1
|
Meroni M, Dongiovanni P. PNPLA3 rs738409 Genetic Variant Inversely Correlates with Platelet Count, Thereby Affecting the Performance of Noninvasive Scores of Hepatic Fibrosis. Int J Mol Sci 2023; 24:15046. [PMID: 37894727 PMCID: PMC10606003 DOI: 10.3390/ijms242015046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/03/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Noninvasive tests (NITs) including platelets (PLTs) have been proposed to replace hepatic biopsy for the diagnosis of nonalcoholic fatty liver disease (NAFLD), or as more recently redefined, metabolic dysfunction-associated steatotic liver disease (MASLD). There has been reported an inverse correlation between PLTs and progressive MASLD, which is also affected by the patatin-like phospholipase domain-containing protein 3 (PNPLA3) rs738409 C>G mutation. However, the correlation between low PLTs and PNPLA3 genotype has been poorly investigated. We stratified 1155 biopsy-proven MASLD patients according to PNPLA3 genotype. The hepatic expression of genes involved in megakaryopoiesis was investigated in n = 167 bariatric patients by RNAseq. PLT count progressively decreased according to the number of PNPLA3 at-risk alleles, irrespective of the presence of advanced fibrosis. The hepatic expression of genes involved in PLT biogenesis was associated with the PNPLA3 GG genotype. Finally, the presence of the PNPLA3 homozygosity flattened the accuracy of fibrosis-4 (FIB-4) in discriminating histological fibrosis stages. The PNPLA3 GG genotype may underpower the accuracy of NITs which include PLT count in identifying those patients with potentially reversible stages of fibrosis.
Collapse
Affiliation(s)
| | - Paola Dongiovanni
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy;
| |
Collapse
|
2
|
Bomfim BCM, Azevedo-Silva J, Caminha G, Santos JPR, Pelajo-Machado M, de Paula Ayres-Silva J. Lectin-based carbohydrate profile of megakaryocytes in murine fetal liver during development. Sci Rep 2023; 13:6729. [PMID: 37185919 PMCID: PMC10130079 DOI: 10.1038/s41598-023-32863-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Hematopoiesis is the process by which blood cells are generated. During embryonic development, these cells migrate through different organs until they reach the bone marrow, their definitive place in adulthood. Around E10.5, the fetal liver starts budding from the gut, where first hematopoietic cells arrive and expand. Hematopoietic cell migration occurs through cytokine stimulation, receptor expression, and glycosylation patterns on the cell surface. In addition, carbohydrates can modulate different cell activation states. For this reason, we aimed to characterize and quantify fetal megakaryocytic cells in mouse fetal liver according to their glycan residues at different gestational ages through lectins. Mouse fetuses between E11.5 and E18.5 were formalin-fixed and, paraffin-embedded, for immunofluorescence analysis using confocal microscopy. The results showed that the following sugar residues were expressed in proliferating and differentiating megakaryocytes in the fetal liver at different gestational ages: α-mannose, α-glucose, galactose, GlcNAc, and two types of complex oligosaccharides. Megakaryocytes also showed three proliferation waves during liver development at E12.5, E14.5, and E18.5. Additionally, the lectins that exhibited high and specific pattern intensities at liver capsules and vessels were shown to be a less time-consuming and robust alternative alternative to conventional antibodies for displaying liver structures such as capsules and vessels, as well as for megakaryocyte differentiation in the fetal liver.
Collapse
Affiliation(s)
| | - Jessyca Azevedo-Silva
- Laboratory of Pathology, Oswaldo Cruz Institute - Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Giulia Caminha
- Laboratory of Pathology, Oswaldo Cruz Institute - Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | | | - Marcelo Pelajo-Machado
- Laboratory of Pathology, Oswaldo Cruz Institute - Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | | |
Collapse
|
3
|
Šrajer Gajdošik M, Kovač Peić A, Begić M, Grbčić P, Brilliant KE, Hixson DC, Josić D. Possible Role of Extracellular Vesicles in Hepatotoxicity of Acetaminophen. Int J Mol Sci 2022; 23:8870. [PMID: 36012131 PMCID: PMC9408656 DOI: 10.3390/ijms23168870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 07/29/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022] Open
Abstract
We examined proteomic profiles of rat liver extracellular vesicles (EVs) shed following treatment with a sub-toxic dose (500 mg/kg) of the pain reliever drug, acetaminophen (APAP). EVs representing the entire complement of hepatic cells were isolated after perfusion of the intact liver and analyzed with LC-MS/MS. The investigation was focused on revealing the function and cellular origin of identified EVs proteins shed by different parenchymal and non-parenchymal liver cells and their possible role in an early response of this organ to a toxic environment. Comparison of EV proteomic profiles from control and APAP-treated animals revealed significant differences. Alpha-1-macroglobulin and members of the cytochrome P450 superfamily were highly abundant proteins in EVs shed by the normal liver. In contrast, proteins like aminopeptidase N, metalloreductase STEAP4, different surface antigens like CD14 and CD45, and most members of the annexin family were detected only in EVs that were shed by livers of APAP-treated animals. In EVs from treated livers, there was almost a complete disappearance of members of the cytochrome P450 superfamily and a major decrease in other enzymes involved in the detoxification of xenobiotics. Additionally, there were proteins that predominated in non-parenchymal liver cells and in the extracellular matrix, like fibronectin, receptor-type tyrosine-protein phosphatase C, and endothelial type gp91. These differences indicate that even treatment with a sub-toxic concentration of APAP initiates dramatic perturbation in the function of this vital organ.
Collapse
Affiliation(s)
| | | | - Marija Begić
- Faculty of Medicine, University Juraj Dobrila of Pula, 52100 Pula, Croatia
| | - Petra Grbčić
- Faculty of Medicine, University Juraj Dobrila of Pula, 52100 Pula, Croatia
| | - Kate E. Brilliant
- Proteomics Core, COBRE CCRD, Rhode Island Hospital, Providence, RI 02903, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Douglas C. Hixson
- Proteomics Core, COBRE CCRD, Rhode Island Hospital, Providence, RI 02903, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Djuro Josić
- Faculty of Medicine, University Juraj Dobrila of Pula, 52100 Pula, Croatia
- Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| |
Collapse
|
4
|
Lawrence M, Shahsavari A, Bornelöv S, Moreau T, McDonald R, Vallance TM, Kania K, Paramor M, Baye J, Perrin M, Steindel M, Jimenez-Gomez P, Penfold C, Mohorianu I, Ghevaert C. Mapping the biogenesis of forward programmed megakaryocytes from induced pluripotent stem cells. SCIENCE ADVANCES 2022; 8:eabj8618. [PMID: 35171685 PMCID: PMC8849335 DOI: 10.1126/sciadv.abj8618] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 12/23/2021] [Indexed: 06/14/2023]
Abstract
Platelet deficiency, known as thrombocytopenia, can cause hemorrhage and is treated with platelet transfusions. We developed a system for the production of platelet precursor cells, megakaryocytes, from pluripotent stem cells. These cultures can be maintained for >100 days, implying culture renewal by megakaryocyte progenitors (MKPs). However, it is unclear whether the MKP state in vitro mirrors the state in vivo, and MKPs cannot be purified using conventional surface markers. We performed single-cell RNA sequencing throughout in vitro differentiation and mapped each state to its equivalent in vivo. This enabled the identification of five surface markers that reproducibly purify MKPs, allowing us insight into their transcriptional and epigenetic profiles. Last, we performed culture optimization, increasing MKP production. Together, this study has mapped parallels between the MKP states in vivo and in vitro and allowed the purification of MKPs, accelerating the progress of in vitro-derived transfusion products toward the clinic.
Collapse
Affiliation(s)
- Moyra Lawrence
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, Cambridge CB2 0AW, UK
- Department of Haematology and NHS Blood and Transplant, University of Cambridge, Cambridge, UK
| | - Arash Shahsavari
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Susanne Bornelöv
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Thomas Moreau
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, Cambridge CB2 0AW, UK
- Department of Haematology and NHS Blood and Transplant, University of Cambridge, Cambridge, UK
- Bit Bio, Discovery Drive, Cambridge Biomedical Campus, Cambridge CB2 0AX, UK
| | - Rebecca McDonald
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Thomas M. Vallance
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Katarzyna Kania
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Maike Paramor
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, Cambridge CB2 0AW, UK
| | - James Baye
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Marion Perrin
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Maike Steindel
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, Cambridge CB2 0AW, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | - Paula Jimenez-Gomez
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Christopher Penfold
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | - Irina Mohorianu
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Cedric Ghevaert
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, Cambridge CB2 0AW, UK
- Department of Haematology and NHS Blood and Transplant, University of Cambridge, Cambridge, UK
| |
Collapse
|
5
|
Goette NP, Borzone FR, Lupi ADD, Chasseing NA, Rubio MF, Costas MA, Heller PG, Marta RF, Lev PR. Megakaryocyte-stromal cell interactions: effect on megakaryocyte proliferation, proplatelet production, and survival. Exp Hematol 2022; 107:24-37. [PMID: 35032592 DOI: 10.1016/j.exphem.2022.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 11/28/2022]
Abstract
Bone marrow stromal cells provide a proper environment for the development of hematologic lineages. The incorporation of different stromal cells to in vitro culture systems would be an attractive model to study megakaryopoiesis and thrombopoiesis. Our objective was to evaluate the participation of different types of stromal cells on in vitro megakaryopoiesis, thrombopoiesis and megakaryocyte (MK) survival. CD34-positive progenitors from umbilical cord blood were differentiated into MK precursors and then co-cultured with umbilical vein endothelial cells (HUVEC), bone marrow mesenchymal stem cells (MSCs), skin fibroblasts (SF) (all human) or mouse fibroblast cell line (L929). The number of MKs (CD61-positive cells) was increased in the presence of HUVEC and SF while L929 cells decreased total and mature MK count. Concerning thrombopoiesis, HUVEC increased proplatelet (PP)-producing MKs, while MSCs, L929 and SF had the opposite effect (immunofluorescence staining and microscopic analysis). MK survival was enhanced in MSC and SF co-cultures, as assessed by evaluation of pyknotic nuclei. However, HUVEC and L929 did not prevent apoptosis of MKs. Reciprocally, the cloning efficiency of MSCs was decreased in the presence of MKs, while the ability of stromal cells (either MSCs or SF) to produce the extracellular matrix proteins type III collagen, fibronectin, dermatan sulfate, heparan sulfate and P4HB was preserved. These data indicate that each stromal cell type performs distinctive functions, which differentially modulate MK growth and platelet production, and, at the same time, that MKs also modify stromal cells behavior. Overall, our results highlight the relevance of considering the influence of stromal cells in MK research as well as the close interplay of different cell types within the bone marrow milieu.
Collapse
Affiliation(s)
- Nora P Goette
- Institute of Medical Research A Lanari, University of Buenos Aires, Buenos Aires, Argentina
| | - Francisco R Borzone
- Laboratory of Immunohematology, Institute of Biology and Experimental Medicine, National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Ailen D Discianni Lupi
- Institute of Medical Research A Lanari, University of Buenos Aires, Buenos Aires, Argentina
| | - Norma A Chasseing
- Laboratory of Immunohematology, Institute of Biology and Experimental Medicine, National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - María F Rubio
- Institute of Medical Research A Lanari, University of Buenos Aires, Buenos Aires, Argentina; Department of Molecular Biology and Apoptosis , Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Mónica A Costas
- Institute of Medical Research A Lanari, University of Buenos Aires, Buenos Aires, Argentina; Department of Molecular Biology and Apoptosis , Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Paula G Heller
- Institute of Medical Research A Lanari, University of Buenos Aires, Buenos Aires, Argentina; Department of Experimental Hematology, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Rosana F Marta
- Institute of Medical Research A Lanari, University of Buenos Aires, Buenos Aires, Argentina; Department of Experimental Hematology, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Paola R Lev
- Institute of Medical Research A Lanari, University of Buenos Aires, Buenos Aires, Argentina; Department of Experimental Hematology, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
6
|
Mack R, Zhang L, Breslin Sj P, Zhang J. The Fetal-to-Adult Hematopoietic Stem Cell Transition and its Role in Childhood Hematopoietic Malignancies. Stem Cell Rev Rep 2021; 17:2059-2080. [PMID: 34424480 DOI: 10.1007/s12015-021-10230-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2021] [Indexed: 01/07/2023]
Abstract
As with most organ systems that undergo continuous generation and maturation during the transition from fetal to adult life, the hematopoietic and immune systems also experience dynamic changes. Such changes lead to many unique features in blood cell function and immune responses in early childhood. The blood cells and immune cells in neonates are a mixture of fetal and adult origin due to the co-existence of both fetal and adult types of hematopoietic stem cells (HSCs) and progenitor cells (HPCs). Fetal blood and immune cells gradually diminish during maturation of the infant and are almost completely replaced by adult types of cells by 3 to 4 weeks after birth in mice. Such features in early childhood are associated with unique features of hematopoietic and immune diseases, such as leukemia, at these developmental stages. Therefore, understanding the cellular and molecular mechanisms by which hematopoietic and immune changes occur throughout ontogeny will provide useful information for the study and treatment of pediatric blood and immune diseases. In this review, we summarize the most recent studies on hematopoietic initiation during early embryonic development, the expansion of both fetal and adult types of HSCs and HPCs in the fetal liver and fetal bone marrow stages, and the shift from fetal to adult hematopoiesis/immunity during neonatal/infant development. We also discuss the contributions of fetal types of HSCs/HPCs to childhood leukemias.
Collapse
Affiliation(s)
- Ryan Mack
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Lei Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Peter Breslin Sj
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA.,Departments of Molecular/Cellular Physiology and Biology, Loyola University Medical Center and Loyola University Chicago, Chicago, IL, 60660, USA
| | - Jiwang Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA.
| |
Collapse
|
7
|
Abstract
Children show a higher incidence of leukaemia compared with young adolescents, yet their cells are less damaged because of their young age. Children with Down syndrome (DS) have an even higher risk of developing leukaemia during the first years of life. The presence of a constitutive trisomy of chromosome 21 (T21) in DS acts as a genetic driver for leukaemia development, however, additional oncogenic mutations are required. Therefore, T21 provides the opportunity to better understand leukaemogenesis in children. Here, we describe the increased risk of leukaemia in DS during childhood from a somatic evolutionary view. According to this idea, cancer is caused by a variation in inheritable phenotypes within cell populations that are subjected to selective forces within the tissue context. We propose a model in which the increased risk of leukaemia in DS children derives from higher rates of mutation accumulation, already present during fetal development, which is further enhanced by changes in selection dynamics within the fetal liver niche. This model could possibly be used to understand the rate-limiting steps of leukaemogenesis early in life.
Collapse
|
8
|
Pirola CJ, Salatino A, Sookoian S. Pleiotropy within gene variants associated with nonalcoholic fatty liver disease and traits of the hematopoietic system. World J Gastroenterol 2021; 27:305-320. [PMID: 33584064 PMCID: PMC7852588 DOI: 10.3748/wjg.v27.i4.305] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/19/2020] [Accepted: 12/28/2020] [Indexed: 02/06/2023] Open
Abstract
Genome-wide association studies of complex diseases, including nonalcoholic fatty liver disease (NAFLD), have demonstrated that a large number of variants are implicated in the susceptibility of multiple traits — a phenomenon known as pleiotropy that is increasingly being explored through phenome-wide association studies. We focused on the analysis of pleiotropy within variants associated with hematologic traits and NAFLD. We used information retrieved from large public National Health and Nutrition Examination Surveys, Genome-wide association studies, and phenome-wide association studies based on the general population and explored whether variants associated with NAFLD also present associations with blood cell-related traits. Next, we applied systems biology approaches to assess the potential biological connection/s between genes that predispose affected individuals to NAFLD and nonalcoholic steatohepatitis, and genes that modulate hematological-related traits—specifically platelet count. We reasoned that this analysis would allow the identification of potential molecular mediators that link NAFLD with platelets. Genes associated with platelet count are most highly expressed in the liver, followed by the pancreas, heart, and muscle. Conversely, genes associated with NAFLD presented high expression levels in the brain, lung, spleen, and colon. Functional mapping, gene prioritization, and functional analysis of the most significant loci (P < 1 × 10-8) revealed that loci involved in the genetic modulation of platelet count presented significant enrichment in metabolic and energy balance pathways. In conclusion, variants in genes influencing NAFLD exhibit pleiotropic associations with hematologic traits, particularly platelet count. Likewise, significant enrichment of related genes with variants influencing platelet traits was noted in metabolic-related pathways. Hence, this approach yields novel mechanistic insights into NAFLD pathogenesis.
Collapse
Affiliation(s)
- Carlos Jose Pirola
- Department of Molecular Genetics and Biology of Complex Diseases, National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Institute of Medical Research (IDIM), Ciudad Autónoma de Buenos Aires C1427ARO, Argentina
- Institute of Medical Research A Lanari, University of Buenos Aires, School of Medicine, Ciudad Autónoma de Buenos Aires, Ciudad Autónoma de Buenos Aires C1427ARO, Argentina
| | - Adrian Salatino
- Department of Molecular Genetics and Biology of Complex Diseases, National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Institute of Medical Research (IDIM), Ciudad Autónoma de Buenos Aires C1427ARO, Argentina
- Institute of Medical Research A Lanari, University of Buenos Aires, School of Medicine, Ciudad Autónoma de Buenos Aires, Ciudad Autónoma de Buenos Aires C1427ARO, Argentina
| | - Silvia Sookoian
- Institute of Medical Research A Lanari, University of Buenos Aires, School of Medicine, Ciudad Autónoma de Buenos Aires, Ciudad Autónoma de Buenos Aires C1427ARO, Argentina
- Department of Clinical and Molecular Hepatology, National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Institute of Medical Research (IDIM), Ciudad Autónoma de Buenos Aires C1427ARO, Argentina
| |
Collapse
|
9
|
Boscher J, Guinard I, Eckly A, Lanza F, Léon C. Blood platelet formation at a glance. J Cell Sci 2020; 133:133/20/jcs244731. [DOI: 10.1242/jcs.244731] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
ABSTRACT
The main function of blood platelets is to ensure hemostasis and prevent hemorrhages. The 1011 platelets needed daily are produced in a well-orchestrated process. However, this process is not yet fully understood and in vitro platelet production is still inefficient. Platelets are produced in the bone marrow by megakaryocytes, highly specialized precursor cells that extend cytoplasmic projections called proplatelets (PPTs) through the endothelial barrier of sinusoid vessels. In this Cell Science at a Glance article and the accompanying poster we discuss the mechanisms and pathways involved in megakaryopoiesis and platelet formation processes. We especially address the – still underestimated – role of the microenvironment of the bone marrow, and present recent findings on how PPT extension in vivo differs from that in vitro and entails different mechanisms. Finally, we recapitulate old but recently revisited evidence that – although bone marrow does produce megakaryocytes and PPTs – remodeling and the release of bona fide platelets, mainly occur in the downstream microcirculation.
Collapse
Affiliation(s)
- Julie Boscher
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, F-67000 Strasbourg, France
| | - Ines Guinard
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, F-67000 Strasbourg, France
| | - Anita Eckly
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, F-67000 Strasbourg, France
| | - François Lanza
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, F-67000 Strasbourg, France
| | - Catherine Léon
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, F-67000 Strasbourg, France
| |
Collapse
|
10
|
Lucotti S, Muschel RJ. Platelets and Metastasis: New Implications of an Old Interplay. Front Oncol 2020; 10:1350. [PMID: 33042789 PMCID: PMC7530207 DOI: 10.3389/fonc.2020.01350] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/26/2020] [Indexed: 12/17/2022] Open
Abstract
During the process of hematogenous metastasis, tumor cells interact with platelets and their precursors megakaryocytes, providing a selection driver for the metastatic phenotype. Cancer cells have evolved a plethora of mechanisms to engage platelet activation and aggregation. Platelet coating of tumor cells in the blood stream promotes the successful completion of multiple steps of the metastatic cascade. Along the same lines, clinical evidence suggests that anti-coagulant therapy might be associated with reduced risk of metastatic disease and better prognosis in cancer patients. Here, we review experimental and clinical literature concerning the contribution of platelets and megakaryocytes to cancer metastasis and provide insights into the clinical relevance of anti-coagulant therapy in cancer treatment.
Collapse
Affiliation(s)
- Serena Lucotti
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
| | - Ruth J Muschel
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
11
|
Soares-da-Silva F, Peixoto M, Cumano A, Pinto-do-Ó P. Crosstalk Between the Hepatic and Hematopoietic Systems During Embryonic Development. Front Cell Dev Biol 2020; 8:612. [PMID: 32793589 PMCID: PMC7387668 DOI: 10.3389/fcell.2020.00612] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/19/2020] [Indexed: 12/14/2022] Open
Abstract
Hematopoietic stem cells (HSCs) generated during embryonic development are able to maintain hematopoiesis for the lifetime, producing all mature blood lineages. HSC transplantation is a widely used cell therapy intervention in the treatment of hematologic, autoimmune and genetic disorders. Its use, however, is hampered by the inability to expand HSCs ex vivo, urging for a better understanding of the mechanisms regulating their physiological expansion. In the adult, HSCs reside in the bone marrow, in specific microenvironments that support stem cell maintenance and differentiation. Conversely, while developing, HSCs are transiently present in the fetal liver, the major hematopoietic site in the embryo, where they expand. Deeper insights on the dynamics of fetal liver composition along development, and on how these different cell types impact hematopoiesis, are needed. Both, the hematopoietic and hepatic fetal systems have been extensively studied, albeit independently. This review aims to explore their concurrent establishment and evaluate to what degree they may cross modulate their respective development. As insights on the molecular networks that govern physiological HSC expansion accumulate, it is foreseeable that strategies to enhance HSC proliferation will be improved.
Collapse
Affiliation(s)
- Francisca Soares-da-Silva
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- Lymphocytes and Immunity Unit, Immunology Department, Pasteur Institute, Paris, France
- INSERM U1223, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Márcia Peixoto
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- Lymphocytes and Immunity Unit, Immunology Department, Pasteur Institute, Paris, France
- INSERM U1223, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Ana Cumano
- Lymphocytes and Immunity Unit, Immunology Department, Pasteur Institute, Paris, France
- INSERM U1223, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Perpetua Pinto-do-Ó
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
12
|
Effects of Delta-Like Noncanonical Notch Ligand 1 Expression of Human Fetal Liver Hepatoblasts on Hematopoietic Progenitors. Stem Cells Int 2019; 2019:7916275. [PMID: 31011334 PMCID: PMC6442310 DOI: 10.1155/2019/7916275] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/14/2018] [Accepted: 01/09/2019] [Indexed: 02/01/2023] Open
Abstract
Although the hepatic and hematopoietic progenitors of the liver are well characterized, the interactions between these two lineages remain mostly elusive. Hepatoblasts express delta-like noncanonical Notch ligand 1 (Dlk1), whose cleaved extracellular domain can become a soluble protein. We assessed the effects of DLK1 gene expression knockdown in cultures of total fetal liver cells. Furthermore, we separated Dlk1+ hepatoblasts from the total liver cell fraction and investigated effects of direct cell contact. Dlk1− cells were cultured either without Dlk1+ hepatoblasts, in direct contact with hepatoblasts, or separated from hepatoblasts by a porous membrane in inserts to inhibit cell contact but allow free exchange of molecules. Expression of the hepatic and hematopoietic genes, colony forming unit potential of various hematopoietic progenitors, and cell numbers and types were investigated. We found that DLK1 knockdown in total fetal liver cell cultures decreased total cell numbers. The expression of hepatic progenitor genes and mature hematopoietic genes was affected. Hematopoietic BFU-E and CFU-GM colony numbers were reduced significantly. The depletion of Dlk1+ hepatoblasts in culture decreased the potential of all hematopoietic progenitors to form colonies of all types and reduced the percentage of mature hematopoietic cells. The addition of hepatoblasts in inserts to Dlk1− cells further decreased the potential to form the CFU-GM and CFU-GEMM colonies and the percentage of mature hematopoietic cells but increased total cell numbers. Conclusively, direct contact of Dlk1 supports hematopoietic progenitor expansion and functionality that cannot be reconstituted in coculture without direct cell contact.
Collapse
|
13
|
Schmelzer E. Hepatic progenitors of the fetal liver: Interactions with hematopoietic stem cells. Differentiation 2019; 106:9-14. [PMID: 30826473 DOI: 10.1016/j.diff.2019.02.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 12/29/2022]
Abstract
The aim of this review is to summarize and give an overview on the findings of signaling between hepatic and hematopoietic progenitors of the liver. To date, there are not many findings published in the field, and the aim of this review is to cover all current publications in this area. The liver is the main site of hematopoiesis during fetal development. However, little is known about how hepatic and other non-hematopoietic progenitors potentially influence hematopoiesis and vice versa. The concurrent peaks of hepatic and hematopoietic progenitor proliferation during development indicate interactions that could possibly be mediated through cell-cell contact, extracellular matrices, cytokines and growth factors, or other signaling molecules. For example, hepatic progenitors, such as hepatic stem cells and hepatoblasts, possess characteristic surface markers that can be cleaved, giving rise to fragments of various lengths. A surface molecule of hepatoblasts has been demonstrated to play an essential role in hematopoiesis. Particularly, these effects on hematopoiesis were distinct, depending on whether it was membrane-bound or cleaved. In this review, the various hepatic and hematopoietic progenitor cell types are concisely described, and the current findings of their potential interactions are summarized.
Collapse
Affiliation(s)
- Eva Schmelzer
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, 3025 East Carson Street, Pittsburgh, PA 15203, USA.
| |
Collapse
|
14
|
Analysis of Hematopoietic Niche in the Mouse Embryo. Methods Mol Biol 2019. [PMID: 30671734 DOI: 10.1007/7651_2018_176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The development, differentiation, and maturation of hematopoietic cells are regulated by the intrinsic and extrinsic regulation. Intrinsic activity is affected by cell autonomous gene expression and extrinsic factors originate from the so-called niche surrounding the hematopoietic cells. It remains unclear why the hematopoietic sites are shifted during embryogenesis. Flow cytometry and immunohistochemistry enable us to study embryonic regulation of hematopoietic niche in the mouse embryo.
Collapse
|