - Claudia Buss
- Development, Health and Disease Research Program, University of California, Irvine, School of Medicine, Irvine, California; Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, California; Department of Medical Psychology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
19
|
Parween S, Alawathugoda TT, Prabakaran AD, Dheen ST, Morse RH, Emerald BS, Ansari SA. Nutrient sensitive protein O-GlcNAcylation modulates the transcriptome through epigenetic mechanisms during embryonic neurogenesis. Life Sci Alliance 2022; 5:5/8/e202201385. [PMID: 35470239 PMCID: PMC9039347 DOI: 10.26508/lsa.202201385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 01/02/2023] Open
Abstract
Protein O-GlcNAcylation is a dynamic, nutrient-sensitive mono-glycosylation deposited on numerous nucleo-cytoplasmic and mitochondrial proteins, including transcription factors, epigenetic regulators, and histones. However, the role of protein O-GlcNAcylation on epigenome regulation in response to nutrient perturbations during development is not well understood. Herein we recapitulated early human embryonic neurogenesis in cell culture and found that pharmacological up-regulation of O-GlcNAc levels during human embryonic stem cells' neuronal differentiation leads to up-regulation of key neurogenic transcription factor genes. This transcriptional de-repression is associated with reduced H3K27me3 and increased H3K4me3 levels on the promoters of these genes, perturbing promoter bivalency possibly through increased EZH2-Thr311 phosphorylation. Elevated O-GlcNAc levels also lead to increased Pol II-Ser5 phosphorylation and affect H2BS112O-GlcNAc and H2BK120Ub1 on promoters. Using an in vivo rat model of maternal hyperglycemia, we show similarly elevated O-GlcNAc levels and epigenetic dysregulations in the developing embryo brains because of hyperglycemia, whereas pharmacological inhibition of O-GlcNAc transferase (OGT) restored these molecular changes. Together, our results demonstrate O-GlcNAc mediated sensitivity of chromatin to nutrient status, and indicate how metabolic perturbations could affect gene expression during neurodevelopment.
Collapse
Affiliation(s)
- Shama Parween
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Thilina T Alawathugoda
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ashok D Prabakaran
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - S Thameem Dheen
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Randall H Morse
- New York State Department of Health, Wadsworth Center, Albany, NY, USA
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Suraiya A Ansari
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates .,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
20
|
Abstract
The study of developmental processes in Rhodnius prolixus has recently advanced with the sequencing of the genome. In this work, we analyze the maternal gene expression driving oogenesis and early embryogenesis in R. prolixus. We examined the transcriptional profile of mRNAs to establish the genes expressed across the ovary, unfertilized eggs and different embryonic stages of R. prolixus until the formation of the germ band anlage (0, 12, 24, and 48 h post egg laying). We identified 81 putative maternal and ovary-related genes and validated their expression by qRT-PCR. We validate the function of the ortholog gene Bicaudal-D (Rp-BicD) by in situ hybridization and parental RNAi. Consistent with a role in oogenesis and early development of R. prolixus, we show that lack of Rp-BicD does not significantly affect oogenesis but impairs the formation of the blastoderm. Based on our findings, we propose three times of action for maternal genes during oogenesis and embryogenesis in R. prolixus.
Collapse
|
21
|
Kanwore K, Kanwore K, Adzika GK, Abiola AA, Guo X, Kambey PA, Xia Y, Gao D. Cancer Metabolism: The Role of Immune Cells Epigenetic Alteration in Tumorigenesis, Progression, and Metastasis of Glioma. Front Immunol 2022; 13:831636. [PMID: 35392088 PMCID: PMC8980436 DOI: 10.3389/fimmu.2022.831636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/28/2022] [Indexed: 12/17/2022] Open
Abstract
Glioma is a type of brain and spinal cord tumor that begins in glial cells that support the nervous system neurons functions. Age, radiation exposure, and family background of glioma constitute are risk factors of glioma initiation. Gliomas are categorized on a scale of four grades according to their growth rate. Grades one and two grow slowly, while grades three and four grow faster. Glioblastoma is a grade four gliomas and the deadliest due to its aggressive nature (accelerated proliferation, invasion, and migration). As such, multiple therapeutic approaches are required to improve treatment outcomes. Recently, studies have implicated the significant roles of immune cells in tumorigenesis and the progression of glioma. The energy demands of gliomas alter their microenvironment quality, thereby inducing heterogeneity and plasticity change of stromal and immune cells via the PI3K/AKT/mTOR pathway, which ultimately results in epigenetic modifications that facilitates tumor growth. PI3K is utilized by many intracellular signaling pathways ensuring the proper functioning of the cell. The activation of PI3K/AKT/mTOR regulates the plasma membrane activities, contributing to the phosphorylation reaction necessary for transcription factors activities and oncogenes hyperactivation. The pleiotropic nature of PI3K/AKT/mTOR makes its activity unpredictable during altered cellular functions. Modification of cancer cell microenvironment affects many cell types, including immune cells that are the frontline cells involved in inflammatory cascades caused by cancer cells via high cytokines synthesis. Typically, the evasion of immunosurveillance by gliomas and their resistance to treatment has been attributed to epigenetic reprogramming of immune cells in the tumor microenvironment, which results from cancer metabolism. Hence, it is speculative that impeding cancer metabolism and/or circumventing the epigenetic alteration of immune cell functions in the tumor microenvironment might enhance treatment outcomes. Herein, from an oncological and immunological perspective, this review discusses the underlying pathomechanism of cell-cell interactions enhancing glioma initiation and metabolism activation and tumor microenvironment changes that affect epigenetic modifications in immune cells. Finally, prospects for therapeutic intervention were highlighted.
Collapse
Affiliation(s)
- Kouminin Kanwore
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou, China.,Xuzhou Key Laboratory of Neurobiology, Department of Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Konimpo Kanwore
- Faculty Mixed of Medicine and Pharmacy, Lomé-Togo, University of Lomé, Lomé, Togo
| | | | - Ayanlaja Abdulrahman Abiola
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou, China.,Xuzhou Key Laboratory of Neurobiology, Department of Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Xiaoxiao Guo
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou, China.,Xuzhou Key Laboratory of Neurobiology, Department of Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Piniel Alphayo Kambey
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou, China.,Xuzhou Key Laboratory of Neurobiology, Department of Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Ying Xia
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou, China.,Xuzhou Key Laboratory of Neurobiology, Department of Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Dianshuai Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou, China.,Xuzhou Key Laboratory of Neurobiology, Department of Anatomy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
22
|
Grinshpan N, Abayed FA, Wahl M, Ner-Gaon H, Manor R, Sagi A, Shay T. The transcriptional landscape of the giant freshwater prawn: Embryonic development and early sexual differentiation mechanisms. Front Endocrinol (Lausanne) 2022; 13:1059936. [PMID: 36568080 PMCID: PMC9767951 DOI: 10.3389/fendo.2022.1059936] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
The giant freshwater prawn pjMacrobrachium rosenbergii is one of the best studied species in aquaculture. However, the transcriptional changes associated with embryonic development and the sexual differentiation mechanism of M. rosenbergii remain to be elucidated. To characterize the embryonic development of this prawn and to determine whether differential expression and differential splicing play roles in the early sexual differentiation of M. rosenbergii, we profiled five developmental days of male and female embryos by RNA sequencing. We identified modules of co-expressed genes representing waves of transcription that correspond to physiological processes in early embryonic development (such as the maternal-to-zygotic transition) up to preparation for life outside the egg (development of muscles, cuticle etc.). Additionally, we found that hundreds of genes are differentially expressed between sexes, most of them uncharacterized, suggesting that the sex differentiation mechanism of M. rosenbergii might contain clade-specific elements. The resulting first-of-a-kind transcriptional map of embryonic development of male and female M. rosenbergii will guide future studies to reveal the roles of specific genes and splicing isoforms in the embryonic development and sexual differentiation process of M. rosenbergii.
Collapse
Affiliation(s)
- Nufar Grinshpan
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Faiza A.A. Abayed
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Melody Wahl
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hadas Ner-Gaon
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Rivka Manor
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Amir Sagi
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- *Correspondence: Amir Sagi, ; Tal Shay,
| | - Tal Shay
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- *Correspondence: Amir Sagi, ; Tal Shay,
| |
Collapse
|
23
|
Sabatier P, Beusch CM, Saei AA, Aoun M, Moruzzi N, Coelho A, Leijten N, Nordenskjöld M, Micke P, Maltseva D, Tonevitsky AG, Millischer V, Carlos Villaescusa J, Kadekar S, Gaetani M, Altynbekova K, Kel A, Berggren PO, Simonson O, Grinnemo KH, Holmdahl R, Rodin S, Zubarev RA. An integrative proteomics method identifies a regulator of translation during stem cell maintenance and differentiation. Nat Commun 2021; 12:6558. [PMID: 34772928 PMCID: PMC8590018 DOI: 10.1038/s41467-021-26879-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 10/25/2021] [Indexed: 12/21/2022] Open
Abstract
Detailed characterization of cell type transitions is essential for cell biology in general and particularly for the development of stem cell-based therapies in regenerative medicine. To systematically study such transitions, we introduce a method that simultaneously measures protein expression and thermal stability changes in cells and provide the web-based visualization tool ProteoTracker. We apply our method to study differences between human pluripotent stem cells and several cell types including their parental cell line and differentiated progeny. We detect alterations of protein properties in numerous cellular pathways and components including ribosome biogenesis and demonstrate that modulation of ribosome maturation through SBDS protein can be helpful for manipulating cell stemness in vitro. Using our integrative proteomics approach and the web-based tool, we uncover a molecular basis for the uncoupling of robust transcription from parsimonious translation in stem cells and propose a method for maintaining pluripotency in vitro.
Collapse
Affiliation(s)
- Pierre Sabatier
- Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, 17177, Sweden
| | - Christian M Beusch
- Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, 17177, Sweden
| | - Amir A Saei
- Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, 17177, Sweden
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Mike Aoun
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, 17177, Sweden
| | - Noah Moruzzi
- The Rolf Luft Research Center for Diabetes and Endocrinology, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, 17176, Sweden
| | - Ana Coelho
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, 17177, Sweden
| | - Niels Leijten
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, Utrecht, 3584 CH, The Netherlands
| | - Magnus Nordenskjöld
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, 171 76, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, 17177, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, 171 76, Sweden
| | - Patrick Micke
- Immunology, Genetics and Pathology, Rudbecklaboratoriet, Uppsala University, Uppsala, 751 85, Sweden
| | - Diana Maltseva
- Faculty of biology and biotechnology, National Research University Higher School of Economics, Myasnitskaya Street, 13/4, Moscow, 117997, Russia
| | - Alexander G Tonevitsky
- Faculty of biology and biotechnology, National Research University Higher School of Economics, Myasnitskaya Street, 13/4, Moscow, 117997, Russia
- Scientific Research Center Bioclinicum, Ugreshskaya str. 2/85, Moscow, 115088, Russia
| | - Vincent Millischer
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, 17177, Sweden
- Translational Psychiatry, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, 171 76, Sweden
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, 1090, Austria
| | - J Carlos Villaescusa
- Neurogenetic Unit, Department of Molecular Medicine and Surgery, Karolinska University Hospital, Stockholm, 171 76, Sweden
- Stem Cell R&D-TRU, Novo Nordisk A/S, Måløv, Denmark
| | - Sandeep Kadekar
- Department of Surgical Sciences, Uppsala University, Uppsala, 752 37, Sweden
| | - Massimiliano Gaetani
- Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, 17177, Sweden
- Chemical Proteomics Core Facility, Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 77, Sweden
- Chemical Proteomics, Science for Life Laboratory (SciLifeLab), Stockholm, 17 177, Sweden
| | | | - Alexander Kel
- geneXplain GmbH, Am Exer 19B, 38302, Wolfenbuettel, Germany
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, 17176, Sweden
| | - Oscar Simonson
- Department of Surgical Sciences, Uppsala University, Uppsala, 752 37, Sweden
- Department of Cardio-thoracic Surgery and Anesthesiology, Uppsala University Hospital, Uppsala, 751 85, Sweden
| | - Karl-Henrik Grinnemo
- Department of Surgical Sciences, Uppsala University, Uppsala, 752 37, Sweden
- Department of Cardio-thoracic Surgery and Anesthesiology, Uppsala University Hospital, Uppsala, 751 85, Sweden
| | - Rikard Holmdahl
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, 17177, Sweden
| | - Sergey Rodin
- Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, 17177, Sweden.
- Department of Surgical Sciences, Uppsala University, Uppsala, 752 37, Sweden.
- Department of Cardio-thoracic Surgery and Anesthesiology, Uppsala University Hospital, Uppsala, 751 85, Sweden.
| | - Roman A Zubarev
- Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, 17177, Sweden.
- Department of Pharmacological & Technological Chemistry, I.M. Sechenov First Moscow State Medical University, Moscow, 119146, Russia.
- The National Medical Research Center for Endocrinology, Moscow, 115478, Russia.
| |
Collapse
|
24
|
Sakae Y, Tanaka M. Metabolism and Sex Differentiation in Animals from a Starvation Perspective. Sex Dev 2021; 15:168-178. [PMID: 34284403 DOI: 10.1159/000515281] [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: 11/04/2020] [Accepted: 02/12/2021] [Indexed: 11/19/2022] Open
Abstract
Animals determine their sex genetically (GSD: genetic sex determination) and/or environmentally (ESD: environmental sex determination). Medaka (Oryzias latipes) employ a XX/XY GSD system, however, they display female-to-male sex reversal in response to various environmental changes such as temperature, hypoxia, and green light. Interestingly, we found that 5 days of starvation during sex differentiation caused female-to-male sex reversal. In this situation, the metabolism of pantothenate and fatty acid synthesis plays an important role in sex reversal. Metabolism is associated with other biological factors such as germ cells, HPG axis, lipids, and epigenetics, and supplys substances and acts as signal transducers. In this review, we discuss the importance of metabolism during sex differentiation and how metabolism contributes to sex differentiation.
Collapse
Affiliation(s)
- Yuta Sakae
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Minoru Tanaka
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| |
Collapse
|
25
|
Maraldi T, Angeloni C, Prata C, Hrelia S. NADPH Oxidases: Redox Regulators of Stem Cell Fate and Function. Antioxidants (Basel) 2021; 10:973. [PMID: 34204425 PMCID: PMC8234808 DOI: 10.3390/antiox10060973] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/15/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
One of the major sources of reactive oxygen species (ROS) generated within stem cells is the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family of enzymes (NOXs), which are critical determinants of the redox state beside antioxidant defense mechanisms. This balance is involved in another one that regulates stem cell fate: indeed, self-renewal, proliferation, and differentiation are decisive steps for stem cells during embryo development, adult tissue renovation, and cell therapy application. Ex vivo culture-expanded stem cells are being investigated for tissue repair and immune modulation, but events such as aging, senescence, and oxidative stress reduce their ex vivo proliferation, which is crucial for their clinical applications. Here, we review the role of NOX-derived ROS in stem cell biology and functions, focusing on positive and negative effects triggered by the activity of different NOX isoforms. We report recent findings on downstream molecular targets of NOX-ROS signaling that can modulate stem cell homeostasis and lineage commitment and discuss the implications in ex vivo expansion and in vivo engraftment, function, and longevity. This review highlights the role of NOX as a pivotal regulator of several stem cell populations, and we conclude that these aspects have important implications in the clinical utility of stem cells, but further studies on the effects of pharmacological modulation of NOX in human stem cells are imperative.
Collapse
Affiliation(s)
- Tullia Maraldi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy;
| | - Cristina Angeloni
- School of Pharmacy, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy;
| | - Cecilia Prata
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum—University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy;
| |
Collapse
|
26
|
Moraes JGN, Behura SK, Geary TW, Spencer TE. Analysis of the uterine lumen in fertility-classified heifers: I. Glucose, prostaglandins, and lipids†. Biol Reprod 2021; 102:456-474. [PMID: 31616913 DOI: 10.1093/biolre/ioz191] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/27/2019] [Accepted: 10/01/2019] [Indexed: 12/18/2022] Open
Abstract
Survival and growth of the bovine conceptus (embryo and associated extraembryonic membranes) are dependent on endometrial secretions or histotroph found in the uterine lumen. Previously, serial embryo transfer was used to classify heifers as high fertile (HF), subfertile (SF), or infertile (IF). Here, we investigated specific histotroph components [glucose, prostaglandins (PGs), and lipids] in the uterine lumen of day 17 pregnant and open fertility-classified heifers. Concentrations of glucose in the uterine lumen were increased by pregnancy but did not differ among fertility-classified heifers. Differences in expression of genes encoding glucose transporters and involved with glycolysis and gluconeogenesis were observed between conceptuses collected from HF and SF heifers. In the uterine lumen, PGE2 and PGF2α were increased by pregnancy, and HF heifers had higher concentrations of PGE2, PGF2α, and 6-keto-PFG1α than SF heifers. Differences were found in expression of genes regulating PG signaling, arachidonic acid metabolism, and peroxisome proliferator-activated receptor signaling among conceptuses and endometrium from fertility-classified heifers. Lipidomics was conducted exclusively in samples from HF heifers, and phosphatidylcholine was the main lipid class that increased in the uterine lumen by pregnancy. Expression of several lipid metabolism genes differed between HF and SF conceptuses, and a number of fatty acids were differentially abundant in the uterine lumen of pregnant HF and SF heifers. These results support the ideas that uterine luminal histotroph impacts conceptus survival and programs its development and is a facet of dysregulated conceptus-endometrial interactions that result in loss of the conceptus in SF cattle during the implantation period of pregnancy establishment.
Collapse
Affiliation(s)
- Joao G N Moraes
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| | - Susanta K Behura
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| | - Thomas W Geary
- USDA-ARS, Fort Keogh Livestock and Range Research Laboratory, Miles City, Montana, USA
| | - Thomas E Spencer
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
27
|
Piñeyro-Ruiz C, Serrano H, Jorge I, Miranda-Valentin E, Pérez-Brayfield MR, Camafeita E, Mesa R, Vázquez J, Jorge JC. A Proteomics Signature of Mild Hypospadias: A Pilot Study. Front Pediatr 2020; 8:586287. [PMID: 33425810 PMCID: PMC7786202 DOI: 10.3389/fped.2020.586287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/24/2020] [Indexed: 12/02/2022] Open
Abstract
Background and Objective: Mild hypospadias is a birth congenital condition characterized by the relocation of the male urethral meatus from its typical anatomical position near the tip of the glans penis, to a lower ventral position up to the brim of the glans corona, which can also be accompanied by foreskin ventral deficiency. For the most part, a limited number of cases have known etiology. We have followed a high-throughput proteomics approach to study the proteome in mild hypospadias patients. Methods: Foreskin samples from patients with mild hypospadias were collected during urethroplasty, while control samples were collected during elective circumcision (n = 5/group). A high-throughput, quantitative proteomics approach based on multiplexed peptide stable isotope labeling (SIL) and liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis was used to ascertain protein abundance changes in hypospadias patients when compared to control samples. Results: A total of 4,815 proteins were quantitated (2,522 with at least two unique peptides). One hundred and thirty-three proteins from patients with mild hypospadias showed significant abundance changes with respect to control samples, where 38 proteins were increased, and 95 proteins were decreased. Unbiased functional biological analysis revealed that both mitochondrial energy production and apoptotic signaling pathways were enriched in mild hypospadias. Conclusions: This first comprehensive proteomics characterization of mild hypospadias shows molecular changes associated with essential cellular processes related to energy production and apoptosis. Further evaluation of the proteome may expand the search of novel candidates in the etiology of mild hypospadias and could also lead to the identification of biomarkers for this congenital urogenital condition.
Collapse
Affiliation(s)
- Coriness Piñeyro-Ruiz
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, PR, United States
| | - Horacio Serrano
- Department of Internal Medicine, School of Medicine, University of Puerto Rico, San Juan, PR, United States
- Clinical Proteomics Laboratory, Internal Medicine Department, Comprehensive Cancer Center (CCC)-Medical Sciences Campus (MSC)-University of Puerto Rico (UPR), San Juan, PR, United States
- Department of Biochemistry, School of Medicine, University of Puerto Rico, San Juan, PR, United States
| | - Inmaculada Jorge
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Eric Miranda-Valentin
- Department of Internal Medicine, School of Medicine, University of Puerto Rico, San Juan, PR, United States
- Clinical Proteomics Laboratory, Internal Medicine Department, Comprehensive Cancer Center (CCC)-Medical Sciences Campus (MSC)-University of Puerto Rico (UPR), San Juan, PR, United States
| | - Marcos R. Pérez-Brayfield
- Department of Surgery, Urology Section, School of Medicine, University of Puerto Rico, San Juan, PR, United States
| | - Emilio Camafeita
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Raquel Mesa
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jesús Vázquez
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Juan Carlos Jorge
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, PR, United States
| |
Collapse
|
28
|
Gurner KH, Truong TT, Harvey AJ, Gardner DK. A combination of growth factors and cytokines alter preimplantation mouse embryo development, foetal development and gene expression profiles. Mol Hum Reprod 2020; 26:953-970. [DOI: 10.1093/molehr/gaaa072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/29/2020] [Indexed: 12/15/2022] Open
Abstract
Abstract
Within the maternal tract, the preimplantation embryo is exposed to an array of growth factors (GFs) and cytokines, most of which are absent from culture media used in clinical IVF. Whilst the addition of individual GFs and cytokines to embryo culture media can improve preimplantation mouse embryo development, there is a lack of evidence on the combined synergistic effects of GFs and cytokines on embryo development and further foetal growth. Therefore, in this study, the effect of a combined group of GFs and cytokines on mouse preimplantation embryo development and subsequent foetal development and gene expression profiles was investigated. Supplementation of embryo culture media with an optimised combination of GFs and cytokines (0.05 ng/ml vascular endothelial GF, 1 ng/ml platelet-derived GF, 0.13 ng/ml insulin-like GF 1, 0.026 ng/ml insulin-like GF 2 and 1 ng/ml granulocyte colony-stimulating factor) had no effect on embryo morphokinetics but significantly increased trophectoderm cell number (P = 0.0002) and total cell number (P = 0.024). Treatment with this combination of GFs and cytokines also significantly increased blastocyst outgrowth area (P < 0.05) and, following embryo transfer, increased foetal weight (P = 0.027), crown-rump length (P = 0.017) and overall morphological development (P = 0.027). RNA-seq analysis of in vitro derived foetuses identified concurrent alterations to the transcriptional profiles of liver and placental tissues compared with those developed in vivo, with greater changes observed in the GF and cytokine treated group. Together these data highlight the importance of balancing the actions of such factors for the regulation of normal development and emphasise the need for further studies investigating this prior to clinical implementation.
Collapse
Affiliation(s)
- Kathryn H Gurner
- School of BioSciences, University of Melbourne, Parkville, VIC 3010, Australia
| | - Thi T Truong
- School of BioSciences, University of Melbourne, Parkville, VIC 3010, Australia
| | - Alexandra J Harvey
- School of BioSciences, University of Melbourne, Parkville, VIC 3010, Australia
| | - David K Gardner
- School of BioSciences, University of Melbourne, Parkville, VIC 3010, Australia
- Melbourne IVF, East Melbourne, VIC 3002, Australia
| |
Collapse
|
29
|
Ly CH, Lynch GS, Ryall JG. A Metabolic Roadmap for Somatic Stem Cell Fate. Cell Metab 2020; 31:1052-1067. [PMID: 32433923 DOI: 10.1016/j.cmet.2020.04.022] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 02/13/2020] [Accepted: 04/29/2020] [Indexed: 01/14/2023]
Abstract
While metabolism was initially thought to play a passive role in cell biology by generating ATP to meet bioenergetic demands, recent studies have identified critical roles for metabolism in the generation of new biomass and provision of obligate substrates for the epigenetic modification of histones and DNA. This review details how metabolites generated through glycolysis and the tricarboxylic acid cycle are utilized by somatic stem cells to support cell proliferation and lineage commitment. Importantly, we also discuss the evolving hypothesis that histones can act as an energy reservoir during times of energy stress. Finally, we discuss how cells integrate both extrinsic metabolic cues and intrinsic metabolic machinery to regulate cell fate.
Collapse
Affiliation(s)
- C Hai Ly
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - James G Ryall
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC 3010, Australia.
| |
Collapse
|
30
|
Hansen JM, Jones DP, Harris C. The Redox Theory of Development. Antioxid Redox Signal 2020; 32:715-740. [PMID: 31891515 PMCID: PMC7047088 DOI: 10.1089/ars.2019.7976] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 12/30/2019] [Indexed: 12/16/2022]
Abstract
Significance: The geological record shows that as atmospheric O2 levels increased, it concomitantly coincided with the evolution of metazoans. More complex, higher organisms contain a more cysteine-rich proteome, potentially as a means to regulate homeostatic responses in a more O2-rich environment. Regulation of redox-sensitive processes to control development is likely to be evolutionarily conserved. Recent Advances: During early embryonic development, the conceptus is exposed to varying levels of O2. Oxygen and redox-sensitive elements can be regulated to promote normal development, defined as changes to cellular mass, morphology, biochemistry, and function, suggesting that O2 is a developmental morphogen. During periods of O2 fluctuation, embryos are "reprogrammed," on the genomic and metabolic levels. Reprogramming imparts changes to particular redox couples (nodes) that would support specific post-translational modifications (PTMs), targeting the cysteine proteome to regulate protein function and development. Critical Issues: Major developmental events such as stem cell expansion, proliferation, differentiation, migration, and cell fate decisions are controlled through oxidative PTMs of cysteine-based redox nodes. As such, timely coordinated redox regulation of these events yields normal developmental outcomes and viable species reproduction. Disruption of normal redox signaling can produce adverse developmental outcomes. Future Directions: Furthering our understanding of the redox-sensitive processes/pathways, the nature of the regulatory PTMs involved in development and periods of activation/sensitivity to specific developmental pathways would greatly support the theory of redox regulation of development, and would also provide rationale and direction to more fully comprehend poor developmental outcomes, such as dysmorphogenesis, functional deficits, and preterm embryonic death.
Collapse
Affiliation(s)
- Jason M. Hansen
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Dean P. Jones
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Craig Harris
- Toxicology Program, Department of Environmental Sciences, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
31
|
Ma H, Hayama T, Van Dyken C, Darby H, Koski A, Lee Y, Gutierrez NM, Yamada S, Li Y, Andrews M, Ahmed R, Liang D, Gonmanee T, Kang E, Nasser M, Kempton B, Brigande J, McGill TJ, Terzic A, Amato P, Mitalipov S. Deleterious mtDNA mutations are common in mature oocytes. Biol Reprod 2020; 102:607-619. [PMID: 31621839 PMCID: PMC7068114 DOI: 10.1093/biolre/ioz202] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/08/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022] Open
Abstract
Heritable mitochondrial DNA (mtDNA) mutations are common, yet only a few recurring pathogenic mtDNA variants account for the majority of known familial cases in humans. Purifying selection in the female germline is thought to be responsible for the elimination of most harmful mtDNA mutations during oogenesis. Here we show that deleterious mtDNA mutations are abundant in ovulated mature mouse oocytes and preimplantation embryos recovered from PolG mutator females but not in their live offspring. This implies that purifying selection acts not in the maternal germline per se, but during post-implantation development. We further show that oocyte mtDNA mutations can be captured and stably maintained in embryonic stem cells and then reintroduced into chimeras, thereby allowing examination of the effects of specific mutations on fetal and postnatal development.
Collapse
Affiliation(s)
- Hong Ma
- Center for Embryonic Cell and Gene Therapy, Oregon Health & Science University, 3303 S.W. Bond Avenue, Portland, Oregon 97239, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 N.W. 185th Avenue, Beaverton, Oregon 97006, USA
| | - Tomonari Hayama
- Center for Embryonic Cell and Gene Therapy, Oregon Health & Science University, 3303 S.W. Bond Avenue, Portland, Oregon 97239, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 N.W. 185th Avenue, Beaverton, Oregon 97006, USA
| | - Crystal Van Dyken
- Center for Embryonic Cell and Gene Therapy, Oregon Health & Science University, 3303 S.W. Bond Avenue, Portland, Oregon 97239, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 N.W. 185th Avenue, Beaverton, Oregon 97006, USA
| | - Hayley Darby
- Center for Embryonic Cell and Gene Therapy, Oregon Health & Science University, 3303 S.W. Bond Avenue, Portland, Oregon 97239, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 N.W. 185th Avenue, Beaverton, Oregon 97006, USA
| | - Amy Koski
- Center for Embryonic Cell and Gene Therapy, Oregon Health & Science University, 3303 S.W. Bond Avenue, Portland, Oregon 97239, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 N.W. 185th Avenue, Beaverton, Oregon 97006, USA
| | - Yeonmi Lee
- Stem Cell Center, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil Songpa-gu, Seoul 05505, Republic of Korea
| | - Nuria Marti Gutierrez
- Center for Embryonic Cell and Gene Therapy, Oregon Health & Science University, 3303 S.W. Bond Avenue, Portland, Oregon 97239, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 N.W. 185th Avenue, Beaverton, Oregon 97006, USA
| | - Satsuki Yamada
- Department of Cardiovascular Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Ying Li
- Center for Embryonic Cell and Gene Therapy, Oregon Health & Science University, 3303 S.W. Bond Avenue, Portland, Oregon 97239, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 N.W. 185th Avenue, Beaverton, Oregon 97006, USA
| | - Michael Andrews
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, 3375 S.W. Terwilliger Blvd, Portland, Oregon 97239, USA
| | - Riffat Ahmed
- Center for Embryonic Cell and Gene Therapy, Oregon Health & Science University, 3303 S.W. Bond Avenue, Portland, Oregon 97239, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 N.W. 185th Avenue, Beaverton, Oregon 97006, USA
| | - Dan Liang
- Center for Embryonic Cell and Gene Therapy, Oregon Health & Science University, 3303 S.W. Bond Avenue, Portland, Oregon 97239, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 N.W. 185th Avenue, Beaverton, Oregon 97006, USA
| | - Thanasup Gonmanee
- Center for Embryonic Cell and Gene Therapy, Oregon Health & Science University, 3303 S.W. Bond Avenue, Portland, Oregon 97239, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 N.W. 185th Avenue, Beaverton, Oregon 97006, USA
| | - Eunju Kang
- Stem Cell Center, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil Songpa-gu, Seoul 05505, Republic of Korea
| | - Mohammed Nasser
- Center for Embryonic Cell and Gene Therapy, Oregon Health & Science University, 3303 S.W. Bond Avenue, Portland, Oregon 97239, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 N.W. 185th Avenue, Beaverton, Oregon 97006, USA
| | - Beth Kempton
- Oregon Hearing Research Center, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, Oregon 97239, USA
| | - John Brigande
- Oregon Hearing Research Center, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, Oregon 97239, USA
| | - Trevor J McGill
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, 3375 S.W. Terwilliger Blvd, Portland, Oregon 97239, USA
| | - Andre Terzic
- Department of Cardiovascular Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Paula Amato
- Center for Embryonic Cell and Gene Therapy, Oregon Health & Science University, 3303 S.W. Bond Avenue, Portland, Oregon 97239, USA
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, Oregon 97239, USA
| | - Shoukhrat Mitalipov
- Center for Embryonic Cell and Gene Therapy, Oregon Health & Science University, 3303 S.W. Bond Avenue, Portland, Oregon 97239, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 N.W. 185th Avenue, Beaverton, Oregon 97006, USA
| |
Collapse
|
32
|
Tsogtbaatar E, Landin C, Minter-Dykhouse K, Folmes CDL. Energy Metabolism Regulates Stem Cell Pluripotency. Front Cell Dev Biol 2020; 8:87. [PMID: 32181250 PMCID: PMC7059177 DOI: 10.3389/fcell.2020.00087] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/31/2020] [Indexed: 12/19/2022] Open
Abstract
Pluripotent stem cells (PSCs) are characterized by their unique capacity for both unlimited self-renewal and their potential to differentiate to all cell lineages contained within the three primary germ layers. While once considered a distinct cellular state, it is becoming clear that pluripotency is in fact a continuum of cellular states, all capable of self-renewal and differentiation, yet with distinct metabolic, mitochondrial and epigenetic features dependent on gestational stage. In this review we focus on two of the most clearly defined states: “naïve” and “primed” PSCs. Like other rapidly dividing cells, PSCs have a high demand for anabolic precursors necessary to replicate their genome, cytoplasm and organelles, while concurrently consuming energy in the form of ATP. This requirement for both anabolic and catabolic processes sufficient to supply a highly adapted cell cycle in the context of reduced oxygen availability, distinguishes PSCs from their differentiated progeny. During early embryogenesis PSCs adapt their substrate preference to match the bioenergetic requirements of each specific developmental stage. This is reflected in different mitochondrial morphologies, membrane potentials, electron transport chain (ETC) compositions, and utilization of glycolysis. Additionally, metabolites produced in PSCs can directly influence epigenetic and transcriptional programs, which in turn can affect self-renewal characteristics. Thus, our understanding of the role of metabolism in PSC fate has expanded from anabolism and catabolism to include governance of the pluripotent epigenetic landscape. Understanding the roles of metabolism and the factors influencing metabolic pathways in naïve and primed pluripotent states provide a platform for understanding the drivers of cell fate during development. This review highlights the roles of the major metabolic pathways in the acquisition and maintenance of the different states of pluripotency.
Collapse
Affiliation(s)
- Enkhtuul Tsogtbaatar
- Stem Cell and Regenerative Metabolism Laboratory, Departments of Cardiovascular Diseases and Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, United States
| | - Chelsea Landin
- Stem Cell and Regenerative Metabolism Laboratory, Departments of Cardiovascular Diseases and Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, United States
| | - Katherine Minter-Dykhouse
- Stem Cell and Regenerative Metabolism Laboratory, Departments of Cardiovascular Diseases and Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, United States
| | - Clifford D L Folmes
- Stem Cell and Regenerative Metabolism Laboratory, Departments of Cardiovascular Diseases and Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, United States
| |
Collapse
|
33
|
Yang Q, Fu W, Wang Y, Miao K, Zhao H, Wang R, Guo M, Wang Z, Tian J, An L. The proteome of IVF-induced aberrant embryo-maternal crosstalk by implantation stage in ewes. J Anim Sci Biotechnol 2020; 11:7. [PMID: 31956410 PMCID: PMC6958772 DOI: 10.1186/s40104-019-0405-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/26/2019] [Indexed: 01/17/2023] Open
Abstract
Background Implantation failure limits the success of in vitro fertilization and embryo transfer (IVF-ET). Well-organized embryo-maternal crosstalk is essential for successful implantation. Previous studies mainly focused on the aberrant development of in vitro fertilized (IVF) embryos. In contrast, the mechanism of IVF-induced aberrant embryo-maternal crosstalk is not well defined. Results In the present study, using ewes as the model, we profiled the proteome that features aberrant IVF embryo-maternal crosstalk following IVF-ET. By comparing in vivo (IVO) and IVF conceptuses, as well as matched endometrial caruncular (C) and intercaruncular (IC) areas, we filtered out 207, 295, and 403 differentially expressed proteins (DEPs) in each comparison. Proteome functional analysis showed that the IVF conceptuses were characterized by the increased abundance of energy metabolism and proliferation-related proteins, and the decreased abundance of methyl metabolism-related proteins. In addition, IVF endometrial C areas showed the decreased abundance of endometrial remodeling and redox homeostasis-related proteins; while IC areas displayed the aberrant abundance of protein homeostasis and extracellular matrix (ECM) interaction-related proteins. Based on these observations, we propose a model depicting the disrupted embryo-maternal crosstalk following IVF-ET: Aberrant energy metabolism and redox homeostasis of IVF embryos, might lead to an aberrant endometrial response to conceptus-derived pregnancy signals, thus impairing maternal receptivity. In turn, the suboptimal uterine environment might stimulate a compensation effect of the IVF conceptuses, which was revealed as enhanced energy metabolism and over-proliferation. Conclusion Systematic proteomic profiling provides insights to understand the mechanisms that underlie the aberrant IVF embryo-maternal crosstalk. This might be helpful to develop practical strategies to prevent implantation failure following IVF-ET.
Collapse
Affiliation(s)
- Qianying Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Wei Fu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Yue Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Kai Miao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Haichao Zhao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Rui Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Min Guo
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Zhilong Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Jianhui Tian
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Lei An
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| |
Collapse
|
34
|
Richard A, Vallin E, Romestaing C, Roussel D, Gandrillon O, Gonin-Giraud S. Erythroid differentiation displays a peak of energy consumption concomitant with glycolytic metabolism rearrangements. PLoS One 2019; 14:e0221472. [PMID: 31483850 PMCID: PMC6726194 DOI: 10.1371/journal.pone.0221472] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 08/07/2019] [Indexed: 12/13/2022] Open
Abstract
Our previous single-cell based gene expression analysis pointed out significant variations of LDHA level during erythroid differentiation. Deeper investigations highlighted that a metabolic switch occurred along differentiation of erythroid cells. More precisely we showed that self-renewing progenitors relied mostly upon lactate-productive glycolysis, and required LDHA activity, whereas differentiating cells, mainly involved mitochondrial oxidative phosphorylation (OXPHOS). These metabolic rearrangements were coming along with a particular temporary event, occurring within the first 24h of erythroid differentiation. The activity of glycolytic metabolism and OXPHOS rose jointly with oxgene consumption dedicated to ATP production at 12-24h of the differentiation process before lactate-productive glycolysis sharply fall down and energy needs decline. Finally, we demonstrated that the metabolic switch mediated through LDHA drop and OXPHOS upkeep might be necessary for erythroid differentiation. We also discuss the possibility that metabolism, gene expression and epigenetics could act together in a circular manner as a driving force for differentiation.
Collapse
Affiliation(s)
- Angélique Richard
- Laboratoire de Biologie et Modélisation de la Cellule, Université de Lyon, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Elodie Vallin
- Laboratoire de Biologie et Modélisation de la Cellule, Université de Lyon, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Caroline Romestaing
- Laboratoire d’Ecologie des Hydrosystèmes Naturels et Anthropisés, Université de Lyon, Université Claude Bernard Lyon 1, ENTPE, Villeurbanne, France
| | - Damien Roussel
- Laboratoire d’Ecologie des Hydrosystèmes Naturels et Anthropisés, Université de Lyon, Université Claude Bernard Lyon 1, ENTPE, Villeurbanne, France
| | - Olivier Gandrillon
- Laboratoire de Biologie et Modélisation de la Cellule, Université de Lyon, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Inria Team Dracula, Inria Center Grenoble Rhône-Alpes, Grenoble, France
| | - Sandrine Gonin-Giraud
- Laboratoire de Biologie et Modélisation de la Cellule, Université de Lyon, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
35
|
Cabello-Rivera D, Sarmiento-Soto H, López-Barneo J, Muñoz-Cabello AM. Mitochondrial Complex I Function Is Essential for Neural Stem/Progenitor Cells Proliferation and Differentiation. Front Neurosci 2019; 13:664. [PMID: 31297047 PMCID: PMC6607990 DOI: 10.3389/fnins.2019.00664] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/11/2019] [Indexed: 12/20/2022] Open
Abstract
Neurogenesis in developing and adult mammalian brain is a tightly regulated process that relies on neural stem cell (NSC) activity. There is increasing evidence that mitochondrial metabolism affects NSC homeostasis and differentiation but the precise role of mitochondrial function in the neurogenic process requires further investigation. Here, we have analyzed how mitochondrial complex I (MCI) dysfunction affects NSC viability, proliferation and differentiation, as well as survival of the neural progeny. We have generated a conditional knockout model (hGFAP-NDUFS2 mice) in which expression of the NDUFS2 protein, essential for MCI function, is suppressed in cells expressing the Cre recombinase under the human glial fibrillary acidic protein promoter, active in mouse radial glial cells (RGCs) and in neural stem cells (NSCs) that reside in adult neurogenic niches. In this model we observed that survival of central NSC population does not appear to be severely affected by MCI dysfunction. However, perinatal brain development was markedly inhibited and Ndufs2 knockout mice died before the tenth postnatal day. In addition, in vitro studies of subventricular zone NSCs showed that active neural progenitors require a functional MCI to produce ATP and to proliferate. In vitro differentiation of neural precursors into neurons and oligodendrocytes was also profoundly affected. These data indicate the need of a correct MCI function and oxidative phosphorylation for glia-like NSC proliferation, differentiation and subsequent oligodendrocyte or neuronal maturation.
Collapse
Affiliation(s)
- Daniel Cabello-Rivera
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain.,Facultad de Medicina, Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Helia Sarmiento-Soto
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain.,Facultad de Medicina, Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain.,Facultad de Medicina, Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ana M Muñoz-Cabello
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain.,Facultad de Medicina, Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
36
|
Yeganeh B, Lee J, Ermini L, Lok I, Ackerley C, Post M. Autophagy is required for lung development and morphogenesis. J Clin Invest 2019; 129:2904-2919. [PMID: 31162135 DOI: 10.1172/jci127307] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 04/12/2019] [Indexed: 12/30/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) remains a major respiratory illness in extremely premature infants. The biological mechanisms leading to BPD are not fully understood, although an arrest in lung development has been implicated. The current study aimed to investigate the occurrence of autophagy in the developing mouse lung and its regulatory role in airway branching and terminal sacculi formation. We found 2 windows of epithelial autophagy activation in the developing mouse lung, both resulting from AMPK activation. Inhibition of AMPK-mediated autophagy led to reduced lung branching in vitro. Conditional deletion of beclin 1 (Becn1) in mouse lung epithelial cells (Becn1Epi-KO), either at early (E10.5) or late (E16.5) gestation, resulted in lethal respiratory distress at birth or shortly after. E10.5 Becn1Epi-KO lungs displayed reduced airway branching and sacculi formation accompanied by impaired vascularization, excessive epithelial cell death, reduced mesenchymal thinning of the interstitial walls, and delayed epithelial maturation. E16.5 Becn1Epi-KO lungs had reduced terminal air sac formation and vascularization and delayed distal epithelial differentiation, a pathology similar to that seen in infants with BPD. Taken together, our findings demonstrate that intrinsic autophagy is an important regulator of lung development and morphogenesis and may contribute to the BPD phenotype when impaired.
Collapse
Affiliation(s)
- Behzad Yeganeh
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children
| | - Joyce Lee
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children.,Institute of Medical Science, and
| | - Leonardo Ermini
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children
| | - Irene Lok
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children
| | - Cameron Ackerley
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children.,Departments of Physiology and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Martin Post
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children.,Institute of Medical Science, and.,Departments of Physiology and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
37
|
Henderson AJ, Finger BJ, Scott AW, Harvey AJ, Green MP. Acute in vitro exposure to environmentally relevant atrazine levels perturbs bovine preimplantation embryo metabolism and cell number. Reprod Toxicol 2019; 87:87-96. [PMID: 31129258 DOI: 10.1016/j.reprotox.2019.05.060] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/07/2019] [Accepted: 05/21/2019] [Indexed: 11/20/2022]
Abstract
Atrazine is a widely used herbicide known to negatively alter endocrine systems and perturb metabolism. Preimplantation exposure to pesticides may adversely affect long-term health, however few studies examine the effect of environmental levels and whether specific periods of development are particularly sensitive. In this study, the effect of acute, preimplantation atrazine exposure (days 3.5-7.5 post-fertilization) at levels detected and deemed safe in drinking water (0.02 and 20 μg/L respectively) on in vitro bovine embryo development, quality, metabolism, and gene expression was investigated. Atrazine exposure had no effect on development or quality, but significantly reduced blastocyst total cell numbers, attributable to a decrease in trophectoderm cells. Notably, atrazine (20 μg/L) markedly increased carbohydrate metabolism. Therefore, short-term exposure to environmentally relevant atrazine concentrations perturbs bovine preimplantation embryo metabolism and cell number, highlighting a potential mechanism by which atrazine can mediate embryo viability and health.
Collapse
Affiliation(s)
- Ashleigh J Henderson
- School of BioSciences, The University of Melbourne, Parkville, Melbourne, VIC, 3010, Australia
| | - Bethany J Finger
- School of BioSciences, The University of Melbourne, Parkville, Melbourne, VIC, 3010, Australia
| | - Alexander W Scott
- School of BioSciences, The University of Melbourne, Parkville, Melbourne, VIC, 3010, Australia
| | - Alexandra J Harvey
- School of BioSciences, The University of Melbourne, Parkville, Melbourne, VIC, 3010, Australia
| | - Mark P Green
- School of BioSciences, The University of Melbourne, Parkville, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
38
|
Abstract
Primordial germ cells (PGCs) must complete a complex and dynamic developmental program during embryogenesis to establish the germline. This process is highly conserved and involves a diverse array of tasks required of PGCs, including migration, survival, sex differentiation, and extensive epigenetic reprogramming. A common theme across many organisms is that PGC success is heterogeneous: only a portion of all PGCs complete all these steps while many other PGCs are eliminated from further germline contribution. The differences that distinguish successful PGCs as a population are not well understood. Here, we examine variation that exists in PGCs as they navigate the many stages of this developmental journey. We explore potential sources of PGC heterogeneity and their potential implications in affecting germ cell behaviors. Lastly, we discuss the potential for PGC development to function as a multistage selection process that assesses heterogeneity in PGCs to refine germline quality.
Collapse
Affiliation(s)
- Daniel H Nguyen
- Department of Obstetrics, Gynecology and Reproductive Science, Center for Reproductive Sciences, Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, United States
| | - Rebecca G Jaszczak
- Department of Obstetrics, Gynecology and Reproductive Science, Center for Reproductive Sciences, Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, United States
| | - Diana J Laird
- Department of Obstetrics, Gynecology and Reproductive Science, Center for Reproductive Sciences, Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, United States.
| |
Collapse
|
39
|
Lotti LV, Vespa S, Pantalone MR, Perconti S, Esposito DL, Visone R, Veronese A, Paties CT, Sanna M, Verginelli F, Nauclér CS, Mariani-Costantini R. A Developmental Perspective on Paragangliar Tumorigenesis. Cancers (Basel) 2019; 11:cancers11030273. [PMID: 30813557 PMCID: PMC6468609 DOI: 10.3390/cancers11030273] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 12/13/2022] Open
Abstract
In this review, we propose that paraganglioma is a fundamentally organized, albeit aberrant, tissue composed of neoplastic vascular and neural cell types that share a common origin from a multipotent mesenchymal-like stem/progenitor cell. This view is consistent with the pseudohypoxic footprint implicated in the molecular pathogenesis of the disease, is in harmony with the neural crest origin of the paraganglia, and is strongly supported by the physiological model of carotid body hyperplasia. Our immunomorphological and molecular studies of head and neck paragangliomas demonstrate in all cases relationships between the vascular and the neural tumor compartments, that share mesenchymal and immature vasculo-neural markers, conserved in derived cell cultures. This immature, multipotent phenotype is supported by constitutive amplification of NOTCH signaling genes and by loss of the microRNA-200s and -34s, which control NOTCH1, ZEB1, and PDGFRA in head and neck paraganglioma cells. Importantly, the neuroepithelial component is distinguished by extreme mitochondrial alterations, associated with collapse of the ΔΨm. Finally, our xenograft models of head and neck paraganglioma demonstrate that mesenchymal-like cells first give rise to a vasculo-angiogenic network, and then self-organize into neuroepithelial-like clusters, a process inhibited by treatment with imatinib.
Collapse
Affiliation(s)
- Lavinia Vittoria Lotti
- Department of Experimental Medicine, "La Sapienza" University, Viale Regina Elena 324, 00161 Rome, Italy.
| | - Simone Vespa
- Center of Sciences on Aging and Translational Medicine (CeSI-MeT), "G. d'Annunzio" University, Via Luigi Polacchi 11, 66100 Chieti, Italy.
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University, Via dei Vestini 31, 66100 Chieti, Italy.
| | - Mattia Russel Pantalone
- Department of Medicine (Solna), Division of Microbial Pathogenesis, BioClinicum, Karolinska Institutet, 17164 Stockholm, Sweden.
| | - Silvia Perconti
- Center of Sciences on Aging and Translational Medicine (CeSI-MeT), "G. d'Annunzio" University, Via Luigi Polacchi 11, 66100 Chieti, Italy.
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University, Via dei Vestini 31, 66100 Chieti, Italy.
| | - Diana Liberata Esposito
- Center of Sciences on Aging and Translational Medicine (CeSI-MeT), "G. d'Annunzio" University, Via Luigi Polacchi 11, 66100 Chieti, Italy.
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University, Via dei Vestini 31, 66100 Chieti, Italy.
| | - Rosa Visone
- Center of Sciences on Aging and Translational Medicine (CeSI-MeT), "G. d'Annunzio" University, Via Luigi Polacchi 11, 66100 Chieti, Italy.
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University, Via dei Vestini 31, 66100 Chieti, Italy.
| | - Angelo Veronese
- Department of Medicine and Aging Sciences, "G. d'Annunzio" University, Via Luigi Polacchi 11, 66100 Chieti, Italy.
| | - Carlo Terenzio Paties
- Department of Oncology-Hematology, Service of Anatomic Pathology, "Guglielmo da Saliceto" Hospital, Via Taverna 49, 29100 Piacenza, Italy.
| | - Mario Sanna
- Skull Base Unit, "Gruppo Otologico" Piacenza-Roma, Via Antonio Emmanueli, 42, 29121 Piacenza, Italy.
| | - Fabio Verginelli
- Department of Pharmacy, "G. d'Annunzio" University, Via dei Vestini 31, 66100 Chieti, Italy.
| | - Cecilia Soderberg Nauclér
- Department of Medicine (Solna), Division of Microbial Pathogenesis, BioClinicum, Karolinska Institutet, 17164 Stockholm, Sweden.
| | - Renato Mariani-Costantini
- Center of Sciences on Aging and Translational Medicine (CeSI-MeT), "G. d'Annunzio" University, Via Luigi Polacchi 11, 66100 Chieti, Italy.
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University, Via dei Vestini 31, 66100 Chieti, Italy.
| |
Collapse
|
40
|
Vlaski-Lafarge M, Loncaric D, Perez L, Labat V, Debeissat C, Brunet de la Grange P, Rossignol R, Ivanovic Z, Bœuf H. Bioenergetic Changes Underline Plasticity of Murine Embryonic Stem Cells. Stem Cells 2019; 37:463-475. [PMID: 30599083 DOI: 10.1002/stem.2965] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/13/2018] [Accepted: 12/03/2018] [Indexed: 12/19/2022]
Abstract
Murine embryonic stem cells (mESCs) are endowed by a time-dependent window of plasticity during their early commitment steps. Indeed, while mESCs deprived of leukemia inhibitory factor (LIF) for 24 hours revert to their naive pluripotent state after subsequent LIF readdition, cells deprived of LIF for 48 hours are no longer efficient in reverting, upon LIF addition, and undergo irreversible differentiation. We investigated undisclosed bioenergetic profiles of early mESC-derived committed cells versus their undifferentiated states in order to reveal specific bioenergetic changes associated with mESC plasticity. Multiparametric bioenergetic analysis revealed that pluripotent (+LIF) and reversibly committed cells (-LIF24h) are energetically flexible, depending on both oxidative phosphorylation (OXPHOS) and glycolysis. They exhibit high mitochondrial respiration in the presence of the main energetic substrates and can also rely on glycolysis in the presence of OXPHOS inhibitor. Inhibition of the glycolysis or mitochondrial respiration does not change drastically the expression of pluripotency genes, which remain well expressed. In addition, cells treated with these inhibitors keep their capacity to differentiate efficiently upon embryoid bodies formation. Transition from metabolically active mESCs to irreversibly committed cells is associated with a clear change in mitochondrial network morphology, to an increase of adenosine triphosphate (ATP) produced from glycolysis and a decline of ATP turnover and of the mitochondrial activity without change in the mitochondrial mass. Our study pointed that plasticity window of mESCs is associated with the bivalent energetic metabolism and potency to shift to glycolysis or OXPHOS on demand. LIF removal provokes glycolytic metabolic orientation and consecutive loss of the LIF-dependent reversion of cells to the pluripotent state. Stem Cells 2019;37:463-475.
Collapse
Affiliation(s)
- Marija Vlaski-Lafarge
- R&D Department, Etablissement Français du Sang Nouvelle-Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux
| | - Darija Loncaric
- R&D Department, Etablissement Français du Sang Nouvelle-Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux
| | - Laura Perez
- R&D Department, Etablissement Français du Sang Nouvelle-Aquitaine, Bordeaux, France
| | - Véronique Labat
- R&D Department, Etablissement Français du Sang Nouvelle-Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux
| | - Christelle Debeissat
- R&D Department, Etablissement Français du Sang Nouvelle-Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux
| | - Philippe Brunet de la Grange
- R&D Department, Etablissement Français du Sang Nouvelle-Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux
| | | | - Zoran Ivanovic
- R&D Department, Etablissement Français du Sang Nouvelle-Aquitaine, Bordeaux, France.,Inserm/U1035, University of Bordeaux
| | - Hélène Bœuf
- Inserm/U1026, University of Bordeaux, Bordeaux, France
| |
Collapse
|
41
|
Perestrelo T, Correia M, Ramalho-Santos J, Wirtz D. Metabolic and Mechanical Cues Regulating Pluripotent Stem Cell Fate. Trends Cell Biol 2018; 28:1014-1029. [DOI: 10.1016/j.tcb.2018.09.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/30/2018] [Accepted: 09/25/2018] [Indexed: 02/07/2023]
|
42
|
Cadete VJJ, Vasam G, Menzies KJ, Burelle Y. Mitochondrial quality control in the cardiac system: An integrative view. Biochim Biophys Acta Mol Basis Dis 2018; 1865:782-796. [PMID: 30472159 DOI: 10.1016/j.bbadis.2018.11.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 10/29/2018] [Accepted: 11/15/2018] [Indexed: 01/26/2023]
Abstract
Recent studies have led to the discovery of multiple mitochondrial quality control (mQC) processes that operate at various scales, ranging from the degradation of proteins by mitochondrial proteases to the degradation of selected cargos or entire organelles in lysosomes. While the mechanisms governing these mQC processes are progressively being delineated, their role and importance remain unclear. Converging evidence however point to a complex system whereby multiple and partly overlapping processes are recruited to orchestrate a cell type specific mQC response that is adapted to the physiological state and level of stress encountered. Knowledge gained from basic model systems of mQC therefore need to be integrated within organ-specific (patho)physiological frameworks. Building on this notion, this article focuses on mQC in the heart, where developmental metabolic reprogramming, sustained contraction, and multiple pathophysiological conditions pose broadly different constraints. We provide an overview of current knowledge of mQC processes, and discuss their implication in cardiac mQC under normal and diseased conditions.
Collapse
Affiliation(s)
- Virgilio J J Cadete
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Goutham Vasam
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Keir J Menzies
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa Brain and Mind Research Institute and Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON, Canada
| | - Yan Burelle
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
43
|
Gatie MI, Kelly GM. Metabolic profile and differentiation potential of extraembryonic endoderm-like cells. Cell Death Discov 2018; 4:42. [PMID: 30302276 PMCID: PMC6158286 DOI: 10.1038/s41420-018-0102-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 08/20/2018] [Accepted: 08/28/2018] [Indexed: 02/08/2023] Open
Abstract
Glucose metabolism has a crucial role for providing substrates required to generate ATP and regulate the epigenetic landscape. We reported that F9 embryonal carcinoma stem-like cells require cytosolic reactive oxygen species to differentiate into extraembryonic endoderm; however, mitochondrial sources were not examined. To extend these studies, we examined the metabolic profile of early and late-passage F9 cells, and show that their ability to differentiate is similar, even though each population has dramatically different metabolic profiles. Differentiated early-passage cells relied on glycolysis, while differentiated late-passage cells transitioned towards oxidative phosphorylation (OXPHOS). Unexpectedly, electron transport chain protein stoichiometry was disrupted in differentiated late-passage cells, whereas genes encoding mitofusion 1 and 2, which promote mitochondrial fusion and favor OXPHOS, were upregulated in differentiated early-passage cells. Despite this, early-passage cells cultured under conditions to promote glycolysis showed enhanced differentiation, whereas promoting OXPHOS in late-passage cells showed a similar trend. Further analysis revealed that the distinct metabolic profiles seen between the two populations is largely associated with changes in genomic integrity, linking metabolism to passage number. Together, these results indicate that passaging has no effect on the potential for F9 cells to differentiate into extraembryonic endoderm; however, it does impact their metabolic profile. Thus, it is imperative to determine the molecular and metabolic status of a stem cell population before considering its utility as a therapeutic tool for regenerative medicine.
Collapse
Affiliation(s)
- Mohamed I Gatie
- 1Department of Biology, Collaborative Graduate Specialization in Developmental Biology, The University of Western Ontario, London, ON Canada
| | - Gregory M Kelly
- 1Department of Biology, Collaborative Graduate Specialization in Developmental Biology, The University of Western Ontario, London, ON Canada.,2Department of Paediatrics, The University of Western Ontario, London, ON Canada.,3Department of Physiology and Pharmacology, The University of Western Ontario, London, ON Canada.,Child Health Research Institute, London, ON Canada.,5Ontario Institute for Regenerative Medicine, Toronto, ON Canada
| |
Collapse
|
44
|
Arrázola MS, Andraini T, Szelechowski M, Mouledous L, Arnauné-Pelloquin L, Davezac N, Belenguer P, Rampon C, Miquel MC. Mitochondria in Developmental and Adult Neurogenesis. Neurotox Res 2018; 36:257-267. [PMID: 30215161 DOI: 10.1007/s12640-018-9942-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 07/18/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022]
Abstract
Generation of new neurons is a tightly regulated process that involves several intrinsic and extrinsic factors. Among them, a metabolic switch from glycolysis to oxidative phosphorylation, together with mitochondrial remodeling, has emerged as crucial actors of neurogenesis. However, although accumulating data raise the importance of mitochondrial morphology and function in neural stem cell proliferation and differentiation during development, information regarding the contribution of mitochondria to adult neurogenesis processes remains limited. In the present review, we discuss recent evidence covering the importance of mitochondrial morphology, function, and energy metabolism in the regulation of neuronal development and adult neurogenesis, and their impact on memory processes.
Collapse
Affiliation(s)
- Macarena S Arrázola
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France. .,Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.
| | - Trinovita Andraini
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France.,Department of Physiology, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Marion Szelechowski
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Lionel Mouledous
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Laetitia Arnauné-Pelloquin
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Noélie Davezac
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Pascale Belenguer
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Claire Rampon
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Marie-Christine Miquel
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
45
|
Chen YM, Li H, Fan Y, Zhang QJ, Li X, Wu LJ, Chen ZJ, Zhu C, Qian LM. Identification of differentially expressed lncRNAs involved in transient regeneration of the neonatal C57BL/6J mouse heart by next-generation high-throughput RNA sequencing. Oncotarget 2018; 8:28052-28062. [PMID: 28427208 PMCID: PMC5438630 DOI: 10.18632/oncotarget.15887] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 02/20/2017] [Indexed: 02/01/2023] Open
Abstract
Previous studies have shown that mammalian cardiac tissue has a regenerative capacity. Remarkably, neonatal mice can regenerate their cardiac tissue for up to 6 days after birth, but this capacity is lost by day 7. In this study, we aimed to explore the expression pattern of long noncoding RNA (lncRNA) during this period and examine the mechanisms underlying this process. We found that 685 lncRNAs and 1833 mRNAs were differentially expressed at P1 and P7 by the next-generation high-throughput RNA sequencing. The coding genes associated with differentially expressed lncRNAs were mainly involved in metabolic processes and cell proliferation, and also were potentially associated with several key regeneration signalling pathways, including PI3K-Akt, MAPK, Hippo and Wnt. In addition, we identified some correlated targets of highly-dysregulated lncRNAs such as Igfbp3, Trnp1, Itgb6, and Pim3 by the coding-noncoding gene co-expression network. These data may offer a reference resource for further investigation about the mechanisms by which lncRNAs regulate cardiac regeneration.
Collapse
Affiliation(s)
- Yu-Mei Chen
- Department of Emergency, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Hua Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Yi Fan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Qi-Jun Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Xing Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Li-Jie Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Zi-Jie Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Chun Zhu
- Department of Pediatrics, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P. R. China
| | - Ling-Mei Qian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| |
Collapse
|
46
|
Hübner D, Jahn K, Pinkert S, Böhnke J, Jung M, Fechner H, Rujescu D, Liebert UG, Claus C. Infection of iPSC Lines with Miscarriage-Associated Coxsackievirus and Measles Virus and Teratogenic Rubella Virus as a Model for Viral Impairment of Early Human Embryogenesis. ACS Infect Dis 2017; 3:886-897. [PMID: 29043768 DOI: 10.1021/acsinfecdis.7b00103] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human induced pluripotent stem cell (iPSC) lines are a promising model for the early phase of human embryonic development. Here, their contribution to the still incompletely understood pathogenesis of congenital virus infections was evaluated. The infection of iPSC lines with miscarriage-associated coxsackievirus B3 (CVB3) and measles virus (MV) was compared to the efficient teratogen rubella virus (RV). While CVB3 and MV were found to be cytopathogenic on iPSC lines, RV replicated without impairment of iPSC colony morphology and integrity. This so far outstanding course of infection enabled maintenance of RV-infected iPSC cultures over several passages and their subsequent differentiation to ectoderm, endoderm, and mesoderm. A modification of the metabolic profile of infected iPSC lines was the only common aspect for all three viruses. This study points toward two important aspects. First, iPSC lines represent a suitable cell culture model for early embryonic virus infection. Second, metabolic activity represents an important means for evaluation of pathogen-associated alterations in iPSC lines.
Collapse
Affiliation(s)
- Denise Hübner
- Institute
of Virology, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany
| | - Kristin Jahn
- Institute
of Virology and Faculty of Life Sciences, University of Leipzig, Talstrasse 33, 04103 Leipzig, Germany
| | - Sandra Pinkert
- Department
of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Janik Böhnke
- Institute
of Virology and Faculty of Life Sciences, University of Leipzig, Talstrasse 33, 04103 Leipzig, Germany
| | - Matthias Jung
- Department
of Psychiatry, Psychotherapy, and Psychosomatics, Martin-Luther-University Halle, Julius-Kühn-Str. 7, 06112 Halle, Germany
| | - Henry Fechner
- Department
of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Dan Rujescu
- Department
of Psychiatry, Psychotherapy, and Psychosomatics, Martin-Luther-University Halle, Julius-Kühn-Str. 7, 06112 Halle, Germany
| | - Uwe Gerd Liebert
- Institute
of Virology, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany
| | - Claudia Claus
- Institute
of Virology, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany
| |
Collapse
|
47
|
Abstract
Metabolic pathways play a vital yet poorly understood role in embryogenesis. In this issue of Developmental Cell, Bulusu et al. (2017) and Oginuma et al. (2017) provide insights into the intricate relationship between metabolism and morphogenesis, showing that glycolysis facilitates body elongation and balances neural and mesodermal differentiation.
Collapse
Affiliation(s)
| | - Andrew C Oates
- The Francis Crick Institute, 1 Midland Road, NW1 1AT London, UK; Institute of Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| |
Collapse
|
48
|
Direct comparison of distinct naive pluripotent states in human embryonic stem cells. Nat Commun 2017; 8:15055. [PMID: 28429706 PMCID: PMC5413953 DOI: 10.1038/ncomms15055] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 02/23/2017] [Indexed: 11/17/2022] Open
Abstract
Until recently, human embryonic stem cells (hESCs) were shown to exist in a state of primed pluripotency, while mouse embryonic stem cells (mESCs) display a naive or primed pluripotent state. Here we show the rapid conversion of in-house-derived primed hESCs on mouse embryonic feeder layer (MEF) to a naive state within 5–6 days in naive conversion media (NCM-MEF), 6–10 days in naive human stem cell media (NHSM-MEF) and 14–20 days using the reverse-toggle protocol (RT-MEF). We further observe enhanced unbiased lineage-specific differentiation potential of naive hESCs converted in NCM-MEF, however, all naive hESCs fail to differentiate towards functional cell types. RNA-seq analysis reveals a divergent role of PI3K/AKT/mTORC signalling, specifically of the mTORC2 subunit, in the different naive hESCs. Overall, we demonstrate a direct evaluation of several naive culture conditions performed in the same laboratory, thereby contributing to an unbiased, more in-depth understanding of different naive hESCs. Human embryonic stem cells (hESCs) in culture display a state of primed pluripotency, but recent protocols have been developed that enable hESCs to adopt a naive-like pluripotent state. Here the authors perform a side-by-side comparison of methods used to culture naive hESCs and confirm the role of PI3K/AKT/mTORC signalling in facilitating the induction of naive pluripotency.
Collapse
|
49
|
Kelly GM, Gatie MI. Mechanisms Regulating Stemness and Differentiation in Embryonal Carcinoma Cells. Stem Cells Int 2017; 2017:3684178. [PMID: 28373885 PMCID: PMC5360977 DOI: 10.1155/2017/3684178] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 01/10/2017] [Accepted: 02/08/2017] [Indexed: 02/06/2023] Open
Abstract
Just over ten years have passed since the seminal Takahashi-Yamanaka paper, and while most attention nowadays is on induced, embryonic, and cancer stem cells, much of the pioneering work arose from studies with embryonal carcinoma cells (ECCs) derived from teratocarcinomas. This original work was broad in scope, but eventually led the way for us to focus on the components involved in the gene regulation of stemness and differentiation. As the name implies, ECCs are malignant in nature, yet maintain the ability to differentiate into the 3 germ layers and extraembryonic tissues, as well as behave normally when reintroduced into a healthy blastocyst. Retinoic acid signaling has been thoroughly interrogated in ECCs, especially in the F9 and P19 murine cell models, and while we have touched on this aspect, this review purposely highlights how some key transcription factors regulate pluripotency and cell stemness prior to this signaling. Another major focus is on the epigenetic regulation of ECCs and stem cells, and, towards that end, this review closes on what we see as a new frontier in combating aging and human disease, namely, how cellular metabolism shapes the epigenetic landscape and hence the pluripotency of all stem cells.
Collapse
Affiliation(s)
- Gregory M. Kelly
- Department of Biology, Molecular Genetics Unit, Western University, London, ON, Canada
- Collaborative Program in Developmental Biology, Western University, London, ON, Canada
- Department of Paediatrics and Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Child Health Research Institute, London, ON, Canada
- Ontario Institute for Regenerative Medicine, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | - Mohamed I. Gatie
- Department of Biology, Molecular Genetics Unit, Western University, London, ON, Canada
- Collaborative Program in Developmental Biology, Western University, London, ON, Canada
| |
Collapse
|
50
|
Li S, Winuthayanon W. Oviduct: roles in fertilization and early embryo development. J Endocrinol 2017; 232:R1-R26. [PMID: 27875265 DOI: 10.1530/joe-16-0302] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 10/11/2016] [Indexed: 12/12/2022]
Abstract
Animal oviducts and human Fallopian tubes are a part of the female reproductive tract that hosts fertilization and pre-implantation development of the embryo. With an increasing understanding of roles of the oviduct at the cellular and molecular levels, current research signifies the importance of the oviduct on naturally conceived fertilization and pre-implantation embryo development. This review highlights the physiological conditions within the oviduct during fertilization, environmental regulation, oviductal fluid composition and its role in protecting embryos and supplying nutrients. Finally, the review compares different aspects of naturally occurring fertilization and assisted reproductive technology (ART)-achieved fertilization and embryo development, giving insight into potential areas for improvement in this technology.
Collapse
Affiliation(s)
- Shuai Li
- School of Molecular BiosciencesCollege of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Wipawee Winuthayanon
- School of Molecular BiosciencesCollege of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| |
Collapse
|