1
|
Usseglio J, Dumur A, Pagès E, Renaudie É, Abélanet A, Brie J, Champion É, Magnaudeix A. Microporous Hydroxyapatite-Based Ceramics Alter the Physiology of Endothelial Cells through Physical and Chemical Cues. J Funct Biomater 2023; 14:460. [PMID: 37754874 PMCID: PMC10531673 DOI: 10.3390/jfb14090460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/15/2023] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
Incorporation of silicate ions in calcium phosphate ceramics (CPC) and modification of their multiscale architecture are two strategies for improving the vascularization of scaffolds for bone regenerative medicine. The response of endothelial cells, actors for vascularization, to the chemical and physical cues of biomaterial surfaces is little documented, although essential. We aimed to characterize in vitro the response of an endothelial cell line, C166, cultivated on the surface CPCs varying either in terms of their chemistry (pure versus silicon-doped HA) or their microstructure (dense versus microporous). Adhesion, metabolic activity, and proliferation were significantly altered on microporous ceramics, but the secretion of the pro-angiogenic VEGF-A increased from 262 to 386 pg/mL on porous compared to dense silicon-doped HA ceramics after 168 h. A tubulogenesis assay was set up directly on the ceramics. Two configurations were designed for discriminating the influence of the chemistry from that of the surface physical properties. The formation of tubule-like structures was qualitatively more frequent on dense ceramics. Microporous ceramics induced calcium depletion in the culture medium (from 2 down to 0.5 mmol/L), which is deleterious for C166. Importantly, this effect might be associated with the in vitro static cell culture. No influence of silicon doping of HA on C166 behavior was detected.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Amandine Magnaudeix
- Université de Limoges, CNRS, Institut de Recherche sur les Céramiques, UMR 7315, F-87000 Limoges, France; (J.U.); (A.D.); (E.P.); (É.R.); (A.A.); (J.B.); (É.C.)
| |
Collapse
|
2
|
Antonelli A, Scarpa ES, Bruzzone S, Astigiano C, Piacente F, Bruschi M, Fraternale A, Di Buduo CA, Balduini A, Magnani M. Anoxia Rapidly Induces Changes in Expression of a Large and Diverse Set of Genes in Endothelial Cells. Int J Mol Sci 2023; 24:ijms24065157. [PMID: 36982232 PMCID: PMC10049254 DOI: 10.3390/ijms24065157] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Sinusoidal endothelial cells are the predominant vascular surface of the bone marrow and constitute the functional hematopoietic niche where hematopoietic stem and progenitor cells receive cues for self-renewal, survival, and differentiation. In the bone marrow hematopoietic niche, the oxygen tension is usually very low, and this condition affects stem and progenitor cell proliferation and differentiation and other important functions of this region. Here, we have investigated in vitro the response of endothelial cells to a marked decrease in O2 partial pressure to understand how the basal gene expression of some relevant biological factors (i.e., chemokines and interleukins) that are fundamental for the intercellular communication could change in anoxic conditions. Interestingly, mRNA levels of CXCL3, CXCL5, and IL-34 genes are upregulated after anoxia exposure but become downmodulated by sirtuin 6 (SIRT6) overexpression. Indeed, the expression levels of some other genes (such as Leukemia Inhibitory Factor (LIF)) that were not significantly affected by 8 h anoxia exposure become upregulated in the presence of SIRT6. Therefore, SIRT6 mediates also the endothelial cellular response through the modulation of selected genes in an extreme hypoxic condition.
Collapse
Affiliation(s)
- Antonella Antonelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | | | - Santina Bruzzone
- Department of Experimental Medicine, Section of Biochemistry, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Cecilia Astigiano
- Department of Experimental Medicine, Section of Biochemistry, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Francesco Piacente
- Department of Experimental Medicine, Section of Biochemistry, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Michela Bruschi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Alessandra Fraternale
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | | | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
- Department of Biomedical Engineering, Tufts University in Boston, Boston, MA 02111, USA
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
- Correspondence:
| |
Collapse
|
3
|
Woods K, Guezguez B. Dynamic Changes of the Bone Marrow Niche: Mesenchymal Stromal Cells and Their Progeny During Aging and Leukemia. Front Cell Dev Biol 2021; 9:714716. [PMID: 34447754 PMCID: PMC8383146 DOI: 10.3389/fcell.2021.714716] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are a heterogenous cell population found in a wide range of tissues in the body, known for their nutrient-producing and immunomodulatory functions. In the bone marrow (BM), these MSCs are critical for the regulation of hematopoietic stem cells (HSC) that are responsible for daily blood production and functional immunity throughout an entire organism's lifespan. Alongside other stromal cells, MSCs form a specialized microenvironment BM tissue called "niche" that tightly controls HSC self-renewal and differentiation. In addition, MSCs are crucial players in maintaining bone integrity and supply of hormonal nutrients due to their capacity to differentiate into osteoblasts and adipocytes which also contribute to cellular composition of the BM niche. However, MSCs are known to encompass a large heterogenous cell population that remains elusive and poorly defined. In this review, we focus on deciphering the BM-MSC biology through recent advances in single-cell identification of hierarchical subsets with distinct functionalities and transcriptional profiles. We also discuss the contribution of MSCs and their osteo-adipo progeny in modulating the complex direct cell-to-cell or indirect soluble factors-mediated interactions of the BM HSC niche during homeostasis, aging and myeloid malignancies. Lastly, we examine the therapeutic potential of MSCs for rejuvenation and anti-tumor remedy in clinical settings.
Collapse
Affiliation(s)
- Kevin Woods
- German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- Department of Hematology and Oncology, University Medical Center Mainz, Mainz, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Borhane Guezguez
- German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- Department of Hematology and Oncology, University Medical Center Mainz, Mainz, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
4
|
Barcia Durán JG, Lu T, Houghton S, Geng F, Schreiner R, Xiang J, Rafii S, Redmond D, Lis R. Endothelial Jak3 expression enhances pro-hematopoietic angiocrine function in mice. Commun Biol 2021; 4:406. [PMID: 33767339 PMCID: PMC7994450 DOI: 10.1038/s42003-021-01846-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 02/12/2021] [Indexed: 02/08/2023] Open
Abstract
Jak3 is the only non-promiscuous member of the Jak family of secondary messengers. Studies to date have focused on understanding and targeting the cell-autonomous role of Jak3 in immunity, while functional Jak3 expression outside the hematopoietic system remains largely unreported. We show that Jak3 is expressed in endothelial cells across hematopoietic and non-hematopoietic organs, with heightened expression in the bone marrow. The bone marrow niche is understood as a network of different cell types that regulate hematopoietic function. We show that the Jak3-/- bone marrow niche is deleterious for the maintenance of long-term repopulating hematopoietic stem cells (LT-HSCs) and that JAK3-overexpressing endothelial cells have increased potential to expand LT-HSCs in vitro. This work may serve to identify a novel function for a highly specific tyrosine kinase in the bone marrow vascular niche and to further characterize the LT-HSC function of sinusoidal endothelium.
Collapse
Affiliation(s)
- José Gabriel Barcia Durán
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Tyler Lu
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Sean Houghton
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Fuqiang Geng
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ryan Schreiner
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Department of Ophthalmology, Margaret Dyson Vision Research Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Jenny Xiang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Shahin Rafii
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY, 10065, USA
| | - David Redmond
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.
| | - Raphaël Lis
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
5
|
Yang CY, Lu RJH, Lee MK, Hsiao FSH, Yen YP, Cheng CC, Hsu PS, Tsai YT, Chen SK, Liu IH, Chen PY, Lin SP. Transcriptome Analysis of Dnmt3l Knock-Out Mice Derived Multipotent Mesenchymal Stem/Stromal Cells During Osteogenic Differentiation. Front Cell Dev Biol 2021; 9:615098. [PMID: 33718357 PMCID: PMC7947861 DOI: 10.3389/fcell.2021.615098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 02/01/2021] [Indexed: 01/01/2023] Open
Abstract
Multipotent mesenchymal stem/stromal cells (MSCs) exhibit great potential for cell-based therapy. Proper epigenomic signatures in MSCs are important for the maintenance and the subsequent differentiation potential. The DNA methyltransferase 3-like (DNMT3L) that was mainly expressed in the embryonic stem (ES) cells and the developing germ cells plays an important role in shaping the epigenetic landscape. Here, we report the reduced colony forming ability and impaired in vitro osteogenesis in Dnmt3l-knockout-mice-derived MSCs (Dnmt3l KO MSCs). By comparing the transcriptome between undifferentiated Dnmt3l KO MSCs and the MSCs from the wild-type littermates, some of the differentially regulated genes (DEGs) were found to be associated with bone-morphology-related phenotypes. On the third day of osteogenic induction, differentiating Dnmt3l KO MSCs were enriched for genes associated with nucleosome structure, peptide binding and extracellular matrix modulation. Differentially expressed transposable elements in many subfamilies reflected the change of corresponding regional epigenomic signatures. Interestingly, DNMT3L protein is not expressed in cultured MSCs. Therefore, the observed defects in Dnmt3l KO MSCs are unlikely a direct effect from missing DNMT3L in this cell type; instead, we hypothesized them as an outcome of the pre-deposited epigenetic signatures from the DNMT3L-expressing progenitors. We observed that 24 out of the 107 upregulated DEGs in Dnmt3l KO MSCs were hypermethylated in their gene bodies of DNMT3L knock-down ES cells. Among these 24 genes, some were associated with skeletal development or homeostasis. However, we did not observe reduced bone development, or reduced bone density through aging in vivo. The stronger phenotype in vitro suggested the involvement of potential spreading and amplification of the pre-deposited epigenetic defects over passages, and the contribution of oxidative stress during in vitro culture. We demonstrated that transient deficiency of epigenetic co-factor in ES cells or progenitor cells caused compromised property in differentiating cells much later. In order to facilitate safer practice in cell-based therapy, we suggest more in-depth examination shall be implemented for cells before transplantation, even on the epigenetic level, to avoid long-term risk afterward.
Collapse
Affiliation(s)
- Chih-Yi Yang
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Rita Jui-Hsien Lu
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, Taiwan.,Department of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Ming-Kang Lee
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Felix Shih-Hsian Hsiao
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan.,Department of Animal Science and Biotechnology, Tunghai University, Taichung, Taiwan
| | - Ya-Ping Yen
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan.,Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chun-Chun Cheng
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Pu-Sheng Hsu
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Yi-Tzang Tsai
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Shih-Kuo Chen
- Department of Life Sciences, National Taiwan University, Taipei, Taiwan
| | - I-Hsuan Liu
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Pao-Yang Chen
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, Taiwan
| | - Shau-Ping Lin
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan.,Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan.,Center for Systems Biology, National Taiwan University, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
6
|
Su X, Jing H, Yu W, Lei F, Wang R, Hu C, Li M, Lin T, Zhou H, Wang F, Liao L. A bone matrix-simulating scaffold to alleviate replicative senescence of mesenchymal stem cells during long-term expansion. J Biomed Mater Res A 2020; 108:1955-1967. [PMID: 32323459 DOI: 10.1002/jbm.a.36958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/15/2020] [Accepted: 04/19/2020] [Indexed: 02/05/2023]
Abstract
Replicative senescence during in vitro augmentation, which is mostly induced by the loss of physiological microenvironment, hinders the application of mesenchymal stem cells (MSCs) in the clinic. Here, we investigated whether MSCs senescence could be prevented by bio-scaffold mimicking the natural tissue matrix. Human umbilical cord mesenchymal stem cells (hUCMSCs) exhibited a senescent phenotype during a long-term passage in the conventional culture dish. To fabricate the bone matrix, a naturally based matrix composed of nano-hydroxyapatite/chitosan/poly lactide-co-glycolide (nHA/CS/PLGA) was produced. Long-term passage resulted in an obvious increase in the expression of senescence markers and a reduction in the expression of master genes involved in tissue regeneration. Functional assay confirmed that nHA/CS/PLGA scaffold preserved the proliferation and differentiation of hUCMSCs even after being passaged 27 times. Moreover, in vivo ectopic bone formation assay revealed that the bone formation of hUCMSCs cultured on the nano-scaffolds for the long term was as robust as the cells in the early passage. In summary, our results demonstrate that nHA/CS/PLGA scaffold effectively preserves the stemness and youth of hUCMSCs in the long-term passage. Taken advantage of its compatibility and bioactivity, nHA/CS/PLGA scaffold is of great potential in large-scale expansion of MSCs for stem cell therapy and tissue engineering.
Collapse
Affiliation(s)
- Xiaoxia Su
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Huan Jing
- Department of Stomatology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Wenting Yu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Fengzhen Lei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Rui Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Cheng Hu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Mujia Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Tingting Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Hong Zhou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Fei Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Li Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Abstract
Aplastic anemia (AA) is a rare and life-threatening bone marrow failure (BMF) that results in peripheral blood cytopenia and reduced bone marrow hematopoietic cell proliferation. The symptoms are similar to myelofibrosis, myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) making diagnosis of AA complicated. The pathogenesis of AA is complex and its mechanism needs to be deciphered on an individualized basis. This review summarizes several contributions made in trying to understand AA pathogenesis in recent years which may be helpful for the development of personalized therapies for AA.
Collapse
Affiliation(s)
- Li Wang
- a Department of Hematology , Affiliated Hospital of Nantong University , Nantong , People's Republic of China
| | - Hong Liu
- a Department of Hematology , Affiliated Hospital of Nantong University , Nantong , People's Republic of China
| |
Collapse
|
8
|
Bello AB, Park H, Lee SH. Current approaches in biomaterial-based hematopoietic stem cell niches. Acta Biomater 2018; 72:1-15. [PMID: 29578087 DOI: 10.1016/j.actbio.2018.03.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 02/07/2018] [Accepted: 03/14/2018] [Indexed: 12/20/2022]
Abstract
Hematopoietic stem cells (HSCs) are multipotent progenitor cells that can differentiate and replenish blood and immune cells. While there is a growing demand for autologous and allogeneic HSC transplantation owing to the increasing incidence of hereditary and hematologic diseases, the low population of HSCs in cord-blood and bone marrow (the main source of HSCs) hinders their medical applicability. Several cytokine and growth factor-based methods have been developed to expand the HSCs in vitro; however, the expansion rate is low, or the expanded cells fail to survive upon engraftment. This is at least in part because the overly simplistic polystyrene culture substrates fail to fully replicate the microenvironments or niches where these stem cells live. Bone marrow niches are multi-dimensional, complex systems that involve both biochemical (cells, growth factors, and cytokines) and physiochemical (stiffness, O2 concentration, and extracellular matrix presentation) factors that regulate the quiescence, proliferation, activation, and differentiation of the HSCs. Although several studies have been conducted on in vitro HSC expansion via 2D and 3D biomaterial-based platforms, additional work is required to engineer an effective biomaterial platform that mimics bone marrow niches. In this study, the factors that regulate the HSC in vivo were explained and their applications in the engineering of a bone marrow biomaterial-based platform were discussed. In addition, current approaches, challenges, and the future direction of a biomaterial-based culture and expansion of the HSC were examined. STATEMENT OF SIGNIFICANCE Hematopoietic stem cells (HSC) are multipotent cells that can differentiate and replace the blood and immune cells of the body. However, in vivo, there is a low population of these cells, and thus their use in biotherapeutic and medical applications is limited (i.e., bone marrow transplantation). In this review, the biochemical factors (growth factors, cytokines, co-existing cells, ECM, gas concentrations, and differential gene expression) that may regulate the over-all fate of HSC, in vivo, were summarized and discussed. Moreover, different conventional and recent biomaterial platforms were reviewed, and their potential in generating a biomaterial-based, BM niche-mimicking platform for the efficient growth and expansion of clinically relevant HSCs in-vitro, was discussed.
Collapse
Affiliation(s)
- Alvin Bacero Bello
- School of Integrative Engineering, Chung-Ang University, Seoul 06911, Republic of Korea; Department of Biomedical Science, CHA University, Seongnam-Si 13488, Republic of Korea
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul 06911, Republic of Korea.
| | - Soo-Hong Lee
- Department of Biomedical Science, CHA University, Seongnam-Si 13488, Republic of Korea.
| |
Collapse
|
9
|
Selman C, Sinclair A, Pedroni SMA, Irvine EE, Michie AM, Withers DJ. Evidence that hematopoietic stem cell function is preserved during aging in long-lived S6K1 mutant mice. Oncotarget 2017; 7:29937-43. [PMID: 27083004 PMCID: PMC5058654 DOI: 10.18632/oncotarget.8729] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 04/05/2016] [Indexed: 11/25/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) signalling pathway plays a highly conserved role in aging; mice lacking ribosomal protein S6 kinase 1 (S6K1−/−) have extended lifespan and healthspan relative to wild type (WT) controls. Exactly how reduced mTOR signalling induces such effects is unclear, although preservation of stem cell function may be important. We show, using gene expression analyses, that there was a reduction in expression of cell cycle genes in young (12 week) and aged (80 week) S6K1−/− BM-derived c-Kit+ cells when compared to age-matched WT mice, suggesting that these cells are more quiescent in S6K1−/− mice. In addition, we investigated hematopoietic stem cell (HSC) frequency and function in young and aged S6K1−/− and WT mice. Young, but not aged, S6K1−/− mice had more LSK (lineage−, c-Kit+, Sca-1+) cells (% of bone marrow (BM)), including the most primitive long-term repopulating HSCs (LT-HSC) relative to WT controls. Donor-derived engraftment of LT-HSCs in recipient mice was unaffected by genotype in young mice, but was enhanced in transplants using LT-HSCs derived from aged S6K1−/− mice. Our results are the first to provide evidence that age-associated HSC functional decline is ameliorated in a long-lived mTOR mutant mouse.
Collapse
Affiliation(s)
- Colin Selman
- Glasgow Ageing Research Network (GARNER), Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Amy Sinclair
- Glasgow Ageing Research Network (GARNER), Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Silvia M A Pedroni
- Metabolic Signaling Group, Medical Research Council Clinical Sciences Centre, Imperial College, London, UK
| | - Elaine E Irvine
- Metabolic Signaling Group, Medical Research Council Clinical Sciences Centre, Imperial College, London, UK
| | - Alison M Michie
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Dominic J Withers
- Metabolic Signaling Group, Medical Research Council Clinical Sciences Centre, Imperial College, London, UK
| |
Collapse
|
10
|
Kurkjian CJ, Guo H, Montgomery ND, Cheng N, Yuan H, Merrill JR, Sempowski GD, Brickey WJ, Ting JPY. The Toll-Like Receptor 2/6 Agonist, FSL-1 Lipopeptide, Therapeutically Mitigates Acute Radiation Syndrome. Sci Rep 2017; 7:17355. [PMID: 29230065 PMCID: PMC5725477 DOI: 10.1038/s41598-017-17729-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/16/2017] [Indexed: 02/03/2023] Open
Abstract
Risks of radiation exposure from nuclear incidents and cancer radiotherapy are undeniable realities. These dangers urgently compel the development of agents for ameliorating radiation–induced injuries. Biologic pathways mediated by myeloid differentiation primary response gene 88 (MyD88), the common adaptor for toll–like receptor (TLR) and Interleukin–1 receptor signaling, are critical for radioprotection. Treating with agonists prior to radiation enhances survival by activating TLR signaling, whereas radiomitigating TLR–activating therapeutics given after exposure are less defined. We examine the radiomitigation capability of TLR agonists and identify one that is superior for its efficacy and reduced toxic consequences compared to other tested agonists. We demonstrate that the synthetic TLR2/6 ligand Fibroblast–stimulating lipopeptide (FSL–1) substantially prolongs survival in both male and female mice when administered 24 hours after radiation and shows MyD88–dependent function. FSL–1 treatment results in accelerated hematopoiesis in bone marrow, spleen and periphery, and augments systemic levels of hematopoiesis–stimulating factors. The ability of FSL–1 to stimulate hematopoiesis is critical, as hematopoietic dysfunction results from a range of ionizing radiation doses. The efficacy of a single FSL–1 dose for alleviating radiation injury while protecting against adverse effects reveals a viable radiation countermeasures agent.
Collapse
Affiliation(s)
- Cathryn J Kurkjian
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Hao Guo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Nathan D Montgomery
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Ning Cheng
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.,Oral Biology Curriculum, School of Dentistry, University of North Carolina, Chapel Hill, NC, USA
| | - Hong Yuan
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Biomedical Imaging Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joseph R Merrill
- Biomedical Imaging Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - W June Brickey
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Jenny P-Y Ting
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA. .,Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
11
|
Zhao Y, Zhang H. Update on the mechanisms of homing of adipose tissue-derived stem cells. Cytotherapy 2017; 18:816-27. [PMID: 27260205 DOI: 10.1016/j.jcyt.2016.04.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 03/11/2016] [Accepted: 04/25/2016] [Indexed: 02/06/2023]
Abstract
Adipose tissue-derived stem cells (ADSCs), which resemble bone marrow mesenchymal stromal cells (BMSCs), have shown great advantages and promise in the field of regenerative medicine. They can be readily harvested in large numbers with low donor-site morbidity. To date, a great number of preclinical and clinical studies have shown ADSCs' safety and efficacy in regenerative medicine. However, a better understanding of the mechanisms of homing of ADSCs is needed to advance the clinical utility of this therapy. In this review, the reports of the homing of ADSCs were searched using Pubmed and Google Scholar to update our knowledge. ADSCs were proved to interact with endothelial cells by expressing the similar integrins with BMSCs. In addition, ADSCs do not possess the dominant ligand for P-selectin, just like BMSCs. Stromal derived factor-1 (SDF-1)/CXCR4 and CXC ligand-5 (CXCL5)/CXCR2 interactions are the two main axes governing ADSCs extravasation from bone marrow vessels. Some more signaling pathways involved in migration of ADSCs have been investigated, including LPA/LPA1 signaling pathway, MAPK/Erk1/2 signaling pathway, RhoA/Rock signaling pathway and PDGF-BB/PDGFR-β signaling pathway. Status quo of a lack of intensive studies on the details of homing of ADSCs should be improved in the near future before clinical application.
Collapse
Affiliation(s)
- Yong Zhao
- Minimally Invasive Urology Center, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Haiyang Zhang
- Minimally Invasive Urology Center, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China; Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA.
| |
Collapse
|
12
|
Junqueira Reis LC, Picanço-Castro V, Paes BCMF, Pereira OA, Gerdes Gyuricza I, de Araújo FT, Morato-Marques M, Moreira LF, Costa EDBO, dos Santos TPM, Covas DT, Pereira Carramaschi LDV, Russo EMDS. Induced Pluripotent Stem Cell for the Study and Treatment of Sickle Cell Anemia. Stem Cells Int 2017; 2017:7492914. [PMID: 28814957 PMCID: PMC5549510 DOI: 10.1155/2017/7492914] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/05/2017] [Indexed: 12/14/2022] Open
Abstract
Sickle cell anemia (SCA) is a monogenic disease of high mortality, affecting millions of people worldwide. There is no broad, effective, and safe definitive treatment for SCA, so the palliative treatments are the most used. The establishment of an in vitro model allows better understanding of how the disease occurs, besides allowing the development of more effective tests and treatments. In this context, iPSC technology is a powerful tool for basic research and disease modeling, and a promise for finding and screening more effective and safe drugs, besides the possibility of use in regenerative medicine. This work obtained a model for study and treatment of SCA using iPSC. Then, episomal vectors were used for reprogramming peripheral blood mononuclear cells to obtain integration-free iPSC. Cells were collected from patients treated with hydroxyurea and without treatment. The iPSCP Bscd lines were characterized for pluripotent and differentiation potential. The iPSC lines were differentiated into HSC, so that we obtained a dynamic and efficient protocol of CD34+CD45+ cells production. We offer a valuable tool for a better understanding of how SCA occurs, in addition to making possible the development of more effective drugs and treatments and providing better understanding of widely used treatments, such as hydroxyurea.
Collapse
Affiliation(s)
- Luiza Cunha Junqueira Reis
- Pharmaceutical Sciences School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
- Blood Center Foundation of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Virgínia Picanço-Castro
- Blood Center Foundation of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Bárbara Cristina Martins Fernandes Paes
- Blood Center Foundation of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
- Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Olívia Ambrozini Pereira
- Philosophy, Sciences and Languages School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | | | | | | | | | - Dimas Tadeu Covas
- Blood Center Foundation of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
- Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | - Elisa Maria de Sousa Russo
- Pharmaceutical Sciences School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
- Blood Center Foundation of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
13
|
Fibroblast growth factor 2 supports osteoblastic niche cells during hematopoietic homeostasis recovery after bone marrow suppression. Cell Commun Signal 2017; 15:25. [PMID: 28662672 PMCID: PMC5492158 DOI: 10.1186/s12964-017-0181-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 06/16/2017] [Indexed: 11/30/2022] Open
Abstract
Background Hematopoietic stem cell (HSC) maintenance requires a specific microenvironment. HSC niches can be activated by tissue damaging chemotherapeutic drugs and various cell signaling molecules such as SDF-1 and FGF, which might also result in bone marrow stress. Recent research has insufficiently shown that endosteal osteolineage cells and other niche constituents recover after marrow injury. Methods We investigated the role of FGF2 in the osteoblastic niche cells during hematopoietic homeostasis recovery after bone marrow injury. Mice were treated with 5-fluorouracil (5FU) to eliminate actively cycling cells in the bone marrow. Primary osteoblasts were isolated and subjected to cell culture. Real-time PCR, western blot and immunohistochemical staining were performed to study niche-related genes, osteoblast markers, and FGF2 signaling. Proliferation rate were analyzed by marker gene Ki67 and colony formation assay. Also, osterix-positive osteoprogenitor cells were isolated by FACS from Osx-GFP-Cre mice after 5FU treatment, and subjected to RNA-sequencing and analyzed for Fgf receptors and niche markers. Results The endosteal osteolineage cells isolated from 5FU-treated mice showed increased expression of the niche-related genes Sdf-1, Jagged-1, Scf, N-cad, Angpt1 and Vcam-1 and the osteoblast marker genes Osx, Opn, Runx2, and Alp, indicating that BM stress upon 5FU treatment activated the osteoblastic niche. Endosteal osteoblast expanded from a single layer to several layers 3 and 6 days after 5FU treatment. During the early recovery phase in 5FU-activated osteoblastic niches increased FGF2 expression and activated its downstream pERK. FGF2 treatment resulted in increased proliferation rate and the expression of niche marker genes in 5FU-activated osteoblastic niche cells. RNA-seq analysis in Osterix-positive osteoprogenitor cells isolated from 5FU-treated Osx-GFP mice showed significantly increased expression of Fgf receptors Fgfr1, 2 and 3. Although osteoblastic niche cells were damaged by 5FU treatment in the beginning, the increased number of OB layers in the recovery phase may be derived from resident osteoprogenitor cells by FGF2 activation under stress. Conclusions Taken together, FGF2 signaling can regulate osteoblastic niche cells to support HSC homeostasis in response to bone marrow damage.
Collapse
|
14
|
Xu Y, Chu N, Qiu X, Gober HJ, Li D, Wang L. The interconnected role of chemokines and estrogen in bone metabolism. Biosci Trends 2016; 10:433-444. [DOI: 10.5582/bst.2016.01072] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Yingping Xu
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Shanghai Medical College, Fudan University
- The Academy of Integrative Medicine of Fudan University
| | - Nan Chu
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Shanghai Medical College, Fudan University
| | - Xuemin Qiu
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Shanghai Medical College, Fudan University
- The Academy of Integrative Medicine of Fudan University
| | | | - Dajin Li
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Shanghai Medical College, Fudan University
- The Academy of Integrative Medicine of Fudan University
| | - Ling Wang
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Shanghai Medical College, Fudan University
- The Academy of Integrative Medicine of Fudan University
| |
Collapse
|
15
|
Shekaran A, Sim E, Tan KY, Chan JKY, Choolani M, Reuveny S, Oh S. Enhanced in vitro osteogenic differentiation of human fetal MSCs attached to 3D microcarriers versus harvested from 2D monolayers. BMC Biotechnol 2015; 15:102. [PMID: 26520400 PMCID: PMC4628389 DOI: 10.1186/s12896-015-0219-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 10/20/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are of great interest in bone regenerative medicine due to their osteogenic potential and trophic effects. However, challenges to large-scale production of MSCs can hinder the translation of MSC therapies. 3D Microcarrier (MC)-based MSC culture presents a scalable and cost-effective alternative to conventional methods of expansion in 2D monolayers. Furthermore, biodegradable MCs may allow for MC-bound MSC delivery without enzymatic harvest for selected applications such as bone healing. However, the effects of cell expansion on microcarriers and enzymatic cell harvest on MSC phenotype and osteogenic differential potential are not well understood. In this study, we characterized human fetal MSCs (hfMSCs) after expansion in 3D microcarrier spinner or 2D monolayer cultures. Following expansion, we compared osteogenic differentiation of cultures seeded with 3D MC-harvested, 3D MC-bound and conventional 2D monolayer (MNL)-harvested cells when cultured in osteogenic induction media on collagen-coated plates. RESULTS Fetal MSCs expanded on both 3D agitated Microcarriers (MC) and 2D Plastic static monolayer (MNL) cultures express high levels of MSC surface markers. MC-harvested hfMSCs displayed higher expression of early osteogenic genes but slower mineralization kinetics compared to MNL-harvested MSCs during osteogenic induction. However, in the comparison between MC-bound and MC-harvested hfMSCs, osteogenic genes were upregulated and mineralization kinetics was accelerated in the former condition. Importantly, 3D MC-bound hfMSCs expressed higher levels of osteogenic genes and displayed either higher or equivalent levels of mineralization, depending on the cell line, compared to the classical monolayer cultures use in the literature (MNL-harvested hfMSCs). CONCLUSION Beyond the processing and scalability advantages of the microcarrier culture, hfMSCs attached to MCs undergo robust osteogenic differentiation and mineralization compared to enzymatically harvested cells. Thus biodegradable/biocompatible MCs which can potentially be used for cell expansion as well as a scaffold for direct in vivo delivery of cells may have advantages over the current methods of monolayer-expansion and delivery post-harvest for bone regeneration applications.
Collapse
Affiliation(s)
- Asha Shekaran
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Eileen Sim
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Kah Yong Tan
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Jerry Kok Yen Chan
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.,Cancer & Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore, 169857, Singapore.,Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Mahesh Choolani
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Shaul Reuveny
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Steve Oh
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore.
| |
Collapse
|
16
|
Wu L, Mo W, Zhang Y, Deng H, Li Y, Zhou R, Zhang L, Pan S, Wang S. Impairment of hematopoietic stem cell niches in patients with aplastic anemia. Int J Hematol 2015; 102:645-53. [PMID: 26440975 DOI: 10.1007/s12185-015-1881-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/29/2015] [Accepted: 09/30/2015] [Indexed: 10/23/2022]
Abstract
Aplastic anemia (AA) is a serious hematological disorder characterized by pancytopenia and defective bone marrow (BM) microenvironment. Previous investigations have demonstrated that there is a defect of angiogenesis in the BM of patients with AA. However, whether abnormalities of the BM microenvironment, particularly the overall specialization of niches, which have been classified into osteoblastic, vascular, and perivascular niches, are involved in the pathogenesis of AA is unknown. In the present study, 46 patients with AA and 15 controls were selected. The cellular elements of the BM microenvironment, including endosteal, vascular, and perivascular cells, were analyzed by immunohistochemical staining in situ. Patients with AA showed markedly fewer endosteal cells [0.33 vs 3.67 per high-power field (hpf); P < 0.05], vascular cells (8.00 vs 12.67 per hpf; P < 0.05), and perivascular cells (7.17 vs 10.67 per hpf; P < 0.05) compared with controls. These data indicate that AA is associated with impaired hematopoietic stem cell niches.
Collapse
Affiliation(s)
- Liangliang Wu
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou Medical University, 1 Panfu Road, Guangzhou, 510180, Guangdong, People's Republic of China
| | - Wenjian Mo
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou Medical University, 1 Panfu Road, Guangzhou, 510180, Guangdong, People's Republic of China
| | - Yuping Zhang
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou Medical University, 1 Panfu Road, Guangzhou, 510180, Guangdong, People's Republic of China
| | - Hui Deng
- Department of Blood Transfusion, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yumiao Li
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou Medical University, 1 Panfu Road, Guangzhou, 510180, Guangdong, People's Republic of China
| | - Ruiqing Zhou
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou Medical University, 1 Panfu Road, Guangzhou, 510180, Guangdong, People's Republic of China
| | - Lu Zhang
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou Medical University, 1 Panfu Road, Guangzhou, 510180, Guangdong, People's Republic of China
| | - Shiyi Pan
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou Medical University, 1 Panfu Road, Guangzhou, 510180, Guangdong, People's Republic of China
| | - Shunqing Wang
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou Medical University, 1 Panfu Road, Guangzhou, 510180, Guangdong, People's Republic of China.
| |
Collapse
|
17
|
Bari S, Chu PPY, Lim A, Fan X, Bunte RM, Li S, Ghosh S, Chiu GNC, Hwang WYK. Mitochondrial superoxide reduction and cytokine secretion skewing by carbon nanotube scaffolds enhance ex vivo expansion of human cord blood hematopoietic progenitors. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1643-56. [DOI: 10.1016/j.nano.2015.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 05/27/2015] [Accepted: 06/06/2015] [Indexed: 01/25/2023]
|
18
|
Chiarini F, Lonetti A, Evangelisti C, Buontempo F, Orsini E, Evangelisti C, Cappellini A, Neri LM, McCubrey JA, Martelli AM. Advances in understanding the acute lymphoblastic leukemia bone marrow microenvironment: From biology to therapeutic targeting. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:449-463. [PMID: 26334291 DOI: 10.1016/j.bbamcr.2015.08.015] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 08/26/2015] [Accepted: 08/27/2015] [Indexed: 02/07/2023]
Abstract
The bone marrow (BM) microenvironment regulates the properties of healthy hematopoietic stem cells (HSCs) localized in specific niches. Two distinct microenvironmental niches have been identified in the BM, the "osteoblastic (endosteal)" and "vascular" niches. Nevertheless, these niches provide sanctuaries where subsets of leukemic cells escape chemotherapy-induced death and acquire a drug-resistant phenotype. Moreover, it is emerging that leukemia cells are able to remodel the BM niches into malignant niches which better support neoplastic cell survival and proliferation. This review focuses on the cellular and molecular biology of microenvironment/leukemia interactions in acute lymphoblastic leukemia (ALL) of both B- and T-cell lineage. We shall also highlight the emerging role of exosomes/microvesicles as efficient messengers for cell-to-cell communication in leukemia settings. Studies on the interactions between the BM microenvironment and ALL cells have led to the discovery of potential therapeutic targets which include cytokines/chemokines and their receptors, adhesion molecules, signal transduction pathways, and hypoxia-related proteins. The complex interplays between leukemic cells and BM microenvironment components provide a rationale for innovative, molecularly targeted therapies, designed to improve ALL patient outcome. A better understanding of the contribution of the BM microenvironment to the process of leukemogenesis and leukemia persistence after initial remission, may provide new targets that will allow destruction of leukemia cells without adversely affecting healthy HSCs. This article is part of a Special Issue entitled: Tumor Microenvironment Regulation of Cancer Cell Survival, Metastasis,Inflammation, and Immune Surveillance edited by Peter Ruvolo and Gregg L. Semenza.
Collapse
Affiliation(s)
- Francesca Chiarini
- Institute of Molecular Genetics, National Research Council, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Annalisa Lonetti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Camilla Evangelisti
- Institute of Molecular Genetics, National Research Council, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Francesca Buontempo
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Ester Orsini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Cecilia Evangelisti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alessandra Cappellini
- Department of Human Social and Health Sciences, University of Cassino, Cassino, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
19
|
ZHAO QIANG, WANG XUEYAO, YU XIAOXIA, ZHAI YINGXIAN, HE XU, WU SHAN, SHI YINGAI. Expression of human telomerase reverse transcriptase mediates the senescence of mesenchymal stem cells through the PI3K/AKT signaling pathway. Int J Mol Med 2015; 36:857-64. [DOI: 10.3892/ijmm.2015.2284] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 07/08/2015] [Indexed: 11/06/2022] Open
|
20
|
Kashyap M, Pore S, Wang Z, Gingrich J, Yoshimura N, Tyagi P. Inflammasomes are important mediators of prostatic inflammation associated with BPH. JOURNAL OF INFLAMMATION-LONDON 2015; 12:37. [PMID: 25991911 PMCID: PMC4436794 DOI: 10.1186/s12950-015-0082-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/08/2015] [Indexed: 11/10/2022]
Abstract
Background There is mounting evidence to support the role of inflammation in benign prostate hyperplasia (BPH), and a recent study reported expression of inflammasome derived cytokine IL-18 in prostate biopsy of BPH patients. Here we examined the expression of inflammasome-derived cytokines and activation of nucleotide-binding oligomerization domain-like receptor with pyrin domain protein 1 (NLRP) 1 inflammasome in a rat model of prostatic inflammation relevant to BPH. Methods Prostatic inflammation was experimentally induced in three-month-old male Sprague–Dawley rats by intraprostatic injection (50 μL) of either 5 % formalin or saline (sham) into the ventral lobes of prostate. 7 days later, prostate and bladder tissue was harvested for analysis of inflammasome by Western blot, immunohistochemistry and downstream cytokine production by Milliplex. Results Expression of interleukins, CXC and CC chemokines were elevated 2-15 fold in formalin injected prostate relative to sham. Significant expression of NLRP1 inflammasome components and caspase-1 in prostate were associated with significant elevation of pro and cleaved forms of IL-1β (25.50 ± 1.16 vs 3.05 ± 0.65 pg/mg of protein) and IL-18 (1646.15 ± 182.61 vs 304.67 ± 103.95 pg/mg of protein). Relative to prostate tissue, the cytokine expression in bladder tissue was much lower and did not involve inflammasome activation. Conclusions Significant upregulation of NLRP1, caspase-1 and downstream cytokines (IL-18 and IL-1β) suggests that a NLRP1 inflammasome is assembled and activated in prostate tissue of this rat model. Recapitulation of findings from human BPH specimens suggests that the inflammasome may perpetuate the inflammatory state associated with BPH. Further clarification of these pathways may offer innovative therapeutic targets for BPH-related inflammation.
Collapse
Affiliation(s)
- Mahendra Kashyap
- Department of Urology, University of Pittsburgh, Pittsburgh, USA
| | - Subrata Pore
- Department of Urology, University of Pittsburgh, Pittsburgh, USA
| | - Zhou Wang
- Department of Urology, University of Pittsburgh, Pittsburgh, USA
| | - Jeffrey Gingrich
- Department of Urology, University of Pittsburgh, Pittsburgh, USA
| | - Naoki Yoshimura
- Department of Urology, University of Pittsburgh, Pittsburgh, USA
| | - Pradeep Tyagi
- Department of Urology, University of Pittsburgh, Pittsburgh, USA
| |
Collapse
|
21
|
Ahmadzadeh A, Kast RE, Ketabchi N, Shahrabi S, Shahjahani M, Jaseb K, Saki N. Regulatory effect of chemokines in bone marrow niche. Cell Tissue Res 2015; 361:401-10. [DOI: 10.1007/s00441-015-2129-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 01/16/2015] [Indexed: 12/31/2022]
|
22
|
Xia J, Xu X, Huang P, He M, Wang X. The potential of CXCL5 as a target for liver cancer - what do we know so far? Expert Opin Ther Targets 2014; 19:141-6. [PMID: 25495348 DOI: 10.1517/14728222.2014.993317] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
CXCL5, epithelial cell derived neutrophil attractant 78, is a CXC chemokine predominantly expressed on epithelial cells. It has specificity for CXCR2 receptors and is involved in the recruitment and activation of neutrophils. CXCL5 is considered a therapeutic target in liver cancer, since treatment with small-interfering RNAs or antibodies against CXCL5 can suppress tumor growth, proliferation, migration and invasion. Experimental evidence demonstrated that CXCL5 antibodies could reduce the tumor growth and synergistically increase the efficiency of the tyrosine kinase inhibitor, Gefitinib, without the addition of toxicity. A number of challenges are encountered and should be considered during the development and clinical application of CXCL5 target-specific drugs. The specificity of CXCL5 as a therapeutic target for certain types and duration of cancer should be more carefully clarified, since it seems that CXCL5 is involved in many molecular pathways and crosstalk between targeted chemokines/receptors. The concept that CXCL5 serves as the therapeutic target for liver cancer was evidenced by preclinical studies, and is the beginning of CXCL5-based drug discovery and development.
Collapse
Affiliation(s)
- Jinglin Xia
- Fudan University, Minhang Hospital , Minhang , China +86 21 64041990 Ext: 5420 ; +86 21 54961729 ;
| | | | | | | | | |
Collapse
|
23
|
Na SS, Aldonza MB, Sung HJ, Kim YI, Son YS, Cho S, Cho JY. Stanniocalcin-2 (STC2): A potential lung cancer biomarker promotes lung cancer metastasis and progression. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1854:668-76. [PMID: 25463045 DOI: 10.1016/j.bbapap.2014.11.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 10/29/2014] [Accepted: 11/05/2014] [Indexed: 12/14/2022]
Abstract
The homodimeric glycoprotein, stanniocalcin 2 (STC2) is previously known to be involved in the regulation of calcium and phosphate transport in the kidney and also reported to play multiple roles in several cancers. However, its function and clinical significance in lung cancer have never been reported and still remain uncertain. Here, we investigated the possibility of STC2 as a lung cancer biomarker and identified its potential role in lung cancer cell growth, metastasis and progression. Proteomic analysis of secretome of primary cultured lung cancer cells revealed higher expression of STC2 in cancers compared to that of adjacent normal cells. RT-PCR and Western blot analyses showed higher mRNA and protein expressions of STC2 in lung cancer tissues compared to the adjacent normal tissues. Knockdown of STC2 in H460 lung cancer cells slowed down cell growth progression and colony formation. Further analysis revealed suppression of migration, invasion and delayed G0/G1 cell cycle progression in the STC2 knockdown cells. STC2 knockdown also attenuated the H202-induced oxidative stress on H460 cell viability with a subsequent increase in intracellular ROS levels, which suggest a protective role of STC2 in redox regulatory system of lung cancer. These findings suggest that STC2 can be a potential lung cancer biomarker and plays a positive role in lung cancer metastasis and progression. This article is part of a Special Issue entitled: Medical Proteomics.
Collapse
Affiliation(s)
- Sang-su Na
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Republic of Korea; Department of Physical Therapy, College of Rehabilitation, Daegu University, Daegu, Republic of Korea
| | - Mark Borris Aldonza
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Hye-Jin Sung
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Yong-In Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Yeon Sung Son
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Republic of Korea
| | - Sukki Cho
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seoungnam-si, Gyeonggi-do, Republic of Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Republic of Korea.
| |
Collapse
|
24
|
Iyer S, Viernes DR, Chisholm JD, Margulies BS, Kerr WG. SHIP1 regulates MSC numbers and their osteolineage commitment by limiting induction of the PI3K/Akt/β-catenin/Id2 axis. Stem Cells Dev 2014; 23:2336-51. [PMID: 24857423 DOI: 10.1089/scd.2014.0122] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Here, we show that Src homology 2-domain-containing inositol 5'-phosphatase 1 (SHIP1) is required for the efficient development of osteoblasts from mesenchymal stem cells (MSCs) such that bone growth and density are reduced in mice that lack SHIP1 expression in MSCs. We find that SHIP1 promotes the osteogenic output of MSCs by limiting activation of the PI3K/Akt/β-catenin pathway required for induction of the MSC stemness factor Id2. In parallel, we demonstrate that mice with myeloid-restricted ablation of SHIP1, including osteoclasts (OCs), show no reduction in bone mass or density. Hence, diminished bone mass and density in the SHIP1-deficient mice results from SHIP deficiency in MSC and osteolineage progenitors. Intriguingly, mice with a SHIP-deficient MSC compartment also exhibit decreased OC numbers. In agreement with our genetic findings we also show that treatment of mice with an SHIP1 inhibitor (SHIPi) significantly reduces bone mass. These findings demonstrate a novel role for SHIP1 in MSC fate determination and bone growth. Further, SHIPi may represent a novel therapeutic approach to limit bone development in osteopetrotic and sclerotic bone diseases.
Collapse
Affiliation(s)
- Sonia Iyer
- 1 Department of Microbiology and Immunology, SUNY Upstate Medical University , Syracuse, New York
| | | | | | | | | |
Collapse
|
25
|
Bone marrow vascular niche: home for hematopoietic stem cells. BONE MARROW RESEARCH 2014; 2014:128436. [PMID: 24822129 PMCID: PMC4009113 DOI: 10.1155/2014/128436] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 02/13/2014] [Accepted: 02/15/2014] [Indexed: 12/19/2022]
Abstract
Though discovered later than osteoblastic niche, vascular niche has been regarded as an alternative indispensable niche operating regulation on hematopoietic stem cells (HSCs). As significant progresses gained on this type niche, it is gradually clear that the main work of vascular niche is undertaking to support hematopoiesis. However, compared to what have been defined in the mechanisms through which the osteoblastic niche regulates hematopoiesis, we know less in vascular niche. In this review, based on research data hitherto we will focus on component foundation and various functions of vascular niche that guarantee the normal hematopoiesis process within bone marrow microenvironments. And the possible pathways raised by various research results through which this environment undergoes its function will be discussed as well.
Collapse
|
26
|
Govey PM, Jacobs JM, Tilton SC, Loiselle AE, Zhang Y, Freeman WM, Waters KM, Karin NJ, Donahue HJ. Integrative transcriptomic and proteomic analysis of osteocytic cells exposed to fluid flow reveals novel mechano-sensitive signaling pathways. J Biomech 2014; 47:1838-45. [PMID: 24720889 DOI: 10.1016/j.jbiomech.2014.03.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 03/11/2014] [Accepted: 03/11/2014] [Indexed: 01/01/2023]
Abstract
Osteocytes, positioned within bone׳s porous structure, are subject to interstitial fluid flow upon whole bone loading. Such fluid flow is widely theorized to be a mechanical signal transduced by osteocytes, initiating a poorly understood cascade of signaling events mediating bone adaptation to mechanical load. The objective of this study was to examine the time course of flow-induced changes in osteocyte gene transcript and protein levels using high-throughput approaches. Osteocyte-like MLO-Y4 cells were subjected to 2h of oscillating fluid flow (1Pa peak shear stress) and analyzed following 0, 2, 8, and 24h post-flow incubation. Transcriptomic microarray analysis, followed by gene ontology pathway analysis, demonstrated fluid flow regulation of genes consistent with both known and unknown metabolic and inflammatory responses in bone. Additionally, two of the more highly up-regulated gene products - chemokines Cxcl1 and Cxcl2, supported by qPCR - have not previously been reported as responsive to fluid flow. Proteomic analysis demonstrated greatest up-regulation of the ATP-producing enzyme NDK, calcium-binding Calcyclin, and G protein-coupled receptor kinase 6. Finally, an integrative pathway analysis merging fold changes in transcript and protein levels predicted signaling nodes not directly detected at the sampled time points, including transcription factors c-Myc, c-Jun, and RelA/NF-κB. These results extend our knowledge of the osteocytic response to fluid flow, most notably up-regulation of Cxcl1 and Cxcl2 as possible paracrine agents for osteoblastic and osteoclastic recruitment. Moreover, these results demonstrate the utility of integrative, high-throughput approaches in place of a traditional candidate approach for identifying novel mechano-sensitive signaling molecules.
Collapse
Affiliation(s)
- Peter M Govey
- Division of Musculoskeletal Sciences, Department of Orthopaedics and Rehabilitation, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Jon M Jacobs
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Susan C Tilton
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Alayna E Loiselle
- Division of Musculoskeletal Sciences, Department of Orthopaedics and Rehabilitation, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Yue Zhang
- Division of Musculoskeletal Sciences, Department of Orthopaedics and Rehabilitation, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Willard M Freeman
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Katrina M Waters
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Norman J Karin
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Henry J Donahue
- Division of Musculoskeletal Sciences, Department of Orthopaedics and Rehabilitation, Penn State College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
27
|
Calvi LM, Link DC. Cellular complexity of the bone marrow hematopoietic stem cell niche. Calcif Tissue Int 2014; 94:112-24. [PMID: 24101231 PMCID: PMC3936515 DOI: 10.1007/s00223-013-9805-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 09/15/2013] [Indexed: 12/17/2022]
Abstract
The skeleton serves as the principal site for hematopoiesis in adult terrestrial vertebrates. The function of the hematopoietic system is to maintain homeostatic levels of all circulating blood cells, including myeloid cells, lymphoid cells, red blood cells, and platelets. This action requires the daily production of more than 500 billion blood cells. The vast majority of these cells are synthesized in the bone marrow, where they arise from a limited number of hematopoietic stem cells (HSCs) that are multipotent and capable of extensive self-renewal. These attributes of HSCs are best demonstrated by marrow transplantation, where even a single HSC can repopulate the entire hematopoietic system. HSCs are therefore adult stem cells capable of multilineage repopulation, poised between cell fate choices which include quiescence, self-renewal, differentiation, and apoptosis. While HSC fate choices are in part determined by multiple stochastic fluctuations of cell autonomous processes, according to the niche hypothesis, signals from the microenvironment are also likely to determine stem cell fate. While it had long been postulated that signals within the bone marrow could provide regulation of hematopoietic cells, it is only in the past decade that advances in flow cytometry and genetic models have allowed for a deeper understanding of the microenvironmental regulation of HSCs. In this review, we will highlight the cellular regulatory components of the HSC niche.
Collapse
Affiliation(s)
- Laura M Calvi
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA,
| | | |
Collapse
|
28
|
Silvestre JS, Smadja DM, Lévy BI. Postischemic revascularization: from cellular and molecular mechanisms to clinical applications. Physiol Rev 2013; 93:1743-802. [PMID: 24137021 DOI: 10.1152/physrev.00006.2013] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After the onset of ischemia, cardiac or skeletal muscle undergoes a continuum of molecular, cellular, and extracellular responses that determine the function and the remodeling of the ischemic tissue. Hypoxia-related pathways, immunoinflammatory balance, circulating or local vascular progenitor cells, as well as changes in hemodynamical forces within vascular wall trigger all the processes regulating vascular homeostasis, including vasculogenesis, angiogenesis, arteriogenesis, and collateral growth, which act in concert to establish a functional vascular network in ischemic zones. In patients with ischemic diseases, most of the cellular (mainly those involving bone marrow-derived cells and local stem/progenitor cells) and molecular mechanisms involved in the activation of vessel growth and vascular remodeling are markedly impaired by the deleterious microenvironment characterized by fibrosis, inflammation, hypoperfusion, and inhibition of endogenous angiogenic and regenerative programs. Furthermore, cardiovascular risk factors, including diabetes, hypercholesterolemia, hypertension, diabetes, and aging, constitute a deleterious macroenvironment that participates to the abrogation of postischemic revascularization and tissue regeneration observed in these patient populations. Thus stimulation of vessel growth and/or remodeling has emerged as a new therapeutic option in patients with ischemic diseases. Many strategies of therapeutic revascularization, based on the administration of growth factors or stem/progenitor cells from diverse sources, have been proposed and are currently tested in patients with peripheral arterial disease or cardiac diseases. This review provides an overview from our current knowledge regarding molecular and cellular mechanisms involved in postischemic revascularization, as well as advances in the clinical application of such strategies of therapeutic revascularization.
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW Dipeptidyl peptidase 4 (DPP4, CD26) is a protease that cleaves selected amino acids at the N-terminal penultimate position and has the potential to alter the protein function. The regulation and roles of DPP4 activity are not well understood; therefore, the purpose of this review is to discuss the recent literature regarding DPP4 regulation, as well as the variety of molecules it may affect, and their potential clinical applications. RECENT FINDINGS Recent insight into the number of proteins that have DPP4 sites, and how DPP4 truncation may alter hematopoiesis based on the protein full length vs. truncated state, has shown that DPP4 truncation of colony-stimulating factors (CSFs) alters their function and that the activity of these CSFs can be enhanced when DPP4 activity is inhibited. DPP4 inhibition has recently been used in a clinical trial to attempt to enhance the engraftment of cord blood cells, and an endogenous DPP4 inhibitor tissue factor pathway inhibitor has been discovered, increasing our understanding of the potential importance of DPP4. SUMMARY DPP4 plays a role in regulating the activity of CSFs and other cytokines involved in hematopoiesis. This information may be useful for enhancing hematopoietic cell transplantation, blood cell recovery after stress, and for understanding the physiology and pathophysiology of blood and other cell systems.
Collapse
|