1
|
Cheng Y, Dang S, Zhang Y, Chen Y, Yu R, Liu M, Jin S, Han A, Katz S, Wang S. Sequencing-free whole genome spatial transcriptomics at molecular resolution in intact tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641951. [PMID: 40161724 PMCID: PMC11952344 DOI: 10.1101/2025.03.06.641951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Recent breakthroughs in spatial transcriptomics technologies have enhanced our understanding of diverse cellular identities, compositions, interactions, spatial organizations, and functions. Yet existing spatial transcriptomics tools are still limited in either transcriptomic coverage or spatial resolution. Leading spatial-capture or spatial-tagging transcriptomics techniques that rely on in-vitro sequencing offer whole-transcriptome coverage, in principle, but at the cost of lower spatial resolution compared to image-based techniques. In contrast, high-performance image-based spatial transcriptomics techniques, which rely on in situ hybridization or in situ sequencing, achieve single-molecule spatial resolution and retain sub-cellular morphologies, but are limited by probe libraries that target only a subset of the transcriptome, typically covering several hundred to a few thousand transcript species. Together, these limitations hinder unbiased, hypothesis-free transcriptomic analyses at high spatial resolution. Here we develop a new image-based spatial transcriptomics technology termed Reverse-padlock Amplicon Encoding FISH (RAEFISH) with whole-genome level coverage while retaining single-molecule spatial resolution in intact tissues. We demonstrate image-based spatial transcriptomics targeting 23,000 human transcript species or 22,000 mouse transcript species, including nearly the entire protein-coding transcriptome and several thousand long-noncoding RNAs, in single cells in cultures and in tissue sections. Our analyses reveal differential subcellular localizations of diverse transcripts, cell-type-specific and cell-type-invariant tissue zonation dependent transcriptome, and gene expression programs underlying preferential cell-cell interactions. Finally, we further develop our technology for direct spatial readout of gRNAs in an image-based high-content CRISPR screen. Overall, these developments provide the research community with a broadly applicable technology that enables high-coverage, high-resolution spatial profiling of both long and short, native and engineered RNA species in many biomedical contexts.
Collapse
Affiliation(s)
- Yubao Cheng
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shengyuan Dang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
- These authors contributed equally to this work
| | - Yuan Zhang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
- These authors contributed equally to this work
| | - Yanbo Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Ruihuan Yu
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
- Present Address: Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Miao Liu
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shengyan Jin
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Ailin Han
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Samuel Katz
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Siyuan Wang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
- M.D.-Ph.D. Program, Yale University, New Haven, CT 06510, USA
- Yale Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT 06510, USA
- Molecular Cell Biology, Genetics and Development Program, Yale University, New Haven, CT 06510, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
- Biochemistry, Quantitative Biology, Biophysics, and Structural Biology Program, Yale University, New Haven, CT 06510, USA
- Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Liver Center, Yale University School of Medicine, New Haven, CT 06510, USA
- Lead contact
| |
Collapse
|
2
|
Murvai VR, Radu CM, Galiș R, Ghitea TC, Tătaru-Copos AF, Vesa AA, Huniadi A. The Relationship Between Thrombophilia and Modifications in First-Trimester Prenatal Screening Markers. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:318. [PMID: 40005435 PMCID: PMC11857445 DOI: 10.3390/medicina61020318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025]
Abstract
Background and Objective: Thrombophilia significantly increases the risk of complications like recurrent pregnancy loss, preeclampsia, IUGR, and stillbirth. Objective: This study aimed to evaluate the impact of inherited thrombophilic mutations on first-trimester screening outcomes, focusing on their relationship with maternal biomarkers and ultrasonographic parameters. Materials and Methods: A prospective observational study was conducted on 105 pregnant women during the first trimester (10-13 weeks of gestation). Genetic testing identified common thrombophilic mutations, including factor V Leiden, prothrombin G20210A, and MTHFR polymorphisms. First-trimester screening parameters, including PAPP-A, free β-hCG, and nuchal translucency (NT), were assessed. Maternal demographic and clinical characteristics, such as parity and smoking status, were recorded. Pearson correlation and risk estimates were calculated to explore associations between thrombophilic mutations, maternal factors, and screening results. Results: Lower parity (≤2) was significantly associated with a reduced risk of low PAPP-A levels (<1.0 MoM) (OR = 0.173; 95% CI: 0.044-0.676). Non-smokers showed a trend toward lower risk of low PAPP-A, although the association was not statistically significant. NT measurements <2.5 mm were consistent with normal fetal development, while maternal factors such as chronic hypertension and a history of small-for-gestational-age infants showed no significant correlations with screening markers. No significant association was observed between thrombophilic mutations and biomarker levels. Conclusions: Parity emerges as a significant factor influencing first-trimester screening outcomes, particularly PAPP-A levels, underscoring the need for tailored risk assessments in multiparous women. While smoking and thrombophilic mutations showed no definitive impact, their potential role in placental dysfunction warrants further investigation. These findings emphasize the importance of integrating maternal characteristics into screening protocols to enhance predictive accuracy and maternal-fetal outcomes.
Collapse
Affiliation(s)
- Viorela Romina Murvai
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 1 University Street, 410087 Oradea, Romania; (V.R.M.); (C.-M.R.); (A.-F.T.-C.); (A.H.)
- Department of Obstetrics and Gynecology, Emergency County Hospital Bihor, 65 Gheorghe Doja Street, 410169 Oradea, Romania
| | - Casandra-Maria Radu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 1 University Street, 410087 Oradea, Romania; (V.R.M.); (C.-M.R.); (A.-F.T.-C.); (A.H.)
| | - Radu Galiș
- Department of Neonatology, Faculty of Medicine and Pharmacy, University of Oradea, 1 University Street, 410087 Oradea, Romania;
| | - Timea Claudia Ghitea
- Pharmacy Department, Faculty of Medicine and Pharmacy, University of Oradea, 1 University Street, 410087 Oradea, Romania
| | - Anca-Florina Tătaru-Copos
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 1 University Street, 410087 Oradea, Romania; (V.R.M.); (C.-M.R.); (A.-F.T.-C.); (A.H.)
| | - Alexandra-Alina Vesa
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 1 University Street, 410087 Oradea, Romania; (V.R.M.); (C.-M.R.); (A.-F.T.-C.); (A.H.)
- Department of Morphological Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 1 University Street, 410087 Oradea, Romania
| | - Anca Huniadi
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 1 University Street, 410087 Oradea, Romania; (V.R.M.); (C.-M.R.); (A.-F.T.-C.); (A.H.)
- Department of Surgical Sciences, Obstetrics and Gynecology, Faculty of Medicine and Pharmacy, University of Oradea, 1 University Street, 410087 Oradea, Romania
| |
Collapse
|
3
|
Cain TL, Derecka M, McKinney-Freeman S. The role of the haematopoietic stem cell niche in development and ageing. Nat Rev Mol Cell Biol 2025; 26:32-50. [PMID: 39256623 DOI: 10.1038/s41580-024-00770-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 09/12/2024]
Abstract
Blood production depends on rare haematopoietic stem cells (HSCs) and haematopoietic stem and progenitor cells (HSPCs) that ultimately take up residence in the bone marrow during development. HSPCs and HSCs are subject to extrinsic regulation by the bone marrow microenvironment, or niche. Studying the interactions between HSCs and their niche is critical for improving ex vivo culturing conditions and genetic manipulation of HSCs, which is pivotal for improving autologous HSC therapies and transplantations. Additionally, understanding how the complex molecular network in the bone marrow is altered during ageing is paramount for developing novel therapeutics for ageing-related haematopoietic disorders. HSCs are unique amongst stem and progenitor cell pools in that they engage with multiple physically distinct niches during their ontogeny. HSCs are specified from haemogenic endothelium in the aorta, migrate to the fetal liver and, ultimately, colonize their final niche in the bone marrow. Recent studies employing single-cell transcriptomics and microscopy have identified novel cellular interactions that govern HSC specification and engagement with their niches throughout ontogeny. New lineage-tracing models and microscopy tools have raised questions about the numbers of HSCs specified, as well as the functional consequences of HSCs interacting with each developmental niche. Advances have also been made in understanding how these niches are modified and perturbed during ageing, and the role of these altered interactions in haematopoietic diseases. In this Review, we discuss these new findings and highlight the questions that remain to be explored.
Collapse
Affiliation(s)
- Terri L Cain
- Department of Haematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Marta Derecka
- Department of Haematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | |
Collapse
|
4
|
Wang J, Liang Y, Xu C, Gao J, Tong J, Shi L. The heterogeneity of erythroid cells: insight at the single-cell transcriptome level. Cell Tissue Res 2024; 397:179-192. [PMID: 38953986 DOI: 10.1007/s00441-024-03903-9] [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: 12/11/2023] [Accepted: 06/19/2024] [Indexed: 07/04/2024]
Abstract
Erythroid cells, the most prevalent cell type in blood, are one of the earliest products and permeate through the entire process of hematopoietic development in the human body, the oxygen-transporting function of which is crucial for maintaining overall health and life support. Previous investigations into erythrocyte differentiation and development have primarily focused on population-level analyses, lacking the single-cell perspective essential for comprehending the intricate pathways of erythroid maturation, differentiation, and the encompassing cellular heterogeneity. The continuous optimization of single-cell transcriptome sequencing technology, or single-cell RNA sequencing (scRNA-seq), provides a powerful tool for life sciences research, which has a particular superiority in the identification of unprecedented cell subgroups, the analyzing of cellular heterogeneity, and the transcriptomic characteristics of individual cells. Over the past decade, remarkable strides have been taken in the realm of single-cell RNA sequencing technology, profoundly enhancing our understanding of erythroid cells. In this review, we systematically summarize the recent developments in single-cell transcriptome sequencing technology and emphasize their substantial impact on the study of erythroid cells, highlighting their contributions, including the exploration of functional heterogeneity within erythroid populations, the identification of novel erythrocyte subgroups, the tracking of different erythroid lineages, and the unveiling of mechanisms governing erythroid fate decisions. These findings not only invigorate erythroid cell research but also offer new perspectives on the management of diseases related to erythroid cells.
Collapse
Affiliation(s)
- Jingwei Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Yipeng Liang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Changlu Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Jie Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Jingyuan Tong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
| | - Lihong Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin, 300020, China.
| |
Collapse
|
5
|
Palis J. Erythropoiesis in the mammalian embryo. Exp Hematol 2024; 136:104283. [PMID: 39048071 DOI: 10.1016/j.exphem.2024.104283] [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: 07/03/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
Red blood cells (RBCs) comprise a critical component of the cardiovascular network, which constitutes the first functional organ system of the developing mammalian embryo. Examination of circulating blood cells in mammalian embryos revealed two distinct types of erythroid cells: large, nucleated "primitive" erythroblasts followed by smaller, enucleated "definitive" erythrocytes. This review describes the current understanding of primitive and definitive erythropoiesis gleaned from studies of mouse and human embryos and induced pluripotent stem cells (iPSCs). Primitive erythropoiesis in the mouse embryo comprises a transient wave of committed primitive erythroid progenitors (primitive erythroid colony-forming cells, EryP-CFC) in the early yolk sac that generates a robust cohort of precursors that mature in the bloodstream and enucleate. In contrast, definitive erythropoiesis has two distinct developmental origins. The first comprises a transient wave of definitive erythroid progenitors (burst-forming units erythroid, BFU-E) that emerge in the yolk sac and seed the fetal liver where they terminally mature to provide the first definitive RBCs. The second comprises hematopoietic stem cell (HSC)-derived BFU-E that terminally mature at sites colonized by HSCs particularly the fetal liver and subsequently the bone marrow. Primitive and definitive erythropoiesis are derived from endothelial identity precursors with distinct developmental origins. Although they share prototypical transcriptional regulation, primitive and definitive erythropoiesis are also characterized by distinct lineage-specific factors. The exquisitely timed, sequential production of primitive and definitive erythroid cells is necessary for the survival and growth of the mammalian embryo.
Collapse
Affiliation(s)
- James Palis
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY.
| |
Collapse
|
6
|
Sommer A, Gomez Perdiguero E. Extraembryonic hematopoietic lineages-to macrophages and beyond. Exp Hematol 2024; 136:104285. [PMID: 39053841 DOI: 10.1016/j.exphem.2024.104285] [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: 06/06/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
The first blood and immune cells in vertebrates emerge in the extraembryonic yolk sac. Throughout the last century, it has become evident that this extraembryonic tissue gives rise to transient primitive and definitive hematopoiesis but not hematopoietic stem cells. More recently, studies have elucidated that yolk sac-derived blood and immune cells are present far longer than originally expected. These cells take over essential roles for the survival and proper organogenesis of the developing fetus up until birth. In this review, we discuss the most recent findings and views on extraembryonic hematopoiesis in mice and humans.
Collapse
Affiliation(s)
- Alina Sommer
- Macrophages and Endothelial Cells Unit, Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, France; Sorbonne Université, Collège Doctoral, Paris, France
| | - Elisa Gomez Perdiguero
- Macrophages and Endothelial Cells Unit, Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, France.
| |
Collapse
|
7
|
Park S, Hunter ES. Modeling the human placenta: in vitro applications in developmental and reproductive toxicology. Crit Rev Toxicol 2024; 54:431-464. [PMID: 39016688 DOI: 10.1080/10408444.2023.2295349] [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: 07/31/2023] [Revised: 11/20/2023] [Accepted: 12/07/2023] [Indexed: 07/18/2024]
Abstract
During its temporary tenure, the placenta has extensive and specialized functions that are critical for pre- and post-natal development. The consequences of chemical exposure in utero can have profound effects on the structure and function of pregnancy-associated tissues and the life-long health of the birthing person and their offspring. However, the toxicological importance and critical functions of the placenta to embryonic and fetal development and maturation have been understudied. This narrative will review early placental development in humans and highlight some in vitro models currently in use that are or can be applied to better understand placental processes underlying developmental toxicity due to in utero environmental exposures.
Collapse
Affiliation(s)
- Sarah Park
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
- Center for Computational Toxicology and Exposure, ORD, US EPA, Research Triangle Park, NC, USA
| | - Edward Sidney Hunter
- Center for Computational Toxicology and Exposure, ORD, US EPA, Research Triangle Park, NC, USA
| |
Collapse
|
8
|
Gunawardena N, Chou ST. Generation of red blood cells from induced pluripotent stem cells. Curr Opin Hematol 2024; 31:115-121. [PMID: 38362913 PMCID: PMC10959681 DOI: 10.1097/moh.0000000000000810] [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] [Indexed: 02/17/2024]
Abstract
PURPOSE OF REVIEW Human induced pluripotent stem cells (iPSCs) are an attractive source to generate in-vitro-derived blood for use as transfusable and reagent red cells. We review recent advancements in the field and the remaining limitations for clinical use. RECENT FINDINGS For iPSC-derived red blood cell (RBC) generation, recent work has optimized culture conditions to omit feeder cells, enhance red cell maturation, and produce cells that mimic fetal or adult-type RBCs. Genome editing provides novel strategies to improve cell yield and create designer RBCs with customized antigen phenotypes. SUMMARY Current protocols support red cell production that mimics embryonic and fetal hematopoiesis and cell yield sufficient for diagnostic RBC reagents. Ongoing challenges to generate RBCs for transfusion include recapitulating definitive erythropoiesis to produce functional adult-type cells, increasing scalability of culture conditions, and optimizing high-density manufacturing capacity.
Collapse
Affiliation(s)
| | - Stella T Chou
- Division of Hematology, Department of Pediatrics
- Division of Transfusion Medicine, Department of Pathology, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
9
|
Boss AL, Chamley LW, Brooks AES, James JL. Human placental vascular and perivascular cell heterogeneity differs between first trimester and term, and in pregnancies affected by foetal growth restriction. Mol Hum Reprod 2023; 29:gaad041. [PMID: 38059603 PMCID: PMC10746841 DOI: 10.1093/molehr/gaad041] [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: 05/25/2023] [Revised: 11/12/2023] [Indexed: 12/08/2023] Open
Abstract
Growth-restricted placentae have a reduced vascular network, impairing exchange of nutrients and oxygen. However, little is known about the differentiation events and cell types that underpin normal/abnormal placental vascular formation and function. Here, we used 23-colour flow cytometry to characterize placental vascular/perivascular populations between first trimester and term, and in foetal growth restriction (FGR). First-trimester endothelial cells had an immature phenotype (CD144+/lowCD36-CD146low), while term endothelial cells expressed mature endothelial markers (CD36+CD146+). At term, a distinct population of CD31low endothelial cells co-expressed mesenchymal markers (CD90, CD26), indicating a capacity for endothelial to mesenchymal transition (EndMT). In FGR, compared with normal pregnancies, endothelial cells constituted 3-fold fewer villous core cells (P < 0.05), contributing to an increased perivascular: endothelial cell ratio (2.6-fold, P < 0.05). This suggests that abnormal EndMT may play a role in FGR. First-trimester endothelial cells underwent EndMT in culture, losing endothelial (CD31, CD34, CD144) and gaining mesenchymal (CD90, CD26) marker expression. Together this highlights how differences in villous core cell heterogeneity and phenotype may contribute to FGR pathophysiology across gestation.
Collapse
Affiliation(s)
- Anna L Boss
- Department of Obstetrics and Gynecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Lawrence W Chamley
- Department of Obstetrics and Gynecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Anna E S Brooks
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Joanna L James
- Department of Obstetrics and Gynecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
10
|
Ozarslan N, Robinson JF, Buarpung S, Kim MY, Ansbro MR, Akram J, Montoya DJ, Kamya MR, Kakuru A, Dorsey G, Rosenthal PJ, Cheng G, Feeney ME, Fisher SJ, Gaw SL. Distinct transcriptional profiles of maternal and fetal placental macrophages at term are associated with gravidity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.25.559419. [PMID: 37808856 PMCID: PMC10557660 DOI: 10.1101/2023.09.25.559419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Maternal intervillous monocytes (MIMs) and fetal Hofbauer cells (HBCs) are myeloid-derived immune cells at the maternal-fetal interface. Little is known regarding the molecular phenotypes and roles of these distinct monocyte/macrophage populations. Here, we used RNA sequencing to investigate the transcriptional profiles of MIMs and HBCs in six normal term pregnancies. Our analyses revealed distinct transcriptomes of MIMs and HBCs. Genes involved in differentiation and cell organization pathways were more highly expressed in MIMs vs. HBCs. In contrast, HBCs had higher expression of genes involved in inflammatory responses and cell surface receptor signaling. Maternal gravidity influenced monocyte programming, as expression of pro-inflammatory molecules was significantly higher in MIMs from multigravidas compared to primigravidas. In HBCs, multigravidas displayed enrichment of gene pathways involved in cell-cell signaling and differentiation. In summary, our results demonstrated that MIMs and HBCs have highly divergent transcriptional signatures, reflecting their distinct origins, locations, functions, and roles in inflammatory responses. Our data further suggested that maternal gravidity influences the gene signatures of MIMs and HBCs, potentially modulating the interplay between tolerance and trained immunity. The phenomenon of reproductive immune memory may play a novel role in the differential susceptibility of primigravidas to pregnancy complications.
Collapse
|
11
|
Calvanese V, Mikkola HKA. The genesis of human hematopoietic stem cells. Blood 2023; 142:519-532. [PMID: 37339578 PMCID: PMC10447622 DOI: 10.1182/blood.2022017934] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/27/2023] [Accepted: 05/13/2023] [Indexed: 06/22/2023] Open
Abstract
Developmental hematopoiesis consists of multiple, partially overlapping hematopoietic waves that generate the differentiated blood cells required for embryonic development while establishing a pool of undifferentiated hematopoietic stem cells (HSCs) for postnatal life. This multilayered design in which active hematopoiesis migrates through diverse extra and intraembryonic tissues has made it difficult to define a roadmap for generating HSCs vs non-self-renewing progenitors, especially in humans. Recent single-cell studies have helped in identifying the rare human HSCs at stages when functional assays are unsuitable for distinguishing them from progenitors. This approach has made it possible to track the origin of human HSCs to the unique type of arterial endothelium in the aorta-gonad-mesonephros region and document novel benchmarks for HSC migration and maturation in the conceptus. These studies have delivered new insights into the intricate process of HSC generation and provided tools to inform the in vitro efforts to replicate the physiological developmental journey from pluripotent stem cells via distinct mesodermal and endothelial intermediates to HSCs.
Collapse
Affiliation(s)
- Vincenzo Calvanese
- Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA
| | - Hanna K. A. Mikkola
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
12
|
Thomas JR, Appios A, Calderbank EF, Yoshida N, Zhao X, Hamilton RS, Moffett A, Sharkey A, Laurenti E, Hanna CW, McGovern N. Primitive haematopoiesis in the human placenta gives rise to macrophages with epigenetically silenced HLA-DR. Nat Commun 2023; 14:1764. [PMID: 36997537 PMCID: PMC10063560 DOI: 10.1038/s41467-023-37383-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 03/13/2023] [Indexed: 04/03/2023] Open
Abstract
The earliest macrophages are generated during embryonic development from erythro-myeloid progenitors (EMPs) via primitive haematopoiesis. Although this process is thought to be spatially restricted to the yolk sac in the mouse, in humans, it remains poorly understood. Human foetal placental macrophages, or Hofbauer cells (HBC), arise during the primitive haematopoietic wave ~18 days post conception and lack expression of human leukocyte antigen (HLA) class II. Here, we identify a population of placental erythro-myeloid progenitors (PEMPs) in the early human placenta that have conserved features of primitive yolk sac EMPs, including the lack of HLF expression. Using in vitro culture experiments we demonstrate that PEMP generate HBC-like cells lacking HLA-DR expression. We find the absence of HLA-DR in primitive macrophages is mediated via epigenetic silencing of class II transactivator, CIITA, the master regulator of HLA class II gene expression. These findings establish the human placenta as an additional site of primitive haematopoiesis.
Collapse
Affiliation(s)
- Jake R Thomas
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
- Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Anna Appios
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Emily F Calderbank
- Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Nagisa Yoshida
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Xiaohui Zhao
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | | | - Ashley Moffett
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Andrew Sharkey
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Elisa Laurenti
- Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Courtney W Hanna
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| | - Naomi McGovern
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
- Department of Pathology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
13
|
Ferraz T, Benton SJ, Zareef I, Aribaloye O, Bloise E, Connor KL. Impact of Co-Occurrence of Obesity and SARS-CoV-2 Infection during Pregnancy on Placental Pathologies and Adverse Birth Outcomes: A Systematic Review and Narrative Synthesis. Pathogens 2023; 12:pathogens12040524. [PMID: 37111410 PMCID: PMC10140965 DOI: 10.3390/pathogens12040524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Obesity is a risk factor for severe COVID-19 disease during pregnancy. We hypothesized that the co-occurrence of high maternal body mass index (BMI) and gestational SARS-CoV-2 infection are detrimental to fetoplacental development. We conducted a systematic review following PRISMA/SWiM guidelines and 13 studies were eligible. In the case series studies (n = 7), the most frequent placental lesions reported in SARS-CoV-2(+) pregnancies with high maternal BMI were chronic inflammation (71.4%, 5/7 studies), fetal vascular malperfusion (FVM) (71.4%, 5/7 studies), maternal vascular malperfusion (MVM) (85.7%, 6/7 studies) and fibrinoids (100%, 7/7 studies). In the cohort studies (n = 4), three studies reported higher rates of chronic inflammation, MVM, FVM and fibrinoids in SARS-CoV-2(+) pregnancies with high maternal BMI (72%, n = 107/149; mean BMI of 30 kg/m2) compared to SARS-CoV-2(−) pregnancies with high BMI (7.4%, n = 10/135). In the fourth cohort study, common lesions observed in placentae from SARS-CoV-2(+) pregnancies with high BMI (n = 187 pregnancies; mean BMI of 30 kg/m2) were chronic inflammation (99%, 186/187), MVM (40%, n = 74/187) and FVM (26%, n = 48/187). BMI and SARS-CoV-2 infection had no effect on birth anthropometry. SARS-CoV-2 infection during pregnancy associates with increased prevalence of placental pathologies, and high BMI in these pregnancies could further affect fetoplacental trajectories.
Collapse
Affiliation(s)
- Thaina Ferraz
- Health Sciences, Carleton University, Ottawa, ON K1S 5B6, Canada
| | | | - Israa Zareef
- Health Sciences, Carleton University, Ottawa, ON K1S 5B6, Canada
| | | | - Enrrico Bloise
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Kristin L. Connor
- Health Sciences, Carleton University, Ottawa, ON K1S 5B6, Canada
- Correspondence: ; Tel.: +1-613-520-2600 (ext. 4202)
| |
Collapse
|
14
|
Kuperwaser F, Avital G, Vaz MJ, Noble KN, Dammann AN, Randis TM, Aronoff DM, Ratner AJ, Yanai I. Host inflammatory dynamics reveal placental immune modulation by Group B Streptococcus during pregnancy. Mol Syst Biol 2023; 19:e11021. [PMID: 36744393 PMCID: PMC9996236 DOI: 10.15252/msb.202211021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 02/07/2023] Open
Abstract
Group B Streptococcus (GBS) is a pathobiont that can ascend to the placenta and cause adverse pregnancy outcomes, in part through production of the toxin β-hemolysin/cytolysin (β-h/c). Innate immune cells have been implicated in the response to GBS infection, but the impact of β-h/c on their response is poorly defined. We show that GBS modulates innate immune cell states by subversion of host inflammation through β-h/c, allowing worse outcomes. We used an ascending mouse model of GBS infection to measure placental cell state changes over time following infection with a β-h/c-deficient and isogenic wild type GBS strain. Transcriptomic analysis suggests that β-h/c-producing GBS elicit a worse phenotype through suppression of host inflammatory signaling in placental macrophages and neutrophils, and comparison of human placental macrophages infected with the same strains recapitulates these results. Our findings have implications for identification of new targets in GBS disease to support host defense against pathogenic challenge.
Collapse
Affiliation(s)
- Felicia Kuperwaser
- Institute for Computational MedicineNYU Grossman School of MedicineNew YorkNYUSA
| | - Gal Avital
- Institute for Computational MedicineNYU Grossman School of MedicineNew YorkNYUSA
| | - Michelle J Vaz
- Department of PediatricsNYU Grossman School of MedicineNew YorkNYUSA
| | - Kristen N Noble
- Division of Neonatology, Department of PediatricsVanderbilt University Medical CenterNashvilleTNUSA
| | - Allison N Dammann
- Renaissance School of Medicine at Stony Brook UniversityStony BrookNYUSA
| | - Tara M Randis
- Departments of Pediatrics and Molecular Medicine, Morsani School of MedicineUniversity of South FloridaFLTampaUSA
| | | | - Adam J Ratner
- Department of PediatricsNYU Grossman School of MedicineNew YorkNYUSA
- Department of MicrobiologyNYU Grossman School of MedicineNew YorkNYUSA
| | - Itai Yanai
- Institute for Computational MedicineNYU Grossman School of MedicineNew YorkNYUSA
- Department of Biochemistry and Molecular PharmacologyNYU Grossman School of MedicineNew YorkNYUSA
| |
Collapse
|
15
|
Yan L, Li J, Wang Y, Zhu Q, Zhao X, He C, Zhu C, Ji S, Zhang Y, MuDanLiFu H, Zhang J. Trophoblastic infiltration of tubal pregnancy may have an association with chronic inflammation of the fallopian tube. Int J Gynaecol Obstet 2023. [PMID: 36607245 DOI: 10.1002/ijgo.14658] [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: 09/20/2022] [Revised: 11/28/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
OBJECTIVE To explore the factors associated with trophoblastic infiltration in ampullary pregnancy from the perspective of clinical and pathologic characteristics. METHODS A single-center, retrospective, clinicopathologic cohort study was conducted in women who were diagnosed with tubal pregnancy and underwent salpingectomy in the International Peace Maternal and Child Health Care Hospital from January 2018 to June 2021. RESULTS A total of 333 eligible women diagnosed with ampullary pregnancy were included in the analysis. Multivariate logistic analysis showed that preoperative β-human chorionic gonadotropin greater than 3000 IU/L (adjusted odds ratio [aOR] 3.77, 95% confidence interval [CI] 2.02-7.03), and vascular remodeling phenomenon (aOR 4.34, 95% CI 2.41-7.83) were positively correlated with the infiltration of extravillous trophoblasts into serosa, while presence of chronic inflammation of the fallopian tube was a negatively corellated factor (aOR 0.49, 95% CI 0.29-0.85). CONCLUSION The depth of trophoblastic infiltration in tubal pregnancy may be related to the presence of chronic inflammation in the fallopian tube. A tubal pregnancy in a tube with chronic salpingitis is more likely to develop into an abortive ectopic pregnancy; whereas in a fallopian tube without chronic inflammation, the risk of it developing into a ruptured ectopic pregnancy increases. Hence, early identification is needed to properly address this dangerous pregnancy situation.
Collapse
Affiliation(s)
- Li Yan
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Key Laboratory Embryo Original Diseases, Shanghai, China
| | - Juan Li
- Department of Pathology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yang Wang
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Key Laboratory Embryo Original Diseases, Shanghai, China
| | - Qian Zhu
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Key Laboratory Embryo Original Diseases, Shanghai, China
| | - Xiaoya Zhao
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Key Laboratory Embryo Original Diseases, Shanghai, China
| | - Chuqing He
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Key Laboratory Embryo Original Diseases, Shanghai, China
| | - Chenfeng Zhu
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Key Laboratory Embryo Original Diseases, Shanghai, China
| | - Sifan Ji
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Key Laboratory Embryo Original Diseases, Shanghai, China
| | - Yiqin Zhang
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Key Laboratory Embryo Original Diseases, Shanghai, China
| | - HaLiSai MuDanLiFu
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Key Laboratory Embryo Original Diseases, Shanghai, China
| | - Jian Zhang
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai Key Laboratory Embryo Original Diseases, Shanghai, China
| |
Collapse
|
16
|
Barrett AN, Huang Z, Aung S, Ho SSY, Roslan NS, Mahyuddin AP, Biswas A, Choolani M. Whole-Chromosome Karyotyping of Fetal Nucleated Red Blood Cells Using the Ion Proton Sequencing Platform. Genes (Basel) 2022; 13:genes13122257. [PMID: 36553524 PMCID: PMC9778445 DOI: 10.3390/genes13122257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/26/2022] [Accepted: 11/26/2022] [Indexed: 12/02/2022] Open
Abstract
The current gold standard for the definitive diagnosis of fetal aneuploidy uses either chorionic villus sampling (CVS) or amniocentesis, both of which are which are invasive procedures carrying a procedure-related risk of miscarriage of up to 0.1-0.2%. Non-invasive prenatal diagnosis using fetal nucleated red blood cells (FNRBCs) isolated from maternal peripheral venous blood would remove this risk of miscarriage since these cells can be isolated from the mother's blood. We aimed to detect whole-chromosome aneuploidies from single nucleated fetal red blood cells using whole-genome amplification followed by massively parallel sequencing performed on a semiconductor sequencing platform. Twenty-six single cells were picked from the placental villi of twelve patients thought to have a normal fetal genotype and who were undergoing elective first-trimester surgical termination of pregnancy. Following karyotyping, it was subsequently found that two of these cases were also abnormal (one trisomy 15 and one mosaic genotype). One single cell from chorionic villus samples for two patients carrying a fetus with trisomy 21 and two single cells from women carrying fetuses with T18 were also picked. Pooled libraries were sequenced on the Ion Proton and data were analysed using Ion Reporter software. We correctly classified fetal genotype in all 24 normal cells, as well as the 2 T21 cells, the 2 T18 cells, and the two T15 cells. The two cells picked from the fetus with a mosaic result by CVS were classified as unaffected, suggesting that this was a case of confined placental mosaicism. Fetal sex was correctly assigned in all cases. We demonstrated that semiconductor sequencing using commercially available software for data analysis can be achieved for the non-invasive prenatal diagnosis of whole-chromosome aneuploidy with 100% accuracy.
Collapse
Affiliation(s)
- Angela N. Barrett
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228, Singapore
| | - Zhouwei Huang
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228, Singapore
| | - Sarah Aung
- iGene Laboratory Pte Ltd., 1 Science Park Road #04-10, The Capricorn, Singapore 117528, Singapore
| | - Sherry S. Y. Ho
- iGene Laboratory Pte Ltd., 1 Science Park Road #04-10, The Capricorn, Singapore 117528, Singapore
| | - Nur Syazana Roslan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228, Singapore
| | - Aniza P. Mahyuddin
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228, Singapore
| | - Arijit Biswas
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228, Singapore
- Department of Obstetrics & Gynaecology, National University Hospital, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228, Singapore
| | - Mahesh Choolani
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228, Singapore
- Department of Obstetrics & Gynaecology, National University Hospital, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228, Singapore
- Correspondence:
| |
Collapse
|
17
|
Testa U, Castelli G, Pelosi E. Clonal Hematopoiesis: Role in Hematologic and Non-Hematologic Malignancies. Mediterr J Hematol Infect Dis 2022; 14:e2022069. [PMID: 36119457 PMCID: PMC9448266 DOI: 10.4084/mjhid.2022.069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/18/2022] [Indexed: 02/08/2023] Open
Abstract
Hematopoietic stem cells (HSCs) ensure the coordinated and balanced production of all hematopoietic cell types throughout life. Aging is associated with a gradual decline of the self-renewal and regenerative potential of HSCs and with the development of clonal hematopoiesis. Clonal hematopoiesis of indeterminate potential (CHIP) defines the clonal expansion of genetically variant hematopoietic cells bearing one or more gene mutations and/or structural variants (such as copy number alterations). CHIP increases exponentially with age and is associated with cancers, including hematologic neoplasia, cardiovascular and other diseases. The presence of CHIP consistently increases the risk of hematologic malignancy, particularly in individuals who have CHIP in association with peripheral blood cytopenia.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
18
|
Abstract
The liver is the major target organ of continued alcohol consumption at risk and resulting alcoholic liver disease (ALD) is the most common liver disease worldwide. The underlying molecular mechanisms are still poorly understood despite decades of scientific effort limiting our abilities to identify those individuals who are at risk to develop the disease, to develop appropriate screening strategies and, in addition, to develop targeted therapeutic approaches. ALD is predestined for the newly evolving translational medicine, as conventional clinical and health care structures seem to be constrained to fully appreciate this disease. This concept paper aims at summarizing the 15 years translational experience at the Center of Alcohol Research in Heidelberg, namely based on the long-term prospective and detailed characterization of heavy drinkers with mortality data. In addition, novel experimental findings will be presented. A special focus will be the long-known hepatic iron accumulation, the somewhat overlooked role of the hematopoietic system and novel insights into iron sensing and the role of hepcidin. Our preliminary work indicates that enhanced red blood cell (RBC) turnover is critical for survival in ALD patients. RBC turnover is not primarily due to vitamin deficiency but rather to ethanol toxicity directly targeted to erythrocytes but also to the bone marrow stem cell compartment. These novel insights also help to explain long-known aspects of ALD such as mean corpuscular volume of erythrocytes (MCV) and elevated aspartate transaminase (GOT/AST) levels. This work also aims at identifying future projects, naming unresolved observations, and presenting novel hypothetical concepts still requiring future validation.
Collapse
|
19
|
Zaugg J, Solenthaler F, Albrecht C. Materno-fetal iron transfer and the emerging role of ferroptosis pathways. Biochem Pharmacol 2022; 202:115141. [PMID: 35700759 DOI: 10.1016/j.bcp.2022.115141] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 11/28/2022]
Abstract
A successful pregnancy and the birth of a healthy baby depend to a great extent on the controlled supply of essential nutrients via the placenta. Iron is essential for mitochondrial energy supply and oxygen distribution via the blood. However, its high reactivity requires tightly regulated transport processes. Disturbances of maternal-fetal iron transfer during pregnancy can aggravate or lead to severe pathological consequences for the mother and the fetus with lifelong effects. Furthermore, high intracellular iron levels due to disturbed gestational iron homeostasis have recently been associated with the non-apoptotic cell death pathway called ferroptosis. Therefore, the investigation of transplacental iron transport mechanisms, their physiological regulation and potential risks are of high clinical importance. The present review summarizes the current knowledge on principles and regulatory mechanisms underlying materno-fetal iron transport and gives insight into common pregnancy conditions in which iron homeostasis is disturbed. Moreover, the significance of the newly emerging ferroptosis pathway and its impact on the regulation of placental iron homeostasis, oxidative stress and gestational diseases will be discussed.
Collapse
Affiliation(s)
- Jonas Zaugg
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Switzerland
| | - Fabia Solenthaler
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Switzerland
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Switzerland.
| |
Collapse
|
20
|
Pinson MR, Tseng AM, Adams A, Lehman TE, Chung K, Gutierrez J, Larin KV, Chambers C, Miranda RC. Prenatal alcohol exposure contributes to negative pregnancy outcomes by altering fetal vascular dynamics and the placental transcriptome. Alcohol Clin Exp Res 2022; 46:1036-1049. [PMID: 35474222 PMCID: PMC9325399 DOI: 10.1111/acer.14846] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/17/2022] [Accepted: 04/18/2022] [Indexed: 11/28/2022]
Abstract
Background Prenatal alcohol exposure (PAE) has been shown to alter fetal blood flow in utero and is also associated with placental insufficiency and intrauterine growth restriction (IUGR), suggesting an underlying connection between perturbed circulation and pregnancy outcomes. Methods Timed‐pregnant C57/BL6NHsd mice, bred in‐house, were exposed by gavage on gestational day 10 (GD10) to ethanol (3 g/kg) or purified water, as a control. Pulse‐wave Doppler ultrasound measurements for umbilical arteries and ascending aorta were obtained post‐gavage (GD12, GD14, GD18) on 2 fetuses/litter. RNA from the non‐decidual (labyrinthine and junctional zone) portion of placentas was isolated and processed for RNA‐seq and subsequent bioinformatic analyses, and the association between transcriptomic changes and fetal phenotypes assessed. Results Exposure to ethanol in pregnant mice on GD10 attenuates umbilical cord blood flow transiently during gestation, and is associated with indices of IUGR, specifically decreased fetal weight and morphometric indices of cranial growth. Moreover, RNA‐seq of the fetal portion of the placenta demonstrated that this single exposure has lasting transcriptomic changes, including upregulation of Tet3, which is associated with spontaneous abortion. Weighted gene co‐expression network analysis (WGCNA) identified erythrocyte differentiation and homeostasis as important pathways associated with improved umbilical cord blood flow as gestation progresses. WGCNA also identified sensory perception of chemical stimulus/odorant and receptor activity as important pathways associated with cranial growth. Conclusion Our data suggest that PAE perturbs the expression of placental genes relevant for placental hematopoiesis and environmental sensing, resulting in transient impairment of umbilical cord blood flow and, subsequently, IUGR.
Collapse
Affiliation(s)
- Marisa R Pinson
- Department of Neuroscience and Experimental Therapeutics, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Alexander M Tseng
- Department of Neuroscience and Experimental Therapeutics, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Amy Adams
- Department of Neuroscience and Experimental Therapeutics, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Tenley E Lehman
- Department of Neuroscience and Experimental Therapeutics, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Karen Chung
- Department of Neuroscience and Experimental Therapeutics, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Jessica Gutierrez
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Kirill V Larin
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Christina Chambers
- Clinical and Translational Research Institute, University of California San Diego, San Diego, California, USA.,Department of Pediatrics, University of California San Diego, San Diego, California, USA
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M College of Medicine, Bryan, Texas, USA.,Women's Health in Neuroscience Program, Texas A&M University College of Medicine, Bryan, Texas, USA.,Interdisciplinary Program of Genetics, Texas A&M University, College Station, Texas, USA
| | | |
Collapse
|
21
|
Physiological Function of the Dynamic Oxygen Signaling Pathway at the Maternal-fetal Interface. J Reprod Immunol 2022; 151:103626. [DOI: 10.1016/j.jri.2022.103626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 02/21/2022] [Accepted: 04/13/2022] [Indexed: 11/21/2022]
|
22
|
Mapping human haematopoietic stem cells from haemogenic endothelium to birth. Nature 2022; 604:534-540. [PMID: 35418685 PMCID: PMC9645817 DOI: 10.1038/s41586-022-04571-x] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/22/2022] [Indexed: 01/19/2023]
Abstract
The ontogeny of human haematopoietic stem cells (HSCs) is poorly defined owing to the inability to identify HSCs as they emerge and mature at different haematopoietic sites1. Here we created a single-cell transcriptome map of human haematopoietic tissues from the first trimester to birth and found that the HSC signature RUNX1+HOXA9+MLLT3+MECOM+HLF+SPINK2+ distinguishes HSCs from progenitors throughout gestation. In addition to the aorta-gonad-mesonephros region, nascent HSCs populated the placenta and yolk sac before colonizing the liver at 6 weeks. A comparison of HSCs at different maturation stages revealed the establishment of HSC transcription factor machinery after the emergence of HSCs, whereas their surface phenotype evolved throughout development. The HSC transition to the liver marked a molecular shift evidenced by suppression of surface antigens reflecting nascent HSC identity, and acquisition of the HSC maturity markers CD133 (encoded by PROM1) and HLA-DR. HSC origin was tracked to ALDH1A1+KCNK17+ haemogenic endothelial cells, which arose from an IL33+ALDH1A1+ arterial endothelial subset termed pre-haemogenic endothelial cells. Using spatial transcriptomics and immunofluorescence, we visualized this process in ventrally located intra-aortic haematopoietic clusters. The in vivo map of human HSC ontogeny validated the generation of aorta-gonad-mesonephros-like definitive haematopoietic stem and progenitor cells from human pluripotent stem cells, and serves as a guide to improve their maturation to functional HSCs.
Collapse
|
23
|
Deng L, Lu Y, Yang D, Yang F, Ruan H, Wei C, Lai K, Pang L. Placental transcriptome sequencing combined with bioinformatics predicts potential genes and circular RNAs associated with hemoglobin Bart's hydrops fetalis syndrome. J Obstet Gynaecol Res 2021; 48:313-327. [PMID: 34935248 DOI: 10.1111/jog.15126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 10/17/2021] [Accepted: 11/28/2021] [Indexed: 02/06/2023]
Abstract
AIM Hemoglobin Bart's hydrops fetalis syndrome (BHFS) is the most severe form of α-thalassemia. Histological alternations can be observed in placenta, but placental transcriptome profile and circular RNAs have not been studied in this disease. The aim of this study was to define the placental transcriptional changes and find relevant circular RNAs in BHFS. METHODS We performed high-throughput RNA sequencing to detect placental samples from fetuses affected by BHFS (n = 5) and normal fetuses (NF, n = 5), quantitative reverse transcription polymerase chain reaction (RT-qPCR), and Sanger sequencing to validate the differentially expressed circRNAs and their potentially related miRNAs (BHFS, n = 22; NF, n = 11). Bioinformatics methods were performed for further analysis. RESULTS Our results showed 152 differentially expressed genes (DEGs), 112 circRNAs, and 45 microRNAs that were differentially expressed. DEGs were found to be involved in Gene Ontology terms related to gas transport, cell adhesion, oxidative stress, organ development, hemopoiesis, and others. RT-qPCR results showed that hsa_circ_0003961 and hsa_circ_0006687 were upregulated (p < 0.05). The competing endogenous RNA and co-expression networks showed that hsa_circ_0003961 and hsa_circ_0006687 were connected with 3 miRNAs and some DEGs, including cell adhesion genes (e.g., CLDN19), hemoglobin related genes (e.g., SOX6 and HBZ) and angiogenesis related genes (e.g., EPHB2). Downregulations of hsa-miR-1299 and hsa-miR-625-5p in ceRNA network were also validated by RT-qPCR. Gene set enrichment analysis results for the two circRNAs showed that some gene sets associated with cell adhesion, hematopoietic system and apoptosis were significantly enriched. CONCLUSIONS Our study characterized the placental transcriptome of BHFS. The circRNAs hsa_circ_0003961 and hsa_circ_0006687 in placenta may be relevant to BHFS.
Collapse
Affiliation(s)
- Lingjie Deng
- Department of Prenatal Diagnosis and Genetic Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yebin Lu
- Guangxi Medical University, Nanning, China.,Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Gaungxi Medical University), Ministry of Education, Nanning, China
| | - Dongmei Yang
- Department of Prenatal Diagnosis and Genetic Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Fang Yang
- Department of Prenatal Diagnosis and Genetic Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Heyun Ruan
- Guangxi Medical University, Nanning, China
| | | | - Ketong Lai
- Guangxi Medical University, Nanning, China
| | - Lihong Pang
- Department of Prenatal Diagnosis and Genetic Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
24
|
James JL, Boss AL, Sun C, Allerkamp HH, Clark AR. From stem cells to spiral arteries: A journey through early placental development. Placenta 2021; 125:68-77. [PMID: 34819240 DOI: 10.1016/j.placenta.2021.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/11/2021] [Accepted: 11/14/2021] [Indexed: 12/19/2022]
Abstract
Early placental development lays the foundation of a healthy pregnancy, and numerous tightly regulated processes must occur for the placenta to meet the increasing nutrient and oxygen exchange requirements of the growing fetus later in gestation. Inadequacies in early placental development can result in disorders such as fetal growth restriction that do not present clinically until the second half of gestation. Indeed, growth restricted placentae exhibit impaired placental development and function, including reduced overall placental size, decreased branching of villi and the blood vessels within them, altered trophoblast function, and impaired uterine vascular remodelling, which together combine to reduce placental exchange capacity. This review explores the importance of early placental development across multiple anatomical aspects of placentation, from the stem cells and lineage hierarchies from which villous core cells and trophoblasts arise, through extravillous trophoblast invasion and spiral artery remodelling, and finally remodelling of the larger uterine vessels.
Collapse
Affiliation(s)
- Joanna L James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand.
| | - Anna L Boss
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Cherry Sun
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Hanna H Allerkamp
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand; Auckland Bioengineering Institute, University of Auckland, New Zealand
| | - Alys R Clark
- Auckland Bioengineering Institute, University of Auckland, New Zealand
| |
Collapse
|
25
|
Erlandsson L, Masoumi Z, Hansson LR, Hansson SR. The roles of free iron, heme, haemoglobin, and the scavenger proteins haemopexin and alpha-1-microglobulin in preeclampsia and fetal growth restriction. J Intern Med 2021; 290:952-968. [PMID: 34146434 DOI: 10.1111/joim.13349] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Preeclampsia (PE) is a complex pregnancy syndrome characterised by maternal hypertension and organ damage after 20 weeks of gestation and is associated with an increased risk of cardiovascular disease later in life. Extracellular haemoglobin (Hb) and its metabolites heme and iron are highly toxic molecules and several defence mechanisms have evolved to protect the tissue. OBJECTIVES We will discuss the roles of free iron, heme, Hb, and the scavenger proteins haemopexin and alpha-1-microglobulin in pregnancies complicated by PE and fetal growth restriction (FGR). CONCLUSION In PE, oxidative stress causes syncytiotrophoblast (STB) stress and increased shedding of placental STB-derived extracellular vesicles (STBEV). The level in maternal circulation correlates with the severity of hypertension and supports the involvement of STBEVs in causing maternal symptoms in PE. In PE and FGR, iron homeostasis is changed, and iron levels significantly correlate with the severity of the disease. The normal increase in plasma volume taking place during pregnancy is less for PE and FGR and therefore have a different impact on, for example, iron concentration, compared to normal pregnancy. Excess iron promotes ferroptosis is suggested to play a role in trophoblast stress and lipotoxicity. Non-erythroid α-globin regulates vasodilation through the endothelial nitric oxide synthase pathway, and hypoxia-induced α-globin expression in STBs in PE placentas is suggested to contribute to hypertension in PE. Underlying placental pathology in PE with and without FGR might be amplified by iron and heme overload causing oxidative stress and ferroptosis. As the placenta becomes stressed, the release of STBEVs increases and affects the maternal vasculature.
Collapse
Affiliation(s)
- Lena Erlandsson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Zahra Masoumi
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Lucas R Hansson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Stefan R Hansson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.,Obstetrics and Gynecology, Skåne University Hospital, Lund/Malmö, Sweden
| |
Collapse
|
26
|
Thomas JR, Naidu P, Appios A, McGovern N. The Ontogeny and Function of Placental Macrophages. Front Immunol 2021; 12:771054. [PMID: 34745147 PMCID: PMC8566952 DOI: 10.3389/fimmu.2021.771054] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 10/05/2021] [Indexed: 01/02/2023] Open
Abstract
The placenta is a fetal-derived organ whose function is crucial for both maternal and fetal health. The human placenta contains a population of fetal macrophages termed Hofbauer cells. These macrophages play diverse roles, aiding in placental development, function and defence. The outer layer of the human placenta is formed by syncytiotrophoblast cells, that fuse to form the syncytium. Adhered to the syncytium at sites of damage, on the maternal side of the placenta, is a population of macrophages termed placenta associated maternal macrophages (PAMM1a). Here we discuss recent developments that have led to renewed insight into our understanding of the ontogeny, phenotype and function of placental macrophages. Finally, we discuss how the application of new technologies within placental research are helping us to further understand these cells.
Collapse
Affiliation(s)
| | | | | | - Naomi McGovern
- Department of Pathology and Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
27
|
Industrially Compatible Transfusable iPSC-Derived RBCs: Progress, Challenges and Prospective Solutions. Int J Mol Sci 2021; 22:ijms22189808. [PMID: 34575977 PMCID: PMC8472628 DOI: 10.3390/ijms22189808] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 02/06/2023] Open
Abstract
Amidst the global shortfalls in blood supply, storage limitations of donor blood and the availability of potential blood substitutes for transfusion applications, society has pivoted towards in vitro generation of red blood cells (RBCs) as a means to solve these issues. Many conventional research studies over the past few decades have found success in differentiating hematopoietic stem and progenitor cells (HSPCs) from cord blood, adult bone marrow and peripheral blood sources. More recently, techniques that involve immortalization of erythroblast sources have also gained traction in tackling this problem. However, the RBCs generated from human induced pluripotent stem cells (hiPSCs) still remain as the most favorable solution due to many of its added advantages. In this review, we focus on the breakthroughs for high-density cultures of hiPSC-derived RBCs, and highlight the major challenges and prospective solutions throughout the whole process of erythropoiesis for hiPSC-derived RBCs. Furthermore, we elaborate on the recent advances and techniques used to achieve cost-effective, high-density cultures of GMP-compliant RBCs, and on their relevant novel applications after downstream processing and purification.
Collapse
|
28
|
The Impact of Hypoxia in Early Pregnancy on Placental Cells. Int J Mol Sci 2021; 22:ijms22189675. [PMID: 34575844 PMCID: PMC8466283 DOI: 10.3390/ijms22189675] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/04/2021] [Accepted: 09/05/2021] [Indexed: 12/14/2022] Open
Abstract
Oxygen levels in the placental microenvironment throughout gestation are not constant, with severe hypoxic conditions present during the first trimester. This hypoxic phase overlaps with the most critical stages of placental development, i.e., blastocyst implantation, cytotrophoblast invasion, and spiral artery remodeling initiation. Dysregulation of any of these steps in early gestation can result in pregnancy loss and/or adverse pregnancy outcomes. Hypoxia has been shown to regulate not only the self-renewal, proliferation, and differentiation of trophoblast stem cells and progenitor cells, but also the recruitment, phenotype, and function of maternal immune cells. In this review, we will summarize how oxygen levels in early placental development determine the survival, fate, and function of several important cell types, e.g., trophoblast stem cells, extravillous trophoblasts, syncytiotrophoblasts, uterine natural killer cells, Hofbauer cells, and decidual macrophages. We will also discuss the cellular mechanisms used to cope with low oxygen tensions, such as the induction of hypoxia-inducible factor (HIF) or mammalian target of rapamycin (mTOR) signals, regulation of the metabolic pathway, and adaptation to autophagy. Understanding the beneficial roles of hypoxia in early placental development will provide insights into the root cause(s) of some pregnancy disorders, such as spontaneous abortion, preeclampsia, and intrauterine growth restriction.
Collapse
|
29
|
Thomas JR, Appios A, Zhao X, Dutkiewicz R, Donde M, Lee CYC, Naidu P, Lee C, Cerveira J, Liu B, Ginhoux F, Burton G, Hamilton RS, Moffett A, Sharkey A, McGovern N. Phenotypic and functional characterization of first-trimester human placental macrophages, Hofbauer cells. J Exp Med 2021; 218:211477. [PMID: 33075123 PMCID: PMC7579740 DOI: 10.1084/jem.20200891] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/30/2020] [Accepted: 09/11/2020] [Indexed: 12/31/2022] Open
Abstract
Hofbauer cells (HBCs) are a population of macrophages found in high abundance within the stroma of the first-trimester human placenta. HBCs are the only fetal immune cell population within the stroma of healthy placenta. However, the functional properties of these cells are poorly described. Aligning with their predicted origin via primitive hematopoiesis, we find that HBCs are transcriptionally similar to yolk sac macrophages. Phenotypically, HBCs can be identified as HLA-DR-FOLR2+ macrophages. We identify a number of factors that HBCs secrete (including OPN and MMP-9) that could affect placental angiogenesis and remodeling. We determine that HBCs have the capacity to play a defensive role, where they are responsive to Toll-like receptor stimulation and are microbicidal. Finally, we also identify a population of placenta-associated maternal macrophages (PAMM1a) that adhere to the placental surface and express factors, such as fibronectin, that may aid in repair.
Collapse
Affiliation(s)
- Jake R Thomas
- Department of Pathology, University of Cambridge, Cambridge, UK.,Centre for Trophoblast Research, Departments of Physiology and Neuroscience, University of Cambridge, Cambridge, UK
| | - Anna Appios
- Department of Pathology, University of Cambridge, Cambridge, UK.,Centre for Trophoblast Research, Departments of Physiology and Neuroscience, University of Cambridge, Cambridge, UK
| | - Xiaohui Zhao
- Centre for Trophoblast Research, Departments of Physiology and Neuroscience, University of Cambridge, Cambridge, UK
| | | | - Maria Donde
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Colin Y C Lee
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Praveena Naidu
- Department of Pathology, University of Cambridge, Cambridge, UK.,Centre for Trophoblast Research, Departments of Physiology and Neuroscience, University of Cambridge, Cambridge, UK
| | - Christopher Lee
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Joana Cerveira
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Bing Liu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China.,State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China.,State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin, China
| | - Florent Ginhoux
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Graham Burton
- Centre for Trophoblast Research, Departments of Physiology and Neuroscience, University of Cambridge, Cambridge, UK
| | - Russell S Hamilton
- Centre for Trophoblast Research, Departments of Physiology and Neuroscience, University of Cambridge, Cambridge, UK.,Department of Genetics, University of Cambridge, Cambridge, UK
| | - Ashley Moffett
- Department of Pathology, University of Cambridge, Cambridge, UK.,Centre for Trophoblast Research, Departments of Physiology and Neuroscience, University of Cambridge, Cambridge, UK
| | - Andrew Sharkey
- Department of Pathology, University of Cambridge, Cambridge, UK.,Centre for Trophoblast Research, Departments of Physiology and Neuroscience, University of Cambridge, Cambridge, UK
| | - Naomi McGovern
- Department of Pathology, University of Cambridge, Cambridge, UK.,Centre for Trophoblast Research, Departments of Physiology and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
30
|
Hypoxia-Induced Alpha-Globin Expression in Syncytiotrophoblasts Mimics the Pattern Observed in Preeclamptic Placentas. Int J Mol Sci 2021; 22:ijms22073357. [PMID: 33806017 PMCID: PMC8036899 DOI: 10.3390/ijms22073357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 12/16/2022] Open
Abstract
Preeclampsia (PE) is a pregnancy disorder associated with placental dysfunction and elevated fetal hemoglobin (HbF). Early in pregnancy the placenta harbors hematopoietic stem and progenitor cells (HSPCs) and is an extramedullary source of erythropoiesis. However, globin expression is not unique to erythroid cells and can be triggered by hypoxia. To investigate the role of the placenta in increasing globin levels previously reported in PE, flow cytometry, histological and immunostaining and in situ analyses were used on placenta samples and ex vivo explant cultures. Our results indicated that in PE pregnancies, placental HSPC homing and erythropoiesis were not affected. Non-erythroid alpha-globin mRNA and protein, but not gamma-globin, were detected in syncytiotrophoblasts and stroma of PE placenta samples. Similarly, alpha-globin protein and mRNA were upregulated in normal placenta explants cultured in hypoxia. The upregulation was independent of HIF1 and NRF2, the two main candidates of globin transcription in non-erythroid cells. Our study is the first to demonstrate alpha-globin mRNA expression in syncytiotrophoblasts in PE, induced by hypoxia. However, gamma-globin was only expressed in erythrocytes. We conclude that alpha-globin, but not HbF, is expressed in placental syncytiotrophoblasts in PE and may contribute to the pathology of the disease.
Collapse
|
31
|
Jiang X, Liu B, Nie Z, Duan L, Xiong Q, Jin Z, Yang C, Chen Y. The role of m6A modification in the biological functions and diseases. Signal Transduct Target Ther 2021; 6:74. [PMID: 33611339 PMCID: PMC7897327 DOI: 10.1038/s41392-020-00450-x] [Citation(s) in RCA: 1155] [Impact Index Per Article: 288.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 12/09/2020] [Indexed: 01/31/2023] Open
Abstract
N6-methyladenosine (m6A) is the most prevalent, abundant and conserved internal cotranscriptional modification in eukaryotic RNAs, especially within higher eukaryotic cells. m6A modification is modified by the m6A methyltransferases, or writers, such as METTL3/14/16, RBM15/15B, ZC3H3, VIRMA, CBLL1, WTAP, and KIAA1429, and, removed by the demethylases, or erasers, including FTO and ALKBH5. It is recognized by m6A-binding proteins YTHDF1/2/3, YTHDC1/2 IGF2BP1/2/3 and HNRNPA2B1, also known as "readers". Recent studies have shown that m6A RNA modification plays essential role in both physiological and pathological conditions, especially in the initiation and progression of different types of human cancers. In this review, we discuss how m6A RNA methylation influences both the physiological and pathological progressions of hematopoietic, central nervous and reproductive systems. We will mainly focus on recent progress in identifying the biological functions and the underlying molecular mechanisms of m6A RNA methylation, its regulators and downstream target genes, during cancer progression in above systems. We propose that m6A RNA methylation process offer potential targets for cancer therapy in the future.
Collapse
Affiliation(s)
- Xiulin Jiang
- grid.419010.d0000 0004 1792 7072Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, 650223 Kunming, Yunnan China ,grid.410726.60000 0004 1797 8419Kunming College of Life Science, University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Baiyang Liu
- grid.419010.d0000 0004 1792 7072Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, 650223 Kunming, Yunnan China ,grid.410726.60000 0004 1797 8419Kunming College of Life Science, University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Zhi Nie
- grid.419010.d0000 0004 1792 7072Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, 650223 Kunming, Yunnan China ,grid.410726.60000 0004 1797 8419Kunming College of Life Science, University of Chinese Academy of Sciences, 100049 Beijing, China ,grid.285847.40000 0000 9588 0960Kunming Medical University, 650500 Kunming, China
| | - Lincan Duan
- grid.285847.40000 0000 9588 0960Kunming Medical University, 650500 Kunming, China
| | - Qiuxia Xiong
- grid.285847.40000 0000 9588 0960Kunming Medical University, 650500 Kunming, China
| | - Zhixian Jin
- grid.285847.40000 0000 9588 0960Kunming Medical University, 650500 Kunming, China
| | - Cuiping Yang
- grid.419010.d0000 0004 1792 7072Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, 650223 Kunming, Yunnan China
| | - Yongbin Chen
- grid.419010.d0000 0004 1792 7072Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, 650223 Kunming, Yunnan China ,grid.9227.e0000000119573309Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223 Kunming, Yunnan China
| |
Collapse
|
32
|
Wu Y, Hirschi KK. Tissue-Resident Macrophage Development and Function. Front Cell Dev Biol 2021; 8:617879. [PMID: 33490082 PMCID: PMC7820365 DOI: 10.3389/fcell.2020.617879] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022] Open
Abstract
Tissue-resident macrophages have been associated with important and diverse biological processes such as native immunity, tissue homeostasis and angiogenesis during development and postnatally. Thus, it is critical to understand the origins and functions of tissue-resident macrophages, as well as mechanisms underlying their regulation. It is now well accepted that murine macrophages are produced during three consecutive waves of hematopoietic development. The first wave of macrophage formation takes place during primitive hematopoiesis, which occurs in the yolk sac, and gives rise to primitive erythroid, megakaryocyte and macrophage progenitors. These “primitive” macrophage progenitors ultimately give rise to microglia in the adult brain. The second wave, which also occurs in the yolk sac, generates multipotent erythro-myeloid progenitors (EMP), which give rise to tissue-resident macrophages. Tissue-resident macrophages derived from EMP reside in diverse niches of different tissues except the brain, and demonstrate tissue-specific functions therein. The third wave of macrophages derives from hematopoietic stem cells (HSC) that are formed in the aorta-gonad-mesonephros (AGM) region of the embryo and migrate to, and colonize, the fetal liver. These HSC-derived macrophages are a long-lived pool that will last throughout adulthood. In this review, we discuss the developmental origins of tissue-resident macrophages, their molecular regulation in specific tissues, and their impact on embryonic development and postnatal homeostasis.
Collapse
Affiliation(s)
- Yinyu Wu
- Department of Medicine, Yale University School of Medicine, New Haven, CT, United States.,Department of Genetics, Yale University School of Medicine, New Haven, CT, United States.,Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, United States.,Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States.,Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| | - Karen K Hirschi
- Department of Medicine, Yale University School of Medicine, New Haven, CT, United States.,Department of Genetics, Yale University School of Medicine, New Haven, CT, United States.,Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, United States.,Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States.,Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States.,Department of Cell Biology, Cardiovascular Research Center, University of Virginia, School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
33
|
Demirci S, Leonard A, Tisdale JF. Hematopoietic stem cells from pluripotent stem cells: Clinical potential, challenges, and future perspectives. Stem Cells Transl Med 2020; 9:1549-1557. [PMID: 32725882 PMCID: PMC7695636 DOI: 10.1002/sctm.20-0247] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/01/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
The generation of hematopoietic stem cells (HSCs) from induced pluripotent stem cells (iPSCs) is an active and promising area of research; however, generating engraftable HSCs remains a major obstacle. Ex vivo HSC derivation from renewable sources such as iPSCs offers an experimental tool for studying developmental hematopoiesis, disease modeling, and drug discovery, and yields tremendous therapeutic potential for malignant and nonmalignant hematological disorders. Although initial attempts mostly recapitulated yolk sac primitive/definitive hematopoiesis with inability to engraft, recent advances suggest the feasibility of engraftable HSC derivation from iPSCs utilizing ectopic transcription factor expression. Strategic development for de novo HSC generation includes further investigations of HSC ontogeny, and elucidation of critical signaling pathways, epigenetic modulations, HSC and iPSC microenvironment, and cell-cell interactions that contribute to stem cell biology and function.
Collapse
Affiliation(s)
- Selami Demirci
- Cellular and Molecular Therapeutics BranchNational Heart Lung and Blood Institutes (NHLBI), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Alexis Leonard
- Cellular and Molecular Therapeutics BranchNational Heart Lung and Blood Institutes (NHLBI), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - John F. Tisdale
- Cellular and Molecular Therapeutics BranchNational Heart Lung and Blood Institutes (NHLBI), National Institutes of Health (NIH)BethesdaMarylandUSA
| |
Collapse
|
34
|
Bernecker C, Ackermann M, Lachmann N, Rohrhofer L, Zaehres H, Araúzo-Bravo MJ, van den Akker E, Schlenke P, Dorn I. Enhanced Ex Vivo Generation of Erythroid Cells from Human Induced Pluripotent Stem Cells in a Simplified Cell Culture System with Low Cytokine Support. Stem Cells Dev 2019; 28:1540-1551. [PMID: 31595840 PMCID: PMC6882453 DOI: 10.1089/scd.2019.0132] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Red blood cell (RBC) differentiation from human induced pluripotent stem cells (hiPSCs) offers great potential for developmental studies and innovative therapies. However, ex vivo erythropoiesis from hiPSCs is currently limited by low efficiency and unphysiological conditions of common culture systems. Especially, the absence of a physiological niche may impair cell growth and lineage-specific differentiation. We here describe a simplified, xeno- and feeder-free culture system for prolonged RBC generation that uses low numbers of supporting cytokines [stem cell factor (SCF), erythropoietin (EPO), and interleukin 3 (IL-3)] and is based on the intermediate development of a “hematopoietic cell forming complex (HCFC).” From this HCFC, CD43+ hematopoietic cells (purity >95%) were continuously released into the supernatant and could be collected repeatedly over a period of 6 weeks for further erythroid differentiation. The released cells were mainly CD34+/CD45+ progenitors with high erythroid colony-forming potential and CD36+ erythroid precursors. A total of 1.5 × 107 cells could be harvested from the supernatant of one six-well plate, showing 100- to 1000-fold amplification during subsequent homogeneous differentiation into GPA+ erythroid cells. Mean enucleation rates near 40% (up to 60%) further confirmed the potency of the system. These benefits may be explained by the generation of a niche within the HCFC that mimics the spatiotemporal signaling of the physiological microenvironment in which erythropoiesis occurs. Compared to other protocols, this method provides lower complexity, less cytokine and medium consumption, higher cellular output, and better enucleation. In addition, slight modifications in cytokine addition shift the system toward continuous generation of granulocytes and macrophages.
Collapse
Affiliation(s)
- Claudia Bernecker
- Department of Blood Group Serology and Transfusion Medicine, Medical University Graz, Graz, Austria
| | - Mania Ackermann
- RG Translational Hematology of Congenital Diseases, Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Nico Lachmann
- RG Translational Hematology of Congenital Diseases, Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Lisa Rohrhofer
- Department of Blood Group Serology and Transfusion Medicine, Medical University Graz, Graz, Austria
| | - Holm Zaehres
- Department of Anatomy and Molecular Embryology, Ruhr-University Bochum, Bochum, Germany
| | - Marcos J Araúzo-Bravo
- Computational Biology and Systems Biomedicine Research Group, Biodonostia Health Research Institute, San Sebastián, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | | | - Peter Schlenke
- Department of Blood Group Serology and Transfusion Medicine, Medical University Graz, Graz, Austria
| | - Isabel Dorn
- Department of Blood Group Serology and Transfusion Medicine, Medical University Graz, Graz, Austria
| |
Collapse
|
35
|
Khalil S, Ariel Gru A, Saavedra AP. Cutaneous extramedullary haematopoiesis: Implications in human disease and treatment. Exp Dermatol 2019; 28:1201-1209. [DOI: 10.1111/exd.14013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 06/26/2019] [Accepted: 07/11/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Shadi Khalil
- Department of Dermatology University of Virginia School of Medicine Charlottesville Virginia
| | - Alejandro Ariel Gru
- Department of Pathology University of Virginia School of Medicine Charlottesville Virginia
| | - Arturo P. Saavedra
- Department of Dermatology University of Virginia School of Medicine Charlottesville Virginia
| |
Collapse
|
36
|
Trakarnsanga K, Ferguson D, Daniels DE, Griffiths RE, Wilson MC, Mordue KE, Gartner A, Andrienko TN, Calvert A, Condie A, McCahill A, Mountford JC, Toye AM, Anstee DJ, Frayne J. Vimentin expression is retained in erythroid cells differentiated from human iPSC and ESC and indicates dysregulation in these cells early in differentiation. Stem Cell Res Ther 2019; 10:130. [PMID: 31036072 PMCID: PMC6489253 DOI: 10.1186/s13287-019-1231-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/02/2019] [Accepted: 04/04/2019] [Indexed: 05/16/2023] Open
Abstract
Background Pluripotent stem cells are attractive progenitor cells for the generation of erythroid cells in vitro as have expansive proliferative potential. However, although embryonic (ESC) and induced pluripotent (iPSC) stem cells can be induced to undergo erythroid differentiation, the majority of cells fail to enucleate and the molecular basis of this defect is unknown. One protein that has been associated with the initial phase of erythroid cell enucleation is the intermediate filament vimentin, with loss of vimentin potentially required for the process to proceed. Methods In this study, we used our established erythroid culture system along with western blot, PCR and interegation of comparative proteomic data sets to analyse the temporal expression profile of vimentin in erythroid cells differentiated from adult peripheral blood stem cells, iPSC and ESC throughout erythropoiesis. Confocal microscopy was also used to examine the intracellular localisation of vimentin. Results We show that expression of vimentin is turned off early during normal adult erythroid cell differentiation, with vimentin protein lost by the polychromatic erythroblast stage, just prior to enucleation. In contrast, in erythroid cells differentiated from iPSC and ESC, expression of vimentin persists, with high levels of both mRNA and protein even in orthochromatic erythroblasts. In the vimentin-positive iPSC orthochromatic erythroblasts, F-actin was localized around the cell periphery; however, in those rare cells captured undergoing enucleation, vimentin was absent and F-actin was re-localized to the enucleosome as found in normal adult orthrochromatic erythroblasts. Conclusion As both embryonic and adult erythroid cells loose vimentin and enucleate, retention of vimentin by iPSC and ESC erythroid cells indicates an intrinsic defect. By analogy with avian erythrocytes which naturally retain vimentin and remain nucleated, retention in iPSC- and ESC-derived erythroid cells may impede enucleation. Our data also provide the first evidence that dysregulation of processes in these cells occurs from the early stages of differentiation, facilitating targeting of future studies. Electronic supplementary material The online version of this article (10.1186/s13287-019-1231-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kongtana Trakarnsanga
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK.,Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Daniel Ferguson
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | - Deborah E Daniels
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK.,NIHR Blood and Transplant Research Unit, University of Bristol, Bristol, BS8 1TD, UK
| | - Rebecca E Griffiths
- Bristol Institute for Transfusion Sciences, National Health Service Blood and Transplant (NHSBT), Bristol, BS34 7QH, UK.,NIHR Blood and Transplant Research Unit, University of Bristol, Bristol, BS8 1TD, UK
| | | | - Kathryn E Mordue
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | - Abi Gartner
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | - Tatyana N Andrienko
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK.,NIHR Blood and Transplant Research Unit, University of Bristol, Bristol, BS8 1TD, UK
| | - Annabel Calvert
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | - Alison Condie
- Scottish National Blood Transfusion Service, Jack Copland Centre, Heriot Watt Research Park, Edinburgh, EH14 4AP, UK
| | - Angela McCahill
- Scottish National Blood Transfusion Service, Jack Copland Centre, Heriot Watt Research Park, Edinburgh, EH14 4AP, UK
| | - Joanne C Mountford
- Scottish National Blood Transfusion Service, Jack Copland Centre, Heriot Watt Research Park, Edinburgh, EH14 4AP, UK
| | - Ashley M Toye
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK.,Bristol Institute for Transfusion Sciences, National Health Service Blood and Transplant (NHSBT), Bristol, BS34 7QH, UK.,NIHR Blood and Transplant Research Unit, University of Bristol, Bristol, BS8 1TD, UK
| | - David J Anstee
- Bristol Institute for Transfusion Sciences, National Health Service Blood and Transplant (NHSBT), Bristol, BS34 7QH, UK.,NIHR Blood and Transplant Research Unit, University of Bristol, Bristol, BS8 1TD, UK
| | - Jan Frayne
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK. .,NIHR Blood and Transplant Research Unit, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
37
|
Huang ZW, Fong CY, Gauthaman K, Sukumar P, Mahyuddin AP, Barrett AN, Bongso A, Choolani M. Biology of human primitive erythroblasts for application in noninvasive prenatal diagnosis. Prenat Diagn 2018; 38:673-684. [PMID: 29876942 DOI: 10.1002/pd.5295] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 05/23/2018] [Accepted: 05/30/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Human primitive erythroblasts produced during early embryogenesis have been found in maternal circulation at early gestation and are considered good target cells for noninvasive prenatal diagnosis. We aimed to gain a better understanding of the biology of primitive erythroblasts and maximize their potential utility for noninvasive prenatal diagnosis. METHODS Cells were obtained from first trimester human placental tissues. Biological properties including surface antigen composition, differentiation, proliferation, enucleation, and degeneration were studied as gestation progressed. A microdroplet culture system was developed to observe the behavior of these cells in vitro. RESULTS Histology showed that primitive erythroblasts undergo maturation from polychromatic to orthochromatic erythroblasts and can differentiate spontaneously in vitro. Cell surface markers and nuclear gene expression suggest that the cells do not possess stemness properties, despite being primitive in nature. They have limited proliferative activity and highly deacetylated chromatin, but a microdroplet culture system can prolong their viability under normoxic conditions. No apoptosis was seen by 11 weeks' gestation, and there was no enucleation in vitro. CONCLUSION These properties confirm that viable cells with intact nuclei can be obtained at very early gestation for genetic analysis.
Collapse
Affiliation(s)
- Zhou-Wei Huang
- National University of Singapore, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, 119228, Singapore
| | - Chui-Yee Fong
- National University of Singapore, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, 119228, Singapore
| | - Kalamegam Gauthaman
- King Abdulaziz University, King Fahd Medical Research Centre, Jeddah, 21589, Saudi Arabia
| | - Ponnusamy Sukumar
- National University of Singapore, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, 119228, Singapore.,Management Development Institute of Singapore, 501 Stirling Road, 148951, Singapore
| | - Aniza P Mahyuddin
- National University of Singapore, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, 119228, Singapore
| | - Angela N Barrett
- National University of Singapore, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, 119228, Singapore
| | - Ariff Bongso
- National University of Singapore, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, 119228, Singapore
| | - Mahesh Choolani
- National University of Singapore, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, 119228, Singapore
| |
Collapse
|
38
|
Soares MJ, Iqbal K, Kozai K. Hypoxia and Placental Development. Birth Defects Res 2018; 109:1309-1329. [PMID: 29105383 DOI: 10.1002/bdr2.1135] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/04/2017] [Indexed: 12/17/2022]
Abstract
Hemochorial placentation is orchestrated through highly regulated temporal and spatial decisions governing the fate of trophoblast stem/progenitor cells. Trophoblast cell acquisition of specializations facilitating invasion and uterine spiral artery remodeling is a labile process, sensitive to the environment, and represents a process that is vulnerable to dysmorphogenesis in pathologic states. Hypoxia is a signal guiding placental development, and molecular mechanisms directing cellular adaptations to low oxygen tension are integral to trophoblast cell differentiation and placentation. Hypoxia can also be used as an experimental tool to investigate regulatory processes controlling hemochorial placentation. These developmental processes are conserved in mouse, rat, and human placentation. Consequently, elements of these developmental events can be modeled and hypotheses tested in trophoblast stem cells and in genetically manipulated rodents. Hypoxia is also a consequence of a failed placenta, yielding pathologies that can adversely affect maternal adjustments to pregnancy, fetal health, and susceptibility to adult disease. The capacity of the placenta for adaptation to environmental challenges highlights the importance of its plasticity in safeguarding a healthy pregnancy. Birth Defects Research 109:1309-1329, 2017.© 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas.,Fetal Health Research, Children's Research Institute, Children's Mercy, Kansas City, Missouri
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Keisuke Kozai
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
39
|
|
40
|
Definitive Erythropoiesis from Pluripotent Stem Cells: Recent Advances and Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1107:1-13. [PMID: 29876866 DOI: 10.1007/5584_2018_228] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Derivation of functional and mature red blood cells (RBCs) with adult globin expression from renewable source such as induced pluripotent stem cells (iPSCs) is of importance from the clinical point of view. Definitive RBC generation can only be succeeded through production of true hematopoietic stem cells (HSCs). There has been a great effort to obtain definitive engraftable HSCs from iPSCs but the results were mostly unsatisfactory due to low, short-term and linage-biased engraftment in mouse models. Moreover, ex vivo differentiation approaches ended up with RBCs with mostly embryonic and fetal globin expression. To establish reliable, standardized and effective laboratory protocols, we need to expand our knowledge about developmental hematopoiesis/erythropoiesis and identify critical regulatory signaling pathways and transcription factors. Once we meet these challenges, we could establish differentiation protocols for massive RBC production for transfusion purposes in the clinical setting, performing drug screening and disease modeling in ex vivo conditions, and investigating the embryological cascade of erythropoiesis. More interestingly, with the introduction of relatively efficient and facile genome editing tools, genetic correction for inherited RBC disorders such as sickle cell disease (SCD) would become possible through iPSCs that can subsequently generate definitive HSCs, which then give rise to definitive RBCs producing β-globin after transplantation.
Collapse
|
41
|
Rossmann MP, Orkin SH, Chute JP. Hematopoietic Stem Cell Biology. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
42
|
Chondrou V, Kolovos P, Sgourou A, Kourakli A, Pavlidaki A, Kastrinou V, John A, Symeonidis A, Ali BR, Papachatzopoulou A, Katsila T, Patrinos GP. Whole transcriptome analysis of human erythropoietic cells during ontogenesis suggests a role of VEGFA gene as modulator of fetal hemoglobin and pharmacogenomic biomarker of treatment response to hydroxyurea in β-type hemoglobinopathy patients. Hum Genomics 2017; 11:24. [PMID: 29061162 PMCID: PMC5654038 DOI: 10.1186/s40246-017-0120-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 10/16/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Human erythropoiesis is characterized by distinct gene expression profiles at various developmental stages. Previous studies suggest that fetal-to-adult hemoglobin switch is regulated by a complex mechanism, in which many key players still remain unknown. Here, we report our findings from whole transcriptome analysis of erythroid cells, isolated from erythroid tissues at various developmental stages in an effort to identify distinct molecular signatures of each erythroid tissue. RESULTS From our in-depth data analysis, pathway analysis, and text mining, we opted to focus on the VEGFA gene, given its gene expression characteristics. Selected VEGFA genomic variants, identified through linkage disequilibrium analysis, were explored further for their association with elevated fetal hemoglobin levels in β-type hemoglobinopathy patients. Our downstream analysis of non-transfusion-dependent β-thalassemia patients, β-thalassemia major patients, compound heterozygous sickle cell disease/β-thalassemia patients receiving hydroxyurea as fetal hemoglobin augmentation treatment, and non-thalassemic individuals indicated that VEGFA genomic variants were associated with disease severity in β-thalassemia patients and hydroxyurea treatment efficacy in SCD/β-thalassemia compound heterozygous patients. CONCLUSIONS Our findings suggest that VEGFA may act as a modifier gene of human globin gene expression and, at the same time, serve as a genomic biomarker in β-type hemoglobinopathy disease severity and hydroxyurea treatment efficacy.
Collapse
Affiliation(s)
- Vasiliki Chondrou
- Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, GR-265 04, Patras, Greece
| | - Petros Kolovos
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | | | - Alexandra Kourakli
- Hematology Division, Department of Internal Medicine, Faculty of Medicine, University of Patras, Patras, Greece
| | - Alexia Pavlidaki
- Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, GR-265 04, Patras, Greece
- Present address: Institut de Génétique et de Biologie Moléculaire et Cellulaire IGBMC/CNRS/INSERM/UDS, 67404 ILLKIRCH, BP 10142, CU de Strasbourg, France
| | - Vlasia Kastrinou
- Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, GR-265 04, Patras, Greece
| | - Anne John
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Argiris Symeonidis
- Hematology Division, Department of Internal Medicine, Faculty of Medicine, University of Patras, Patras, Greece
| | - Bassam R Ali
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | | | - Theodora Katsila
- Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, GR-265 04, Patras, Greece
| | - George P Patrinos
- Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, GR-265 04, Patras, Greece.
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.
| |
Collapse
|
43
|
Ivanovs A, Rybtsov S, Ng ES, Stanley EG, Elefanty AG, Medvinsky A. Human haematopoietic stem cell development: from the embryo to the dish. Development 2017; 144:2323-2337. [PMID: 28676567 DOI: 10.1242/dev.134866] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Haematopoietic stem cells (HSCs) emerge during embryogenesis and give rise to the adult haematopoietic system. Understanding how early haematopoietic development occurs is of fundamental importance for basic biology and medical sciences, but our knowledge is still limited compared with what we know of adult HSCs and their microenvironment. This is particularly true for human haematopoiesis, and is reflected in our current inability to recapitulate the development of HSCs from pluripotent stem cells in vitro In this Review, we discuss what is known of human haematopoietic development: the anatomical sites at which it occurs, the different temporal waves of haematopoiesis, the emergence of the first HSCs and the signalling landscape of the haematopoietic niche. We also discuss the extent to which in vitro differentiation of human pluripotent stem cells recapitulates bona fide human developmental haematopoiesis, and outline some future directions in the field.
Collapse
Affiliation(s)
- Andrejs Ivanovs
- Institute for Stem Cell Research, MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK.,Institute of Anatomy and Anthropology, Riga Stradiņš University, Riga LV-1007, Latvia
| | - Stanislav Rybtsov
- Institute for Stem Cell Research, MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Elizabeth S Ng
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, Victoria 3052, Australia.,Department of Anatomy and Developmental Biology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Edouard G Stanley
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, Victoria 3052, Australia.,Department of Anatomy and Developmental Biology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria 3800, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Andrew G Elefanty
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, Victoria 3052, Australia .,Department of Anatomy and Developmental Biology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria 3800, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Alexander Medvinsky
- Institute for Stem Cell Research, MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| |
Collapse
|
44
|
Liu X, Zhang Y, Ni M, Cao H, Signer RA, Li D, Li M, Gu Z, Hu Z, Dickerson KE, Weinberg SE, Chandel NS, DeBerardinis RJ, Zhou F, Shao Z, Xu J. Regulation of mitochondrial biogenesis in erythropoiesis by mTORC1-mediated protein translation. Nat Cell Biol 2017; 19:626-638. [PMID: 28504707 PMCID: PMC5771482 DOI: 10.1038/ncb3527] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 04/06/2017] [Indexed: 12/15/2022]
Abstract
Advances in genomic profiling present new challenges of explaining how changes in DNA and RNA are translated into proteins linking genotype to phenotype. Here we compare the genome-scale proteomic and transcriptomic changes in human primary haematopoietic stem/progenitor cells and erythroid progenitors, and uncover pathways related to mitochondrial biogenesis enhanced through post-transcriptional regulation. Mitochondrial factors including TFAM and PHB2 are selectively regulated through protein translation during erythroid specification. Depletion of TFAM in erythroid cells alters intracellular metabolism, leading to elevated histone acetylation, deregulated gene expression, and defective mitochondria and erythropoiesis. Mechanistically, mTORC1 signalling is enhanced to promote translation of mitochondria-associated transcripts through TOP-like motifs. Genetic and pharmacological perturbation of mitochondria or mTORC1 specifically impairs erythropoiesis in vitro and in vivo. Our studies support a mechanism for post-transcriptional control of erythroid mitochondria and may have direct relevance to haematologic defects associated with mitochondrial diseases and ageing.
Collapse
Affiliation(s)
- Xin Liu
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yuannyu Zhang
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Key Laboratory of Computational Biology, Collaborative Innovation Center for Genetics and Developmental Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Min Ni
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hui Cao
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Robert A.J. Signer
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Dan Li
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mushan Li
- Key Laboratory of Computational Biology, Collaborative Innovation Center for Genetics and Developmental Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhimin Gu
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zeping Hu
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kathryn E. Dickerson
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Samuel E. Weinberg
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Navdeep S. Chandel
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ralph J. DeBerardinis
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Feng Zhou
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Zhen Shao
- Key Laboratory of Computational Biology, Collaborative Innovation Center for Genetics and Developmental Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jian Xu
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
45
|
Masoumi Z, Familari M, Källén K, Ranstam J, Olofsson P, Hansson SR. Fetal hemoglobin in umbilical cord blood in preeclamptic and normotensive pregnancies: A cross-sectional comparative study. PLoS One 2017; 12:e0176697. [PMID: 28453539 PMCID: PMC5409527 DOI: 10.1371/journal.pone.0176697] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/14/2017] [Indexed: 12/14/2022] Open
Abstract
Preeclampsia (PE) is associated with increased fetal hemoglobin (HbF) in the maternal circulation but its source is unknown. To investigate whether excessive HbF is produced in the placenta or the fetus, the concentration of HbF (cHbF) in the arterial and venous umbilical cord blood (UCB) was compared in 15825 normotensive and 444 PE pregnancies. The effect of fetal gender on cHbF was also evaluated in both groups. Arterial and venous UCB sampled immediately after birth at 36-42 weeks of gestation were analyzed for total Hb concentration (ctHb) (g/L) and HbF% using a Radiometer blood gas analyzer. Non-parametric tests were used for statistical comparison and P values < 0.05 were considered significant. Our results indicated higher cHbF in venous compared to arterial UCB in both normotensive (118.90 vs 117.30) and PE (126.75 vs 120.12) groups. In PE compared to normotensive pregnancies, a significant increase was observed in arterial and venous ctHb (171.00 vs 166.00 and 168.00 vs 163.00, respectively) while cHbF was only significantly increased in venous UCB (126.75 vs 118.90). The pattern was similar in both genders. These results indicate a substantial placental contribution to HbF levels in UCB, which increases in PE and is independent of fetal gender, suggesting the elevated cHbF evident in PE results from placental dysfunction.
Collapse
Affiliation(s)
- Zahra Masoumi
- Department of Clinical Sciences Lund, Division of Obstetrics and Gynecology, Lund University, Lund, Sweden
| | - Mary Familari
- School of Biosciences, University of Melbourne, Parkville, Australia
| | - Karin Källén
- Center for Reproductive Epidemiology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Jonas Ranstam
- Department of Clinical Sciences Lund, Division of Orthopedics, Faculty of Medicine, Lund University, Lund, Sweden
| | - Per Olofsson
- Department of Clinical Sciences Malmö, Division of Obstetrics and Gynecology, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Stefan R. Hansson
- Department of Clinical Sciences Lund, Division of Obstetrics and Gynecology, Lund University, Lund, Sweden
- Department of Clinical Sciences Malmö, Division of Obstetrics and Gynecology, Lund University, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
46
|
Gunnarsson R, Åkerström B, Hansson SR, Gram M. Recombinant alpha-1-microglobulin: a potential treatment for preeclampsia. Drug Discov Today 2017; 22:736-743. [DOI: 10.1016/j.drudis.2016.12.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 11/21/2016] [Accepted: 12/08/2016] [Indexed: 01/31/2023]
|
47
|
Schroeder DI, Schmidt RJ, Crary-Dooley FK, Walker CK, Ozonoff S, Tancredi DJ, Hertz-Picciotto I, LaSalle JM. Placental methylome analysis from a prospective autism study. Mol Autism 2016; 7:51. [PMID: 28018572 PMCID: PMC5159983 DOI: 10.1186/s13229-016-0114-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/28/2016] [Indexed: 11/10/2022] Open
Abstract
Background Autism spectrum disorders (ASD) are increasingly prevalent neurodevelopmental disorders that are behaviorally diagnosed in early childhood. Most ASD cases likely arise from a complex mixture of genetic and environmental factors, an interface where the epigenetic marks of DNA methylation may be useful as risk biomarkers. The placenta is a potentially useful surrogate tissue characterized by a methylation pattern of partially methylated domains (PMDs) and highly methylated domains (HMDs) reflective of methylation patterns observed in the early embryo. Methods In this study, we investigated human term placentas from the MARBLES (Markers of Autism Risk in Babies: Learning Early Signs) prospective study by whole genome bisulfite sequencing. We also examined the utility of PMD/HMDs in detecting methylation differences consistent with ASD diagnosis at age three. Results We found that while human placental methylomes have highly reproducible PMD and HMD locations, there is a greater variation between individuals in methylation levels over PMDs than HMDs due to both sampling and individual variability. In a comparison of methylation differences in placental samples from 24 ASD and 23 typically developing (TD) children, a HMD containing a putative fetal brain enhancer near DLL1 was found to reach genome-wide significance and was validated for significantly higher methylation in ASD by pyrosequencing. Conclusions These results suggest that the placenta could be an informative surrogate tissue for predictive ASD biomarkers in high-risk families. Electronic supplementary material The online version of this article (doi:10.1186/s13229-016-0114-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Diane I Schroeder
- Department of Medical Microbiology and Immunology, Genome Center, Davis, CA 95616 USA
| | - Rebecca J Schmidt
- Department of Public Health Sciences, University of California, Davis, CA 95616 USA ; MIND Institute, University of California, Davis, CA 95616 USA
| | | | - Cheryl K Walker
- Department of Obstetrics and Gynecology, University of California, Davis, CA 95616 USA ; MIND Institute, University of California, Davis, CA 95616 USA
| | - Sally Ozonoff
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA 95616 USA ; MIND Institute, University of California, Davis, CA 95616 USA
| | - Daniel J Tancredi
- Department of Pediatrics, University of California, Davis, CA 95616 USA
| | - Irva Hertz-Picciotto
- Department of Public Health Sciences, University of California, Davis, CA 95616 USA ; MIND Institute, University of California, Davis, CA 95616 USA
| | - Janine M LaSalle
- Department of Medical Microbiology and Immunology, Genome Center, Davis, CA 95616 USA ; MIND Institute, University of California, Davis, CA 95616 USA
| |
Collapse
|
48
|
Capellera-Garcia S, Flygare J. Direct lineage reprogramming: a useful addition to the blood cell research toolbox. Expert Rev Hematol 2016; 10:107-109. [PMID: 27967256 DOI: 10.1080/17474086.2017.1272409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Sandra Capellera-Garcia
- a Department of Molecular Medicine and Gene Therapy , Lund Stem Cell Center, Lund University , Lund , Sweden
| | - Johan Flygare
- a Department of Molecular Medicine and Gene Therapy , Lund Stem Cell Center, Lund University , Lund , Sweden
| |
Collapse
|
49
|
Kim AR, Sankaran VG. Development of autologous blood cell therapies. Exp Hematol 2016; 44:887-94. [PMID: 27345108 DOI: 10.1016/j.exphem.2016.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 12/21/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation and blood cell transfusions are performed commonly in patients with a variety of blood disorders. Unfortunately, these donor-derived cell therapies are constrained due to limited supplies, infectious risk factors, a lack of appropriately matched donors, and the risk of immunologic complications from such products. The use of autologous cell therapies has been proposed to overcome these shortcomings. One can derive such therapies directly from hematopoietic stem and progenitor cells of individuals, which can then be manipulated ex vivo to produce the desired modifications or differentiated to produce a particular target population. Alternatively, pluripotent stem cells, which have a theoretically unlimited self-renewal capacity and an ability to differentiate into any desired cell type, can be used as an autologous starting source for such manipulation and differentiation approaches. Such cell products can also be used as a delivery vehicle for therapeutics. In this review, we highlight recent advances and discuss ongoing challenges for the in vitro generation of autologous hematopoietic cells that can be used for cell therapy.
Collapse
Affiliation(s)
- Ah Ram Kim
- Division of Hematology/Oncology, Boston Children's Hospital, and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Vijay G Sankaran
- Division of Hematology/Oncology, Boston Children's Hospital, and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
| |
Collapse
|
50
|
Abstract
Optimal iron nutrition in utero is essential for development of the fetus and helps establish birth iron stores adequate to sustain growth in early infancy. In species with hemochorial placentas, such as humans and rodents, iron in the maternal circulation is transferred to the fetus by directly contacting placental syncytiotrophoblasts. Early kinetic studies provided valuable data on the initial uptake of maternal transferrin, an iron-binding protein, by the placenta. However, the remaining steps of iron trafficking across syncytiotrophoblasts and through the fetal endothelium into the fetal blood remain poorly characterized. Over the last 20 years, identification of transmembrane iron transporters and the iron regulatory hormone hepcidin has greatly expanded the knowledge of cellular iron transport and its regulation by systemic iron status. In addition, emerging human and animal data demonstrating comprised fetal iron stores in severe maternal iron deficiency challenge the classic dogma of exclusive fetal control over the transfer process and indicate that maternal and local signals may play a role in regulating this process. This review compiles current data on the kinetic, molecular, and regulatory aspects of placental iron transport and considers new questions and knowledge gaps raised by these advances.
Collapse
Affiliation(s)
- Chang Cao
- C. Cao and M.D. Fleming are with the Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Mark D Fleming
- C. Cao and M.D. Fleming are with the Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, USA.
| |
Collapse
|