1
|
Ricci F, Di Credico A, Gaggi G, Iannetti G, Ghinassi B, Gallina S, Olshansky B, Di Baldassarre A. Metoprolol disrupts inflammatory response of human cardiomyocytes via β-arrestin2 biased agonism and NF-κB signaling modulation. Biomed Pharmacother 2023; 168:115804. [PMID: 39491416 DOI: 10.1016/j.biopha.2023.115804] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/14/2023] [Accepted: 10/26/2023] [Indexed: 11/05/2024] Open
Abstract
AIMS Recent evidence supports non-class cardioprotective effects of metoprolol against neutrophil-mediated ischemia-reperfusion injury during exacerbated inflammation. Whether metoprolol exerts direct anti-inflammatory effect on cardiomyocytes is unknown. Accordingly, we aimed to investigate the direct anti-inflammatory effects of metoprolol in a cellular model of human induced pluripotent stem cell-derived cardiomyocytes (hiCMs) and to explore the role of β-arrestin2 (β-ARR2) biased agonism signaling pathway. METHODS AND RESULTS hiCMs were treated with TNF-α for 24 h, followed by 4-hour treatment with metoprolol or esmolol. Electrical response of hiCMs to β1-selective blockade was assessed by microelectrode arrays technology. The effect on inflammatory and adhesion molecule expression was evaluated in wild-type and β-ARR2 silenced hiCMs. To silence β-ARR2 expression, hiCMs were transfected with a specific small interfering RNA targeting β-ARR2 mRNA and preventing its translation. TNF-α stimulation boosted the expression of IκB, NF-κB, IL1β, IL6, and VCAM1 in hiCMs. TNF-α-treated hiCMs showed similar physiological responses to metoprolol and esmolol, with no difference in field potential duration and beat period recorded. Adding metoprolol significantly decreased inflammatory response patterns in wild-type hiCMs by dampening TNF-α induced expression of NF-κB, IL1β, and IL6, but not in β-ARR2-knockout hiCMs. A similar response was not observed in presence of β1-selective blockade with esmolol. CONCLUSIONS Metoprolol exerts a non-class direct anti-inflammatory effect on hi-CMs. β1-selective blockade with metoprolol disrupts inflammatory responses induced by TNF-α and induces significant inhibition of NF-κB signaling cascade via β-ARR2 biased agonism. If confirmed at clinical level, metoprolol could be tested and repurposed to treat cardiac inflammatory disorders.
Collapse
Affiliation(s)
- Fabrizio Ricci
- Department of Neuroscience, Imaging and Clinical Sciences, G.d'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy and University Cardiology Division, Heart Department, SS Annunziata University Hospital, Chieti, Italy; Department of Clinical Sciences, Lund University, 214 28 Malmö, Sweden
| | - Andrea Di Credico
- Department of Medicine and Aging Sciences, and Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| | - Giulia Gaggi
- Department of Medicine and Aging Sciences, and Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| | - Giovanni Iannetti
- Department of Neuroscience, Imaging and Clinical Sciences, G.d'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy and University Cardiology Division, Heart Department, SS Annunziata University Hospital, Chieti, Italy
| | - Barbara Ghinassi
- Department of Medicine and Aging Sciences, and Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| | - Sabina Gallina
- Department of Neuroscience, Imaging and Clinical Sciences, G.d'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy and University Cardiology Division, Heart Department, SS Annunziata University Hospital, Chieti, Italy
| | | | - Angela Di Baldassarre
- Department of Medicine and Aging Sciences, and Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), G. D'Annunzio University of Chieti-Pescara, 66100 Chieti, Italy.
| |
Collapse
|
2
|
Di Credico A, Gaggi G, Bucci I, Ghinassi B, Di Baldassarre A. The Effects of Combined Exposure to Bisphenols and Perfluoroalkyls on Human Perinatal Stem Cells and the Potential Implications for Health Outcomes. Int J Mol Sci 2023; 24:15018. [PMID: 37834465 PMCID: PMC10573528 DOI: 10.3390/ijms241915018] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023] Open
Abstract
The present study investigates the impact of two endocrine disruptors, namely Bisphenols (BPs) and Perfluoroalkyls (PFs), on human stem cells. These chemicals leach from plastic, and when ingested through contaminated food and water, they interfere with endogenous hormone signaling, causing various diseases. While the ability of BPs and PFs to cross the placental barrier and accumulate in fetal serum has been documented, the exact consequences for human development require further elucidation. The present research work explored the effects of combined exposure to BPs (BPA or BPS) and PFs (PFOS and PFOA) on human placenta (fetal membrane mesenchymal stromal cells, hFM-MSCs) and amniotic fluid (hAFSCs)-derived stem cells. The effects of the xenobiotics were assessed by analyzing cell proliferation, mitochondrial functionality, and the expression of genes involved in pluripotency and epigenetic regulation, which are crucial for early human development. Our findings demonstrate that antenatal exposure to BPs and/or PFs may alter the biological characteristics of perinatal stem cells and fetal epigenome, with potential implications for health outcomes at birth and in adulthood. Further research is necessary to comprehend the full extent of these effects and their long-term consequences.
Collapse
Affiliation(s)
- Andrea Di Credico
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy; (A.D.C.); (I.B.); (B.G.); (A.D.B.)
- Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- UdA TechLab Center (UdATech), 66100 Chieti, Italy
| | - Giulia Gaggi
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy; (A.D.C.); (I.B.); (B.G.); (A.D.B.)
- Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- UdA TechLab Center (UdATech), 66100 Chieti, Italy
| | - Ines Bucci
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy; (A.D.C.); (I.B.); (B.G.); (A.D.B.)
- Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Barbara Ghinassi
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy; (A.D.C.); (I.B.); (B.G.); (A.D.B.)
- Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- UdA TechLab Center (UdATech), 66100 Chieti, Italy
| | - Angela Di Baldassarre
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy; (A.D.C.); (I.B.); (B.G.); (A.D.B.)
- Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- UdA TechLab Center (UdATech), 66100 Chieti, Italy
| |
Collapse
|
3
|
Gaggi G, Di Credico A, Barbagallo F, Ballerini P, Ghinassi B, Di Baldassarre A. Antenatal Exposure to Plastic Pollutants: Study of the Bisphenols and Perfluoroalkyls Effects on Human Stem Cell Models. EXPOSURE AND HEALTH 2023. [DOI: 10.1007/s12403-023-00586-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/07/2023] [Accepted: 07/05/2023] [Indexed: 09/02/2023]
Abstract
AbstractEndocrine disruptors (EDs), such as Bisphenols (BPs) and Perfluoroalkyls (PFs), are a class of plastic pollutants widely used in industrial applications. Human exposure to these molecules usually occurs through ingestion of contaminated food and water. Once entered the human body they can interfere with endogenous hormone signaling, leading to a wide spectrum of diseases. It has been reported that BPs and PFs can cross the placental barrier accumulating in the fetal serum, but the detrimental consequences for human development remain to be clarified. Here we analyze the effects of different doses of bisphenol A and S (BPA, BPS) perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) on proliferation and mitochondrial health on different types of stem cells: through an integrated approach that combines data from pluripotent stem cells (hiPSCs) with that from the “environment” in which the embryo develops (fetal annexes-derived perinatal stem cells) we verified the potential developmental toxicity of the in utero EDs exposure. Data obtained showed that overall, BPs, and PFs tended to increase the proliferation rate of perinatal stem cells; a similar response was observed in hiPSCs exposed to very low doses of BPs and PFs, while at higher concentrations these chemicals were toxic; in addition, both the BPs and the PFs exerted a mitotoxic effects hiPSCs at all the concentration studied. All these data suggest that antenatal exposure to BPs and PFs, also at very low concentrations, may modify the biological characteristics of stem cells present in both the developing fetus and the fetal annexes, thus perturbing normal human development.
Collapse
|
4
|
Liu Q, Luo L, Ren C, Zou M, Yang S, Cai B, Wu L, Wang Y, Fu S, Hua X, Tang N, Huang S, Huang X, Xin W, Chen F, Zhang X. The opposing roles of the mTOR signaling pathway in different phases of human umbilical cord blood-derived CD34 + cell erythropoiesis. Stem Cells 2020; 38:1492-1505. [PMID: 32871057 PMCID: PMC7693065 DOI: 10.1002/stem.3268] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 06/25/2020] [Accepted: 07/10/2020] [Indexed: 02/05/2023]
Abstract
As an indispensable, even lifesaving practice, red blood cell (RBC) transfusion is challenging due to several issues, including supply shortage, immune incompatibility, and blood-borne infections since donated blood is the only source of RBCs. Although large-scale in vitro production of functional RBCs from human stem cells is a promising alternative, so far, no such system has been reported to produce clinically transfusable RBCs due to the poor understanding of mechanisms of human erythropoiesis, which is essential for the optimization of in vitro erythrocyte generation system. We previously reported that inhibition of mammalian target of rapamycin (mTOR) signaling significantly decreased the percentage of erythroid progenitor cells in the bone marrow of wild-type mice. In contrast, rapamycin treatment remarkably improved terminal maturation of erythroblasts and anemia in a mouse model of β-thalassemia. In the present study, we investigated the effect of mTOR inhibition with rapamycin from different time points on human umbilical cord blood-derived CD34+ cell erythropoiesis in vitro and the underlying mechanisms. Our data showed that rapamycin treatment significantly suppressed erythroid colony formation in the commitment/proliferation phase of erythropoiesis through inhibition of cell-cycle progression and proliferation. In contrast, during the maturation phase of erythropoiesis, mTOR inhibition dramatically promoted enucleation and mitochondrial clearance by enhancing autophagy. Collectively, our results suggest contrasting roles for mTOR in regulating different phases of human erythropoiesis.
Collapse
Affiliation(s)
- Qian Liu
- Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
- Shantou University Medical CollegeShantouPeople's Republic of China
| | - Linhong Luo
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Chunhong Ren
- Department of International Medical ServiceThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Muping Zou
- Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Siqin Yang
- Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Bozhi Cai
- Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Libiao Wu
- Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Yunsheng Wang
- Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Shan Fu
- Shantou University Medical CollegeShantouPeople's Republic of China
| | - Xu Hua
- Shantou University Medical CollegeShantouPeople's Republic of China
| | - Nianping Tang
- Shantou University Medical CollegeShantouPeople's Republic of China
| | - Shiping Huang
- Shantou University Medical CollegeShantouPeople's Republic of China
| | - Xianxi Huang
- Intensive Care Unit, The First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Wen Xin
- Beijing TransGen Biotech Co., Ltd.BeijingPeople's Republic of China
| | - Feiheng Chen
- Department of HematologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Xin Zhang
- Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
- Shantou University Medical CollegeShantouPeople's Republic of China
- Department of CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
- Laboratory of Medical Molecular ImagingThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| |
Collapse
|
5
|
Federici G, Varricchio L, Martelli F, Falchi M, Picconi O, Francescangeli F, Contavalli P, Girelli G, Tafuri A, Petricoin EF, Mazzarini M, Zeuner A, Migliaccio AR. Phosphoproteomic Landscaping Identifies Non-canonical cKIT Signaling in Polycythemia Vera Erythroid Progenitors. Front Oncol 2019; 9:1245. [PMID: 31824842 PMCID: PMC6883719 DOI: 10.3389/fonc.2019.01245] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/29/2019] [Indexed: 01/08/2023] Open
Abstract
Although stem cell factor (SCF)/cKIT interaction plays key functions in erythropoiesis, cKIT signaling in human erythroid cells is still poorly defined. To provide new insights into cKIT-mediated erythroid expansion in development and disease, we performed phosphoproteomic profiling of primary erythroid progenitors from adult blood (AB), cord blood (CB), and Polycythemia Vera (PV) at steady-state and upon SCF stimulation. While AB and CB, respectively, activated transient or sustained canonical cKIT-signaling, PV showed a non-canonical signaling including increased mTOR and ERK1 and decreased DEPTOR. Accordingly, screening of FDA-approved compounds showed increased PV sensitivity to JAK, cKIT, and MEK inhibitors. Moreover, differently from AB and CB, in PV the mature 145kDa-cKIT constitutively associated with the tetraspanin CD63 and was not endocytosed upon SCF stimulation, contributing to unrestrained cKIT signaling. These results identify a clinically exploitable variegation of cKIT signaling/metabolism that may contribute to the great erythroid output occurring during development and in PV.
Collapse
Affiliation(s)
| | - Lilian Varricchio
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Fabrizio Martelli
- National Center for Preclinical and Clinical Research and Evaluation of Pharmaceutical Drugs, Istituto Superiore di Sanità, Rome, Italy
| | - Mario Falchi
- National HIV/AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Orietta Picconi
- National HIV/AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | | | - Paola Contavalli
- Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Gabriella Girelli
- Immunohematology and Transfusion Medicine Unit, "La Sapienza" University of Rome, Rome, Italy
| | - Agostino Tafuri
- Sant'Andrea Hospital-La Sapienza, Department of Clinic and Molecular Medicine "La Sapienza" University of Rome, Rome, Italy
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, United States
| | - Maria Mazzarini
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, Bologna, Italy
| | - Ann Zeuner
- Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Anna Rita Migliaccio
- Myeloproliferative Neoplasm Research Consortium, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
6
|
|
7
|
Uchida N, Washington KN, Mozer B, Platner C, Ballantine J, Skala LP, Raines L, Shvygin A, Hsieh MM, Mitchell LG, Tisdale JF. RNA Trans-Splicing Targeting Endogenous β-Globin Pre-Messenger RNA in Human Erythroid Cells. Hum Gene Ther Methods 2017; 28:91-99. [PMID: 28267358 DOI: 10.1089/hgtb.2016.077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Sickle cell disease results from a point mutation in exon 1 of the β-globin gene (total 3 exons). Replacing sickle β-globin exon 1 (and exon 2) with a normal sequence by trans-splicing is a potential therapeutic strategy. Therefore, this study sought to develop trans-splicing targeting β-globin pre-messenger RNA among human erythroid cells. Binding domains from random β-globin sequences were comprehensively screened. Six candidates had optimal binding, and all targeted intron 2. Next, lentiviral vectors encoding RNA trans-splicing molecules were constructed incorporating a unique binding domain from these candidates, artificial 5' splice site, and γ-globin cDNA, and trans-splicing was evaluated in CD34+ cell-derived erythroid cells from healthy individuals. Lentiviral transduction was efficient, with vector copy numbers of 9.7 to 15.3. The intended trans-spliced RNA product, including exon 3 of endogenous β-globin and γ-globin, was detected at the molecular level. Trans-splicing efficiency was improved to 0.07-0.09% by longer binding domains, including the 5' splice site of intron 2. In summary, screening was performed to select efficient binding domains for trans-splicing. Detectable levels of trans-splicing were obtained for endogenous β-globin RNA in human erythroid cells. These methods provide the basis for future trans-splicing directed gene therapy.
Collapse
Affiliation(s)
- Naoya Uchida
- 1 Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH) , Bethesda, Maryland
| | | | - Brian Mozer
- 3 Office of Research Integrity , Office of the Assistant Secretary for Health, Rockville, Maryland
| | - Charlotte Platner
- 1 Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH) , Bethesda, Maryland
| | - Josiah Ballantine
- 1 Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH) , Bethesda, Maryland
| | - Luke P Skala
- 1 Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH) , Bethesda, Maryland
| | - Lydia Raines
- 1 Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH) , Bethesda, Maryland
| | - Anna Shvygin
- 1 Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH) , Bethesda, Maryland
| | - Matthew M Hsieh
- 1 Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH) , Bethesda, Maryland
| | | | - John F Tisdale
- 1 Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH) , Bethesda, Maryland
| |
Collapse
|
8
|
Uchida N, Haro-Mora JJ, Fujita A, Lee DY, Winkler T, Hsieh MM, Tisdale JF. Efficient Generation of β-Globin-Expressing Erythroid Cells Using Stromal Cell-Derived Induced Pluripotent Stem Cells from Patients with Sickle Cell Disease. Stem Cells 2016; 35:586-596. [PMID: 27739611 DOI: 10.1002/stem.2517] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 09/23/2016] [Accepted: 09/28/2016] [Indexed: 12/15/2022]
Abstract
Human embryonic stem (ES) cells and induced pluripotent stem (iPS) cells represent an ideal source for in vitro modeling of erythropoiesis and a potential alternative source for red blood cell transfusions. However, iPS cell-derived erythroid cells predominantly produce ε- and γ-globin without β-globin production. We recently demonstrated that ES cell-derived sacs (ES sacs), known to express hemangioblast markers, allow for efficient erythroid cell generation with β-globin production. In this study, we generated several iPS cell lines derived from bone marrow stromal cells (MSCs) and peripheral blood erythroid progenitors (EPs) from sickle cell disease patients, and evaluated hematopoietic stem/progenitor cell (HSPC) generation after iPS sac induction as well as subsequent erythroid differentiation. MSC-derived iPS sacs yielded greater amounts of immature hematopoietic progenitors (VEGFR2 + GPA-), definitive HSPCs (CD34 + CD45+), and megakaryoerythroid progenitors (GPA + CD41a+), as compared to EP-derived iPS sacs. Erythroid differentiation from MSC-derived iPS sacs resulted in greater amounts of erythroid cells (GPA+) and higher β-globin (and βS-globin) expression, comparable to ES sac-derived cells. These data demonstrate that human MSC-derived iPS sacs allow for more efficient erythroid cell generation with higher β-globin production, likely due to heightened emergence of immature progenitors. Our findings should be important for iPS cell-derived erythroid cell generation. Stem Cells 2017;35:586-596.
Collapse
Affiliation(s)
- Naoya Uchida
- Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Juan J Haro-Mora
- Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Atsushi Fujita
- Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Duck-Yeon Lee
- Biochemistry Core, NHLBI, NIH, Bethesda, Maryland, USA
| | | | - Matthew M Hsieh
- Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - John F Tisdale
- Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
9
|
D'Amico MA, Ghinassi B, Izzicupo P, Di Ruscio A, Di Baldassarre A. IL-6 Activates PI3K and PKCζ Signaling and Determines Cardiac Differentiation in Rat Embryonic H9c2 Cells. J Cell Physiol 2016. [PMID: 26205888 DOI: 10.1002/jcp.25101] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION IL-6 influences several biological processes, including cardiac stem cell and cardiomyocyte physiology. Although JAK-STAT3 activation is the defining feature of IL-6 signaling, signaling molecules such as PI3K, PKCs, and ERK1/2 are also activated and elicit different responses. Moreover, most studies on the specific role of these signaling molecules focus on the adult heart, and few studies are available on the biological effects evoked by IL-6 in embryonic cardiomyocytes. AIM The aim of this study was to clarify the biological response of embryonic heart derived cells to IL-6 by analyzing the morphological modifications and the signaling cascades evoked by the cytokine in H9c2 cells. RESULTS IL-6 stimulation determined the terminal differentiation of H9c2 cells, as evidenced by the increased expression of cardiac transcription factors (NKX2.5 and GATA4), structural proteins (α-myosin heavy chain and cardiac Troponin T) and the gap junction protein Connexin 43. This process was mediated by the rapid modulation of PI3K, Akt, PTEN, and PKCζ phosphorylation levels. PI3K recruitment was an upstream event in the signaling cascade and when PI3K was inhibited, IL-6 failed to modify PKCζ, PTEN, and Akt phosphorylation. Blocking PKCζ activity affected only PTEN and Akt. Finally, the overexpression of a constitutively active form of PKCζ in H9c2 cells largely mimicked the morphological and molecular effects evoked by IL-6. CONCLUSIONS This study demonstrated that IL-6 induces the cardiac differentiation of H9c2 embryonic cells though a signaling cascade that involves PI3K, PTEN, and PKCζ activities.
Collapse
Affiliation(s)
- Maria Angela D'Amico
- Department of Medicine and Aging Sciences, Section of Human Morphology, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Barbara Ghinassi
- Department of Medicine and Aging Sciences, Section of Human Morphology, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Pascal Izzicupo
- Department of Medicine and Aging Sciences, Section of Human Morphology, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Annalisa Di Ruscio
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Angela Di Baldassarre
- Department of Medicine and Aging Sciences, Section of Human Morphology, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
10
|
Fujita A, Uchida N, Haro-Mora JJ, Winkler T, Tisdale J. β-Globin-Expressing Definitive Erythroid Progenitor Cells Generated from Embryonic and Induced Pluripotent Stem Cell-Derived Sacs. Stem Cells 2016; 34:1541-52. [PMID: 26866725 DOI: 10.1002/stem.2335] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 01/19/2016] [Indexed: 01/10/2023]
Abstract
Human embryonic stem (ES) cells and induced pluripotent stem (iPS) cells represent a potential alternative source for red blood cell transfusion. However, when using traditional methods with embryoid bodies, ES cell-derived erythroid cells predominantly express embryonic type ɛ-globin, with lesser fetal type γ-globin and very little adult type β-globin. Furthermore, no β-globin expression is detected in iPS cell-derived erythroid cells. ES cell-derived sacs (ES sacs) have been recently used to generate functional platelets. Due to its unique structure, we hypothesized that ES sacs serve as hemangioblast-like progenitors capable to generate definitive erythroid cells that express β-globin. With our ES sac-derived erythroid differentiation protocol, we obtained ∼120 erythroid cells per single ES cell. Both primitive (ɛ-globin expressing) and definitive (γ- and β-globin expressing) erythroid cells were generated from not only ES cells but also iPS cells. Primitive erythropoiesis is gradually switched to definitive erythropoiesis during prolonged ES sac maturation, concurrent with the emergence of hematopoietic progenitor cells. Primitive and definitive erythroid progenitor cells were selected on the basis of glycophorin A or CD34 expression from cells within the ES sacs before erythroid differentiation. This selection and differentiation strategy represents an important step toward the development of in vitro erythroid cell production systems from pluripotent stem cells. Further optimization to improve expansion should be required for clinical application. Stem Cells 2016;34:1541-1552.
Collapse
Affiliation(s)
- Atsushi Fujita
- Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Naoya Uchida
- Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Juan J Haro-Mora
- Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | | | - John Tisdale
- Molecular and Clinical Hematology Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
11
|
Liu YX, Dong X, Gong F, Su N, Li SB, Zhang HT, Liu JL, Xue JH, Ji SP, Zhang Z. Promotion of Erythropoietic Differentiation in Hematopoietic Stem Cells by SOCS3 Knock-Down. PLoS One 2015; 10:e0135259. [PMID: 26252772 PMCID: PMC4529111 DOI: 10.1371/journal.pone.0135259] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 07/20/2015] [Indexed: 01/08/2023] Open
Abstract
Suppressor of cytokine signaling 3 (SOCS3) plays an important role in mice fetal liver erythropoiesis, but the roles of SOCS3 in human hematopoietic stem cells (HSCs) have not been well investigated. In the present study, lentiviral small interference RNA expression vectors (shRNA) of SOCS3 were constructed and stably transferred into HSCs. We found that SOCS3 knockdown induced erythroid expansion in HSCs. Conversely, Ectopic expression of SOCS3 in progenitor cells blocked erythroid expansion and erythroid colony formation of HSCs. To further explore the involved mechanism, we compared gene expression profiles of SOCS3-shRNA tranduced HSCs with that of control HSCs by whole genome microarrays. The results indicated that cell developmental process related genes, especially hematopoietic lineage-specific genes, associated with the responses to SOCS3 in HSCs.Downexpression of SOCS3 in HSCs or differentiated erythroid progenitor cells induced a transcriptional program enriched for erythroid development relative genes. Our results proved that SOCS3 down-expression induced lineage commitment towards erythroid progenitor cell fate by activation of erythroid-specific gene in HSCs and provided new insight into the mechanism of erythropoietic development.
Collapse
Affiliation(s)
- Yu-xiao Liu
- First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
| | - Xing Dong
- Third Military Medical University, Chongqing, China
- General Hospital of Beijing Military Command, Beijing, China
| | - Feng Gong
- Beijing Institution of Transfusion Medicine, Beijing, China
| | - Ning Su
- First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
| | - Su-bo Li
- Beijing Institution of Transfusion Medicine, Beijing, China
| | - Hai-tao Zhang
- First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
| | - Jia-ling Liu
- First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
| | - Jing-hui Xue
- First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
- * E-mail: (JHX); (SPJ); (ZWZ)
| | - Shou-ping Ji
- Beijing Institution of Transfusion Medicine, Beijing, China
- * E-mail: (JHX); (SPJ); (ZWZ)
| | - Zhi–wen Zhang
- First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
- * E-mail: (JHX); (SPJ); (ZWZ)
| |
Collapse
|
12
|
Follo MY, Faenza I, Piazzi M, Blalock WL, Manzoli L, McCubrey JA, Cocco L. Nuclear PI-PLCβ1: an appraisal on targets and pathology. Adv Biol Regul 2013; 54:2-11. [PMID: 24296032 DOI: 10.1016/j.jbior.2013.11.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 11/08/2013] [Indexed: 11/16/2022]
Abstract
Lipid signalling molecules are essential components of the processes that allow one extracellular signal to be transferred inside the nucleus, where specific lipid second messengers elicit reactions capable of regulating gene transcription, DNA replication or repair and DNA cleavage, eventually resulting in cell growth, differentiation, apoptosis or many other cell functions. Nuclear inositides are independently regulated, suggesting that the nucleus constitutes a functionally distinct compartment of inositol lipids metabolism. Indeed, nuclear inositol lipids themselves can modulate nuclear processes, such as transcription and pre-mRNA splicing, growth, proliferation, cell cycle regulation and differentiation. Nuclear PI-PLCβ1 is a key molecule for nuclear inositide signalling, where it plays a role in cell cycle progression, proliferation and differentiation. Here we review the targets and possible involvement of nuclear PI-PLCβ1 in human physiology and pathology.
Collapse
Affiliation(s)
- Matilde Y Follo
- Department of Biomedical and Neuromotor Sciences, Cellular Signalling Laboratory, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy.
| | - Irene Faenza
- Department of Biomedical and Neuromotor Sciences, Cellular Signalling Laboratory, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Manuela Piazzi
- Department of Biomedical and Neuromotor Sciences, Cellular Signalling Laboratory, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - William L Blalock
- CNR - Consiglio Nazionale delle Ricerche, Istituto di Genetica Molecolare and SC Laboratorio di Biologia Cellulare Muscoloscheletrica, IOR, Bologna, Italy
| | - Lucia Manzoli
- Department of Biomedical and Neuromotor Sciences, Cellular Signalling Laboratory, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Cellular Signalling Laboratory, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy.
| |
Collapse
|
13
|
Yang YR, Follo MY, Cocco L, Suh PG. The physiological roles of primary phospholipase C. Adv Biol Regul 2013; 53:232-241. [PMID: 24041464 DOI: 10.1016/j.jbior.2013.08.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 08/10/2013] [Indexed: 06/02/2023]
Abstract
The roles of phosphoinositide-specific phospholipase C (PLC) have been extensively investigated in diverse cell lines and pathological conditions. Among the PLC isozmes, primary PLCs, PLC-β and PLC-γ, are directly activated by receptor activation, unlike other secondary PLCs (PLC-ɛ, PLC-δ1, and PLC-η1). PLC-β isozymes are activated by G protein couple receptor and PLC-γ isozymes are activated by receptor tyrosine kinase (RTK). Primary PLCs are differentially expressed in different tissues, suggesting their specific roles in diverse tissues and regulate a variety of physiological and pathophysiological functions. Thus, dysregulation of phospholipases contributes to a number of human diseases and primary PLCs have been identified as therapeutic targets for prevention and treatment of diseases. Here we review the roles of primary PLCs in physiology and their impact in pathology.
Collapse
Affiliation(s)
- Yong Ryoul Yang
- School of Nano-Biotechnology and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 689-798, Republic of Korea
| | | | | | | |
Collapse
|
14
|
Faenza I, Fiume R, Piazzi M, Colantoni A, Cocco L. Nuclear inositide specific phospholipase C signalling - interactions and activity. FEBS J 2013; 280:6311-21. [PMID: 23890371 DOI: 10.1111/febs.12450] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 06/26/2013] [Accepted: 07/18/2013] [Indexed: 01/07/2023]
Abstract
Evidence accumulated over the past 20 years has highlighted the presence of an autonomous nuclear inositol lipid metabolism, and suggests that lipid signalling molecules are important components of signalling pathways operating within the nucleus. Nuclear polyphosphoinositide (PI) signalling relies on the synthesis and metabolism of phosphatidylinositol 4,5-bisphosphate, which can modulate the activity of effector proteins and is a substrate of signalling enzymes. The regulation of the nuclear PI pool is totally independent from the plasma membrane counterpart, suggesting that the nucleus constitutes a functionally distinct compartment of inositol lipids metabolism. Among the nuclear enzymes involved in PI metabolism, inositide specific phospholipase C (PI-PLC) has been one of the most extensively studied. Several isoforms of PI-PLCs have been identified in the nucleus, namely PI-PLC-β1, γ1, δ1 and ζ; however, the β1 isozyme is the best characterized. In the present review, we focus on the signal transduction-related metabolism of nuclear PI-PLC and review the most convincing evidence for PI-PLC expression and activity being involved in differentiation and proliferation programmes in several cell systems. Moreover, nuclear PI-PLC is an intermediate effector and interactor for nuclear inositide signalling. The inositide cycle exists and shows a biological role inside the nucleus. It is an autonomous lipid-dependent signalling system, independently regulated with respect to the one at the plasma membrane counterpart, and is involved in cell cycle progression and differentiation.
Collapse
Affiliation(s)
- Irene Faenza
- Cell Signaling Laboratory, Department of Biomedical Science (DIBINEM), University of Bologna, Italy
| | | | | | | | | |
Collapse
|
15
|
Okonko DO, Marley SB, Anker SD, Poole-Wilson PA, Gordon MY. Suppression of erythropoiesis in patients with chronic heart failure and anaemia of unknown origin: evidence of an immune basis. Int J Cardiol 2013; 166:664-71. [DOI: 10.1016/j.ijcard.2011.11.081] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 11/24/2011] [Accepted: 11/26/2011] [Indexed: 12/17/2022]
|
16
|
In vitro large scale production of human mature red blood cells from hematopoietic stem cells by coculturing with human fetal liver stromal cells. BIOMED RESEARCH INTERNATIONAL 2013; 2013:807863. [PMID: 23484161 PMCID: PMC3581122 DOI: 10.1155/2013/807863] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 12/02/2012] [Indexed: 01/09/2023]
Abstract
In vitro models of human erythropoiesis are useful in studying the mechanisms of erythroid differentiation in normal and pathological conditions. Here we describe an erythroid liquid culture system starting from cord blood derived hematopoietic stem cells (HSCs). HSCs were cultured for more than 50 days in erythroid differentiation conditions and resulted in a more than 10(9)-fold expansion within 50 days under optimal conditions. Homogeneous erythroid cells were characterized by cell morphology, flow cytometry, and hematopoietic colony assays. Furthermore, terminal erythroid maturation was improved by cosculturing with human fetal liver stromal cells. Cocultured erythroid cells underwent multiple maturation events, including decrease in size, increase in glycophorin A expression, and nuclear condensation. This process resulted in extrusion of the pycnotic nuclei in up to 80% of the cells. Importantly, they possessed the capacity to express the adult definitive β -globin chain upon further maturation. We also show that the oxygen equilibrium curves of the cord blood-differentiated red blood cells (RBCs) are comparable to normal RBCs. The large number and purity of erythroid cells and RBCs produced from cord blood make this method useful for fundamental research in erythroid development, and they also provide a basis for future production of available RBCs for transfusion.
Collapse
|
17
|
Follo MY, Marmiroli S, Faenza I, Fiume R, Ramazzotti G, Martelli AM, Gobbi P, McCubrey JA, Finelli C, Manzoli FA, Cocco L. Nuclear phospholipase C β1 signaling, epigenetics and treatments in MDS. Adv Biol Regul 2012; 53:2-7. [PMID: 23058275 DOI: 10.1016/j.jbior.2012.09.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 09/13/2012] [Indexed: 11/19/2022]
Abstract
Myelodysplastic syndromes (MDS), clonal hematopoietic stem-cell disorders mainly affecting older adult patients, show ineffective hematopoiesis in one or more of the lineages of the bone marrow. Most MDS are characterized by anemia, and a number of cases progresses to acute myeloid leukemia (AML). Indeed, the molecular mechanisms underlying the MDS evolution to AML are still unclear, even though the nuclear signaling elicited by PI-PLCβ1 has been demonstrated to play an important role in the control of the balance between cell cycle progression and apoptosis in MDS cells. Here we review both the role of epigenetic therapy on PI-PLCβ1 promoter and the changes in PI-PLCβ1 expression in MDS patients treated for anemia.
Collapse
MESH Headings
- Apoptosis/drug effects
- Bone Marrow/drug effects
- Bone Marrow/metabolism
- Bone Marrow/pathology
- Cell Cycle/drug effects
- Cell Nucleus/drug effects
- Cell Nucleus/genetics
- Cell Nucleus/metabolism
- Epigenesis, Genetic/drug effects
- Erythropoietin/therapeutic use
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Myelodysplastic Syndromes/complications
- Myelodysplastic Syndromes/drug therapy
- Myelodysplastic Syndromes/genetics
- Myelodysplastic Syndromes/pathology
- Phosphatidylinositols/metabolism
- Phospholipase C beta/genetics
- Phospholipase C beta/metabolism
- Promoter Regions, Genetic
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Matilde Y Follo
- Cellular Signalling Laboratory, Department of Human Anatomical Sciences, University of Bologna, Bologna, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Activation of nuclear inositide signalling pathways during erythropoietin therapy in low-risk MDS patients. Leukemia 2012; 26:2474-82. [PMID: 22596089 DOI: 10.1038/leu.2012.133] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Inositide signaling pathways can have a role in the Myelodysplastic Syndromes (MDS) progression to acute myeloid leukemia. Erythropoietin (EPO) is currently used in low-risk MDS, where it successfully corrects anemia in 50-70% of patients. However, some MDS patients are refractory to this treatment and little is known about the exact molecular mechanisms underlying the effect of EPO in these subjects. Here, we investigated the role of inositide pathways in low-risk MDS treated with EPO, mainly focusing on the Akt/PI-PLC (Phosphoinositide-Phospholipase C) gamma1 axis, which is activated by the EPO receptor, and PI-PLCbeta1/Cyclin D3 signaling, as Cyclin D3 is associated with hematopoietic proliferation and differentiation. Interestingly, EPO responder patients showed a specific activation of both the Akt/PI-PLCgamma1 pathway and beta-Globin gene expression, while nonresponders displayed an increase in PI-PLCbeta1 signaling. Moreover, in normal CD34+ cells induced to erythroid differentiation, PI-PLCbeta1 overexpression abrogated both EPO-induced Akt phosphorylation and beta-Globin expression. Overall, these findings suggest that PI-PLCbeta1 can act as a negative regulator of erythroid differentiation and confirm the involvement of Akt/PI-PLCgamma1 pathway in EPO signaling, therefore contributing to the comprehension of the effect of EPO in low-risk MDS and possibly paving the way to the identification of MDS patients at higher risk of refractoriness to EPO treatment.
Collapse
|
19
|
Chronic IFN-γ production in mice induces anemia by reducing erythrocyte life span and inhibiting erythropoiesis through an IRF-1/PU.1 axis. Blood 2011; 118:2578-88. [DOI: 10.1182/blood-2010-10-315218] [Citation(s) in RCA: 136] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Abstract
Anemia of chronic disease is a complication accompanying many inflammatory diseases. The proinflammatory cytokine IFN-γ has been implicated in this form of anemia, but the underlying mechanism remains unclear. Here we describe a novel mouse model for anemia of chronic disease, in which enhanced CD27-mediated costimulation strongly increases the formation of IFN-γ–producing effector T cells, leading to a progressive anemia. We demonstrate that the anemia in these mice is fully dependent on IFN-γ and that this cytokine reduces both the life span and the formation of red blood cells. Molecular analysis revealed that IFN-γ induces expression of the transcription factors of interferon regulatory factor-1 (IRF-1) and PU.1 in both murine and human erythroid precursors. We found that, on IFN-γ stimulation, IRF-1 binds to the promoter of SPI.1 (PU.1) and induces PU.1 expression, leading to inhibition of erythropoiesis. Notably, down-regulation of either IRF-1 or PU.1 expression is sufficient to overcome IFN-γ–induced inhibition of erythropoiesis. These findings reveal a molecular mechanism by which chronic exposure to IFN-γ induces anemia.
Collapse
|
20
|
Migliaccio G, Sanchez M, Leblanc A, Masiello F, Tirelli V, Migliaccio AR, Najfeld V, Whitsett C. Long-term storage does not alter functionality of in vitro generated human erythroblasts: implications for ex vivo generated erythroid transfusion products. Transfusion 2009; 49:2668-79. [PMID: 19659677 DOI: 10.1111/j.1537-2995.2009.02329.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Cultured human erythroid cells derived in vitro may represent alternative transfusion products. It is unknown, however, if these ex vivo expanded erythroid cells remain functional or develop genetic abnormalities after storage. STUDY DESIGN AND METHODS Using mononuclear cells from four adult blood donors, erythroblasts were generated ex vivo in expansion cultures supplemented with stem cell factor, interleukin-3, erythropoietin (EPO), dexamethasone, and estradiol. The viability and in vitro function of freshly expanded or short (1-2 months)- and long (8 years)-term-stored erythroblasts cryopreserved in dimethyl sulfoxide were compared. Erythroblast function was defined as ability to proliferate in expansion media and mature in response to EPO. Cell number was determined manually and expressed as fold increase. Viability was assessed by trypan blue and propidium iodide exclusion. Maturation was evaluated by morphologic analyses and CD36/CD235a expression profiling. Cytogenetic evaluation included karyotype and multicolor fluorescence in situ hybridization analyses. RESULTS Equivalent numbers (>80%) of erythroblasts were viable after short- and long-term storage. Freshly expanded and short- and long-term-stored erythroblasts equally doubled in number (fold increase, 2.4) retaining an immature phenotype (23% of the cells were CD36(high)CD235a(neg)) when cultured for 4 days under expansion conditions. The numbers of freshly expanded and short-term-stored erythroblasts that matured when exposed for 4 days to EPO were also similar (approx. 22% of the cells became CD36(neg)CD235a(high)). In spite of the massive amplification, ex vivo generated erythroblasts demonstrated a normal (46,XY) karyotype with no obvious genomic rearrangements. CONCLUSION Ex vivo expanded erythroblasts remain functional and genetically normal after long-term storage.
Collapse
Affiliation(s)
- Giovanni Migliaccio
- Division of Hematology and Oncology, Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Follo MY, Finelli C, Clissa C, Mongiorgi S, Bosi C, Martinelli G, Baccarani M, Manzoli L, Martelli AM, Cocco L. Phosphoinositide-Phospholipase C β1 Mono-Allelic Deletion Is Associated With Myelodysplastic Syndromes Evolution Into Acute Myeloid Leukemia. J Clin Oncol 2009; 27:782-790. [DOI: 10.1200/jco.2008.19.3748] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
Purpose To evaluate the association between the presence of phosphoinositide-phospholipase C β1 (PI-PLCβ1) mono-allelic deletion with the clinical outcome of myelodysplastic syndromes (MDS) patients. Methods PI-PLCβ1, PI-PLCβ4, and PI-PLCγ1 cytogenetic investigations were performed on 80 newly diagnosed MDS patients (18 low risk, 26 intermediate 1, 18 intermediate 2, 18 high risk) comparing the results with the clinical outcome of the patients. Moreover, fluorescent in situ hybridization results were validated by real-time polymerase chain reaction (PCR). Finally, PI-PLCβ1 gene and protein expression were assessed by both real-time PCR and immunocytochemical experiments. Results Collectively, 35 (43.75%) of 80 of the MDS patients showed a specific mono-allelic deletion of PI-PLCβ1. Kaplan-Meier analysis revealed a significant association (P < .0001) between the PI-PLCβ1 mono-allelic deletion and a higher risk of evolution into acute myeloid leukemia (AML), since 23 of 35 MDS patients (65.7%) bearing the PI-PLCβ1 mono-allelic deletion evolved into AML. Even in multivariate analysis, the PI-PLCβ1 mono-allelic deletion retained a higher significance, with a P < .001, as a prognostic factor of evolution into AML (odds ratio [OR] 1.83; 95% CI, 2.26 to 17.24; P = .00045). Finally, PI-PLCβ1 deletion was related to an altered gene and protein expression. Conclusion PI-PLCβ1 mono-allelic deletion is associated with a worse clinical outcome in MDS patients, hinting at the identification of a new group at higher risk of AML evolution and representing a reliable prognostic tool. Moreover, targeting PI-PLCβ1 pathways might emerge as a new therapeutic strategy for MDS.
Collapse
Affiliation(s)
- Matilde Y. Follo
- From the Cellular Signalling Laboratory, Department of Human Anatomical Sciences; Institute of Hematology and Medical Oncology “L. e A. Seràgnoli”, University of Bologna; Hematology Unit, Ospedale Civile di Piacenza; and the Istituto per i Trapianti d'Organo e l'Immunocitologia del CNR, Sezione di Bologna, Bologna, Italy
| | - Carlo Finelli
- From the Cellular Signalling Laboratory, Department of Human Anatomical Sciences; Institute of Hematology and Medical Oncology “L. e A. Seràgnoli”, University of Bologna; Hematology Unit, Ospedale Civile di Piacenza; and the Istituto per i Trapianti d'Organo e l'Immunocitologia del CNR, Sezione di Bologna, Bologna, Italy
| | - Cristina Clissa
- From the Cellular Signalling Laboratory, Department of Human Anatomical Sciences; Institute of Hematology and Medical Oncology “L. e A. Seràgnoli”, University of Bologna; Hematology Unit, Ospedale Civile di Piacenza; and the Istituto per i Trapianti d'Organo e l'Immunocitologia del CNR, Sezione di Bologna, Bologna, Italy
| | - Sara Mongiorgi
- From the Cellular Signalling Laboratory, Department of Human Anatomical Sciences; Institute of Hematology and Medical Oncology “L. e A. Seràgnoli”, University of Bologna; Hematology Unit, Ospedale Civile di Piacenza; and the Istituto per i Trapianti d'Organo e l'Immunocitologia del CNR, Sezione di Bologna, Bologna, Italy
| | - Costanza Bosi
- From the Cellular Signalling Laboratory, Department of Human Anatomical Sciences; Institute of Hematology and Medical Oncology “L. e A. Seràgnoli”, University of Bologna; Hematology Unit, Ospedale Civile di Piacenza; and the Istituto per i Trapianti d'Organo e l'Immunocitologia del CNR, Sezione di Bologna, Bologna, Italy
| | - Giovanni Martinelli
- From the Cellular Signalling Laboratory, Department of Human Anatomical Sciences; Institute of Hematology and Medical Oncology “L. e A. Seràgnoli”, University of Bologna; Hematology Unit, Ospedale Civile di Piacenza; and the Istituto per i Trapianti d'Organo e l'Immunocitologia del CNR, Sezione di Bologna, Bologna, Italy
| | - Michele Baccarani
- From the Cellular Signalling Laboratory, Department of Human Anatomical Sciences; Institute of Hematology and Medical Oncology “L. e A. Seràgnoli”, University of Bologna; Hematology Unit, Ospedale Civile di Piacenza; and the Istituto per i Trapianti d'Organo e l'Immunocitologia del CNR, Sezione di Bologna, Bologna, Italy
| | - Lucia Manzoli
- From the Cellular Signalling Laboratory, Department of Human Anatomical Sciences; Institute of Hematology and Medical Oncology “L. e A. Seràgnoli”, University of Bologna; Hematology Unit, Ospedale Civile di Piacenza; and the Istituto per i Trapianti d'Organo e l'Immunocitologia del CNR, Sezione di Bologna, Bologna, Italy
| | - Alberto M. Martelli
- From the Cellular Signalling Laboratory, Department of Human Anatomical Sciences; Institute of Hematology and Medical Oncology “L. e A. Seràgnoli”, University of Bologna; Hematology Unit, Ospedale Civile di Piacenza; and the Istituto per i Trapianti d'Organo e l'Immunocitologia del CNR, Sezione di Bologna, Bologna, Italy
| | - Lucio Cocco
- From the Cellular Signalling Laboratory, Department of Human Anatomical Sciences; Institute of Hematology and Medical Oncology “L. e A. Seràgnoli”, University of Bologna; Hematology Unit, Ospedale Civile di Piacenza; and the Istituto per i Trapianti d'Organo e l'Immunocitologia del CNR, Sezione di Bologna, Bologna, Italy
| |
Collapse
|
22
|
Dorn I, Lazar-Karsten P, Boie S, Ribbat J, Hartwig D, Driller B, Kirchner H, Schlenke P. In vitro proliferation and differentiation of human CD34+ cells from peripheral blood into mature red blood cells with two different cell culture systems. Transfusion 2008; 48:1122-32. [DOI: 10.1111/j.1537-2995.2008.01653.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
23
|
Tong Q, Hirschler-Laszkiewicz I, Zhang W, Conrad K, Neagley DW, Barber DL, Cheung JY, Miller BA. TRPC3 is the erythropoietin-regulated calcium channel in human erythroid cells. J Biol Chem 2008; 283:10385-95. [PMID: 18276585 DOI: 10.1074/jbc.m710231200] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Erythropoietin (Epo) stimulates a significant increase in the intracellular calcium concentration ([Ca(2+)](i)) through activation of the murine transient receptor potential channel TRPC2, but TRPC2 is a pseudogene in humans. TRPC3 expression increases on normal human erythroid progenitors during differentiation. Here, we determined that erythropoietin regulates calcium influx through TRPC3. Epo stimulation of HEK 293T cells transfected with Epo receptor and TRPC3 resulted in a dose-dependent increase in [Ca(2+)](i), which required extracellular calcium influx. Treatment with the phospholipase C (PLC) inhibitor U-73122 or down-regulation of PLCgamma1 by RNA interference inhibited the Epo-stimulated increase in [Ca(2+)](i) in TRPC3-transfected HEK 293T cells and in primary human erythroid precursors, demonstrating a requirement for PLC. TRPC3 associated with PLCgamma, and substitution of predicted PLCgamma Src homology 2 binding sites (Y226F, Y555F, Y648F, and Y674F) on TRPC3 reduced the interaction of TRPC3 with PLCgamma and inhibited the rise in [Ca(2+)](i). Substitution of Tyr(226) alone with phenylalanine significantly reduced the Epo-stimulated increase in [Ca(2+)](i) but not the association of PLCgamma with TRPC3. PLC activation results in production of inositol 1,4,5-trisphosphate (IP(3)). To determine whether IP(3) is involved in Epo activation of TRPC3, TRPC3 mutants were prepared with substitution or deletion of COOH-terminal IP(3) receptor (IP(3)R) binding domains. In cells expressing TRPC3 with mutant IP(3)R binding sites and Epo receptor, interaction of IP(3)R with TRPC3 was abolished, and Epo-modulated increase in [Ca(2+)](i) was reduced. Our data demonstrate that Epo modulates TRPC3 activation through a PLCgamma-mediated process that requires interaction of PLCgamma and IP(3)R with TRPC3. They also show that TRPC3 Tyr(226) is critical in Epo-dependent activation of TRPC3. These data demonstrate a redundancy of TRPC channel activation mechanisms by widely different agonists.
Collapse
Affiliation(s)
- Qin Tong
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Lanuti P, Bertagnolo V, Gaspari AR, Ciccocioppo F, Pierdomenico L, Bascelli A, Sabatino G, Miscia S, Marchisio M. Parallel regulation of PKC-alpha and PKC-delta characterizes the occurrence of erythroid differentiation from human primary hematopoietic progenitors. Exp Hematol 2007; 34:1624-34. [PMID: 17157158 DOI: 10.1016/j.exphem.2006.07.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2006] [Revised: 07/13/2006] [Accepted: 07/24/2006] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Erythroid differentiation is a process characterized by modulation of different proteins including phosphoinositide-related enzymes such as protein kinase C (PKC) isoforms. Because in different cell lines PKC-alpha and PKC-delta have been reported to be involved in the mechanisms controlling proliferation and differentiation, the aim of this study was to examine the relative involvement of these PKC isoforms in the development of CD235a+ erythroid cells from human healthy hematopoietic progenitors. MATERIALS AND METHODS Erythroid differentiation from human primary hematopoietic progenitor cells was achieved by adopting the human erythroblasts mass amplification culture. Expression and activity of PKC isoforms and their relationship with proliferation and differentiation were investigated by morphologic analysis, reverse-transcriptase polymerase chain reaction, Western blotting, multiparametric flow cytometry, and transfection experiments. RESULTS PKC-alpha was found expressed and phosphorylated in cells undergoing both proliferation and differentiation, although PKC-delta, largely expressed and activated during proliferation, was evidently downregulated during differentiation. Overexpression of PKC-delta-CAT scarcely influenced the development of glycophorin-A (CD235a)+ erythroid cells from hematopoietic progenitors, although overexpression of PKC-alpha-CAT strongly induced the development of CD235a+ erythroid cells. On the other hand, in PKC-alpha-CAT-transfected cells, pharmacologic inhibition of PKC-delta further increased the number of CD235a+ cells, although inhibition of PKC-alpha resulted in an evident impairment of the development of CD235a+ erythroid cells. CONCLUSIONS Our results indicate that the suppression or at least a strong downregulation of PKC-delta, concomitant to PKC-alpha expression and activity, might be a cofactor to be further investigated and might be involved in the events regulating erythropoietin-induced erythroid differentiation from human primary hematopoietic progenitor cells.
Collapse
Affiliation(s)
- Paola Lanuti
- Cell Signalling Unit, Section of Human Anatomy, Department of Biomorphology, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Marchisio M, Santavenere E, Paludi M, Gaspari AR, Lanuti P, Bascelli A, Ercolino E, Di Baldassarre A, Miscia S. Erythroid cell differentiation is characterized by nuclear matrix localization and phosphorylation of protein kinases C (PKC) alpha, delta, and zeta. J Cell Physiol 2005; 205:32-6. [PMID: 15880451 DOI: 10.1002/jcp.20364] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Protein kinases C (PKC) zeta expression and phosphorylation at nuclear level during dimethyl sulfoxide (DMSO)-induced differentiation in Friend erythroleukemia cells have been previously reported, suggesting a possible role of this PKC isoform in the DMSO-related signaling. In order to shed more light on this tantalizing topic, we investigated PKC intracellular and sub-cellular localization and activity during DMSO-induced erythroid differentiation. Results indicated that at least PKC alpha, zeta, and delta are strongly and temporally involved in the DMSO-induced differentiation signals since their expression and phosphorylation, though at different extents, were observed during treatments. Intriguingly, while PKC alpha and zeta associate to the nuclear matrix during the differentiation event, PKC delta appears to be residentially associated to the nuclear matrix. Furthermore, an evident downregulation of the beta-globin gene transcription (differentiation hallmark) was detected upon a progressive inhibition of these PKC isoforms by means of specific inhibitors, indicating, therefore, that PKC alpha, zeta, and delta phosphorylation play a crucial role in the control of erythroid differentiation.
Collapse
Affiliation(s)
- Marco Marchisio
- Cell Signalling Unit, Section of Human Anatomy, Department of Biomorfologia, University "G.d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|