1
|
Jones HN, Williams A, Wilson RL. A human cytotrophoblast-villous endothelium-fetal organ multi-cell model and the impact on gene and protein expression in placenta cytotrophoblast, fetal hepatocytes and fetal kidney epithelial cells. Placenta 2025; 168:35-45. [PMID: 40505174 DOI: 10.1016/j.placenta.2025.05.029] [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: 04/14/2025] [Revised: 05/18/2025] [Accepted: 05/31/2025] [Indexed: 06/18/2025]
Abstract
INTRODUCTION Appropriate fetal growth requires multi-directional, coordinated communication between maternal, placental, and fetal systems. Disruptions in these signaling arms can have deleterious consequences for fetal growth and lead to fetal developmental adaptations associated with short- and long-term morbidities. This proof-of-concept human cell model study aimed to identify the effects of altered trophoblast culture conditions and human insulin-like 1 growth factor (hIGF1) nanoparticle gene therapy on fetal liver hepatocytes and kidney epithelial cells. METHODS We utilized human cell lines: BeWo choriocarcinoma cells (trophoblast), Human Placental Micro-Vascular Endothelial Cells, and WRL68 (hepatocytes) or HEK293T/17 (kidney epithelium), in a co-culture model designed to mimic cytotrophoblast-villous endothelium-fetal organ communication. RESULTS Trophoblast stress response mechanisms were increased by culturing BeWo cells in growth media without fetal bovine serum (FBS). BeWo cells were also cultured without FBS and treated with a hIGF1 nanoparticle gene therapy which is known to mitigate cellular stress mechanisms. BeWo cells without FBS support had increased expression of cellular stress mechanisms but not when IGF1 was over-expressed with a transient hIGF1 nanoparticle gene therapy. BeWo cells without FBS and without FBS + hIGF1 nanoparticle gene therapy had increased expression of gluconeogenesis and glycolysis rate-limiting enzymes. Gene and protein expression in fetal liver and kidney cells was not impacted by increased trophoblast stress or hIGF1 nanoparticle gene therapy. DISCUSSION Our data demonstrated that cytotrophoblast, cultured without FBS support, turn on mechanisms involved in glucose production. Whether this is reflected in vivo remains uninvestigated but may represent a placental compensation mechanism in complicated pregnancies.
Collapse
Affiliation(s)
- Helen N Jones
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, FL, USA; Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Alyssa Williams
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, FL, USA; Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Rebecca L Wilson
- Center for Research in Perinatal Outcomes, College of Medicine, University of Florida, Gainesville, FL, USA; Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
2
|
Xun Y, Jiang Y, Khalid A, Tian Z, Rios J, Zhang Z. KBTBD2 controls bone development by regulating IGF-1 signaling during osteoblast differentiation. Cell Death Differ 2025; 32:1099-1111. [PMID: 39562829 PMCID: PMC12162822 DOI: 10.1038/s41418-024-01416-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024] Open
Abstract
Kelch repeat and BTB (POZ) domain-containing 2 (KBTBD2) is known for its pivotal role in metabolic regulation, particularly in adipocytes. However, its significance in skeletal development has remained elusive. Here, we uncover a previously unrecognized function of KBTBD2 in bone formation. Conditional knockout of Kbtbd2 in embryonic osteochondroprogenitor cells or osteoblasts results in impaired osteogenic differentiation, leading to reduced skeletal growth and mineralization. Mechanistically, the loss of KBTBD2 during osteogenesis leads to the accumulation of p85α, a regulatory subunit encoded by phosphoinositide-3-kinase regulatory subunit 1 (Pik3r1), which exerts a potent inhibitory effect on insulin-like growth factor 1 (IGF-1)-induced activation of AKT. Moreover, our study extends the understanding of KBTBD2's relevance beyond bone biology to the context of SHORT syndrome, a rare genetic disorder marked by short stature and various physical abnormalities. We demonstrate that p85α harboring the p.(Arg649Trp) mutation, most frequently found in SHORT syndrome patients, exhibits reduced binding to KBTBD2, leading to impaired IGF-1-mediated activation of AKT. These findings reveal that KBTBD2 is essential in bone formation via regulating the IGF-1 signaling pathway and suggest loss of KBTBD2-mediated regulation of p85α as a potential mechanism for SHORT syndrome.
Collapse
Affiliation(s)
- Yu Xun
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yiao Jiang
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Aysha Khalid
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Zeru Tian
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jonathan Rios
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhao Zhang
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
3
|
Zheng S, Chen X, Fang J, Li Y, Xiao X, Zhang X, Zhang L, Cheng Y, Hao L. The role of insulin-like growth factor-1 in lactation. Gene 2025; 962:149577. [PMID: 40404070 DOI: 10.1016/j.gene.2025.149577] [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: 01/03/2025] [Revised: 04/21/2025] [Accepted: 05/18/2025] [Indexed: 05/24/2025]
Abstract
Generally, Insulin-like growth factor 1 (IGF-1) is believed to regulate lactation activity by promoting cell proliferation and differentiation. With the advancement of research, IGF-1 has been discovered to play an important role in different stages of lactation. In actual animal production, lactation ability directly affects milk yield and milk quality, which not only affects the survival and future growth of pups, but also is an important economic trait of some animals. In this paper, it is introduced that IGF-1 plays an important role in the whole lactation process, and what factors are involved in the regulation of IGF-1 in this process and how to improve lactation ability through IGF-1 in animal production, providing a theoretical basis for further exploration of IGF-1 in lactation, and also brings a theoretical foundation for the improvement of animal lactation ability.
Collapse
Affiliation(s)
- Shuo Zheng
- College of Animal Science, Jilin University, Changchun 130062, China
| | - Xi Chen
- College of Animal Science, Jilin University, Changchun 130062, China
| | - JiaY Fang
- College of Animal Science, Jilin University, Changchun 130062, China
| | - Yi Li
- College of Animal Science, Jilin University, Changchun 130062, China
| | - XingY Xiao
- College of Animal Science, Jilin University, Changchun 130062, China
| | - XunM Zhang
- College of Animal Science, Jilin University, Changchun 130062, China
| | - LiB Zhang
- College of Animal Science, Jilin University, Changchun 130062, China
| | - YunY Cheng
- College of Public Health, Jilin University, Changchun 130061, China
| | - LinL Hao
- College of Animal Science, Jilin University, Changchun 130062, China.
| |
Collapse
|
4
|
Xu W, Fu H, Meng X, Sun Y, Ning F, Du Z. Cloning and Spatiotemporal Expression Analysis of IGF1R Gene cDNA in Alopex lagopus (Arctic Fox). Life (Basel) 2025; 15:796. [PMID: 40430222 PMCID: PMC12113172 DOI: 10.3390/life15050796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/07/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
This study aimed to clarify the sequence characteristics and spatiotemporal expression patterns of the insulin-like growth factor 1 receptor (IGF1R) gene in Alopex lagopus (Arctic fox), thereby addressing the existing knowledge gap regarding IGF1R-mediated growth regulation in this species. The findings establish a crucial foundation for subsequent investigations into the correlation between this gene and Arctic fox growth traits. Specific primers were designed based on the cDNA sequence of the canine IGF1R gene (Accession No. XM_545828). The full-length coding sequence (CDS) of the Arctic fox IGF1R gene (1617 bp, encoding 538 amino acids) was successfully cloned via RT-PCR. Phylogenetic analysis using the Unweighted Pair Group Method with Arithmetic Mean (UPGMA) algorithm revealed a 99% sequence homology in the IGF1R gene between the Arctic fox and canine, confirmed their closest evolutionary relationship. Protein characterization showed that the IGF1R protein has a molecular weight of 60.62 kDa (theoretical isoelectric point pI = 5.15), containing one fibronectin type-III domain and one tyrosine kinase domain, classifying it as an acidic hydrophilic transmembrane protein. Phosphorylation site prediction identified 27 phosphorylation sites, with secondary structures dominated by α-helices (26.39%) and random coils (52.79%). The IGF1R gene displayed significant tissue-specific expression variations across 12 examined tissues in Arctic foxes: highest expression levels in testis, minimal expression in stomach, and no detectable expression in duodenum. Spatiotemporal expression analysis revealed that in 2-, 4-, and 6-month-old individuals, hepatic IGF1R exhibited a progressive increase, testicular expression reached peak levels at 6 months, and skeletal muscle demonstrated transient upregulation peaking at 4 months. These spatiotemporal expression patterns suggest that IGF1R may participate in metabolism and organ developmental processes during critical growth stages of Arctic foxes through tissue-specific regulation.
Collapse
Affiliation(s)
| | | | | | | | - Fangyong Ning
- College of Animal Sciences and Technology, Northeast Agricultural University, Harbin 150030, China; (W.X.); (H.F.); (X.M.); (Y.S.)
| | - Zhiheng Du
- College of Animal Sciences and Technology, Northeast Agricultural University, Harbin 150030, China; (W.X.); (H.F.); (X.M.); (Y.S.)
| |
Collapse
|
5
|
Di Chiara M, Spiriti C, Gloria F, Laccetta G, Dito L, Gharbiya M, Rizzo G, Terrin G. Fetal Distress as a Determinant for Refeeding Syndrome in Preterm Neonates. Nutrients 2025; 17:1417. [PMID: 40362726 PMCID: PMC12073788 DOI: 10.3390/nu17091417] [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: 03/03/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: Preterm neonates receiving parenteral nutrition (PN) are at risk of developing refeeding syndrome (RS). Risk factors and the related consequences remain largely undefined. In particular, the reason why only some preterm neonates out of a group receiving the same nutritional protocol will develop RS is yet to be fully understood. The aims of this study were to explore the clinical and nutritional factors contributing to RS and to assess the clinical consequences of this condition. Methods: A retrospective study was conducted, including all newborns with gestational age ≤ 34 weeks and/or body birth weight ≤ 1500 g who were consecutively admitted to the neonatal intensive care unit (NICU) of "Umberto I" Hospital, Sapienza University of Rome, from 2015 to 2022. The population was divided into two groups comprising newborns who developed RS (cases) and infants who did not develop the condition (controls) up to the first 2 weeks of life. The enrolled newborns were compared for clinical and nutritional factors and main morbidities. Results: A total of 412 neonates were enrolled, consisting of 53 cases and 359 controls. The main prenatal risk factor for RS was found to be fetal distress (p = 0.028). The occurrence of RS was identified as statistically significantly associated (p = 0.010; p = 0.007) with the development of extrauterine growth restriction (EUGR) and retinopathy of prematurity (ROP). Conclusions: Fetal distress is the predominant perinatal risk factor associated with the development of RS in preterm neonates managed with early currently recommended PN. These findings suggest an increased risk of ROP and EUGR in preterm neonates with RS.
Collapse
Affiliation(s)
- Maria Di Chiara
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University, 00161 Rome, Italy; (C.S.); (F.G.); (G.L.); (L.D.); (G.R.); (G.T.)
| | - Caterina Spiriti
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University, 00161 Rome, Italy; (C.S.); (F.G.); (G.L.); (L.D.); (G.R.); (G.T.)
| | - Flavia Gloria
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University, 00161 Rome, Italy; (C.S.); (F.G.); (G.L.); (L.D.); (G.R.); (G.T.)
| | - Gianluigi Laccetta
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University, 00161 Rome, Italy; (C.S.); (F.G.); (G.L.); (L.D.); (G.R.); (G.T.)
| | - Lucia Dito
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University, 00161 Rome, Italy; (C.S.); (F.G.); (G.L.); (L.D.); (G.R.); (G.T.)
| | - Magda Gharbiya
- Department of Sense Organs, Sapienza University of Rome, 155, Viale del Policlinico, 00161 Rome, Italy;
| | - Giuseppe Rizzo
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University, 00161 Rome, Italy; (C.S.); (F.G.); (G.L.); (L.D.); (G.R.); (G.T.)
| | - Gianluca Terrin
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University, 00161 Rome, Italy; (C.S.); (F.G.); (G.L.); (L.D.); (G.R.); (G.T.)
| |
Collapse
|
6
|
Yan S, Gao C, Tian K, Xiao C, Shi J, Jia X, Wang K, Sun G, Li D, Li W, Kang X. Comparative population genomics analysis for chicken body sizes using genome-wide single nucleotide polymorphisms. Anim Biosci 2025; 38:600-611. [PMID: 39482999 PMCID: PMC11917417 DOI: 10.5713/ab.24.0347] [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: 05/23/2024] [Revised: 08/27/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
OBJECTIVE This study aims to investigate the selection history, genome regions, and candidate genes associated with different chicken body sizes, thereby providing insights into the genetic basis of complex economic traits such as chicken body size and growth. METHODS In this study, a total of 217 individuals from eight breeds were selected. According to body size, they were divided into two groups: large chickens and bantam chickens, with four breeds in each group. Firstly, we investigate population structure by principal component analysis (PCA), phylogenetic tree, and ancestry component analysis. Next, we recognize runs of homozygosity (ROH) islands through calculating ROH. Finally, we carry out selection signatures analysis utilizing population differentiation index and nucleic acid diversity. RESULTS The population structure analysis show that large and bantam chickens are clearly separated. Large chickens are clustered together, the bantam chickens are relatively dispersed. The results of ROH island analysis show that 48 and 56 ROH islands were identified in large and bantam chickens respectively. Among the interesting ROH islands, a total of eight candidate genes were identified. In selection signatures analysis, a total of 322 selected genes were annotated in large chickens, such as POU1F1, BMP10, enrichment in 16 gene ontology (GO) terms. In bantam chickens, a total of 447 selected genes were annotated, such as IGF1, GRB10, enrichment in 20 GO terms and 2 Kyoto encyclopedia of genes and genomes pathways. The haplotype analysis results show that GRB10 has differences in chickens of different body sizes. CONCLUSION By population structure, ROH islands, and selection signatures analysis, we have identified multiple genes associated with chicken body size, growth, and development (such as BMP10, IGF1, GRB10, etc). This provides a theoretical reference for the subsequent development of molecular markers for chicken body size and the analysis of the genetic mechanism of chicken body size.
Collapse
Affiliation(s)
- Sensen Yan
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046,
China
- The Shennong Laboratory, Zhengzhou 450046,
China
| | - Chaoqun Gao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046,
China
- The Shennong Laboratory, Zhengzhou 450046,
China
| | - Kaiyuan Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046,
China
- The Shennong Laboratory, Zhengzhou 450046,
China
| | - Chengpeng Xiao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046,
China
- The Shennong Laboratory, Zhengzhou 450046,
China
| | - Junlai Shi
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046,
China
- The Shennong Laboratory, Zhengzhou 450046,
China
| | - Xintao Jia
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046,
China
- The Shennong Laboratory, Zhengzhou 450046,
China
| | - Kejun Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046,
China
| | - Guirong Sun
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046,
China
- The Shennong Laboratory, Zhengzhou 450046,
China
| | - Donghua Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046,
China
- The Shennong Laboratory, Zhengzhou 450046,
China
| | - Wenting Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046,
China
- The Shennong Laboratory, Zhengzhou 450046,
China
| | - Xiangtao Kang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046,
China
- The Shennong Laboratory, Zhengzhou 450046,
China
| |
Collapse
|
7
|
Ferraretti G, Rill A, Abondio P, Smith K, Ojeda-Granados C, De Fanti S, Alberti M, Izzi M, Sherpa PT, Cocco P, Tiriticco M, Di Marcello M, Dezi A, Gnecchi-Ruscone GA, Natali L, Corcelli A, Marinelli G, Garagnani P, Peluzzi D, Luiselli D, Pettener D, Sarno S, Sazzini M. Convergent evolution of complex adaptive traits modulates angiogenesis in high-altitude Andean and Himalayan human populations. Commun Biol 2025; 8:377. [PMID: 40050470 PMCID: PMC11885840 DOI: 10.1038/s42003-025-07813-6] [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] [Received: 03/11/2024] [Accepted: 02/25/2025] [Indexed: 03/09/2025] Open
Abstract
Convergent adaptations represent paradigmatic examples of the capacity of natural selection to influence organisms' biology. However, the possibility to investigate the genetic determinants underpinning convergent complex adaptive traits has been offered only recently by methods for inferring polygenic adaptations from genomic data. Relying on this approach, we demonstrate how high-altitude Andean human groups experienced pervasive selective events at angiogenic pathways, which resemble those previously attested for Himalayan populations despite partial convergence at the single-gene level was observed. This provides additional evidence for the drivers of convergent evolution of enhanced blood perfusion in populations exposed to hypobaric hypoxia for thousands of years.
Collapse
Affiliation(s)
- Giulia Ferraretti
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Aina Rill
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
- Josep Carreras Leukaemia Research Institute, PhD Programme in Biomedicine, University of Barcelona, Barcelona, Spain
| | - Paolo Abondio
- Department of Cultural Heritage, Ravenna Campus, University of Bologna, Ravenna, Italy
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Kyra Smith
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Claudia Ojeda-Granados
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences and Advanced Technologies "GF Ingrassia", University of Catania, Catania, Italy
| | - Sara De Fanti
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Marta Alberti
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Massimo Izzi
- Complex Operative Unit of Endocrinology and Diabetes Care, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Paolo Cocco
- Explora Nunaat International, Montorio al Vomano, Teramo, Italy
| | | | | | - Agnese Dezi
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Bari, Italy
| | - Guido Alberto Gnecchi-Ruscone
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Archaeo- and Palaeogenetics, Institute for Archaeological Sciences, Department of Geosciences & Senckenberg Centre for Human Evolution and Palaeoenvironment, University of Tübingen, Tübingen, Germany
| | - Luca Natali
- Explora Nunaat International, Montorio al Vomano, Teramo, Italy
- Italian Institute of Human Paleontology, Rome, Italy
| | - Angela Corcelli
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | | | - Paolo Garagnani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Davide Peluzzi
- Explora Nunaat International, Montorio al Vomano, Teramo, Italy
| | - Donata Luiselli
- Department of Cultural Heritage, Ravenna Campus, University of Bologna, Ravenna, Italy
| | - Davide Pettener
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Stefania Sarno
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Marco Sazzini
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy.
- Interdepartmental Centre Alma Mater Research Institute on Global Changes and Climate Change, University of Bologna, Bologna, Italy.
| |
Collapse
|
8
|
Miao J, Zhang Y, Su C, Zheng Q, Guo J. Insulin-Like Growth Factor Signaling in Alzheimer's Disease: Pathophysiology and Therapeutic Strategies. Mol Neurobiol 2025; 62:3195-3225. [PMID: 39240280 PMCID: PMC11790777 DOI: 10.1007/s12035-024-04457-1] [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: 04/24/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia among the elderly population, posing a significant public health challenge due to limited therapeutic options that merely delay cognitive decline. AD is associated with impaired energy metabolism and reduced neurotrophic signaling. The insulin-like growth factor (IGF) signaling pathway, crucial for central nervous system (CNS) development, metabolism, repair, cognition, and emotion regulation, includes IGF-1, IGF-2, IGF-1R, IGF-2R, insulin receptor (IR), and six insulin-like growth factor binding proteins (IGFBPs). Research has identified abnormalities in IGF signaling in individuals with AD and AD models. Dysregulated expression of IGFs, receptors, IGFBPs, and disruptions in downstream phosphoinositide 3-kinase-protein kinase B (PI3K/AKT) and mitogen-activated protein kinase (MAPK) pathways collectively increase AD susceptibility. Studies suggest modulating the IGF pathway may ameliorate AD pathology and cognitive decline. This review explores the CNS pathophysiology of IGF signaling in AD progression and assesses the potential of targeting the IGF system as a novel therapeutic strategy. Further research is essential to elucidate how aberrant IGF signaling contributes to AD development, understand underlying molecular mechanisms, and evaluate the safety and efficacy of IGF-based treatments.
Collapse
Affiliation(s)
- Jie Miao
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yanli Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Department of Neurology, Sixth Hospital of Shanxi Medical University (General Hospital of Tisco), Taiyuan, 030001, Shanxi, China
| | - Chen Su
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Qiandan Zheng
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Junhong Guo
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
9
|
Cannarella R, Suryavanshi M, Calogero AE, Desai N, Lundy SD, Miller AW. Deciphering Sperm-Carried IGF2 Transcript Variants: Cloning, qPCR Detection, and Variant Analysis. Endocrinology 2025; 166:bqaf035. [PMID: 39950974 DOI: 10.1210/endocr/bqaf035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Indexed: 03/06/2025]
Abstract
CONTEXT The insulin-like growth factor 2 (IGF2) gene, a paternally imprinted gene inactive in oocytes, plays a vital role in early embryo development. While 5 IGF2 variants have been described, the specific variants expressed in human spermatozoa compared to granulosa cells (GCs) remain unclear. OBJECTIVE To characterize the quantity and variants of IGF2 transcripts expressed in human spermatozoa. METHODS Post-gradient sperm samples were collected from 2 healthy, fertile men with normal semen parameters, while GCs were isolated following an oocyte retrieval procedure of a woman undergoing in vitro fertilization due to male factor infertility. RNA extraction, cDNA synthesis, PCR amplification, and cloning were performed. PCR products were ligated into PCR4-TOPO vectors and transformed into Escherichia coli DH-10α. A total of 96 positive clones (32 per sample) were characterized via Sanger sequencing to identify variants. Quantitative PCR (qPCR) with gene-specific primers analyzed transcript quantities, single nucleotide polymorphisms (SNPs), product sizes, and melting temperatures. RESULTS Of the 96 true-positive IGF2 cDNA clones, 14 distinct variants were identified, including deletions, insertions, and SNPs, resulting in amino acid sequence changes. Two common variants were present in both sperm and GCs, while 2 were GC-specific, and the remaining were exclusive to spermatozoa. Some clustered with known NCBI variants, while others formed 2 novel phylogenetic clusters. CONCLUSION This study expands the repertoire of IGF2 variants and highlights differences between spermatozoa and GC transcripts. It is the first to analyze IGF2 variants in sperm from fertile men, paving the way for future research into their role in embryogenesis.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania 95123, Italy
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Mangesh Suryavanshi
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania 95123, Italy
| | - Nina Desai
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Women's Health Institute, Cleveland Clinic, Beachwood, OH 44122, USA
| | - Scott D Lundy
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Aaron W Miller
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44106, USA
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
10
|
Wang P, Sheng X, Xia X, Wang F, Li R, Ahmed Z, Chen N, Lei C, Ma Z. The genomic landscape of short tandem repeats in cattle. Anim Genet 2025; 56:e13498. [PMID: 39692037 DOI: 10.1111/age.13498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024]
Abstract
Short tandem repeats (STRs) are abundant and have high mutation rates across cattle genomes; however, comprehensive exploration of cattle STRs is needed. Here, we constructed a comprehensive map of 467 553 polymorphic STRs (pSTRs) constructed from 423 cattle genomes representing 59 breeds worldwide. We observed that pSTRs in coding sequences and 5'UTRs (Untranslated Regions) were under strong selective constraints and exhibited a relatively low level of diversity. Furthermore, we found that these pSTRs underwent more contraction than expansion. Population analysis showed a strong positive correlation (R = 1) between pSTR diversity and single nucleotide polymorphic heterozygosity. We also investigated STR differences between taurine and indicine cattle and detected 2301 highly divergent STRs, which might relate to immune, endocrine and neurodevelopmental pathways. In summary, our large-scale study characterizes the spectrum of STRs in cattle, expands the scale of known cattle STR variation and provides novel insights into differences among various cattle subspecies.
Collapse
Affiliation(s)
- Pengfei Wang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xin Sheng
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Xiaoting Xia
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Fuwen Wang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Ruizhe Li
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Zulfiqar Ahmed
- Department of Livestock and Poultry Production, Faculty of Veterinary and Animal Sciences, University of Poonch Rawalakot, Azad Jammu and Kashmir, Pakistan
| | - Ningbo Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Zhijie Ma
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| |
Collapse
|
11
|
Zhang M, Traspov A, Yang J, Zheng M, Kharzinova VR, Ai H, Zinovieva NA, Huang L. Genomic and transcriptomic insights into vitamin A-induced thermogenesis and gene reuse as a cold adaptation strategy in wild boars. Commun Biol 2025; 8:116. [PMID: 39856249 PMCID: PMC11759952 DOI: 10.1038/s42003-025-07536-8] [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: 09/08/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Wild boars inhabit diverse climates, including frigid regions like Siberia, but their migration history and cold adaptation mechanisms into high latitudes remain poorly understood. We constructed the most comprehensive wild boar whole-genome variant dataset to date, comprising 124 samples from tropical to frigid zones, among which 47 Russian, 8 South Chinese and 3 Vietnamese wild boars were newly supplemented. We also gathered 75 high-quality RNA-seq datasets from 10 tissues of 6 wild boars from Russia and 6 from southern China. Demographic analysis revealed the appearance of Russian wild boars in Far East of Asia (RUA) and Europe (RUE) after the last glacial maximum till ~ 10 thousand years ago. Recent gene flow (<100 years) from RUA to RUE reflects human-mediated introductions. Cold-region wild boars exhibit strong selection signatures indicative of genetic adaptation to cold climates. Further pathway and transcriptomic analyses reveal a novel cold resistance mechanism centered on enhanced vitamin A metabolism and catalysis, involving the reuse of UGT2B31 and rhythm regulation by ANGPTL8, RLN3 and ZBTB20. This may compensate for the pig's lack of brown fat/UCP1 thermogenesis. These findings provide new insights into the molecular basis of cold adaptation and improve our understanding of Eurasian wild boar migration history.
Collapse
Affiliation(s)
- Mingpeng Zhang
- National Key Laboratory for Swine genetic improvement and production technology, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi Province, P.R. China
- Key Laboratory of Biodiversity Conservation and Bioresource Utilization of Jiangxi Province, College of Life Sciences, Jiangxi Normal University, Nanchang, Jiangxi Province, P.R. China
| | - Aleksei Traspov
- L.K. Ernst Federal Research Center for Animal Husbandry, Dubrovitsy, Podolsk Municipal District, Moscow Region, Podolsk, Russia
| | - Jiawen Yang
- National Key Laboratory for Swine genetic improvement and production technology, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi Province, P.R. China
| | - Min Zheng
- National Key Laboratory for Swine genetic improvement and production technology, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi Province, P.R. China
| | - Veronika R Kharzinova
- L.K. Ernst Federal Research Center for Animal Husbandry, Dubrovitsy, Podolsk Municipal District, Moscow Region, Podolsk, Russia
| | - Huashui Ai
- National Key Laboratory for Swine genetic improvement and production technology, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi Province, P.R. China.
| | - Natalia A Zinovieva
- L.K. Ernst Federal Research Center for Animal Husbandry, Dubrovitsy, Podolsk Municipal District, Moscow Region, Podolsk, Russia.
| | - Lusheng Huang
- National Key Laboratory for Swine genetic improvement and production technology, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi Province, P.R. China.
| |
Collapse
|
12
|
Brusco De Freitas M, Poulsen GJ, Jess T. Anthropometric Trajectories in Children Prior to Development of Inflammatory Bowel Disease. JAMA Netw Open 2025; 8:e2455158. [PMID: 39821394 PMCID: PMC11742528 DOI: 10.1001/jamanetworkopen.2024.55158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/09/2024] [Indexed: 01/19/2025] Open
Abstract
Importance Poor nutrition and growth in childhood have short-term and long-term consequences, so understanding the timing of the onset of an impaired nutritional status is crucial for diagnosing and treating inflammatory bowel disease (IBD) at its earliest stage. Objective To assess anthropometric trajectories before a pediatric diagnosis of IBD and growth recovery after diagnosis. Design, Setting, and Participants This population-based cohort study included children born in Denmark from January 1, 1997, through December 31, 2015, with weight and length or height measurements at birth and at least 1 length or height and weight measurement at school age based on the Danish Medical Birth Register and the Danish National Child Health Register. Within this population, all individuals diagnosed with IBD at ages 5 to 17 years, according to the Danish National Patient Register, were identified. Data were analyzed from October 13, 2023, to April 17, 2024. Exposure A pediatric diagnosis of IBD compared with the corresponding population without the disease. Main Outcomes and Measures The outcome measures were z scores for length or height, weight, and body mass index (BMI [calculated as weight in kilograms divided by height in meters squared]) before and after pediatric IBD diagnosis compared with reference and sibling populations. Results The final study population included 916 133 individuals (51.2% male) with a median of 3 pairs of length or height and weight measurements collected (IQR, 2-6 pairs). Of those, 1522 (median age, 14.3 years [IQR, 11.8-16.3 years]; 763 female [50.1%]) were diagnosed with IBD (851 [55.9%] with Crohn disease [CD] and 671 [44.1%] with ulcerative colitis [UC]). Compared with children without IBD, individuals with a later diagnosis of CD had declining anthropometric measures 3 years (weight: mean, -0.12 g [95% CI, -0.20 to -0.03 g]; BMI: mean, -0.13 [95% CI, -0.21 to -0.04]) and 1 year (length or height: mean, -0.20 cm [95% CI, -0.29 to -0.10 cm]) prior to diagnosis, whereas this was observed 1 year prior to a diagnosis of UC for weight (mean, -0.12 g [95% CI, -0.22 to -0.02 g]) and BMI (mean, -0.13 [95% CI, -0.23 to -0.03]). Deviating anthropometric patterns persisted after diagnosis, with the slowest recovery observed in children with CD. Conclusions and Relevance The findings of this large-scale population-based cohort study of anthropometrics in children suggest impaired nutritional status as assessed by weight up to 3 years and by length or height 1 year before a diagnosis of CD and by weight up to 1 year before a diagnosis of UC. These findings emphasize that the onset of pediatric IBD may occur years prior to diagnosis, that growth recovery may first occur after diagnosis and treatment, and that frequent nutritional screenings may help ensure a healthy transition to adulthood.
Collapse
Affiliation(s)
- Maiara Brusco De Freitas
- Center for Molecular Prediction of Inflammatory Bowel Disease, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark
| | - Gry Juul Poulsen
- Center for Molecular Prediction of Inflammatory Bowel Disease, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark
| | - Tine Jess
- Center for Molecular Prediction of Inflammatory Bowel Disease, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark
- Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
13
|
Khan MZ, Zugaza JL, Torres Aleman I. The signaling landscape of insulin-like growth factor 1. J Biol Chem 2025; 301:108047. [PMID: 39638246 PMCID: PMC11748690 DOI: 10.1016/j.jbc.2024.108047] [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: 07/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
The sheer amplitude of biological actions of insulin-like growth factor I (IGF-1) affecting all types of cells in all tissues suggests a vast signaling landscape for this ubiquitous humoral signal. While the canonical signaling pathways primarily involve the Ras/MAPK and PI3K/AKT cascades, the evolutionary conservation of insulin-like peptides (ILPs) and their pathways hints at the potential for novel functions to emerge over time. Indeed, the evolutionary trajectory of ILPs opens the possibility of either novel functions for these two pathways, novel downstream routes, or both. Evidence supporting this notion includes observations of neofunctionalization in bony fishes or crustaceans, and the involvement of ILPs pathways in invertebrate eusociality or in vertebrate bone physiology, respectively. Such evolutionary processes likely contribute to the rich diversity of ILPs signaling observed today. Moreover, the interplay between conserved signaling pathways, such as those implicated in aging (predominantly involving the PI3K-AKT route), and lesser known pathways, such as those mediated by biased G-protein coupled receptors and others even less known, may underpin the context-dependent actions characteristic of ILPs signaling. While canonical IGF-1 signaling is often assumed to account for the intracellular pathways utilized by this growth factor, a comprehensive analysis of all the pathways mediated by the IGF-1 receptor (IGF-1R) remains lacking. This review aims to explore both canonical and non-canonical routes of IGF-1R action across various cell types, offering a detailed examination of the mechanisms underlying IGF-1 signaling and highlighting the significant gaps in our current understanding.
Collapse
Affiliation(s)
- Muhammad Zahid Khan
- Achucarro Basque Center for Neuroscience, Leioa, Spain; CIBERNED, Madrid, Spain
| | - Jose Luis Zugaza
- Achucarro Basque Center for Neuroscience, Leioa, Spain; Ikerbasque Science Foundation, Bilbao, Spain
| | - Ignacio Torres Aleman
- Achucarro Basque Center for Neuroscience, Leioa, Spain; CIBERNED, Madrid, Spain; Ikerbasque Science Foundation, Bilbao, Spain.
| |
Collapse
|
14
|
Hayes CA, Wilson D, De Leon MA, Mustapha MJ, Morales S, Odden MC, Ashpole NM. Insulin-like growth factor-1 and cognitive health: Exploring cellular, preclinical, and clinical dimensions. Front Neuroendocrinol 2025; 76:101161. [PMID: 39536910 DOI: 10.1016/j.yfrne.2024.101161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
Age and insulin-like growth factor-1 (IGF-1) have an inverse association with cognitive decline and dementia. IGF-1 is known to have important pleiotropic functions beginning in neurodevelopment and extending into adulthood such as neurogenesis. At the cellular level, IGF-1 has pleiotropic signaling mechanisms through the IGF-1 receptor on neurons and neuroglia to attenuate inflammation, promote myelination, maintain astrocytic functions for homeostatic balances, and neuronal synaptogenesis. In preclinical rodent models of aging and transgenic models of IGF-1, increased IGF-1 improves cognition in a variety of behavioral paradigms along with reducing IGF-1 via knockout models being able to induce cognitive impairment. At the clinical levels, most studies highlight that increased levels of IGF-1 are associated with better cognition. This review provides a comprehensive and up-to-date evaluation of the association between IGF-1 and cognition at the cellular signaling levels, preclinical, and clinical levels.
Collapse
Affiliation(s)
- Cellas A Hayes
- Department of Epidemiology and Population Health, School of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA.
| | - Destiny Wilson
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
| | - Miguel A De Leon
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
| | | | - Sharon Morales
- Department of Biomedical Science, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Michelle C Odden
- Department of Epidemiology and Population Health, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Nicole M Ashpole
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
| |
Collapse
|
15
|
Lou Q, Jiang K, Wang X, Pan Y, Qiu G, Wu B, Yuan L, Xie S, Chen J, Xu Q, Zhao M, Jiang L. IGF1R signaling in perinatal mesenchymal stem cells determines definitive hematopoiesis in bone marrow. Blood 2024; 144:2773-2787. [PMID: 39437540 DOI: 10.1182/blood.2024024258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 09/06/2024] [Accepted: 09/21/2024] [Indexed: 10/25/2024] Open
Abstract
ABTRACT During the transition from embryonic to adult life, the sites of hematopoiesis undergo dynamic shifts across various tissues. In adults, although bone marrow (BM) becomes the primary site for definitive hematopoiesis, the establishment of the BM niche for accommodating hematopoietic stem cells (HSCs) remains incompletely understood. Here, we reveal that perinatal BM mesenchymal stem cells (BMSCs) exhibit highly activated insulin-like growth factor 1 receptor (IGF1R) signaling compared with adult BMSCs (aBMSCs). Deletion of Igf1r in perinatal BMSCs (pBMSCs) hinders the transition of HSCs from the fetal liver to the BM in perinatal mice and disrupts hematopoiesis in adult individuals. Conversely, the deletion of Igf1r in aBMSCs, adipocytes, osteoblasts, or endothelial cells does not affect HSCs in the BM. Mechanistically, IGF1R signaling activates the transcription factor nuclear factor of activated T cells c1 in pBMSCs, which upregulates CXCL12 and other niche factors for HSC retention. Overall, IGF1R signaling in pBMSCs regulates the development of the BM niche for hematopoiesis.
Collapse
Affiliation(s)
- Qi Lou
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Kaizheng Jiang
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaoqi Wang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Yuan Pan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Guo Qiu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Binghuo Wu
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lisha Yuan
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Siyu Xie
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jian Chen
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Quanhui Xu
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Meng Zhao
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Linjia Jiang
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
16
|
Gao Y, Feng X, Diao S, Liu Y, Zhong Z, Cai X, Li G, Teng J, Liu X, Li J, Zhang Z. Deciphering genetic characteristics of South China and North China indigenous pigs through selection signatures. BMC Genomics 2024; 25:1191. [PMID: 39695929 DOI: 10.1186/s12864-024-11119-y] [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: 09/26/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Indigenous pig breeds in China have accumulated significant genetic diversity due to regional selection pressures. Investigating the selection signatures of these populations helps to understand their adaptive evolution and contributes to genetic improvement programs. RESULTS We collected whole-genome sequencing data from 133 individuals, including South China and North China indigenous pigs and Asian wild boars. After data filtering, we retained 31,521,978 high-quality SNPs. Population structure analysis using PCA revealed distinct genetic clustering among these populations. Selection signature detection identified 5,227 loci under selection in South China indigenous pigs and 5,800 in North China indigenous pigs compared to Asian wild boars. Candidate genes were enriched in immune response pathways, reproductive traits, and pigmentation pathways. South China indigenous pigs exhibited selection signals for fat deposition and immune responses, while North China indigenous pigs showed stronger signals related to growth, blood physiology, and reproductive performance. Additionally, key genes such as MC1R and KIT were associated with coat color variation, and IGF1R and IGF2R were linked to growth regulation. CONCLUSION Our results demonstrate that indigenous pigs in China have undergone selection for distinct traits aligned with their regional environments and farming systems. South China indigenous pigs have been selected for traits related to fat deposition and immunity, while North China indigenous pigs have been selected for growth and reproductive traits. The findings offer crucial insights into the genetic architecture of indigenous pig breeds, providing a valuable foundation for future genetic breeding programs.
Collapse
Affiliation(s)
- Yahui Gao
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xueyan Feng
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Shuqi Diao
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yuqiang Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Zhanming Zhong
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaotian Cai
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Guangzhen Li
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jinyan Teng
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaohong Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Jiaqi Li
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Zhe Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
17
|
Bøtkjær JA, Poulsen LLC, Noer PR, Grøndahl ML, Englund ALM, Franks S, Hardy K, Oxvig C, Andersen CY. Dynamics of IGF Signaling During the Ovulatory Peak in Women Undergoing Ovarian Stimulation. J Clin Endocrinol Metab 2024; 110:e160-e167. [PMID: 38436415 DOI: 10.1210/clinem/dgae132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/04/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
CONTEXT Insulin-like growth factor (IGF) signaling is known to affect human ovarian follicular function during growth and development. However, the role of the IGF system is unknown during the ovulatory peak, which is characterized by profound changes in granulosa cell (GCs) mitosis and function. OBJECTIVE How is the IGF system expressed and regulated during the midcycle surge in women? METHODS Follicular fluid (FF) and GCs were collected during the ovulatory peak from 2 specific time points. One sample was obtained before oocyte pickup (OPU): before ovulation trigger (OT) (T = 0 hours) or at 12, 17, or 32 hours after OT, and 1 sample was obtained at OPU 36 hours after OT. Fifty women undergoing ovarian stimulation at a university hospital were included. Gene expression profiles were assessed by microarray analysis of GCs. IGF-related proteins in the FF were assessed by immunoassay or by determination of activity with a proteinase assay. RESULTS Gene expression of proteins promoting IGF activity (ie, IGF2, PAPP-A, and IRS1) together with proliferation markers were downregulated on a transcriptional level in GCs after OT, whereas proteins inhibiting the IGF signal (ie, IGFBPs, IGF2, and STC1) were upregulated. STC1 gene expression and protein levels were greatly upregulated after OT with a parallel steep downregulation of PAPP-A proteolytic activity. CONCLUSION These data suggest that downregulation of IGF signaling mediated by increased STC1 expression is instrumental for the sudden cessation in GC proliferation and onset of differentiation during the ovulatory peak.
Collapse
Affiliation(s)
- Jane Alrø Bøtkjær
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, 2100 Copenhagen, Denmark
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Liv la Cour Poulsen
- Fertility Clinic, Zealand University Hospital, 4600 Køge, Denmark
- Fertility Clinic, University Hospital of Copenhagen, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| | - Pernille Rimmer Noer
- Department of Molecular Biology and Genetics, University of Aarhus, 8000 Aarhus, Denmark
| | - Marie Louise Grøndahl
- Fertility Clinic, University Hospital of Copenhagen, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| | | | - Stephen Franks
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Kate Hardy
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, University of Aarhus, 8000 Aarhus, Denmark
| | - Claus Yding Andersen
- Fertility Clinic, University Hospital of Copenhagen, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
18
|
Yoshimura Y, Watanabe T, Nakamura K, Futatsugi A, Mikoshiba K, Hiyama TY. High-temperature exposure during the early embryonic stage lowers core body temperature after growth via a hypothalamic Igfbp2-dependent mechanism. Sci Rep 2024; 14:29586. [PMID: 39627352 PMCID: PMC11615319 DOI: 10.1038/s41598-024-80252-1] [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: 08/08/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024] Open
Abstract
The mechanisms underlying individual differences in core body temperature (Tc) are unexplained by genetic factors and poorly understood. Here, we investigated whether the environmental temperature during early development affects postnatal Tc. Mouse embryos were cultured from pronuclear to blastocyst stage in either standard (37 °C) or high (38 °C) temperature, and the Tc of each grown-up adult was measured. The adult 38 °C-incubated mice showed lower Tc than the 37 °C group without changes in activity levels. In the hypothalamus of the 38 °C group, insulin-like growth factor 1 (Igf1) and IGF binding protein 2 (Igfbp2) gene expression increased. The decrease in Tc in the wild-type 38 °C group was alleviated by brain neuron-specific Igfbp2 knockout. This suggests that IGFBP2 binds to IGF-1 and, inhibits its binding to the receptor, thereby interfering with the thermogenic signaling of IGF-1. These results suggest that one of the factors determining individual postnatal Tc is the ambient temperature of embryos at an early developmental stage, which could affect epigenetic changes, such as DNA methylation, leading to alterations in the Igf1 and Igfbp2 gene expressions in adulthood.
Collapse
Affiliation(s)
- Yuki Yoshimura
- Department of Integrative Physiology, Tottori University Graduate School and Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan.
| | - Tatsuo Watanabe
- Department of Integrative Physiology, Tottori University Graduate School and Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan
| | - Kazuomi Nakamura
- Advanced Medicine, Innovation and Clinical Research Center, Tottori University Hospital, 36-1 Nishi-cho, Yonago, Tottori, 683-8504, Japan
| | - Akira Futatsugi
- Department of Basic Medical Sciences, Kobe City College of Nursing, 3-4 Gakuen-nishi-machi, Nishi-ku, Kobe, Hyogo, 651-2103, Japan
| | - Katsuhiko Mikoshiba
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China
- Faculty of Science, Toho University, Miyama 2-2-1, Funabashi, Chiba, 274-8510, Japan
| | - Takeshi Y Hiyama
- Department of Integrative Physiology, Tottori University Graduate School and Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan.
- International Platform for Dryland Research and Education, Tottori University, 1390 Hamasaka, Tottori, Tottori, 680-0001, Japan.
| |
Collapse
|
19
|
Lee VY, Nils AVM, Arruda BP, Xavier GF, Nogueira MI, Motta-Teixeira LC, Takada SH. Spontaneous running wheel exercise during pregnancy prevents later neonatal-anoxia-induced somatic and neurodevelopmental alterations. IBRO Neurosci Rep 2024; 17:263-279. [PMID: 39310269 PMCID: PMC11414703 DOI: 10.1016/j.ibneur.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction About 15-20 % of babies that suffer perinatal asphyxia die and around 25 % of the survivors exhibit permanent neural outcomes. Minimization of this global health problem has been warranted. This study investigated if the offspring of pregnant female rats allowed to spontaneously exercise on running wheels along a 11-day pregnancy period were protected for somatic and neurodevelopmental disturbs that usually follow neonatal anoxia. Methods spontaneous exercise was applied to female rats which were housed in cages allowing free access to running wheels along a 11-day pregnancy period. Their offspring were submitted to anoxia 24-36 h after birth. Somatic and sensory-motor development of the pups were recorded until postnatal day 21 (P21). Myelin basic protein (MBP)-stained areas of sensory and motor cortices were measured at P21. Neuronal nuclei (NeuN)-immunopositive cells and synapsin-I levels in hippocampal formation were estimated at P21 and P75. Results gestational exercise and / or neonatal anoxia increased the weight and the size of the pups. In addition, gestational exercise accelerated somatic and sensory-motor development of the pups and protected them against neonatal-anoxia-induced delay in development. Further, neonatal anoxia reduced MBP stained area in the secondary motor cortex and decreased hippocampal neuronal estimates and synapsin-I levels at P21; gestational exercise prevented these effects. Therefore, spontaneous exercise along pregnancy is a valuable strategy to prevent neonatal-anoxia-induced disturbs in the offspring. Conclusion spontaneous gestational running wheel exercise protects against neonatal anoxia-induced disturbs in the offspring, including (1) physical and neurobehavioral developmental impairments, and (2) hippocampal and cortical changes. Thus, spontaneous exercise during pregnancy may represent a valuable strategy to prevent disturbs which usually follow neonatal anoxia.
Collapse
Affiliation(s)
- Vitor Yonamine Lee
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 2415, Sao Paulo, SP 05508-900, Brazil
| | - Aline Vilar Machado Nils
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, R. do Matão, Travessa 14, 101, Sao Paulo 05508-900, Brazil
| | - Bruna Petrucelli Arruda
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Alameda da Universidade, s/n, Bloco Delta, São Bernardo do Campo, SP 09606-070, Brazil
| | - Gilberto Fernando Xavier
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, R. do Matão, Travessa 14, 101, Sao Paulo 05508-900, Brazil
| | - Maria Inês Nogueira
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 2415, Sao Paulo, SP 05508-900, Brazil
| | - Lívia Clemente Motta-Teixeira
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 2415, Sao Paulo, SP 05508-900, Brazil
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, R. do Matão, Travessa 14, 101, Sao Paulo 05508-900, Brazil
- Departamento de Ciências Fisiológicas, Faculdade de Ciências Médicas da Santa Casa de São Paulo, R. Jaguaribe, 155 - Vila Buarque, Sao Paulo, SP 01224-001, Brazil
| | - Silvia Honda Takada
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 2415, Sao Paulo, SP 05508-900, Brazil
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Alameda da Universidade, s/n, Bloco Delta, São Bernardo do Campo, SP 09606-070, Brazil
| |
Collapse
|
20
|
Wiens KR, Wasti N, Ulloa OO, Klegeris A. Diversity of Microglia-Derived Molecules with Neurotrophic Properties That Support Neurons in the Central Nervous System and Other Tissues. Molecules 2024; 29:5525. [PMID: 39683685 DOI: 10.3390/molecules29235525] [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: 10/02/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Microglia, the brain immune cells, support neurons by producing several established neurotrophic molecules including glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF). Modern analytical techniques have identified numerous phenotypic states of microglia, each associated with the secretion of a diverse set of substances, which likely include not only canonical neurotrophic factors but also other less-studied molecules that can interact with neurons and provide trophic support. In this review, we consider the following eight such candidate cytokines: oncostatin M (OSM), leukemia inhibitory factor (LIF), activin A, colony-stimulating factor (CSF)-1, interleukin (IL)-34, growth/differentiation factor (GDF)-15, fibroblast growth factor (FGF)-2, and insulin-like growth factor (IGF)-2. The available literature provides sufficient evidence demonstrating murine cells produce these cytokines and that they exhibit neurotrophic activity in at least one neuronal model. Several distinct types of neurotrophic activity are identified that only partially overlap among the cytokines considered, reflecting either their distinct intrinsic properties or lack of comprehensive studies covering the full spectrum of neurotrophic effects. The scarcity of human-specific studies is another significant knowledge gap revealed by this review. Further studies on these potential microglia-derived neurotrophic factors are warranted since they may be used as targeted treatments for diverse neurological disorders.
Collapse
Affiliation(s)
- Kennedy R Wiens
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Naved Wasti
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Omar Orlando Ulloa
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Andis Klegeris
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| |
Collapse
|
21
|
Wilson RL, Schmidt JK, Davenport BN, Ren E, Keding LT, Shaw SA, Schotzko ML, Antony KM, Simmons HA, Golos TG, Jones HN. Placental gene therapy in nonhuman primates: a pilot study of maternal, placental, and fetal response to non-viral, polymeric nanoparticle delivery of IGF1. Mol Hum Reprod 2024; 30:gaae038. [PMID: 39499161 PMCID: PMC11562130 DOI: 10.1093/molehr/gaae038] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/14/2024] [Indexed: 11/07/2024] Open
Abstract
Currently, there are no placenta-targeted treatments to alter the in utero environment for administration to pregnant women who receive a diagnosis of fetal growth restriction (FGR). Water-soluble polymers have a distinguished record of clinical relevance outside of pregnancy. We have demonstrated the effective delivery of polymer-based nanoparticles containing a non-viral human insulin-like growth factor 1 (IGF1) transgene to correct placental insufficiency in small animal models of FGR. Our goals were to extend these studies to a proof-of-concept study in the pregnant macaque, establish feasibility of nanoparticle-mediated gene therapy delivery to trophoblasts, and investigate the acute maternal, placental, and fetal responses to treatment. Pregnant macaques underwent ultrasound-guided intraplacental injections of nanoparticles (GFP- or IGF1-expressing plasmid under the control of the trophoblast-specific PLAC1 promoter complexed with a HPMA-DMEAMA co-polymer) at approximately gestational day 100 (term = 165 days). Fetectomy was performed 24 h (GFP; n = 1), 48 h (IGF1; n = 3) or 10 days (IGF1; n = 3) after nanoparticle delivery. Routine pathological assessment was performed on biopsied maternal tissues and placental and fetal tissues. Maternal blood was analyzed for complete blood count (CBC), immunomodulatory proteins and growth factors, progesterone (P4), and estradiol (E2). Placental ERK/AKT/mTOR signaling was assessed using Western blot and qPCR. Fluorescent microscopy and in situ hybridization confirmed placental uptake and transient transgene expression in villous syncytiotrophoblast. No off-target expression was observed in either maternal or fetal tissues. Histopathological assessment of the placenta recorded observations not necessarily related to the IGF1 nanoparticle treatment. In maternal blood, CBCs, P4, and E2 remained within the normal range for pregnant macaques across the treatment period. Changes to placental ERK and AKT signaling at 48 h and 10 days after IGF1 nanoparticle treatment indicated an upregulation in placental homeostatic mechanisms to prevent overactivity in the normal pregnancy environment. The lack of adverse maternal reaction to nanoparticle-mediated IGF1 treatment, combined with changes in placental signaling to maintain homeostasis, indicates no deleterious impact of treatment during the acute phase of study.
Collapse
Affiliation(s)
- Rebecca L Wilson
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Research in Perinatal Outcomes, University of Florida, Gainesville, FL, USA
| | - Jenna Kropp Schmidt
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Baylea N Davenport
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Research in Perinatal Outcomes, University of Florida, Gainesville, FL, USA
| | - Emily Ren
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Logan T Keding
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Sarah A Shaw
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Michele L Schotzko
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Kathleen M Antony
- Department of Obstetrics and Gynecology, University of Wisconsin–Madison, Madison, WI, USA
| | - Heather A Simmons
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, USA
- Department of Obstetrics and Gynecology, University of Wisconsin–Madison, Madison, WI, USA
- Department of Comparative Biosciences, University of Wisconsin–Madison, Madison, WI, USA
| | - Helen N Jones
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Research in Perinatal Outcomes, University of Florida, Gainesville, FL, USA
| |
Collapse
|
22
|
Giunti S, Blanco MG, De Rosa MJ, Rayes D. The ketone body β-hydroxybutyrate ameliorates neurodevelopmental deficits in the GABAergic system of daf-18/PTEN Caenorhabditis elegans mutants. eLife 2024; 13:RP94520. [PMID: 39422188 PMCID: PMC11488850 DOI: 10.7554/elife.94520] [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] [Indexed: 10/19/2024] Open
Abstract
A finely tuned balance between excitation and inhibition (E/I) is essential for proper brain function. Disruptions in the GABAergic system, which alter this equilibrium, are a common feature in various types of neurological disorders, including autism spectrum disorders (ASDs). Mutations in Phosphatase and Tensin Homolog (PTEN), the main negative regulator of the phosphatidylinositol 3-phosphate kinase/Akt pathway, are strongly associated with ASD. However, it is unclear whether PTEN deficiencies can differentially affect inhibitory and excitatory signaling. Using the Caenorhabditis elegans neuromuscular system, where both excitatory (cholinergic) and inhibitory (GABAergic) inputs regulate muscle activity, we found that daf-18/PTEN mutations impact GABAergic (but not cholinergic) neurodevelopment and function. This selective impact results in a deficiency in inhibitory signaling. The defects observed in the GABAergic system in daf-18/PTEN mutants are due to reduced activity of DAF-16/FOXO during development. Ketogenic diets (KGDs) have proven effective for disorders associated with E/I imbalances. However, the mechanisms underlying their action remain largely elusive. We found that a diet enriched with the ketone body β-hydroxybutyrate during early development induces DAF-16/FOXO activity, therefore improving GABAergic neurodevelopment and function in daf-18/PTEN mutants. Our study provides valuable insights into the link between PTEN mutations and neurodevelopmental defects and delves into the mechanisms underlying the potential therapeutic effects of KGDs.
Collapse
Affiliation(s)
- Sebastián Giunti
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) (UNS-CONICET), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y TécnicasBahia BlancaArgentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)Bahia BlancaArgentina
| | - María Gabriela Blanco
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) (UNS-CONICET), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y TécnicasBahia BlancaArgentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)Bahia BlancaArgentina
| | - María José De Rosa
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) (UNS-CONICET), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y TécnicasBahia BlancaArgentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)Bahia BlancaArgentina
| | - Diego Rayes
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) (UNS-CONICET), Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y TécnicasBahia BlancaArgentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional Del Sur (UNS)Bahia BlancaArgentina
| |
Collapse
|
23
|
Bertels JC, He G, Long F. Metabolic reprogramming in skeletal cell differentiation. Bone Res 2024; 12:57. [PMID: 39394187 PMCID: PMC11470040 DOI: 10.1038/s41413-024-00374-0] [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: 05/16/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 10/13/2024] Open
Abstract
The human skeleton is a multifunctional organ made up of multiple cell types working in concert to maintain bone and mineral homeostasis and to perform critical mechanical and endocrine functions. From the beginning steps of chondrogenesis that prefigures most of the skeleton, to the rapid bone accrual during skeletal growth, followed by bone remodeling of the mature skeleton, cell differentiation is integral to skeletal health. While growth factors and nuclear proteins that influence skeletal cell differentiation have been extensively studied, the role of cellular metabolism is just beginning to be uncovered. Besides energy production, metabolic pathways have been shown to exert epigenetic regulation via key metabolites to influence cell fate in both cancerous and normal tissues. In this review, we will assess the role of growth factors and transcription factors in reprogramming cellular metabolism to meet the energetic and biosynthetic needs of chondrocytes, osteoblasts, or osteoclasts. We will also summarize the emerging evidence linking metabolic changes to epigenetic modifications during skeletal cell differentiation.
Collapse
Affiliation(s)
- Joshua C Bertels
- Department of Surgery, Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Guangxu He
- Department of Surgery, Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Orthopedics, The Second Xiangya Hospital, Changsha, Hunan, China
| | - Fanxin Long
- Department of Surgery, Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Moorwood K, Smith FM, Garfield AS, Cowley M, Holt LJ, Daly RJ, Ward A. Grb7, Grb10 and Grb14, encoding the growth factor receptor-bound 7 family of signalling adaptor proteins have overlapping functions in the regulation of fetal growth and post-natal glucose metabolism. BMC Biol 2024; 22:221. [PMID: 39343875 PMCID: PMC11441139 DOI: 10.1186/s12915-024-02018-5] [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: 02/02/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND The growth factor receptor bound protein 7 (Grb7) family of signalling adaptor proteins comprises Grb7, Grb10 and Grb14. Each can interact with the insulin receptor and other receptor tyrosine kinases, where Grb10 and Grb14 inhibit insulin receptor activity. In cell culture studies they mediate functions including cell survival, proliferation, and migration. Mouse knockout (KO) studies have revealed physiological roles for Grb10 and Grb14 in glucose-regulated energy homeostasis. Both Grb10 KO and Grb14 KO mice exhibit increased insulin signalling in peripheral tissues, with increased glucose and insulin sensitivity and a modestly increased ability to clear a glucose load. In addition, Grb10 strongly inhibits fetal growth such that at birth Grb10 KO mice are 30% larger by weight than wild type littermates. RESULTS Here, we generate a Grb7 KO mouse model. We show that during fetal development the expression patterns of Grb7 and Grb14 each overlap with that of Grb10. Despite this, Grb7 and Grb14 did not have a major role in influencing fetal growth, either alone or in combination with Grb10. At birth, in most respects both Grb7 KO and Grb14 KO single mutants were indistinguishable from wild type, while Grb7:Grb10 double knockout (DKO) were near identical to Grb10 KO single mutants and Grb10:Grb14 DKO mutants were slightly smaller than Grb10 KO single mutants. In the developing kidney Grb7 had a subtle positive influence on growth. An initial characterisation of Grb7 KO adult mice revealed sexually dimorphic effects on energy homeostasis, with females having a significantly smaller renal white adipose tissue depot and an enhanced ability to clear glucose from the circulation, compared to wild type littermates. Males had elevated fasted glucose levels with a trend towards smaller white adipose depots, without improved glucose clearance. CONCLUSIONS Grb7 and Grb14 do not have significant roles as inhibitors of fetal growth, unlike Grb10, and instead Grb7 may promote growth of the developing kidney. In adulthood, Grb7 contributes subtly to glucose mediated energy homeostasis, raising the possibility of redundancy between all three adaptors in physiological regulation of insulin signalling and glucose handling.
Collapse
Affiliation(s)
- Kim Moorwood
- Department of Life Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Florentia M Smith
- Department of Life Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Alastair S Garfield
- Department of Life Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Michael Cowley
- Department of Life Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK
- Present Address: Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Campus, Box 7633, Raleigh, NC, 27695, USA
| | - Lowenna J Holt
- Cancer Research Program, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, NSW, 2010, Australia
| | - Roger J Daly
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Andrew Ward
- Department of Life Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| |
Collapse
|
25
|
Sandovici I, Fernandez-Twinn DS, Campbell N, Cooper WN, Sekita Y, Zvetkova I, Ferland-McCollough D, Prosser HM, Oyama LM, Pantaleão LC, Cimadomo D, Barbosa de Queiroz K, Cheuk CSK, Smith NM, Kay RG, Antrobus R, Hoelle K, Ma MKL, Smith NH, Geyer SH, Reissig LF, Weninger WJ, Siddle K, Willis AE, Lam BYH, Bushell M, Ozanne SE, Constância M. Overexpression of Igf2-derived Mir483 inhibits Igf1 expression and leads to developmental growth restriction and metabolic dysfunction in mice. Cell Rep 2024; 43:114750. [PMID: 39283743 PMCID: PMC7617298 DOI: 10.1016/j.celrep.2024.114750] [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: 01/10/2023] [Revised: 06/04/2024] [Accepted: 08/28/2024] [Indexed: 09/28/2024] Open
Abstract
Mir483 is a conserved and highly expressed microRNA in placental mammals, embedded within the Igf2 gene. Its expression is dysregulated in a number of human diseases, including metabolic disorders and certain cancers. Here, we investigate the developmental regulation and function of Mir483 in vivo. We find that Mir483 expression is dependent on Igf2 transcription and the regulation of the Igf2/H19 imprinting control region. Transgenic Mir483 overexpression in utero causes fetal, but not placental, growth restriction through insulin-like growth factor 1 (IGF1) and IGF2 and also causes cardiovascular defects leading to fetal death. Overexpression of Mir483 post-natally results in growth stunting through IGF1 repression, increased hepatic lipid production, and excessive adiposity. IGF1 infusion rescues the post-natal growth restriction. Our findings provide insights into the function of Mir483 as a growth suppressor and metabolic regulator and suggest that it evolved within the INS-IGF2-H19 transcriptional region to limit excessive tissue growth through repression of IGF signaling.
Collapse
Affiliation(s)
- Ionel Sandovici
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Denise S Fernandez-Twinn
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Niamh Campbell
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Wendy N Cooper
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Yoichi Sekita
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Ilona Zvetkova
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | | | - Haydn M Prosser
- The Wellcome Trust Sanger Institute, Genome Campus, Hinxton, UK
| | - Lila M Oyama
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Departmento de Fisiologia, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil
| | - Lucas C Pantaleão
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Danilo Cimadomo
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; Laboratory of Developmental Biology, Department of Biology and Biotechnology "Lazzaro Spallanzani," University of Pavia, Pavia, Italy
| | - Karina Barbosa de Queiroz
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Cecilia S K Cheuk
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Nicola M Smith
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Richard G Kay
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Katharina Hoelle
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Marcella K L Ma
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Noel H Smith
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Stefan H Geyer
- Center for Anatomy and Cell Biology, Division of Anatomy, Medical University of Vienna, Vienna, Austria
| | - Lukas F Reissig
- Center for Anatomy and Cell Biology, Division of Anatomy, Medical University of Vienna, Vienna, Austria
| | - Wolfgang J Weninger
- Center for Anatomy and Cell Biology, Division of Anatomy, Medical University of Vienna, Vienna, Austria
| | - Kenneth Siddle
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Anne E Willis
- Medical Research Council Toxicology Unit, University of Leicester, Leicester, UK
| | - Brian Y H Lam
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Martin Bushell
- Medical Research Council Toxicology Unit, University of Leicester, Leicester, UK
| | - Susan E Ozanne
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Miguel Constância
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
26
|
Ruan X, Jin X, Sun F, Pi J, Jinghu Y, Lin X, Zhang N, Chen G. IGF signaling pathway in bone and cartilage development, homeostasis, and disease. FASEB J 2024; 38:e70031. [PMID: 39206513 DOI: 10.1096/fj.202401298r] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
The skeleton plays a fundamental role in the maintenance of organ function and daily activities. The insulin-like growth factor (IGF) family is a group of polypeptide substances with a pronounced role in osteoblast differentiation, bone development, and metabolism. Disturbance of the IGFs and the IGF signaling pathway is inextricably linked with assorted developmental defects, growth irregularities, and jeopardized skeletal structure. Recent findings have illustrated the significance of the action of the IGF signaling pathway via growth factors and receptors and its interactions with dissimilar signaling pathways (Wnt/β-catenin, BMP, TGF-β, and Hh/PTH signaling pathways) in promoting the growth, survival, and differentiation of osteoblasts. IGF signaling also exhibits profound influences on cartilage and bone development and skeletal homeostasis via versatile cell-cell interactions in an autocrine, paracrine, and endocrine manner systemically and locally. Our review summarizes the role and regulatory function as well as a potentially integrated gene network of the IGF signaling pathway with other signaling pathways in bone and cartilage development and skeletal homeostasis, which in turn provides an enlightening insight into visualizing bright molecular targets to be eligible for designing effective drugs to handle bone diseases and maladies, such as osteoporosis, osteoarthritis, and dwarfism.
Collapse
Affiliation(s)
- Xinyi Ruan
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiuhui Jin
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Fuju Sun
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jiashun Pi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yihan Jinghu
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xinyi Lin
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Nenghua Zhang
- Clinical Laboratory, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
27
|
Cannarella R, Rando OJ, Condorelli RA, Chamayou S, Romano S, Guglielmino A, Yin Q, Hans TG, Mancuso F, Arato I, Bellucci C, Luca G, Lundy SD, La Vignera S, Calogero AE. Sperm-carried IGF2: towards the discovery of a spark contributing to embryo growth and development. Mol Hum Reprod 2024; 30:gaae034. [PMID: 39312692 PMCID: PMC11975288 DOI: 10.1093/molehr/gaae034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/06/2024] [Indexed: 09/25/2024] Open
Abstract
Spermatozoa have been shown to carry key RNAs which, according to animal evidence, seem to play a role in early embryo development. In this context, a potential key growth regulator is insulin-like growth factor 2 (IGF2), a highly conserved paternally expressed imprinted gene involved in cell growth and proliferation which, recent observations indicate, is expressed in human spermatozoa. We herein hypothesized that sperm IGF2 gene expression and transmission at fertilization is required to support early embryo development. To test this hypothesis, we analyzed sperm IGF2 mRNA levels in the same semen aliquot used for homologous assisted reproductive technique (ART) in infertile couples and correlated these levels with embryo morphokinetics. To find a mechanistic explanation for the observed results, the transcriptomes of blastocysts obtained after injection of Igf2 mRNA in mouse parthenotes were analyzed. Sperm IGF2 mRNA negatively correlated with time of 2-cell stage (t2), t3, t4, t5, and time of expanded blastocyst (tEB), independently of maternal age, body mass index, anti-Müllerian hormone levels, and oocyte quality. An IGF2 mRNA index >4.9 predicted the ability of the embryos to reach the blastocyst stage on Day 5, with a sensitivity of 100% and a specificity of 71.6% (AUC 0.845; P < 0.001). In the animal study, transcriptome analysis demonstrated that 65 and 36 genes were, respectively, up- and down-regulated in the experimental group compared to the control group. These genes belong to pathways that regulate early embryo development, thus supporting the findings found in humans. This study has the potential to challenge the longstanding tenet that spermatozoa are simply vehicles carrying paternal DNA. Instead, it suggests that IGF2 mRNA in healthy spermatozoa provides critical support for early embryo development. Pre-ART sperm-carried IGF2 mRNA levels may be used as a marker to predict the chances of obtaining blastocysts to be transferred for infertile couples undergoing ART.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Oliver J Rando
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | | | - Simona Romano
- Centro HERA—Unità di Medicina della Riproduzione, Catania, Italy
| | | | - Qiangzong Yin
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Tobias Gustafsson Hans
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Francesca Mancuso
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Iva Arato
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Catia Bellucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giovanni Luca
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Scott D Lundy
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
28
|
Kuna M, Soares MJ. Cited2 is a key regulator of placental development and plasticity. Bioessays 2024; 46:e2300118. [PMID: 38922923 PMCID: PMC11331489 DOI: 10.1002/bies.202300118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
The biology of trophoblast cell lineage development and placentation is characterized by the involvement of several known transcription factors. Central to the action of a subset of these transcriptional regulators is CBP-p300 interacting transactivator with Glu/Asp-rich carboxy-terminal domain 2 (CITED2). CITED2 acts as a coregulator modulating transcription factor activities and affecting placental development and adaptations to physiological stressors. These actions of CITED2 on the trophoblast cell lineage and placentation are conserved across the mouse, rat, and human. Thus, aspects of CITED2 biology in hemochorial placentation can be effectively modeled in the mouse and rat. In this review, we present information on the conserved role of CITED2 in the biology of placentation and discuss the use of CITED2 as a tool to discover new insights into regulatory mechanisms controlling placental development.
Collapse
Affiliation(s)
- Marija Kuna
- Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Michael J. Soares
- Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy, Kansas City, MO
| |
Collapse
|
29
|
Antounians L, Figueira RL, Kukreja B, Litvack ML, Zani-Ruttenstock E, Khalaj K, Montalva L, Doktor F, Obed M, Blundell M, Wu T, Chan C, Wagner R, Lacher M, Wilson MD, Post M, Kalish BT, Zani A. Fetal hypoplastic lungs have multilineage inflammation that is reversed by amniotic fluid stem cell extracellular vesicle treatment. SCIENCE ADVANCES 2024; 10:eadn5405. [PMID: 39058789 PMCID: PMC11277482 DOI: 10.1126/sciadv.adn5405] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 06/21/2024] [Indexed: 07/28/2024]
Abstract
Antenatal administration of extracellular vesicles from amniotic fluid stem cells (AFSC-EVs) reverses features of pulmonary hypoplasia in models of congenital diaphragmatic hernia (CDH). However, it remains unknown which lung cellular compartments and biological pathways are affected by AFSC-EV therapy. Herein, we conducted single-nucleus RNA sequencing (snRNA-seq) on rat fetal CDH lungs treated with vehicle or AFSC-EVs. We identified that intra-amniotically injected AFSC-EVs reach the fetal lung in rats with CDH, where they promote lung branching morphogenesis and epithelial cell differentiation. Moreover, snRNA-seq revealed that rat fetal CDH lungs have a multilineage inflammatory signature with macrophage enrichment, which is reversed by AFSC-EV treatment. Macrophage enrichment in CDH fetal rat lungs was confirmed by immunofluorescence, flow cytometry, and inhibition studies with GW2580. Moreover, we validated macrophage enrichment in human fetal CDH lung autopsy samples. Together, this study advances knowledge on the pathogenesis of pulmonary hypoplasia and further evidence on the value of an EV-based therapy for CDH fetuses.
Collapse
Affiliation(s)
- Lina Antounians
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Rebeca Lopes Figueira
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Bharti Kukreja
- Neurosciences and Mental Health Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
| | - Michael L. Litvack
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
| | - Elke Zani-Ruttenstock
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Kasra Khalaj
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Louise Montalva
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Fabian Doktor
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Mikal Obed
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Matisse Blundell
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Taiyi Wu
- Neurosciences and Mental Health Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
| | - Cadia Chan
- Genetics and Genome Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, Canada
| | - Richard Wagner
- Department of Pediatric Surgery, Leipzig University, Leipzig 04109, Germany
| | - Martin Lacher
- Department of Pediatric Surgery, Leipzig University, Leipzig 04109, Germany
| | - Michael D. Wilson
- Genetics and Genome Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, Canada
| | - Martin Post
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5T 1P5, Canada
| | - Brian T. Kalish
- Neurosciences and Mental Health Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, Canada
- Division of Neonatology, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Augusto Zani
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
- Department of Surgery, University of Toronto, Toronto M5T 1P5, Canada
| |
Collapse
|
30
|
Wilson BM, Ko FC, Moran MM, Adra A, Rasmussen MB, Thymann T, Sangild PT, Sumner DR. Skeletal consequences of preterm birth in pigs as a model for preterm infants. J Bone Miner Res 2024; 39:791-803. [PMID: 38655758 PMCID: PMC11472249 DOI: 10.1093/jbmr/zjae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 03/18/2024] [Accepted: 04/22/2024] [Indexed: 04/26/2024]
Abstract
Preterm birth affects about 10% of all live births with many resultant health challenges, including metabolic bone disease of prematurity (MBDP), which is characterized by elevated alkaline phosphatase, suppressed phosphate, and deficient skeletal development. Because of the lack of an animal model, very little is known about bone structure, strength, and quality after preterm birth. This study investigated the utility of a pig model to replicate clinical features of preterm birth, including MBDP, and sought to determine if early postnatal administration of IGF-1 was an effective treatment. Preterm pigs, born by caesarean section at 90% gestation, were reared in intensive care facilities (respiratory, thermoregulatory, and nutritional support) and compared with sow-reared term pigs born vaginally. Preterm pigs were systemically treated with vehicle or IGF-1 (recombinant human IGF-1/BP-3, 2.25 mg/kg/d). Tissues were collected at postnatal days 1, 5, and 19 (the normal weaning period in pigs). Most bone-related outcomes were affected by preterm birth throughout the study period, whereas IGF-1 supplementation had almost no effect. By day 19, alkaline phosphatase was elevated, phosphate and calcium were reduced, and the bone resorption marker C-terminal crosslinks of type I collagen was elevated in preterm pigs compared to term pigs. Preterm pigs also had decrements in femoral cortical cross-sectional properties, consistent with reduced whole-bone strength. Thus, the preterm pig model replicates many features of preterm bone development in infants, including features of MBDP, and allows for direct interrogation of skeletal tissues, enhancing the field's ability to examine underlying mechanisms.
Collapse
Affiliation(s)
- Brittany M Wilson
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, 60612, United States
| | - Frank C Ko
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, 60612, United States
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, 60612, United States
| | - Meghan M Moran
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, 60612, United States
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, 60612, United States
| | - Amal Adra
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, 60612, United States
| | - Martin B Rasmussen
- Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, DK-1958, Denmark
| | - Thomas Thymann
- Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, DK-1958, Denmark
| | - Per T Sangild
- Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, DK-1958, Denmark
- Department of Neonatology, Rigshospitalet, Copenhagen, DK-2100, Denmark
- Department of Pediatrics, Odense University Hospital, Odense, DK-5000, Denmark
- Faculty of Theology, University of Copenhagen, Copenhagen, DK-2300, Denmark
| | - Dale Rick Sumner
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, 60612, United States
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, 60612, United States
| |
Collapse
|
31
|
Kawashima-Sonoyama Y, Wada K, Yamamoto K, Fujimoto M, Namba N, Taketani T. Clinical characteristics of and growth hormone treatment effects on short stature with type 1 insulin-like growth factor receptor (IGF1R) gene alteration. Endocr J 2024; 71:687-694. [PMID: 38710621 DOI: 10.1507/endocrj.ej23-0680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/08/2024] Open
Abstract
Short stature with IGF-1 receptor (IGF1R) gene alteration is known as small-for-gestational-age (SGA) short stature with elevated serum IGF1 levels. Its prevalence and clinical characteristics remain unclear. No adapted treatment is available for short stature related to IGF1R gene alteration in Japan, and genetic testing is not yet widely accessible. We investigated short stature with IGF1R gene alterations and analyzed the clinical data of 13 patients using the results of questionnaires issued to the Japanese Society for Pediatric Endocrinology. Four cases were caused by a deletion of chromosome 15q26.3, and eight were caused by heterozygous pathogenic variants in the IGF1R gene. Cases with deletions showed a more severe degree of growth impairment (-4.5 ± 0.43 SD) than those caused by pathological variants (-2.71 ± 0.15 SD) and were accompanied by neurodevelopmental delay. However, cases caused by pathological variants lacked distinctive features. Only three of the 12 cases demonstrated serum IGF1 values exceeding +2 SD, and the other three had values below 0 SD. Four patients did not meet the criteria for SGA at birth. Six patients received GH therapy for SGA short stature and showed improvement in growth rate without any side effects or elevated serum IGF1 levels during treatment. Elevated IGF1 levels (over +2 SD) after GH treatment should be considered a suspicious finding. Owing to the lack of distinctive features, there was a possibility of undiagnosed cases of this condition. Promoting genetic testing and clinical trials on GH administration for this condition is recommended.
Collapse
Affiliation(s)
| | - Keisuke Wada
- Department of Pediatrics, Shimane University Faculty of Medicine, Shimane 693-8501, Japan
| | - Kei Yamamoto
- Department of Pediatrics, Shimane University Faculty of Medicine, Shimane 693-8501, Japan
| | - Masanobu Fujimoto
- Division of Pediatrics & Perinatology, Tottori University Faculty of Medicine, Tottori 683-8504, Japan
| | - Noriyuki Namba
- Division of Pediatrics & Perinatology, Tottori University Faculty of Medicine, Tottori 683-8504, Japan
| | - Takeshi Taketani
- Department of Pediatrics, Shimane University Faculty of Medicine, Shimane 693-8501, Japan
| |
Collapse
|
32
|
Zhou H, Fick K, Patel V, Hilton LR, Kim HW, Bagi Z, Weintraub NL, Chen W. AGPAT3 deficiency impairs adipocyte differentiation and leads to a lean phenotype in mice. Am J Physiol Endocrinol Metab 2024; 327:E69-E80. [PMID: 38717361 PMCID: PMC11390115 DOI: 10.1152/ajpendo.00012.2024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/01/2024] [Accepted: 05/01/2024] [Indexed: 06/22/2024]
Abstract
Acylglycerophosphate acyltransferases (AGPATs) catalyze the de novo formation of phosphatidic acid to synthesize glycerophospholipids and triglycerides. AGPATs demonstrate unique physiological roles despite a similar biochemical function. AGPAT3 is highly expressed in the testis, kidney, and liver, with intermediate expression in adipose tissue. Loss of AGPAT3 is associated with reproductive abnormalities and visual dysfunction. However, the role of AGPAT3 in adipose tissue and whole body metabolism has not been investigated. We found that male Agpat3 knockout (KO) mice exhibited reduced body weights with decreased white and brown adipose tissue mass. Such changes were less pronounced in the female Agpat3-KO mice. Agpat3-KO mice have reduced plasma insulin growth factor 1 (IGF1) and insulin levels and diminished circulating lipid metabolites. They manifested intact glucose homeostasis and insulin sensitivity despite a lean phenotype. Agpat3-KO mice maintained an energy balance with normal food intake, energy expenditure, and physical activity, except for increased water intake. Their adaptive thermogenesis was also normal despite reduced brown adipose mass and triglyceride content. Mechanistically, Agpat3 was elevated during mouse and human adipogenesis and enriched in adipocytes. Agpat3-knockdown 3T3-L1 cells and Agpat3-deficient mouse embryonic fibroblasts (MEFs) have impaired adipogenesis in vitro. Interestingly, pioglitazone treatment rescued the adipogenic deficiency in Agpat3-deficient cells. We conclude that AGPAT3 regulates adipogenesis and adipose development. It is possible that adipogenic impairment in Agpat3-deficient cells potentially leads to reduced adipose mass. Findings from this work support the unique role of AGPAT3 in adipose tissue.NEW & NOTEWORTHY AGPAT3 deficiency results in male-specific growth retardation. It reduces adipose tissue mass but does not significantly impact glucose homeostasis or energy balance, except for influencing water intake in mice. Like AGPAT2, AGPAT3 is upregulated during adipogenesis, potentially by peroxisome proliferator-activated receptor gamma (PPARγ). Loss of AGPAT3 impairs adipocyte differentiation, which could be rescued by pioglitazone. Overall, AGPAT3 plays a significant role in regulating adipose tissue mass, partially involving its influence on adipocyte differentiation.
Collapse
Affiliation(s)
- Hongyi Zhou
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Kendra Fick
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Vijay Patel
- Department of Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Lisa Renee Hilton
- Department of Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Ha Won Kim
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Neal L Weintraub
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Weiqin Chen
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
33
|
Nagaya M, Uchikura A, Nakano K, Watanabe M, Matsunari H, Umeyama K, Mizuno N, Nishimura T, Nakauchi H, Nagashima H. Generation of insulin-like growth factor 1 receptor-knockout pigs as a potential system for interspecies organogenesis. Regen Ther 2024; 26:783-791. [PMID: 39309395 PMCID: PMC11416208 DOI: 10.1016/j.reth.2024.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND To overcome organ shortage during transplantation, interspecies organ generation via blastocyst complementation has been proposed, although not yet in evolutionarily distant species. To establish high levels of chimerism, low chimerism is required early in development, followed by high chimerism, to effectively complement the organ niche. Very few human cells are expected to contribute to chimerism in heterologous animals. Previous studies had demonstrated increased donor chimerism in both intra- and interspecies chimeras in rodents, using insulin-like growth factor 1 receptor (Igf1r) knockout (KO) mice; deletion of the Igf1r gene in the mouse host embryo created a cell-competitive niche. The current study aimed to generate IGF1R-KO pigs and evaluate whether they have the same phenotype as Igf1r-KO mice. METHODS To generate IGF1R-KO pigs, genome-editing molecules were injected into the cytoplasm of pig zygotes. The fetuses were evaluated at 104 days of gestation. RESULTS IGF1R-KO pigs were generated successfully. Their phenotypes were almost identical to those of Igf1r-KO mice, including small lungs and enlarged endodermal organs in fetuses, and they were highly reproducible. CONCLUSIONS Pigs may allow the generation of organs using blastocyst complementation with developmentally-compatible xenogeneic pluripotent stem cells over a large evolutionary distance.
Collapse
Affiliation(s)
- Masaki Nagaya
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
| | - Ayuko Uchikura
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
| | - Kazuaki Nakano
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
- PorMedTec Co. Ltd., 2-3227 Mita, Tama-ku, Kawasaki, Kanagawa, 214-0034, Japan
| | - Masahito Watanabe
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
- PorMedTec Co. Ltd., 2-3227 Mita, Tama-ku, Kawasaki, Kanagawa, 214-0034, Japan
| | - Hitomi Matsunari
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
- PorMedTec Co. Ltd., 2-3227 Mita, Tama-ku, Kawasaki, Kanagawa, 214-0034, Japan
| | - Kazuhiro Umeyama
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
- PorMedTec Co. Ltd., 2-3227 Mita, Tama-ku, Kawasaki, Kanagawa, 214-0034, Japan
| | - Naoaki Mizuno
- Division of Stem Cell Therapy, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
- Stem Cell Therapy Laboratory, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, 113-8510 Tokyo, Japan
| | - Toshiya Nishimura
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA94305, USA
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
- Stem Cell Therapy Laboratory, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, 113-8510 Tokyo, Japan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA94305, USA
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
- Laboratory of Developmental Engineering, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
- PorMedTec Co. Ltd., 2-3227 Mita, Tama-ku, Kawasaki, Kanagawa, 214-0034, Japan
| |
Collapse
|
34
|
Millward DJ. Post-natal muscle growth and protein turnover: a narrative review of current understanding. Nutr Res Rev 2024; 37:141-168. [PMID: 37395180 DOI: 10.1017/s0954422423000124] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
A model explaining the dietary-protein-driven post-natal skeletal muscle growth and protein turnover in the rat is updated, and the mechanisms involved are described, in this narrative review. Dietary protein controls both bone length and muscle growth, which are interrelated through mechanotransduction mechanisms with muscle growth induced both from stretching subsequent to bone length growth and from internal work against gravity. This induces satellite cell activation, myogenesis and remodelling of the extracellular matrix, establishing a growth capacity for myofibre length and cross-sectional area. Protein deposition within this capacity is enabled by adequate dietary protein and other key nutrients. After briefly reviewing the experimental animal origins of the growth model, key concepts and processes important for growth are reviewed. These include the growth in number and size of the myonuclear domain, satellite cell activity during post-natal development and the autocrine/paracrine action of IGF-1. Regulatory and signalling pathways reviewed include developmental mechanotransduction, signalling through the insulin/IGF-1-PI3K-Akt and the Ras-MAPK pathways in the myofibre and during mechanotransduction of satellite cells. Likely pathways activated by maximal-intensity muscle contractions are highlighted and the regulation of the capacity for protein synthesis in terms of ribosome assembly and the translational regulation of 5-TOPmRNA classes by mTORC1 and LARP1 are discussed. Evidence for and potential mechanisms by which volume limitation of muscle growth can occur which would limit protein deposition within the myofibre are reviewed. An understanding of how muscle growth is achieved allows better nutritional management of its growth in health and disease.
Collapse
Affiliation(s)
- D Joe Millward
- Department of Nutritional Sciences, School of Biosciences & Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
35
|
Moorwood K, Smith FM, Garfield AS, Ward A. Imprinted Grb10, encoding growth factor receptor bound protein 10, regulates fetal growth independently of the insulin-like growth factor type 1 receptor (Igf1r) and insulin receptor (Insr) genes. BMC Biol 2024; 22:127. [PMID: 38816743 PMCID: PMC11140863 DOI: 10.1186/s12915-024-01926-w] [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: 11/21/2023] [Accepted: 05/22/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Optimal size at birth dictates perinatal survival and long-term risk of developing common disorders such as obesity, type 2 diabetes and cardiovascular disease. The imprinted Grb10 gene encodes a signalling adaptor protein capable of inhibiting receptor tyrosine kinases, including the insulin receptor (Insr) and insulin-like growth factor type 1 receptor (Igf1r). Grb10 restricts fetal growth such that Grb10 knockout (KO) mice are at birth some 25-35% larger than wild type. Using a mouse genetic approach, we test the widely held assumption that Grb10 influences growth through interaction with Igf1r, which has a highly conserved growth promoting role. RESULTS Should Grb10 interact with Igf1r to regulate growth Grb10:Igf1r double mutant mice should be indistinguishable from Igf1r KO single mutants, which are around half normal size at birth. Instead, Grb10:Igf1r double mutants were intermediate in size between Grb10 KO and Igf1r KO single mutants, indicating additive effects of the two signalling proteins having opposite actions in separate pathways. Some organs examined followed a similar pattern, though Grb10 KO neonates exhibited sparing of the brain and kidneys, whereas the influence of Igf1r extended to all organs. An interaction between Grb10 and Insr was similarly investigated. While there was no general evidence for a major interaction for fetal growth regulation, the liver was an exception. The liver in Grb10 KO mutants was disproportionately overgrown with evidence of excess lipid storage in hepatocytes, whereas Grb10:Insr double mutants were indistinguishable from Insr single mutants or wild types. CONCLUSIONS Grb10 acts largely independently of Igf1r or Insr to control fetal growth and has a more variable influence on individual organs. Only the disproportionate overgrowth and excess lipid storage seen in the Grb10 KO neonatal liver can be explained through an interaction between Grb10 and the Insr. Our findings are important for understanding how positive and negative influences on fetal growth dictate size and tissue proportions at birth.
Collapse
Affiliation(s)
- Kim Moorwood
- Department of Life Sciences, University of Bath, Building 4 South, Claverton Down, Bath, BA2 7AY, United Kingdom
| | - Florentia M Smith
- Department of Life Sciences, University of Bath, Building 4 South, Claverton Down, Bath, BA2 7AY, United Kingdom
| | - Alastair S Garfield
- Department of Life Sciences, University of Bath, Building 4 South, Claverton Down, Bath, BA2 7AY, United Kingdom
| | - Andrew Ward
- Department of Life Sciences, University of Bath, Building 4 South, Claverton Down, Bath, BA2 7AY, United Kingdom.
| |
Collapse
|
36
|
Luo YE, Villani KR, Lei H, Kuo LY, Imery I, Stoker BE, Fatima N, Noles SM, Moore CM, Barton ER. Ablation of specific insulin-like growth factor I forms reveals the importance of cleavage for regenerative capacity and glycosylation for skeletal muscle storage. FASEB J 2024; 38:e23634. [PMID: 38679876 PMCID: PMC11107140 DOI: 10.1096/fj.202302512rr] [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: 12/05/2023] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 05/01/2024]
Abstract
Insulin-like growth factor-I (IGF-I) facilitates mitotic and anabolic actions in all tissues. In skeletal muscle, IGF-I can promote growth and resolution of damage by promoting satellite cell proliferation and differentiation, suppressing inflammation, and enhancing fiber formation. While the most well-characterized form of IGF-I is the mature protein, alternative splicing and post-translational modification complexity lead to several additional forms of IGF-I. Previous studies showed muscle efficiently stores glycosylated pro-IGF-I. However, non-glycosylated forms display more efficient IGF-I receptor activation in vitro, suggesting that the removal of the glycosylated C terminus is a necessary step to enable increased activity. We employed CRISPR-Cas9 gene editing to ablate IGF-I glycosylation sites (2ND) or its cleavage site (3RA) in mice to determine the necessity of glycosylation or cleavage for IGF-I function in postnatal growth and during muscle regeneration. 3RA mice had the highest circulating and muscle IGF-I content, whereas 2ND mice had the lowest levels compared to wild-type mice. After weaning, 4-week-old 2ND mice exhibited higher body and skeletal muscle mass than other strains. However, by 16 weeks of age, muscle and body size differences disappeared. Even though 3RA mice had more IGF-I stored in muscle in homeostatic conditions, regeneration was delayed after cardiotoxin-induced injury, with prolonged necrosis most evident at 5 days post injury (dpi). In contrast, 2ND displayed improved regeneration with reduced necrosis, and greater fiber size and muscle mass at 11 and 21 dpi. Overall, these results demonstrate that while IGF-I glycosylation may be important for storage, cleavage is needed to enable IGF-I to be used for efficient activity in postnatal growth and following acute injury.
Collapse
Affiliation(s)
- Yangyi E. Luo
- Applied Physiology and Kinesiology, University of Florida, Gainesville, FL USA
- Myology Institute, University of Florida, Gainesville, FL USA
| | - Katelyn R. Villani
- Applied Physiology and Kinesiology, University of Florida, Gainesville, FL USA
- Myology Institute, University of Florida, Gainesville, FL USA
| | - Hanqin Lei
- Applied Physiology and Kinesiology, University of Florida, Gainesville, FL USA
| | - Li-Ying Kuo
- Applied Physiology and Kinesiology, University of Florida, Gainesville, FL USA
| | - Ian Imery
- Applied Physiology and Kinesiology, University of Florida, Gainesville, FL USA
| | - Bradley E. Stoker
- Applied Physiology and Kinesiology, University of Florida, Gainesville, FL USA
| | - Naureen Fatima
- Applied Physiology and Kinesiology, University of Florida, Gainesville, FL USA
| | - Steven M. Noles
- Applied Physiology and Kinesiology, University of Florida, Gainesville, FL USA
| | - Cara M. Moore
- Animal Care Services, University of Florida, Gainesville, FL USA
| | - Elisabeth R. Barton
- Applied Physiology and Kinesiology, University of Florida, Gainesville, FL USA
- Myology Institute, University of Florida, Gainesville, FL USA
| |
Collapse
|
37
|
Douglas JC, Sekulovski N, Arreola MR, Oh Y, Hayashi K, MacLean JA. Normal Ovarian Function in Subfertile Mouse with Amhr2-Cre-Driven Ablation of Insr and Igf1r. Genes (Basel) 2024; 15:616. [PMID: 38790245 PMCID: PMC11121541 DOI: 10.3390/genes15050616] [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: 04/19/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Insulin receptor signaling promotes cell differentiation, proliferation, and growth which are essential for oocyte maturation, embryo implantation, endometrial decidualization, and placentation. The dysregulation of insulin signaling in women with metabolic syndromes including diabetes exhibits poor pregnancy outcomes that are poorly understood. We utilized the Cre/LoxP system to target the tissue-specific conditional ablation of insulin receptor (Insr) and insulin-like growth factor-1 receptor (Igf1r) using an anti-Mullerian hormone receptor 2 (Amhr2) Cre-driver which is active in ovarian granulosa and uterine stromal cells. Our long-term goal is to examine insulin-dependent molecular mechanisms that underlie diabetic pregnancy complications, and our conditional knockout models allow for such investigation without confounding effects of ligand identity, source and cross-reactivity, or global metabolic status within dams. Puberty occurred with normal timing in all conditional knockout models. Estrous cycles progressed normally in Insrd/d females but were briefly stalled in diestrus in Igf1rd/d and double receptor (DKO) mice. The expression of vital ovulatory genes (Lhcgr, Pgr, Ptgs2) was not significantly different in 12 h post-hCG superovulated ovaries in knockout mice. Antral follicles exhibited an elevated apoptosis of granulosa cells in Igf1rd/d and DKO mice. However, the distribution of ovarian follicle subtypes and subsequent ovulations was normal in all insulin receptor mutants compared to littermate controls. While ovulation was normal, all knockout lines were subfertile suggesting that the loss of insulin receptor signaling in the uterine stroma elicits implantation and decidualization defects responsible for subfertility in Amhr2-Cre-derived insulin receptor mutants.
Collapse
Affiliation(s)
- Jenna C. Douglas
- Center for Reproductive Biology, School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA; (J.C.D.)
| | - Nikola Sekulovski
- Department of Physiology, Southern Illinois University, Carbondale, IL 62901, USA
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Madison R. Arreola
- Center for Reproductive Biology, School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA; (J.C.D.)
| | - Yeongseok Oh
- Center for Reproductive Biology, School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA; (J.C.D.)
| | - Kanako Hayashi
- Center for Reproductive Biology, School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA; (J.C.D.)
| | - James A. MacLean
- Center for Reproductive Biology, School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA; (J.C.D.)
- Department of Physiology, Southern Illinois University, Carbondale, IL 62901, USA
| |
Collapse
|
38
|
Rasmussen MB, Holgersen K, Pankratova S, Bæk O, Burrin DG, Thymann T, Sangild PT. Gut development following insulin-like growth factor-1 supplementation to preterm pigs. Pediatr Res 2024; 95:1528-1535. [PMID: 38086951 PMCID: PMC11126387 DOI: 10.1038/s41390-023-02949-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/31/2023] [Accepted: 11/18/2023] [Indexed: 05/26/2024]
Abstract
BACKGROUND Reduced insulin-like growth factor-1 (IGF-1) levels may contribute to impaired organ development in preterm infants. Using preterm pigs as a model, we hypothesized that IGF-1 supplementation improves health and gut development during the first three weeks of life. METHODS First, clinical and organ endpoints were compared between artificially-reared, cesarean-delivered preterm pigs and vaginally-delivered, sow-reared term pigs at 5, 9 and 19 days. Next, preterm pigs were treated with recombinant human IGF-1 for 19 days (2.25 mg/kg/day, systemically). RESULTS Relative to term pigs, preterm pigs had lower body weight, fat, bone contents, relative weights of liver and spleen and a longer and thinner intestine at 19 days. Preterm birth reduced intestinal villi heights and peptidase activities, but only at 5 and 9 days. In preterm pigs, IGF-1 reduced mortality primarily occurring from gastrointestinal complications and with a tendency towards salvaging smaller pigs. IGF-1 supplementation also increased spleen and kidney weights, small intestine length and maltase to lactase activity, reflecting gut maturation. CONCLUSION Preterm birth affects body composition and gut maturation in the first 1-2 weeks, but differences are marginal thereafter. Supplemental IGF-1 may improve gut health in pigs and infants in the first few weeks after preterm birth. IMPACT Insulin-like growth factor 1 (IGF-1) supplementation may improve gut health and development in prematurity, but whether the effects are sustained beyond the immediate postnatal period is unclear. In preterm pigs, the prematurity effects on IGF-1 and gut health deficiencies are most pronounced during the first week of life and diminishes thereafter. In preterm pigs, IGF-1 supplementation beyond the first week of life reduced mortality. The present study provides evidence of a sustained effect of IGF-1 supplementation on the gastrointestinal tract after the immediate postnatal period.
Collapse
Affiliation(s)
- Martin Bo Rasmussen
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Kristine Holgersen
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark
| | - Stanislava Pankratova
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Ole Bæk
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Douglas G Burrin
- US Department of Agriculture/Agricultural Research Service and Department of Pediatrics, Baylor College of Medicine/Texas Children's Hospital, Children's Nutrition Research Center, Houston, TX, USA
| | - Thomas Thymann
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Per Torp Sangild
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark.
- Faculty of Theology, University of Copenhagen, Copenhagen, Denmark.
- Department of Neonatology, Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
39
|
Dai Y, Peng Y, Lu Z, Mao T, Chen K, Lu X, Liu K, Zhou X, Hu W, Wang H. Prenatal prednisone exposure impacts liver development and function in fetal mice and its characteristics. Toxicol Sci 2024; 199:63-80. [PMID: 38439560 DOI: 10.1093/toxsci/kfae027] [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] [Indexed: 03/06/2024] Open
Abstract
Prednisone, a widely used glucocorticoid drug in human and veterinary medicine, has been reported to cause developmental toxicity. However, systematic studies about the effect of prednisone on fetal liver development are still unclear. We investigated the potential effects of maternal exposure to clinically equivalent doses of prednisone during different gestational stages on cell proliferation and apoptosis, cell differentiation, glucose and lipid metabolism, and hematopoiesis in the liver of fetal mice, and explored the potential mechanisms. Results showed that prenatal prednisone exposure (PPE) could suppress cell proliferation, inhibit hepatocyte differentiation, and promote cholangiocyte differentiation in the fetal liver. Meanwhile, PPE could result in the enhancement of glyconeogenesis and bile acid synthesis and the inhibition of fatty acid β-oxidation and hematopoiesis in the fetal liver. Further analysis found that PPE-induced alterations in liver development had obvious stage and sex differences. Overall, the alteration in fetal liver development and function induced by PPE was most pronounced during the whole pregnancy (GD0-18), and the males were relatively more affected than the females. Additionally, fetal hepatic insulin-like growth factor 1 (IGF1) signaling pathway was inhibited by PPE. In conclusion, PPE could impact fetal liver development and multiple functions, and these alterations might be partially related to the inhibition of IGF1 signaling pathway.
Collapse
Affiliation(s)
- Yongguo Dai
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei Province 430071, China
| | - Yu Peng
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei Province 430071, China
| | - Zhengjie Lu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei Province 430071, China
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
| | - Tongyun Mao
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei Province 430071, China
| | - Kaiqi Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei Province 430071, China
| | - Xiaoqian Lu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei Province 430071, China
| | - Kexin Liu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei Province 430071, China
| | - Xinli Zhou
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei Province 430071, China
| | - Wen Hu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei Province 430071, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei Province 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei Province 430071, China
| |
Collapse
|
40
|
Höglund A, Henriksen R, Churcher AM, Guerrero-Bosagna CM, Martinez-Barrio A, Johnsson M, Jensen P, Wright D. The regulation of methylation on the Z chromosome and the identification of multiple novel Male Hyper-Methylated regions in the chicken. PLoS Genet 2024; 20:e1010719. [PMID: 38457441 PMCID: PMC10954189 DOI: 10.1371/journal.pgen.1010719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 03/20/2024] [Accepted: 01/31/2024] [Indexed: 03/10/2024] Open
Abstract
DNA methylation is a key regulator of eukaryote genomes, and is of particular relevance in the regulation of gene expression on the sex chromosomes, with a key role in dosage compensation in mammalian XY systems. In the case of birds, dosage compensation is largely absent, with it being restricted to two small Male Hyper-Methylated (MHM) regions on the Z chromosome. To investigate how variation in DNA methylation is regulated on the Z chromosome we utilised a wild x domestic advanced intercross in the chicken, with both hypothalamic methylomes and transcriptomes assayed in 124 individuals. The relatively large numbers of individuals allowed us to identify additional genomic MHM regions on the Z chromosome that were significantly differentially methylated between the sexes. These regions appear to down-regulate local gene expression in males, but not remove it entirely (unlike the lncRNAs identified in the initial MHM regions). These MHM regions were further tested and the most balanced genes appear to show decreased expression in males, whilst methylation appeared to be far more correlated with gene expression in the less balanced, as compared to the most balanced genes. In addition, quantitative trait loci (QTL) that regulate variation in methylation on the Z chromosome, and those loci that regulate methylation on the autosomes that derive from the Z chromosome were mapped. Trans-effect hotspots were also identified that were based on the autosomes but affected the Z, and also one that was based on the Z chromosome but that affected both autosomal and sex chromosome DNA methylation regulation. We show that both cis and trans loci that originate from the Z chromosome never exhibit an interaction with sex, whereas trans loci originating from the autosomes but affecting the Z chromosome always display such an interaction. Our results highlight how additional MHM regions are actually present on the Z chromosome, and they appear to have smaller-scale effects on gene expression in males. Quantitative variation in methylation is also regulated both from the autosomes to the Z chromosome, and from the Z chromosome to the autosomes.
Collapse
Affiliation(s)
- Andrey Höglund
- Science for Life Laboratory, Department of Environmental Science, Stockholm University, Stockholm, Sweden
| | - Rie Henriksen
- AVIAN Behavioural Genomics and Physiology Group, Linköping University, Linköping, Sweden
| | | | - Carlos M. Guerrero-Bosagna
- Physiology and Environmental Toxicology Program, Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| | | | - Martin Johnsson
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Per Jensen
- AVIAN Behavioural Genomics and Physiology Group, Linköping University, Linköping, Sweden
| | - Dominic Wright
- AVIAN Behavioural Genomics and Physiology Group, Linköping University, Linköping, Sweden
| |
Collapse
|
41
|
Qiao L, Lu C, Zang T, Dzyuba B, Shao J. Maternal GLP-1 receptor activation inhibits fetal growth. Am J Physiol Endocrinol Metab 2024; 326:E268-E276. [PMID: 38197791 PMCID: PMC11193516 DOI: 10.1152/ajpendo.00361.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 01/11/2024]
Abstract
Glucagon-like peptide 1 (GLP-1) regulates food intake, insulin production, and metabolism. Our recent study demonstrated that pancreatic α-cells-secreted (intraislet) GLP-1 effectively promotes maternal insulin secretion and metabolic adaptation during pregnancy. However, the role of circulating GLP-1 in maternal energy metabolism remains largely unknown. Our study aims to investigate systemic GLP-1 response to pregnancy and its regulatory effect on fetal growth. Using C57BL/6 mice, we observed a gradual decline in maternal blood GLP-1 concentrations. Subsequent administration of the GLP-1 receptor agonist semaglutide (Sem) to dams in late pregnancy revealed a modest decrease in maternal food intake during initial treatment. At the same time, no significant alterations were observed in maternal body weight or fat mass. Notably, Sem-treated dams exhibited a significant decrease in fetal body weight, which persisted even following the restoration of maternal blood glucose levels. Despite no observable change in placental weight, a marked reduction in the placenta labyrinth area from Sem-treated dams was evident. Our investigation further demonstrated a substantial decrease in the expression levels of various pivotal nutrient transporters within the placenta, including glucose transporter one and sodium-neutral amino acid transporter one, after Sem treatment. In addition, Sem injection led to a notable reduction in the capillary area, number, and surface densities within the labyrinth. These findings underscore the crucial role of modulating circulating GLP-1 levels in maternal adaptation, emphasizing the inhibitory effects of excessive GLP-1 receptor activation on both placental development and fetal growth.NEW & NOTEWORTHY Our study reveals a progressive decline in maternal blood glucagon-like peptide 1 (GLP-1) concentration. GLP-1 receptor agonist injection in late pregnancy significantly reduced fetal body weight, even after restoration of maternal blood glucose concentration. GLP-1 receptor activation significantly reduced the placental labyrinth area, expression of some nutrient transporters, and capillary development. Our study indicates that reducing maternal blood GLP-1 levels is a physiological adaptation process that benefits placental development and fetal growth.
Collapse
Affiliation(s)
- Liping Qiao
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States
| | - Cindy Lu
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States
| | - Tianyi Zang
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States
| | - Brianna Dzyuba
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States
| | - Jianhua Shao
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States
| |
Collapse
|
42
|
Hann SH, Kim SY, Kim YL, Jo YW, Kang JS, Park H, Choi SY, Kong YY. Depletion of SMN protein in mesenchymal progenitors impairs the development of bone and neuromuscular junction in spinal muscular atrophy. eLife 2024; 12:RP92731. [PMID: 38318851 PMCID: PMC10945524 DOI: 10.7554/elife.92731] [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] [Indexed: 02/07/2024] Open
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disorder characterized by the deficiency of the survival motor neuron (SMN) protein, which leads to motor neuron dysfunction and muscle atrophy. In addition to the requirement for SMN in motor neurons, recent studies suggest that SMN deficiency in peripheral tissues plays a key role in the pathogenesis of SMA. Using limb mesenchymal progenitor cell (MPC)-specific SMN-depleted mouse models, we reveal that SMN reduction in limb MPCs causes defects in the development of bone and neuromuscular junction (NMJ). Specifically, these mice exhibited impaired growth plate homeostasis and reduced insulin-like growth factor (IGF) signaling from chondrocytes, rather than from the liver. Furthermore, the reduction of SMN in fibro-adipogenic progenitors (FAPs) resulted in abnormal NMJ maturation, altered release of neurotransmitters, and NMJ morphological defects. Transplantation of healthy FAPs rescued the morphological deterioration. Our findings highlight the significance of mesenchymal SMN in neuromusculoskeletal pathogenesis of SMA and provide insights into potential therapeutic strategies targeting mesenchymal cells for the treatment of SMA.
Collapse
Affiliation(s)
- Sang-Hyeon Hann
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seon-Yong Kim
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Ye Lynne Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young-Woo Jo
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jong-Seol Kang
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyerim Park
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Se-Young Choi
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Young-Yun Kong
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
43
|
Estrella CAS, Gatford KL, Xiang R, Javadmanesh A, Ghanipoor-Samami M, Nattrass GS, Shuaib E, McAllister MM, Beckman I, Thomsen DA, Clifton VL, Owens JA, Roberts CT, Hiendleder S, Kind KL. Asymmetric growth-limiting development of the female conceptus. Front Endocrinol (Lausanne) 2024; 14:1306513. [PMID: 38362586 PMCID: PMC10867182 DOI: 10.3389/fendo.2023.1306513] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/08/2023] [Indexed: 02/17/2024] Open
Abstract
Introduction Sex differences in prenatal growth may contribute to sex-dependent programming effects on postnatal phenotype. Methods We integrated for the first time phenotypic, histomorphological, clinico-chemical, endocrine and gene expression analyses in a single species, the bovine conceptus at mid-gestation. Results We demonstrate that by mid-gestation, before the onset of accelerated growth, the female conceptus displays asymmetric lower growth compared to males. Female fetuses were smaller with lower ponderal index and organ weights than males. However, their brain:body weight, brain:liver weight and heart:body weight ratios were higher than in males, indicating brain and heart 'sparing'. The female placenta weighed less and had lower volumes of trophoblast and fetal connective tissue than the male placenta. Female umbilical cord vessel diameters were smaller, and female-specific relationships of body weight and brain:liver weight ratios with cord vessel diameters indicated that the umbilico-placental vascular system creates a growth-limiting environment where blood flow is redistributed to protect brain and heart growth. Clinico-chemical indicators of liver perfusion support this female-specific growth-limiting phenotype, while lower insulin-like growth factor 2 (IGF2) gene expression in brain and heart, and lower circulating IGF2, implicate female-specific modulation of key endocrine mediators by nutrient supply. Conclusion This mode of female development may increase resilience to environmental perturbations in utero and contribute to sex-bias in programming outcomes including susceptibility to non-communicable diseases.
Collapse
Affiliation(s)
- Consuelo Amor S. Estrella
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Epigenetics and Genetics Group and Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Kathryn L. Gatford
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Ruidong Xiang
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Epigenetics and Genetics Group and Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Ali Javadmanesh
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Epigenetics and Genetics Group and Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Mani Ghanipoor-Samami
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Epigenetics and Genetics Group and Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Greg S. Nattrass
- South Australian Research and Development Institute, Livestock Systems, Roseworthy, SA, Australia
| | - Entesar Shuaib
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Epigenetics and Genetics Group and Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Milton M. McAllister
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Ian Beckman
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Dana A. Thomsen
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Epigenetics and Genetics Group and Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Vicki L. Clifton
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Mater Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Julie A. Owens
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Deakin University, Geelong, VIC, Australia
| | - Claire T. Roberts
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Flinders University, College of Medicine and Public Health, Adelaide, SA, Australia
| | - Stefan Hiendleder
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Epigenetics and Genetics Group and Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - Karen L. Kind
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Epigenetics and Genetics Group and Davies Research Centre, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| |
Collapse
|
44
|
Zhou W, Zhang N, Huang K, Lin H, Tu J, Zheng C, Que P, Chiang CY, Martinez J, Naerhulan H, Székely T, Zhang Z, Liu Y. Divergent Selection in Low Recombination Regions Shapes the Genomic Islands in Two Incipient Shorebird Species. Mol Biol Evol 2024; 41:msae006. [PMID: 38225175 PMCID: PMC10835341 DOI: 10.1093/molbev/msae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 01/17/2024] Open
Abstract
Speciation in the face of gene flow is usually associated with a heterogeneous genomic landscape of divergence in nascent species pairs. However, multiple factors, such as divergent selection and local recombination rate variation, can influence the formation of these genomic islands. Examination of the genomic landscapes of species pairs that are still in the early stages of speciation provides an insight into this conundrum. In this study, population genomic analyses were undertaken using a wide range of sampling and whole-genome resequencing data from 96 unrelated individuals of Kentish plover (Charadrius alexandrinus) and white-faced plover (Charadrius dealbatus). We suggest that the two species exhibit varying levels of population admixture along the Chinese coast and on the Taiwan Island. Genome-wide analyses for introgression indicate that ancient introgression had occurred in Taiwan population, and gene flow is still ongoing in mainland coastal populations. Furthermore, we identified a few genomic regions with significant levels of interspecific differentiation and local recombination suppression, which contain several genes potentially associated with disease resistance, coloration, and regulation of plumage molting and thus may be relevant to the phenotypic and ecological divergence of the two nascent species. Overall, our findings suggest that divergent selection in low recombination regions may be a main force in shaping the genomic islands in two incipient shorebird species.
Collapse
Affiliation(s)
- Wenjun Zhou
- State Key Laboratory of Biocontrol, School of Ecology, Sun Yat-sen University, Guangzhou, China
- Department of Zoology, University of Cambridge, Cambridge, UK
| | - Nan Zhang
- State Key Laboratory of Biocontrol, School of Ecology, Sun Yat-sen University, Guangzhou, China
| | - Kaichi Huang
- Department of Botany and Biodiversity Research Centre, University of British Columbia, Vancouver, Bc, Canada
| | - Hongzhou Lin
- State Key Laboratory of Biocontrol, School of Ecology, Sun Yat-sen University, Guangzhou, China
| | - Jie Tu
- State Key Laboratory of Biocontrol, School of Ecology, Sun Yat-sen University, Guangzhou, China
| | - Chenqing Zheng
- State Key Laboratory of Biocontrol, School of Ecology, Sun Yat-sen University, Guangzhou, China
| | - Pinjia Que
- Ministry of Education Key Laboratory for Biodiversity Sciences and Ecological Engineering, College of Life Sciences, Beijing Normal University, Beijing, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu Research Base of Giant Panda Breeding, Sichuan Academy of Giant Panda, Chengdu, China
| | - Chung-Yu Chiang
- Department of Environmental Science, Tunghai University, Taichung, Taiwan, China
| | | | - Halimubieke Naerhulan
- Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, UK
| | - Tamás Székely
- State Key Laboratory of Biocontrol, School of Ecology, Sun Yat-sen University, Guangzhou, China
- Ministry of Education Key Laboratory for Biodiversity Sciences and Ecological Engineering, College of Life Sciences, Beijing Normal University, Beijing, China
- Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, UK
| | - Zhengwang Zhang
- Ministry of Education Key Laboratory for Biodiversity Sciences and Ecological Engineering, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yang Liu
- State Key Laboratory of Biocontrol, School of Ecology, Sun Yat-sen University, Guangzhou, China
- Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, UK
| |
Collapse
|
45
|
Sarver DC, Garcia-Diaz J, Saqib M, Riddle RC, Wong GW. Tmem263 deletion disrupts the GH/IGF-1 axis and causes dwarfism and impairs skeletal acquisition. eLife 2024; 12:RP90949. [PMID: 38241182 PMCID: PMC10945605 DOI: 10.7554/elife.90949] [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] [Indexed: 01/21/2024] Open
Abstract
Genome-wide association studies (GWAS) have identified a large number of candidate genes believed to affect longitudinal bone growth and bone mass. One of these candidate genes, TMEM263, encodes a poorly characterized plasma membrane protein. Single nucleotide polymorphisms in TMEM263 are associated with bone mineral density in humans and mutations are associated with dwarfism in chicken and severe skeletal dysplasia in at least one human fetus. Whether this genotype-phenotype relationship is causal, however, remains unclear. Here, we determine whether and how TMEM263 is required for postnatal growth. Deletion of the Tmem263 gene in mice causes severe postnatal growth failure, proportional dwarfism, and impaired skeletal acquisition. Mice lacking Tmem263 show no differences in body weight within the first 2 weeks of postnatal life. However, by P21 there is a dramatic growth deficit due to a disrupted growth hormone (GH)/insulin-like growth factor 1 (IGF-1) axis, which is critical for longitudinal bone growth. Tmem263-null mice have low circulating IGF-1 levels and pronounced reductions in bone mass and growth plate length. The low serum IGF-1 in Tmem263-null mice is associated with reduced hepatic GH receptor (GHR) expression and GH-induced JAK2/STAT5 signaling. A deficit in GH signaling dramatically alters GH-regulated genes and feminizes the liver transcriptome of Tmem263-null male mice, with their expression profile resembling wild-type female, hypophysectomized male, and Stat5b-null male mice. Collectively, our data validates the causal role for Tmem263 in regulating postnatal growth and raises the possibility that rare mutations or variants of TMEM263 may potentially cause GH insensitivity and impair linear growth.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Jean Garcia-Diaz
- Department of Orthopaedic Surgery, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Orthopaedics, University of Maryland School of MedicineBaltimoreUnited States
- Cell and Molecular Medicine graduate program, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Muzna Saqib
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Ryan C Riddle
- Department of Orthopaedic Surgery, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Orthopaedics, University of Maryland School of MedicineBaltimoreUnited States
- Research and Development Service, Baltimore Veterans Administration Medical CenterBaltimoreUnited States
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
46
|
Choi JW, Kim SW, Kim HS, Kang MJ, Kim SA, Han JY, Kim H, Ku SY. Effects of Melatonin, GM-CSF, IGF-1, and LIF in Culture Media on Embryonic Development: Potential Benefits of Individualization. Int J Mol Sci 2024; 25:751. [PMID: 38255823 PMCID: PMC10815572 DOI: 10.3390/ijms25020751] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
The implantation of good-quality embryos to the receptive endometrium is essential for successful live birth through in vitro fertilization (IVF). The higher the quality of embryos, the higher the live birth rate per cycle, and so efforts have been made to obtain as many high-quality embryos as possible after fertilization. In addition to an effective controlled ovarian stimulation process to obtain high-quality embryos, the composition of the embryo culture medium in direct contact with embryos in vitro is also important. During embryonic development, under the control of female sex hormones, the fallopian tubes and endometrium create a microenvironment that supplies the nutrients and substances necessary for embryos at each stage. During this process, the development of the embryo is finely regulated by signaling molecules, such as growth factors and cytokines secreted from the epithelial cells of the fallopian tube and uterine endometrium. The development of embryo culture media has continued since the first successful human birth through IVF in 1978. However, there are still limitations to mimicking a microenvironment similar to the reproductive organs of women suitable for embryo development in vitro. Efforts have been made to overcome the harsh in vitro culture environment and obtain high-quality embryos by adding various supplements, such as antioxidants and growth factors, to the embryo culture medium. Recently, there has been an increase in the number of studies on the effect of supplementation in different clinical situations such as old age, recurrent implantation failure (RIF), and unexplained infertility; in addition, anticipation of the potential benefits from individuation is rising. This article reviews the effects of representative supplements in culture media on embryo development.
Collapse
Affiliation(s)
- Jung-Won Choi
- Laboratory of In Vitro Fertilization, Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (J.-W.C.); (H.-S.K.); (M.-J.K.); (S.-A.K.)
| | - Sung-Woo Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (S.-W.K.); (J.-Y.H.); (H.K.)
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hee-Sun Kim
- Laboratory of In Vitro Fertilization, Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (J.-W.C.); (H.-S.K.); (M.-J.K.); (S.-A.K.)
| | - Moon-Joo Kang
- Laboratory of In Vitro Fertilization, Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (J.-W.C.); (H.-S.K.); (M.-J.K.); (S.-A.K.)
| | - Sung-Ah Kim
- Laboratory of In Vitro Fertilization, Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (J.-W.C.); (H.-S.K.); (M.-J.K.); (S.-A.K.)
| | - Ji-Yeon Han
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (S.-W.K.); (J.-Y.H.); (H.K.)
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hoon Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (S.-W.K.); (J.-Y.H.); (H.K.)
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (S.-W.K.); (J.-Y.H.); (H.K.)
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea
| |
Collapse
|
47
|
Dobbins TW, Swanson RM, Dennis AA, Rivera JD, Dinh TTN, Lemley CO, Burnett DD. Melatonin supplementation to sows in mid to late gestation affects offspring circadian, myogenic, and growth factor transcript abundance in pre and postnatal skeletal muscle. J Anim Sci 2024; 102:skae377. [PMID: 39679952 DOI: 10.1093/jas/skae377] [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: 05/21/2024] [Accepted: 12/15/2024] [Indexed: 12/17/2024] Open
Abstract
The neuroendocrine hormone melatonin is associated with circadian rhythms and has antioxidant and vasodilative properties. In cattle, melatonin rescues fetal growth during maternal nutrient restriction in a seasonally dependent manner, but melatonin research in swine is limited. The objective of this study was to evaluate the effects of dietary melatonin supplementation during mid to late gestation on circadian rhythm and muscle growth and development of the longissimus dorsi in utero and postnatally. Sows received 20 mg of dietary melatonin daily (MEL) or no melatonin supplement (CON). Experiment 1 supplemented sows from gestational age (dGA) 38 ± 1 to 99 ± 1, experiment 2 supplemented sows from 41 to 106 ± 1 dGA, and experiment 3 supplemented sows from 60 dGA to farrowing. At harvest, morphometric measurements of all fetuses were taken, while the small (SM), medium (MED), and large (LG) piglets from each litter were used for further analysis. Prenatal data were analyzed using the MIXED procedure of SAS, and postnatal data were analyzed using the GLIMMIX procedure. Fetal morphometrics were analyzed for fixed the effect of treatment, and transcript abundance was analyzed for treatment, time, and size. Postnatal parameters were analyzed for fixed effects of treatment, size, and production stage. In experiment 1, MEL increased (P = 0.016) Period 1 (PER1) transcript abundance in the evening (PM) compared to the morning (AM). In experiment 1, myogenin (MYOG) transcript abundance was increased (P = 0.033) in MEL fetuses in the AM compared to MEL in the PM. Myogenic factor 5 (MYF5) and paired box 7 (PAX7) were increased (P = 0.016) in the PM. Fetuses from MEL-treated sows had increased (P < 0.05) BW, curve crown-rump length, and head circumference in experiment 2. In experiment 2, MEL increased (P = 0.012) PER1 and Period 2 (PER2) transcript abundance in the PM. In experiment 2, myoblast differentiation 1 (MYOD) was increased (P = 0.016) in SM and MED fetuses, while MYF5 and PAX7 were increased (P = 0.019) in SM fetuses. Postnatal BW was increased (P = 0.025) in MED and LG MEL-treated offspring compared to CON. Insulin-like growth factor 1 (IGF1) was downregulated (P = 0.050) in MEL-treated offspring, while insulin-like growth factor 1 receptor (IGF1R) was upregulated (P = 0.009) in MEL offspring. These results indicate that maternal melatonin supplementation during gestation modulates fetal circadian regulatory genes and alters myogenic genes during growth.
Collapse
Affiliation(s)
- Thomas W Dobbins
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA
| | - Rebecca M Swanson
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA
| | - Amberly A Dennis
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA
| | - J Daniel Rivera
- Department of Animal Science, University of Arkansas System Division of Agriculture, Southwest Research and Extension Center, Hope, AR 71801, USA
| | - Thu T N Dinh
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA
| | - Caleb O Lemley
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA
| | - Derris D Burnett
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA
| |
Collapse
|
48
|
Gao K, Han S, Li Z, Luo Z, Lv S, Choe HM, Paek HJ, Quan B, Kang J, Yin X. Analysis of metabolome and transcriptome of longissimus thoracis and subcutaneous adipose tissues reveals the regulatory mechanism of meat quality in MSTN mutant castrated male finishing pigs. Meat Sci 2024; 207:109370. [PMID: 37864922 DOI: 10.1016/j.meatsci.2023.109370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 08/21/2023] [Accepted: 10/13/2023] [Indexed: 10/23/2023]
Abstract
The underlying mechanism of myostatin (MSTN) gene mutation impact on porcine carcass and meat quality has not yet been fully understood. The meat quality trait testing of the second filial generation wild-type (WT) and homozygous MSTN mutant (MSTN-/-) castrated male finishing pigs, and RNA-seq and metabolomics on the longissimus thoracis (LT) and subcutaneous adipose tissues (SAT) were performed. Compared with WT pigs, MSTN-/- pigs had higher carcass lean percentage and lower backfat thickness (all P < 0.01), and also had lower shear force (P < 0.01) and meat redness (P < 0.05). The gene and metabolite expression profiles were different between two groups. Metabolites and genes related to purine metabolism (such as xanthine metabolite (P < 0.05), AMPD3 and XDH genes (all padj < 0.01)), PI3K/Akt/mTOR signaling pathway (such as Phe-Phe and Glu-Glu metabolites (all P < 0.05), WNT4 and AKT2 genes (all padj < 0.01)), antioxidant related pathway (such as GPX2, GPX3, and GPX7 genes (all padj < 0.01)), and extracellular matrix related pathway (such as COL1A1 and COL3A1 genes (all padj < 0.01)) were significantly altered in LT. While metabolites and genes associated to lipid metabolism (such as trans-elaidic acid and PE(18:1(9Z)/0:0) metabolites (all P < 0.05), ACOX1, ACAT1 and HADH genes (all padj < 0.01)) were significantly changed in SAT. This study revealed the biological mechanisms of homozygous MSTN mutation regulated porcine carcass and meat quality, such as lean meat percentage, fat deposition and tenderness, which provides reference for the utilization of MSTN-/- pigs.
Collapse
Affiliation(s)
- Kai Gao
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Yanbian University, Yanji 133002, China; Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji 133002, China
| | - Shengzhong Han
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji 133002, China
| | - Zhouyan Li
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji 133002, China
| | - Zhaobo Luo
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji 133002, China
| | - Sitong Lv
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Yanbian University, Yanji 133002, China; Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji 133002, China
| | - Hak Myong Choe
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji 133002, China
| | - Hyo Jin Paek
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji 133002, China
| | - Biaohu Quan
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji 133002, China
| | - Jindan Kang
- Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji 133002, China
| | - Xijun Yin
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Yanbian University, Yanji 133002, China; Jilin Provincial Key Laboratory of Transgenic Animal and Embryo Engineering, Yanbian University, Yanji 133002, China.
| |
Collapse
|
49
|
Bartke A, Hascup E, Hascup K. Responses to Many Anti-Aging Interventions Are Sexually Dimorphic. World J Mens Health 2024; 42:29-38. [PMID: 37118966 PMCID: PMC10782120 DOI: 10.5534/wjmh.230015] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/23/2023] [Accepted: 02/28/2023] [Indexed: 04/30/2023] Open
Abstract
There is increasing appreciation that sex differences are not limited to reproductive organs or traits related to reproduction and that sex is an important biological variable in most characteristics of a living organism. The biological process of aging and aging-related traits are no exception and exhibit numerous, often major, sex differences. This article explores one aspect of these differences, namely sex differences in the responses to anti-aging interventions. Aging can be slowed down and/or postponed by a variety of environmental ("lifestyle"), genetic or pharmacological interventions. Although many, particularly older studies utilized only one sex of experimental animals, there is considerable evidence that responses to these interventions can be very different in females and males. Calorie restriction (CR), that is reducing food intake without malnutrition can extend longevity in both sexes, but specific metabolic alterations and health benefits induced by CR are not the same in women and men. In laboratory mice, several of the genetic alterations that reduce insulin-like growth factor I (IGF-1) signaling extend longevity more effectively in females or in females only. Beneficial effects of rapamycin, an inhibitor of mTOR signaling, on mouse longevity are greater in females. In contrast, several anti-aging compounds, including a weak estrogen, 17 alpha estradiol, extend longevity of male, but not female, mice. Apparently, fundamental mechanisms of aging are not identical in females and males and it is essential to use both sexes in studies aimed at identifying novel anti-aging interventions. Recommendations for lifestyle modifications, drugs, and dietary supplements to maintain good health and functionality into advanced age and to live longer will likely need to be tailored to the sex of the user.
Collapse
Affiliation(s)
- Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA.
| | - Erin Hascup
- Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Department of Neurology, Neurosciences Institute, Southern Illinois University School of Medicine, Springfield, IL, USA
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin Hascup
- Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Department of Neurology, Neurosciences Institute, Southern Illinois University School of Medicine, Springfield, IL, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
50
|
Hayes E, Winston N, Stocco C. Molecular crosstalk between insulin-like growth factors and follicle-stimulating hormone in the regulation of granulosa cell function. Reprod Med Biol 2024; 23:e12575. [PMID: 38571513 PMCID: PMC10988955 DOI: 10.1002/rmb2.12575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 04/05/2024] Open
Abstract
Background The last phase of folliculogenesis is driven by follicle-stimulating hormone (FSH) and locally produced insulin-like growth factors (IGFs), both essential for forming preovulatory follicles. Methods This review discusses the molecular crosstalk of the FSH and IGF signaling pathways in regulating follicular granulosa cells (GCs) during the antral-to-preovulatory phase. Main findings IGFs were considered co-gonadotropins since they amplify FSH actions in GCs. However, this view is not compatible with data showing that FSH requires IGFs to stimulate GCs, that FSH renders GCs sensitive to IGFs, and that FSH signaling interacts with factors downstream of AKT to stimulate GCs. New evidence suggests that FSH and IGF signaling pathways intersect at several levels to regulate gene expression and GC function. Conclusion FSH and locally produced IGFs form a positive feedback loop essential for preovulatory follicle formation in all species. Understanding the mechanisms by which FSH and IGFs interact to control GC function will help design new interventions to optimize follicle maturation, perfect treatment of ovulatory defects, improve in vitro fertilization, and develop new contraceptive approaches.
Collapse
Affiliation(s)
- Emily Hayes
- Department of Physiology and BiophysicsUniversity of Illinois Chicago College of MedicineChicagoIllinoisUSA
| | - Nicola Winston
- Department of Obstetrics and GynecologyUniversity of Illinois Chicago College of MedicineChicagoIllinoisUSA
| | - Carlos Stocco
- Department of Physiology and BiophysicsUniversity of Illinois Chicago College of MedicineChicagoIllinoisUSA
- Department of Obstetrics and GynecologyUniversity of Illinois Chicago College of MedicineChicagoIllinoisUSA
| |
Collapse
|