1
|
Burzi IS, Parchi PD, Barachini S, Pardini E, Sardo Infirri G, Montali M, Petrini I. Hypoxia Promotes the Stemness of Mesangiogenic Progenitor Cells and Prevents Osteogenic but not Angiogenic Differentiation. Stem Cell Rev Rep 2024; 20:1830-1842. [PMID: 38914791 PMCID: PMC11457687 DOI: 10.1007/s12015-024-10749-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2024] [Indexed: 06/26/2024]
Abstract
The stem cell niche in the bone marrow is a hypoxic environment, where the low oxygen tension preserves the pluripotency of stem cells. We have identified mesangiogenic progenitor cells (MPC) exhibiting angiogenic and mesenchymal differentiation capabilities in vitro. The effect of hypoxia on MPC has not been previously explored. In this study, MPCs were isolated from volunteers' bone marrow and cultured under both normoxic and hypoxic conditions (3% O2). MPCs maintained their characteristic morphology and surface marker expression (CD18 + CD31 + CD90-CD73-) under hypoxia. However, hypoxic conditions led to reduced MPC proliferation in primary cultures and hindered their differentiation into mesenchymal stem cells (MSCs) upon exposure to differentiative medium. First passage MSCs derived from MPC appeared unaffected by hypoxia, exhibiting no discernible differences in proliferative potential or cell cycle. However, hypoxia impeded the subsequent osteogenic differentiation of MSCs, as evidenced by decreased hydroxyapatite deposition. Conversely, hypoxia did not impact the angiogenic differentiation potential of MPCs, as demonstrated by spheroid-based assays revealing comparable angiogenic sprouting and tube-like formation capabilities under both hypoxic and normoxic conditions. These findings indicate that hypoxia preserves the stemness phenotype of MPCs, inhibits their differentiation into MSCs, and hampers their osteogenic maturation while leaving their angiogenic potential unaffected. Our study sheds light on the intricate effects of hypoxia on bone marrow-derived MPCs and their differentiation pathways.
Collapse
Affiliation(s)
- Irene Sofia Burzi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Via Savi 2, 56125, Pisa, Italy
| | - Paolo Domenico Parchi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Via Savi 2, 56125, Pisa, Italy
| | - Serena Barachini
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56125, Pisa, Italy
| | - Eleonora Pardini
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Via Savi 2, 56125, Pisa, Italy
| | - Gisella Sardo Infirri
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Via Savi 2, 56125, Pisa, Italy
| | - Marina Montali
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56125, Pisa, Italy
| | - Iacopo Petrini
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Via Savi 2, 56125, Pisa, Italy.
| |
Collapse
|
2
|
Kumar S, Yadav V, Sharma N, Sethi A. HypoxamiR-210-3p regulates mesenchymal stem cells proliferation via P53 & Akt. Mol Cell Biochem 2024; 479:2119-2129. [PMID: 37620743 DOI: 10.1007/s11010-023-04834-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023]
Abstract
Transplanted stem cells (˃95%) into ischemic myocardium die because of unfavourable conditions. Moreover, hypoxia role in the cell cycle regulation has been studied in transformed/immortalized cell lines which may have altered cell cycle regulators and/or mutated and, can't be transplanted in patients. We quest to find out the mechanism of cell cycle regulation in mesenchymal stem cells (MSC) to regulate its survival and proliferation in repair processes. Additionally, critically analysed role of hypoxamiR-210-3p, and cell cycle regulators that can regulate cell proliferation under hypoxic conditions. Bone marrow-derived MSC (BM-MSC) isolated from young male Fischer-344 rats by flushing the cavity of femur and propagated in vitro under 1% hypoxia for 72 h showed an increased in cell proliferation ( > 30%, p < 0.05) compared to normoxia. miR-210-3p, role in cell proliferation under hypoxic condition was confirmed by knockdown. Loss of function studies with transfection of anti-mir-210-3p, we observed decrease in proliferation of BM-MSC under hypoxia. Furthermore, BM-MSC proliferation due to miR-210-3p was confirmed using CFSE assay and flow cytometry, in which more cells were observed in S-phase. Mechanistically, western blot analysis showed miR-210-3p inhibition upregulates p53 and p21 expression and subsequent decrease in pAkt under hypoxia. On contrary, CFSE and Western blot under normoxic conditions showed downregulation of p53 and p21 whilst upregulation of pAkt indicated the key role of miR-210-3p in BM-MSC proliferation. Our results demonstrate the role of miR-210-3p in BM-MSC proliferation under both hypoxic and normoxic conditions and illustrate the potential mechanism via the regulation of pAkt, p53 and p21.
Collapse
Affiliation(s)
- Sanjay Kumar
- Department of Biochemistry, Medical College, All India Institute of Medical Sciences, Bathinda, India.
- Division of Regenerative Medicine, Department of Pathology and Laboratory Medicine, Center of Excellence (CoE) Cardiovascular Diseases, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45229, USA.
- Department of Biochemistry, Medical College, All India Institute of Medical Sciences, Bathinda, Punjab, 151001, India.
| | - Varsha Yadav
- Department of Biochemistry, Medical College, All India Institute of Medical Sciences, Bathinda, India
| | - Namrta Sharma
- Department of Biochemistry, Medical College, All India Institute of Medical Sciences, Bathinda, India
| | - Anshika Sethi
- Department of Biochemistry, Medical College, All India Institute of Medical Sciences, Bathinda, India
| |
Collapse
|
3
|
谢 鸥, 李 珊, 罗 洋, 王 乐. [Protective effects of 2-methoxyestradiol against hypoxic pulmonary hypertension in neonatal rats]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:757-764. [PMID: 39014954 PMCID: PMC11562046 DOI: 10.7499/j.issn.1008-8830.2401078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/31/2024] [Indexed: 07/18/2024]
Abstract
OBJECTIVES To investigate the protective effects of 2-methoxyestradiol (2ME) against hypoxic pulmonary hypertension (HPH) in neonatal rats. METHODS Ninety-six Wistar neonatal rats were randomly divided into a normoxia group, a hypoxia group, and a hypoxia + 2ME group, with each group further subdivided into 3-day, 7-day, 14-day, and 21-day subgroups, containing eight rats each. The hypoxia and hypoxia + 2ME groups received daily subcutaneous injections of saline and 2ME (240 μg/kg), respectively, while the normoxia group was raised in a normoxic environment with daily saline injections. Right ventricular systolic pressure (RVSP) was measured using the direct pressure method. Pulmonary vascular morphology was assessed using hematoxylin and eosin staining, with metrics including the percentage of medial thickness of small pulmonary arteries relative to the external diameter (MT%) and the cross-sectional area of the media of small pulmonary arteries relative to the total cross-sectional area (MA%). Immunohistochemistry was used to detect the expression levels of hypoxia-inducible factor-1α (HIF-1α) and proliferating cell nuclear antigen (PCNA) proteins, while real-time quantitative PCR was used to to assess HIF-1α and PCNA mRNA levels. RESULTS Compared to the normoxia group, the hypoxia and hypoxia + 2ME groups showed increased RVSP and upregulated HIF-1α and PCNA protein and mRNA expression levels at 3, 7, 14, and 21 days after hypoxia (P<0.05). Furthermore, at 7, 14, and 21 days after hypoxia, the hypoxia group showed increased MT% and MA% (P<0.05). In comparison to the hypoxia group, the hypoxia + 2ME group exhibited reduced RVSP and downregulated HIF-1α and PCNA protein and mRNA expression levels, along with decreased MT% and MA% at 7, 14, and 21 days after hypoxia (P<0.05). CONCLUSIONS 2ME may protect against HPH in neonatal rats by inhibiting the expression of HIF-1α and PCNA and reducing pulmonary vascular remodeling. Citation:Chinese Journal of Contemporary Pediatrics, 2024, 26(7): 757-764.
Collapse
Affiliation(s)
| | | | | | - 乐 王
- 新疆医科大学第一附属医院新生儿科,新疆乌鲁木齐830054
| |
Collapse
|
4
|
Abraham M, Kori I, Vishwakarma U, Goel S. Comprehensive assessment of goat adipose tissue-derived mesenchymal stem cells cultured in different media. Sci Rep 2024; 14:8380. [PMID: 38600175 PMCID: PMC11006890 DOI: 10.1038/s41598-024-58465-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 03/29/2024] [Indexed: 04/12/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have demonstrated potential in treating livestock diseases that are unresponsive to conventional therapies. MSCs derived from goats, a valuable model for studying orthopaedic disorders in humans, offer insights into bone formation and regeneration. Adipose tissue-derived MSCs (ADSCs) are easily accessible and have a high capacity for expansion. Although the choice of culture media significantly influences the biological properties of MSCs, the optimal media for goat ADSCs (gADSCs) remains unclear. This study aimed to assess the effects of four commonly used culture media on gADSCs' culture characteristics, stem cell-specific immunophenotype, and differentiation. Results showed that MEM, DMEM/F12, and DMEM-LG were superior in maintaining cell morphology and culture parameters of gADSCs, such as cell adherence, metabolic activity, colony-forming potential, and population doubling. Conversely, DMEM-HG exhibited poor performance across all evaluated parameters. The gADSCs cultured in DMEM/F12 showed enhanced early proliferation and lower apoptosis. The cell surface marker distribution exhibited superior characteristics in gADSCs cultured in MEM and DMEM/F12. In contrast, the distribution was inferior in gADSCs cultured in DMEM-LG. DMEM/F12 and DMEM-LG culture media demonstrated a significantly higher potential for chondrogenic differentiation and DMEM-LG for osteogenic differentiation. In conclusion, DMEM/F12 is a suitable culture medium for propagating gADSCs as it effectively maintains cell morphology, growth parameters, proliferation and lower apoptosis while exhibiting desirable expression patterns of MSC-specific markers. These findings contribute to optimising culture conditions for gADSCs, enhancing their potential applications in disease treatment and regenerative medicine.
Collapse
Affiliation(s)
- Michelle Abraham
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
| | - Ibraz Kori
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
- DBT-Regional Centre for Biotechnology (RCB), Faridabad, Haryana, India
| | - Utkarsha Vishwakarma
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
- DBT-Regional Centre for Biotechnology (RCB), Faridabad, Haryana, India
| | - Sandeep Goel
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India.
- DBT-Regional Centre for Biotechnology (RCB), Faridabad, Haryana, India.
| |
Collapse
|
5
|
Lacher SE, Skon-Hegg C, Ruis BL, Krznarich J, Slattery M. An antioxidant response element regulates the HIF1α axis in breast cancer cells. Free Radic Biol Med 2023; 204:243-251. [PMID: 37179033 PMCID: PMC10321210 DOI: 10.1016/j.freeradbiomed.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023]
Abstract
The redox sensitive transcription factor NRF2 is a central regulator of the transcriptional response to reactive oxygen species (ROS). NRF2 is widely recognized for its ROS-responsive upregulation of antioxidant genes that are essential for mitigating the damaging effects of oxidative stress. However, multiple genome-wide approaches have suggested that NRF2's regulatory reach extends well beyond the canonical antioxidant genes, with the potential to regulate many noncanonical target genes. Recent work from our lab and others suggests HIF1A, which encodes the hypoxia-responsive transcription factor HIF1α, is one such noncanonical NRF2 target. These studies found that NRF2 activity is associated with high HIF1A expression in multiple cellular contexts, HIF1A expression is partially dependent on NRF2, and there is a putative NRF2 binding site (antioxidant response element, or ARE) approximately 30 kilobases upstream of HIF1A. These findings all support a model in which HIF1A is a direct target of NRF2, but did not confirm the functional importance of the upstream ARE in HIF1A expression. Here we use CRISPR/Cas9 genome editing to mutate this ARE in its genomic context and test the impact on HIF1A expression. We find that mutation of this ARE in a breast cancer cell line (MDA-MB-231) eliminates NRF2 binding and decreases HIF1A expression at the transcript and protein levels, and disrupts HIF1α target genes as well as phenotypes driven by these HIF1α targets. Taken together, these results indicate that this NRF2 targeted ARE plays an important role in the expression of HIF1A and activity of the HIF1α axis in MDA-MB-231 cells.
Collapse
Affiliation(s)
- Sarah E Lacher
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA.
| | - Cara Skon-Hegg
- Whiteside Institute for Clinical Research, St. Luke's Hospital, University of Minnesota Medical School, Duluth, MN, 55812, USA
| | - Brian L Ruis
- Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Jennifer Krznarich
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA
| | - Matthew Slattery
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA.
| |
Collapse
|
6
|
Rahimi B, Panahi M, Lotfi H, Khalili M, Salehi A, Saraygord-Afshari N, Alizadeh E. Sodium selenite preserves rBM-MSCs' stemness, differentiation potential, and immunophenotype and protects them against oxidative stress via activation of the Nrf2 signaling pathway. BMC Complement Med Ther 2023; 23:131. [PMID: 37098557 PMCID: PMC10127330 DOI: 10.1186/s12906-023-03952-7] [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: 09/11/2022] [Accepted: 04/10/2023] [Indexed: 04/27/2023] Open
Abstract
BACKGROUND The physiological level of reactive oxygen species (ROS) is necessary for many cellular functions. However, during the in-vitro manipulations, cells face a high level of ROS, leading to reduced cell quality. Preventing this abnormal ROS level is a challenging task. Hence, here we evaluated the effect of sodium selenite supplementation on the antioxidant potential, stemness capacity, and differentiation of rat-derived Bone Marrow MSCs (rBM-MSCs) and planned to check our hypothesis on the molecular pathways and networks linked to sodium selenite's antioxidant properties. METHODS MTT assay was used to assess the rBM-MSCs cells' viability following sodium selenite supplementation (concentrations of: 0.001, 0.01, 0.1, 1, 10 µM). The expression level of OCT-4, NANOG, and SIRT1 was explored using qPCR. The adipocyte differentiation capacity of MSCs was checked after Sodium Selenite treatment. The DCFH-DA assay was used to determine intracellular ROS levels. Sodium selenite-related expression of HIF-1α, GPX, SOD, TrxR, p-AKT, Nrf2, and p38 markers was determined using western blot. Significant findings were investigated by the String tool to picture the probable molecular network. RESULTS Media supplemented with 0.1 µM sodium selenite helped to preserve rBM-MSCs multipotency and keep their surface markers presentation; this also reduced the ROS level and improved the rBM-MSCs' antioxidant and stemness capacity. We observed enhanced viability and reduced senescence for rBM-MSCs. Moreover, sodium selenite helped in rBM-MSCs cytoprotection by regulating the expression of HIF-1 of AKT, Nrf2, SOD, GPX, and TrxR markers. CONCLUSIONS We showed that sodium selenite could help protect MSCs during in-vitro manipulations, probably via the Nrf2 pathway.
Collapse
Affiliation(s)
- Bahareh Rahimi
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences (IUMS), Shahid Hemmat Highway, Tehran, 1449614535, Iran
| | - Mohammad Panahi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajie Lotfi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mostafa Khalili
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Astireh Salehi
- Biology Department, Islamic Azad University, Sanandaj Branch, Sanandaj, Iran
| | - Neda Saraygord-Afshari
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences (IUMS), Shahid Hemmat Highway, Tehran, 1449614535, Iran.
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
7
|
Li J, Li X, Guo Q. Drug Resistance in Cancers: A Free Pass for Bullying. Cells 2022; 11:3383. [PMID: 36359776 PMCID: PMC9654341 DOI: 10.3390/cells11213383] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 08/13/2023] Open
Abstract
The cancer burden continues to grow globally, and drug resistance remains a substantial challenge in cancer therapy. It is well established that cancerous cells with clonal dysplasia generate the same carcinogenic lesions. Tumor cells pass on genetic templates to subsequent generations in evolutionary terms and exhibit drug resistance simply by accumulating genetic alterations. However, recent evidence has implied that tumor cells accumulate genetic alterations by progressively adapting. As a result, intratumor heterogeneity (ITH) is generated due to genetically distinct subclonal populations of cells coexisting. The genetic adaptive mechanisms of action of ITH include activating "cellular plasticity", through which tumor cells create a tumor-supportive microenvironment in which they can proliferate and cause increased damage. These highly plastic cells are located in the tumor microenvironment (TME) and undergo extreme changes to resist therapeutic drugs. Accordingly, the underlying mechanisms involved in drug resistance have been re-evaluated. Herein, we will reveal new themes emerging from initial studies of drug resistance and outline the findings regarding drug resistance from the perspective of the TME; the themes include exosomes, metabolic reprogramming, protein glycosylation and autophagy, and the relates studies aim to provide new targets and strategies for reversing drug resistance in cancers.
Collapse
Affiliation(s)
| | | | - Qie Guo
- The Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| |
Collapse
|
8
|
Nie Z, Chen M, Gao Y, Huang D, Cao H, Peng Y, Guo N, Wang F, Zhang S. Ferroptosis and Tumor Drug Resistance: Current Status and Major Challenges. Front Pharmacol 2022; 13:879317. [PMID: 35668934 PMCID: PMC9163417 DOI: 10.3389/fphar.2022.879317] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Ferroptosis is a novel type of regulated cell death, whose unique metabolic characteristics are commonly used to evaluate the conditions of various diseases especially in tumors. Accumulating evidence supports that ferroptosis can regulate tumor development, metastasis, and therapeutic responses. Considering to the important role of chemotherapy in tumor treatment, drug resistance has become the most serious challenge. Revealing the molecular mechanism of ferroptosis is expected to solve tumor drug resistance and find new therapies to treat cancers. In this review, we discuss the relationship between ferroptosis and tumor drug resistance, summarize the abnormal ferroptosis in tissues of different cancer types and current research progress and challenges in overcoming treatment resistance, and explore the concept of targeting ferroptosis to improve tumor treatment outcomes.
Collapse
Affiliation(s)
- Zhenyu Nie
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Mei Chen
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Yuanhui Gao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Denggao Huang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Hui Cao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Yanling Peng
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Na Guo
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Fei Wang
- Department of Urology, Hainan General Hospital, Affiliated Hainan Hospital of Hainan Medical University, Haikou, China
- *Correspondence: Shufang Zhang, ; Fei Wang,
| | - Shufang Zhang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
- *Correspondence: Shufang Zhang, ; Fei Wang,
| |
Collapse
|
9
|
Expression of proliferation-related genes in BM-MSC-treated ALL cells in hypoxia condition is regulated under the influence of epigenetic factors in-vitro. Med Oncol 2022; 39:88. [PMID: 35581482 DOI: 10.1007/s12032-022-01671-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/12/2022] [Indexed: 10/18/2022]
Abstract
Mesenchymal stem cells affect ALL cell biology under hypoxic conditions. We studied survival, proliferation, expression, and promoter methylation levels of essential genes involved in expanding MOLT-4 cells co-cultured with BM-MSC under the hypoxic condition. Here, MOLT-4 cells were co-cultured with BMMSCs under hypoxic conditions. First, the apoptosis rate was evaluated by Flow cytometry. Then, MOLT-4 cells' proliferation rate was assessed using MTT assay, and the expressions and methylation rates of genes were determined by qRT-PCR and MS-qPCR, respectively. The results showed that although MOLT-4 cells proliferation and survival rates were reduced under hypoxic conditions, this reduction was not statistically significant. Also, we showed that hypoxic conditions caused upregulation of candidate genes and affected their methylation status. Besides, it was revealed that Pontin was downregulated, while KDM3A, SKP2, and AURKA had an upward trend in the presence of MOLT-4 cells plus BM-MSC. The co-culture of leukemia cells with BMMSCs under hypoxic conditions may be a potential therapeutic approach for ALL.
Collapse
|
10
|
Hypoxia Induces DPSC Differentiation versus a Neurogenic Phenotype by the Paracrine Mechanism. Biomedicines 2022; 10:biomedicines10051056. [PMID: 35625792 PMCID: PMC9138575 DOI: 10.3390/biomedicines10051056] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/28/2022] [Accepted: 04/28/2022] [Indexed: 12/10/2022] Open
Abstract
As previously described by several authors, dental pulp stem cells (DPSCs), when adequately stimulated, may acquire a neuronal-like phenotype acting as a favorable source of stem cells in the generation of nerves. Besides, it is known that hypoxia conditioning is capable of stimulating cell differentiation as well as survival and self-renewal, and that multiple growth factors, including Epidermal Growth factor (EGF) and basic fibroblast growth factor (bFGF), are often involved in the induction of the neuronal differentiation of progenitor cells. In this work, we investigated the role of hypoxia in the commitment of DPSCs into a neuronal phenotype. These cells were conditioned with hypoxia (O2 1%) for 5 and 16 days; subsequently, we analyzed the proliferation rate and morphology, and tested the cells for neural and stem markers. Moreover, we verified the possible autocrine/paracrine role of DPSCs in the induction of neural differentiation by comparing the secretome profile of the hypoxic and normoxic conditioned media (CM). Our results showed that the hypoxia-mediated DPSC differentiation was time dependent. Moreover, conditioned media (CM derived from DPSCs stimulated by hypoxia were able, in turn, to induce the neural differentiation of SH-SY5Y neuroblastoma cells and undifferentiated DPSCs. In conclusion, under the herein-mentioned conditions, hypoxia seems to favor the differentiation of DPSCs into neuron-like cells. In this way, we confirm the potential clinical utility of differentiated neuronal DPSCs, and we also suggest the even greater potential of CM-derived-hypoxic DPSCs that could more readily be used in regenerative therapies.
Collapse
|
11
|
Wu D, Liu L, Fu S, Zhang J. Osteostatin improves the Osteogenic differentiation of mesenchymal stem cells and enhances angiogenesis through HIF-1α under hypoxia conditions in vitro. Biochem Biophys Res Commun 2022; 606:100-107. [PMID: 35339748 DOI: 10.1016/j.bbrc.2022.02.085] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Hypoxia conditions induced by bone defects would prolong the duration of bone regeneration. The effect of osteostatin (OST) on the osteogenic differentiation of mesenchymal stem cells (MSCs) and angiogenesis under hypoxia conditions remain unexplored. METHODS SPF mice were obtained, and MSCs were isolated from bone marrow. MSCs were treated with 1% oxygen for hypoxia induction, and 200 nM of OST was used to treat cells under nomorxia or hypoxia conditions. Cell proliferation was evaluated using CCK8 assay, and trypan blue staining was implemented for determining cell death ratio. Alkaline phosphatase activity and alizarin redS staining was conducted to histologically evaluated osteogenic differentiation. Flow cytometry was used for the detection of CD31hiEmcnhi cells (Type H ECs), whose migration was detected by Transwell assay and angiogenesis was measured by tube formation assay. Protein level was measured by western blotting and mRNA level was monitored via RT-qPCR. RESULTS The MSC proliferation was enhanced by OST under hypoxia conditions. The osteogenic differentiation of MSCs was decreased under hypoxia conditions, and treatment of OST significantly reversed its inhibitory effect. The hypoxia treated culture medium of MSCs promoted the proliferation, migration, and angiogenesis of type H ECs, while the effects were further strengthened by OST addition. HIF-1α was found to be upregulated in hypoxia treated MSCs, whereas silencing of HIF-1α had reversed effects on the angiogenic capacity of Type H ECs. CONCLUSION OST improved the proliferation and osteogenic differentiation of MSCs and further promoted angiogenesis of type H ECs through upregulating HIF-1α expression.
Collapse
Affiliation(s)
- Dongjin Wu
- Department of Spine Surgery, The Second Hospital of Shandong University, Shandong, China
| | - Liyan Liu
- Department of Nephrology, The Fifth People's Hospital of Jinan, Shandong, China
| | - Shenglong Fu
- Department of Orthopaedics, The Fifth People's Hospital of Jinan, Shandong, China
| | - Jun Zhang
- Department of Orthopaedics, The Fifth People's Hospital of Jinan, Shandong, China.
| |
Collapse
|
12
|
Wang P, Zhu P, Yu C, Wu J. The Proliferation and Stemness of Peripheral Blood-Derived Mesenchymal Stromal Cells Were Enhanced by Hypoxia. Front Endocrinol (Lausanne) 2022; 13:873662. [PMID: 35634504 PMCID: PMC9134856 DOI: 10.3389/fendo.2022.873662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/05/2022] [Indexed: 01/08/2023] Open
Abstract
This study aimed to address the dilemma of low peripheral blood-derived mesenchymal stromal cell (PBMSC) activity and reduced phenotype in bone or cartilage tissue engineering. Rat PBMSCs (rPBMSCs) were obtained by density gradient centrifugation, and stromal cell characteristics were confirmed by flow cytometry (FCM) and multi-differentiation potential induction experiments. Cell growth curve, viability experiments, and clone formation experiments were performed by [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] (MTS) and cell counting, and the cell cycle was confirmed by cell FCM. The proliferation signal pathway and stemness-related proteins were detected by molecular methods including Western blot and real-time polymerase chain reaction. CD73, CD90, and CD105 were highly expressed, and CD14, CD19, CD34, CD45, and HLA-DR were barely expressed in rPBMSCs. rPBMSCs possessed the potential to differentiate into chondrocytes, adipocytes, and osteoblasts under their respective induction conditions. Cell growth curve and viability experiments were performed under hypoxic conditions: 19% O2, 5% O2, and 1% O2. Specifically, 5% O2 accelerated the proliferation and expression of the stemness of PBMSCs. Cycle experiments proved that hypoxia promoted the cell transition from the G1 phase to the S phase. Molecular experiments confirmed that 5% O2 hypoxia significantly elevated the expressions of hypoxia-inducible factor 1α and β-catenin and simultaneously the expressions of cycle-related genes including CyclinE/CDK2 and stemness-related genes including Nanog and SOX2. The appropriate concentration of hypoxia (i.e., 5% O2) enhanced the proliferation and stemness of rPBMSCs and increased the multidirectional differentiation potential of stromal cells. The proposed culture method could improve the viability and maintain the phenotype of rPBMSCs in cartilage or bone tissue engineering.
Collapse
Affiliation(s)
- Pengzhen Wang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
- *Correspondence: Pengzhen Wang,
| | - Pingping Zhu
- Department of Neurology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Chaosheng Yu
- Department of Otorhinolaryngology, Guangzhou Red Cross Medicine, Jinan University, Guangzhou, China
| | - Jian Wu
- Department of Otorhinolaryngology, Guangzhou Red Cross Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
13
|
Li Y, Wei Y, Gu L. Effect of hypoxia on proliferation and glucocorticoid resistance of T-cell acute lymphoblastic leukaemia. ACTA ACUST UNITED AC 2021; 26:775-784. [PMID: 34565306 DOI: 10.1080/16078454.2021.1980689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Hypoxia is emerging as a key factor in the biology of leukaemia. Here, we want to clarify the impact of hypoxia on the proliferation of T-cell acute lymphoblastic leukaemia (T-ALL) cells and the response to chemotherapy. METHODS T-ALL cells were cultured under normoxic and hypoxic conditions. MTT assay and trypan blue staining technique was used to detect cell viability and proliferation. In vitro sensitivity to glucocorticoid was assessed by IC50. CDI was used to analyze the combined effects of glucocorticoid and hypoxia. Flow cytometry was performed to detect apoptosis and cell cycle. Western blotting was performed to detect the protein expression associated with hypoxia. RESULTS Hypoxia of 1% O2 resulted different impact on cell viability and proliferation to different T-ALL cell lines, reduced, unaffected or induced, according to their different metabolic phenotype. All the cell lines showed an induction of key enzymes in glycolysis pathway following hypoxia exposure, although different effector proteins were induced in different cell lines. In GC-sensitive cells, acute hypoxia made no effect on the IC50 of dexamethasone, but chronic hypoxia may improve cell survival and induce GC resistance. However, acute hypoxia induced a higher GC resistance in GC-resistant T-ALL cells and showed an antagonistic effect while combined with high-dose dexamethasone. CONCLUSION T-ALL cells adapt well to hypoxic environment. Hypoxia may influence leukaemic cell proliferation. More importantly, hypoxia contributes to GC resistance in T-ALL blasts, especially in refractory/relapsed T-ALL.
Collapse
Affiliation(s)
- Yuanyuan Li
- Laboratory of Hematology/Oncology, Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Joint laboratory of West China Second University Hospital, Sichuan University and School of Life Science, Fudan University for Pulmonary Development and Disease, Chengdu, People's Republic of China.,NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, People's Republic of China
| | - Yi Wei
- West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Ling Gu
- Laboratory of Hematology/Oncology, Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Joint laboratory of West China Second University Hospital, Sichuan University and School of Life Science, Fudan University for Pulmonary Development and Disease, Chengdu, People's Republic of China.,NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
14
|
Feng XD, Zhu JQ, Zhou JH, Lin FY, Feng B, Shi XW, Pan QL, Yu J, Li LJ, Cao HC. Hypoxia-inducible factor-1α-mediated upregulation of CD99 promotes the proliferation of placental mesenchymal stem cells by regulating ERK1/2. World J Stem Cells 2021; 13:317-330. [PMID: 33959221 PMCID: PMC8080541 DOI: 10.4252/wjsc.v13.i4.317] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/11/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND As human placenta-derived mesenchymal stem cells (hP-MSCs) exist in a physiologically hypoxic microenvironment, various studies have focused on the influence of hypoxia. However, the underlying mechanisms remain to be further explored. AIM The aim was to reveal the possible mechanisms by which hypoxia enhances the proliferation of hP-MSCs. METHODS A hypoxic cell incubator (2.5% O2) was used to mimic a hypoxic microenvironment. Cell counting kit-8 and 5-ethynyl-20-deoxyuridine incorporation assays were used to assay the proliferation of hP-MSCs. The cell cycle was profiled by flow cytometry. Transcriptome profiling of hP-MSCs under hypoxia was performed by RNA sequencing. CD99 mRNA expression was assayed by reverse transcription-polymerase chain reaction. Small interfering RNA-mediated hypoxia-inducible factor 1α (HIF-1α) or CD99 knockdown of hP-MSCs, luciferase reporter assays, and the ERK1/2 signaling inhibitor PD98059 were used in the mechanistic analysis. Protein expression was assayed by western blotting; immunofluorescence assays were conducted to evaluate changes in expression levels. RESULTS Hypoxia enhanced hP-MSC proliferation, increased the expression of cyclin E1, cyclin-dependent kinase 2, and cyclin A2, and decreased the expression of p21. Under hypoxia, CD99 expression was increased by HIF-1α. CD99-specific small interfering RNA or the ERK1/2 signaling inhibitor PD98059 abrogated the hypoxia-induced increase in cell proliferation. CONCLUSION Hypoxia promoted hP-MSCs proliferation in a manner dependent on CD99 regulation of the MAPK/ERK signaling pathway in vitro.
Collapse
Affiliation(s)
- Xu-Dong Feng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Jia-Qi Zhu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Jia-Hang Zhou
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Fei-Yan Lin
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Bing Feng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Xiao-Wei Shi
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Qiao-Ling Pan
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Jiong Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Lan-Juan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Hong-Cui Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China.
| |
Collapse
|
15
|
Labedz-Maslowska A, Bryniarska N, Kubiak A, Kaczmarzyk T, Sekula-Stryjewska M, Noga S, Boruczkowski D, Madeja Z, Zuba-Surma E. Multilineage Differentiation Potential of Human Dental Pulp Stem Cells-Impact of 3D and Hypoxic Environment on Osteogenesis In Vitro. Int J Mol Sci 2020; 21:ijms21176172. [PMID: 32859105 PMCID: PMC7504399 DOI: 10.3390/ijms21176172] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023] Open
Abstract
Human dental pulp harbours unique stem cell population exhibiting mesenchymal stem/stromal cell (MSC) characteristics. This study aimed to analyse the differentiation potential and other essential functional and morphological features of dental pulp stem cells (DPSCs) in comparison with Wharton’s jelly-derived MSCs from the umbilical cord (UC-MSCs), and to evaluate the osteogenic differentiation of DPSCs in 3D culture with a hypoxic microenvironment resembling the stem cell niche. Human DPSCs as well as UC-MSCs were isolated from primary human tissues and were subjected to a series of experiments. We established a multiantigenic profile of DPSCs with CD45−/CD14−/CD34−/CD29+/CD44+/CD73+/CD90+/CD105+/Stro-1+/HLA-DR− (using flow cytometry) and confirmed their tri-lineage osteogenic, chondrogenic, and adipogenic differentiation potential (using qRT-PCR and histochemical staining) in comparison with the UC-MSCs. The results also demonstrated the potency of DPSCs to differentiate into osteoblasts in vitro. Moreover, we showed that the DPSCs exhibit limited cardiomyogenic and endothelial differentiation potential. Decreased proliferation and metabolic activity as well as increased osteogenic differentiation of DPSCs in vitro, attributed to 3D cell encapsulation and low oxygen concentration, were also observed. DPSCs exhibiting elevated osteogenic potential may serve as potential candidates for a cell-based product for advanced therapy, particularly for bone repair. Novel tissue engineering approaches combining DPSCs, 3D biomaterial scaffolds, and other stimulating chemical factors may represent innovative strategies for pro-regenerative therapies.
Collapse
Affiliation(s)
- Anna Labedz-Maslowska
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.L.-M.); (N.B.); (A.K.); (S.N.); (Z.M.)
| | - Natalia Bryniarska
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.L.-M.); (N.B.); (A.K.); (S.N.); (Z.M.)
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Andrzej Kubiak
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.L.-M.); (N.B.); (A.K.); (S.N.); (Z.M.)
- Institute of Nuclear Physics, Polish Academy of Sciences, 31-342 Krakow, Poland
| | - Tomasz Kaczmarzyk
- Department of Oral Surgery, Faculty of Medicine, Jagiellonian University Medical College, 31-155 Krakow, Poland;
| | - Malgorzata Sekula-Stryjewska
- Laboratory of Stem Cell Biotechnology, Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland;
| | - Sylwia Noga
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.L.-M.); (N.B.); (A.K.); (S.N.); (Z.M.)
- Laboratory of Stem Cell Biotechnology, Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland;
| | | | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.L.-M.); (N.B.); (A.K.); (S.N.); (Z.M.)
| | - Ewa Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (A.L.-M.); (N.B.); (A.K.); (S.N.); (Z.M.)
- Correspondence: ; Tel.: +48-12-664-61-80
| |
Collapse
|
16
|
Wang W, Wang F, Hao R, Wang A, Sharshov K, Druzyaka A, Lancuo Z, Shi Y, Feng S. First de novo whole genome sequencing and assembly of the bar-headed goose. PeerJ 2020; 8:e8914. [PMID: 32292659 PMCID: PMC7144584 DOI: 10.7717/peerj.8914] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 03/15/2020] [Indexed: 12/23/2022] Open
Abstract
Background The bar-headed goose (Anser indicus) mainly inhabits the plateau wetlands of Asia. As a specialized high-altitude species, bar-headed geese can migrate between South and Central Asia and annually fly twice over the Himalayan mountains along the central Asian flyway. The physiological, biochemical and behavioral adaptations of bar-headed geese to high-altitude living and flying have raised much interest. However, to date, there is still no genome assembly information publicly available for bar-headed geese. Methods In this study, we present the first de novo whole genome sequencing and assembly of the bar-headed goose, along with gene prediction and annotation. Results 10X Genomics sequencing produced a total of 124 Gb sequencing data, which can cover the estimated genome size of bar-headed goose for 103 times (average coverage). The genome assembly comprised 10,528 scaffolds, with a total length of 1.143 Gb and a scaffold N50 of 10.09 Mb. Annotation of the bar-headed goose genome assembly identified a total of 102 Mb (8.9%) of repetitive sequences, 16,428 protein-coding genes, and 282 tRNAs. In total, we determined that there were 63 expanded and 20 contracted gene families in the bar-headed goose compared with the other 15 vertebrates. We also performed a positive selection analysis between the bar-headed goose and the closely related low-altitude goose, swan goose (Anser cygnoides), to uncover its genetic adaptations to the Qinghai-Tibetan Plateau. Conclusion We reported the currently most complete genome sequence of the bar-headed goose. Our assembly will provide a valuable resource to enhance further studies of the gene functions of bar-headed goose. The data will also be valuable for facilitating studies of the evolution, population genetics and high-altitude adaptations of the bar-headed geese at the genomic level.
Collapse
Affiliation(s)
- Wen Wang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xi'ning, Qinghai, China
| | - Fang Wang
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xi'ning, Qinghai, China
| | - Rongkai Hao
- Novogene Bioinformatics Institute, Beijing, China
| | - Aizhen Wang
- College of Eco-Environmental Engineering, Qinghai University, Xi'ning, Qinghai, China
| | - Kirill Sharshov
- Research Institute of Experimental and Clinical Medicine, Novosibirsk, Russia
| | - Alexey Druzyaka
- Institute of Systematics and Ecology of Animals, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Zhuoma Lancuo
- School of Finance and Economics, Qinghai University, Xi'ning, Qinghai, China
| | - Yuetong Shi
- KunLun College of Qinghai University, Xi'ning, Qinghai, China
| | - Shuo Feng
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xi'ning, Qinghai, China
| |
Collapse
|
17
|
Jung AR, Park YH, Kim GE, Kim MY, Jeon SH, Kim HY, Kim SY, Oh SH, Lee JY. Stem Cell/Oxygen-Releasing Microparticle Enhances Erectile Function in a Cavernous Nerve Injury Model. Tissue Eng Part A 2020; 27:50-62. [PMID: 32122268 DOI: 10.1089/ten.tea.2019.0240] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Erectile dysfunction caused by damage to the cavernous nerve is a common complication of radical prostatectomy for patients with localized prostate cancer. Various studies have investigated repair of damaged tissue and prevention of fibrosis in the corpus cavernosum using stem cell therapy. However, stem cell therapy has limitations, including insufficient nutrient and oxygen supply to transplanted stem cells. This study investigated whether stem cell/oxygen-releasing hollow microparticles (HPs) had therapeutic effect on erectile dysfunction in a rat model of bilateral cavernous nerve injury (BCNI). Therapeutic effects were observed in the BCNI model at 1, 2, and 4 weeks postcavernous nerve injury. Erectile function further improved after treatment with stem cell/oxygen-releasing HP system compared with treatment with only stem cells at 4 weeks. Stem cell/oxygen-releasing HP system increased cyclic guanosine monophosphate (cGMP) level and neuronal nitric oxide synthase (nNOS), endothelial nitric oxide synthase (eNOS), α-smooth muscle actin (α-SMA), and muscarinic acetylcholine receptor 3 (M3) expression while decreasing fibrosis and apoptosis in the corpus cavernosum. Our results clearly show that stem cell survival increases around transplanted stem cell/oxygen-releasing hybrid system site. Taken together, an oxygen-releasing HP system supported prolonged stem cell survival, sustaining the paracrine effect of the stem cells, and consequently enhancing erectile function. These findings show promise with regard to prolonged stem cell survival in stem cell applications for various diseases and types of tissue damage. Impact statement In this study, we used an oxygen-releasing hollow microparticles (HPs) system with stem cells to attempt to overcome certain limitations of stem cell therapy, including insufficient nutrient and oxygen supplies for transplanted stem cells. Our results demonstrated that a stem cell/oxygen-releasing HP hybrid system could further improve erectile function, cyclic guanosine monophosphate (cGMP) level, and NOS level in a bilateral cavernous nerve injury rat model through prolonged stem cell survival. Our data suggest that a stem cell/oxygen-releasing HP system is a promising clinical treatment option for postprostatectomy erectile dysfunction. Furthermore, this system may be relevant in different disease therapies and regenerative medicine.
Collapse
Affiliation(s)
- Ae Ryang Jung
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Urology, Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yong Hyun Park
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ga Eun Kim
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Urology, Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mee Young Kim
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Urology, Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Hwan Jeon
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ho Yong Kim
- Department of Nanobiomedical Science, Dankook University, Cheonan, Republic of Korea
| | - So Young Kim
- Department of Nanobiomedical Science, Dankook University, Cheonan, Republic of Korea
| | - Se Heang Oh
- Department of Nanobiomedical Science, Dankook University, Cheonan, Republic of Korea.,Department of Pharmaceutical Engineering, Dankook University, Cheonan, Republic of Korea
| | - Ji Youl Lee
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Urology, Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
18
|
Hou W, Kaczorowski A, Lantwin P, Kippenberger M, Schütz V, Franke D, Dieffenbacher SC, Hohenfellner M, Duensing S. Microenvironment-Derived FGF-2 Stimulates Renal Cell Carcinoma Cell Proliferation through Modulation of p27Kip1: Implications for Spatial Niche Formation and Functional Intratumoral Heterogeneity. Pathobiology 2020; 87:114-124. [PMID: 32229735 DOI: 10.1159/000506709] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/20/2020] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND/OBJECTIVE Clear cell renal cell carcinoma (ccRCC) is characterized by a high degree of functional intratumoral heterogeneity (ITH). This is highlighted by the finding that tumor cell proliferation and intracellular signaling occur preferentially in the tumor periphery. The driving forces for such a spatial organization are largely unknown. Herein, we investigate the role of the tumor microenvironment in the control of tumor cell proliferation and functional ITH. METHODS Conditioned media (CM) derived from nonmalignant peritumoral kidney tissue were used to stimulate RCC cells in vitro. A neutralization assay was used to characterize the role of FGF-2 in the CM. The molecular mechanisms underlying the action of CM on RCC cells were investigated using immunoblotting, flow cytometry and immunofluorescence microscopy. Lastly, a series of ccRCCs were stained for Ki-67 and p27Kip1, and expression was analyzed in both tumor periphery and center. RESULTS We show that CM derived from nonmalignant kidney cells adjacent to an RCC can downregulate the expression of the CDK inhibitor p27Kip1 through enhanced protein degradation in an FGF-2-dependent fashion. FGF-2 functions mainly through the PI3K/AKT pathway downstream of its receptors, and RCC cells with constitutively high AKT activity show not only an enhanced degradation of p27Kip1 through the Emi1-Skp2 axis, but also a subcellular mislocalization of p27Kip1 to the cytoplasmic compartment. Such a mislocalization was also detected in the tumor periphery in vivo suggesting that p27Kip1 plays an important role in shaping this spatial niche. CONCLUSIONS Our results suggest that the tumor microenvironment is involved in shaping the tumor peripheral niche by stimulating the enhanced proliferation that is characteristic for this zone.
Collapse
Affiliation(s)
- Weibin Hou
- Section of Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Heidelberg, Germany
| | - Adam Kaczorowski
- Section of Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Heidelberg, Germany
| | - Philippa Lantwin
- Section of Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Heidelberg, Germany
| | - Maximilian Kippenberger
- Section of Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Heidelberg, Germany
| | - Viktoria Schütz
- Department of Urology, University Hospital Heidelberg, Heidelberg, Germany
| | - Desiree Franke
- Department of Urology, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | - Stefan Duensing
- Section of Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Heidelberg, Germany, .,Department of Urology, University Hospital Heidelberg, Heidelberg, Germany,
| |
Collapse
|
19
|
Succinate Supplement Elicited "Pseudohypoxia" Condition to Promote Proliferation, Migration, and Osteogenesis of Periodontal Ligament Cells. Stem Cells Int 2020; 2020:2016809. [PMID: 32215014 PMCID: PMC7085835 DOI: 10.1155/2020/2016809] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/01/2020] [Accepted: 02/25/2020] [Indexed: 01/03/2023] Open
Abstract
Most mesenchymal stem cells reside in a niche of low oxygen tension. Iron-chelating agents such as CoCl2 and deferoxamine have been utilized to mimic hypoxia and promote cell growth. The purpose of the present study was to explore whether a supplement of succinate, a natural metabolite of the tricarboxylic acid (TCA) cycle, can mimic hypoxia condition to promote human periodontal ligament cells (hPDLCs). Culturing hPDLCs in hypoxia condition promoted cell proliferation, migration, and osteogenic differentiation; moreover, hypoxia shifted cell metabolism from oxidative phosphorylation to glycolysis with accumulation of succinate in the cytosol and its release into culture supernatants. The succinate supplement enhanced hPDLC proliferation, migration, and osteogenesis with decreased succinate dehydrogenase (SDH) expression and activity, as well as increased hexokinase 2 (HK2) and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), suggesting metabolic reprogramming from oxidative phosphorylation to glycolysis in a normal oxygen condition. The succinate supplement in cell cultures promoted intracellular succinate accumulation while stabilizing hypoxia inducible factor-1α (HIF-1α), leading to a state of pseudohypoxia. Moreover, we demonstrate that hypoxia-induced proliferation was G-protein-coupled receptor 91- (GPR91-) dependent, while exogenous succinate-elicited proliferation involved the GPR91-dependent and GPR91-independent pathway. In conclusion, the succinate supplement altered cell metabolism in hPDLCs, induced a pseudohypoxia condition, and enhanced proliferation, migration, and osteogenesis of mesenchymal stem cells in vitro.
Collapse
|
20
|
Effects of Flow Rate on Mesenchymal Stem Cell Oxygen Consumption Rates in 3D Bone-Tissue-Engineered Constructs Cultured in Perfusion Bioreactor Systems. FLUIDS 2020. [DOI: 10.3390/fluids5010030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Bone grafts represent a multibillion-dollar industry, with over a million grafts occurring each year. Common graft types are associated with issues such as donor site morbidity in autologous grafts and immunological response in allogenic grafts. Bone-tissue-engineered constructs are a logical approach to combat the issues commonly encountered with these bone grafting techniques. When creating bone-tissue-engineered constructs, monitoring systems are required to determine construct characteristics, such as cellularity and cell type. This study aims to expand on the current predictive metrics for these characteristics, specifically analyzing the effects of media flow rate on oxygen uptake rates (OURs) of mesenchymal stem cells seeded on poly(L-lactic acid) (PLLA) scaffolds cultured in a flow perfusion bioreactor. To do this, oxygen consumption rates were measured for cell/scaffold constructs at varying flow rates ranging from 150 to 750 microliters per minute. Residence time analyses were performed for this bioreactor at these flow rates. Average observed oxygen uptake rates of stem cells in perfusion bioreactors were shown to increase with increased oxygen availability at higher flow rates. The residence time analysis helped identify potential pitfalls in current bioreactor designs, such as the presence of channeling. Furthermore, this analysis shows that oxygen uptake rates have a strong linear correlation with residence times of media in the bioreactor setup, where cells were seen to exhibit a maximum oxygen uptake rate of 3 picomoles O2/hr/cell.
Collapse
|
21
|
Lei Y, Chen T, Li Y, Shang M, Zhang Y, Jin Y, Yu Q, Guo F, Wang T. O-GlcNAcylation of PFKFB3 is required for tumor cell proliferation under hypoxia. Oncogenesis 2020; 9:21. [PMID: 32060258 PMCID: PMC7021673 DOI: 10.1038/s41389-020-0208-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 02/05/2020] [Accepted: 02/05/2020] [Indexed: 12/27/2022] Open
Abstract
The protein O-GlcNAcylation catalysed by O-GlcNAc transferase (OGT) is tightly regulated by glucose availability. It is upregulated and essential for tumor cell proliferation under hypoxic conditions. However, the mechanism behind is still unclear. Here, we showed that the glycolytic regulator 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB3), which also promotes cell cycle progression in the nucleus, was O-GlcNAcylated in response to hypoxia. The O-GlcNAcylation of PFKFB3 could compete phosphorylation by hypoxia-activated ERK at the same modification site Ser172. Phosphorylated PFKFB3 could interact with the protein G3BP2 and retain in the cytosol; this in turn led to the accumulation of hypoxia-induced-P27 in the nucleus resulting in the cell cycle arrest. Such a pathway was compromised by high level of PFKFB3 O-GlcNAcylation in tumor cells contributing to cell cycle progression. Consistently, the PFKFB3-Ser172 phosphorylation level inversely correlated with the OGT level in pancreatic cancer patients. Our findings uncovered an O-GlcNAcylation mediated mechanism to promote tumor cell proliferation under metabolic stress, linking the aberrant OGT activity to tumorigenesis in pancreatic cancer.
Collapse
Affiliation(s)
- Yinrui Lei
- Department of Pharmacology And Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.,Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Tao Chen
- Endoscopy Center, Shanghai East Hospital, Tongji University, 150Jimo Road, Shanghai, 200120, China
| | - Yeyi Li
- Department of Pharmacology And Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Man Shang
- Department of Pharmacology And Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yan Zhang
- Department of Pharmacology And Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yuepeng Jin
- Department of General Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Qiujing Yu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Fang Guo
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Ting Wang
- Department of Pharmacology And Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China. .,The Institute of Cell Metabolism and Disease, Shanghai Key Laboratory of Pancreatic Cancer, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 201620, China.
| |
Collapse
|
22
|
Khasawneh RR, Abu-El-Rub E, Serhan AO, Serhan BO, Abu-El-Rub H. Cross talk between 26S proteasome and mitochondria in human mesenchymal stem cells' ability to survive under hypoxia stress. J Physiol Sci 2019; 69:1005-1017. [PMID: 31679117 PMCID: PMC10716940 DOI: 10.1007/s12576-019-00720-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/16/2019] [Indexed: 12/19/2022]
Abstract
Mesenchymal stem cells (MSCs) are regarded as unique cells which play an imperative role in the field of regenerative medicine. They are characterized by the self-renewal capacity, multi-lineage differentiation abilities and immunomodulation properties which render them perfectly ideal cell type for treating a wide range of chronic diseases. Despite these enchanted features, there are many hurdles that need to be circumvented to ensure their long-term survival and viability after transplantation. Recently, hypoxia has been indicated as one of the most baffling stress conditions that can affect the survival rate of MSCs either positively or negatively depending on the level of hypoxia. MSCs can survive well under moderate hypoxia, but die shortly if they were exposed to severe hypoxia without clearly convincing explanation for this enigma. The current study reveals a novel mechanism of 26S proteasome in controlling the ability of BM-MSCs to withstand hypoxic stress by maintaining proper mitochondrial function. The results indicated that 26S proteasome remains functioning once BM- MSCs are exposed to moderate hypoxia (2.5%O2) and preserves their survival and proliferation mediated by intact mitochondrial performance, whereas 26S proteasome becomes inactive when BM-MSCs faces severe hypoxia that lead to poor mitochondrial function and less chance to survive longer. The outcomes of this study demonstrated the importance of 26S proteasome machinery in enhancing the resistance of BM-MSCs to hypoxic stress condition which may help in better planning future studies that target this system. Schematic representation summarizing the findings of the current study. 26S proteasome function preservation in normoxia and moderate hypoxia leads to maintain appropriate proliferation and mitochondrial activity in human BM-MSCs and promote their survival. On the opposite side, severe hypoxia disrupts the 26S proteasome function leading to significant reduction in the proliferation, survival and mitochondrial dynamics in human BM-MSCs causing their death.
Collapse
Affiliation(s)
- Ramada R Khasawneh
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Ejlal Abu-El-Rub
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan.
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada.
| | | | - Bashar Omar Serhan
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Hadeel Abu-El-Rub
- Forensic Medicine and Toxicology Department, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
23
|
Hypoxia and Hypoxia-Inducible Factors in Kidney Injury and Repair. Cells 2019; 8:cells8030207. [PMID: 30823476 PMCID: PMC6468851 DOI: 10.3390/cells8030207] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 02/21/2019] [Accepted: 02/25/2019] [Indexed: 02/07/2023] Open
Abstract
Acute kidney injury (AKI) is a major kidney disease characterized by an abrupt loss of renal function. Accumulating evidence indicates that incomplete or maladaptive repair after AKI can result in kidney fibrosis and the development and progression of chronic kidney disease (CKD). Hypoxia, a condition of insufficient supply of oxygen to cells and tissues, occurs in both acute and chronic kidney diseases under a variety of clinical and experimental conditions. Hypoxia-inducible factors (HIFs) are the "master" transcription factors responsible for gene expression in hypoxia. Recent researches demonstrate that HIFs play an important role in kidney injury and repair by regulating HIF target genes, including microRNAs. However, there are controversies regarding the pathological roles of HIFs in kidney injury and repair. In this review, we describe the regulation, expression, and functions of HIFs, and their target genes and related functions. We also discuss the involvement of HIFs in AKI and kidney repair, presenting HIFs as effective therapeutic targets.
Collapse
|
24
|
Tasev D, Dekker-Vroling L, van Wijhe M, Broxterman HJ, Koolwijk P, van Hinsbergh VWM. Hypoxia Impairs Initial Outgrowth of Endothelial Colony Forming Cells and Reduces Their Proliferative and Sprouting Potential. Front Med (Lausanne) 2018; 5:356. [PMID: 30619865 PMCID: PMC6306419 DOI: 10.3389/fmed.2018.00356] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 12/06/2018] [Indexed: 01/09/2023] Open
Abstract
Vascular homeostasis and regeneration in ischemic tissue relies on intrinsic competence of the tissue to rapidly recruit endothelial cells for vascularization. The mononuclear cell (MNC) fraction of blood contains circulating progenitors committed to endothelial lineage. These progenitors give rise to endothelial colony-forming cells (ECFCs) that actively participate in neovascularization of ischemic tissue. To evaluate if the initial clonal outgrowth of ECFCs from cord (CB) and peripheral blood (PB) was stimulated by hypoxic conditions, MNCs obtained from CB and PB were subjected to 20 and 1% O2 cell culture conditions. Clonal outgrowth was followed during a 30 day incubation period. Hypoxia impaired the initial outgrowth of ECFC colonies from CB and also reduced their number that were developing from PB MNCs. Three days of oxygenation (20% O2) prior to hypoxia could overcome the initial CB-ECFC outgrowth. Once proliferating and subcultured the CB-ECFCs growth was only modestly affected by hypoxia; proliferation of PB-ECFCs was reduced to a similar extent (18-30% reduction). Early passages of subcultured CB- and PB-ECFCs contained only viable cells and few if any senescent cells. Tube formation by subcultured PB-ECFCs was also markedly inhibited by continuous exposure to 1% O2. Gene expression profiles point to regulation of the cell cycle and metabolism as major altered gene clusters. Finally we discuss our counterintuitive observations in the context of the important role that hypoxia has in promoting neovascularization.
Collapse
Affiliation(s)
- Dimitar Tasev
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Laura Dekker-Vroling
- Department of Medical Oncology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Michiel van Wijhe
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Henk J Broxterman
- Department of Medical Oncology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Pieter Koolwijk
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Victor W M van Hinsbergh
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| |
Collapse
|
25
|
Su J, Fang M, Tian B, Luo J, Jin C, Wang X, Ning Z, Li X. Atorvastatin protects cardiac progenitor cells from hypoxia-induced cell growth inhibition via MEG3/miR-22/HMGB1 pathway. Acta Biochim Biophys Sin (Shanghai) 2018; 50:1257-1265. [PMID: 30481260 DOI: 10.1093/abbs/gmy133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Indexed: 12/14/2022] Open
Abstract
Heart failure (HF) induced by ischemia myocardial infarction (MI) is one of the major causes of morbidity and mortality all around the world. Atorvastatin, a hydroxymethylglutaryl coenzyme A reductase inhibitor, has been demonstrated to benefit patients with ischemic or non-ischemic-induced HF, but the mechanism is still poorly understood. Increasing evidence indicates that lncRNAs play important role in variety of human disease. However, the role and underlying molecular mechanisms remain largely unclear. In our work, we applied 0.5% O2 to generate a hypoxia cardiac progenitor cell (CPC) model. Then, CCK8 and EdU assays were employed to investigate the role of atorvastatin in hypoxia CPC cell model. We found that hypoxia inhibits CPC viability and proliferation through modulating MEG3 expression, while atorvastatin application can protect CPCs from hypoxia-induced injury through inhibiting MEG3 expression. Then, we demonstrated that repression of MEG3 inhibited the hypoxia-induced injury of CPCs and overexpression of MEG3 inhibited the protective effect of atorvastatin in the hypoxia-induced injury of CPCs. Furthermore, our study illustrated that atorvastatin played its role in CPC viability and proliferation by modulating the expression of HMGB1 through the MEG3/miR-22 pathway. Our study, for the first time, uncovered the molecular mechanism of atorvastatin's protective role in cardiomyocytes under hypoxia condition, which may provide an exploitable target in developing effective therapy drugs for MI patients.
Collapse
Affiliation(s)
- Jinwen Su
- Department of Cardiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201138, China
| | - Ming Fang
- Department of Cardiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201138, China
| | - Bei Tian
- Department of Cardiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201138, China
| | - Jun Luo
- Department of Cardiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201138, China
| | - Can Jin
- Department of Cardiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201138, China
| | - Xuejun Wang
- Department of Cardiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201138, China
| | - Zhongping Ning
- Department of Cardiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201138, China
| | - Xinming Li
- Department of Cardiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201138, China
| |
Collapse
|
26
|
Hypoxic culture enhances the expansion of rat bone marrow-derived mesenchymal stem cells via the regulatory pathways of cell division and apoptosis. In Vitro Cell Dev Biol Anim 2018; 54:666-676. [PMID: 30136033 DOI: 10.1007/s11626-018-0281-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/12/2018] [Indexed: 02/07/2023]
Abstract
This study aimed to examine the proliferative behavior and molecular mechanisms of rat bone marrow-derived MSCs (rBMSCs) cultured under three different oxygen concentrations. Passaged rBMSCs exhibited significantly greater proliferation rates at 1% O2 and 5% O2 than those at 18% O2 and the cells exposed to 1% O2 showed the highest proliferative potential, which was evidenced by the growth curves, colony-forming efficiencies, and CCK-8 absorbance values. The rBMSCs grown under hypoxic culture conditions (1% O2 and 5% O2) had the increased percentage of cells in S + G2/M-phase and the decreased apoptotic index, compared with normoxia (18% O2). It was revealed for the first time that there were more phosphohistone H3 (PHH3)-positive cells and higher expressions of proliferating cell nuclear antigen (PCNA) in the hypoxic cultures of rBMSCs than in the normoxic culture. Hypoxia upregulated the anti-apoptotic protein Bcl-2 and downregulated the pro-apoptotic proteins Bax and the cleaved caspase-3 in cultured rBMSCs. The levels of hypoxia-inducible factor-1α (HIF-1α) and phosphorylated extracellular signal-regulated kinase 1/2 (p-ERK1/2) were increased in the hypoxic-cultured rBMSCs. Nevertheless, no significant difference was observed in p53 level of rBMSCs between different oxygen concentrations. In conclusion, the hypoxia exerts a promoting effect on the in vitro expansion of rBMSCs via several signaling and molecular pathways involved in the control of cell cycle and apoptosis.
Collapse
|
27
|
Pacheco-Velázquez SC, Robledo-Cadena DX, Hernández-Reséndiz I, Gallardo-Pérez JC, Moreno-Sánchez R, Rodríguez-Enríquez S. Energy Metabolism Drugs Block Triple Negative Breast Metastatic Cancer Cell Phenotype. Mol Pharm 2018; 15:2151-2164. [DOI: 10.1021/acs.molpharmaceut.8b00015] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
| | | | | | | | - Rafael Moreno-Sánchez
- Departamento de Bioquímica, Instituto Nacional de Cardiología, 14080 Tlalpan, CDMX, Mexico
| | - Sara Rodríguez-Enríquez
- Departamento de Bioquímica, Instituto Nacional de Cardiología, 14080 Tlalpan, CDMX, Mexico
- Laboratorio de Medicina Traslacional, Instituto Nacional de Cancerología, 14080 Tlalpan, CDMX, Mexico
| |
Collapse
|
28
|
Comparative Study on In Vitro Culture of Mouse Bone Marrow Mesenchymal Stem Cells. Stem Cells Int 2018; 2018:6704583. [PMID: 29760732 PMCID: PMC5924976 DOI: 10.1155/2018/6704583] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 02/08/2018] [Accepted: 02/25/2018] [Indexed: 12/11/2022] Open
Abstract
In vitro culture of mesenchymal stem cells (MSCs) from mouse bone marrow (BM) has been hampered because of the low yield of MSCs during isolation and the contamination of hematopoietic cells during expansion. The lack of specific mouse BM-MSC markers increases the difficulty. Several techniques have been reported to improve the purity and in vitro growth of mouse BM-MSCs. However, systematic report on comparison of characteristics in primary BM-MSCs between different culture conditions is rare. Here, we studied the effects of oxygen concentrations and initial medium replacement intervals, along with cell passages, on mouse BM-MSCs isolated with differential adhesion method. BM-MSCs exhibited elevated proliferative and clonogenic abilities in 5% oxygen compared with 10% and 21% oxygen, as well as a better expression of the MSC marker Sca-1. Adipogenic and osteogenetic differentiation of BM-MSCs can be observed in both 21% and 5% oxygen. Adipogenic differentiation appeared stronger under normoxia conditions. BM-MSCs showed increased proliferative capacity and adipogenic/osteogenetic differentiation potential when initial medium replacement interval was 4 days compared with 1 day. As passage number increased, cells were more MSC-like in morphology and in expression of surface markers (positive for CD29, CD44, and Sca-1 and negative for CD11b, CD19, and CD45). These data provide new insight into optimizing the culture method and understanding the biological characteristics of mouse BM-MSCs during in vitro expansion.
Collapse
|
29
|
Tishevskaya NV, Babaeva AG, Gevorkyan NM. Effect of lymphocyte morphogenetic activity on organism reactivity and resistibility. Russ J Dev Biol 2018. [DOI: 10.1134/s106236041801006x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
30
|
Antony S, Jiang G, Wu Y, Meitzler JL, Makhlouf HR, Haines DC, Butcher D, Hoon DS, Ji J, Zhang Y, Juhasz A, Lu J, Liu H, Dahan I, Konate M, Roy KK, Doroshow JH. NADPH oxidase 5 (NOX5)-induced reactive oxygen signaling modulates normoxic HIF-1α and p27 Kip1 expression in malignant melanoma and other human tumors. Mol Carcinog 2017; 56:2643-2662. [PMID: 28762556 PMCID: PMC5675809 DOI: 10.1002/mc.22708] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 07/19/2017] [Accepted: 07/28/2017] [Indexed: 12/14/2022]
Abstract
NADPH oxidase 5 (NOX5) generated reactive oxygen species (ROS) have been implicated in signaling cascades that regulate cancer cell proliferation. To evaluate and validate NOX5 expression in human tumors, we screened a broad range of tissue microarrays (TMAs), and report substantial overexpression of NOX5 in malignant melanoma and cancers of the prostate, breast, and ovary. In human UACC-257 melanoma cells that possesses high levels of functional endogenous NOX5, overexpression of NOX5 resulted in enhanced cell growth, increased numbers of BrdU positive cells, and increased γ-H2AX levels. Additionally, NOX5-overexpressing (stable and inducible) UACC-257 cells demonstrated increased normoxic HIF-1α expression and decreased p27Kip1 expression. Similarly, increased normoxic HIF-1α expression and decreased p27Kip1 expression were observed in stable NOX5-overexpressing clones of KARPAS 299 human lymphoma cells and in the human prostate cancer cell line, PC-3. Conversely, knockdown of endogenous NOX5 in UACC-257 cells resulted in decreased cell growth, decreased HIF-1α expression, and increased p27Kip1 expression. Likewise, in an additional human melanoma cell line, WM852, and in PC-3 cells, transient knockdown of endogenous NOX5 resulted in increased p27Kip1 and decreased HIF-1α expression. Knockdown of endogenous NOX5 in UACC-257 cells resulted in decreased Akt and GSK3β phosphorylation, signaling pathways known to modulate p27Kip1 levels. In summary, our findings suggest that NOX5 expression in human UACC-257 melanoma cells could contribute to cell proliferation due, in part, to the generation of high local concentrations of extracellular ROS that modulate multiple pathways that regulate HIF-1α and networks that signal through Akt/GSK3β/p27Kip1 .
Collapse
Affiliation(s)
- Smitha Antony
- Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMaryland
| | - Guojian Jiang
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer InstituteBethesdaMaryland
| | - Yongzhong Wu
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer InstituteBethesdaMaryland
| | - Jennifer L. Meitzler
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer InstituteBethesdaMaryland
| | - Hala R. Makhlouf
- Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMaryland
| | - Diana C. Haines
- Pathology/Histotechnology Laboratory, Leidos Inc./Frederick National Laboratory for Cancer ResearchNational Cancer InstituteFrederickMaryland
| | - Donna Butcher
- Pathology/Histotechnology Laboratory, Leidos Inc./Frederick National Laboratory for Cancer ResearchNational Cancer InstituteFrederickMaryland
| | - Dave S. Hoon
- Department of Molecular OncologyJohn Wayne Cancer InstituteSanta MonicaCalifornia
| | - Jiuping Ji
- Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMaryland
| | - Yiping Zhang
- Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMaryland
| | - Agnes Juhasz
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer InstituteBethesdaMaryland
| | - Jiamo Lu
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer InstituteBethesdaMaryland
| | - Han Liu
- Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMaryland
| | - Iris Dahan
- Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMaryland
| | - Mariam Konate
- Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMaryland
| | - Krishnendu K. Roy
- Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMaryland
| | - James H. Doroshow
- Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMaryland
- Developmental Therapeutics Branch, Center for Cancer ResearchNational Cancer InstituteBethesdaMaryland
| |
Collapse
|
31
|
MGF E peptide pretreatment improves the proliferation and osteogenic differentiation of BMSCs via MEK-ERK1/2 and PI3K-Akt pathway under severe hypoxia. Life Sci 2017; 189:52-62. [DOI: 10.1016/j.lfs.2017.09.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 08/29/2017] [Accepted: 09/15/2017] [Indexed: 12/15/2022]
|
32
|
Parandavar E, Yazdanparast R. Differential impact of various reactive oxygen species (ROS) on HIF-1α/p53 direct interaction in SK-N-MC neuroblastoma cells. Cell Biosci 2017; 7:52. [PMID: 29051813 PMCID: PMC5633900 DOI: 10.1186/s13578-017-0180-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 09/30/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND A vital property of eukaryotic cells physiology is their rather quick response to variation of oxygen tension, mainly by a transcription factor known as hypoxia-inducible factor-1 (HIF-1). Aside from its transcriptional regulation, other mechanisms, such as post translational modifications and protein-protein interactions, the interaction between HIF-1α and p53 has attracted more attention mainly due to simultaneous enhancement in the protein levels of these two anti- and pro-apoptotic vital transcriptional factors within the ROS-stressed cells. METHODS In this study, we measured cell viability following exposure of the cells to H2O2, menadione and Cobalt Chloride by MTT, and ROS content was measured under the same condition. The immunoblotting technique has been used to establish the presence and amount of Caspase, HIF-1α and p53 proteins. Then, the effect of different ROS on interaction between HIF-1α and p53 proteins was examined by co-immunoprecipitation. RESULTS The results showed that cells viability and intracellular ROS content were modulated in response to menadione, H2O2 and Cobalt Chloride. These agents had different influence on HIF-1α signaling pathways as well as its interactions with p53 protein. It appeared that direct communication between HIF-1α and p53 proteins by ROS stresses, under both normoxic and hypoxic conditions, was governed by HIF-1α at a certain induced level. CONCLUSIONS Our data indicated that stabilization, a prerequisite for communication, of HIF-1α is dependent to the types of free radicals.
Collapse
Affiliation(s)
- Elham Parandavar
- Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384 Tehran, Iran
| | - Razieh Yazdanparast
- Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384 Tehran, Iran
| |
Collapse
|
33
|
Hypoxia induces the dysfunction of human endothelial colony-forming cells via HIF-1α signaling. Respir Physiol Neurobiol 2017; 247:87-95. [PMID: 28964937 DOI: 10.1016/j.resp.2017.09.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/29/2017] [Accepted: 09/22/2017] [Indexed: 12/15/2022]
Abstract
Endothelial injury is considered as a trigger of pulmonary vascular lesions in the pathogenesis of hypoxic pulmonary hypertension (HPH). Although endothelial colony-forming cells (ECFCs) have vascular regeneration potential to maintain endothelial integrity, hypoxia-induced precise alteration in ECFCs function remains controversial. This study investigated the impact of hypoxia on human ECFCs function in vitro and the underlying mechanism. We found that hypoxia inhibited ECFCs proliferation, migration and angiogenesis. Compared with no treatment, the expression of hypoxia inducible factor-1α (HIF-1α) in hypoxia-treated ECFCs was increased, with an up-regulation of p27 and a down-regulation of cyclin D1. The over-secreted vascular endothelial growth factor (VEGF) was detected, with the imbalanced expression of fetal liver kinase 1 (flk-1) and fms related tyrosine kinase 1 (flt-1). Hypoxia-induced changes in ECFCs could be reversed by HIF-1α inhibitor KC7F2. These data suggest that HIF-1α holds the key in regulating ECFCs function which may open a new perspective of ECFCs in HPH management.
Collapse
|
34
|
Hypoxia Enhances Cell Properties of Human Mesenchymal Stem Cells. Tissue Eng Regen Med 2017; 14:595-604. [PMID: 30603513 DOI: 10.1007/s13770-017-0068-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/04/2017] [Accepted: 06/29/2017] [Indexed: 12/18/2022] Open
Abstract
Atmospheric (in vitro) oxygen pressure is around 150 mm Hg (20% O2), whereas physiologic (in vivo) oxygen pressure ranges between 5 and 50 mm Hg (0.7-7% O2). The normoxic environment in cell culture does not refer to a physiological stem cell niche. The aim of this study is to investigate the effect of oxygen concentration on cell properties of human mesenchymal stem cells (MSCs). We analyzed cell proliferation rate, senescence, immunophenotype, stemness gene expression and differentiation potency with human urine stem cells (USCs), dental pulp stem cells (DPSCs), amniotic fluid stem cells (AFSCs), and bone marrow stromal cells (BMSCs). USCs, DPSCs, AFSCs and BMSCs were cultured under either 5% O2 hypoxic or 20% O2 normoxic conditions for 5 days. MSCs cultured under hypoxia showed significantly increased proliferation rate and high percentage of S-phase cells, compared to normoxic condition. In real-time PCR assay, the cells cultured under hypoxia expressed higher level of Oct4, C-Myc, Nanog, Nestin and HIF-1α. In immunophenotype analysis, MSCs cultured under hypoxia maintained higher level of the MSC surface markers, and lower hematopoietic markers. Senescence was inhibited under hypoxia. Hypoxia enhances osteogenic differentiation efficiency compared to normoxia. Hypoxia showed enhanced cell proliferation rate, retention of stem cell properties, inhibition of senescence, and increased differentiation ability compared to normoxia.
Collapse
|
35
|
Pezzi A, Amorin B, Laureano Á, Valim V, Dahmer A, Zambonato B, Sehn F, Wilke I, Bruschi L, Silva MALD, Filippi-Chiela E, Silla L. Effects Of Hypoxia in Long-Term In Vitro Expansion of Human Bone Marrow Derived Mesenchymal Stem Cells. J Cell Biochem 2017; 118:3072-3079. [PMID: 28240374 DOI: 10.1002/jcb.25953] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 02/23/2017] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSC) are considered multipotent stromal, non-hematopoietic cells with properties of self-renovation and differentiation. Optimal conditions for culture of MSC have been under investigation. The oxygen tension used for cultivation has been studied and appears to play an important role in biological behavior of mesenchymal cells. The aim is characterize MSC in hypoxia and normoxia conditions comparing their morphological and functional characteristics. Bone marrow-derived mesenchymal stem cells obtained from 15 healthy donors and cultured. MSC obtained from each donor were separated into two cultivation conditions normoxia (21% O2 ) and hypoxia (three donors at 1%, three donors at 2%, five donors at 3%, and four donors at 4% O2 ) up to second passage. MSC were evaluated for proliferation, differentiation, immunophenotyping, size and cell complexity, oxidative stress, mitochondrial activity, and autophagy. Culture conditions applied did not seem to affect immunophenotypic features and cellular plasticity. However, cells subjected to hypoxia showed smaller size and greater cellular complexity, besides lower proliferation (P < 0.002). Furthermore, cells cultured in low O2 tension had lower mitochondrial activity (P < 0.03) and a reduced tendency to autophagy, although oxidative stress did not vary among groups (P < 0.39). Oxygen tension seems to be a key regulator of cellular adaptation in vitro, and metabolic effects underlying this variable remain undescribed. Heterogeneity or even lack of results on the impact of oxygen concentration used for expanding MSC highlights the need for further research, in order to optimize conditions of cultivation and expansion and achieve greater safety and therapeutic efficacy. J. Cell. Biochem. 118: 3072-3079, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Annelise Pezzi
- Celular Therapy Center of Hospital de Clinicas de Porto Alegre, Center for Experimental Research, Porto Alegre, Brazil.,Post-graduation: Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Bruna Amorin
- Celular Therapy Center of Hospital de Clinicas de Porto Alegre, Center for Experimental Research, Porto Alegre, Brazil.,Centro Universitário Ritter dos Reis, Porto Alegre, Brazil
| | - Álvaro Laureano
- Celular Therapy Center of Hospital de Clinicas de Porto Alegre, Center for Experimental Research, Porto Alegre, Brazil.,Post-graduation: Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Vanessa Valim
- Celular Therapy Center of Hospital de Clinicas de Porto Alegre, Center for Experimental Research, Porto Alegre, Brazil.,Post-graduation: Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Alice Dahmer
- Celular Therapy Center of Hospital de Clinicas de Porto Alegre, Center for Experimental Research, Porto Alegre, Brazil.,Post-graduation: Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Bruna Zambonato
- Celular Therapy Center of Hospital de Clinicas de Porto Alegre, Center for Experimental Research, Porto Alegre, Brazil.,Post-graduation: Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Filipe Sehn
- Celular Therapy Center of Hospital de Clinicas de Porto Alegre, Center for Experimental Research, Porto Alegre, Brazil.,Post-graduation: Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Ianaê Wilke
- Celular Therapy Center of Hospital de Clinicas de Porto Alegre, Center for Experimental Research, Porto Alegre, Brazil.,Post-graduation: Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Maria Aparecida Lima da Silva
- Celular Therapy Center of Hospital de Clinicas de Porto Alegre, Center for Experimental Research, Porto Alegre, Brazil
| | | | - Lucia Silla
- Celular Therapy Center of Hospital de Clinicas de Porto Alegre, Center for Experimental Research, Porto Alegre, Brazil.,Post-graduation: Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,Hematology and Bone Marrow Transplantation of Hospital de Clinicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
36
|
Abstract
BACKGROUND Oxygen (O2) homeostasis is an indispensable requirement of eukaryotes. O2 concentration in cellular milieu is defined as normoxia (∼21% O2), physoxia (∼1-13% O2) or hypoxia (∼0.1-1% O2). Hypoxia, a striking micro-environmental feature in tumorigenesis, is countered by tumor cells via induction of O2 governed transcription factor, hypoxia inducible factor-1 (HIF-1). Post discovery, HIF-1 has emerged as a promising anticancer therapeutic target during the last two decades. Recent reports have highlighted that enhanced levels of HIF-1 correlate with tumor metastasis leading to poor patient prognosis. MATERIAL AND METHODS A systematic search in PubMed and SciFinder for the literature on HIF-1 biology and therapeutic importance in cancer was carried out. RESULTS This review highlights the initial description as well as the recent insights into HIF-1 biology and regulation. We have focused on emerging data regarding varied classes of HIF-1 target genes affecting various levels of crosstalk among tumorigenic pathways. We have emphasized on the fact that HIF-1 acts as a networking hub coordinating activities of multiple signaling molecules influencing tumorigenesis. Emerging evidences indicate role of many HIF-induced proteomic and genomic alterations in malignant progression by mediating a myriad of genes stimulating angiogenesis, anaerobic metabolism and survival of cancer cells in O2-deficient microenvironment. CONCLUSIONS Better understanding of the crucial role of HIF-1 in carcinogenesis could offer promising new avenues to researchers and aid in elucidating various open issues regarding the use of HIF-1 as an anticancer therapeutic target. In spite of large efforts in this field, many questions still remain unanswered. Hence, future investigations are necessary to devise, assess and refine methods for translating previous research efforts into novel clinical practices in cancer treatment.
Collapse
Affiliation(s)
- Sourabh Soni
- Pharmacology and Toxicology Lab, Food and Nutraceuticals Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India
- Academy of Scientific and Innovative Research, New Delhi, India
| | - Yogendra S. Padwad
- Pharmacology and Toxicology Lab, Food and Nutraceuticals Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India
- Academy of Scientific and Innovative Research, New Delhi, India
| |
Collapse
|
37
|
Ma C, Liu C, Li X, Lu T, Bai C, Fan Y, Guan W, Guo Y. Cryopreservation and multipotential characteristics evaluation of a novel type of mesenchymal stem cells derived from Small Tailed Han Sheep fetal lung tissue. Cryobiology 2017; 75:7-14. [PMID: 28284665 DOI: 10.1016/j.cryobiol.2017.03.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 03/06/2017] [Indexed: 10/20/2022]
Abstract
Lung mesenchymal stem cells (L-MSCs) characterized by plasticity, reduced relative immune privilege and high anti-fibrosis characteristics play the crucial role in lung tissue regenerative processes. However, up to date, the multi-differentiation potentials and application values of L-MSCs are still uncertain. In the current study, the Small Tailed Han Sheep embryo L-MSCs line from 12 samples, stocking 124 cryogenically-preserved vials, was successfully established by using primary culture and cell cryopreservation techniques. Isolated L-MSCs were morphologically consistent with fibroblasts, could be passaged for at least 18 passages and more than 91.8% of cells were diploid (2n = 54) analyze by G-banding. The majority of cells were in the G0/G1 phase (70.5-91.2%), and the growth curves were all typically sigmoidal. Moreover, L-MSCs were found to express pluripotent genes Oct4, Nanog and MSCs-associated genes β-integrin, CD29, CD44, CD71, CD73 and CD90, while the expressions of hematopoietic cell markers CD34 and CD45 were negative. In addtion, the L-MSCs could be differentiated into cells of three layers with induction medium in vitro, which confirmed their multilineage differentiation potential. The secretion of urea and ALB showed the differentiated hepatocytes still possessed the detoxification function. These results indicated that the isolated L-MSCs displayed typical characteristics of mesenchymal stem cells and that the culture conditions were suitable for their maintenance of stemness and their proliferation in vitro.
Collapse
Affiliation(s)
- Caiyun Ma
- Institute of Beijing Animal Science and Veterinary, Chinese Academy of Agricultural Science, Beijing, 100193, China
| | - Changqing Liu
- Institute of Beijing Animal Science and Veterinary, Chinese Academy of Agricultural Science, Beijing, 100193, China; Department of Life Science, Department of laboratory medicine, Bengbu Medical College, Bengbu, 233030, China
| | - Xiangchen Li
- Institute of Beijing Animal Science and Veterinary, Chinese Academy of Agricultural Science, Beijing, 100193, China
| | - Taofeng Lu
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China
| | - Chunyu Bai
- Institute of Beijing Animal Science and Veterinary, Chinese Academy of Agricultural Science, Beijing, 100193, China
| | - Yanan Fan
- Institute of Beijing Animal Science and Veterinary, Chinese Academy of Agricultural Science, Beijing, 100193, China
| | - Weijun Guan
- Institute of Beijing Animal Science and Veterinary, Chinese Academy of Agricultural Science, Beijing, 100193, China.
| | - Yu Guo
- Institute of Beijing Animal Science and Veterinary, Chinese Academy of Agricultural Science, Beijing, 100193, China; Department of Life Science, Department of laboratory medicine, Bengbu Medical College, Bengbu, 233030, China.
| |
Collapse
|
38
|
HIF-1α induction during reperfusion avoids maladaptive repair after renal ischemia/reperfusion involving miR127-3p. Sci Rep 2017; 7:41099. [PMID: 28106131 PMCID: PMC5247697 DOI: 10.1038/srep41099] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 12/15/2016] [Indexed: 12/11/2022] Open
Abstract
Ischemia/reperfusion (I/R) leads to Acute Kidney Injury. HIF-1α is a key factor during organ response to I/R. We previously demonstrated that HIF-1α is induced during renal reperfusion, after ischemia. Here we investigate the role of HIF-1α and the HIF-1α dependent mechanisms in renal repair after ischemia. By interference of HIF-1α in a rat model of renal I/R, we observed loss of expression and mis-localization of e-cadherin and induction of α-SMA, MMP-13, TGFβ, and collagen I. Moreover, we demonstrate that HIF-1α inhibition promotes renal cell infiltrates by inducing IL-1β, TNF-α, MCP-1 and VCAM-1, through NFkB activity. In addition, HIF-1α inhibition induced proximal tubule cells proliferation but it did not induce compensatory apoptosis, both in vivo. In vitro, HIF-1α knockdown in HK2 cells subjected to hypoxia/reoxygenation (H/R) promote cell entry into S phase, correlating with in vivo data. HIF-1α interference leads to downregulation of miR-127-3p and induction of its target gene Bcl6 in vivo. Moreover, modulation of miR-127-3p in HK2 cells subjected to H/R results in EMT regulation: miR127-3p inhibition promote loss of e-cadherin and induction of α-SMA and collagen I. In conclusion, HIF-1α induction during reperfusion is a protector mechanism implicated in a normal renal tissue repair after I/R.
Collapse
|
39
|
Sandvig I, Gadjanski I, Vlaski-Lafarge M, Buzanska L, Loncaric D, Sarnowska A, Rodriguez L, Sandvig A, Ivanovic Z. Strategies to Enhance Implantation and Survival of Stem Cells After Their Injection in Ischemic Neural Tissue. Stem Cells Dev 2017; 26:554-565. [PMID: 28103744 DOI: 10.1089/scd.2016.0268] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
High post-transplantation cell mortality is the main limitation of various approaches that are aimed at improving regeneration of injured neural tissue by an injection of neural stem cells (NSCs) and mesenchymal stromal cells (MStroCs) in and/or around the lesion. Therefore, it is of paramount importance to identify efficient ways to increase cell transplant viability. We have previously proposed the "evolutionary stem cell paradigm," which explains the association between stem cell anaerobic/microaerophilic metabolic set-up and stem cell self-renewal and inhibition of differentiation. Applying these principles, we have identified the main critical point in the collection and preparation of these cells for experimental therapy: exposure of the cells to atmospheric O2, that is, to oxygen concentrations that are several times higher than the physiologically relevant ones. In this way, the primitive anaerobic cells become either inactivated or adapted, through commitment and differentiation, to highly aerobic conditions (20%-21% O2 in atmospheric air). This inadvertently compromises the cells' survival once they are transplanted into normal tissue, especially in the hypoxic/anoxic/ischemic environment, which is typical of central nervous system (CNS) lesions. In addition to the findings suggesting that stem cells can shift to glycolysis and can proliferate in anoxia, recent studies also propose that stem cells may be able to proliferate in completely anaerobic or ischemic conditions by relying on anaerobic mitochondrial respiration. In this systematic review, we propose strategies to enhance the survival of NSCs and MStroCs that are implanted in hypoxic/ischemic neural tissue by harnessing their anaerobic nature and maintaining as well as enhancing their anaerobic properties via appropriate ex vivo conditioning.
Collapse
Affiliation(s)
- Ioanna Sandvig
- 1 Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ivana Gadjanski
- 2 Innovation Center, Faculty of Mechanical Engineering, University of Belgrade , Belgrade, Serbia .,3 Belgrade Metropolitan University , Belgrade, Serbia
| | - Marija Vlaski-Lafarge
- 4 French Blood Institute (EFS) , Aquitaine-Limousin Branch, Bordeaux, France .,5 U1035 INSERM/Bordeaux University , Bordeaux Cedex, France
| | - Leonora Buzanska
- 6 Stem Cell Bioengineering Unit, Mossakowski Medical Research Centre Polish Academy Sciences, Warsaw, Poland
| | - Darija Loncaric
- 4 French Blood Institute (EFS) , Aquitaine-Limousin Branch, Bordeaux, France .,5 U1035 INSERM/Bordeaux University , Bordeaux Cedex, France
| | - Ana Sarnowska
- 6 Stem Cell Bioengineering Unit, Mossakowski Medical Research Centre Polish Academy Sciences, Warsaw, Poland
| | - Laura Rodriguez
- 4 French Blood Institute (EFS) , Aquitaine-Limousin Branch, Bordeaux, France .,5 U1035 INSERM/Bordeaux University , Bordeaux Cedex, France
| | - Axel Sandvig
- 1 Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway .,7 Division of Pharmacology and Clinical Neurosciences, Department of Neurosurgery and Clinical Neurophysiology, Umeå University Hospital , Umeå, Sweden
| | - Zoran Ivanovic
- 4 French Blood Institute (EFS) , Aquitaine-Limousin Branch, Bordeaux, France .,5 U1035 INSERM/Bordeaux University , Bordeaux Cedex, France
| |
Collapse
|