1
|
Wacka E, Wawrzyniak-Gramacka E, Tylutka A, Morawin B, Gutowicz M, Zembron-Lacny A. The Role of Inflammation in Age-Associated Changes in Red Blood System. Int J Mol Sci 2023; 24:ijms24108944. [PMID: 37240288 DOI: 10.3390/ijms24108944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/08/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Aging-related anemia contributes to frailty syndrome, cognitive decline and early mortality. The study aim was to evaluate inflammaging in relation to anemia as a prognostic indicator in affected older patients. The participants (73.0 ± 7.2 years) were allocated into anemic (n = 47) and non-anemic (n = 66) groups. The hematological variables RBC, MCV, MCH, RDW, iron and ferritin were significantly lower, whereas erythropoietin EPO and transferrin Tf tended toward higher values in the anemic group. Approx. 26% of individuals demonstrated transferrin saturation TfS < 20%, which clearly indicates age-related iron deficiency. The cut-off values for pro-inflammatory cytokine IL-1β, TNFα and hepcidin were 5.3 ng/mL, 97.7 ng/mL and 9.4 ng/mL, respectively. High IL-1β negatively affected Hb concentration (rs = -0.581, p < 0.0001). Relatively high odds ratios were observed for IL-1β (OR = 72.374, 95%Cl 19.688-354.366) and peripheral blood mononuclear cells CD34 (OR = 3.264, 95%Cl 1.263-8.747) and CD38 (OR = 4.398, 95%Cl 1.701-11.906), which together indicates a higher probability of developing anemia. The results endorse the interplay between inflammatory status and iron metabolism and demonstrated a high usefulness of IL-1β in identification of the underlying causes of anemia, while CD34 and CD38 appeared useful in compensatory response assessment and, in the longer term, as part of a comprehensive approach to anemia monitoring in older adults.
Collapse
Affiliation(s)
- Eryk Wacka
- Department of Applied and Clinical Physiology, Collegium Medicum University of Zielona Gora, 65-417 Zielona Gora, Poland
| | - Edyta Wawrzyniak-Gramacka
- Department of Applied and Clinical Physiology, Collegium Medicum University of Zielona Gora, 65-417 Zielona Gora, Poland
| | - Anna Tylutka
- Department of Applied and Clinical Physiology, Collegium Medicum University of Zielona Gora, 65-417 Zielona Gora, Poland
| | - Barbara Morawin
- Department of Applied and Clinical Physiology, Collegium Medicum University of Zielona Gora, 65-417 Zielona Gora, Poland
| | - Marzena Gutowicz
- Department of Applied and Clinical Physiology, Collegium Medicum University of Zielona Gora, 65-417 Zielona Gora, Poland
| | - Agnieszka Zembron-Lacny
- Department of Applied and Clinical Physiology, Collegium Medicum University of Zielona Gora, 65-417 Zielona Gora, Poland
| |
Collapse
|
2
|
Shivam S, El-Matbouli M, Kumar G. Kinetics of Parasite-Specific Antibody and B-Cell-Associated Gene Expression in Brown Trout, Salmo trutta during Proliferative Kidney Disease. BIOLOGY 2021; 10:1244. [PMID: 34943159 PMCID: PMC8698486 DOI: 10.3390/biology10121244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 01/20/2023]
Abstract
Tetracapsuloides bryosalmonae, a myxozoan endoparasite often causes chronic infection in brown trout. Antiparasite immunity mediated by antibodies and B cells is known as an important determinant of host survival and parasite proliferation during chronic infections. Accordingly, studying their time course during proliferative kidney disease (PKD) might be helpful in improving our understanding of its chronic nature. Therefore, we conducted this study to examine parasite specific serum antibody and B-cell-mediated response in laboratory-infected brown trout at different time points. Brown trout were exposed to the spores of T. bryosalmonae, derived from infected bryozoans. Samples were collected at different time points and processed for indirect ELISA, histopathology, and qRT-PCR. T. bryosalmonae specific antibody was detected at 4 weeks post exposure (wpe) and it persisted until 17 wpe. Additionally, the expressions of C4A, CD34, CD79A, BLNK, CD74, BCL7, and CD22 were differentially regulated in the important immune organs, kidney and spleen. To our knowledge, this is the first study addressing anti-T. bryosalmonae antibody response in brown trout at different time points. The results from this study provide valuable insights into the processes leading to changes in B cell development, inflammation and antibody production during the course of PKD in brown trout.
Collapse
Affiliation(s)
- Saloni Shivam
- Clinical Division of Fish Medicine, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
- Central Marine Fisheries Research Institute, Karwar 581301, India
| | - Mansour El-Matbouli
- Clinical Division of Fish Medicine, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Gokhlesh Kumar
- Clinical Division of Fish Medicine, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| |
Collapse
|
3
|
Kapoor S, Shenoy SP, Bose B. CD34 cells in somatic, regenerative and cancer stem cells: Developmental biology, cell therapy, and omics big data perspective. J Cell Biochem 2019; 121:3058-3069. [PMID: 31886574 DOI: 10.1002/jcb.29571] [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: 07/29/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022]
Abstract
The transmembrane phosphoglycoprotein protein CD34 has conventionally been regarded as a marker for hematopoietic progenitors. Its expression on these cells has been leveraged for cell therapy applications in various hematological disorders. More recently, the expression of CD34 has also been reported on cells of nonhematopoietic origin. The list includes somatic cells such as endothelial cells, fibrocytes and interstitial cells and regenerative stem cells such as corneal keratocytes, muscle satellite cells, and muscle-derived stem cells. Furthermore, its expression on some cancer stem cells (CSCs) has also been reported. Till date, the functional roles of this molecule have been implicated in a multitude of cellular processes including cell adhesion, signal transduction, and maintenance of progenitor phenotype. However, the complete understanding about this molecule including its developmental origins, its embryonic connection, and associated functions is far from complete. Here, we review our present understanding of the structure and putative functions of the CD34 molecule based upon our literature survey. We also probed various biological databases to retrieve data related to the expression and associated molecular functions of CD34. Such information, upon synthesis, is hence likely to provide the suitability of such cells for cell therapy. Moreover, we have also covered the existing cell therapy and speculated cell therapy applications of CD34+ cells isolated from various lineages. We have also attempted here to speculate the role(s) of CD34 on CSCs. Finally, we discuss number of large-scale proteomics and transcriptomics studies that have been performed using CD34+ cells.
Collapse
Affiliation(s)
- Saketh Kapoor
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Sudheer P Shenoy
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| |
Collapse
|
4
|
Salati S, Genovese E, Carretta C, Zini R, Bartalucci N, Prudente Z, Pennucci V, Ruberti S, Rossi C, Rontauroli S, Enzo E, Calabresi L, Balliu M, Mannarelli C, Bianchi E, Guglielmelli P, Tagliafico E, Vannucchi AM, Manfredini R. Calreticulin Ins5 and Del52 mutations impair unfolded protein and oxidative stress responses in K562 cells expressing CALR mutants. Sci Rep 2019; 9:10558. [PMID: 31332222 PMCID: PMC6646313 DOI: 10.1038/s41598-019-46843-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 07/03/2019] [Indexed: 12/26/2022] Open
Abstract
Somatic mutations of calreticulin (CALR) have been described in approximately 60–80% of JAK2 and MPL unmutated Essential Thrombocythemia and Primary Myelofibrosis patients. CALR is an endoplasmic reticulum (ER) chaperone responsible for proper protein folding and calcium retention. Recent data demonstrated that the TPO receptor (MPL) is essential for the development of CALR mutant-driven Myeloproliferative Neoplasms (MPNs). However, the precise mechanism of action of CALR mutants haven’t been fully unraveled. In this study, we showed that CALR mutants impair the ability to respond to the ER stress and reduce the activation of the pro-apoptotic pathway of the unfolded protein response (UPR). Moreover, our data demonstrated that CALR mutations induce increased sensitivity to oxidative stress, leading to increase oxidative DNA damage. We finally demonstrated that the downmodulation of OXR1 in CALR-mutated cells could be one of the molecular mechanisms responsible for the increased sensitivity to oxidative stress mediated by mutant CALR. Altogether, our data identify novel mechanisms collaborating with MPL activation in CALR-mediated cellular transformation. CALR mutants negatively impact on the capability of cells to respond to oxidative stress leading to genomic instability and on the ability to react to ER stress, causing resistance to UPR-induced apoptosis.
Collapse
Affiliation(s)
- Simona Salati
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Genovese
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Chiara Carretta
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberta Zini
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Niccolò Bartalucci
- CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Zelia Prudente
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Pennucci
- Institute for Cell and Gene Therapy & Center for Chronic Immunodeficiency, University of Freiburg, Freiburg, Germany
| | - Samantha Ruberti
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Chiara Rossi
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sebastiano Rontauroli
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Enzo
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Calabresi
- CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Manjola Balliu
- CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Carmela Mannarelli
- CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Elisa Bianchi
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Paola Guglielmelli
- CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Enrico Tagliafico
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy.,Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandro M Vannucchi
- CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Rossella Manfredini
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
5
|
Nakanishi M, Mitchell RR, Benoit YD, Orlando L, Reid JC, Shimada K, Davidson KC, Shapovalova Z, Collins TJ, Nagy A, Bhatia M. Human Pluripotency Is Initiated and Preserved by a Unique Subset of Founder Cells. Cell 2019; 177:910-924.e22. [PMID: 30982595 DOI: 10.1016/j.cell.2019.03.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/21/2018] [Accepted: 03/05/2019] [Indexed: 12/12/2022]
Abstract
The assembly of organized colonies is the earliest manifestation in the derivation or induction of pluripotency in vitro. However, the necessity and origin of this assemblance is unknown. Here, we identify human pluripotent founder cells (hPFCs) that initiate, as well as preserve and establish, pluripotent stem cell (PSC) cultures. PFCs are marked by N-cadherin expression (NCAD+) and reside exclusively at the colony boundary of primate PSCs. As demonstrated by functional analysis, hPFCs harbor the clonogenic capacity of PSC cultures and emerge prior to commitment events or phenotypes associated with pluripotent reprogramming. Comparative single-cell analysis with pre- and post-implantation primate embryos revealed hPFCs share hallmark properties with primitive endoderm (PrE) and can be regulated by non-canonical Wnt signaling. Uniquely informed by primate embryo organization in vivo, our study defines a subset of founder cells critical to the establishment pluripotent state.
Collapse
Affiliation(s)
- Mio Nakanishi
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Ryan R Mitchell
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Yannick D Benoit
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Luca Orlando
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Jennifer C Reid
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Kenichi Shimada
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Kathryn C Davidson
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Zoya Shapovalova
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Tony J Collins
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Andras Nagy
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Mickie Bhatia
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada.
| |
Collapse
|
6
|
Noroozinia F, Mahmoudzadeh L, Gharalari FH, Makhdoomi K, Abbasi A. Relationship between interstitial CD34 positive cells and active phase of lupus nephritis. Eur J Rheumatol 2018; 5:254-257. [PMID: 30308141 PMCID: PMC6267746 DOI: 10.5152/eurjrheum.2018.18067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/08/2018] [Indexed: 11/22/2022] Open
Abstract
Objective Lupus nephritis is one of the most serious and common complications of systemic lupus erythematosus. It has an unpredictable course, and the type, severity, and activity of renal lesions cannot be assessed only by clinical and laboratory findings. The aim of the present study was to determine the relationship between the expression of CD34 and the histopathological findings of lupus nephritis. Methods A total of 73 renal biopsy samples of patients with a diagnosis of lupus nephritis were examined for CD34 expression by immunohistochemistry. Samples without staining were considered as 0, mild staining as 1+, moderate as 2+, and strong staining as 3+. The relationship between CD34 expression and histopathological and clinical data (including activity index, chronicity index, lupus nephritis class, age, sex, blood pressure, complete blood count, renal function tests, and serological findings) was analyzed. Results The mean age of the patients was 29.3±11.3 years. CD34 was expressed in all of the cases but with different intensities. There was a significant relationship between the expression of CD34 and the activity index, as a strong expression was seen in lower activity indices (p<0.001). CD34 expression was correlated with patients’ white blood cell (WBC) count and systolic blood pressure (SBP). Patients with strong (score 3) CD34 expression had higher SBPs and lower WBC counts (p=0.03 and 0.04, respectively). Conclusion A strong interstitial expression of CD34 was observed in lower activity indices. It seems that CD34 expression could play a protective role in lupus nephritis and could reduce renal activity.
Collapse
Affiliation(s)
- Farahnaz Noroozinia
- Department of Pathology, Urmia University of Medical Sciences School of Medicine, Urmia, Iran
| | - Leila Mahmoudzadeh
- Department of Pathology, Urmia University of Medical Sciences School of Medicine, Urmia, Iran
| | | | - Khadijeh Makhdoomi
- Nephrology and Renal Transplant Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Ata Abbasi
- Department of Pathology, Urmia University of Medical Sciences School of Medicine, Urmia, Iran
| |
Collapse
|
7
|
Salati S, Prudente Z, Genovese E, Pennucci V, Rontauroli S, Bartalucci N, Mannarelli C, Ruberti S, Zini R, Rossi C, Bianchi E, Guglielmelli P, Tagliafico E, Vannucchi AM, Manfredini R. Calreticulin Affects Hematopoietic Stem/Progenitor Cell Fate by Impacting Erythroid and Megakaryocytic Differentiation. Stem Cells Dev 2018; 27:225-236. [PMID: 29258411 DOI: 10.1089/scd.2017.0137] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Calreticulin (CALR) is a chaperone protein that localizes primarily to the endoplasmic reticulum (ER) lumen where it is responsible for the control of proper folding of neo-synthesized glycoproteins and the retention of calcium. Recently, mutations affecting exon 9 of the CALR gene have been described in approximately 40% of patients with myeloproliferative neoplasms (MPNs). Although the role of mutated CALR in the development of MPNs has begun to be clarified, there are still no data available on the function of wild-type (WT) CALR during physiological hematopoiesis. To shed light on the role of WT CALR during normal hematopoiesis, we performed gene silencing and overexpression experiments in hematopoietic stem progenitor cells (HSPCs). Our results showed that CALR overexpression is able to affect physiological hematopoiesis by enhancing both erythroid and megakaryocytic (MK) differentiation. In agreement with overexpression data, CALR silencing caused a significant decrease in both erythroid and MK differentiation of human HSPCs. Gene expression profiling (GEP) analysis showed that CALR is able to affect the expression of several genes involved in HSPC differentiation toward both the erythroid and MK lineages. Moreover, GEP data also highlighted the modulation of several genes involved in ER stress response, unfolded protein response (UPR), and DNA repair, and of several genes already described to play a role in MPN development, such as proinflammatory cytokines and hematological neoplasm-related markers. Altogether, our data unraveled a new and unexpected role for CALR in the regulation of normal hematopoietic differentiation. Moreover, by showing the impact of CALR on the expression of genes involved in several biological processes already described in cellular transformation, our data strongly suggest a more complex role for CALR in MPN development that goes beyond the activation of the THPO receptor and involves ER stress response, UPR, and DNA repair.
Collapse
Affiliation(s)
- Simona Salati
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| | - Zelia Prudente
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Genovese
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Pennucci
- Institute for Cell and Gene Therapy & Center for Chronic Immunodeficiency, University of Freiburg, Freiburg, Germany
| | - Sebastiano Rontauroli
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| | - Niccolò Bartalucci
- CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Carmela Mannarelli
- CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Samantha Ruberti
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| | - Roberta Zini
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| | - Chiara Rossi
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| | - Elisa Bianchi
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| | - Paola Guglielmelli
- CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Enrico Tagliafico
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandro M Vannucchi
- CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, Department of Experimental and Clinical Medicine, AOU Careggi, University of Florence, Florence, Italy
| | - Rossella Manfredini
- Centre for Regenerative Medicine "Stefano Ferrari," University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
8
|
Zhu LF, Xiao M, Chen YQ, Wang LY, Luo XF, Yuan XH, Ren JH, Chen ZZ, Hu JD, Yang T. In vitro effects of reprogramming factors on the expressions of pluripotent genes and CD 34 gene in human acute promyelocytic leukemia HL-60 cells. Genomics 2017; 109:331-335. [PMID: 28533192 DOI: 10.1016/j.ygeno.2017.05.006] [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] [Received: 03/06/2017] [Revised: 04/28/2017] [Accepted: 05/18/2017] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Our study aims to explore the in vitro effects of reprogramming factors on the expressions of pluripotent genes and CD34 gene in HL-60 cells. METHODS According to the construction of lentiviral vector LV-OSCK of reprogramming factors (Oct-4, Sox2, Klf4, c-Myc), 293T cells were transfected to detect virus titer. The endogenous pluripotent genes (Oct4, SOX2, c-Myc and Klf4) and CD34 mRNA and protein expressions were detected by AP staining, immunofluorescence staining, qRT-PCR and flow cytometry. RESULTS Expressions of Oct4, SOX2, c-Myc and Klf4 were 0.220±0.013, 0.186±0.009, 0.287±0.015 and 0.153±0.007. These levels were significantly higher in the experimental group than the control and blank groups. CD34 protein expression in the experimental group was also discovered to be significantly higher than the other two groups. CONCLUSION The reprogramming factors could increase the expressions of pluripotent genes and CD34 gene in HL-60 cells.
Collapse
Affiliation(s)
- Liang-Fang Zhu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Min Xiao
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Yong-Quan Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Ling-Yan Wang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Xiao-Feng Luo
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Xiao-Hong Yuan
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Jin-Hua Ren
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Zhi-Zhe Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Jian-Da Hu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China
| | - Ting Yang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou 350001, PR China.
| |
Collapse
|
9
|
Daniele G, Simonetti G, Fusilli C, Iacobucci I, Lonoce A, Palazzo A, Lomiento M, Mammoli F, Marsano RM, Marasco E, Mantovani V, Quentmeier H, Drexler HG, Ding J, Palumbo O, Carella M, Nadarajah N, Perricone M, Ottaviani E, Baldazzi C, Testoni N, Papayannidis C, Ferrari S, Mazza T, Martinelli G, Storlazzi CT. Epigenetically induced ectopic expression of UNCX impairs the proliferation and differentiation of myeloid cells. Haematologica 2017; 102:1204-1214. [PMID: 28411256 PMCID: PMC5566027 DOI: 10.3324/haematol.2016.163022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 04/12/2017] [Indexed: 11/09/2022] Open
Abstract
We here describe a leukemogenic role of the homeobox gene UNCX,
activated by epigenetic modifications in acute myeloid leukemia (AML). We found
the ectopic activation of UNCX in a leukemia patient harboring
a t(7;10)(p22;p14) translocation, in 22 of 61 of additional cases [a
total of 23 positive patients out of 62 (37.1%)], and in 6 of 75
(8%) of AML cell lines. UNCX is embedded within a
low-methylation region (canyon) and encodes for a transcription factor involved
in somitogenesis and neurogenesis, with specific expression in the eye, brain,
and kidney. UNCX expression turned out to be associated, and
significantly correlated, with DNA methylation increase at its canyon borders
based on data in our patients and in archived data of patients from The Cancer
Genome Atlas. UNCX-positive and -negative patients displayed
significant differences in their gene expression profiles. An enrichment of
genes involved in cell proliferation and differentiation, such as
MAP2K1 and CCNA1, was revealed. Similar
results were obtained in UNCX-transduced CD34+
cells, associated with low proliferation and differentiation arrest.
Accordingly, we showed that UNCX expression characterizes
leukemia cells at their early stage of differentiation, mainly M2 and M3
subtypes carrying wild-type NPM1. We also observed that
UNCX expression significantly associates with an increased
frequency of acute promyelocytic leukemia with PML-RARA and AML
with t(8;21)(q22;q22.1); RUNX1-RUNX1T1 classes, according to
the World Health Organization disease classification. In summary, our findings
suggest a novel leukemogenic role of UNCX, associated with
epigenetic modifications and with impaired cell proliferation and
differentiation in AML.
Collapse
Affiliation(s)
- Giulia Daniele
- Department of Biology, University of Bari "A. Moro", Italy
| | - Giorgia Simonetti
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Caterina Fusilli
- IRCCS Casa Sollievo della Sofferenza, Bioinformatics Unit, San Giovanni Rotondo, Italy
| | - Ilaria Iacobucci
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Angelo Lonoce
- Department of Biology, University of Bari "A. Moro", Italy
| | | | - Mariana Lomiento
- Department of Life Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Fabiana Mammoli
- Department of Life Science, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Elena Marasco
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Vilma Mantovani
- Center for Applied Biomedical Research (CRBA), S. Orsola-Malpighi Hospital, Bologna, Italy.,Unit of Medical Genetics, Department of Medical and Surgical Sciences, S. Orsola-Malpighi Hospital University of Bologna, Italy
| | - Hilmar Quentmeier
- Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Department of Human and Animal Cell Lines, Braunschweig, Germany
| | - Hans G Drexler
- Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Department of Human and Animal Cell Lines, Braunschweig, Germany
| | - Jie Ding
- Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Department of Human and Animal Cell Lines, Braunschweig, Germany
| | - Orazio Palumbo
- Medical Genetics Unit, IRCCS "Casa Sollievo della Sofferenza (CSS)" Hospital, San Giovanni Rotondo, Italy
| | - Massimo Carella
- Medical Genetics Unit, IRCCS "Casa Sollievo della Sofferenza (CSS)" Hospital, San Giovanni Rotondo, Italy
| | | | - Margherita Perricone
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Emanuela Ottaviani
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Carmen Baldazzi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Nicoletta Testoni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Cristina Papayannidis
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Sergio Ferrari
- Department of Life Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Tommaso Mazza
- IRCCS Casa Sollievo della Sofferenza, Bioinformatics Unit, San Giovanni Rotondo, Italy
| | - Giovanni Martinelli
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | | |
Collapse
|
10
|
Zini R, Rossi C, Norfo R, Pennucci V, Barbieri G, Ruberti S, Rontauroli S, Salati S, Bianchi E, Manfredini R. miR-382-5p Controls Hematopoietic Stem Cell Differentiation Through the Downregulation of MXD1. Stem Cells Dev 2016; 25:1433-43. [DOI: 10.1089/scd.2016.0150] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Roberta Zini
- Centre for Regenerative Medicine “Stefano Ferrari,” Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Chiara Rossi
- Centre for Regenerative Medicine “Stefano Ferrari,” Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Ruggiero Norfo
- Centre for Regenerative Medicine “Stefano Ferrari,” Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Haematopoietic Stem Cell Biology Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Valentina Pennucci
- Centre for Regenerative Medicine “Stefano Ferrari,” Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Greta Barbieri
- Centre for Regenerative Medicine “Stefano Ferrari,” Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Samantha Ruberti
- Centre for Regenerative Medicine “Stefano Ferrari,” Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sebastiano Rontauroli
- Centre for Regenerative Medicine “Stefano Ferrari,” Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Simona Salati
- Centre for Regenerative Medicine “Stefano Ferrari,” Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisa Bianchi
- Centre for Regenerative Medicine “Stefano Ferrari,” Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Rossella Manfredini
- Centre for Regenerative Medicine “Stefano Ferrari,” Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
11
|
Maintenance of the functional integrity of mouse hematopoiesis by EED and promotion of leukemogenesis by EED haploinsufficiency. Sci Rep 2016; 6:29454. [PMID: 27432459 PMCID: PMC4949429 DOI: 10.1038/srep29454] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/20/2016] [Indexed: 12/16/2022] Open
Abstract
Polycomb repressive complex 2 (PRC2) participates in transcriptional repression through methylation of histone H3K27. The WD-repeat protein embryonic ectoderm development (EED) is a non-catalytic but an essential component of PRC2 and its mutations were identified in hematopoietic malignancies. To clarify the role(s) of EED in adult hematopoiesis and leukemogenesis, we generated Eed conditional knockout (EedΔ/Δ) mice. EedΔ/Δ mice died in a short period with rapid decrease of hematopoietic cells. Hematopoietic stem/progenitor cells (HSPCs) were markedly decreased with impaired bone marrow (BM) repopulation ability. Cell cycle analysis of HSPCs demonstrated increased S-phase fraction coupled with suppressed G0/G1 entry. Genes encoding cell adhesion molecules are significantly enriched in EedΔ/Δ HSPCs, and consistently, EedΔ/Δ HSPCs exhibited increased attachment to a major extracellular matrix component, fibronectin. Thus, EED deficiency increases proliferation on one side but promotes quiescence possibly by enhanced adhesion to the hematopoietic niche on the other, and these conflicting events would lead to abnormal differentiation and functional defect of EedΔ/Δ HSPCs. In addition, Eed haploinsufficiency induced hematopoietic dysplasia, and Eed heterozygous mice were susceptible to malignant transformation and developed leukemia in cooperation with Evi1 overexpression. Our results demonstrated differentiation stage-specific and dose-dependent roles of EED in normal hematopoiesis and leukemogenesis.
Collapse
|
12
|
Salati S, Zini R, Nuzzo S, Guglielmelli P, Pennucci V, Prudente Z, Ruberti S, Rontauroli S, Norfo R, Bianchi E, Bogani C, Rotunno G, Fanelli T, Mannarelli C, Rosti V, Salmoiraghi S, Pietra D, Ferrari S, Barosi G, Rambaldi A, Cazzola M, Bicciato S, Tagliafico E, Vannucchi AM, Manfredini R. Integrative analysis of copy number and gene expression data suggests novel pathogenetic mechanisms in primary myelofibrosis. Int J Cancer 2016; 138:1657-69. [PMID: 26547506 DOI: 10.1002/ijc.29920] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 10/15/2015] [Accepted: 10/23/2015] [Indexed: 12/27/2022]
Abstract
Primary myelofibrosis (PMF) is a Myeloproliferative Neoplasm (MPN) characterized by megakaryocyte hyperplasia, progressive bone marrow fibrosis, extramedullary hematopoiesis and transformation to Acute Myeloid Leukemia (AML). A number of phenotypic driver (JAK2, CALR, MPL) and additional subclonal mutations have been described in PMF, pointing to a complex genomic landscape. To discover novel genomic lesions that can contribute to disease phenotype and/or development, gene expression and copy number signals were integrated and several genomic abnormalities leading to a concordant alteration in gene expression levels were identified. In particular, copy number gain in the polyamine oxidase (PAOX) gene locus was accompanied by a coordinated transcriptional up-regulation in PMF patients. PAOX inhibition resulted in rapid cell death of PMF progenitor cells, while sparing normal cells, suggesting that PAOX inhibition could represent a therapeutic strategy to selectively target PMF cells without affecting normal hematopoietic cells' survival. Moreover, copy number loss in the chromatin modifier HMGXB4 gene correlates with a concomitant transcriptional down-regulation in PMF patients. Interestingly, silencing of HMGXB4 induces megakaryocyte differentiation, while inhibiting erythroid development, in human hematopoietic stem/progenitor cells. These results highlight a previously un-reported, yet potentially interesting role of HMGXB4 in the hematopoietic system and suggest that genomic and transcriptional imbalances of HMGXB4 could contribute to the aberrant expansion of the megakaryocytic lineage that characterizes PMF patients.
Collapse
Affiliation(s)
- Simona Salati
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| | - Roberta Zini
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| | - Simona Nuzzo
- Center for Genome Research, University of Modena and Reggio Emilia, via Campi N.287, Modena, 41125, Italy
| | - Paola Guglielmelli
- Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Valentina Pennucci
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| | - Zelia Prudente
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| | - Samantha Ruberti
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| | - Sebastiano Rontauroli
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| | - Ruggiero Norfo
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| | - Elisa Bianchi
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| | - Costanza Bogani
- Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Giada Rotunno
- Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Tiziana Fanelli
- Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Carmela Mannarelli
- Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Vittorio Rosti
- IRCCS Policlinico S.Matteo Foundation, Center for the Study of Myelofibrosis, Pavia, Italy
| | | | - Daniela Pietra
- Department of Hematology Oncology, IRCCS Policlinico San Matteo Foundation & University of Pavia, Pavia, Italy
| | - Sergio Ferrari
- Center for Genome Research, University of Modena and Reggio Emilia, via Campi N.287, Modena, 41125, Italy
| | - Giovanni Barosi
- IRCCS Policlinico S.Matteo Foundation, Center for the Study of Myelofibrosis, Pavia, Italy
| | | | - Mario Cazzola
- Department of Hematology Oncology, IRCCS Policlinico San Matteo Foundation & University of Pavia, Pavia, Italy
| | - Silvio Bicciato
- Center for Genome Research, University of Modena and Reggio Emilia, via Campi N.287, Modena, 41125, Italy
| | - Enrico Tagliafico
- Center for Genome Research, University of Modena and Reggio Emilia, via Campi N.287, Modena, 41125, Italy
| | - Alessandro M Vannucchi
- Department of Experimental and Clinical Medicine, Laboratorio Congiunto MMPC, University of Florence, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Rossella Manfredini
- Life Sciences Department University of Modena and Reggio Emilia, Centre for Regenerative Medicine, via Gottardi N.100, Modena, 41125, Italy
| |
Collapse
|
13
|
Bhatia S, Reister S, Mahotka C, Meisel R, Borkhardt A, Grinstein E. Control of AC133/CD133 and impact on human hematopoietic progenitor cells through nucleolin. Leukemia 2015; 29:2208-20. [PMID: 26183533 DOI: 10.1038/leu.2015.146] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 05/19/2015] [Accepted: 05/29/2015] [Indexed: 01/01/2023]
Abstract
AC133 is a prominent surface marker of CD34+ and CD34- hematopoietic stem/progenitor cell (HSPC) subsets. AC133+ HSPCs contain high progenitor cell activity and are capable of hematopoietic reconstitution. Furthermore, AC133 is used for prospective isolation of tumor-initiating cells in several hematological malignancies. Nucleolin is a multifunctional factor of growing and cancer cells, which is aberrantly active in certain hematological neoplasms, and serves as a candidate molecular target for cancer therapy. Nucleolin is involved in gene transcription and RNA metabolism and is prevalently expressed in HSPCs, as opposed to differentiated hematopoietic tissue. The present study dissects nucleolin-mediated activation of surface AC133 and its cognate gene CD133, via specific interaction of nucleolin with the tissue-dependent CD133 promoter P1, as a mechanism that crucially contributes to AC133 expression in CD34+ HSPCs. In mobilized peripheral blood (MPB)-derived HSPCs, nucleolin elevates colony-forming unit (CFU) frequencies and enriches granulocyte-macrophage CFUs. Furthermore, nucleolin amplifies long-term culture-initiating cells and also promotes long-term, cytokine-dependent maintenance of hematopoietic progenitor cells. Active β-catenin, active Akt and Bcl-2 levels in MPB-derived HSPCs are nucleolin-dependent, and effects of nucleolin on these cells partially rely on β-catenin activity. The study provides new insights into molecular network relevant to stem/progenitor cells in normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- S Bhatia
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Heinrich Heine University, Düsseldorf, Germany
| | - S Reister
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Heinrich Heine University, Düsseldorf, Germany
| | - C Mahotka
- Institute of Pathology, Heinrich Heine University, Düsseldorf, Germany
| | - R Meisel
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Heinrich Heine University, Düsseldorf, Germany
| | - A Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Heinrich Heine University, Düsseldorf, Germany
| | - E Grinstein
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
14
|
Laranjeira P, Rodrigues R, Carvalheiro T, Constanço C, Vitória H, Matarraz S, Trindade H, Órfão A, Paiva A. Expression of CD44 and CD35 during normal and myelodysplastic erythropoiesis. Leuk Res 2015; 39:361-70. [DOI: 10.1016/j.leukres.2014.12.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 11/14/2014] [Accepted: 12/16/2014] [Indexed: 01/09/2023]
|
15
|
miRNA-mRNA integrative analysis in primary myelofibrosis CD34+ cells: role of miR-155/JARID2 axis in abnormal megakaryopoiesis. Blood 2014; 124:e21-32. [PMID: 25097177 DOI: 10.1182/blood-2013-12-544197] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by megakaryocyte (MK) hyperplasia, bone marrow fibrosis, and abnormal stem cell trafficking. PMF may be associated with somatic mutations in JAK2, MPL, or CALR. Previous studies have shown that abnormal MKs play a central role in the pathophysiology of PMF. In this work, we studied both gene and microRNA (miRNA) expression profiles in CD34(+) cells from PMF patients. We identified several biomarkers and putative molecular targets such as FGR, LCN2, and OLFM4. By means of miRNA-gene expression integrative analysis, we found different regulatory networks involved in the dysregulation of transcriptional control and chromatin remodeling. In particular, we identified a network gathering several miRNAs with oncogenic potential (eg, miR-155-5p) and targeted genes whose abnormal function has been previously associated with myeloid neoplasms, including JARID2, NR4A3, CDC42, and HMGB3. Because the validation of miRNA-target interactions unveiled JARID2/miR-155-5p as the strongest relationship in the network, we studied the function of this axis in normal and PMF CD34(+) cells. We showed that JARID2 downregulation mediated by miR-155-5p overexpression leads to increased in vitro formation of CD41(+) MK precursors. These findings suggest that overexpression of miR-155-5p and the resulting downregulation of JARID2 may contribute to MK hyperplasia in PMF.
Collapse
|
16
|
Abstract
Stroke is a leading cause of morbidity in the developed world and results in chronic disability in many cases. The literature related to the critical factors that regulate tissue self-regeneration in stroke is still limited, which restricts effective therapy. However, optimism in this area has been provided by recent research. The mechanisms involved in tissue regeneration and the mode of the participation of stem/progenitor cells and soluble protein neurotrophic factors in this process may yield a more complete understanding of the nature of stroke. This review summarizes the current understanding of both cellular and humoral issues with a particular emphasis on how these issues contribute to tissue regeneration in stroke.
Collapse
Affiliation(s)
- Bogusław Machalinski
- Department of General Pathology, Pomeranian Medical University, Al. Powstancow Wlkp. 72, Szczecin 70-111, Poland
| |
Collapse
|
17
|
Aiello FB, Graciotti L, Procopio AD, Keller JR, Durum SK. Stemness of T cells and the hematopoietic stem cells: fate, memory, niche, cytokines. Cytokine Growth Factor Rev 2013; 24:485-501. [PMID: 24231048 PMCID: PMC6390295 DOI: 10.1016/j.cytogfr.2013.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Stem cells are able to generate both cells that differentiate and cells that remain undifferentiated but potentially have the same developmental program. The prolonged duration of the protective immune memory for infectious diseases such as polio, small pox, and measles, suggested that memory T cells may have stem cell properties. Understanding the molecular basis for the life-long persistence of memory T cells may be useful to project targeted therapies for immune deficiencies and infectious diseases and to formulate vaccines. In the last decade evidence from different laboratories shows that memory T cells may share self-renewal pathways with bone marrow hematopoietic stem cells. In stem cells the intrinsic self-renewal activity, which depends on gene expression, is known to be modulated by extrinsic signals from the environment that may be tissue specific. These extrinsic signals for stemness of memory T cells include cytokines such as IL-7 and IL-15 and there are other cytokine signals for maintaining the cytokine signature (TH1, TH2, etc.) of memory T cells. Intrinsic and extrinsic pathways that might be common to bone marrow hematopoietic stem cells and memory T lymphocytes are discussed and related to self-renewal functions.
Collapse
Affiliation(s)
- Francesca B Aiello
- Laboratory of Molecular Immunoregulation, Frederick, MD 21702, USA; Department of Medicine and Aging Sciences, University of Chieti-Pescara, 66013 Chieti, Italy.
| | | | | | | | | |
Collapse
|
18
|
Faridi F, Ponnusamy K, Quagliano-Lo Coco I, Chen-Wichmann L, Grez M, Henschler R, Wichmann C. Aberrant epigenetic regulators control expansion of human CD34+ hematopoietic stem/progenitor cells. Front Genet 2013; 4:254. [PMID: 24348510 PMCID: PMC3842847 DOI: 10.3389/fgene.2013.00254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 11/08/2013] [Indexed: 12/02/2022] Open
Abstract
Transcription is a tightly regulated process ensuring the proper expression of numerous genes regulating all aspects of cellular behavior. Transcription factors regulate multiple genes including other transcription factors that together control a highly complex gene network. The transcriptional machinery can be “hijacked” by oncogenic transcription factors, thereby leading to malignant cell transformation. Oncogenic transcription factors manipulate a variety of epigenetic control mechanisms to fulfill gene regulatory and cell transforming functions. These factors assemble epigenetic regulators at target gene promoter sequences, thereby disturbing physiological gene expression patterns. Retroviral vector technology and the availability of “healthy” human hematopoietic CD34+ progenitor cells enable the generation of pre-leukemic cell models for the analysis of aberrant human hematopoietic progenitor cell expansion mediated by leukemogenic transcription factors. This review summarizes recent findings regarding the mechanism by which leukemogenic gene products control human hematopoietic CD34+ progenitor cell expansion by disrupting the normal epigenetic program.
Collapse
Affiliation(s)
- Farnaz Faridi
- Department of Transfusion Medicine, Cell Therapeutics and Hemostasis, Ludwig-Maximilian University Hospital Munich, Germany
| | - Kanagaraju Ponnusamy
- Department of Transfusion Medicine, Cell Therapeutics and Hemostasis, Ludwig-Maximilian University Hospital Munich, Germany ; Institute of Transfusion Medicine and Immunohematology, Goethe University Frankfurt, Germany
| | | | - Linping Chen-Wichmann
- Department of Transfusion Medicine, Cell Therapeutics and Hemostasis, Ludwig-Maximilian University Hospital Munich, Germany
| | - Manuel Grez
- Institute for Biomedical Research Georg-Speyer-Haus, Frankfurt, Germany
| | - Reinhard Henschler
- Department of Transfusion Medicine, Cell Therapeutics and Hemostasis, Ludwig-Maximilian University Hospital Munich, Germany
| | - Christian Wichmann
- Department of Transfusion Medicine, Cell Therapeutics and Hemostasis, Ludwig-Maximilian University Hospital Munich, Germany
| |
Collapse
|
19
|
Salati S, Lisignoli G, Manferdini C, Pennucci V, Zini R, Bianchi E, Norfo R, Facchini A, Ferrari S, Manfredini R. Co-culture of hematopoietic stem/progenitor cells with human osteblasts favours mono/macrophage differentiation at the expense of the erythroid lineage. PLoS One 2013; 8:e53496. [PMID: 23349713 PMCID: PMC3551919 DOI: 10.1371/journal.pone.0053496] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 11/29/2012] [Indexed: 12/25/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are located in the bone marrow in a specific microenvironment referred as the hematopoietic stem cell niche, where HSCs interact with a variety of stromal cells. Though several components of the stem cell niche have been identified, the regulatory mechanisms through which such components regulate the stem cell fate are still unknown. In order to address this issue, we investigated how osteoblasts (OBs) can affect the molecular and functional phenotype of Hematopoietic Stem/Progenitor Cells (HSPCs) and vice versa. For this purpose, human CD34+ cells were cultured in direct contact with primary human OBs. Our data showed that CD34+ cells cultured with OBs give rise to higher total cell numbers, produce more CFUs and maintain a higher percentage of CD34+CD38- cells compared to control culture. Moreover, clonogenic assay and long-term culture results showed that co-culture with OBs induces a strong increase in mono/macrophage precursors coupled to a decrease in the erythroid ones. Finally, gene expression profiling (GEP) allowed us to study which signalling pathways were activated in the hematopoietic cell fraction and in the stromal cell compartment after coculture. Such analysis allowed us to identify several cytokine-receptor networks, such as WNT pathway, and transcription factors, as TWIST1 and FOXC1, that could be activated by co-culture with OBs and could be responsible for the biological effects reported above. Altogether our results indicate that OBs are able to affect HPSCs on 2 different levels: on one side, they increase the immature progenitor pool in vitro, on the other side, they favor the expansion of the mono/macrophage precursors at the expense of the erythroid lineage.
Collapse
Affiliation(s)
- Simona Salati
- Centre for Regenerative Medicine Stefano Ferrari, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Comparative analysis of differentially expressed genes in Sika deer antler at different stages. Mol Biol Rep 2012; 40:1665-76. [DOI: 10.1007/s11033-012-2216-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Accepted: 10/09/2012] [Indexed: 10/27/2022]
|
21
|
Zini R, Norfo R, Ferrari F, Bianchi E, Salati S, Pennucci V, Sacchi G, Carboni C, Ceccherelli GB, Tagliafico E, Ferrari S, Manfredini R. Valproic acid triggers erythro/megakaryocyte lineage decision through induction of GFI1B and MLLT3 expression. Exp Hematol 2012; 40:1043-1054.e6. [PMID: 22885124 DOI: 10.1016/j.exphem.2012.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 08/01/2012] [Accepted: 08/05/2012] [Indexed: 11/28/2022]
Abstract
Histone deacetylase inhibitors represent a family of targeted anticancer compounds that are widely used against hematological malignancies. So far little is known about their effects on normal myelopoiesis. Therefore, in order to investigate the effect of histone deacetylase inhibitors on the myeloid commitment of hematopoietic stem/progenitor cells, we treated CD34(+) cells with valproic acid (VPA). Our results demonstrate that VPA treatment induces H4 histone acetylation and hampers cell cycle progression in CD34(+) cells sustaining high levels of CD34 protein expression. In addition, our data show that VPA treatment promotes erythrocyte and megakaryocyte differentiation. In fact, we demonstrate that VPA treatment is able to induce the expression of growth factor-independent protein 1B (GFI1B) and of mixed-lineage leukemia translocated to chromosome 3 protein (MLLT3), which are crucial regulators of erythrocyte and megakaryocyte differentiation, and that the up-regulation of these genes is mediated by the histone hyperacetylation at their promoter sites. Finally, we show that GFI1B inhibition impairs erythroid and megakaryocyte differentiation induced by VPA, while MLLT3 silencing inhibits megakaryocyte commitment only. As a whole, our data suggest that VPA sustains the expression of stemness-related markers in hematopoietic stem/progenitor cells and is able to interfere with hematopoietic lineage commitment by enhancing erythrocyte and megakaryocyte differentiation and by inhibiting the granulocyte and mono-macrophage maturation.
Collapse
Affiliation(s)
- Roberta Zini
- Centre for Regenerative Medicine, Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Insuasti-Beltran G, Steidler NL, Kang H, Reichard KK. CD34+ megakaryocytes (≥30%) are associated with megaloblastic anaemia and non-acute myeloid neoplasia. Histopathology 2012; 61:694-701. [DOI: 10.1111/j.1365-2559.2012.04259.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
23
|
Purinergic signaling inhibits human acute myeloblastic leukemia cell proliferation, migration, and engraftment in immunodeficient mice. Blood 2011; 119:217-26. [PMID: 22053107 DOI: 10.1182/blood-2011-07-370775] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Extracellular ATP and UTP nucleotides increase the proliferation and engraftment potential of normal human hematopoietic stem cells via the engagement of purinergic receptors (P2Rs). In the present study, we show that ATP and UTP have strikingly opposite effects on human acute myeloblastic leukemia (AML) cells. Leukemic cells express P2Rs. ATP-stimulated leukemic cells, but not normal CD34+ cells, undergo down-regulation of genes involved in cell proliferation and migration, whereas cell-cycle inhibitors are up-regulated. Functionally, ATP induced the inhibition of proliferation and accumulation of AML cells, but not of normal cells, in the G0 phase of the cell cycle. Exposure to ATP or UTP inhibited AML-cell migration in vitro. In vivo, xenotransplantation experiments demonstrated that the homing and engraftment capacity of AML blasts and CD34+CD38- cells to immunodeficient mice BM was significantly inhibited by pretreatment with nucleotides. P2R-expression analysis and pharmacologic profiling suggested that the inhibition of proliferation by ATP was mediated by the down-regulation of the P2X7R, which is up-regulated on untreated blasts, whereas the inhibition of chemotaxis was mainly mediated via P2Y2R and P2Y4R subtypes. We conclude that, unlike normal cells, P2R signaling inhibits leukemic cells and therefore its pharmacologic modulation may represent a novel therapeutic strategy.
Collapse
|
24
|
Tenedini E, Roncaglia E, Ferrari F, Orlandi C, Bianchi E, Bicciato S, Tagliafico E, Ferrari S. Integrated analysis of microRNA and mRNA expression profiles in physiological myelopoiesis: role of hsa-mir-299-5p in CD34+ progenitor cells commitment. Cell Death Dis 2011; 1:e28. [PMID: 21364636 PMCID: PMC3032330 DOI: 10.1038/cddis.2010.5] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Hematopoiesis entails a series of hierarchically organized events that proceed throughout cell specification and terminates with cell differentiation. Commitment needs the transcription factors' effort, which, in concert with microRNAs, drives cell fate and responds to promiscuous patterns of gene expression by turning on lineage-specific genes and repressing alternate lineage transcripts. We obtained microRNA profiles from human CD34+ hematopoietic progenitor cells and in vitro differentiated erythroblasts, megakaryoblasts, monoblasts and myeloblast precursors that we analyzed together with their gene expression profiles. The integrated analysis of microRNA-mRNA expression levels highlighted an inverse correlation between microRNAs specifically upregulated in one single-cell progeny and their putative target genes, which resulted in downregulation. Among the upregulated lineage-enriched microRNAs, hsa-miR-299-5p emerged as having a role in controlling CD34+ progenitor fate, grown in multilineage culture conditions. Gain- and loss-of-function experiments revealed that hsa-miR-299-5p participates in the regulation of hematopoietic progenitor fate, modulating megakaryocytic-granulocytic versus erythroid-monocytic differentiation.
Collapse
Affiliation(s)
- E Tenedini
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Grinstein E, Mahotka C, Borkhardt A. Rb and nucleolin antagonize in controlling human CD34 gene expression. Cell Signal 2011; 23:1358-65. [PMID: 21440621 DOI: 10.1016/j.cellsig.2011.03.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 03/17/2011] [Indexed: 01/12/2023]
Abstract
Retinoblastoma protein (Rb) controls cell proliferation, differentiation, survival and gene expression and it has a central role in the signaling network that provides a cell cycle checkpoint in the G1 phase of the cell cycle. Studies in mice have shown that Rb regulates interactions between hematopoietic stem cells and their bone marrow microenvironment and it acts as a critical regulator of hematopoietic stem and progenitor cells under stress. In human hematopoiesis, the CD34 protein is expressed on a subset of progenitor cells capable of self-renewal, multilineage differentiation, and hematopoietic reconstitution, and CD34 has a role in the differentiation of hematopoietic cells. Here we find that, in CD34-positive hematopoietic cells, Rb controls the human CD34 promoter region by antagonizing the CD34 promoter factor nucleolin to provide a mechanism that links expression of endogenous CD34 to cell cycle progression. Our study suggests a direct involvement of Rb in the transcriptional program of human CD34-positive hematopoietic stem/progenitor cells, thus providing further insights into the molecular network relevant to the features of these cells.
Collapse
Affiliation(s)
- Edgar Grinstein
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Center for Child and Adolescent Health, Heinrich Heine University, Düsseldorf, Germany
| | | | | |
Collapse
|
26
|
Abstract
The c-myb transcription factor is highly expressed in immature hematopoietic cells and down-regulated during differentiation. To define its role during the hematopoietic lineage commitment, we silenced c-myb in human CD34(+) hematopoietic stem/progenitor cells. Noteworthy, c-myb silencing increased the commitment capacity toward the macrophage and megakaryocyte lineages, whereas erythroid differentiation was impaired, as demonstrated by clonogenic assay, morphologic and immunophenotypic data. Gene expression profiling and computational analysis of promoter regions of genes modulated in c-myb-silenced CD34(+) cells identified the transcription factors Kruppel-Like Factor 1 (KLF1) and LIM Domain Only 2 (LMO2) as putative targets, which can account for c-myb knockdown effects. Indeed, chromatin immunoprecipitation and luciferase reporter assay demonstrated that c-myb binds to KLF1 and LMO2 promoters and transactivates their expression. Consistently, the retroviral vector-mediated overexpression of either KLF1 or LMO2 partially rescued the defect in erythropoiesis caused by c-myb silencing, whereas only KLF1 was also able to repress the megakaryocyte differentiation enhanced in Myb-silenced CD34(+) cells. Our data collectively demonstrate that c-myb plays a pivotal role in human primary hematopoietic stem/progenitor cells lineage commitment, by enhancing erythropoiesis at the expense of megakaryocyte diffentiation. Indeed, we identified KLF1 and LMO2 transactivation as the molecular mechanism underlying Myb-driven erythroid versus megakaryocyte cell fate decision.
Collapse
|
27
|
Bonanno G, Mariotti A, Procoli A, Corallo M, Scambia G, Pierelli L, Rutella S. Interleukin-21 induces the differentiation of human umbilical cord blood CD34-lineage- cells into pseudomature lytic NK cells. BMC Immunol 2009; 10:46. [PMID: 19712464 PMCID: PMC2743656 DOI: 10.1186/1471-2172-10-46] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2009] [Accepted: 08/27/2009] [Indexed: 12/28/2022] Open
Abstract
Background Umbilical cord blood (UCB) is enriched with transplantable CD34+ cells. In addition to CD34-expressing haematopoietic stem cells (HSC), human UCB contains a rare population of CD34-lineage- cells endowed with the ability to differentiate along the T/NK pathway in response to interleukin (IL)-15 and a stromal cell support. IL-21 is a crucial regulator of NK cell function, whose influence on IL-15-induced differentiation of CD34-lineage- cells has not been investigated previously. The present study was designed and conducted to address whether IL-21 might replace the stromal cell requirements and foster the IL-15-induced NK differentiation of human UCB CD34-lineage- cells. Results CD34-lineage- cells were maintained in liquid culture with Flt3-L and SCF, with the addition of IL-15 and IL-21, either alone or in combination. Cultures were established in the absence of feeder cells or serum supplementation. Cytokine-treated cells were used to evaluate cell surface phenotype, expression of molecular determinants of lymphoid/NK cell differentiation, secretion of IFN-γ, GM-CSF, TNF-α and CCL3/MIP-1α, and cytolytic activity against NK-sensitive tumour cell targets. CD34-lineage- cells proliferated vigorously in response to IL-15 and IL-21 but not to IL-21 alone, and up-regulated phosphorylated Stat1 and Stat3 proteins. CD34-lineage- cells expanded by IL-21 in combination with IL-15 acquired lymphoid morphology and killer-cell immunoglobulin-like receptor (KIR)-CD56+CD16-/+ phenotype, consistent with pseudo-mature NK cells. IL-21/IL-15-differentiated cells expressed high levels of mRNA for Bcl-2, GATA-3 and Id2, a master switch required for NK-cell development, and harboured un-rearranged TCRγ genes. From a functional standpoint, IL-21/IL-15-treated cells secreted copious amounts of IFN-γ, GM-CSF and CCL3/MIP-1α, and expressed cell surface CD107a upon contact with NK-sensitive tumour targets, a measure of exocytosis of NK secretory granules. Conclusion This study underpins a novel role for IL-21 in the differentiation of pseudo-mature lytic NK cells in a synergistic context with IL-15, and identifies a potential strategy to expand functional NK cells for immunotherapy.
Collapse
Affiliation(s)
- Giuseppina Bonanno
- Department of Gynaecology, Catholic University Medical School, Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
28
|
da Silva CL, Gonçalves R, Porada CD, Ascensão JL, Zanjani ED, Cabral JMS, Almeida-Porada G. Differences amid bone marrow and cord blood hematopoietic stem/progenitor cell division kinetics. J Cell Physiol 2009; 220:102-11. [PMID: 19277981 DOI: 10.1002/jcp.21736] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Human hematopoietic stem/progenitor cells (HSC) isolated based upon specific patterns of CD34 and CD38 expression, despite phenotypically identical, were found to be functionally heterogeneous, raising the possibility that reversible expression of these antigens may occur during cellular activation and/or proliferation. In these studies, we combined PKH67 tracking with CD34/CD38 immunostaining to compare cell division kinetics between human bone marrow (BM) and cord blood (CB)-derived HSC expanded in a serum-free/stromal-based system for 14 days (d), and correlated CD34 and CD38 expression with the cell divisional history. CB cells began dividing 24 h earlier than BM cells, and significantly higher numbers underwent mitosis during the time in culture. By d10, over 55% of the CB-cells reached the ninth generation, whereas BM-cells were mostly distributed between the fifth and seventh generation. By d14, all CB cells had undergone multiple cell divisions, while 0.7-3.8% of BM CD34(+) cells remained quiescent. Furthermore, the percentage of BM cells expressing CD34 decreased from 60.8 +/- 6.3% to 30.6 +/- 6.7% prior to initiating division, suggesting that downmodulation of this antigen occurred before commencement of proliferation. Moreover, with BM, all primitive CD34(+)CD38(-) cells present at the end of culture arose from proliferating CD34(+)CD38(+) cells that downregulated CD38 expression, while in CB, a CD34(+)CD38(-) population was maintained throughout culture. These studies show that BM and CB cells differ significantly in cell division kinetics and expression of CD34 and CD38, and that the inherent modulation of these antigens during ex vivo expansion may lead to erroneous quantification of the stem cell content of the expanded graft.
Collapse
|