151
|
Kuppers DA, Arora S, Lim Y, Lim AR, Carter LM, Corrin PD, Plaisier CL, Basom R, Delrow JJ, Wang S, Hansen He H, Torok-Storb B, Hsieh AC, Paddison PJ. N 6-methyladenosine mRNA marking promotes selective translation of regulons required for human erythropoiesis. Nat Commun 2019; 10:4596. [PMID: 31601799 PMCID: PMC6787028 DOI: 10.1038/s41467-019-12518-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 08/26/2019] [Indexed: 12/30/2022] Open
Abstract
Many of the regulatory features governing erythrocyte specification, maturation, and associated disorders remain enigmatic. To identify new regulators of erythropoiesis, we utilize a functional genomic screen for genes affecting expression of the erythroid marker CD235a/GYPA. Among validating hits are genes coding for the N6-methyladenosine (m6A) mRNA methyltransferase (MTase) complex, including, METTL14, METTL3, and WTAP. We demonstrate that m6A MTase activity promotes erythroid gene expression programs through selective translation of ~300 m6A marked mRNAs, including those coding for SETD histone methyltransferases, ribosomal components, and polyA RNA binding proteins. Remarkably, loss of m6A marks results in dramatic loss of H3K4me3 marks across key erythroid-specific KLF1 transcriptional targets (e.g., Heme biosynthesis genes). Further, each m6A MTase subunit and a subset of their mRNAs targets are required for human erythroid specification in primary bone-marrow derived progenitors. Thus, m6A mRNA marks promote the translation of a network of genes required for human erythropoiesis.
Collapse
Affiliation(s)
- Daniel A Kuppers
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Sonali Arora
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Yiting Lim
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Andrea R Lim
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, 98195, USA
| | - Lucas M Carter
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Philip D Corrin
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Christopher L Plaisier
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85281, USA
| | - Ryan Basom
- Genomics Shared Resource, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Jeffrey J Delrow
- Genomics Shared Resource, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Shiyan Wang
- Princess Margaret Cancer Centre/University Health Network, Toronto, ON, Canada
| | - Housheng Hansen He
- Princess Margaret Cancer Centre/University Health Network, Toronto, ON, Canada
| | - Beverly Torok-Storb
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Andrew C Hsieh
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
- School of Medicine, University of Washington, Seattle, WA, 98195, USA.
| | - Patrick J Paddison
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| |
Collapse
|
152
|
Fan X, Wu C, Truitt LL, Espinoza DA, Sellers S, Bonifacino A, Zhou Y, Cordes SF, Krouse A, Metzger M, Donahue RE, Lu R, Dunbar CE. Clonal tracking of erythropoiesis in rhesus macaques. Haematologica 2019; 105:1813-1824. [PMID: 31582555 PMCID: PMC7327626 DOI: 10.3324/haematol.2019.231811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/03/2019] [Indexed: 12/25/2022] Open
Abstract
The classical model of hematopoietic hierarchies is being reconsidered on the basis of data from in vitro assays and single cell expression profiling. Recent experiments suggested that the erythroid lineage might differentiate directly from multipotent hematopoietic stem cells / progenitors or from a highly biased subpopulation of stem cells, rather than transiting through common myeloid progenitors or megakaryocyte-erythrocyte progenitors. We genetically barcoded autologous rhesus macaque stem and progenitor cells, allowing quantitative tracking of the in vivo clonal output of thousands of individual cells over time following transplantation. CD34+ cells were lentiviral-transduced with a high diversity barcode library, with the barcode in an expressed region of the provirus, allowing barcode retrieval from DNA or RNA, with each barcode representing an individual stem or progenitor cell clone. Barcode profiles from bone marrow CD45-CD71+ maturing nucleated red blood cells were compared with other lineages purified from the same bone marrow sample. There was very high correlation of barcode contributions between marrow nucleated red blood cells and other lineages, with the highest correlation between nucleated red blood cells and myeloid lineages, whether at earlier or later time points post transplantation, without obvious clonal contributions from highly erythroid-biased or restricted clones. A similar profile occurred even under stressors such as aging or erythropoietin stimulation. RNA barcode analysis on circulating mature red blood cells followed over long time periods demonstrated stable erythroid clonal contributions. Overall, in this nonhuman primate model with great relevance to human hematopoiesis, we documented continuous production of erythroid cells from multipotent, non-biased hematopoietic stem cell clones at steady-state or under stress.
Collapse
Affiliation(s)
- Xing Fan
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, MA, USA
| | - Chuanfeng Wu
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, MA, USA
| | - Lauren L Truitt
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, MA, USA
| | - Diego A Espinoza
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, MA, USA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephanie Sellers
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, MA, USA
| | - Aylin Bonifacino
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, MA, USA
| | - Yifan Zhou
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, MA, USA.,Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Stefan F Cordes
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, MA, USA
| | - Allen Krouse
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, MA, USA
| | - Mark Metzger
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, MA, USA
| | - Robert E Donahue
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, MA, USA
| | - Rong Lu
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Cynthia E Dunbar
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, MA, USA
| |
Collapse
|
153
|
Microphysiological systems in the evaluation of hematotoxicities during drug development. CURRENT OPINION IN TOXICOLOGY 2019. [DOI: 10.1016/j.cotox.2019.09.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
154
|
Gomes AC, Moreira AC, Silva T, Neves JV, Mesquita G, Almeida AA, Barreira-Silva P, Fernandes R, Resende M, Appelberg R, Rodrigues PNS, Gomes MS. IFN-γ–Dependent Reduction of Erythrocyte Life Span Leads to Anemia during Mycobacterial Infection. THE JOURNAL OF IMMUNOLOGY 2019; 203:2485-2496. [DOI: 10.4049/jimmunol.1900382] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 09/04/2019] [Indexed: 12/26/2022]
|
155
|
Affiliation(s)
- Tomas Ganz
- From the Departments of Medicine and Pathology, David Geffen School of Medicine at UCLA, Los Angeles
| |
Collapse
|
156
|
Infused wild-type macrophages reside and self-renew in the liver to rescue the hemolysis and anemia of Hmox1-deficient mice. Blood Adv 2019; 2:2732-2743. [PMID: 30337301 DOI: 10.1182/bloodadvances.2018019737] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 09/05/2018] [Indexed: 12/13/2022] Open
Abstract
Heme oxygenase 1 (HMOX1), the inducible enzyme that catabolizes the degradation of heme into biliverdin, iron, and carbon monoxide, plays an essential role in the clearance of senescent and damaged red blood cells, systemic iron homeostasis, erythropoiesis, vascular hemostasis, and oxidative and inflammatory stress responses. In humans, HMOX1 deficiency causes a rare and lethal disease, characterized by severe anemia, intravascular hemolysis, as well as vascular and tissue damage. Hmox1 knockout (KO) mice recapitulated the phenotypes of HMOX1-deficiency patients and could be rescued by bone marrow (BM) transplantation that engrafted donor's hematopoietic stem cells into the recipient animals after myeloablation. To find better therapy and elucidate the contribution of macrophages to the pathogenesis of HMOX1-deficiency disease, we infused wild-type (WT) macrophages into Hmox1 KO mice. Results showed that WT macrophages engrafted and proliferated in the livers of Hmox1 KO mice, which corrected the microcytic anemia, rescued the intravascular hemolysis, restored iron homeostasis, eliminated kidney iron overload and tissue damage, and provided long-term protection. These results showed that a single macrophage infusion delivered a long-term curative effect in Hmox1 KO mice, obviating the need for BM transplantation, and suggested that the HMOX1 disease stems mainly from the loss of viable reticuloendothelial macrophages. Our work provides new insights into the etiology of HMOX1 deficiency and demonstrates the potential of infusion of WT macrophages to prevent disease in patients with HMOX1 deficiency and potentially other macrophage-related diseases.
Collapse
|
157
|
Scully EJ, Shabani E, Rangel GW, Grüring C, Kanjee U, Clark MA, Chaand M, Kurita R, Nakamura Y, Ferreira MU, Duraisingh MT. Generation of an immortalized erythroid progenitor cell line from peripheral blood: A model system for the functional analysis of Plasmodium spp. invasion. Am J Hematol 2019; 94:963-974. [PMID: 31148215 PMCID: PMC6984401 DOI: 10.1002/ajh.25543] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/20/2019] [Accepted: 05/28/2019] [Indexed: 02/06/2023]
Abstract
Malaria pathogenesis is caused by the replication of Plasmodium parasites within the red blood cells (RBCs) of the vertebrate host. This selective pressure has favored the evolution of protective polymorphisms in erythrocyte proteins, a subset of which serve as cognate receptors for parasite invasion ligands. Recently, the generation of RBCs from immortalized hematopoietic stem cells (HSCs) has offered a more tractable system for genetic manipulation and long-term in vitro culture, enabling elucidation of the functional determinants of host susceptibility in vitro. Here we report the generation of an immortalized erythroid progenitor cell line (EJ cells) from as few as 100 000 peripheral blood mononuclear cells. It offers a robust method for the creation of customized model systems from small volumes of peripheral blood. The EJ cell differentiation mirrored erythropoiesis of primary HSCs, yielding orthochromatic erythroblasts and enucleated RBCs after eight days (ejRBCs). The ejRBCs supported invasion by both P. vivax and P. falciparum. To demonstrate the genetic tractability of this system, we used CRISPR/Cas9 to disrupt the Duffy Antigen/Receptor for Chemokines (DARC) gene, which encodes the canonical receptor of P. vivax in humans. Invasion of P. vivax into this DARC-knockout cell line was strongly inhibited providing direct genetic evidence that P. vivax requires DARC for RBC invasion. Further, genetic complementation of DARC restored P. vivax invasion. Taken together, the peripheral blood immortalization method presented here offers the capacity to generate biologically representative model systems for studies of blood-stage malaria invasion from the peripheral blood of donors harboring unique genetic backgrounds, or rare polymorphisms.
Collapse
Affiliation(s)
- Erik J. Scully
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, United States of America
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America
- Broad Institute, Cambridge, MA, United States of America
| | - Estela Shabani
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America
| | - Gabriel W. Rangel
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America
| | - Christof Grüring
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America
| | - Usheer Kanjee
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America
| | - Martha A. Clark
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America
| | - Mudit Chaand
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America
| | - Ryo Kurita
- Department of Research and Development, Central Blood Institute, Japanese Red Cross Society, Tokyo 135-8521, Japan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, Ibaraki 305-0074, Japan
| | - Marcelo U Ferreira
- Department of Parasitology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, BR 05508-900
| | - Manoj T. Duraisingh
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America
| |
Collapse
|
158
|
Pretini V, Koenen MH, Kaestner L, Fens MHAM, Schiffelers RM, Bartels M, Van Wijk R. Red Blood Cells: Chasing Interactions. Front Physiol 2019; 10:945. [PMID: 31417415 PMCID: PMC6684843 DOI: 10.3389/fphys.2019.00945] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/09/2019] [Indexed: 12/13/2022] Open
Abstract
Human red blood cells (RBC) are highly differentiated cells that have lost all organelles and most intracellular machineries during their maturation process. RBC are fundamental for the nearly all basic physiologic dynamics and they are key cells in the body's respiratory system by being responsible for the oxygen transport to all cells and tissues, and delivery of carbon dioxide to the lungs. With their flexible structure RBC are capable to deform in order to travel through all blood vessels including very small capillaries. Throughout their in average 120 days lifespan, human RBC travel in the bloodstream and come in contact with a broad range of different cell types. In fact, RBC are able to interact and communicate with endothelial cells (ECs), platelets, macrophages, and bacteria. Additionally, they are involved in the maintenance of thrombosis and hemostasis and play an important role in the immune response against pathogens. To clarify the mechanisms of interaction of RBC and these other cells both in health and disease as well as to highlight the role of important key players, we focused our interest on RBC membrane components such as ion channels, proteins, and phospholipids.
Collapse
Affiliation(s)
- Virginia Pretini
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Theoretical Medicine and Biosciences, Saarland University, Homburg, Germany
| | - Mischa H. Koenen
- Department of Laboratory of Translational Immunology and Department of Pediatric Immunology, Wilhelmina Children’s Hospital, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Lars Kaestner
- Theoretical Medicine and Biosciences, Saarland University, Homburg, Germany
- Experimental Physics, Saarland University, Saarbrücken, Germany
| | - Marcel H. A. M. Fens
- Department of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Raymond M. Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Marije Bartels
- Paediatric Haematology Department, Wilhelmina Children’s Hospital, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Richard Van Wijk
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
159
|
Abstract
The maintenance of a healthy and functional mitochondrial network is critical during development as well as throughout life in the response to physiological adaptations and stress conditions. Owing to their role in energy production, mitochondria are exposed to high levels of reactive oxygen species, making them particularly vulnerable to mitochondrial DNA mutations and protein misfolding. Given that mitochondria are formed from proteins encoded by both nuclear and mitochondrial genomes, an additional layer of complexity is inherent in the coordination of protein synthesis and the mitochondrial import of nuclear-encoded proteins. For these reasons, mitochondria have evolved multiple systems of quality control to ensure that the requisite number of functional mitochondria are present to meet the demands of the cell. These pathways work to eliminate damaged mitochondrial proteins or parts of the mitochondrial network by mitophagy and renew components by adding protein and lipids through biogenesis, collectively resulting in mitochondrial turnover. Mitochondrial quality control mechanisms are multi-tiered, operating at the protein, organelle and cell levels. Herein, we discuss mitophagy in different physiological contexts and then relate it to other quality control pathways, including the unfolded protein response, shedding of vesicles, proteolysis, and degradation by the ubiquitin-proteasome system. Understanding how these pathways contribute to the maintenance of mitochondrial homeostasis could provide insights into the development of targeted treatments when these systems fail in disease.
Collapse
|
160
|
A systems biology pipeline identifies regulatory networks for stem cell engineering. Nat Biotechnol 2019; 37:810-818. [PMID: 31267104 DOI: 10.1038/s41587-019-0159-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 05/16/2019] [Indexed: 12/18/2022]
Abstract
A major challenge for stem cell engineering is achieving a holistic understanding of the molecular networks and biological processes governing cell differentiation. To address this challenge, we describe a computational approach that combines gene expression analysis, previous knowledge from proteomic pathway informatics and cell signaling models to delineate key transitional states of differentiating cells at high resolution. Our network models connect sparse gene signatures with corresponding, yet disparate, biological processes to uncover molecular mechanisms governing cell fate transitions. This approach builds on our earlier CellNet and recent trajectory-defining algorithms, as illustrated by our analysis of hematopoietic specification along the erythroid lineage, which reveals a role for the EGF receptor family member, ErbB4, as an important mediator of blood development. We experimentally validate this prediction and perturb the pathway to improve erythroid maturation from human pluripotent stem cells. These results exploit an integrative systems perspective to identify new regulatory processes and nodes useful in cell engineering.
Collapse
|
161
|
Zhang J, Zhao H, Wu K, Peng Y, Han X, Zhang H, Liang L, Chen H, Hu J, Qu X, Zhang S, Chen L, Liu J. Knockdown of spliceosome U2AF1 significantly inhibits the development of human erythroid cells. J Cell Mol Med 2019; 23:5076-5086. [PMID: 31144421 PMCID: PMC6652819 DOI: 10.1111/jcmm.14370] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 03/14/2019] [Accepted: 04/21/2019] [Indexed: 11/30/2022] Open
Abstract
U2AF1 (U2AF35) is the small subunit of the U2 auxiliary factor (U2AF) that constitutes the U2 snRNP (small nuclear ribonucleoproteins) of the spliceosome. Here, we examined the function of U2AF1 in human erythropoiesis. First, we examined the expression of U2AF1 during in vitro human erythropoiesis and showed that U2AF1 was highly expressed in the erythroid progenitor burst-forming-unit erythroid (BFU-E) cell stage. A colony assay revealed that U2AF1 knockdown cells failed to form BFU-E and colony-forming-unit erythroid (CFU-E) colonies. Our results further showed that knockdown of U2AF1 significantly inhibited cell growth and induced apoptosis in erythropoiesis. Additionally, knockdown of U2AF1 also delayed terminal erythroid differentiation. To explore the molecular basis of the impaired function of erythroid development, RNA-seq was performed and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis results showed that several biological pathways, including the p53 signalling pathway, MAPK signalling pathway and haematopoietic cell lineage, were involved, with the p53 signalling pathway showing the greatest involvement. Western blot analysis revealed an increase in the protein levels of downstream targets of p53 following U2AF1 knockdown. The data further showed that depletion of U2AF1 altered alternatively spliced apoptosis-associated gene transcripts in CFU-E cells. Our findings elucidate the role of U2AF1 in human erythropoiesis and reveal the underlying mechanisms.
Collapse
Affiliation(s)
- Jieying Zhang
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Huizhi Zhao
- School of Life Science, Zhengzhou University, Zhengzhou, China
| | - Kunlu Wu
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Yuanliang Peng
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Xu Han
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Huan Zhang
- School of Life Science, Zhengzhou University, Zhengzhou, China
| | - Long Liang
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Huiyong Chen
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Jingping Hu
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Xiaoli Qu
- School of Life Science, Zhengzhou University, Zhengzhou, China
| | - Shijie Zhang
- School of Life Science, Zhengzhou University, Zhengzhou, China
| | - Lixiang Chen
- School of Life Science, Zhengzhou University, Zhengzhou, China
| | - Jing Liu
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
162
|
Houwing ME, de Pagter PJ, van Beers EJ, Biemond BJ, Rettenbacher E, Rijneveld AW, Schols EM, Philipsen JNJ, Tamminga RYJ, van Draat KF, Nur E, Cnossen MH. Sickle cell disease: Clinical presentation and management of a global health challenge. Blood Rev 2019; 37:100580. [PMID: 31128863 DOI: 10.1016/j.blre.2019.05.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 05/17/2019] [Accepted: 05/17/2019] [Indexed: 01/12/2023]
Abstract
Sickle cell disease is an autosomal recessive, multisystem disorder, characterised by chronic haemolytic anaemia, painful episodes of vaso-occlusion, progressive organ failure and a reduced life expectancy. Sickle cell disease is the most common monogenetic disease, with millions affected worldwide. In well-resourced countries, comprehensive care programs have increased life expectancy of sickle cell disease patients, with almost all infants surviving into adulthood. Therapeutic options for sickle cell disease patients are however, still scarce. Predictors of sickle cell disease severity and a better understanding of pathophysiology and (epi)genetic modifiers are warranted and could lead to more precise management and treatment. This review provides an extensive summary of the pathophysiology and management of sickle cell disease and encompasses the characteristics, complications and current and future treatment options of the disease.
Collapse
Affiliation(s)
- M E Houwing
- Department of Paediatric Haematology, Erasmus University Medical Center - Sophia Children's Hospital, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands.
| | - P J de Pagter
- Department of Paediatric Haematology, Erasmus University Medical Center - Sophia Children's Hospital, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands.
| | - E J van Beers
- Department of Internal Medicine and Dermatology, Van Creveldkliniek, University Medical Center Utrecht, Internal mail no C.01.412, 3508, GA, Utrecht, the Netherlands.
| | - B J Biemond
- Department of Internal Medicine and Clinical Haematology, Amsterdam University Medical Centers, Meibergdreef 9, 1105, AZ, Amsterdam, the Netherlands.
| | - E Rettenbacher
- Department of Paediatric Haematology, Radboud University Medical Center - Amalia Children's Hospital, Geert Grooteplein Zuid 10, 6500, HB, Nijmegen, the Netherlands.
| | - A W Rijneveld
- Department of Haematology, Erasmus University Medical Center, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands.
| | - E M Schols
- Department of Haematology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, the Netherlands.
| | - J N J Philipsen
- Department of Cell Biology, Erasmus University Medical Center, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands.
| | - R Y J Tamminga
- Department of Paediatric Oncology and Haematology, University Medical Center Groningen - Beatrix Children's Hospital, Postbus 30001, 9700, RB, Groningen, the Netherlands..
| | - K Fijn van Draat
- Department of Paediatric Haematology, Amsterdam University Medical Centers - Emma Children's Hospital, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Department of Plasma Proteins, Sanquin Research, the Netherlands.
| | - E Nur
- Department of Internal Medicine and Clinical Haematology, Amsterdam University Medical Centers, Meibergdreef 9, 1105, AZ, Amsterdam, the Netherlands.
| | - M H Cnossen
- Department of Paediatric Haematology, Erasmus University Medical Center - Sophia Children's Hospital, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands.
| |
Collapse
|
163
|
Kesharwani R, Sarmah D, Kaur H, Mounika L, Verma G, Pabbala V, Kotian V, Kalia K, Borah A, Dave KR, Yavagal DR, Bhattacharya P. Interplay between Mitophagy and Inflammasomes in Neurological Disorders. ACS Chem Neurosci 2019; 10:2195-2208. [PMID: 30917655 DOI: 10.1021/acschemneuro.9b00117] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Mitophagy and inflammasomes have a pivotal role in the development of neuropathology. Molecular mechanisms behind mitophagy and inflammasomes are well-understood, but lacunae prevail in understanding the crosstalk between them in various neurological disorders. As mitochondrial dysfunction is the prime event in neurodegeneration, the clearance of impaired mitochondria is one of the main tasks for maintaining cell integrity in the majority of neuropathologies. Along with it, inflammasome activation also plays a major role, which is usually followed by mitochondrial dysfunction. The present review highlights basics of autophagy, mitophagy, and inflammasomes and the molecular mechanisms involved, and more importantly, it tries to elaborate the interplay between mitophagy and inflammasomes in various neurological disorders. This will help in upgrading the reader's understanding in exploring the link between mitophagy and inflammasomes, which has dealt with limitations in past studies.
Collapse
Affiliation(s)
- Radhika Kesharwani
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382 355, Gujarat, India
| | - Deepaneeta Sarmah
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382 355, Gujarat, India
| | - Harpreet Kaur
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382 355, Gujarat, India
| | - Leela Mounika
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382 355, Gujarat, India
| | - Geetesh Verma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382 355, Gujarat, India
| | - Veeresh Pabbala
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382 355, Gujarat, India
| | - Vignesh Kotian
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382 355, Gujarat, India
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382 355, Gujarat, India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar-788 011, Assam, India
| | - Kunjan R. Dave
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Dileep R. Yavagal
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382 355, Gujarat, India
| |
Collapse
|
164
|
Hernandez JA, Castro VL, Reyes-Nava N, Montes LP, Quintana AM. Mutations in the zebrafish hmgcs1 gene reveal a novel function for isoprenoids during red blood cell development. Blood Adv 2019; 3:1244-1254. [PMID: 30987969 PMCID: PMC6482358 DOI: 10.1182/bloodadvances.2018024539] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 03/09/2019] [Indexed: 12/22/2022] Open
Abstract
Erythropoiesis is the process by which new red blood cells (RBCs) are formed and defects in this process can lead to anemia or thalassemia. The GATA1 transcription factor is an established mediator of RBC development. However, the upstream mechanisms that regulate the expression of GATA1 are not completely characterized. Cholesterol is 1 potential upstream mediator of GATA1 expression because previously published studies suggest that defects in cholesterol synthesis disrupt RBC differentiation. Here we characterize RBC development in a zebrafish harboring a single missense mutation in the hmgcs1 gene (Vu57 allele). hmgcs1 encodes the first enzyme in the cholesterol synthesis pathway and mutation of hmgcs1 inhibits cholesterol synthesis. We analyzed the number of RBCs in hmgcs1 mutants and their wild-type siblings. Mutation of hmgcs1 resulted in a decrease in the number of mature RBCs, which coincides with reduced gata1a expression. We combined these experiments with pharmacological inhibition and confirmed that cholesterol and isoprenoid synthesis are essential for RBC differentiation, but that gata1a expression is isoprenoid dependent. Collectively, our results reveal 2 novel upstream regulators of RBC development and suggest that appropriate cholesterol homeostasis is critical for primitive erythropoiesis.
Collapse
Affiliation(s)
- Jose A Hernandez
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX
| | - Victoria L Castro
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX
| | - Nayeli Reyes-Nava
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX
| | - Laura P Montes
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX
| | - Anita M Quintana
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX
| |
Collapse
|
165
|
Kim M, Civin CI, Kingsbury TJ. MicroRNAs as regulators and effectors of hematopoietic transcription factors. WILEY INTERDISCIPLINARY REVIEWS-RNA 2019; 10:e1537. [PMID: 31007002 DOI: 10.1002/wrna.1537] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 03/24/2019] [Accepted: 04/03/2019] [Indexed: 12/17/2022]
Abstract
Hematopoiesis is a highly-regulated development process orchestrated by lineage-specific transcription factors that direct the generation of all mature blood cells types, including red blood cells, megakaryocytes, granulocytes, monocytes, and lymphocytes. Under homeostatic conditions, the hematopoietic system of the typical adult generates over 1011 blood cells daily throughout life. In addition, hematopoiesis must be responsive to acute challenges due to blood loss or infection. MicroRNAs (miRs) cooperate with transcription factors to regulate all aspects of hematopoiesis, including stem cell maintenance, lineage selection, cell expansion, and terminal differentiation. Distinct miR expression patterns are associated with specific hematopoietic lineages and stages of differentiation and functional analyses have elucidated essential roles for miRs in regulating cell transitions, lineage selection, maturation, and function. MiRs function as downstream effectors of hematopoietic transcription factors and as upstream regulators to control transcription factor levels. Multiple miRs have been shown to play essential roles. Regulatory networks comprised of differentially expressed lineage-specific miRs and hematopoietic transcription factors are involved in controlling the quiescence and self-renewal of hematopoietic stem cells as well as proliferation and differentiation of lineage-specific progenitor cells during erythropoiesis, myelopoiesis, and lymphopoiesis. This review focuses on hematopoietic miRs that function as upstream regulators of central hematopoietic transcription factors required for normal hematopoiesis. This article is categorized under: RNA in Disease and Development > RNA in Development Regulatory RNAs/RNAi/Riboswitches > Regulatory RNAs.
Collapse
Affiliation(s)
- MinJung Kim
- Department of Pediatrics, Center for Stem Cell Biology and Regenerative Medicine, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Curt I Civin
- Department of Pediatrics and Physiology, Center for Stem Cell Biology and Regenerative Medicine, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Tami J Kingsbury
- Department of Physiology, Center for Stem Cell Biology and Regenerative Medicine, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
166
|
Jafari H, Hesami S, Safi M, Ghasemi F, Banan M. Expression and hydroxyurea-triggered induction of EGFP upon CRISPR/Cas9-mediated integration into the γ-globin gene of K562 cells. Biotechnol Lett 2019; 41:691-700. [DOI: 10.1007/s10529-019-02671-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/27/2019] [Indexed: 02/08/2023]
|
167
|
Shokri G, Kouhkan F, Nojehdehi S, Soleimani M, Pourfathollah AA, Nikougoftar Zarif M, Tamaddon M, Obeidi N. Simultaneous regulation of miR-451 and miR-191 led to erythroid fate decision of mouse embryonic stem cell. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:432-438. [PMID: 31168349 PMCID: PMC6535200 DOI: 10.22038/ijbms.2019.27919.6795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 10/27/2018] [Indexed: 01/13/2023]
Abstract
OBJECTIVES Various microRNAs (miRNAs) are expressed during development of mammalian cells, when they aid in modulating gene expression by mediating mRNA transcript cleavage and/or regulation of translation rate. miR-191 and miR-451 have been shown to be critical regulators of hematopoiesis and have important roles in the induction of erythroid fate decision. So, the aim of this study is investigation of the miR-191 and miR-451 roles in the controlling mouse embryonic stem cell (mESC) differentiation toward the erythroid lineage. MATERIALS AND METHODS mESCs were infected with either pCDH-miR-Off-191 viruses in pCDH-miR-Off-191 group or simultaneously with pCDH-miR-Off-191 and pCDH-miR-451 lentiviruses in simultaneous group. Then, the expression profiles of erythroid specific transcription factors and globin genes were analyzed using QRT-PCR on day 14 and 21 of differentiation. Flow cytometry analysis was used to evaluate of TER119 and CD235a erythroid specific surface markers. RESULTS Gata-1, Klf-1, Epor and globin chains were found to be expressed in pCDH-miR-Off-191 and in simultaneous groups. The majority of globin chains showed changes in their expression levels with progression of differentiation from day 14 to day 21. Flow cytometry results showed that miR-451 up- regulation and miR-191 down-regulation is associated with the expression of TER119 and CD235a. Of these two groups analyzed, simultaneous group was most significantly potent in stimulation of erythroid fate decision of mESCs. CONCLUSION Together, present data demonstrate that down-regulation of miR-191 alone can enhance the differentiation of mESCs. However, the simultaneous effect of miR-451up-regulation and miR-191 down-regulation is much stronger and can have more practical use in artificial blood production.
Collapse
Affiliation(s)
| | | | | | - Masoud Soleimani
- Department of Hematology, School of Medicine, Tarbiat Modares University, Tehran, Iran
| | | | - Mahin Nikougoftar Zarif
- Blood Transfusion Research Center, High Institute for Education and Research in Transfusion Medicine, Tehran, Iran
| | | | - Narges Obeidi
- Department of Hematology, School of Para Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
168
|
Yang C, Hashimoto M, Lin QXX, Tan DQ, Suda T. Sphingosine-1-phosphate signaling modulates terminal erythroid differentiation through the regulation of mitophagy. Exp Hematol 2019; 72:47-59.e1. [DOI: 10.1016/j.exphem.2019.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/10/2019] [Accepted: 01/11/2019] [Indexed: 01/17/2023]
|
169
|
Liang L, Peng Y, Zhang J, Zhang Y, Roy M, Han X, Xiao X, Sun S, Liu H, Nie L, Kuang Y, Zhu Z, Deng J, Xia Y, Sankaran VG, Hillyer CD, Mohandas N, Ye M, An X, Liu J. Deubiquitylase USP7 regulates human terminal erythroid differentiation by stabilizing GATA1. Haematologica 2019; 104:2178-2187. [PMID: 30872372 PMCID: PMC6821630 DOI: 10.3324/haematol.2018.206227] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 03/13/2019] [Indexed: 01/01/2023] Open
Abstract
Ubiquitination is an enzymatic post-translational modification that affects protein fate. The ubiquitin-proteasome system (UPS) was first discovered in reticulocytes where it plays important roles in reticulocyte maturation. Recent studies have revealed that ubiquitination is a dynamic and reversible process and that deubiquitylases are capable of removing ubiquitin from their protein substrates. Given the fact that the UPS is highly active in reticulocytes, it is speculated that deubiquitylases may play important roles in erythropoiesis. Yet, the role of deubiquitylases in erythropoiesis remains largely unexplored. In the present study, we found that the expression of deubiquitylase USP7 is significantly increased during human terminal erythroid differentiation. We further showed that interfering with USP7 function, either by short hairpin RNA-mediated knockdown or USP7-specific inhibitors, impaired human terminal erythroid differentiation due to decreased GATA1 level and that restoration of GATA1 levels rescued the differentiation defect. Mechanistically, USP7 deficiency led to a decreased GATA1 protein level that could be reversed by proteasome inhibitors. Furthermore, USP7 interacts directly with GATA1 and catalyzes the removal of K48-linked poly ubiquitylation chains conjugated onto GATA1, thereby stabilizing GATA1 protein. Collectively, our findings have identified an important role of a deubiquitylase in human terminal erythroid differentiation by stabilizing GATA1, the master regulator of erythropoiesis.
Collapse
Affiliation(s)
- Long Liang
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Yuanliang Peng
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Jieying Zhang
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Laboratory of Membrane Biology, New York Blood Center, New York, NY, USA
| | - Yibin Zhang
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Mridul Roy
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Xu Han
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Xiaojuan Xiao
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Shuming Sun
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Hong Liu
- Xiangya Hospital, Central South University, Changsha, China
| | - Ling Nie
- Xiangya Hospital, Central South University, Changsha, China
| | - Yijin Kuang
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Zesen Zhu
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Jinghui Deng
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Vijay G Sankaran
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Narla Mohandas
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY, USA
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY, USA .,School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Jing Liu
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China .,Erythropoiesis Research Center, Central South University, Changsha, China
| |
Collapse
|
170
|
Activation of the vitamin D receptor transcription factor stimulates the growth of definitive erythroid progenitors. Blood Adv 2019; 2:1207-1219. [PMID: 29844206 DOI: 10.1182/bloodadvances.2018017533] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/15/2018] [Indexed: 12/30/2022] Open
Abstract
The pathways that regulate the growth of erythroid progenitors are incompletely understood. In a computational analysis of gene expression changes during erythroid ontogeny, the vitamin D receptor (Vdr) nuclear hormone receptor transcription factor gene was identified in fetal and adult stages, but not at the embryonic stage of development. Vdr was expressed in definitive erythroid (EryD) progenitors and was downregulated during their maturation. Activation of Vdr signaling by the vitamin D3 agonist calcitriol increased the outgrowth of EryD colonies from fetal liver and adult bone marrow, maintained progenitor potential, and delayed erythroid maturation, as revealed by clonogenic assays, suspension culture, cell surface phenotype, and gene expression analyses. The early (cKit+CD71lo/neg), but not the late (cKit+CD71hi), EryD progenitor subset of LinnegcKit+ cells was responsive to calcitriol. Culture of cKit+CD71lo/neg progenitors in the presence of both vitamin D3 and glucocorticoid receptor ligands resulted in an increase in proliferation that was at least additive compared with either ligand alone. Lentivirus shRNA-mediated knockdown of Vdr expression abrogated the stimulation of early erythroid progenitor growth by calcitriol. These findings suggest that Vdr has a cell-intrinsic function in early erythroid progenitors. Targeting of downstream components of the Vdr signaling pathway may lead to new approaches for the expansion of erythroid progenitors ex vivo.
Collapse
|
171
|
Barbarani G, Fugazza C, Strouboulis J, Ronchi AE. The Pleiotropic Effects of GATA1 and KLF1 in Physiological Erythropoiesis and in Dyserythropoietic Disorders. Front Physiol 2019; 10:91. [PMID: 30809156 PMCID: PMC6379452 DOI: 10.3389/fphys.2019.00091] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/25/2019] [Indexed: 01/19/2023] Open
Abstract
In the last few years, the advent of new technological approaches has led to a better knowledge of the ontogeny of erythropoiesis during development and of the journey leading from hematopoietic stem cells (HSCs) to mature red blood cells (RBCs). Our view of a well-defined hierarchical model of hematopoiesis with a near-homogeneous HSC population residing at the apex has been progressively challenged in favor of a landscape where HSCs themselves are highly heterogeneous and lineages separate earlier than previously thought. The coordination of these events is orchestrated by transcription factors (TFs) that work in a combinatorial manner to activate and/or repress their target genes. The development of next generation sequencing (NGS) has facilitated the identification of pathological mutations involving TFs underlying hematological defects. The examples of GATA1 and KLF1 presented in this review suggest that in the next few years the number of TF mutations associated with dyserythropoietic disorders will further increase.
Collapse
Affiliation(s)
- Gloria Barbarani
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi Milano-Bicocca, Milan, Italy
| | - Cristina Fugazza
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi Milano-Bicocca, Milan, Italy
| | - John Strouboulis
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Antonella E Ronchi
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi Milano-Bicocca, Milan, Italy
| |
Collapse
|
172
|
Grosso RA, Caldarone PVS, Sánchez MC, Chiabrando GA, Colombo MI, Fader CM. Hemin induces autophagy in a leukemic erythroblast cell line through the LRP1 receptor. Biosci Rep 2019; 39:BSR20181156. [PMID: 30523204 PMCID: PMC6328880 DOI: 10.1042/bsr20181156] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/31/2018] [Accepted: 11/07/2018] [Indexed: 01/04/2023] Open
Abstract
Hemin is an erythropoietic inductor capable of inducing autophagy in erythroid-like cell lines. Low-density lipoprotein receptor-related protein 1 (LRP1) is a transmembrane receptor involved in a wide range of cellular processes, such as proliferation, differentiation, and metabolism. Our aim was to evaluate whether LRP1 is responsible for hemin activity in K562 cells, with the results demonstrating a three-fold increase in LRP1 gene expression levels (P-values <0.001) when assessed by quantitative real-time RT-PCR (qRT-PCR). Moreover, a 70% higher protein amount was observed compared with control condition (P-values <0.01) by Western blot (WB). Time kinetic assays demonstrated a peak in light chain 3 (LC3) II (LC3II) levels after 8 h of hemin stimulation and the localization of LRP1 in the autophagosome structures. Silencing LRP1 by siRNA decreased drastically the hemin-induced autophagy activity by almost 80% compared with control cells (P-values <0.01). Confocal localization and biochemical analysis indicated a significant redistribution of LRP1 from early endosomes and recycling compartments to late endosomes and autophagolysosomes, where the receptor is degraded. We conclude that LRP1 is responsible for hemin-induced autophagy activity in the erythroblastic cell line and that hemin-LRP1 complex activation promotes a self-regulation of the receptor. Our results suggest that hemin, via the LRP1 receptor, favors erythroid maturation by inducing an autophagic response, making it a possible therapeutic candidate to help in the treatment of hematological disorders.
Collapse
Affiliation(s)
- Ruben Adrian Grosso
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Paula Virginia Subirada Caldarone
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5016, Argentina
| | - María Cecilia Sánchez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5016, Argentina
| | - Gustavo Alberto Chiabrando
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5016, Argentina
| | - María Isabel Colombo
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Claudio Marcelo Fader
- Universidad Nacional de Cuyo, Facultad de Odontología, Mendoza, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
173
|
Maea expressed by macrophages, but not erythroblasts, maintains postnatal murine bone marrow erythroblastic islands. Blood 2019; 133:1222-1232. [PMID: 30674470 DOI: 10.1182/blood-2018-11-888180] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/18/2019] [Indexed: 12/11/2022] Open
Abstract
The erythroblastic island (EI), formed by a central macrophage and developing erythroblasts (EBs), was first described decades ago and was recently shown to play an in vivo role in homeostatic and pathological erythropoiesis. The exact molecular mechanisms, however, mediating the interactions between macrophages and EBs remain unclear. Macrophage-EB attacher (Maea) has previously been suggested to mediate homophilic adhesion bounds bridging macrophages and EBs. Maea-deficient mice die perinatally with anemia and defective erythrocyte enucleation, suggesting a critical role in fetal erythropoiesis. Here, we generated conditional knockout mouse models of Maea to assess its cellular and postnatal contributions. Deletion of Maea in macrophages using Csf1r-Cre or CD169-Cre caused severe reductions of bone marrow (BM) macrophages, EBs, and in vivo island formation, whereas its deletion in the erythroid lineage using Epor-Cre had no such phenotype, suggesting a dominant role of Maea in the macrophage for BM erythropoiesis. Interestingly, Maea deletion in spleen macrophages did not alter their numbers or functions. Postnatal Maea deletion using Mx1-Cre or function inhibition using a novel monoclonal antibody also impaired BM erythropoiesis. These results indicate that Maea contributes to adult BM erythropoiesis by regulating the maintenance of macrophages and their interaction with EBs via an as-yet-unidentified EB receptor.
Collapse
|
174
|
Säwen P, Eldeeb M, Erlandsson E, Kristiansen TA, Laterza C, Kokaia Z, Karlsson G, Yuan J, Soneji S, Mandal PK, Rossi DJ, Bryder D. Murine HSCs contribute actively to native hematopoiesis but with reduced differentiation capacity upon aging. eLife 2018; 7:41258. [PMID: 30561324 PMCID: PMC6298771 DOI: 10.7554/elife.41258] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/28/2018] [Indexed: 12/12/2022] Open
Abstract
A hallmark of adult hematopoiesis is the continuous replacement of blood cells with limited lifespans. While active hematopoietic stem cell (HSC) contribution to multilineage hematopoiesis is the foundation of clinical HSC transplantation, recent reports have questioned the physiological contribution of HSCs to normal/steady-state adult hematopoiesis. Here, we use inducible lineage tracing from genetically marked adult HSCs and reveal robust HSC-derived multilineage hematopoiesis. This commences via defined progenitor cells, but varies substantially in between different hematopoietic lineages. By contrast, adult HSC contribution to hematopoietic cells with proposed fetal origins is neglible. Finally, we establish that the HSC contribution to multilineage hematopoiesis declines with increasing age. Therefore, while HSCs are active contributors to native adult hematopoiesis, it appears that the numerical increase of HSCs is a physiologically relevant compensatory mechanism to account for their reduced differentiation capacity with age. As far as we know, all adult blood cells derive from blood stem cells that are located in the bone marrow. These stem cells can produce red blood cells, white blood cells and platelets – the cells fragments that form blood clots to stop bleeding. They can also regenerate, producing more stem cells to support future blood cell production. But, our understanding of the system may be incomplete. The easiest way to study blood cell production is to watch what happens after a bone marrow transplant. Before a transplant, powerful chemotherapy kills the existing stem cells. This forces the transplanted stem cells to restore the whole system from scratch, allowing scientists to study blood cell production in fine detail. But completely replacing the bone marrow puts major stress on the body, and this may alter the way that the stem cells behave. To understand how adult stem cells keep the blood ticking over on a day-to-day basis, experiments also need to look at healthy animals. Säwén et al. now describe a method to follow bone marrow stem cells as they produce blood cells in adult mice. The technique, known as lineage tracing, leaves an indelible mark, a red glow, on the stem cells. The cells pass this mark on every time they divide, leaving a lasting trace in every blood cell that they produce. Tracking the red-glowing cells over time reveals which types of blood cells the stem cells make as well as provides estimates on the timing and extent of these processes. It has previously been suggested that a few types of specialist blood cells, like brain-specific immune cells, originate from cells other than adult blood stem cells. As expected, the adult stem cells did not produce such cells. But, just as seen in transplant experiments, the stem cells were able to produce all the other major blood cell types. They made platelets at the fastest rate, followed by certain types of white blood cells and red blood cells. As the mice got older, the stem cells started to slow down, producing fewer blood cells each. To compensate, the number of stem cells increased, helping to keep blood cell numbers up. This alternative approach to studying blood stem cells shows how the system behaves in a more natural environment. Away from the stresses of transplant, the technique revealed that blood stem cells are not immune to aging. In the future, understanding more about the system in its natural state could lead to ways to boost blood stem cells as we get older.
Collapse
Affiliation(s)
- Petter Säwen
- Division of Molecular Hematology, Department of Laboratory Medicine, Medical Faculty, Lund University, Lund, Sweden
| | - Mohamed Eldeeb
- Division of Molecular Hematology, Department of Laboratory Medicine, Medical Faculty, Lund University, Lund, Sweden
| | - Eva Erlandsson
- Division of Molecular Hematology, Department of Laboratory Medicine, Medical Faculty, Lund University, Lund, Sweden
| | - Trine A Kristiansen
- Division of Molecular Hematology, Department of Laboratory Medicine, Medical Faculty, Lund University, Lund, Sweden
| | - Cecilia Laterza
- StemTherapy, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Zaal Kokaia
- StemTherapy, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Göran Karlsson
- Division of Molecular Hematology, Department of Laboratory Medicine, Medical Faculty, Lund University, Lund, Sweden.,StemTherapy, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Joan Yuan
- Division of Molecular Hematology, Department of Laboratory Medicine, Medical Faculty, Lund University, Lund, Sweden.,StemTherapy, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Shamit Soneji
- Division of Molecular Hematology, Department of Laboratory Medicine, Medical Faculty, Lund University, Lund, Sweden.,StemTherapy, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Pankaj K Mandal
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States.,Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, Massachusetts, United States
| | - Derrick J Rossi
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States.,Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, Massachusetts, United States
| | - David Bryder
- Division of Molecular Hematology, Department of Laboratory Medicine, Medical Faculty, Lund University, Lund, Sweden.,StemTherapy, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden.,Sahlgrenska Cancer Center, Gothenburg University, Gothenburg, Sweden
| |
Collapse
|
175
|
Guo L, Wang Q, Weng L, Hauser LA, Strawser CJ, Mesaros C, Lynch DR, Blair IA. Characterization of a new N-terminally acetylated extra-mitochondrial isoform of frataxin in human erythrocytes. Sci Rep 2018; 8:17043. [PMID: 30451920 PMCID: PMC6242848 DOI: 10.1038/s41598-018-35346-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 11/02/2018] [Indexed: 01/18/2023] Open
Abstract
Frataxin is a highly conserved protein encoded by the frataxin (FXN) gene. The full-length 210-amino acid form of protein frataxin (1-210; isoform A) expressed in the cytosol of cells rapidly translocates to the mitochondria, where it is converted to the mature form (81-210) by mitochondrial processing peptidase. Mature frataxin (81-210) is a critically important protein because it facilitates the assembly of mitochondrial iron-sulfur cluster protein complexes such as aconitase, lipoate synthase, and succinate dehydrogenases. Decreased expression of frataxin protein is responsible for the devastating rare genetic disease of Friedreich's ataxia. The mitochondrial form of frataxin has long been thought to be present in erythrocytes even though paradoxically, erythrocytes lack mitochondria. We have discovered that erythrocyte frataxin is in fact a novel isoform of frataxin (isoform E) with 135-amino acids and an N-terminally acetylated methionine residue. There is three times as much isoform E in erythrocytes (20.9 ± 6.4 ng/mL) from the whole blood of healthy volunteers (n = 10) when compared with the mature mitochondrial frataxin present in other blood cells (7.1 ± 1.0 ng/mL). Isoform E lacks a mitochondrial targeting sequence and so is distributed to both cytosol and the nucleus when expressed in cultured cells. When extra-mitochondrial frataxin isoform E is expressed in HEK 293 cells, it is converted to a shorter isoform identical to the mature frataxin found in mitochondria, which raises the possibility that it is involved in disease etiology. The ability to specifically quantify extra-mitochondrial and mitochondrial isoforms of frataxin in whole blood will make it possible to readily follow the natural history of diseases such as Friedreich's ataxia and monitor the efficacy of therapeutic interventions.
Collapse
Affiliation(s)
- Lili Guo
- Penn SRP Center and Center of Excellence in Environmental Toxicology Center, Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
| | - Qingqing Wang
- Penn SRP Center and Center of Excellence in Environmental Toxicology Center, Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
| | - Liwei Weng
- Penn SRP Center and Center of Excellence in Environmental Toxicology Center, Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Lauren A Hauser
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Cassandra J Strawser
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Clementina Mesaros
- Penn SRP Center and Center of Excellence in Environmental Toxicology Center, Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
| | - David R Lynch
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Ian A Blair
- Penn SRP Center and Center of Excellence in Environmental Toxicology Center, Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States.
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States.
| |
Collapse
|
176
|
In vitroAntimicrobial andin vivoLead Acetate Poison Abatement Study ofGarcinia kolaHeckel. JOURNAL OF TAIBAH UNIVERSITY FOR SCIENCE 2018. [DOI: 10.1016/j.jtusci.2017.06.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
177
|
Brown A, Baird MR, Yip MC, Murray J, Shao S. Structures of translationally inactive mammalian ribosomes. eLife 2018; 7:40486. [PMID: 30355441 PMCID: PMC6226290 DOI: 10.7554/elife.40486] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 10/12/2018] [Indexed: 12/23/2022] Open
Abstract
The cellular levels and activities of ribosomes directly regulate gene expression during numerous physiological processes. The mechanisms that globally repress translation are incompletely understood. Here, we use electron cryomicroscopy to analyze inactive ribosomes isolated from mammalian reticulocytes, the penultimate stage of red blood cell differentiation. We identify two types of ribosomes that are translationally repressed by protein interactions. The first comprises ribosomes sequestered with elongation factor 2 (eEF2) by SERPINE mRNA binding protein 1 (SERBP1) occupying the ribosomal mRNA entrance channel. The second type are translationally repressed by a novel ribosome-binding protein, interferon-related developmental regulator 2 (IFRD2), which spans the P and E sites and inserts a C-terminal helix into the mRNA exit channel to preclude translation. IFRD2 binds ribosomes with a tRNA occupying a noncanonical binding site, the ‘Z site’, on the ribosome. These structures provide functional insights into how ribosomal interactions may suppress translation to regulate gene expression.
Collapse
Affiliation(s)
- Alan Brown
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States
| | - Matthew R Baird
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States
| | - Matthew Cj Yip
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Jason Murray
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Sichen Shao
- Department of Cell Biology, Harvard Medical School, Boston, United States
| |
Collapse
|
178
|
Andrade DC, Haine L, Toledo C, Diaz HS, Quintanilla RA, Marcus NJ, Iturriaga R, Richalet JP, Voituron N, Del Rio R. Ventilatory and Autonomic Regulation in Sleep Apnea Syndrome: A Potential Protective Role for Erythropoietin? Front Physiol 2018; 9:1440. [PMID: 30374309 PMCID: PMC6196773 DOI: 10.3389/fphys.2018.01440] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/21/2018] [Indexed: 12/20/2022] Open
Abstract
Obstructive sleep apnea (OSA) is the most common form of sleep disordered breathing and is associated with wide array of cardiovascular morbidities. It has been proposed that during OSA, the respiratory control center (RCC) is affected by exaggerated afferent signals coming from peripheral/central chemoreceptors which leads to ventilatory instability and may perpetuate apnea generation. Treatments focused on decreasing hyperactivity of peripheral/central chemoreceptors may be useful to improving ventilatory instability in OSA patients. Previous studies indicate that oxidative stress and inflammation are key players in the increased peripheral/central chemoreflex drive associated with OSA. Recent data suggest that erythropoietin (Epo) could also be involved in modulating chemoreflex activity as functional Epo receptors are constitutively expressed in peripheral and central chemoreceptors cells. Additionally, there is some evidence that Epo has anti-oxidant/anti-inflammatory effects. Accordingly, we propose that Epo treatment during OSA may reduce enhanced peripheral/central chemoreflex drive and normalize the activity of the RCC which in turn may help to abrogate ventilatory instability. In this perspective article we discuss the potential beneficial effects of Epo administration on ventilatory regulation in the setting of OSA.
Collapse
Affiliation(s)
- David C Andrade
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Investigación en Fisiología del Ejercicio, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Liasmine Haine
- Laboratoire Hypoxie and Poumon - EA2363, Université Paris 13, Paris, France
| | - Camilo Toledo
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Hugo S Diaz
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Investigación Biomédica, Universidad Autónoma de Chile, Santiago, Chile
| | | | - Noah J Marcus
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA, United States
| | - Rodrigo Iturriaga
- Laboratorio de Neurobiología, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jean-Paul Richalet
- Laboratoire Hypoxie and Poumon - EA2363, Université Paris 13, Paris, France
| | - Nicolas Voituron
- Laboratoire Hypoxie and Poumon - EA2363, Université Paris 13, Paris, France
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
179
|
Kocaturk NM, Gozuacik D. Crosstalk Between Mammalian Autophagy and the Ubiquitin-Proteasome System. Front Cell Dev Biol 2018; 6:128. [PMID: 30333975 PMCID: PMC6175981 DOI: 10.3389/fcell.2018.00128] [Citation(s) in RCA: 277] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/13/2018] [Indexed: 12/16/2022] Open
Abstract
Autophagy and the ubiquitin-proteasome system (UPS) are the two major intracellular quality control and recycling mechanisms that are responsible for cellular homeostasis in eukaryotes. Ubiquitylation is utilized as a degradation signal by both systems, yet, different mechanisms are in play. The UPS is responsible for the degradation of short-lived proteins and soluble misfolded proteins whereas autophagy eliminates long-lived proteins, insoluble protein aggregates and even whole organelles (e.g., mitochondria, peroxisomes) and intracellular parasites (e.g., bacteria). Both the UPS and selective autophagy recognize their targets through their ubiquitin tags. In addition to an indirect connection between the two systems through ubiquitylated proteins, recent data indicate the presence of connections and reciprocal regulation mechanisms between these degradation pathways. In this review, we summarize these direct and indirect interactions and crosstalks between autophagy and the UPS, and their implications for cellular stress responses and homeostasis.
Collapse
Affiliation(s)
- Nur Mehpare Kocaturk
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Devrim Gozuacik
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabanci University, Istanbul, Turkey
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul, Turkey
| |
Collapse
|
180
|
Yang L, Li LC, Wang X, Wang WH, Wang YC, Xu CR. The contributions of mesoderm-derived cells in liver development. Semin Cell Dev Biol 2018; 92:63-76. [PMID: 30193996 DOI: 10.1016/j.semcdb.2018.09.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/31/2018] [Accepted: 09/02/2018] [Indexed: 02/07/2023]
Abstract
The liver is an indispensable organ for metabolism and drug detoxification. The liver consists of endoderm-derived hepatobiliary lineages and various mesoderm-derived cells, and interacts with the surrounding tissues and organs through the ventral mesentery. Liver development, from hepatic specification to liver maturation, requires close interactions with mesoderm-derived cells, such as mesothelial cells, hepatic stellate cells, mesenchymal cells, liver sinusoidal endothelial cells and hematopoietic cells. These cells affect liver development through precise signaling events and even direct physical contact. Through the use of new techniques, emerging studies have recently led to a deeper understanding of liver development and its related mechanisms, especially the roles of mesodermal cells in liver development. Based on these developments, the current protocols for in vitro hepatocyte-like cell induction and liver-like tissue construction have been optimized and are of great importance for the treatment of liver diseases. Here, we review the roles of mesoderm-derived cells in the processes of liver development, hepatocyte-like cell induction and liver-like tissue construction.
Collapse
Affiliation(s)
- Li Yang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Lin-Chen Li
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xin Wang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China
| | - Wei-Hua Wang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Yan-Chun Wang
- Haidian Maternal & Child Health Hospital, Beijing, 100080, China
| | - Cheng-Ran Xu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, China.
| |
Collapse
|
181
|
Tangprasittipap A, Kaewprommal P, Sripichai O, Sathirapongsasuti N, Satirapod C, Shaw PJ, Piriyapongsa J, Hongeng S. Comparison of gene expression profiles between human erythroid cells derived from fetal liver and adult peripheral blood. PeerJ 2018; 6:e5527. [PMID: 30186694 PMCID: PMC6120446 DOI: 10.7717/peerj.5527] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 08/07/2018] [Indexed: 12/23/2022] Open
Abstract
Background A key event in human development is the establishment of erythropoietic progenitors in the bone marrow, which is accompanied by a fetal-to-adult switch in hemoglobin expression. Understanding of this event could lead to medical application, notably treatment of sickle cell disease and β-thalassemia. The changes in gene expression of erythropoietic progenitor cells as they migrate from the fetal liver and colonize the bone marrow are still rather poorly understood, as primary fetal liver (FL) tissues are difficult to obtain. Methods We obtained human FL tissue and adult peripheral blood (AB) samples from Thai subjects. Primary CD34+ cells were cultured in vitro in a fetal bovine serum-based culture medium. After 8 days of culture, erythroid cell populations were isolated by flow cytometry. Gene expression in the FL- and AB-derived cells was studied by Affymetrix microarray and reverse-transcription quantitative PCR. The microarray data were combined with that from a previous study of human FL and AB erythroid development, and meta-analysis was performed on the combined dataset. Results FL erythroid cells showed enhanced proliferation and elevated fetal hemoglobin relative to AB cells. A total of 1,391 fetal up-regulated and 329 adult up-regulated genes were identified from microarray data generated in this study. Five hundred ninety-nine fetal up-regulated and 284 adult up-regulated genes with reproducible patterns between this and a previous study were identified by meta-analysis of the combined dataset, which constitute a core set of genes differentially expressed between FL and AB erythroid cells. In addition to these core genes, 826 and 48 novel genes were identified only from data generated in this study to be FL up- and AB up-regulated, respectively. The in vivo relevance for some of these novel genes was demonstrated by pathway analysis, which showed novel genes functioning in pathways known to be important in proliferation and erythropoiesis, including the mitogen-activated protein kinase (MAPK) and the phosphatidyl inositol 3 kinase (PI3K)-Akt pathways. Discussion The genes with upregulated expression in FL cells, which include many novel genes identified from data generated in this study, suggest that cellular proliferation pathways are more active in the fetal stage. Erythroid progenitor cells may thus undergo a reprogramming during ontogenesis in which proliferation is modulated by changes in expression of key regulators, primarily MYC, and others including insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3), neuropilin and tolloid-like 2 (NETO2), branched chain amino acid transaminase 1 (BCAT1), tenascin XB (TNXB) and proto-oncogene, AP-1 transcription factor subunit (JUND). This reprogramming may thus be necessary for acquisition of the adult identity and switching of hemoglobin expression.
Collapse
Affiliation(s)
| | - Pavita Kaewprommal
- Biostatistics and Bioinformatics Laboratory, Genome Technology Research Unit, National Center for Genetic Engineering and Biotechnology, Pathum Thani, Thailand
| | - Orapan Sripichai
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | | | | | - Philip J Shaw
- Protein-Ligand Engineering and Molecular Biology Laboratory, Medical Molecular Biology Research Unit, National Center for Genetic Engineering and Biotechnology, Pathum Thani, Thailand
| | - Jittima Piriyapongsa
- Biostatistics and Bioinformatics Laboratory, Genome Technology Research Unit, National Center for Genetic Engineering and Biotechnology, Pathum Thani, Thailand
| | - Suradej Hongeng
- Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
182
|
Alattar N, Daud H, Al-Majmaie R, Zeulla D, Al-Rubeai M, Rice JH. Surface-enhanced Raman scattering for rapid hematopoietic stem cell differentiation analysis. APPLIED OPTICS 2018; 57:E184-E189. [PMID: 30117870 DOI: 10.1364/ao.57.00e184] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/29/2018] [Indexed: 05/27/2023]
Abstract
Raman-spectroscopy-based methods, such as surface-enhanced Raman spectroscopy, are a well-evolved method to molecular fingerprint cell types. Here we demonstrate that surface-enhanced Raman spectroscopy can enable us to distinguish cell development stages of bone marrow hematopoietic stem cells towards red blood cells through the identification of specific surface-enhanced Raman spectroscopy biomarkers. The approach taken here is to allow cells to take in gold nanoparticles as Raman enhancement platforms for kinetic structural observations presented here through the view of the multidimensional parameter contribution, thereby enabling profiling of bone marrow hematopoietic stem cells acquired from proliferation (stage one), differentiation (stage two), and mature red blood cells (stage three).
Collapse
|
183
|
Moore KS, von Lindern M. RNA Binding Proteins and Regulation of mRNA Translation in Erythropoiesis. Front Physiol 2018; 9:910. [PMID: 30087616 PMCID: PMC6066521 DOI: 10.3389/fphys.2018.00910] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/21/2018] [Indexed: 12/12/2022] Open
Abstract
Control of gene expression in erythropoiesis has to respond to signals that may emerge from intracellular processes or environmental factors. Control of mRNA translation allows for relatively rapid modulation of protein synthesis from the existing transcriptome. For instance, the protein synthesis rate needs to be reduced when reactive oxygen species or unfolded proteins accumulate in the cells, but also when iron supply is low or when growth factors are lacking in the environment. In addition, regulation of mRNA translation can be important as an additional layer of control on top of gene transcription, in which RNA binding proteins (RBPs) can modify translation of a set of transcripts to the cell’s actual protein requirement. The 5′ and 3′ untranslated regions of mRNA (5′UTR, 3′UTR) contain binding sites for general and sequence specific translation factors. They also contain secondary structures that may hamper scanning of the 5′UTR by translation complexes or may help to recruit translation factors. In addition, the term 5′UTR is not fully correct because many transcripts contain small open reading frames in their 5′UTR that are translated and contribute to regulation of mRNA translation. It is becoming increasingly clear that the transcriptome only partly predicts the proteome. The aim of this review is (i) to summarize how the availability of general translation initiation factors can selectively regulate transcripts because the 5′UTR contains secondary structures or short translated sequences, (ii) to discuss mechanisms that control the length of the mRNA poly(A) tail in relation to mRNA translation, and (iii) to give examples of sequence specific RBPs and their targets. We focused on transcripts and RBPs required for erythropoiesis. Whereas differentiation of erythroblasts to erythrocytes is orchestrated by erythroid transcription factors, the production of erythrocytes needs to respond to the availability of growth factors and nutrients, particularly the availability of iron.
Collapse
Affiliation(s)
- Kat S Moore
- Department of Hematopoiesis, Sanquin Research, and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, Netherlands
| | - Marieke von Lindern
- Department of Hematopoiesis, Sanquin Research, and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, Netherlands
| |
Collapse
|
184
|
Dahlin JS, Hamey FK, Pijuan-Sala B, Shepherd M, Lau WWY, Nestorowa S, Weinreb C, Wolock S, Hannah R, Diamanti E, Kent DG, Göttgens B, Wilson NK. A single-cell hematopoietic landscape resolves 8 lineage trajectories and defects in Kit mutant mice. Blood 2018; 131:e1-e11. [PMID: 29588278 PMCID: PMC5969381 DOI: 10.1182/blood-2017-12-821413] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/16/2018] [Indexed: 12/19/2022] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) maintain the adult blood system, and their dysregulation causes a multitude of diseases. However, the differentiation journeys toward specific hematopoietic lineages remain ill defined, and system-wide disease interpretation remains challenging. Here, we have profiled 44 802 mouse bone marrow HSPCs using single-cell RNA sequencing to provide a comprehensive transcriptional landscape with entry points to 8 different blood lineages (lymphoid, megakaryocyte, erythroid, neutrophil, monocyte, eosinophil, mast cell, and basophil progenitors). We identified a common basophil/mast cell bone marrow progenitor and characterized its molecular profile at the single-cell level. Transcriptional profiling of 13 815 HSPCs from the c-Kit mutant (W41/W41) mouse model revealed the absence of a distinct mast cell lineage entry point, together with global shifts in cell type abundance. Proliferative defects were accompanied by reduced Myc expression. Potential compensatory processes included upregulation of the integrated stress response pathway and downregulation of proapoptotic gene expression in erythroid progenitors, thus providing a template of how large-scale single-cell transcriptomic studies can bridge between molecular phenotypes and quantitative population changes.
Collapse
Affiliation(s)
- Joakim S Dahlin
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Fiona K Hamey
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Blanca Pijuan-Sala
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Mairi Shepherd
- Department of Haematology, University of Cambridge, Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom; and
| | - Winnie W Y Lau
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Sonia Nestorowa
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Caleb Weinreb
- Department of Systems Biology, Harvard Medical School, Boston, MA
| | - Samuel Wolock
- Department of Systems Biology, Harvard Medical School, Boston, MA
| | - Rebecca Hannah
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Evangelia Diamanti
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - David G Kent
- Department of Haematology, University of Cambridge, Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom; and
| | - Berthold Göttgens
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Nicola K Wilson
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
| |
Collapse
|
185
|
Different effects of granulocyte colony-stimulating factor and erythropoietin on erythropoiesis. Stem Cell Res Ther 2018; 9:119. [PMID: 29720275 PMCID: PMC5930863 DOI: 10.1186/s13287-018-0877-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/08/2018] [Accepted: 04/16/2018] [Indexed: 11/24/2022] Open
Abstract
Background Red blood cells are the most abundant cells in the blood that deliver oxygen to the whole body. Erythropoietin (EPO), a positive regulator of erythropoiesis, is currently the major treatment for chronic anemia. Granulocyte colony-stimulating factor (G-CSF) is a multifunctional cytokine and a well-known regulator of hematopoietic stem cell proliferation, differentiation, and mobilization. The use of EPO in combination with G-CSF has been reported to synergistically improve erythroid responses in a group of patients with myelodysplastic syndromes who did not respond to EPO treatment alone; however, the mechanism remains unclear. Methods C57BL/6 J mice injected with G-CSF or EPO were used to compare the erythropoiesis status and the efficiency of erythroid mobilization by flow cytometry. Results In this study, we found that G-CSF induced more orthochromatophilic erythroblast production than did EPO in the bone marrow and spleen. In addition, in contrast to EPO treatments, G-CSF treatments enhanced the efficiency of the mobilization of newly synthesized reticulocytes into peripheral blood. Our results demonstrated that the effects of G-CSF on erythropoiesis and erythrocytic mobilization were independent of EPO secretion and, in contrast to EPO, G-CSF promoted progression of erythropoiesis through transition of early stage R2 (basophilic erythroblasts) to late stage R4 (orthochromatophilic erythroblasts). Conclusions We demonstrate for the first time that G-CSF treatments induce a faster erythropoiesis-enhancing response than that of EPO. These findings suggest an alternative approach to treating acute anemia, especially when patients are experiencing a clinical emergency in remote areas without proper blood bank supplies.
Collapse
|
186
|
Patel VS, Ete Chan M, Rubin J, Rubin CT. Marrow Adiposity and Hematopoiesis in Aging and Obesity: Exercise as an Intervention. Curr Osteoporos Rep 2018; 16:105-115. [PMID: 29476393 PMCID: PMC5866776 DOI: 10.1007/s11914-018-0424-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Changes in the bone marrow microenvironment, which accompany aging and obesity, including increased marrow adiposity, can compromise hematopoiesis. Here, we review deleterious shifts in molecular, cellular, and tissue activity and consider the potential of exercise to slow degenerative changes associated with aging and obesity. RECENT FINDINGS While bone marrow hematopoietic stem cells (HSC) are increased in frequency and myeloid-biased with age, the effect of obesity on HSC proliferation and differentiation remains controversial. HSC from both aged and obese environment have reduced hematopoietic reconstitution capacity following bone marrow transplant. Increased marrow adiposity affects HSC function, causing upregulation of myelopoiesis and downregulation of lymphopoiesis. Exercise, in contrast, can reduce marrow adiposity and restore hematopoiesis. The impact of marrow adiposity on hematopoiesis is determined mainly through correlations. Mechanistic studies are needed to determine a causative relationship between marrow adiposity and declines in hematopoiesis, which could aid in developing treatments for conditions that arise from disruptions in the marrow microenvironment.
Collapse
Affiliation(s)
- Vihitaben S Patel
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794-2580, USA
| | - M Ete Chan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794-2580, USA
| | - Janet Rubin
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Clinton T Rubin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794-2580, USA.
| |
Collapse
|
187
|
Zivot A, Lipton JM, Narla A, Blanc L. Erythropoiesis: insights into pathophysiology and treatments in 2017. Mol Med 2018; 24:11. [PMID: 30134792 PMCID: PMC6016880 DOI: 10.1186/s10020-018-0011-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 03/02/2018] [Indexed: 12/20/2022] Open
Abstract
Erythropoiesis is a tightly-regulated and complex process originating in the bone marrow from a multipotent stem cell and terminating in a mature, enucleated erythrocyte.Altered red cell production can result from the direct impairment of medullary erythropoiesis, as seen in the thalassemia syndromes, inherited bone marrow failure as well as in the anemia of chronic disease. Alternatively, in disorders such as sickle cell disease (SCD) as well as enzymopathies and membrane defects, medullary erythropoiesis is not, or only minimally, directly impaired. Despite these differences in pathophysiology, therapies have traditionally been non-specific, limited to symptomatic control of anemia via packed red blood cell (pRBC) transfusion, resulting in iron overload and the eventual need for iron chelation or splenectomy to reduce defective red cell destruction. Likewise, in polycythemia vera overproduction of red cells has historically been dealt with by non-specific myelosuppression or phlebotomy. With a deeper understanding of the molecular mechanisms underlying disease pathophysiology, new therapeutic targets have been identified including induction of fetal hemoglobin, interference with aberrant signaling pathways and gene therapy for definitive cure. This review, utilizing some representative disorders of erythropoiesis, will highlight novel therapeutic modalities currently in development for treatment of red cell disorders.
Collapse
Affiliation(s)
- Andrea Zivot
- Laboratory of Developmental Erythropoiesis, Center for Autoimmune, Musculoskeletal, and Hematopoietic Diseases, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
- Division of Pediatrics Hematology/Oncology and Stem Cell Transplantation, Cohen Children's Medical Center, New Hyde Park, NY, 11040, USA
| | - Jeffrey M Lipton
- Laboratory of Developmental Erythropoiesis, Center for Autoimmune, Musculoskeletal, and Hematopoietic Diseases, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
- Division of Pediatrics Hematology/Oncology and Stem Cell Transplantation, Cohen Children's Medical Center, New Hyde Park, NY, 11040, USA
- Stanford University School of Medicine, Stanford, CA, USA
| | - Anupama Narla
- Department of Molecular Medicine and Pediatrics, Donald and Barbara Zucker School of Medicine at Hofstra Northwell, Hempstead, NY, 11549, USA
| | - Lionel Blanc
- Laboratory of Developmental Erythropoiesis, Center for Autoimmune, Musculoskeletal, and Hematopoietic Diseases, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
- Division of Pediatrics Hematology/Oncology and Stem Cell Transplantation, Cohen Children's Medical Center, New Hyde Park, NY, 11040, USA.
- Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
188
|
Fye HKS, Mrosso P, Bruce L, Thézénas ML, Davis S, Fischer R, Rwegasira GL, Makani J, Kessler BM. A robust mass spectrometry method for rapid profiling of erythrocyte ghost membrane proteomes. Clin Proteomics 2018; 15:14. [PMID: 29588628 PMCID: PMC5863380 DOI: 10.1186/s12014-018-9190-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/13/2018] [Indexed: 11/13/2022] Open
Abstract
Background Red blood cell (RBC) physiology is directly linked to many human disorders associated with low tissue oxygen levels or anemia including chronic obstructive pulmonary disease, congenital heart disease, sleep apnea and sickle cell anemia. Parasites such as Plasmodium spp. and phylum Apicomplexa directly target RBCs, and surface molecules within the RBC membrane are critical for pathogen interactions. Proteomics of RBC membrane ‘ghost’ fractions has therefore been of considerable interest, but protocols described to date are either suboptimal or too extensive to be applicable to a larger set of clinical cohorts. Methods Here, we describe an optimised erythrocyte isolation protocol from blood, tested for various storage conditions and explored using different fractionation conditions for isolating ghost RBC membranes. Liquid chromatography mass spectrometry (LC–MS) analysis on a Q-Exactive Orbitrap instrument was used to profile proteins isolated from the comparative conditions. Data analysis was run on the MASCOT and MaxQuant platforms to assess their scope and diversity. Results The results obtained demonstrate a robust method for membrane enrichment enabling consistent MS based characterisation of > 900 RBC membrane proteins in single LC–MS/MS analyses. Non-detergent based membrane solubilisation methods using the tissue and supernatant fractions of isolated ghost membranes are shown to offer effective haemoglobin removal as well as diverse recovery including erythrocyte membrane proteins of high and low abundance. Conclusions The methods described in this manuscript propose a medium to high throughput framework for membrane proteome profiling by LC–MS of potential applicability to larger clinical cohorts in a variety of disease contexts. Electronic supplementary material The online version of this article (10.1186/s12014-018-9190-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Haddy K S Fye
- 1Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ UK
| | - Paul Mrosso
- 2Muhimbili Wellcome Programme, Muhimbili University of Health and Allied Sciences, PO Box 65001, Dar es Salaam, Tanzania
| | - Lesley Bruce
- 3Bristol Institute for Transfusion Sciences, NHS Blood and Transplant, 500 North Bristol Park, Filton, Bristol, BS34 7QH UK
| | - Marie-Laëtitia Thézénas
- 1Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ UK
| | - Simon Davis
- 1Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ UK
| | - Roman Fischer
- 1Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ UK
| | - Gration L Rwegasira
- 2Muhimbili Wellcome Programme, Muhimbili University of Health and Allied Sciences, PO Box 65001, Dar es Salaam, Tanzania
| | - Julie Makani
- 2Muhimbili Wellcome Programme, Muhimbili University of Health and Allied Sciences, PO Box 65001, Dar es Salaam, Tanzania
| | - Benedikt M Kessler
- 1Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ UK
| |
Collapse
|
189
|
Lippi G, Turcato G, Cervellin G, Sanchis-Gomar F. Red blood cell distribution width in heart failure: A narrative review. World J Cardiol 2018; 10:6-14. [PMID: 29487727 PMCID: PMC5827617 DOI: 10.4330/wjc.v10.i2.6] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 01/23/2018] [Accepted: 02/05/2018] [Indexed: 02/07/2023] Open
Abstract
The red blood cell distribution width (RDW) is a simple, rapid, inexpensive and straightforward hematological parameter, reflecting the degree of anisocytosis in vivo. The currently available scientific evidence suggests that RDW assessment not only predicts the risk of adverse outcomes (cardiovascular and all-cause mortality, hospitalization for acute decompensation or worsened left ventricular function) in patients with acute and chronic heart failure (HF), but is also a significant and independent predictor of developing HF in patients free of this condition. Regarding the biological interplay between impaired hematopoiesis and cardiac dysfunction, many of the different conditions associated with increased heterogeneity of erythrocyte volume (i.e., ageing, inflammation, oxidative stress, nutritional deficiencies and impaired renal function), may be concomitantly present in patients with HF, whilst anisocytosis may also directly contribute to the development and worsening of HF. In conclusion, the longitudinal assessment of RDW changes over time may be considered an efficient measure to help predicting the risk of both development and progression of HF.
Collapse
|
190
|
Halder UC. Bone marrow stem cells to destroy circulating HIV: a hypothetical therapeutic strategy. ACTA ACUST UNITED AC 2018; 25:3. [PMID: 29445623 PMCID: PMC5800069 DOI: 10.1186/s40709-018-0075-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 01/27/2018] [Indexed: 12/19/2022]
Abstract
Human immunodeficiency virus (HIV) still poses enigmatic threats to human life. This virus has mastered in bypassing anti retroviral therapy leading to patients’ death. Circulating viruses are phenomenal for the disease outcome. This hypothesis proposes a therapeutic strategy utilizing receptor-integrated hematopoietic, erythroid and red blood cells. Here, HIV specific receptors trap circulating viruses that enter erythrocyte cytoplasm and form inactive integration complex. This model depicts easy, effective removal of circulating HIV without any adverse effect.
Collapse
Affiliation(s)
- Umesh Chandra Halder
- Department of Zoology, Raniganj Girls' College, Searsole, Rajbari, Raniganj, Paschim Barddhaman, West Bengal 713358 India
| |
Collapse
|
191
|
Ostrowski D, Heinrich R. Alternative Erythropoietin Receptors in the Nervous System. J Clin Med 2018; 7:E24. [PMID: 29393890 PMCID: PMC5852440 DOI: 10.3390/jcm7020024] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 01/24/2018] [Accepted: 01/26/2018] [Indexed: 12/18/2022] Open
Abstract
In addition to its regulatory function in the formation of red blood cells (erythropoiesis) in vertebrates, Erythropoietin (Epo) contributes to beneficial functions in a variety of non-hematopoietic tissues including the nervous system. Epo protects cells from apoptosis, reduces inflammatory responses and supports re-establishment of compromised functions by stimulating proliferation, migration and differentiation to compensate for lost or injured cells. Similar neuroprotective and regenerative functions of Epo have been described in the nervous systems of both vertebrates and invertebrates, indicating that tissue-protective Epo-like signaling has evolved prior to its erythropoietic function in the vertebrate lineage. Epo mediates its erythropoietic function through a homodimeric Epo receptor (EpoR) that is also widely expressed in the nervous system. However, identification of neuroprotective but non-erythropoietic Epo splice variants and Epo derivatives indicated the existence of other types of Epo receptors. In this review, we summarize evidence for potential Epo receptors that might mediate Epo's tissue-protective function in non-hematopoietic tissue, with focus on the nervous system. In particular, besides EpoR, we discuss three other potential neuroprotective Epo receptors: (1) a heteroreceptor consisting of EpoR and common beta receptor (βcR), (2) the Ephrin (Eph) B4 receptor and (3) the human orphan cytokine receptor-like factor 3 (CRLF3).
Collapse
Affiliation(s)
- Daniela Ostrowski
- Department of Biology, Truman State University, Kirksville, MO 63501, USA.
| | - Ralf Heinrich
- Department of Cellular Neurobiology, Institute for Zoology, Georg-August-University Göttingen, 37073 Göttingen, Germany.
| |
Collapse
|
192
|
McIver SC, Hewitt KJ, Gao X, Mehta C, Zhang J, Bresnick EH. Dissecting Regulatory Mechanisms Using Mouse Fetal Liver-Derived Erythroid Cells. Methods Mol Biol 2018; 1698:67-89. [PMID: 29076084 DOI: 10.1007/978-1-4939-7428-3_4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Multipotent hematopoietic stem cells differentiate into an ensemble of committed progenitor cells that produce the diverse blood cells essential for life. Physiological mechanisms governing hematopoiesis, and mechanistic aberrations underlying non-malignant and malignant hematologic disorders, are often very similar in mouse and man. Thus, mouse models provide powerful systems for unraveling mechanisms that control hematopoietic stem/progenitor cell (HSPC) function in their resident microenvironments in vivo. Ex vivo systems, involving the culture of HSPCs generated in vivo, allow one to dissociate microenvironment-based and cell intrinsic mechanisms, and therefore have considerable utility. Dissecting mechanisms controlling cellular proliferation and differentiation is facilitated by the use of primary cells, since mutations and chromosome aberrations in immortalized and cancer cell lines corrupt normal mechanisms. Primary erythroid precursor cells can be expanded or differentiated in culture to yield large numbers of progeny at discrete maturation stages. We described a robust method for isolation, culture, and analysis of primary mouse erythroid precursor cells and their progeny.
Collapse
Affiliation(s)
- Skye C McIver
- Department of Cell and Regenerative Biology, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, 4009 WIMR, 1111 Highland Ave, Madison, WI, 53705, USA.,UW-Madison Blood Research Program, University of Wisconsin, Madison, WI, 53705, USA
| | - Kyle J Hewitt
- Department of Cell and Regenerative Biology, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, 4009 WIMR, 1111 Highland Ave, Madison, WI, 53705, USA.,UW-Madison Blood Research Program, University of Wisconsin, Madison, WI, 53705, USA
| | - Xin Gao
- Department of Cell and Regenerative Biology, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, 4009 WIMR, 1111 Highland Ave, Madison, WI, 53705, USA.,UW-Madison Blood Research Program, University of Wisconsin, Madison, WI, 53705, USA
| | - Charu Mehta
- Department of Cell and Regenerative Biology, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, 4009 WIMR, 1111 Highland Ave, Madison, WI, 53705, USA.,UW-Madison Blood Research Program, University of Wisconsin, Madison, WI, 53705, USA
| | - Jing Zhang
- UW-Madison Blood Research Program, University of Wisconsin, Madison, WI, 53705, USA.,McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, 53705, USA
| | - Emery H Bresnick
- Department of Cell and Regenerative Biology, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, 4009 WIMR, 1111 Highland Ave, Madison, WI, 53705, USA. .,UW-Madison Blood Research Program, University of Wisconsin, Madison, WI, 53705, USA.
| |
Collapse
|
193
|
Vinjamur DS, Bauer DE. Growing and Genetically Manipulating Human Umbilical Cord Blood-Derived Erythroid Progenitor (HUDEP) Cell Lines. Methods Mol Biol 2018; 1698:275-284. [PMID: 29076097 DOI: 10.1007/978-1-4939-7428-3_17] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The recently established human umbilical cord blood-derived erythroid progenitor (HUDEP) cell lines have equipped red blood cell researchers with valuable in vitro models of erythroid development. Of the three established HUDEP cell lines, HUDEP-2 cells express predominantly adult β-globin and most closely resemble adult erythroid cells. This chapter describes culture protocols for the maintenance and erythroid differentiation of HUDEP-2 cells. Methods to genetically manipulate HUDEP-2 cells using a CRISPR/Cas9 nuclease-based approach are also discussed.
Collapse
Affiliation(s)
- Divya S Vinjamur
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Daniel E Bauer
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA. .,Dana-Farber Cancer Institute, Harvard Medical School, Harvard Stem Cell Institute, Karp 8211, 1 Blackfan Circle, Boston, MA, 02115, USA.
| |
Collapse
|
194
|
Mumau MD, Vanderbeck AN, Lynch ED, Golec SB, Emerson SG, Punt JA. Identification of a Multipotent Progenitor Population in the Spleen That Is Regulated by NR4A1. THE JOURNAL OF IMMUNOLOGY 2017; 200:1078-1087. [PMID: 29282309 DOI: 10.4049/jimmunol.1701250] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/21/2017] [Indexed: 01/06/2023]
Abstract
The developmental fate of hematopoietic stem and progenitor cells is influenced by their physiological context. Although most hematopoietic stem and progenitor cells are found in the bone marrow of the adult, some are found in other tissues, including the spleen. The extent to which the fate of stem cells is determined by the tissue in which they reside is not clear. In this study, we identify a new progenitor population, which is enriched in the mouse spleen, defined by cKit+CD71lowCD24high expression. This previously uncharacterized population generates exclusively myeloid lineage cells, including erythrocytes, platelets, monocytes, and neutrophils. These multipotent progenitors of the spleen (MPPS) develop from MPP2, a myeloid-biased subset of hematopoietic progenitors. We find that NR4A1, a transcription factor expressed by myeloid-biased long term-hematopoietic stem cells, guides the lineage specification of MPPS. In vitro, NR4A1 expression regulates the potential of MPPS to differentiate into erythroid cells. MPPS that express NR4A1 differentiate into a variety of myeloid lineages, whereas those that do not express NR4A1 primarily develop into erythroid cells. Similarly, in vivo, after adoptive transfer, Nr4a1-deficient MPPS contribute more to erythrocyte and platelet populations than do wild-type MPPS. Finally, unmanipulated Nr4a1-/- mice harbor significantly higher numbers of erythroid progenitors in the spleen compared with wild-type mice. Together, our data show that NR4A1 expression by MPPS limits erythropoiesis and megakaryopoeisis, permitting development to other myeloid lineages. This effect is specific to the spleen, revealing a unique molecular pathway that regulates myeloid bias in an extramedullary niche.
Collapse
Affiliation(s)
- Melanie D Mumau
- Herbert Irving Comprehensive Cancer Research Center, Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032
| | - Ashley N Vanderbeck
- Herbert Irving Comprehensive Cancer Research Center, Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032
| | - Elizabeth D Lynch
- Herbert Irving Comprehensive Cancer Research Center, Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032
| | - Sophia B Golec
- Herbert Irving Comprehensive Cancer Research Center, Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032
| | - Stephen G Emerson
- Herbert Irving Comprehensive Cancer Research Center, Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032
| | - Jennifer A Punt
- Herbert Irving Comprehensive Cancer Research Center, Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032
| |
Collapse
|
195
|
Kamran N, Li Y, Sierra M, Alghamri MS, Kadiyala P, Appelman HD, Edwards M, Lowenstein PR, Castro MG. Melanoma induced immunosuppression is mediated by hematopoietic dysregulation. Oncoimmunology 2017; 7:e1408750. [PMID: 29399415 PMCID: PMC5790391 DOI: 10.1080/2162402x.2017.1408750] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/16/2017] [Accepted: 11/17/2017] [Indexed: 01/10/2023] Open
Abstract
Tumors are associated with expansion of immunosuppressive cells such as tumor associated macrophages (TAMs), regulatory T cells (Tregs) and myeloid derived suppressor cells (MDSCs). These cells promote tumor growth, angiogenesis, metastasis and immune escape. Cancer patients frequently present symptoms such as anemia, leukocytosis and/or cytopenia; associated with poor prognosis. To uncover tumor-mediated hematopoietic abnormalities and identify novel targets that can be harnessed to improve tumor-specific immune responses, we investigated the hematopoietic stem and progenitor cell compartment in melanoma bearing mice. We show that melanoma growth results in expansion of myeloid lineages such as MDSCs, macrophages and DCs along with a reduction in mature RBCs and platelets. Mature B lymphocytes in the blood and BM of melanoma mice were also reduced. Mice bearing melanoma showed extramedullary hematopoiesis in the spleen. Increased expansion of myeloid lineages occurred directly at the level of stem and progenitor cells. The reduction in mature B lymphocytes resulted from a block at the Pro-B cell stage in the bone marrow. Addition of recombinant IL-3 to bone marrow cells resulted in the expansion of committed myeloid progenitors including common myeloid precursors, granulocyte-monocyte precursors and megakaryocyte-erythrocyte precursors. In vivo, IL-3 receptor stimulation in melanoma bearing mice using an IL-3 antibody also resulted in a robust expansion of committed myeloid progenitors and hematopoietic stem cells. Collectively our findings demonstrate that tumor growth plays a pivotal role in reprogramming the host immune system by impacting hematopoiesis directly at the level of stem cell compartment.
Collapse
Affiliation(s)
- Neha Kamran
- Department of Neurosurgery, University of Michigan Medical School, MSRB II, RM 4570 C, 1150 West Medical Center Drive, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, MSRB II, RM 4570 C, 1150 West Medical Center Drive, Ann Arbor, MI, USA
| | - Youping Li
- Department of Neurosurgery, University of Michigan Medical School, MSRB II, RM 4570 C, 1150 West Medical Center Drive, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, MSRB II, RM 4570 C, 1150 West Medical Center Drive, Ann Arbor, MI, USA
| | - Maria Sierra
- Department of Neurosurgery, University of Michigan Medical School, MSRB II, RM 4570 C, 1150 West Medical Center Drive, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, MSRB II, RM 4570 C, 1150 West Medical Center Drive, Ann Arbor, MI, USA
| | - Mahmoud S. Alghamri
- Department of Neurosurgery, University of Michigan Medical School, MSRB II, RM 4570 C, 1150 West Medical Center Drive, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, MSRB II, RM 4570 C, 1150 West Medical Center Drive, Ann Arbor, MI, USA
| | - Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School, MSRB II, RM 4570 C, 1150 West Medical Center Drive, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, MSRB II, RM 4570 C, 1150 West Medical Center Drive, Ann Arbor, MI, USA
| | - Henry D. Appelman
- Department of Pathology, University of Michigan Medical School, 5220 Med Sci I, Ann Arbor, MI, USA
| | - Marta Edwards
- Department of Neurosurgery, University of Michigan Medical School, MSRB II, RM 4570 C, 1150 West Medical Center Drive, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, MSRB II, RM 4570 C, 1150 West Medical Center Drive, Ann Arbor, MI, USA
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, MSRB II, RM 4570 C, 1150 West Medical Center Drive, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, MSRB II, RM 4570 C, 1150 West Medical Center Drive, Ann Arbor, MI, USA
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan Medical School, MSRB II, RM 4570 C, 1150 West Medical Center Drive, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, MSRB II, RM 4570 C, 1150 West Medical Center Drive, Ann Arbor, MI, USA
| |
Collapse
|
196
|
Abstract
Iron is an essential trace element, but it is also toxic in excess, and thus mammals have developed elegant mechanisms for keeping both cellular and whole-body iron concentrations within the optimal physiologic range. In the diet, iron is either sequestered within heme or in various nonheme forms. Although the absorption of heme iron is poorly understood, nonheme iron is transported across the apical membrane of the intestinal enterocyte by divalent metal-ion transporter 1 (DMT1) and is exported into the circulation via ferroportin 1 (FPN1). Newly absorbed iron binds to plasma transferrin and is distributed around the body to sites of utilization with the erythroid marrow having particularly high iron requirements. Iron-loaded transferrin binds to transferrin receptor 1 on the surface of most body cells, and after endocytosis of the complex, iron enters the cytoplasm via DMT1 in the endosomal membrane. This iron can be used for metabolic functions, stored within cytosolic ferritin, or exported from the cell via FPN1. Cellular iron concentrations are modulated by the iron regulatory proteins (IRPs) IRP1 and IRP2. At the whole-body level, dietary iron absorption and iron export from the tissues into the plasma are regulated by the liver-derived peptide hepcidin. When tissue iron demands are high, hepcidin concentrations are low and vice versa. Too little or too much iron can have important clinical consequences. Most iron deficiency reflects an inadequate supply of iron in the diet, whereas iron excess is usually associated with hereditary disorders. These disorders include various forms of hemochromatosis, which are characterized by inadequate hepcidin production and, thus, increased dietary iron intake, and iron-loading anemias whereby both increased iron absorption and transfusion therapy contribute to the iron overload. Despite major recent advances, much remains to be learned about iron physiology and pathophysiology.
Collapse
Affiliation(s)
- Gregory J Anderson
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia; and Schools of
- Chemistry and Molecular Bioscience and
- Medicine, University of Queensland, St. Lucia, Queensland, Australia
| | - David M Frazer
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia; and Schools of
| |
Collapse
|
197
|
Bozler J, Kacsoh BZ, Bosco G. Nematocytes: Discovery and characterization of a novel anculeate hemocyte in Drosophila falleni and Drosophila phalerata. PLoS One 2017; 12:e0188133. [PMID: 29141015 PMCID: PMC5687758 DOI: 10.1371/journal.pone.0188133] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 11/01/2017] [Indexed: 12/27/2022] Open
Abstract
Immune challenges, such as parasitism, can be so pervasive and deleterious that they constitute an existential threat to a species' survival. In response to these ecological pressures, organisms have developed a wide array of novel behavioral, cellular, and molecular adaptations. Research into these immune defenses in model systems has resulted in a revolutionary understanding of evolution and functional biology. As the field has expanded beyond the limited number of model organisms our appreciation of evolutionary innovation and unique biology has widened as well. With this in mind, we have surveyed the hemolymph of several non-model species of Drosophila. Here we identify and describe a novel hemocyte, type-II nematocytes, found in larval stages of numerous Drosophila species. Examined in detail in Drosophila falleni and Drosophila phalerata, we find that these remarkable cells are distinct from previously described hemocytes due to their anucleate state (lacking a nucleus) and unusual morphology. Type-II nematocytes are long, narrow cells with spindle-like projections extending from a cell body with high densities of mitochondria and microtubules, and exhibit the ability to synthesize proteins. These properties are unexpected for enucleated cells, and together with our additional characterization, we demonstrate that these type-II nematocytes represent a biological novelty. Surprisingly, despite the absence of a nucleus, we observe through live cell imaging that these cells remain motile with a highly dynamic cellular shape. Furthermore, these cells demonstrate the ability to form multicellular structures, which we suggest may be a component of the innate immune response to macro-parasites. In addition, live cell imaging points to a large nucleated hemocyte, type-I nematocyte, as the progenitor cell, leading to enucleation through a budding or asymmetrical division process rather than nuclear ejection: This study is the first to report such a process of enucleation. Here we describe these cells in detail for the first time and examine their evolutionary history in Drosophila.
Collapse
Affiliation(s)
- Julianna Bozler
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Balint Z. Kacsoh
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Giovanni Bosco
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| |
Collapse
|
198
|
Liu Z, Feng Q, Sun P, Lu Y, Yang M, Zhang X, Jin X, Li Y, Lu SJ, Quan C. Genome-wide DNA methylation drives human embryonic stem cell erythropoiesis by remodeling gene expression dynamics. Epigenomics 2017; 9:1543-1558. [PMID: 29135282 DOI: 10.2217/epi-2017-0039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To investigate the role of DNA methylation during erythrocyte production by human embryonic stem cells (hESCs). METHODS We employed an erythroid differentiation model from hESCs, and then tracked the genome-wide DNA methylation maps and gene expression patterns through an Infinium HumanMethylation450K BeadChip and an Ilumina Human HT-12 v4 Expression Beadchip, respectively. RESULTS A negative correlation between DNA methylation and gene expression was substantially enriched during the later differentiation stage and was present in both the promoter and the gene body. Moreover, erythropoietic genes with differentially methylated CpG sites that were primarily enriched in nonisland regions were upregulated, and demethylation of their gene bodies was associated with the presence of enhancers and DNase I hypersensitive sites. Finally, the components of JAK-STAT-NF-κB signaling were DNA hypomethylated and upregulated, which targets the key genes for erythropoiesis. CONCLUSION Erythroid lineage commitment by hESCs requires genome-wide DNA methylation modifications to remodel gene expression dynamics.
Collapse
Affiliation(s)
- Zhijing Liu
- Department of Pathology, Qingdao Municipal Hospital, Affiliated with Qingdao University, 1 Jiaozhou Road, Qingdao 266000, Shandong, China
| | - Qiang Feng
- Vcanbio Center for Translational Biotechnology, 21 Strathmore Road, Natick, MA 01760, USA
| | - Pengpeng Sun
- Department of Critical Care Medicine, Qingdao Center Medical Group, Affiliated with Qingdao University, 127 Siliunan Road, Qingdao 266042, Shandong, China
| | - Yan Lu
- Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun 130021, China
| | - Minlan Yang
- Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun 130021, China
| | - Xiaowei Zhang
- Center for Translational Medicine, Central Hospital of Zibo, Affiliated with Shandong University, 54 Gongqingtuan Road, Zibo 255000, Shandong, China
| | - Xiangshu Jin
- Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun 130021, China
| | - Yulin Li
- Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun 130021, China
| | - Shi-Jiang Lu
- Vcanbio Center for Translational Biotechnology, 21 Strathmore Road, Natick, MA 01760, USA
| | - Chengshi Quan
- Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun 130021, China
| |
Collapse
|
199
|
Javan GT, Salhotra A, Finley SJ, Soni S. Erythroblast macrophage protein (Emp): Past, present, and future. Eur J Haematol 2017; 100:3-9. [DOI: 10.1111/ejh.12983] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Gulnaz T. Javan
- Physical Sciences Department Forensic Science Program Alabama State University Montgomery AL USA
| | | | - Sheree J. Finley
- Physical Sciences Department Alabama State University Montgomery AL USA
| | - Shivani Soni
- Department of Biological Sciences California State University Fullerton CA USA
- Department of Biological Science Schmid College of Science and Technology Chapman University Irvine CA USA
| |
Collapse
|
200
|
Reichel C, Gmeiner G, Thevis M. Antibody‐based strategies for the detection of Luspatercept (ACE‐536) in human serum. Drug Test Anal 2017; 9:1721-1730. [DOI: 10.1002/dta.2302] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/08/2017] [Accepted: 09/12/2017] [Indexed: 02/01/2023]
Affiliation(s)
- Christian Reichel
- Doping Control LaboratorySeibersdorf Labor GmbH An der Bundesstrasse 60 A‐2444 Seibersdorf Austria
| | - Günter Gmeiner
- Doping Control LaboratorySeibersdorf Labor GmbH An der Bundesstrasse 60 A‐2444 Seibersdorf Austria
| | - Mario Thevis
- Institute of Biochemistry / Center for Preventive Doping Research, German Sport University Cologne Am Sportpark Muengersdorf 6 50933 Cologne Germany
- European Monitoring Center for Emerging Doping AgentsGerman Sport University Cologne Am Sportpark Muengersdorf 6 50933 Cologne Germany
| |
Collapse
|