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: 7] [Impact Index Per Article: 3.5] [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
|
Simulated microgravity affects stroma-dependent ex vivo myelopoiesis. Tissue Cell 2023; 80:101987. [PMID: 36481580 DOI: 10.1016/j.tice.2022.101987] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 11/25/2022]
Abstract
Microgravity is known negatively affect physiology of living beings, including hematopoiesis. Dysregulation of hematopoietic cells and supporting stroma relationships in bone marrow niche may be in charge. We compared the efficacy of ex vivo expansion of hematopoietic stem and progenitor cells (HSPCs) in presence of native or osteocommitted MSCs under simulated microgravity (Smg) using Random Positioning Machine (RPM). In comparison with 1 g, a decrease of MSC-associated HSPCs and an increase of floating HSPCs was observed after 7 days of Smg exposure. Among floating HSPCs, primitive progenitors were presented by late CD34+/133-. Total CFUs as well as erythroid (BFU-E) and granulocytic (CFU-G) numbers were lower. MSC-associated primitive HSPCs demonstrated increased proportion of late CD34+/133- in expense of early CD34-/133+. Osteo-MSCs preferentially supported late primitive CD34+ and more committed HSPCs as followed from increase of CFUs, and CD235a+ erythroid progenitors. Under Smg, an increased VEGF, eotaxin, and GRO-a levels, and a decrease in RANTES were found in the osteo-MSC-HSPC co-cultures. IL-6,-8, -13, G-CSF, GRO-a, MCP-3, MIP-1b, VEGF increased in co-culture with osteo-MSCs vs intact MSCs. Based on the findings, the misbalance between primitive/committed HSPCs and a decrease in hematopoiesis-supportive activity of osteocommitted cells are supposed to underline hematopoietic disorders during space flights.
Collapse
|
3
|
Urao N, Liu J, Takahashi K, Ganesh G. Hematopoietic Stem Cells in Wound Healing Response. Adv Wound Care (New Rochelle) 2022; 11:598-621. [PMID: 34353116 PMCID: PMC9419985 DOI: 10.1089/wound.2021.0065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Significance: Emerging evidence has shown a link between the status of hematopoietic stem cells (HSCs) and wound healing responses. Thus, better understanding HSCs will contribute to further advances in wound healing research. Recent Advances: Myeloid cells such as neutrophils and monocyte-derived macrophages are critical players in the process of wound healing. HSCs actively respond to wound injury and other tissue insults, including infection and produce the effector myeloid cells, and a failing of the HSC response can result in impaired wound healing. Technological advances such as transcriptome at single-cell resolution, epigenetics, three-dimensional imaging, transgenic animals, and animal models, have provided novel concepts of myeloid generation (myelopoiesis) from HSCs, and have revealed cell-intrinsic and -extrinsic mechanisms that can impact HSC functions in the context of health conditions. Critical Issues: The newer concepts include-the programmed cellular fate at a differentiation stage that is used to be considered as the multilineage, the signaling pathways that can activate HSCs directly and indirectly, the mechanisms that can deteriorate HSCs, the roles and remodeling of the surrounding environment for HSCs and their progenitors (the niche). Future Directions: The researches on HSCs, which produce blood cells, should contribute to the development of blood biomarkers predicting a risk of chronic wounds, which may transform clinical practice of wound care with precision medicine for patients at high risk of poor healing.
Collapse
Affiliation(s)
- Norifumi Urao
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, New York, USA.,Correspondence: Department of Pharmacology, State University of New York Upstate Medical University, 766 Irving Avenue, Weiskotten Hall Room 5322, Syracuse, NY 13210, USA.
| | - Jinghua Liu
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Kentaro Takahashi
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Gayathri Ganesh
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
4
|
New Perspectives to Improve Mesenchymal Stem Cell Therapies for Drug-Induced Liver Injury. Int J Mol Sci 2022; 23:ijms23052669. [PMID: 35269830 PMCID: PMC8910533 DOI: 10.3390/ijms23052669] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 02/06/2023] Open
Abstract
Drug-induced liver injury (DILI) is one of the leading causes of acute liver injury. Many factors may contribute to the susceptibility of patients to this condition, making DILI a global medical problem that has an impact on public health and the pharmaceutical industry. The use of mesenchymal stem cells (MSCs) has been at the forefront of regenerative medicine therapies for many years, including MSCs for the treatment of liver diseases. However, there is currently a huge gap between these experimental approaches and their application in clinical practice. In this concise review, we focus on the pathophysiology of DILI and highlight new experimental approaches conceived to improve cell-based therapy by the in vitro preconditioning of MSCs and/or the use of cell-free products as treatment for this liver condition. Finally, we discuss the advantages of new approaches, but also the current challenges that must be addressed in order to develop safer and more effective procedures that will allow cell-based therapies to reach clinical practice, enhancing the quality of life and prolonging the survival time of patients with DILI.
Collapse
|
5
|
Rebuilding the hematopoietic stem cell niche: Recent developments and future prospects. Acta Biomater 2021; 132:129-148. [PMID: 33813090 DOI: 10.1016/j.actbio.2021.03.061] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 12/20/2022]
Abstract
Hematopoietic stem cells (HSCs) have proven their clinical relevance in stem cell transplantation to cure patients with hematological disorders. Key to their regenerative potential is their natural microenvironment - their niche - in the bone marrow (BM). Developments in the field of biomaterials enable the recreation of such environments with increasing preciseness in the laboratory. Such artificial niches help to gain a fundamental understanding of the biophysical and biochemical processes underlying the interaction of HSCs with the materials in their environment and the disturbance of this interplay during diseases affecting the BM. Artificial niches also have the potential to multiply HSCs in vitro, to enable the targeted differentiation of HSCs into mature blood cells or to serve as drug-testing platforms. In this review, we will introduce the importance of artificial niches followed by the biology and biophysics of the natural archetype. We will outline how 2D biomaterials can be used to dissect the complexity of the natural niche into individual parameters for fundamental research and how 3D systems evolved from them. We will present commonly used biomaterials for HSC research and their applications. Finally, we will highlight two areas in the field of HSC research, which just started to unlock the possibilities provided by novel biomaterials, in vitro blood production and studying the pathophysiology of the niche in vitro. With these contents, the review aims to give a broad overview of the different biomaterials applied for HSC research and to discuss their potentials, challenges and future directions in the field. STATEMENT OF SIGNIFICANCE: Hematopoietic stem cells (HSCs) are multipotent cells responsible for maintaining the turnover of all blood cells. They are routinely applied to treat patients with hematological diseases. This high clinical relevance explains the necessity of multiplication or differentiation of HSCs in the laboratory, which is hampered by the missing natural microenvironment - the so called niche. Biomaterials offer the possibility to mimic the niche and thus overcome this hurdle. The review introduces the HSC niche in the bone marrow and discusses the utility of biomaterials in creating artificial niches. It outlines how 2D systems evolved into sophisticated 3D platforms, which opened the gateway to applications such as, expansion of clinically relevant HSCs, in vitro blood production, studying niche pathologies and drug testing.
Collapse
|
6
|
Nlrp3 Inflammasome Signaling Regulates the Homing and Engraftment of Hematopoietic Stem Cells (HSPCs) by Enhancing Incorporation of CXCR4 Receptor into Membrane Lipid Rafts. Stem Cell Rev Rep 2021; 16:954-967. [PMID: 32661868 PMCID: PMC7456406 DOI: 10.1007/s12015-020-10005-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Fast and efficient homing and engraftment of hematopoietic stem progenitor cells (HSPCs) is crucial for positive clinical outcomes from transplantation. We found that this process depends on activation of the Nlrp3 inflammasome, both in the HSPCs to be transplanted and in the cells in the recipient bone marrow (BM) microenvironment. For the first time we provide evidence that functional deficiency in the Nlrp3 inflammasome in transplanted cells or in the host microenvironment leads to defective homing and engraftment. At the molecular level, functional deficiency of the Nlrp3 inflammasome in HSPCs leads to their defective migration in response to the major BM homing chemoattractant stromal-derived factor 1 (SDF-1) and to other supportive chemoattractants, including sphingosine-1-phosphate (S1P) and extracellular adenosine triphosphate (eATP). We report that activation of the Nlrp3 inflammasome increases autocrine release of eATP, which promotes incorporation of the CXCR4 receptor into membrane lipid rafts at the leading surface of migrating cells. On the other hand, a lack of Nlrp3 inflammasome expression in BM conditioned for transplantation leads to a decrease in expression of SDF-1 and danger-associated molecular pattern molecules (DAMPs), which are responsible for activation of the complement cascade (ComC), which in turn facilitates the homing and engraftment of HSPCs.
Collapse
|
7
|
Bessy T, Itkin T, Passaro D. Bioengineering the Bone Marrow Vascular Niche. Front Cell Dev Biol 2021; 9:645496. [PMID: 33996805 PMCID: PMC8113773 DOI: 10.3389/fcell.2021.645496] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/23/2021] [Indexed: 01/01/2023] Open
Abstract
The bone marrow (BM) tissue is the main physiological site for adult hematopoiesis. In recent years, the cellular and matrix components composing the BM have been defined with unprecedent resolution, both at the molecular and structural levels. With the expansion of this knowledge, the possibility of reproducing a BM-like structure, to ectopically support and study hematopoiesis, becomes a reality. A number of experimental systems have been implemented and have displayed the feasibility of bioengineering BM tissues, supported by cells of mesenchymal origin. Despite being known as an abundant component of the BM, the vasculature has been largely disregarded for its role in regulating tissue formation, organization and determination. Recent reports have highlighted the crucial role for vascular endothelial cells in shaping tissue development and supporting steady state, emergency and malignant hematopoiesis, both pre- and postnatally. Herein, we review the field of BM-tissue bioengineering with a particular focus on vascular system implementation and integration, starting from describing a variety of applicable in vitro models, ending up with in vivo preclinical models. Additionally, we highlight the challenges of the field and discuss the clinical perspectives in terms of adoptive transfer of vascularized BM-niche grafts in patients to support recovering hematopoiesis.
Collapse
Affiliation(s)
- Thomas Bessy
- Leukemia and Niche Dynamics Laboratory, Université de Paris, Institut Cochin, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Paris, France
| | - Tomer Itkin
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Diana Passaro
- Leukemia and Niche Dynamics Laboratory, Université de Paris, Institut Cochin, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Paris, France
| |
Collapse
|
8
|
Ye J, Tang X, Long Y, Chu Z, Zhou Q, Lin B. The effect of hypoxia on the proliferation capacity of dermal papilla cell by regulating lactate dehydrogenase. J Cosmet Dermatol 2020; 20:684-690. [PMID: 32599679 PMCID: PMC7891641 DOI: 10.1111/jocd.13578] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/20/2022]
Abstract
Hypoxia is of great significance for stem cells to maintain the proliferation and differentiation capacity. As a specialized mesenchymal component of the hair follicle (HF), the dermal papilla cell (DPC) not only regulates HF cycle, but also plays a pivotal role in differentiating hair follicle stem cell(HFSC) into HF. However, whether hypoxia could affect DPCs on proliferation or metabolism remains unclear. In our study, DPCs were cultured in normoxia (20%O2) or hypoxia (5%O2). Cell viability assays were performed, and lactate dehydrogenase (LDH) activity and lactate level in DPCs were detected. After that, LDH was overexpressed or knocked down in DPCs; then, the expression of protein markers (ALP, Ki‐67) was assessed by Western blotting, and cell proliferation was also detected after overexpression or knockdown of LDH. Hypoxia did show positive effect on proliferation of DPCs. The LDH activity of DPCs cultured under hypoxic condition was significantly higher than that of cultured under normoxic condition. Overexpression of LDH significantly up‐regulates the expression of ALP and Ki‐67 compared with knockdown and negative control. Cell proliferation was also promoted in DPCs with elevated LDH. Our findings showed that the proliferation activity of DPCs could be stimulated under hypoxia. Meanwhile, LDH plays an important role in maintaining the activity of DPCs in hypoxic condition.
Collapse
Affiliation(s)
- Jun Ye
- Department of emergency surgery, The affiliated ZhuZhou Hospital of Xiangya School of Medicine, CSU, ZhuZhou, China
| | - Xiaoli Tang
- Department of emergency surgery, The affiliated ZhuZhou Hospital of Xiangya School of Medicine, CSU, ZhuZhou, China
| | - Yunzhu Long
- Infectious Department, The affiliated ZhuZhou Hospital of Xiangya School of Medicine, CSU, ZhuZhou, China
| | - Zhou Chu
- Department of public health, The affiliated ZhuZhou Hospital of Xiangya School of Medicine, CSU, ZhuZhou, China
| | - Qing Zhou
- Infectious Department, The affiliated ZhuZhou Hospital of Xiangya School of Medicine, CSU, ZhuZhou, China
| | - Bojie Lin
- Department of Plastic and Aesthetic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
9
|
Andreeva E, Andrianova I, Bobyleva P, Gornostaeva A, Ezdakova M, Golikova E, Buravkova L. Adipose tissue-derived stromal cells retain immunosuppressive and angiogenic activity after coculture with cord blood hematopoietic precursors. Eur J Cell Biol 2020; 99:151069. [PMID: 31982141 DOI: 10.1016/j.ejcb.2020.151069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/25/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
Adipose-tissue derived stromal cells (ASCs) are currently considered as a full value alternative source of bone marrow MSCs for prevention of graft-versus-host disease (GVHD) after hematopoietic stem cell transplantation due to their immunosuppressive potential. Besides, ASCs are known to support ex vivo expansion of hematopoietic stem and progenitor cells (HSPCs). Ex vivo expansion enables to amplify significantly the number of HSPCs of different commitment. Mononuclear cells (MNCs) from cord blood (cb) contain HSPCs and are easily assessed. The rarity of those HSPCs is a serious limitation of its application in cell therapy. Here we expanded cbMNCs in stroma-dependent setting to generate heterocellular associates consisting of ASCs and undifferentiated and low committed hematopoietic cbHSPCs. A part of cbHSPCs in associates demonstrated a primitive phenotype confirmed by formation of "cobblestone areas". ASCs associated with cbHSPCs demonstrated up-regulation of immunosuppressive indoleamine 2,3-dioxygenase (IDO), leukemia inhibitory factor (LIF), cyclooxygenase-2 (PTGS2) genes. ASC-cbHSPCs as well as ASCs provoked the suppression of HLA-DR activation and apoptosis of mitogen-stimulated T cells. VEGF transcription and secretion were elevated providing stimulation of blood vessel formation in ovo. Thus, ASCs retain immunosuppressive and proangiogenic capacities evidencing "third party" potential along with the effective support of ex vivo expansion of cbHSPCs. Above functions expand the relevance of ASCs for needs of regenerative medicine.
Collapse
Affiliation(s)
- Elena Andreeva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye Shosse, 76a, 123007 Moscow, Russia.
| | - Irina Andrianova
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye Shosse, 76a, 123007 Moscow, Russia
| | - Polina Bobyleva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye Shosse, 76a, 123007 Moscow, Russia; Faculty of Basic Medicine, Moscow State University, Lomonosovsky Prospekt, 31-5, 117192, Moscow, Russia
| | - Aleksandra Gornostaeva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye Shosse, 76a, 123007 Moscow, Russia
| | - Maria Ezdakova
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye Shosse, 76a, 123007 Moscow, Russia
| | - Ekaterina Golikova
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye Shosse, 76a, 123007 Moscow, Russia
| | - Ludmila Buravkova
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye Shosse, 76a, 123007 Moscow, Russia; Faculty of Basic Medicine, Moscow State University, Lomonosovsky Prospekt, 31-5, 117192, Moscow, Russia
| |
Collapse
|
10
|
Lu Z, Jiang X, Chen M, Feng L, Kang YJ. An oxygen-releasing device to improve the survival of mesenchymal stem cells in tissue engineering. Biofabrication 2019; 11:045012. [PMID: 31315098 DOI: 10.1088/1758-5090/ab332a] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Supplying oxygen to inner areas of cell constructs to support cell proliferation and metabolism is a major challenge in tissue engineering involving stem cells. Developing devices that incorporate oxygen release materials to increase the availability of the localized oxygen supply is therefore key to addressing this limitation. Herein, we designed and developed a 3D-printed oxygen-releasing device composed of an alginate hydrogel scaffold combined with an oxygen-generating biomaterial (calcium peroxide) to improve the oxygen supply of the microenvironment for culturing adipose tissue-derived stem cells. The results demonstrated that the 3D-printed oxygen-releasing device alleviated hypoxia, maintained oxygen availability, and ensured proliferation of the embedded cells, whilst also reducing hypoxia-induced apoptosis. The introduction of this 3D-printed oxygen-releasing device could enhance the survival of embedded stem cells.
Collapse
|
11
|
Tissue "Hypoxia" and the Maintenance of Leukemia Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1143:129-145. [PMID: 31338818 DOI: 10.1007/978-981-13-7342-8_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The relationship of the homing of normal hematopoietic stem cells (HSC) in the bone marrow to specific environmental conditions, referred to as the stem cell niche (SCN), has been intensively studied over the last three decades. These conditions include the action of a number of molecular and cellular players, as well as critical levels of nutrients, oxygen and glucose in particular, involved in energy production. These factors are likely to act also in leukemias, due to the strict analogy between the hierarchical structure of normal hematopoietic cell populations and that of leukemia cell populations. This led to propose that leukemic growth is fostered by cells endowed with stem cell properties, the leukemia stem cells (LSC), a concept readily extended to comprise the cancer stem cells (CSC) of solid tumors. Two alternative routes have been proposed for CSC generation, that is, the oncogenic staminalization (acquisition of self-renewal) of a normal progenitor cell (the "CSC in normal progenitor cell" model) and the oncogenic transformation of a normal (self-renewing) stem cell (the "CSC in normal stem cell" model). The latter mechanism, in the hematological context, makes LSC derive from HSC, suggesting that LSC share SCN homing with HSC. This chapter is focused on the availability of oxygen and glucose in the regulation of LSC maintenance within the SCN. In this respect, the most critical aspect in view of the outcome of therapy is the long-term maintenance of the LSC subset capable to sustain minimal residual disease and the related risk of relapse of disease.
Collapse
|
12
|
Andreeva E, Andrianova I, Sotnezova E, Gornostaeva A, Khorkova S, Buravkova L. Hematopoiesis-supportive function of growth-arrested human adipose-tissue stromal cells under physiological hypoxia. J Biosci Bioeng 2018; 127:647-654. [PMID: 30503171 DOI: 10.1016/j.jbiosc.2018.10.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/05/2018] [Accepted: 10/15/2018] [Indexed: 12/22/2022]
Abstract
Ex vivo expansion of hematopoietic progenitors is considered as an attractive tool to increase the number of stem and progenitor cells (HSPCs) for cell therapy. The efficacy of ex vivo expansion is strongly depends on the feeder cell activity to mimic hematopoietic microenvironment. Here we demonstrated, that combination of mitomycin C-induced growth arrest and tissue-related O2 (physiological hypoxia) modulated stromal capacity of adipose tissue derived stromal cells (ASCs). Growth arrest did not affect viability, stromal phenotype and multilineage potential of ASCs permanently expanded at tissue-related O2. Meanwhile, the PCR analysis revealed an up-regulation of genes, encoded molecules of cell-cell (ICAM1, HCAM/CD44) and cell-matrix adhesion (ITGs), extracellular matrix production (COLs) and remodeling (MMPs, HAS1) in growth-arrested ASCs at physiological hypoxia in comparison with ambient O2 (20%). The number of ICAM-1 positive ASCs was increased under low O2 as well. These alterations contributed into the ex vivo expansion of cord blood HSPCs providing the preferential production of primitive HSPCs. The number of cobblestone area forming cell (CAFC) colonies was 1.5-fold higher at physiological hypoxia (p < 0.05). CAFCs considered as long-term culture-initiating cells (LTC-IC) known to support long-term hematopoiesis restoration in vivo. The presented data may be applicable in the development of upscale protocols of HSPC expansion.
Collapse
Affiliation(s)
- Elena Andreeva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye shosse, 76a, 123007 Moscow, Russia.
| | - Irina Andrianova
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye shosse, 76a, 123007 Moscow, Russia
| | - Elena Sotnezova
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye shosse, 76a, 123007 Moscow, Russia
| | - Aleksandra Gornostaeva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye shosse, 76a, 123007 Moscow, Russia
| | - Svetlana Khorkova
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye shosse, 76a, 123007 Moscow, Russia
| | - Ludmila Buravkova
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye shosse, 76a, 123007 Moscow, Russia; Faculty of Basic Medicine, Moscow State University, Lomonosovsky Prospekt, 31-5, 117192 Moscow, Russia
| |
Collapse
|
13
|
Bahsoun S, Coopman K, Forsyth NR, Akam EC. The Role of Dissolved Oxygen Levels on Human Mesenchymal Stem Cell Culture Success, Regulatory Compliance, and Therapeutic Potential. Stem Cells Dev 2018; 27:1303-1321. [DOI: 10.1089/scd.2017.0291] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Soukaina Bahsoun
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Karen Coopman
- Centre for Biological Engineering, Loughborough University, Loughborough, United Kingdom
| | - Nicholas R. Forsyth
- Guy Hilton Research Centre, Institute for Science and Technology in Medicine, Keele University, Keele, United Kingdom
| | - Elizabeth C. Akam
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| |
Collapse
|
14
|
Buravkova LB, Andreeva ER, Lobanova MV, Cotnezova EV, Grigoriev AI. The Differential Expression of Adhesion Molecule and Extracellular Matrix Genes in Mesenchymal Stromal Cells after Interaction with Cord Blood Hematopoietic Progenitors. DOKL BIOCHEM BIOPHYS 2018; 479:69-71. [DOI: 10.1134/s1607672918020047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Indexed: 12/18/2022]
|
15
|
Irigoyen M, García-Ruiz JC, Berra E. The hypoxia signalling pathway in haematological malignancies. Oncotarget 2018; 8:36832-36844. [PMID: 28415662 PMCID: PMC5482702 DOI: 10.18632/oncotarget.15981] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/27/2017] [Indexed: 12/25/2022] Open
Abstract
Haematological malignancies are tumours that affect the haematopoietic and the lymphatic systems. Despite the huge efforts to eradicate these tumours, the percentage of patients suffering resistance to therapies and relapse still remains significant. The tumour environment favours drug resistance of cancer cells, and particularly of cancer stem/initiating cells. Hypoxia promotes aggressiveness, metastatic spread and relapse in most of the solid tumours. Furthermore, hypoxia is associated with worse prognosis and resistance to conventional treatments through activation of the hypoxia-inducible factors. Haematological malignancies are not considered solid tumours, and therefore, the role of hypoxia in these diseases was initially presumed to be inconsequential. However, hypoxia is a hallmark of the haematopoietic niche. Here, we will review the current understanding of the role of both hypoxia and hypoxia-inducible factors in different haematological tumours.
Collapse
Affiliation(s)
- Marta Irigoyen
- Centro de Investigación Cooperativa en Biociencias CIC bioGUNE, Derio, Spain
| | - Juan Carlos García-Ruiz
- Servicio de Hematología y Hemoterapia, BioCruces Health Research Institute, Hospital Universitario Cruces, Spain
| | - Edurne Berra
- Centro de Investigación Cooperativa en Biociencias CIC bioGUNE, Derio, Spain
| |
Collapse
|
16
|
Chen YY, Wu CL, Shen SH. High Signal in Bone Marrow on Diffusion-Weighted Imaging of Female Pelvis: Correlation With Anemia and Fibroid-Associated Symptoms. J Magn Reson Imaging 2018; 48:1024-1033. [PMID: 29504179 DOI: 10.1002/jmri.26002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 02/15/2018] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The diffusion-weighted imaging (DWI) signals of the female pelvic bone marrow show great variability and are usually high in female patients with fibroid-associated symptoms and anemia. PURPOSE To ascertain clinical factors contributing to high signal intensity in the bone marrow of the female pelvis on DWI. STUDY TYPE Retrospective case-control study. SUBJECTS A single-institution review of 221 female patients underwent a pelvic magnetic resonance study from December 2012 to July 2014. FIELD STRENGTH/SEQUENCE 1.5T/DWI (b = 0 and 1000) and apparent diffusion coefficient (ADC). ASSESSMENT The ADC of pelvic bone marrow and the muscle-normalized signal intensity (SI) on DWI (mnDWI) were measured. A brightness grading scale ranging from 0 to 4 was used for pelvic bone assessment. Clinical factors, namely, age, the lowest hemoglobin level in the last 6 months, the presence of large uterine fibroids, and/or adenomyosis and fibroid-associated symptoms were recorded. STATISTICAL TESTS The relationships between the brightness grade and clinical factors were evaluated through multinomial logistic regression, and correlations of mnDWI and the ADC with the clinical factors were analyzed through the Kruskal-Wallis test, Jonckheere's trend test, and the Mann-Whitney U-test with Bonferroni correction. RESULTS Age and the hemoglobin level were inversely associated with the bone marrow brightness grade on DWI (both P < 0.05), whereas the presence of fibroid-associated symptoms showed a positive association (P = 0.028). The ADC and mnDWI in women younger than 50 years were significantly higher than those in older women (both P < 0.0001). The ADC had no significant correlation with anemia (P = 0.511), whereas mnDWI increased as the severity of anemia increased (P = 0.00154). DATA CONCLUSION Our study showed an association of high DWI SI of pelvic bone marrow with anemia in premenopausal women. LEVEL OF EVIDENCE 4 Technical Efficacy Stage 3 J. Magn. Reson. Imaging 2018;48:1024-1033.
Collapse
Affiliation(s)
- Ying-Yuan Chen
- Department of Radiology, National Yang-Ming University Hospital, Yilan County, Taiwan, ROC.,School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Ching-Lan Wu
- Department of Radiology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC.,School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Shu-Huei Shen
- Department of Radiology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC.,School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| |
Collapse
|
17
|
Tiwari A, Wong CS, Nekkanti LP, Deane JA, McDonald C, Li J, Pham Y, Sutherland AE, Jenkin G, Kirkland MA. Controlling the Effective Oxygen Tension Experienced by Cells Using a Dynamic Culture Technique for Hematopoietic Ex Vivo Expansion. ACTA ACUST UNITED AC 2018; 44:2A.11.1-2A.11.13. [PMID: 29512129 DOI: 10.1002/cpsc.42] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Clinical hematopoietic stem/progenitor cell (HSPC) transplantation outcomes are strongly correlated with the number of cells infused. Hence, to generate sufficient HSPCs for transplantation, the best culture parameters for expansion are critical. It is generally assumed that the defined oxygen (O2 ) set for the incubator reflects the pericellular O2 to which cells are being exposed. Studies have shown that low O2 tension maintains an undifferentiated state, but the expansion rate may be constrained because of limited diffusion in a static culture system. A combination of low ambient O2 and dynamic culture conditions has been developed to increase the reconstituting capacity of human HSPCs. In this unit, the protocols for serum-free expansion of HSPCs at 5% and 20% O2 in static and dynamic nutrient flow mode are described. Finally, the impact of O2 tension on HSPC expansion in vitro by flow cytometry and colony forming assays and in vivo through engraftment using a murine model is assessed. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Abhilasha Tiwari
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Cynthia S Wong
- Institute for Frontier Materials, Deakin University, Geelong, Victoria, Australia
| | - Lakshmi P Nekkanti
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - James A Deane
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Courtney McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Jingang Li
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Yen Pham
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Amy E Sutherland
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Department of Obstetrics & Gynaecology, Southern Clinical School, Monash University, Clayton, Victoria, Australia
| | - Mark A Kirkland
- Institute for Frontier Materials, Deakin University, Geelong, Victoria, Australia
| |
Collapse
|
18
|
Pathogen reduction through additive-free short-wave UV light irradiation retains the optimal efficacy of human platelet lysate for the expansion of human bone marrow mesenchymal stem cells. PLoS One 2017; 12:e0181406. [PMID: 28763452 PMCID: PMC5538655 DOI: 10.1371/journal.pone.0181406] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 06/30/2017] [Indexed: 12/17/2022] Open
Abstract
Background We recently developed and characterized a standardized and clinical grade human Platelet Lysate (hPL) that constitutes an advantageous substitute for fetal bovine serum (FBS) for human mesenchymal stem cell (hMSC) expansion required in cell therapy procedures, avoiding xenogenic risks (virological and immunological) and ethical issues. Because of the progressive use of pathogen-reduced (PR) labile blood components, and the requirement of ensuring the viral safety of raw materials for cell therapy products, we evaluated the impact of the novel procedure known as THERAFLEX UV-Platelets for pathogen reduction on hPL quality (growth factors content) and efficacy (as a medium supplement for hMSC expansion). This technology is based on short-wave ultraviolet light (UV-C) that induces non-reversible damages in DNA and RNA of pathogens while preserving protein structures and functions, and has the main advantage of not needing the addition of any photosensitizing additives (that might secondarily interfere with hMSCs). Methodology / Principal findings We applied the THERAFLEX UV-Platelets procedure on fresh platelet concentrates (PCs) suspended in platelet additive solution and prepared hPL from these treated PCs. We compared the quality and efficacy of PR-hPL with the corresponding non-PR ones. We found no impact on the content of five cytokines tested (EGF, bFGF, PDGF-AB, VEGF and IGF-1) but a significant decrease in TGF-ß1 (-21%, n = 11, p<0.01). We performed large-scale culture of hMSCs from bone marrow (BM) during three passages and showed that hPL or PR-hPL at 8% triggered comparable BM-hMSC proliferation as FBS at 10% plus bFGF. Moreover, after proliferation of hMSCs in an hPL- or PR-hPL-containing medium, their profile of membrane marker expression, their clonogenic potential and immunosuppressive properties were maintained, in comparison with BM-hMSCs cultured under FBS conditions. The potential to differentiate towards the adipogenic and osteogenic lineages of hMSCs cultured in parallel in the three conditions also remained identical. Conclusion / Significance We demonstrated the feasibility of using UV-C-treated platelets to subsequently obtain pathogen-reduced hPL, while preserving its optimal quality and efficacy for hMSC expansion in cell therapy applications.
Collapse
|
19
|
Mesenchymal Stem Cells Transplantation following Partial Hepatectomy: A New Concept to Promote Liver Regeneration-Systematic Review of the Literature Focused on Experimental Studies in Rodent Models. Stem Cells Int 2017; 2017:7567958. [PMID: 28386285 PMCID: PMC5366767 DOI: 10.1155/2017/7567958] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 02/09/2017] [Accepted: 02/14/2017] [Indexed: 02/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are an attractive source for regenerative medicine because they are easily accessible through minimally invasive methods and have the potential to enhance liver regeneration (LG) and improve liver function, following partial hepatectomy (PH) and acute or chronic liver injury. A systematic review of the literature was conducted for articles published up to September 1st, 2016, using the MEDLINE database. The keywords that were used in various combinations were as follows: “Mesenchymal stem cells”, “transplantation”, “stem cells”, “adipose tissue derived stem cells”, “bone marrow-derived stem cells”, “partial hepatectomy”, “acute liver failure”, “chronic liver failure”, “liver fibrosis”, “liver cirrhosis”, “rats”, “mice”, and “liver regeneration”. All introduced keywords were searched for separately in MeSH Database to control relevance and terminological accuracy and validity. A total of 41 articles were identified for potential inclusion and reviewed in detail. After a strict selection process, a total of 28 articles were excluded, leaving 13 articles to form the basis of this systematic review. MSCs transplantation promoted LG and improved liver function. Furthermore, MSCs had the ability to differentiate in hepatocyte-like cells, increase survival, and protect hepatocytes by paracrine mechanisms. MSCs transplantation may provide beneficial effects in the process of LG after PH and acute or chronic liver injury. They may represent a new therapeutic option to treat posthepatectomy acute liver failure.
Collapse
|
20
|
Wahl EA, Schenck TL, Machens HG, Balmayor ER. VEGF released by deferoxamine preconditioned mesenchymal stem cells seeded on collagen-GAG substrates enhances neovascularization. Sci Rep 2016; 6:36879. [PMID: 27830734 PMCID: PMC5103276 DOI: 10.1038/srep36879] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/24/2016] [Indexed: 01/01/2023] Open
Abstract
Hypoxia preconditioning of mesenchymal stem cells (MSCs) has been shown to promote wound healing through HIF-1α stabilization. Preconditioned MSCs can be applied to three-dimensional biomaterials to further enhance the regenerative properties. While environmentally induced hypoxia has proven difficult in clinical settings, this study compares the wound healing capabilities of adipose derived (Ad) MSCs seeded on a collagen-glycosaminoglycan (GAG) dermal substrate exposed to either environmental hypoxia or FDA approved deferoxamine mesylate (DFO) to stabilize HIF-1α for wound healing. The release of hypoxia related reparative factors by the cells on the collagen-GAG substrate was evaluated to detect if DFO produces results comparable to environmentally induced hypoxia to facilitate optimal clinical settings. VEGF release increased in samples exposed to DFO. While the SDF-1α release was lower in cells exposed to environmental hypoxia in comparison to cells cultured in DFO in vitro. The AdMSC seeded biomaterial was further evaluated in a murine model. The implants where harvested after 1 days for histological, inflammatory, and protein analysis. The application of DFO to the cells could mimic and enhance the wound healing capabilities of environmentally induced hypoxia through VEGF expression and promises a more viable option in clinical settings that is not merely restricted to the laboratory.
Collapse
Affiliation(s)
- Elizabeth A Wahl
- Department of Plastic Surgery and Hand Surgery, University Hospital rechts der Isar, Technische Universität München, Munich, 81675, Germany
| | - Thilo L Schenck
- Department of Plastic Surgery and Hand Surgery, University Hospital rechts der Isar, Technische Universität München, Munich, 81675, Germany
| | - Hans-Günther Machens
- Department of Plastic Surgery and Hand Surgery, University Hospital rechts der Isar, Technische Universität München, Munich, 81675, Germany
| | - Elizabeth R Balmayor
- Department of Plastic Surgery and Hand Surgery, University Hospital rechts der Isar, Technische Universität München, Munich, 81675, Germany.,Institute for Advanced Study, Technische Universität München, Garching, 85748, Germany
| |
Collapse
|
21
|
Lodge KM, Thompson AAR, Chilvers ER, Condliffe AM. Hypoxic regulation of neutrophil function and consequences for Staphylococcus aureus infection. Microbes Infect 2016; 19:166-176. [PMID: 27789256 DOI: 10.1016/j.micinf.2016.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/07/2016] [Accepted: 10/12/2016] [Indexed: 01/22/2023]
Abstract
Staphylococcal infection and neutrophilic inflammation can act in concert to establish a profoundly hypoxic environment. In this review we summarise how neutrophils and Staphylococcus aureus are adapted to function under hypoxic conditions, with a particular focus on the impaired ability of hypoxic neutrophils to effect Staphylococcus aureus killing.
Collapse
Affiliation(s)
- Katharine M Lodge
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge University Hospitals, Hills Road, Cambridge, CB2 0QQ, UK
| | - A A Roger Thompson
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Edwin R Chilvers
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge University Hospitals, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Alison M Condliffe
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| |
Collapse
|
22
|
Tiwari A, Wong CS, Nekkanti LP, Deane JA, McDonald C, Jenkin G, Kirkland MA. Impact of Oxygen Levels on Human Hematopoietic Stem and Progenitor Cell Expansion. Stem Cells Dev 2016; 25:1604-1613. [PMID: 27539189 DOI: 10.1089/scd.2016.0153] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Oxygen levels are an important variable during the in vitro culture of stem cells. There has been increasing interest in the use of low oxygen to maximize proliferation and, in some cases, effect differentiation of stem cell populations. It is generally assumed that the defined pO2 in the incubator reflects the pO2 to which the stem cells are being exposed. However, we demonstrate that the pO2 experienced by cells in static culture can change dramatically during the course of culture as cell numbers increase and as the oxygen utilization by cells exceeds the diffusion of oxygen through the media. Dynamic culture (whereby the cell culture plate is in constant motion) largely eliminates this effect, and a combination of low ambient oxygen and dynamic culture results in a fourfold increase in reconstituting capacity of human hematopoietic stem cells compared with those cultured in static culture at ambient oxygen tension. Cells cultured dynamically at 5% oxygen exhibited the best expansion: 30-fold increase by flow cytometry, 120-fold increase by colony assay, and 11% of human CD45 engraftment in the bone marrow of NOD/SCID mice. To our knowledge, this is the first study to compare individual and combined effects of oxygen and static or dynamic culture on hematopoietic ex vivo expansion. Understanding and controlling the effective oxygen tension experienced by cells may be important in clinical stem cell expansion systems, and these results may have relevance to the interpretation of low oxygen culture studies.
Collapse
Affiliation(s)
- Abhilasha Tiwari
- 1 The Ritchie Centre, Hudson Institute of Medical Research , Clayton, Australia
| | | | - Lakshmi P Nekkanti
- 1 The Ritchie Centre, Hudson Institute of Medical Research , Clayton, Australia
| | - James A Deane
- 1 The Ritchie Centre, Hudson Institute of Medical Research , Clayton, Australia .,3 Department of Obstetrics and Gynaecology, Southern Clinical School, Monash University , Clayton, Australia
| | - Courtney McDonald
- 1 The Ritchie Centre, Hudson Institute of Medical Research , Clayton, Australia
| | - Graham Jenkin
- 1 The Ritchie Centre, Hudson Institute of Medical Research , Clayton, Australia .,3 Department of Obstetrics and Gynaecology, Southern Clinical School, Monash University , Clayton, Australia
| | | |
Collapse
|
23
|
Quantitative proteomic analysis and comparison of two bone marrow stromal cell lines using the SILAC method. Exp Hematol 2016; 44:1059-1071. [PMID: 27539861 DOI: 10.1016/j.exphem.2016.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/11/2016] [Accepted: 08/04/2016] [Indexed: 01/10/2023]
Abstract
Two human bone marrow stromal cell lines, HS5 and HS27a, co-cultured with myeloid cells, have frequently been used in studies of cross talk between cells in the bone marrow microenvironment and hematopoietic cells. Altered expression of proteins is typically associated with cell-cell signal transduction and regulation of cellular functions. Many studies have focused on key proteins that contribute to functional differences in cell co-culture models, but global quantitative proteome analysis of HS5 and HS27a has not been performed. We employed the stable isotope labeling by amino acids in cell culture (SILAC) method using two stable isotopes each of arginine and lysine to label proteins in the two cell lines. Labeled proteins were analyzed by 2-D ultrahigh-resolution liquid chromatography- LTQ/Orbitrap mass spectrometry. Among 4,213 unique identified and annotated proteins in the cell lines, 1,462 were detected in two independent experiments. Of these, 69 exhibited significant upregulation and 48 significant downregulation (>95% confidence) in HS27a relative to HS5 cells. Gene ontology term and pathway analysis indicated that the differentially regulated proteins were involved in cellular movement, cell-to-cell signaling and interaction, and hematologic system development and function. A total of 55 items were identified in both genomic and proteomic databases. Quantitative reverse transcription polymerase chain reaction and Western blotting were performed on 7 proteins randomly selected from 28 differentially expressed proteins that were identified in both databases and were involved in the top networks/pathways. We observed a decrease in apoptosis in co-cultured KG1a cells when integrin αV was inhibited in HS27a cells, which suggested the functional role of integrin αV in the co-culture system. The integrated genomic/proteomic approach described here, and the identified proteins, will provide a useful basis for further elucidation of molecular mechanisms in the bone marrow microenvironment and for ongoing studies of cross talk among stromal cells and myeloma cells in co-culture systems.
Collapse
|
24
|
Sotnezova E, Andreeva E, Grigoriev A, Buravkova L. Ex Vivo Expansion of Hematopoietic Stem and Progenitor Cells from Umbilical Cord Blood. Acta Naturae 2016; 8:6-16. [PMID: 27795840 PMCID: PMC5081707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Indexed: 12/02/2022] Open
Abstract
Transplantation of umbilical cord blood cells is currently widely used in modern cell therapy. However, the limited number of hematopoietic stem and progenitor cells (HSPCs) and prolonged time of recovery after the transplantation are significant limitations in the use of cord blood. Ex vivo expansion with various cytokine combinations is one of the most common approaches for increasing the number of HSPCs from one cord blood unit. In addition, there are protocols that enable ex vivo amplification of cord blood cells based on native hematopoietic microenvironmental cues, including stromal components and the tissue-relevant oxygen level. The newest techniques for ex vivo expansion of HSPCs are based on data from the elucidation of the molecular mechanisms governing the hematopoietic niche function. Application of these methods has provided an improvement of several important clinical outcomes. Alternative methods of cord blood transplantation enhancement based on optimization of HPSC homing and engraftment in patient tissues have also been successful. The goal of the present review is to analyze recent methodological approaches to cord blood HSPC ex vivo amplification.
Collapse
Affiliation(s)
- E.V. Sotnezova
- Institute of Biomedical Problems of the Russian Academy of Sciences, Khoroshovskoye shosse 76A, Moscow, 123007, Russia
| | - E.R. Andreeva
- Institute of Biomedical Problems of the Russian Academy of Sciences, Khoroshovskoye shosse 76A, Moscow, 123007, Russia
| | - A.I. Grigoriev
- Institute of Biomedical Problems of the Russian Academy of Sciences, Khoroshovskoye shosse 76A, Moscow, 123007, Russia
| | - L.B. Buravkova
- Institute of Biomedical Problems of the Russian Academy of Sciences, Khoroshovskoye shosse 76A, Moscow, 123007, Russia
| |
Collapse
|
25
|
Wang Z, Du Z, Cai H, Ye Z, Fan J, Tan WS. Low oxygen tension favored expansion and hematopoietic reconstitution of CD34+CD38−cells expanded from human cord blood-derived CD34+Cells. Biotechnol J 2016; 11:945-53. [DOI: 10.1002/biot.201500497] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 12/08/2015] [Accepted: 03/17/2016] [Indexed: 01/09/2023]
Affiliation(s)
- Ziyan Wang
- State Key Laboratory of Bioreactor Engineering; East China University of Science and Technology; Shanghai China
| | - Zheng Du
- State Key Laboratory of Bioreactor Engineering; East China University of Science and Technology; Shanghai China
| | - Haibo Cai
- State Key Laboratory of Bioreactor Engineering; East China University of Science and Technology; Shanghai China
| | - Zhaoyang Ye
- State Key Laboratory of Bioreactor Engineering; East China University of Science and Technology; Shanghai China
| | - Jinli Fan
- State Key Laboratory of Bioreactor Engineering; East China University of Science and Technology; Shanghai China
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering; East China University of Science and Technology; Shanghai China
| |
Collapse
|
26
|
Deynoux M, Sunter N, Hérault O, Mazurier F. Hypoxia and Hypoxia-Inducible Factors in Leukemias. Front Oncol 2016; 6:41. [PMID: 26955619 PMCID: PMC4767894 DOI: 10.3389/fonc.2016.00041] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/08/2016] [Indexed: 01/10/2023] Open
Abstract
Despite huge improvements in the treatment of leukemia, the percentage of patients suffering relapse still remains significant. Relapse most often results from a small number of leukemic stem cells (LSCs) within the bone marrow, which are able to self-renew, and therefore reestablish the full tumor. The marrow microenvironment contributes considerably in supporting the protection and development of leukemic cells. LSCs share specific niches with normal hematopoietic stem cells with the niche itself being composed of a variety of cell types, including mesenchymal stem/stromal cells, bone cells, immune cells, neuronal cells, and vascular cells. A hallmark of the hematopoietic niche is low oxygen partial pressure, indeed this hypoxia is necessary for the long-term maintenance of hematopoietic stem/progenitor cells. Hypoxia is a strong signal, principally maintained by members of the hypoxia-inducible factor (HIF) family. In solid tumors, it has been well established that hypoxia triggers intrinsic metabolic changes and microenvironmental modifications, such as the stimulation of angiogenesis, through activation of HIFs. As leukemia is not considered a “solid” tumor, the role of oxygen in the disease was presumed to be inconsequential and remained long overlooked. This view has now been revised since hypoxia has been shown to influence leukemic cell proliferation, differentiation, and resistance to chemotherapy. However, the role of HIF proteins remains controversial with HIFs being considered as either oncogenes or tumor suppressor genes, depending on the study and model. The purpose of this review is to highlight our knowledge of hypoxia and HIFs in leukemic development and therapeutic resistance and to discuss the recent hypoxia-based strategies proposed to eradicate leukemias.
Collapse
Affiliation(s)
- Margaux Deynoux
- Génétique, Immunothérapie, Chimie et Cancer (GICC) UMR 7292, CNRS, UFR de Médecine, Université François-Rabelais de Tours , Tours , France
| | - Nicola Sunter
- Génétique, Immunothérapie, Chimie et Cancer (GICC) UMR 7292, CNRS, UFR de Médecine, Université François-Rabelais de Tours , Tours , France
| | - Olivier Hérault
- Génétique, Immunothérapie, Chimie et Cancer (GICC) UMR 7292, CNRS, UFR de Médecine, Université François-Rabelais de Tours, Tours, France; Service d'Hématologie Biologique, Centre Hospitalier Régional Universitaire de Tours, Tours, France
| | - Frédéric Mazurier
- Génétique, Immunothérapie, Chimie et Cancer (GICC) UMR 7292, CNRS, UFR de Médecine, Université François-Rabelais de Tours , Tours , France
| |
Collapse
|
27
|
Slone WL, Moses BS, Evans R, Piktel D, Martin KH, Petros W, Craig M, Gibson LF. Modeling Chemotherapy Resistant Leukemia In Vitro. J Vis Exp 2016:e53645. [PMID: 26891147 DOI: 10.3791/53645] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
It is well established that the bone marrow microenvironment provides a unique site of sanctuary for hematopoietic diseases that both initiate and progress in this site. The model presented in the current report utilizes human primary bone marrow stromal cells and osteoblasts as two representative cell types from the marrow niche that influence tumor cell phenotype. The in vitro co-culture conditions described for human leukemic cells with these primary niche components support the generation of a chemoresistant subpopulation of tumor cells that can be efficiently recovered from culture for analysis by diverse techniques. A strict feeding schedule to prevent nutrient fluxes followed by gel type 10 cross-linked dextran (G10) particles recovery of the population of tumor cells that have migrated beneath the adherent bone marrow stromal cells (BMSC) or osteoblasts (OB) generating a "phase dim" (PD) population of tumor cells, provides a consistent source of purified therapy resistant leukemic cells. This clinically relevant population of tumor cells can be evaluated by standard methods to investigate apoptotic, metabolic, and cell cycle regulatory pathways as well as providing a more rigorous target in which to test novel therapeutic strategies prior to pre-clinical investigations targeted at minimal residual disease.
Collapse
Affiliation(s)
- William L Slone
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine
| | - Blake S Moses
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine
| | - Rebecca Evans
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine
| | - Debbie Piktel
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine
| | - Karen H Martin
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine; Department of Neurobiology and Anatomy, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine
| | - William Petros
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine
| | - Michael Craig
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine
| | - Laura F Gibson
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine; Department of Microbiology, Immunology and Cell Biology, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine;
| |
Collapse
|
28
|
Moirangthem RD, Singh S, Adsul A, Jalnapurkar S, Limaye L, Kale VP. Hypoxic niche-mediated regeneration of hematopoiesis in the engraftment window is dominantly affected by oxygen tension in the milieu. Stem Cells Dev 2015; 24:2423-36. [PMID: 26107807 PMCID: PMC4599134 DOI: 10.1089/scd.2015.0112] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The bone marrow (BM) microenvironment or the hematopoietic stem cell (HSC) niche is normally hypoxic, which maintains HSC quiescence. Paradoxically, transplanted HSCs rapidly proliferate in this niche. Pretransplant myelosuppression results in a substantial rise in oxygen levels in the marrow microenvironment due to reduced cellularity and consequent low oxygen consumption. Therefore, it may be construed that the rapid proliferation of the engrafted HSCs in the BM niche is facilitated by the transiently elevated oxygen tension in this milieu during the “engraftment window.” To determine whether oxygen tension dominantly affects the regeneration of hematopoiesis in the BM niche, we created an “oxygen-independent hypoxic niche” by treating BM-derived mesenchymal stromal cells (BMSCs) with a hypoxia-mimetic compound, cobalt chloride (CoCl2) and cocultured them with BM-derived HSC-enriched cells under normoxic conditions (HSCs; CoCl2-cocultures). Cocultures with untreated BMSCs incubated under normoxia (control- cocultures) or hypoxia (1% O2; hypoxic-cocultures) were used as comparators. Biochemical analyses showed that though, both CoCl2 and hypoxia evoked comparable signals in the BMSCs, the regeneration of hematopoiesis in their respective cocultures was radically different. The CoCl2-BMSCs supported robust hematopoiesis, while the hypoxic-BMSCs exerted strong inhibition. The hematopoiesis-supportive ability of CoCl2-BMSCs was abrogated if the CoCl2-cocultures were incubated under hypoxia, demonstrating that the prevalent oxygen tension in the milieu dominantly affects the outcome of the HSC-BM niche interactions. Our data suggest that pharmacologically delaying the reestablishment of hypoxia in the BM may boost post-transplant regeneration of hematopoiesis.
Collapse
Affiliation(s)
| | - Shweta Singh
- Stem Cell Lab, National Centre for Cell Science , Pune, India
| | - Ashwini Adsul
- Stem Cell Lab, National Centre for Cell Science , Pune, India
| | | | - Lalita Limaye
- Stem Cell Lab, National Centre for Cell Science , Pune, India
| | | |
Collapse
|
29
|
Spinello I, Quaranta MT, Paolillo R, Pelosi E, Cerio AM, Saulle E, Lo Coco F, Testa U, Labbaye C. Differential hypoxic regulation of the microRNA-146a/CXCR4 pathway in normal and leukemic monocytic cells: impact on response to chemotherapy. Haematologica 2015; 100:1160-71. [PMID: 26045293 DOI: 10.3324/haematol.2014.120295] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 05/28/2015] [Indexed: 02/06/2023] Open
Abstract
High expression of the chemokine receptor 4, CXCR4, associated with a negative prognosis in acute myeloid leukemia, is related to hypoxia. Because CXCR4 expression is under the post-transcriptional control of microRNA-146a in normal and leukemic monocytic cells, we first investigated the impact of hypoxia on microRNA-146a and CXCR4 expression during monocytopoiesis and in acute monocytic leukemia. We then analyzed the effects of hypoxia on drug sensitivity of CXCR4-expressing leukemic cells. We found that microRNA-146a is a target of hypoxia-inducible factor-1α or -2α in relation to the stage of monocytopoiesis and the level of hypoxia, and demonstrated the regulation of the microRNA-146a/CXCR4 pathway by hypoxia in monocytes derived from CD34(+) cells. Thus, in myeloid leukemic cell lines, hypoxia-mediated control of the microRNA-146a/CXCR4 pathway depends only on the capacity of hypoxia-inducible factor-1α to up-regulate microRNA-146a, which in turn decreases CXCR4 expression. However, at variance with normal monocytic cells and leukemic cell lines, in acute monocytic leukemia overexpressing CXCR4, hypoxia up-modulates microRNA-146a but fails to down-modulate CXCR4 expression. We then investigated the effect of hypoxia on the response of leukemic cells to chemotherapy alone or in combination with stromal-derived factor-1α. We found that hypoxia increases stromal-derived factor-1α-induced survival of leukemic cells by decreasing their sensitivity to anti-leukemic drugs. Altogether, our results demonstrate that hypoxia-mediated regulation of microRNA-146a, which controls CXCR4 expression in monocytic cells, is lost in acute monocytic leukemia, thus contributing to maintaining CXCR4 overexpression and protecting the cells from anti-leukemic drugs in the hypoxic bone marrow microenvironment.
Collapse
Affiliation(s)
- Isabella Spinello
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, University of Rome "Tor Vergata", Rome, Italy
| | - Maria Teresa Quaranta
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, University of Rome "Tor Vergata", Rome, Italy
| | - Rosa Paolillo
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, University of Rome "Tor Vergata", Rome, Italy
| | - Elvira Pelosi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, University of Rome "Tor Vergata", Rome, Italy
| | - Anna Maria Cerio
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, University of Rome "Tor Vergata", Rome, Italy
| | - Ernestina Saulle
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, University of Rome "Tor Vergata", Rome, Italy
| | - Francesco Lo Coco
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy Fondazione Santa Lucia, Rome, Italy
| | - Ugo Testa
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, University of Rome "Tor Vergata", Rome, Italy
| | - Catherine Labbaye
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
30
|
Paolillo R, Spinello I, Quaranta MT, Pasquini L, Pelosi E, Lo Coco F, Testa U, Labbaye C. Human TM9SF4 Is a New Gene Down-Regulated by Hypoxia and Involved in Cell Adhesion of Leukemic Cells. PLoS One 2015; 10:e0126968. [PMID: 25961573 PMCID: PMC4427288 DOI: 10.1371/journal.pone.0126968] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/09/2015] [Indexed: 12/19/2022] Open
Abstract
Background The transmembrane 9 superfamily protein member 4, TM9SF4, belongs to the TM9SF family of proteins highly conserved through evolution. TM9SF4 homologs, previously identified in many different species, were mainly involved in cellular adhesion, innate immunity and phagocytosis. In human, the function and biological significance of TM9SF4 are currently under investigation. However, TM9SF4 was found overexpressed in human metastatic melanoma and in a small subset of acute myeloid leukemia (AMLs) and myelodysplastic syndromes, consistent with an oncogenic function of this gene. Purpose and Results In this study, we first analyzed the expression and regulation of TM9SF4 in normal and leukemic cells and identified TM9SF4 as a gene highly expressed in human quiescent CD34+ hematopoietic progenitor cells (HPCs), regulated during monocytic and granulocytic differentiation of HPCs, both lineages giving rise to mature myeloid cells involved in adhesion, phagocytosis and immunity. Then, we found that TM9SF4 is markedly overexpressed in leukemic cells and in AMLs, particularly in M2, M3 and M4 AMLs (i.e., in AMLs characterized by the presence of a more or less differentiated granulocytic progeny), as compared to normal CD34+ HPCs. Proliferation and differentiation of HPCs occurs in hypoxia, a physiological condition in bone marrow, but also a crucial component of cancer microenvironment. Here, we investigated the impact of hypoxia on TM9SF4 expression in leukemic cells and identified TM9SF4 as a direct target of HIF-1α, downregulated in these cells by hypoxia. Then, we found that the hypoxia-mediated downregulation of TM9SF4 expression is associated with a decrease of cell adhesion of leukemic cells to fibronectin, thus demonstrating that human TM9SF4 is a new molecule involved in leukemic cell adhesion. Conclusions Altogether, our study reports for the first time the expression of TM9SF4 at the level of normal and leukemic hematopoietic cells and its marked expression at the level of AMLs displaying granulocytic differentiation.
Collapse
MESH Headings
- Apoptosis/drug effects
- Bone Marrow Cells/drug effects
- Bone Marrow Cells/metabolism
- Bone Marrow Cells/pathology
- Cell Adhesion/drug effects
- Cell Cycle/drug effects
- Cell Differentiation/drug effects
- Cell Hypoxia
- Cell Proliferation/drug effects
- Cloning, Molecular
- Fibronectins/metabolism
- Gene Expression Regulation, Leukemic
- Granulocytes/drug effects
- Granulocytes/metabolism
- Granulocytes/pathology
- HEK293 Cells
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Monocytes/drug effects
- Monocytes/metabolism
- Monocytes/pathology
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Oxygen/pharmacology
- Primary Cell Culture
- Promoter Regions, Genetic
- Signal Transduction
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Rosa Paolillo
- Department of Hematology, Oncology and Molecular Medicine of Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Isabella Spinello
- Department of Hematology, Oncology and Molecular Medicine of Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Maria Teresa Quaranta
- Department of Hematology, Oncology and Molecular Medicine of Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Luca Pasquini
- Department of Hematology, Oncology and Molecular Medicine of Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Elvira Pelosi
- Department of Hematology, Oncology and Molecular Medicine of Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Francesco Lo Coco
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
- Fondazione Santa Lucia, Rome, Italy
| | - Ugo Testa
- Department of Hematology, Oncology and Molecular Medicine of Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Catherine Labbaye
- Department of Hematology, Oncology and Molecular Medicine of Istituto Superiore di Sanità, 00161, Rome, Italy
- * E-mail:
| |
Collapse
|
31
|
Andreeva ER, Andrianova IV, Sotnezova EV, Buravkov SV, Bobyleva PI, Romanov YA, Buravkova LB. Human adipose-tissue derived stromal cells in combination with hypoxia effectively support ex vivo expansion of cord blood haematopoietic progenitors. PLoS One 2015; 10:e0124939. [PMID: 25919031 PMCID: PMC4412539 DOI: 10.1371/journal.pone.0124939] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 03/20/2015] [Indexed: 12/12/2022] Open
Abstract
The optimisation of haematopoietic stem and progenitor cell expansion is on demand in modern cell therapy. In this work, haematopoietic stem/progenitor cells (HSPCs) have been selected from unmanipulated cord blood mononuclear cells (cbMNCs) due to adhesion to human adipose-tissue derived stromal cells (ASCs) under standard (20%) and tissue-related (5%) oxygen. ASCs efficiently maintained viability and supported further HSPC expansion at 20% and 5% O2. During co-culture with ASCs, a new floating population of differently committed HSPCs (HSPCs-1) grew. This suspension was enriched with СD34+ cells up to 6 (20% O2) and 8 (5% O2) times. Functional analysis of HSPCs-1 revealed cobble-stone area forming cells (CAFCs) and lineage-restricted colony-forming cells (CFCs). The number of CFCs was 1.6 times higher at tissue-related O2, than in standard cultivation (20% O2). This increase was related to a rise in the number of multipotent precursors - BFU-E, CFU-GEMM and CFU-GM. These changes were at least partly ensured by the increased concentration of MCP-1 and IL-8 at 5% O2. In summary, our data demonstrated that human ASCs enables the selection of functionally active HSPCs from unfractionated cbMNCs, the further expansion of which without exogenous cytokines provides enrichment with CD34+ cells. ASCs efficiently support the viability and proliferation of cord blood haematopoietic progenitors of different commitment at standard and tissue-related O2 levels at the expense of direct and paracrine cell-to-cell interactions.
Collapse
Affiliation(s)
- Elena R. Andreeva
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
- * E-mail:
| | - Irina V. Andrianova
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
- Institute of Experimental Cardiology, Cardiology Research Center, Moscow, Russia
| | - Elena V. Sotnezova
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | | | - Polina I. Bobyleva
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Yury A. Romanov
- Institute of Experimental Cardiology, Cardiology Research Center, Moscow, Russia
| | | |
Collapse
|
32
|
Yu Q, Liu L, Lin J, Wang Y, Xuan X, Guo Y, Hu S. SDF-1α/CXCR4 Axis Mediates The Migration of Mesenchymal Stem Cells to The Hypoxic-Ischemic Brain Lesion in A Rat Model. CELL JOURNAL 2015; 16:440-7. [PMID: 25685734 PMCID: PMC4297482 DOI: 10.22074/cellj.2015.504] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 11/06/2013] [Indexed: 12/16/2022]
Abstract
Objective Transplantation of mesenchymal stem cells (MSCs) can promote functional
recovery of the brain after hypoxic-ischemic brain damage (HIBD). However, the mechanism regulating MSC migration to a hypoxic-ischemic lesion is poorly understood. Interaction between stromal cell-derived factor-1α (SDF-1α) and its cognate receptor CXC
chemokine receptor 4 (CXCR4) is crucial for homing and migration of multiple stem cell
types. In this study, we investigate the potential role of SDF-1α/CXCR4 axis in mediating
MSC migration in an HIBD model.
Materials and Methods In this experimental study, we first established the animal model of HIBD using the neonatal rat. Bone marrow MSCs were cultured and labeled with
5-bromo-21-deoxyuridine (BrdU) after which 6×106 cells were intravenously injected into
the rat. BrdU positive MSCs in the hippocampus were detected by immunohistochemical
analyses. The expression of hypoxia-inducible factor-1α (HIF-1α) and SDF-1α in the hippocampus of hypoxic-ischemic rats was detected by Western blotting. To investigate the
role of hypoxia and SDF-1α on migration of MSCs in vitro, MSCs isolated from normal
rats were cultured in a hypoxic environment (PO2=1%). Migration of MSCs was detected
by the transwell assay. The expression of CXCR4 was tested using Western blotting and
flow cytometry.
Results BrdU-labeled MSCs were found in the rat brain, which suggested that transplanted MSCs migrated to the site of the hypoxic-ischemic brain tissue. HIF-1α and SDF-1α significantly increased in the hippocampal formations of HIBD rats in a time-dependent
manner. They peaked on day 7 and were stably expressed until day 21. Migration of MSCs
in vitro was promoted by SDF-1α under hypoxia and inhibited by the CXCR4 inhibitor
AMD3100. The expression of CXCR4 on MSCs was elevated by hypoxia stimulation as
well as microdosage treatment of SDF-1α. Conclusion This observation illustrates that SDF-1α/CXCR4 axis mediate the migration
of MSCs to a hypoxic-ischemic brain lesion in a rat model.
Collapse
Affiliation(s)
- Qin Yu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China ; Institute of Bioengineering, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Lizhen Liu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Lin
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Wang
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaobo Xuan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Guo
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shaojun Hu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
33
|
Huang Y, Chen B, Zhang J. Oxygen tension variation in ischemic gastrocnemius muscle, marrow, and different hypoxic conditions in vitro. Med Sci Monit 2014; 20:2171-6. [PMID: 25372971 PMCID: PMC4301230 DOI: 10.12659/msm.892354] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Bone marrow stromal cells (BMSCs) play an important role in ischemic limb angiogenesis. BMSCs cultured in vitro can be exposed to oxygen tension much higher than that experienced in vivo. This study assessed oxygen tension in bone marrow and ischemic muscle in vivo, and then identified an appropriate oxygen concentration for culturing BMSCs. MATERIAL/METHODS Unilateral hind limb ischemia was surgically induced in 30 mice, and tissue oxygen tension in bilateral gastrocnemius muscles and femoral bone marrow was monitored in vivo using a micro-electrode at 24 hours, 1 week, 2 weeks, and 3 weeks after modeling. Media used for culturing normal marrow, muscle, and artery tissue were incubated with various oxygen concentrations, and O2 tension was continuously monitored. Oxygen tension in aortic arterial blood was monitored using a micro-electrode and blood gas analyzer, and the results were compared. RESULTS Oxygen tension in ischemic gastrocnemius muscle reached a nadir at 1 week after ischemic modeling, when histological changes were most noticeable. Culture media incubated with 3%, 6%, and 14% oxygen (the normal oxygen levels of bone marrow, muscle, and arterial blood, respectively) required 9, 6, and 2 hours, respectively, to reach an equilibrated oxygen tension, and oxygen tension was elevated by 1.6-, 1.2-, and 0.4-fold, respectively, upon re-exposure of the media to air. CONCLUSIONS Physiological oxygen tension differs in different tissues. A 3% O2 concentration mimics the physiological O2 exposure experienced by BMSCs and represents the hypoxic concentration. Culture medium incubated under hypoxic conditions requires a prolonged period of time to regain equilibrated oxygen tension.
Collapse
Affiliation(s)
- Ying Huang
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China (mainland)
| | - Bing Chen
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China (mainland)
| | - Jian Zhang
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China (mainland)
| |
Collapse
|
34
|
Yamamoto Y, Shiraishi M, Fujita M, Kojima I, Tanaka Y, Tachibana S. Triiodothyronine suppresses activin-induced differentiation of erythroleukemia K562 cells under hypoxic conditions. Mol Cell Biochem 2014; 391:217-24. [PMID: 24604674 DOI: 10.1007/s11010-014-2005-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 02/21/2014] [Indexed: 12/13/2022]
Abstract
Thyroid hormone stimulates erythropoietic differentiation. However, severe anemia is sometimes seen in patients with hyperthyroidism, and the mechanisms have not been fully elucidated. Bone marrow is comprised about 2-8% oxygen, and the characteristics of hematopoietic stem cells have been shown to be influenced under hypoxia. Hypoxia-inducible factor-1 is a critical mediator of cellular responses to hypoxia and an important mediator in signal transduction of thyroid hormone [triiodothyronine (T3)]. The aim of this study was to investigate the effect of T3 on erythropoiesis under hypoxia mimicking physiological conditions in the bone marrow. We maintained human erythroleukemia K562 cells under hypoxic atmosphere (2% O₂) and examined their cellular characteristics. Compared to that under normal atmospheric conditions, cells under hypoxia showed a reduction in the proliferation rate and increase in the hemoglobin content or benzidine-positive rate, indicating promotion of erythroid differentiation. T3 had no effect on hypoxia-induced erythroid differentiation, but significantly inhibited activin A/erythroid differentiation factor-induced erythroid differentiation. Moreover, GATA2 mRNA expression was suppressed in association with erythroid differentiation, while T3 significantly diminished that suppression. These results suggest that T3 has a direct suppressive effect on erythroid differentiation under hypoxic conditions.
Collapse
Affiliation(s)
- Yoritsuna Yamamoto
- Division of Environmental Medicine, National Defense Medical College Research Institute, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan,
| | | | | | | | | | | |
Collapse
|
35
|
The effect of hypoxia on the stemness and differentiation capacity of PDLC and DPC. BIOMED RESEARCH INTERNATIONAL 2014; 2014:890675. [PMID: 24701587 PMCID: PMC3950753 DOI: 10.1155/2014/890675] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 01/08/2014] [Indexed: 02/07/2023]
Abstract
Introduction. Stem cells are regularly cultured under normoxic conditions. However, the physiological oxygen tension in the stem cell niche is known to be as low as 1-2% oxygen, suggesting that hypoxia has a distinct impact on stem cell maintenance. Periodontal ligament cells (PDLCs) and dental pulp cells (DPCs) are attractive candidates in dental tissue regeneration. It is of great interest to know whether hypoxia plays a role in maintaining the stemness and differentiation capacity of PDLCs and DPCs. Methods. PDLCs and DPCs were cultured either in normoxia (20% O2) or hypoxia (2% O2). Cell viability assays were performed and the expressions of pluripotency markers (Oct-4, Sox2, and c-Myc) were detected by qRT-PCR and western blotting. Mineralization, glycosaminoglycan (GAG) deposition, and lipid droplets formation were assessed by Alizarin red S, Safranin O, and Oil red O staining, respectively. Results. Hypoxia did not show negative effects on the proliferation of PDLCs and DPCs. The pluripotency markers and differentiation potentials of PDLCs and DPCs significantly increased in response to hypoxic environment. Conclusions. Our findings suggest that hypoxia plays an important role in maintaining the stemness and differentiation capacity of PDLCs and DPCs.
Collapse
|
36
|
Christoph M, Ibrahim K, Hesse K, Augstein A, Schmeisser A, Braun-Dullaeus RC, Simonis G, Wunderlich C, Quick S, Strasser RH, Poitz DM. Local inhibition of hypoxia-inducible factor reduces neointima formation after arterial injury in ApoE-/- mice. Atherosclerosis 2014; 233:641-647. [PMID: 24561491 DOI: 10.1016/j.atherosclerosis.2014.01.048] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 01/22/2014] [Accepted: 01/24/2014] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Hypoxia plays a pivotal role in development and progression of restenosis after vascular injury. Under hypoxic conditions the hypoxia-inducible factors (HIFs) are the most important transcription factors for the adaption to reduced oxygen supply. Therefore the aim of the study was to investigate the effect of a local HIF-inhibition and overexpression on atherosclerotic plaque development in a murine vascular injury model. METHODS AND RESULTS After wire-induced vascular injury in ApoE-/- mice a transient, local inhibition of HIF as well as an overexpression approach of the different HIF-subunits (HIF-1α, HIF-2α) by adenoviral infection was performed. The local inhibition of the HIF-pathway using a dominant-negative mutant dramatically reduced the extent of neointima formation. The diminished plaque size was associated with decreased expression of the well-known HIF-target genes vascular endothelial growth factor-A (VEGF-A) and its receptors Flt-1 and Flk-1. In contrast, the local overexpression of HIF-1α and HIF-2α further increased the plaque size after wire-induced vascular injury. CONCLUSIONS Local HIF-inhibition decreases and HIF-α overexpression increases the injury induced neointima formation. These findings provide new insight into the pathogenesis of atherosclerosis and may lead to new therapeutic options for the treatment of in stent restenosis.
Collapse
MESH Headings
- Adenoviridae
- Animals
- Apolipoproteins E/deficiency
- Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors
- Basic Helix-Loop-Helix Transcription Factors/biosynthesis
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/physiology
- Coronary Restenosis
- Disease Models, Animal
- Endothelium, Vascular/injuries
- Femoral Artery/injuries
- Femoral Artery/pathology
- Genetic Vectors
- Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors
- Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/physiology
- Male
- Mice
- Mice, Knockout
- Neointima/prevention & control
- Plaque, Atherosclerotic/etiology
- Plaque, Atherosclerotic/prevention & control
- Signal Transduction
- Transduction, Genetic
- Up-Regulation
- Vascular Endothelial Growth Factor A/biosynthesis
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor Receptor-1/biosynthesis
- Vascular Endothelial Growth Factor Receptor-1/genetics
- Vascular Endothelial Growth Factor Receptor-2/biosynthesis
- Vascular Endothelial Growth Factor Receptor-2/genetics
Collapse
Affiliation(s)
- Marian Christoph
- University of Dresden, Heart Center, University Hospital, Germany
| | - Karim Ibrahim
- University of Dresden, Heart Center, University Hospital, Germany
| | - Kathleen Hesse
- University of Dresden, Heart Center, University Hospital, Germany
| | - Antje Augstein
- University of Dresden, Heart Center, University Hospital, Germany
| | | | | | - Gregor Simonis
- University of Dresden, Heart Center, University Hospital, Germany
| | | | - Silvio Quick
- University of Dresden, Heart Center, University Hospital, Germany
| | - Ruth H Strasser
- University of Dresden, Heart Center, University Hospital, Germany
| | - David M Poitz
- University of Dresden, Heart Center, University Hospital, Germany.
| |
Collapse
|
37
|
Poitz DM, Augstein A, Hesse K, Christoph M, Ibrahim K, Braun-Dullaeus RC, Strasser RH, Schmeißer A. Regulation of the HIF-system in human macrophages--differential regulation of HIF-α subunits under sustained hypoxia. Mol Immunol 2013; 57:226-35. [PMID: 24176785 DOI: 10.1016/j.molimm.2013.10.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 09/23/2013] [Accepted: 10/01/2013] [Indexed: 02/01/2023]
Abstract
Macrophages are often associated to pathophysiological processes and were found at hypoxic areas. However, cell adaption greatly depends on hypoxia-inducible factors (HIF). Activation of these transcription factors is induced by heterodimerization of an α-(HIF-1α, -2α, -3α) and HIF-1β subunit. The main regulatory pathway is represented by α-subunit stability. Beside, little is known about the exact mechanisms of fine-tuning in Hif-regulation. The present study characterizes the hypoxia-induced regulation of HIF-1α and -2α in human macrophages. The hypoxic increase of both subunits is initially mediated by protein stabilization. Sustained hypoxia caused a distinct regulation of HIF-1α and -2α. The striking increase of HIF-2α protein expression was contrasted by a dramatic decrease of HIF-1α. The long-term downregulation of HIF-1α is due to downregulation of its mRNA. This decrease was accompanied by increased expression of ahif, a natural cis-antisense transcript of HIF-1α. The ahif-transcript was strongly inducible by hypoxia and rapidly degraded under reoxygenation. Using an adenoviral overexpression and siRNA silencing approach revealed that the targeted regulation of ahif is mediated by the HIF-system itself. Furthermore it could be shown that ahif indeed is able to modulate the hypoxic expression of HIF-1α and influences the expression of the HIF-target gene Enolase-2. Taken together, this study characterizes a new regulation process of the HIF-transcription factor-system in human macrophages under hypoxia. For the first time evidence is provided that ahif is regulated by the HIF-system and influences HIF-1α expression in primary human macrophages.
Collapse
Affiliation(s)
- David M Poitz
- Department of Internal Medicine and Cardiology, Dresden University of Technology, Fetscherstr. 74, 01307 Dresden, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Garg A, Newsome PN. Bone marrow mesenchymal stem cells and liver regeneration: believe the hypoxia! Stem Cell Res Ther 2013; 4:108. [PMID: 24004909 PMCID: PMC4056659 DOI: 10.1186/scrt319] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
There are limited data on the efficacy of mesenchymal stem cells (MSCs) in models of extensive hepatic resection. In the previous issue of Stem Cell Research &Therapy, Yu and colleagues demonstrate that transient hypoxic preconditioning of MSCs improves their efficacy in a rat model of massive hepatectomy. This effect appears to be mediated, in part, by increased vascular endothelial growth factor (VEGF) production by the preconditioned MSCs as well as the injured liver. Neutralizing VEGF antibodies ameliorated the benefit of hypoxia-preconditioned MSCs, establishing VEGF as a key mediator of their benefit. This novel approach merits further exploration both mechanistically and to establish the functional advantages of MSCs in other injury settings.
Collapse
|
39
|
Chiba H, Ataka K, Iba K, Nagaishi K, Yamashita T, Fujimiya M. Diabetes impairs the interactions between long-term hematopoietic stem cells and osteopontin-positive cells in the endosteal niche of mouse bone marrow. Am J Physiol Cell Physiol 2013; 305:C693-703. [PMID: 23885062 DOI: 10.1152/ajpcell.00400.2012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hematopoietic stem cells (HSCs) are maintained, and their division/proliferation and quiescence are regulated in the microenvironments, niches, in the bone marrow. Although diabetes is known to induce abnormalities in HSC mobilization and proliferation through chemokine and chemokine receptors, little is known about the interaction between long-term HSCs (LT-HSCs) and osteopontin-positive (OPN) cells in endosteal niche. To examine this interaction, LT-HSCs and OPN cells were isolated from streptozotocin-induced diabetic and nondiabetic mice. In diabetic mice, we observed a reduction in the number of LT-HSCs and OPN cells and impaired expression of Tie2, β-catenin, and N-cadherin on LT-HSCs and β1-integrin, β-catenin, angiopoietin-1, and CXCL12 on OPN cells. In an in vitro coculture system, LT-HSCs isolated from nondiabetic mice exposed to diabetic OPN cells showed abnormal mRNA expression levels of Tie2 and N-cadherin. Conversely, in LT-HSCs derived from diabetic mice exposed to nondiabetic OPN cells, the decreased mRNA expressions of Tie2, β-catenin, and N-cadherin were restored to normal levels. The effects of diabetic or nondiabetic OPN cells on LT-HSCs shown in this coculture system were confirmed by the coinjection of LT-HSCs and OPN cells into bone marrow of irradiated nondiabetic mice. Our results provide new insight into the treatment of diabetes-induced LT-HSC abnormalities and suggest that the replacement of OPN cells may represent a novel treatment strategy.
Collapse
Affiliation(s)
- Hironori Chiba
- Department of Orthopedic Surgery. Sapporo Medical University Faculty of Medicine, Sapporo, Japan, and
| | | | | | | | | | | |
Collapse
|
40
|
The evolving role of the aryl hydrocarbon receptor (AHR) in the normophysiology of hematopoiesis. Stem Cell Rev Rep 2013; 8:1223-35. [PMID: 22628113 DOI: 10.1007/s12015-012-9384-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In addition to its role as a toxicological signal mediator, the Aryl Hydrocarbon Receptor (AHR) is also a transcription factor known to regulate cellular responses to oxidative stress and inflammation through transcriptional regulation of molecules involved in the signaling of nucear factor-erythroid 2-related factor-2 (Nrf2), p53 (TRP53), retinoblastoma (RB1), and NFκB. Recent research suggests that AHR activation of these signaling pathways may provide the molecular basis for understanding AHR's evolving role in endogenous developmental functions during hematopoietic stem-cell maintenance and differentiation. Recent developments into the hematopoietic roles for AHR are reviewed, aiming to reconcile divergent findings as to the endogenous function of AHR in hematopoiesis. Potential mechanistic explanations for AHR's involvement in hematopoietic differentiation are discussed, focusing on its known role as a cell cycle mediator and its interactions with Hypoxia-inducible transcription factor-1 alpha (HIF1-α). Understanding the physiological mechanisms of AHR activation and signaling have far reaching implications ranging from explaining the action of various toxicological agents to providing novel ways to expand stem cell populations ex vivo for use in transplant therapies.
Collapse
|
41
|
Pakravan N. Tumorigenesis: cell defense against hypoxia? Oncol Rev 2013; 7:e1. [PMID: 25992222 PMCID: PMC4419620 DOI: 10.4081/oncol.2013.e1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 03/02/2013] [Accepted: 04/02/2013] [Indexed: 01/01/2023] Open
Abstract
Microenvironmental elements can directly contribute to the induction and the maintenance of tumor. Oxygen is the main element in the cell microenvironment and hypoxia can affect the process of tumorigenesis. In response to hypoxia, cells change their pattern and characteristics. These changes suggest that it is not just adaptation, but some sort of cell defense against hypoxia. If hypoxia is corrected, then cell defense mechanisms are interrupted. An examination of the process of tumorigenesis helps to design better therapeutic strategies. A systematic review of the English literature was conducted by searching PubMed, Google Scholar, and ISI Web databases for studies on changes that defend and help cells to live in a hypoxic microenvironment. Cells respond to hypoxia by de-differentiation and an increase in heat shock proteins. Angiogenesis and deviation of inflammatory response in favor of hypoxic cell survival also defend and save the oxygen-starved cells from death. Finally, anti-angiogenic therapies and more hypoxia enhance metastasis, as tumors with low oxygen concentration are more malignant than tumors with high oxygen concentration. All these enable cells to migrate away from low oxygen areas and seek a more conducive microenvironment. Therapies that make the microenvironment more hypoxic need to be revised. This has been done for anti-angiogenic therapies, previously considered to be anti-tumor approaches. Effective therapies may be correcting therapies which direct the tumor microenvironment towards natural physical/chemical condition. Correcting therapies either bring back tumor cells to a normal form (correct tumor cells) or help the immune system to eradicate tumor cells which can not be corrected.
Collapse
Affiliation(s)
- Nafiseh Pakravan
- Department of Pathobiology, Medical School, Alborz University of Medical Sciences , Karaj, Iran
| |
Collapse
|
42
|
Williams K, Motiani K, Giridhar PV, Kasper S. CD44 integrates signaling in normal stem cell, cancer stem cell and (pre)metastatic niches. Exp Biol Med (Maywood) 2013; 238:324-38. [PMID: 23598979 PMCID: PMC11037417 DOI: 10.1177/1535370213480714] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The stem cell niche provides a regulatory microenvironment for cells as diverse as totipotent embryonic stem cells to cancer stem cells (CSCs) which exhibit stem cell-like characteristics and have the capability of regenerating the bulk of tumor cells while maintaining self-renewal potential. The transmembrane glycoprotein CD44 is a common component of the stem cell niche and exists as a standard isoform (CD44s) and a range of variant isoforms (CD44v) generated though alternative splicing. CD44 modulates signal transduction through post-translational modifications as well as interactions with hyaluronan, extracellular matrix molecules and growth factors and their cognate receptor tyrosine kinases. While the function of CD44 in hematopoietic stem cells has been studied in considerable detail, our knowledge of CD44 function in tissue-derived stem cell niches remains limited. Here we review CD44s and CD44v in both hematopoietic and tissue-derived stem cell niches, focusing on their roles in regulating stem cell behavior including self-renewal and differentiation in addition to cell-matrix interactions and signal transduction during cell migration and tumor progression. Determining the role of CD44 and CD44v in normal stem cell, CSC and (pre)metastatic niches and elucidating their unique functions could provide tools and therapeutic strategies for treating diseases as diverse as fibrosis during injury repair to cancer progression.
Collapse
Affiliation(s)
- Karin Williams
- Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, OH 45267
| | - Karan Motiani
- Division of Urology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267
| | | | - Susan Kasper
- Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, OH 45267
| |
Collapse
|
43
|
Impact of lenalidomide on the functional properties of human mesenchymal stromal cells. Exp Hematol 2012; 40:867-76. [DOI: 10.1016/j.exphem.2012.06.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 05/15/2012] [Accepted: 06/11/2012] [Indexed: 12/11/2022]
|
44
|
Microcapillary culture method: a novel tool for in vitro expansion of stem cells from scarce sources. Arch Med Res 2012; 43:423-30. [PMID: 22959977 DOI: 10.1016/j.arcmed.2012.08.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 07/05/2012] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIMS Although increasing numbers of studies report the derivation of stem cells from a variety of different tissues, derivation efficiencies greatly vary among different studies even for the same tissue source. Hence, a consistent and efficient isolation protocol has not yet been established to date. Several factors have so far been documented that influence and limit mesenchymal stem cell (MSC) isolation and cultivation, including the age and gender of the tissue donor, origin of the tissue, amount of sampled tissue material and cell culture characteristics including the choice of basal media, serum, gas composition, etc. The aim of the study was to investigate the microcapillary culture method (MCM) to establish an efficient and consistent isolation as well as cultivation protocol by comparing the results with other classic culture systems (flasks, center wells). METHODS MSCs isolated from adipose tissue of different donors were observed comparatively under different culture systems (flasks, center wells, microcapillary tubes) and their proliferation and differentiation were investigated. Flow cytometry was used for immunophenotypic characterization of derived cells and histochemical staining (Oil Red O and Alizarin Red S) was applied for determining their differentiation capacity. RESULTS It has been shown for the first time that AD-MSCs can consistently and efficiently be derived from a scarce amount of adipose tissue by MCM. CONCLUSIONS Further and similar studies should be performed to determine whether this methodology can also be applicable for other MSC sources.
Collapse
|
45
|
Razban V, Lotfi AS, Soleimani M, Ahmadi H, Massumi M, Khajeh S, Ghaedi M, Arjmand S, Najavand S, Khoshdel A. HIF-1α Overexpression Induces Angiogenesis in Mesenchymal Stem Cells. Biores Open Access 2012; 1:174-83. [PMID: 23514846 PMCID: PMC3559201 DOI: 10.1089/biores.2012.9905] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Stem cell therapy continues to be an innovative and promising strategy for heart failure. Stem cell injection alone, however, is hampered by poor cell survival and differentiation. This study was aimed to explore the possibility of improving stem cell therapy through genetic modification of stem cells, in order for them to promote angiogenesis in an auto- and paracrine manner under hypoxic conditions. Hypoxia inducible factor-1α was overexpressed in bone marrow-derived mesenchymal stem cells (MSCs) by stable transduction using a lentiviral vector. Under hypoxic and normoxic conditions, the vascular endothelial growth factor (VEGF) concentration in the cells' supernatant was measured by an enzyme-linked immunosorbent assay. Migration was assayed by wound healing and c-Met expression by flow cytometry. Tube formation was evaluated on a Matrigel basement membrane. The concentration of VEGF was significantly increased in the supernatant of HIF-1α-overexpressing MSCs; this medium was significantly more effective in inducing endothelial cell migration compared to untransduced MSCs. Transduced cells showed increased levels of c-Met expression and were more efficient at tube formation. However, no indication of differentiation toward an endothelial phenotype was observed. This study indicated that genetic modification of MSCs by HIF-1α overexpression has the potential to improve components of the angiogenesis process under a hypoxic condition by paracrine and autocrine mechanisms.
Collapse
Affiliation(s)
- Vahid Razban
- National Institute of Genetic Engineering and Biotechnology (NIGEB) , Tehran, Iran
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Stopp S, Bornhäuser M, Ugarte F, Wobus M, Kuhn M, Brenner S, Thieme S. Expression of the melanoma cell adhesion molecule in human mesenchymal stromal cells regulates proliferation, differentiation, and maintenance of hematopoietic stem and progenitor cells. Haematologica 2012; 98:505-13. [PMID: 22801967 DOI: 10.3324/haematol.2012.065201] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The melanoma cell adhesion molecule defines mesenchymal stromal cells in the human bone marrow that regenerate bone and establish a hematopoietic microenvironment in vivo. The role of the melanoma cell adhesion molecule in primary human mesenchymal stromal cells and the maintenance of hematopoietic stem and progenitor cells during ex vivo culture has not yet been demonstrated. We applied RNA interference or ectopic overexpression of the melanoma cell adhesion molecule in human mesenchymal stromal cells to evaluate the effect of the melanoma cell adhesion molecule on their proliferation and differentiation as well as its influence on co-cultivated hematopoietic stem and progenitor cells. Knockdown and overexpression of the melanoma cell adhesion molecule affected several characteristics of human mesenchymal stromal cells related to osteogenic differentiation, proliferation, and migration. Furthermore, knockdown of the melanoma cell adhesion molecule in human mesenchymal stromal cells stimulated the proliferation of hematopoietic stem and progenitor cells, and strongly reduced the formation of long-term culture-initiating cells. In contrast, melanoma cell adhesion molecule-overexpressing human mesenchymal stromal cells provided a supportive microenvironment for hematopoietic stem and progenitor cells. Expression of the melanoma cell adhesion molecule increased the adhesion of hematopoietic stem and progenitor cells to human mesenchymal stromal cells and their migration beneath the monolayer of human mesenchymal stromal cells. Our results demonstrate that the expression of the melanoma cell adhesion molecule in human mesenchymal stromal cells determines their fate and regulates the maintenance of hematopoietic stem and progenitor cells through direct cell-cell contact.
Collapse
Affiliation(s)
- Sabine Stopp
- Medical Clinic and Policlinic I, Dresden, Germany
| | | | | | | | | | | | | |
Collapse
|
47
|
Ribeiro AJS, Tottey S, Taylor RWE, Bise R, Kanade T, Badylak SF, Dahl KN. Mechanical characterization of adult stem cells from bone marrow and perivascular niches. J Biomech 2012; 45:1280-7. [PMID: 22349118 DOI: 10.1016/j.jbiomech.2012.01.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 01/20/2012] [Accepted: 01/29/2012] [Indexed: 12/11/2022]
Abstract
Therapies using adult stem cells often require mechanical manipulation such as injection or incorporation into scaffolds. However, force-induced rupture and mechanosensitivity of cells during manipulation is largely ignored. Here, we image cell mechanical structures and perform a biophysical characterization of three different types of human adult stem cells: bone marrow CD34+ hematopoietic, bone marrow mesenchymal and perivascular mesenchymal stem cells. We use micropipette aspiration to characterize cell mechanics and quantify deformation of subcellular structures under force and its contribution to global cell deformation. Our results suggest that CD34+ cells are mechanically suitable for injection systems since cells transition from solid- to fluid-like at constant aspiration pressure, probably due to a poorly developed actin cytoskeleton. Conversely, mesenchymal stem cells from the bone marrow and perivascular niches are more suitable for seeding into biomaterial scaffolds since they are mechanically robust and have developed cytoskeletal structures that may allow cellular stable attachment and motility through solid porous environments. Among these, perivascular stem cells cultured in 6% oxygen show a developed cytoskeleton but a more compliant nucleus, which can facilitate the penetration into pores of tissues or scaffolds. We confirm the relevance of our measurements using cell motility and migration assays and measure survival of injected cells. Since different types of adult stem cells can be used for similar applications, we suggest considering mechanical properties of stem cells to match optimal mechanical characteristics of therapies.
Collapse
Affiliation(s)
- Alexandre J S Ribeiro
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburg, PA 15213, United States
| | | | | | | | | | | | | |
Collapse
|
48
|
Cools J. A fly's view of the hematopoietic niche. Haematologica 2012; 97:155. [PMID: 22298818 DOI: 10.3324/haematol.2012.062182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
49
|
Darzynkiewicz Z, Balazs EA. Genome integrity, stem cells and hyaluronan. Aging (Albany NY) 2012; 4:78-88. [PMID: 22383371 PMCID: PMC3314170 DOI: 10.18632/aging.100438] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2012] [Accepted: 02/24/2012] [Indexed: 12/12/2022]
Abstract
Faithful preservation of genome integrity is the critical mission of stem cells as well as of germ cells. Reviewed are the following mechanisms involved in protecting DNA in these cells: (a) The efflux machinery that can pump out variety of genotoxins in ATP-dependent manner; (b) the mechanisms maintaining minimal metabolic activity which reduces generation of reactive oxidants, by-products of aerobic respiration; (c) the role of hypoxic niche of stem cells providing a gradient of variable oxygen tension; (d)(e) the presence of hyaluronan (HA) and HA receptors on stem cells and in the niche; (f) the role of role of HA in protecting DNA from oxidative damage; (g) the specific role of HA that may play a role protecting DNA in stem cells; (h) the interactions of HA with sperm cells and oocytes that also may shield their DNA from oxidative damage, and (e) mechanisms by which HA exerts the anti-oxidant activity. While HA has multitude of functions its anti-oxidant capabilities are often overlooked but may be of significance in preservation of integrity of stem and germ cells genome.
Collapse
Affiliation(s)
- Zbigniew Darzynkiewicz
- Brander Cancer Research Institute & Department of Pathology, New York Medical College, Valhalla, NY 10595, USA.
| | | |
Collapse
|