1
|
Sclerostin Depletion Induces Inflammation in the Bone Marrow of Mice. Int J Mol Sci 2021; 22:ijms22179111. [PMID: 34502021 PMCID: PMC8431516 DOI: 10.3390/ijms22179111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/18/2021] [Accepted: 08/20/2021] [Indexed: 01/25/2023] Open
Abstract
Romosozumab, a humanized monoclonal antibody specific for sclerostin (SOST), has been approved for treatment of postmenopausal women with osteoporosis at a high risk for fracture. Previous work in sclerostin global knockout (Sost-/-) mice indicated alterations in immune cell development in the bone marrow (BM), which could be a possible side effect in romosozumab-treated patients. Here, we examined the effects of short-term sclerostin depletion in the BM on hematopoiesis in young mice receiving sclerostin antibody (Scl-Ab) treatment for 6 weeks, and the effects of long-term Sost deficiency on wild-type (WT) long-term hematopoietic stem cells transplanted into older cohorts of Sost-/- mice. Our analyses revealed an increased frequency of granulocytes in the BM of Scl-Ab-treated mice and WT→Sost-/- chimeras, indicating myeloid-biased differentiation in Sost-deficient BM microenvironments. This myeloid bias extended to extramedullary hematopoiesis in the spleen and was correlated with an increase in inflammatory cytokines TNFα, IL-1α, and MCP-1 in Sost-/- BM serum. Additionally, we observed alterations in erythrocyte differentiation in the BM and spleen of Sost-/- mice. Taken together, our current study indicates novel roles for Sost in the regulation of myelopoiesis and control of inflammation in the BM.
Collapse
|
2
|
Pehlivan M, Caliskan C, Yuce Z, Sercan HO. sFRP1 Expression Regulates Wnt signaling in Chronic Myeloid Leukemia K562 Cells. Anticancer Agents Med Chem 2021; 22:1354-1362. [PMID: 34030621 DOI: 10.2174/1871520621666210524162145] [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: 12/10/2020] [Revised: 03/23/2021] [Accepted: 04/12/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Wnt signaling cascades play important roles in cell fate decisions and their deregulation has been documented in many diseases, including malignant tumors and leukemia. One mechanism of aberrant Wnt signaling is the silencing of Wnt inhibitors through epigenetic mechanisms. The sFRPs are one of the most studied Wnt inhibitors; and the sFRP1 loss is known in many hematological malignancies. Therefore, we aimed to compare the expression of Wnt related genes in the presence and absence of sFRP1 in chronic myeloid leukemia (CML) cell line. OBJECTIVE It is important to understand how sFRP1 and sFRP1 perform on CML to design new agents and strategies for resistant and advanced forms of CML. MATERIALS AND METHODS We used K562 cells, which normally do not express sFRP1 and its sFRP1 expressing subclone K562s. Total RNA was isolated from K562 and K562s cell lines end converted cDNA. PCR Array experiments performed using Human Wnt Signaling Pathway Plus RT2 Profiler™ kit. Wnt signaling pathway activation was studied by western blot for downstream signaling targets. RESULTS The WNT3, LRP6, PRICKLE1 and BTRC expressions were significantly decreased in the presence of sFRP1; while WNT5B increased. The sFRP1 expression inhibited stabilization of total β-catenin protein and downstream effector phosphorylation of noncanonical Wnt/PCP signaling; whereas Ca2+/PKC signaling remained active. CONCLUSION
Collapse
Affiliation(s)
- Melek Pehlivan
- Vocational School of Health Services, Izmir Katip Celebi University, Izmir, Turkey
| | - Ceyda Caliskan
- Department of Biomedical Sciences, University of Sheffield, Sheffield, United Kingdom
| | - Zeynep Yuce
- Dokuz Eylul University, Department of Medical Biology and Genetics, Izmir. Turkey
| | - Hakki O Sercan
- Dokuz Eylul University, Department of Medical Biology and Genetics, Izmir. Turkey
| |
Collapse
|
3
|
Prostaglandin E 2 Enhances Aged Hematopoietic Stem Cell Function. Stem Cell Rev Rep 2021; 17:1840-1854. [PMID: 33974233 DOI: 10.1007/s12015-021-10177-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2021] [Indexed: 10/21/2022]
Abstract
Aging of hematopoiesis is associated with increased frequency and clonality of hematopoietic stem cells (HSCs), along with functional compromise and myeloid bias, with donor age being a significant variable in survival after HSC transplantation. No clinical methods currently exist to enhance aged HSC function, and little is known regarding how aging affects molecular responses of HSCs to biological stimuli. Exposure of HSCs from young fish, mice, nonhuman primates, and humans to 16,16-dimethyl prostaglandin E2 (dmPGE2) enhances transplantation, but the effect of dmPGE2 on aged HSCs is unknown. Here we show that ex vivo pulse of bone marrow cells from young adult (3 mo) and aged (25 mo) mice with dmPGE2 prior to serial competitive transplantation significantly enhanced long-term repopulation from aged grafts in primary and secondary transplantation (27 % increase in chimerism) to a similar degree as young grafts (21 % increase in chimerism; both p < 0.05). RNA sequencing of phenotypically-isolated HSCs indicated that the molecular responses to dmPGE2 are similar in young and old, including CREB1 activation and increased cell survival and homeostasis. Common genes within these pathways identified likely key mediators of HSC enhancement by dmPGE2 and age-related signaling differences. HSC expression of the PGE2 receptor EP4, implicated in HSC function, increased with age in both mRNA and surface protein. This work suggests that aging does not alter the major dmPGE2 response pathways in HSCs which mediate enhancement of both young and old HSC function, with significant implications for expanding the therapeutic potential of elderly HSC transplantation.
Collapse
|
4
|
Zarrabi M, Afzal E, Asghari MH, Ebrahimi M. Combination of SB431542, Chir9901, and Bpv as a novel supplement in the culture of umbilical cord blood hematopoietic stem cells. Stem Cell Res Ther 2020; 11:474. [PMID: 33168035 PMCID: PMC7650159 DOI: 10.1186/s13287-020-01945-8] [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: 06/27/2020] [Accepted: 09/20/2020] [Indexed: 01/11/2023] Open
Abstract
Background Small molecule compounds have been well recognized for their promising power in the generation, expansion, and maintenance of embryonic or adult stem cells. The aim of this study was to identify a novel combination of small molecules in order to optimize the ex vivo expansion of umbilical cord blood-derived CD34+ cells. Methods Considering the most important signaling pathways involved in the self-renewal of hematopoietic stem cells, CB-CD34+ cells were expanded with cytokines in the presence of seven small molecules including SB, PD, Chir, Bpv, Pur, Pμ, and NAM. The eliminativism approach was used to find the best combination of selected small molecules for effective ex vivo expansion of CD34+ cell. In each step, proliferation, self-renewal, and clonogenic potential of the expanded cells as well as expression of some hematopoietic stem cell-related genes were studied. Finally, the engraftment potential of expanded cells was also examined by the mouse intra-uterine transplantation model. Results Our data shows that the simultaneous use of SB431542 (TGF-β inhibitor), Chir9901 (GSK3 inhibitor), and Bpv (PTEN inhibitor) resulted in a 50-fold increase in the number of CD34+CD38− cells. This was further reflected in approximately 3 times the increase in the clonogenic potential of the small molecule cocktail-expanded cells. These cells, also, showed a 1.5-fold higher engraftment potential in the peripheral blood of the NMRI model of in utero transplantation. These results are in total conformity with the upregulation of HOXB4, GATA2, and CD34 marker gene as well as the CXCR4 homing gene. Conclusion Taken together, our findings introduce a novel combination of small molecules to improve the yield of existing protocols used in the expansion of hematopoietic stem cells.
Collapse
Affiliation(s)
- Morteza Zarrabi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box, Tehran, 19395-4644, Iran.,Royan Stem Cell Technology Company, Cord Blood Bank, Tehran, Iran
| | - Elaheh Afzal
- Royan Stem Cell Technology Company, Cord Blood Bank, Tehran, Iran
| | - Mohammad Hassan Asghari
- Animal Core Facility, Reproductive Biomedicine Research Center, Royan Institute for Animal Biotechnology, ACECR, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box, Tehran, 19395-4644, Iran.
| |
Collapse
|
5
|
Optimizing BIO feeding strategy promotes ex vivo expansion of human hematopoietic stem and progenitor cells. J Biosci Bioeng 2020; 131:190-197. [PMID: 33127294 DOI: 10.1016/j.jbiosc.2020.09.020] [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: 07/16/2020] [Revised: 09/18/2020] [Accepted: 09/28/2020] [Indexed: 11/21/2022]
Abstract
Ex vivo expansion is critical in facilitating the application of hematopoietic/progenitor stem cells (HSPCs) for regenerative therapies. Wnt signaling is implicated in the expansion and self-renewal maintenance of HSPCs. However, a reasonable method to regulate Wnt signaling in ex vivo cultures to achieve robust expansion of HSPCs has not yet been investigated. Here, cord blood-derived CD34+ cells were cultured with the activator of Wnt signaling 6-bromoindirubin-3'-oxime (BIO) under the following conditions: vehicle control (group A); BIO was added to the culture on days 0, 4, and 7 (group B); and BIO was added to the culture on days 0 and 7 (group C). Initial BIO treatment promoted the expansion of CD34+ cells on day 4. However, BIO supplementation on days 0 and 4 in group B attenuated HSPC expansion on day 7, while enhancing the multilineage commit potential and secondary expansion ability of expanded CD34+ cells. Based on this finding, an optimized BIO feeding strategy (group C) was proposed to support substantial expansion of HSPCs. After 10 days of culture, the expansion fold of CD34+ cells was 28.70 ± 0.46-folds, which was significantly higher than group A (16.20 ± 0.72-folds, p < 0.05). Moreover, the optimized BIO feeding strategy achieved increased primitive HSPC expansion without the loss of biological functions. Mechanistically, the optimized BIO feeding strategy avoided the excessive activation of Wnt observed in group B while maintaining a moderate level of intracellular β-catenin. These results provide an experimental and theoretical basis for Wnt regulation in ex vivo culture process and a potential strategy to expand HSPCs for transplantation.
Collapse
|
6
|
Pehlivan M, Soyoz M, Cerci B, Coven HIK, Yuce Z, Sercan HO. sFRP1 Expression Induces miRNAs That Modulate Wnt Signaling in Chronic Myeloid Leukemia Cells. Mol Biol 2020. [DOI: 10.1134/s0026893320040135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
7
|
Abstract
PURPOSE OF REVIEW We reviewed recent progress on the role of sclerostin (SOST) and its effects on the immune system in order to summarize the current state of knowledge in osteoimmunology, in regard to hematopoiesis, lymphopoiesis, and inflammation. RECENT FINDINGS Changes in sclerostin levels affect distinct niches within the bone marrow that support hematopoietic stem cells and B cell development. Sclerostin's regulation of adipogenesis could also be important for immune cell maintenance with age. Surprisingly, B cell development in the bone marrow is influenced by Sost produced by mesenchymal stem cells and osteoblasts, but not by osteocytes. Additionally, extramedullary hematopoiesis in the spleen and increased pro-inflammatory cytokine levels in the bone marrow are observed in global Sost-/- mice. In addition to changes in bone marrow density, sclerostin depletion affects B lymphopoiesis and myelopoiesis, as well as other changes within the bone marrow cavity that could affect hematopoiesis. It is therefore important to monitor for hematopoietic changes in patients receiving sclerostin-depleting therapies.
Collapse
Affiliation(s)
- Cristine Donham
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, 95343, USA
| | - Jennifer O Manilay
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, 95343, USA.
- Dept. of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, 5200, Lake Road Merced, North, CA, 95343, USA.
| |
Collapse
|
8
|
Herrmann M, Jakob F. Bone Marrow Niches for Skeletal Progenitor Cells and their Inhabitants in Health and Disease. Curr Stem Cell Res Ther 2019; 14:305-319. [PMID: 30674266 DOI: 10.2174/1574888x14666190123161447] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/04/2018] [Accepted: 01/02/2019] [Indexed: 12/19/2022]
Abstract
The bone marrow hosts skeletal progenitor cells which have most widely been referred to as Mesenchymal Stem or Stromal Cells (MSCs), a heterogeneous population of adult stem cells possessing the potential for self-renewal and multilineage differentiation. A consensus agreement on minimal criteria has been suggested to define MSCs in vitro, including adhesion to plastic, expression of typical surface markers and the ability to differentiate towards the adipogenic, osteogenic and chondrogenic lineages but they are critically discussed since the differentiation capability of cells could not always be confirmed by stringent assays in vivo. However, these in vitro characteristics have led to the notion that progenitor cell populations, similar to MSCs in bone marrow, reside in various tissues. MSCs are in the focus of numerous (pre)clinical studies on tissue regeneration and repair. Recent advances in terms of genetic animal models enabled a couple of studies targeting skeletal progenitor cells in vivo. Accordingly, different skeletal progenitor cell populations could be identified by the expression of surface markers including nestin and leptin receptor. While there are still issues with the identity of, and the overlap between different cell populations, these studies suggested that specific microenvironments, referred to as niches, host and maintain skeletal progenitor cells in the bone marrow. Dynamic mutual interactions through biological and physical cues between niche constituting cells and niche inhabitants control dormancy, symmetric and asymmetric cell division and lineage commitment. Niche constituting cells, inhabitant cells and their extracellular matrix are subject to influences of aging and disease e.g. via cellular modulators. Protective niches can be hijacked and abused by metastasizing tumor cells, and may even be adapted via mutual education. Here, we summarize the current knowledge on bone marrow skeletal progenitor cell niches in physiology and pathophysiology. We discuss the plasticity and dynamics of bone marrow niches as well as future perspectives of targeting niches for therapeutic strategies.
Collapse
Affiliation(s)
- Marietta Herrmann
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Clinics Wuerzburg, Wuerzburg, Germany.,Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
9
|
Chicana B, Donham C, Millan AJ, Manilay JO. Wnt Antagonists in Hematopoietic and Immune Cell Fate: Implications for Osteoporosis Therapies. Curr Osteoporos Rep 2019; 17:49-58. [PMID: 30835038 PMCID: PMC6715281 DOI: 10.1007/s11914-019-00503-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW We reviewed the current literature on the roles of the Wnt antagonists sclerostin (Sost) and sclerostin-containing domain protein 1 (Sostdc1) on bone homeostasis, the relationship of the hypoxia-inducible factor (Hif) and von Hippel-Lindau (Vhl) pathways on Sost expression, and how changes in bone induced by depletion of Sost, Sostdc1, and Vhl affect hematopoietic cells. RECENT FINDINGS B cell development is adversely affected in Sost-knockout mice and is more severely affected in Vhl-knockout mice. Inflammation in the Sost-/- bone microenvironment could alter hematopoietic stem cell behavior. Sostdc1-/- mice display defects in natural killer cell development and cytotoxicity. Depletion of Sost and Sostdc1 have effects on immune cell function that warrant investigation in patients receiving Wnt antagonist-depleting therapies for treatment of bone diseases. Additional clinical applications for manipulation of Wnt antagonists include cancer immunotherapies, stem cell transplantation, and directed differentiation to immune lineages.
Collapse
Affiliation(s)
- Betsabel Chicana
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA, USA
| | - Cristine Donham
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA, USA
| | - Alberto J Millan
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA, USA
| | - Jennifer O Manilay
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA, USA.
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, 5200 North Lake Road, Merced, CA, 95343, USA.
| |
Collapse
|
10
|
Said NM. Three gold indicators for breast cancer prognosis: a case-control study with ROC analysis for novel ratios related to CBC with (ALP and LDH). Mol Biol Rep 2019; 46:2013-2027. [PMID: 30706358 DOI: 10.1007/s11033-019-04650-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 01/24/2019] [Indexed: 02/06/2023]
Abstract
Science is still unable to develop a specific strategy for predicting breast cancer in humans. Several attempts are done to obtain the best and closest prognostic predictive biomarkers for breast cancer. The present study aimed to evaluate the impact of novel ratios calculated between the blood indices with CA15.3, alkaline phosphatase and lactate dehydrogenase as prognostic biomarkers in breast cancer. This study was conducted on two groups (Breast cancer Patients group in comparison to a control group who has no tumor family history). All the volunteers are subjected to the routine analysis included liver and kidney function tests, complete blood count with blood indices, tumor markers (CA15.3) assessment, alkaline phosphatase, and lactate dehydrogenase analysis. Thirty different ratios were calculated in the present research between blood indices and three inexpensive serum biomarkers; CA15.3, alkaline phosphatase and lactate dehydrogenase. Fifteen ratios of them were significant in breast cancer group than the control group. Three ratios (PDW/lymphocytes, MPV/lymphocytes, and ALP/RDW) of them gave a sensitivity of 100% with high specificity as indicators for breast cancer incidence. The correlation between significant ratios was very interesting. The more interesting was in the results of subgroup analysis which showed that the ALP/RDW ratio is more specific for pre-menopause while PDW/lymphocytes ratio is more specific for post-menopause. The ratios PDW/lymphocytes, MPV/lymphocytes, and ALP/RDW can be used as prognostic biomarkers in breast cancer patients. The interesting advantage in the results depends on the availability of these indicators in routine blood analysis and will not increase the cost of the diagnostic plan.
Collapse
Affiliation(s)
- Noha Mohamed Said
- Biochemistry Division, Chemistry Department, Faculty of Science, Zagzig University, Zagazig, Egypt.
| |
Collapse
|
11
|
Pehlivan M, Çalışkan C, Yüce Z, Sercan HO. Secreted Wnt antagonists in leukemia: A road yet to be paved. Leuk Res 2018; 69:24-30. [PMID: 29625321 DOI: 10.1016/j.leukres.2018.03.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 02/21/2018] [Accepted: 03/23/2018] [Indexed: 11/20/2022]
Abstract
Wnt signaling has been a topic of research for many years for its diverse and fundamental functions in physiological (such as embryogenesis, organogenesis, proliferation, tissue repair and cellular differentiation) and pathological (carcinogenesis, congenital/genetic diseases, and tissue degeneration) processes. Wnt signaling pathway aberrations are associated with both solid tumors and hematological malignancies. Unregulated Wnt signaling observed in malignancies may be due to a wide spectrum of abnormalities, from mutations in the genes of key players to epigenetic modifications of Wnt antagonists. Of these, Wnt antagonists are gaining significant attention for their potential of being targets for treatment and inhibition of Wnt signaling. In this review, we discuss and summarize the significance of Wnt signaling antagonists in the pathogenesis and treatment of hematological malignancies.
Collapse
Affiliation(s)
- Melek Pehlivan
- Vocational School of Health Services, Izmir Katip Celebi University, Izmir, Turkey.
| | - Ceyda Çalışkan
- Izmir Institute of Technology, Faculty of Science, Department of Molecular Biology & Genetics, Izmir, Turkey.
| | - Zeynep Yüce
- Dokuz Eylul University Faculty of Medicine, Department of Medical Biology and Genetics, Izmir, Turkey.
| | - Hakki Ogun Sercan
- Dokuz Eylul University Faculty of Medicine, Department of Medical Biology and Genetics, Izmir, Turkey.
| |
Collapse
|
12
|
Batsali AK, Pontikoglou C, Koutroulakis D, Pavlaki KI, Damianaki A, Mavroudi I, Alpantaki K, Kouvidi E, Kontakis G, Papadaki HA. Differential expression of cell cycle and WNT pathway-related genes accounts for differences in the growth and differentiation potential of Wharton's jelly and bone marrow-derived mesenchymal stem cells. Stem Cell Res Ther 2017; 8:102. [PMID: 28446235 PMCID: PMC5406919 DOI: 10.1186/s13287-017-0555-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/14/2017] [Accepted: 04/05/2017] [Indexed: 12/25/2022] Open
Abstract
Background In view of the current interest in exploring the clinical use of mesenchymal stem cells (MSCs) from different sources, we performed a side-by-side comparison of the biological properties of MSCs isolated from the Wharton’s jelly (WJ), the most abundant MSC source in umbilical cord, with bone marrow (BM)-MSCs, the most extensively studied MSC population. Methods MSCs were isolated and expanded from BM aspirates of hematologically healthy donors (n = 18) and from the WJ of full-term neonates (n = 18). We evaluated, in parallel experiments, the MSC immunophenotypic, survival and senescence characteristics as well as their proliferative potential and cell cycle distribution. We also assessed the expression of genes associated with the WNT- and cell cycle-signaling pathway and we performed karyotypic analysis through passages to evaluate the MSC genomic stability. The hematopoiesis-supporting capacity of MSCs from both sources was investigated by evaluating the clonogenic cells in the non-adherent fraction of MSC co-cultures with BM or umbilical cord blood-derived CD34+ cells and by measuring the hematopoietic cytokines levels in MSC culture supernatants. Finally, we evaluated the ability of MSCs to differentiate into adipocytes and osteocytes and the effect of the WNT-associated molecules WISP-1 and sFRP4 on the differentiation potential of WJ-MSCs. Results Both ex vivo-expanded MSC populations showed similar morphologic, immunophenotypic, survival and senescence characteristics and acquired genomic alterations at low frequency during passages. WJ-MSCs exhibited higher proliferative potential, possibly due to upregulation of genes that stimulate cell proliferation along with downregulation of genes related to cell cycle inhibition. WJ-MSCs displayed inferior lineage priming and differentiation capacity toward osteocytes and adipocytes, compared to BM-MSCs. This finding was associated with differential expression of molecules related to WNT signaling, including WISP1 and sFRP4, the respective role of which in the differentiation potential of WJ-MSCs was specifically investigated. Interestingly, treatment of WJ-MSCs with recombinant human WISP1 or sFRP4 resulted in induction of osteogenesis and adipogenesis, respectively. WJ-MSCs exhibited inferior hematopoiesis-supporting potential probably due to reduced production of stromal cell-Derived Factor-1α, compared to BM-MSCs. Conclusions Overall, these data are anticipated to contribute to the better characterization of WJ-MSCs and BM-MSCs for potential clinical applications.
Collapse
Affiliation(s)
- Aristea K Batsali
- University of Crete School of Medicine, Heraklion, Greece.,Graduate Program "Molecular Basis of Human Disease", University of Crete School of Medicine, Heraklion, Greece
| | | | - Dimitrios Koutroulakis
- Department of Obstetrics and Gynecology, University of Crete School of Medicine, Heraklion, Greece
| | | | | | - Irene Mavroudi
- University of Crete School of Medicine, Heraklion, Greece
| | - Kalliopi Alpantaki
- Department of Orthopedics and Traumatology, University of Crete School of Medicine, Heraklion, Greece
| | | | - George Kontakis
- Department of Orthopedics and Traumatology, University of Crete School of Medicine, Heraklion, Greece
| | | |
Collapse
|
13
|
Lin X, Dong R, Diao S, Yu G, Wang L, Li J, Fan Z. SFRP2 enhanced the adipogenic and neuronal differentiation potentials of stem cells from apical papilla. Cell Biol Int 2017; 41:534-543. [PMID: 28244619 DOI: 10.1002/cbin.10757] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 02/24/2017] [Indexed: 01/04/2023]
Abstract
Dental tissue-derived mesenchymal stem cells (MSCs) are easily obtained and considered as a favorable cell source for tissue engineering, but the regulation of direct differentiation is unknown, which restricts their application. The present study investigated the effect of SFRP2, a Wnt signaling modulator, on MSC differentiation using stem cells from apical papilla (SCAPs). The cells were cultured in specific inducing medium for adipogenic, neurogenic, or chondrogenic differentiation. Over-expression of SFRP2 via retroviral infection enhanced the adipogenic and neurogenic differentiation of SCAPs. While inhibit of Wnt pathway by IWR1-endo could enhance the neurogenic differentiation potentials of SCAPs, similar with the function of SFRP2. In addition, over-expression of SFRP2 up-regulated the expression of stemness-related genes SOX2 and OCT4. Furthermore, SOX2 and OCT4 expression was significantly inhibited after lentiviral silencing of SFRP2 in SCAPs. Therefore, our results suggest that SFRP2 enhances the adipogenic and neurogenic differentiation potentials of SCAPs by up-regulating SOX2 and OCT4. Moreover, the effect of SFRP2 in neurogenic differentiation of SCAPs maybe also associated with Wnt inhibition. Our results provided useful information about the molecular mechanism underlying directed differentiation in dental tissue-derived MSCs.
Collapse
Affiliation(s)
- Xiao Lin
- Laboratory of Molecular Signaling and Stem Cells Therapy, Capital Medical University School of Stomatology, Beijing, China.,Department of Implant Dentistry, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Rui Dong
- Laboratory of Molecular Signaling and Stem Cells Therapy, Capital Medical University School of Stomatology, Beijing, China
| | - Shu Diao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Capital Medical University School of Stomatology, Beijing, China.,Department of Pediatric Dentistry, Capital Medical University School of Stomatology, Beijing, China
| | - Guoxia Yu
- Laboratory of Molecular Signaling and Stem Cells Therapy, Capital Medical University School of Stomatology, Beijing, China.,Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China.,Department of Stomatology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Liping Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Capital Medical University School of Stomatology, Beijing, China
| | - Jun Li
- Department of Implant Dentistry, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Capital Medical University School of Stomatology, Beijing, China
| |
Collapse
|
14
|
The thyroid hormone nuclear receptors and the Wnt/β-catenin pathway: An intriguing liaison. Dev Biol 2017; 422:71-82. [DOI: 10.1016/j.ydbio.2017.01.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 12/26/2016] [Accepted: 01/04/2017] [Indexed: 12/11/2022]
|
15
|
Pettinato G, Ramanathan R, Fisher RA, Mangino MJ, Zhang N, Wen X. Scalable Differentiation of Human iPSCs in a Multicellular Spheroid-based 3D Culture into Hepatocyte-like Cells through Direct Wnt/β-catenin Pathway Inhibition. Sci Rep 2016; 6:32888. [PMID: 27616299 PMCID: PMC5018737 DOI: 10.1038/srep32888] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 08/16/2016] [Indexed: 12/26/2022] Open
Abstract
Treatment of acute liver failure by cell transplantation is hindered by a shortage of human hepatocytes. Current protocols for hepatic differentiation of human induced pluripotent stem cells (hiPSCs) result in low yields, cellular heterogeneity, and limited scalability. In the present study, we have developed a novel multicellular spheroid-based hepatic differentiation protocol starting from embryoid bodies of hiPSCs (hiPSC-EBs) for robust mass production of human hepatocyte-like cells (HLCs) using two novel inhibitors of the Wnt pathway. The resultant hiPSC-EB-HLCs expressed liver-specific genes, secreted hepatic proteins such as Albumin, Alpha Fetoprotein, and Fibrinogen, metabolized ammonia, and displayed cytochrome P450 activities and functional activities typical of mature primary hepatocytes, such as LDL storage and uptake, ICG uptake and release, and glycogen storage. Cell transplantation of hiPSC-EB-HLC in a rat model of acute liver failure significantly prolonged the mean survival time and resolved the liver injury when compared to the no-transplantation control animals. The transplanted hiPSC-EB-HLCs secreted human albumin into the host plasma throughout the examination period (2 weeks). Transplantation successfully bridged the animals through the critical period for survival after acute liver failure, providing promising clues of integration and full in vivo functionality of these cells after treatment with WIF-1 and DKK-1.
Collapse
Affiliation(s)
- Giuseppe Pettinato
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, USA
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Rajesh Ramanathan
- Department of Surgery, Virginia Commonwealth University Medical Center, Richmond, VA, USA
| | - Robert A Fisher
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Martin J. Mangino
- Department of Surgery, Virginia Commonwealth University Medical Center, Richmond, VA, USA
| | - Ning Zhang
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Xuejun Wen
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, USA
- Shanghai East Hospital, The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai 200120, People’s Republic of China
| |
Collapse
|
16
|
Van Camp JK, Beckers S, Zegers D, Verhulst SL, Van Hoorenbeeck K, Massa G, Verrijken A, Desager KN, Van Gaal LF, Van Hul W. Nucleotide variation of sFRP5 gene is not associated with obesity in children and adolescents. Mol Biol Rep 2016; 43:1041-7. [PMID: 27497818 DOI: 10.1007/s11033-016-4050-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 07/29/2016] [Indexed: 12/12/2022]
Abstract
Because sFRP5 was shown to be an important extracellular modulator of the Wnt pathway, regulating adipogenesis, we wanted to investigate the role of sFRP5 variants in human, monogenic obesity by performing mutation analysis. We screened the complete sFRP5 coding region in 622 obese children and adolescents and 503 lean control individuals by high-resolution melting curve analysis and direct sequencing. We found a total of 15 sequence variants in sFRP5, 10 of which resulted in a non-synonymous amino acid change. Five of these variants were, to our knowledge, not previously reported. For one of the variants (c.-3G>A), we identified a trend towards association between the variant frequency and the obese phenotype. We argue that, when looking at conservation and location inside known protein domains, several of the identified variants (D103N, A113V, K212N and H317L), may affect sFRP5 protein function. In addition, we found c.-3G>A, residing in the Kozak sequence, with a lower frequency in cases compared to controls. However, functional studies investigating the effect of sFRP5 variants on protein function are necessary to determine the true role of sFRP5 genetic variation in human, monogenic obesity.
Collapse
Affiliation(s)
- Jasmijn K Van Camp
- Department of Medical Genetics, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Sigri Beckers
- Department of Medical Genetics, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Doreen Zegers
- Department of Medical Genetics, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Stijn L Verhulst
- Department of Paediatrics, Antwerp University Hospital, Wilrijkstraat 10, 2650, Antwerp, Belgium
| | - Kim Van Hoorenbeeck
- Department of Paediatrics, Antwerp University Hospital, Wilrijkstraat 10, 2650, Antwerp, Belgium
| | - Guy Massa
- Department of Paediatrics, Jessa Hospital, Stadsomvaart 11, 3500, Hasselt, Belgium
| | - An Verrijken
- Department of Endocrinology, Diabetology and Metabolic Diseases, Antwerp University Hospital, Wilrijkstraat 10, 2650, Antwerp, Belgium
| | - Kristine N Desager
- Department of Paediatrics, Antwerp University Hospital, Wilrijkstraat 10, 2650, Antwerp, Belgium
| | - Luc F Van Gaal
- Department of Endocrinology, Diabetology and Metabolic Diseases, Antwerp University Hospital, Wilrijkstraat 10, 2650, Antwerp, Belgium
| | - Wim Van Hul
- Department of Medical Genetics, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium.
| |
Collapse
|
17
|
Cain CJ, Gaborit N, Lwin W, Barruet E, Ho S, Bonnard C, Hamamy H, Shboul M, Reversade B, Kayserili H, Bruneau BG, Hsiao EC. Loss of Iroquois homeobox transcription factors 3 and 5 in osteoblasts disrupts cranial mineralization. Bone Rep 2016; 5:86-95. [PMID: 27453922 PMCID: PMC4926823 DOI: 10.1016/j.bonr.2016.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 02/02/2016] [Indexed: 01/13/2023] Open
Abstract
Cranial malformations are a significant cause of perinatal morbidity and mortality. Iroquois homeobox transcription factors (IRX) are expressed early in bone tissue formation and facilitate patterning and mineralization of the skeleton. Mice lacking Irx5 appear grossly normal, suggesting that redundancy within the Iroquois family. However, global loss of both Irx3 and Irx5 in mice leads to significant skeletal malformations and embryonic lethality from cardiac defects. Here, we study the bone-specific functions of Irx3 and Irx5 using Osx-Cre to drive osteoblast lineage-specific deletion of Irx3 in Irx5(-/-) mice. Although we found that the Osx-Cre transgene alone could also affect craniofacial mineralization, newborn Irx3 (flox/flox) /Irx5(-/-)/Osx-Cre (+) mice displayed additional mineralization defects in parietal, interparietal, and frontal bones with enlarged sutures and reduced calvarial expression of osteogenic genes. Newborn endochondral long bones were largely unaffected, but we observed marked reductions in 3-4-week old bone mineral content of Irx3 (flox/flox) /Irx5(-/-)/Osx-Cre (+) mice. Our findings indicate that IRX3 and IRX5 can work together to regulate mineralization of specific cranial bones. Our results also provide insight into the causes of the skeletal changes and mineralization defects seen in Hamamy syndrome patients carrying mutations in IRX5.
Collapse
Affiliation(s)
- Corey J Cain
- Department of Medicine, Division of Endocrinology and Metabolism, Institute for Human Genetics, Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94143-0794, USA
| | - Nathalie Gaborit
- Inserm, UMR 1087, l'institut du thorax, Nantes, France; CNRS, UMR 6291, Nantes, France; Université de Nantes, France
| | - Wint Lwin
- Department of Medicine, Division of Endocrinology and Metabolism, Institute for Human Genetics, Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94143-0794, USA
| | - Emilie Barruet
- Department of Medicine, Division of Endocrinology and Metabolism, Institute for Human Genetics, Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94143-0794, USA
| | - Samantha Ho
- Department of Medicine, Division of Endocrinology and Metabolism, Institute for Human Genetics, Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94143-0794, USA
| | - Carine Bonnard
- Human Embryology and Genetics Laboratory, Institute of Medical Biology, ASTAR, Singapore 138648, Singapore
| | - Hanan Hamamy
- Department of Genetic Medicine and Development, Geneva University, Geneva 1211, Switzerland
| | - Mohammad Shboul
- Human Embryology and Genetics Laboratory, Institute of Medical Biology, ASTAR, Singapore 138648, Singapore
| | - Bruno Reversade
- Human Embryology and Genetics Laboratory, Institute of Medical Biology, ASTAR, Singapore 138648, Singapore
| | - Hülya Kayserili
- Medical Genetics Department, Koc University School of Medicine, Rumelifeneri Yolu, Sarıyer, Istanbul 34450, Turkey; Medical Genetics Department, Istanbul Medical Faculty, Istanbul University Topkapi, Fatih, 34093 lstanbul, Turkey
| | - Benoit G Bruneau
- Gladstone Institute for Cardiovascular Disease, San Francisco, CA 94158, USA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Edward C Hsiao
- Department of Medicine, Division of Endocrinology and Metabolism, Institute for Human Genetics, Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94143-0794, USA
| |
Collapse
|
18
|
Cain CJ, Valencia JT, Ho S, Jordan K, Mattingly A, Morales BM, Hsiao EC. Increased Gs Signaling in Osteoblasts Reduces Bone Marrow and Whole-Body Adiposity in Male Mice. Endocrinology 2016; 157:1481-94. [PMID: 26901092 PMCID: PMC4816728 DOI: 10.1210/en.2015-1867] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/16/2016] [Indexed: 12/21/2022]
Abstract
Bone is increasingly recognized as an endocrine organ that can regulate systemic hormones and metabolism through secreted factors. Although bone loss and increased adiposity appear to be linked clinically, whether conditions of increased bone formation can also change systemic metabolism remains unclear. In this study, we examined how increased osteogenesis affects metabolism by using an engineered G protein-coupled receptor, Rs1, to activate Gs signaling in osteoblastic cells in ColI(2.3)(+)/Rs1(+) transgenic mice. We previously showed that these mice have dramatically increased bone formation resembling fibrous dysplasia of the bone. We found that total body fat was significantly reduced starting at 3 weeks of age. Furthermore, ColI(2.3)(+)/Rs1(+) mice showed reduced O2 consumption and respiratory quotient measures without effects on food intake and energy expenditure. The mice had significantly decreased serum triacylglycerides, leptin, and adiponectin. Resting glucose and insulin levels were unchanged; however, glucose and insulin tolerance tests revealed increased sensitivity to insulin. The mice showed resistance to fat accumulation from a high-fat diet. Furthermore, ColI(2.3)(+)/Rs1(+) mouse bones had dramatically reduced mature adipocyte differentiation, increased Wingless/Int-1 (Wnt) signaling, and higher osteoblastic glucose utilization than controls. These findings suggest that osteoblasts can influence both local and peripheral adiposity in conditions of increased bone formation and suggest a role for osteoblasts in the regulation of whole-body adiposity and metabolic homeostasis.
Collapse
Affiliation(s)
- Corey J Cain
- Department of Medicine, Division of Endocrinology and Metabolism; Institute for Human Genetics; and Program in Craniofacial Biology (C.J.C., S.H., K.J., A.M., B.M.M., and E.C.H.); and the Biomedical Sciences Graduate Program (J.T.V. and E.C.H.); University of California, San Francisco, San Francisco, California 94143-0794
| | - Joel T Valencia
- Department of Medicine, Division of Endocrinology and Metabolism; Institute for Human Genetics; and Program in Craniofacial Biology (C.J.C., S.H., K.J., A.M., B.M.M., and E.C.H.); and the Biomedical Sciences Graduate Program (J.T.V. and E.C.H.); University of California, San Francisco, San Francisco, California 94143-0794
| | - Samantha Ho
- Department of Medicine, Division of Endocrinology and Metabolism; Institute for Human Genetics; and Program in Craniofacial Biology (C.J.C., S.H., K.J., A.M., B.M.M., and E.C.H.); and the Biomedical Sciences Graduate Program (J.T.V. and E.C.H.); University of California, San Francisco, San Francisco, California 94143-0794
| | - Kate Jordan
- Department of Medicine, Division of Endocrinology and Metabolism; Institute for Human Genetics; and Program in Craniofacial Biology (C.J.C., S.H., K.J., A.M., B.M.M., and E.C.H.); and the Biomedical Sciences Graduate Program (J.T.V. and E.C.H.); University of California, San Francisco, San Francisco, California 94143-0794
| | - Aaron Mattingly
- Department of Medicine, Division of Endocrinology and Metabolism; Institute for Human Genetics; and Program in Craniofacial Biology (C.J.C., S.H., K.J., A.M., B.M.M., and E.C.H.); and the Biomedical Sciences Graduate Program (J.T.V. and E.C.H.); University of California, San Francisco, San Francisco, California 94143-0794
| | - Blanca M Morales
- Department of Medicine, Division of Endocrinology and Metabolism; Institute for Human Genetics; and Program in Craniofacial Biology (C.J.C., S.H., K.J., A.M., B.M.M., and E.C.H.); and the Biomedical Sciences Graduate Program (J.T.V. and E.C.H.); University of California, San Francisco, San Francisco, California 94143-0794
| | - Edward C Hsiao
- Department of Medicine, Division of Endocrinology and Metabolism; Institute for Human Genetics; and Program in Craniofacial Biology (C.J.C., S.H., K.J., A.M., B.M.M., and E.C.H.); and the Biomedical Sciences Graduate Program (J.T.V. and E.C.H.); University of California, San Francisco, San Francisco, California 94143-0794
| |
Collapse
|
19
|
Zhang H, Yu C, Chen M, Li Z, Tian S, Jiang J, Sun C. miR-522 contributes to cell proliferation of hepatocellular carcinoma by targeting DKK1 and SFRP2. Tumour Biol 2016; 37:11321-9. [PMID: 26960688 DOI: 10.1007/s13277-016-4995-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/25/2016] [Indexed: 01/23/2023] Open
Abstract
The morbidity and mortality of hepatocellular carcinoma (HCC) is very high, finding new therapeutic targets are critical for HCC treatment. miR-522 has been demonstrated to be upregulated in HCC tissues, but its role in HCC progression remains to be elucidated. In this report, we found miR-522 was upregulated in HCC cells and tissues, miR-522 overexpression promoted cell proliferation, colony formation, and cell cycle progression, whereas knockdown of miR-522 reduced these effects. We also analyzed the expression of several key cell cycle regulatory proteins and found overexpression of miR-522-inhibited cell cycle inhibitors p21 and p27 expression and enhanced cyclin D1 expression and the level of Rb phosphorylation, vice versa. These suggested miR-522-accelerated G1/S transition. DKK1 (dickkopf-1) and SFRP2 (secreted frizzled-related protein 2) were the targets of miR-522, their expression was inversely with miR-522 in HCC tissues. DKK1 and SFRP2 the antagonists of Wnt signaling, suggesting miR-522-promoted HCC progression through activating Wnt signaling. miR-522 might be a valuable target for HCC therapy.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, 28 Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou, China
| | - Chao Yu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, 28 Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou, China
| | - Meiyuan Chen
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, 28 Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou, China
| | - Zhu Li
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, 28 Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou, China
| | - Se Tian
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, 28 Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou, China
| | - Jianxin Jiang
- Department of Hepatic-Biliary-Pancreatic Surgery, Hubei Cancer Hospital, 116 Zhuodaoquan south road, Hongshan district, Wuhan, 430079, Hubei, People's Republic of China.
| | - Chengyi Sun
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, 28 Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou, China.
| |
Collapse
|
20
|
Zöller M. CD44, Hyaluronan, the Hematopoietic Stem Cell, and Leukemia-Initiating Cells. Front Immunol 2015; 6:235. [PMID: 26074915 PMCID: PMC4443741 DOI: 10.3389/fimmu.2015.00235] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 04/30/2015] [Indexed: 12/14/2022] Open
Abstract
CD44 is an adhesion molecule that varies in size due to glycosylation and insertion of so-called variant exon products. The CD44 standard isoform (CD44s) is highly expressed in many cells and most abundantly in cells of the hematopoietic system, whereas expression of CD44 variant isoforms (CD44v) is more restricted. CD44s and CD44v are known as stem cell markers, first described for hematopoietic stem cells and later on confirmed for cancer- and leukemia-initiating cells. Importantly, both abundantly expressed CD44s as well as CD44v actively contribute to the maintenance of stem cell features, like generating and embedding in a niche, homing into the niche, maintenance of quiescence, and relative apoptosis resistance. This is surprising, as CD44 is not a master stem cell gene. I here will discuss that the functional contribution of CD44 relies on its particular communication skills with neighboring molecules, adjacent cells and, last not least, the surrounding matrix. In fact, it is the interaction of the hyaluronan receptor CD44 with its prime ligand, which strongly assists stem cells to fulfill their special and demanding tasks. Recent fundamental progress in support of this “old” hypothesis, which may soon pave the way for most promising new therapeutics, is presented for both hematopoietic stem cell and leukemia-initiating cell. The contribution of CD44 to the generation of a stem cell niche, to homing of stem cells in their niche, to stem cell quiescence and apoptosis resistance will be in focus.
Collapse
Affiliation(s)
- Margot Zöller
- Department of Tumor Cell Biology, University Hospital of Surgery , Heidelberg , Germany
| |
Collapse
|
21
|
Raghavachari N. Gene expression profiling of hematopoietic stem cells (HSCs). Methods Mol Biol 2015; 1185:91-119. [PMID: 25062624 DOI: 10.1007/978-1-4939-1133-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Transcriptomic analysis to decipher the molecular phenotype of hematopoietic stem cells, regulatory mechanisms directing their life cycle, and the molecular signals mediating proliferation, mobilization, migration, and differentiation is believed to unravel disease-specific disturbances in hematological diseases and assist in the development of novel cell-based clinical therapies in this era of genomic medicine. The recent advent in genomic tools and technologies is now enabling the study of such comprehensive transcriptional characterization of cell types in a robust and successful manner. This chapter describes detailed protocols for isolating RNA from purified population of hematopoietic cells and gene expression profiling of those purified cells using both microarrays (Affymetrix) and RNA-Seq technology (Illumina Platform).
Collapse
Affiliation(s)
- Nalini Raghavachari
- Division of Geriatrics and Clinical Gerontology, National Institute on Aging, Gateway Building, Suite 3C307, 7201 Wisconsin Avenue, Bethesda, MD, 20892-9205, USA,
| |
Collapse
|
22
|
Toscani D, Bolzoni M, Accardi F, Aversa F, Giuliani N. The osteoblastic niche in the context of multiple myeloma. Ann N Y Acad Sci 2014; 1335:45-62. [DOI: 10.1111/nyas.12578] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Denise Toscani
- Myeloma Unit, Department of Clinical and Experimental Medicine; University of Parma; Parma Italy
| | - Marina Bolzoni
- Myeloma Unit, Department of Clinical and Experimental Medicine; University of Parma; Parma Italy
| | - Fabrizio Accardi
- Myeloma Unit, Department of Clinical and Experimental Medicine; University of Parma; Parma Italy
| | - Franco Aversa
- Myeloma Unit, Department of Clinical and Experimental Medicine; University of Parma; Parma Italy
| | - Nicola Giuliani
- Myeloma Unit, Department of Clinical and Experimental Medicine; University of Parma; Parma Italy
| |
Collapse
|
23
|
Tokuda Y, Tanaka M, Yagi T, Tashiro K. The defect of SFRP2 modulates an influx of extracellular calcium in B lymphocytes. BMC Res Notes 2014; 7:780. [PMID: 25370898 PMCID: PMC4242488 DOI: 10.1186/1756-0500-7-780] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 10/24/2014] [Indexed: 11/13/2022] Open
Abstract
Background In the Wnt pathway, the secreted frizzled-related protein 2 (SFRP2) is thought to act as one of the several competitive inhibitors of Wnt. However, the precise role of SFRP2 is still poorly understood especially in B lymphocytes. Here, we investigated the function of SFRP2, comparing the SFRP2 defective as well as normal B lymphocytes in mice. Results We demonstrated that calcium influx from extracellular to intracellular space in splenic B cells was clearly affected by the defect of SFRP2. In addition, the phosphorylation of phospholipase Cγ2 was observed to be reduced in SFRP2 defective splenic B cells with B cell receptor stimulation. Conclusions SFRP2 is suggested to modulate the influx from extracellular calcium in the B cell receptor signaling pathway. Electronic supplementary material The online version of this article (doi:10.1186/1756-0500-7-780) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | - Kei Tashiro
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, 465 kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan.
| |
Collapse
|
24
|
Wang CQ, Krishnan V, Tay LS, Chin DWL, Koh CP, Chooi JY, Nah GSS, Du L, Jacob B, Yamashita N, Lai SK, Tan TZ, Mori S, Tanuichi I, Tergaonkar V, Ito Y, Osato M. Disruption of Runx1 and Runx3 leads to bone marrow failure and leukemia predisposition due to transcriptional and DNA repair defects. Cell Rep 2014; 8:767-82. [PMID: 25066130 DOI: 10.1016/j.celrep.2014.06.046] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 05/02/2014] [Accepted: 06/23/2014] [Indexed: 11/30/2022] Open
Abstract
The RUNX genes encode transcription factors involved in development and human disease. RUNX1 and RUNX3 are frequently associated with leukemias, yet the basis for their involvement in leukemogenesis is not fully understood. Here, we show that Runx1;Runx3 double-knockout (DKO) mice exhibited lethal phenotypes due to bone marrow failure and myeloproliferative disorder. These contradictory clinical manifestations are reminiscent of human inherited bone marrow failure syndromes such as Fanconi anemia (FA), caused by defective DNA repair. Indeed, Runx1;Runx3 DKO cells showed mitomycin C hypersensitivity, due to impairment of monoubiquitinated-FANCD2 recruitment to DNA damage foci, although FANCD2 monoubiquitination in the FA pathway was unaffected. RUNX1 and RUNX3 interact with FANCD2 independently of CBFβ, suggesting a nontranscriptional role for RUNX in DNA repair. These findings suggest that RUNX dysfunction causes DNA repair defect, besides transcriptional misregulation, and promotes the development of leukemias and other cancers.
Collapse
Affiliation(s)
- Chelsia Qiuxia Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; Institute of Molecular and Cell Biology, A(∗)STAR, Singapore 138673, Singapore
| | - Vaidehi Krishnan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Lavina Sierra Tay
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Desmond Wai Loon Chin
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Cai Ping Koh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Jing Yuan Chooi
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Giselle Sek Suan Nah
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; Institute of Molecular and Cell Biology, A(∗)STAR, Singapore 138673, Singapore
| | - Linsen Du
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Bindya Jacob
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Namiko Yamashita
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Soak Kuan Lai
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Seiichi Mori
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Ichiro Tanuichi
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Vinay Tergaonkar
- Institute of Molecular and Cell Biology, A(∗)STAR, Singapore 138673, Singapore.
| | - Yoshiaki Ito
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; Institute of Molecular and Cell Biology, A(∗)STAR, Singapore 138673, Singapore.
| | - Motomi Osato
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; Institute of Molecular and Cell Biology, A(∗)STAR, Singapore 138673, Singapore; Institute of Bioengineering and Nanotechnology, A(∗)STAR, Singapore 138669, Singapore.
| |
Collapse
|
25
|
Nagel S, Meyer C, Kaufmann M, Drexler HG, MacLeod RAF. Deregulated FOX genes in Hodgkin lymphoma. Genes Chromosomes Cancer 2014; 53:917-33. [PMID: 25043849 DOI: 10.1002/gcc.22204] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 06/18/2014] [Accepted: 06/29/2014] [Indexed: 12/27/2022] Open
Abstract
FOX genes encode transcription factors which regulate basic developmental processes during embryogenesis and in the adult. Several FOX genes show deregulated expression in particular malignancies, representing oncogenes or tumor suppressors. Here, we screened six Hodgkin lymphoma (HL) cell lines for FOX gene activity by comparative microarray profiling, revealing overexpression of FOXC1 and FOXD1, and reduced transcription of FOXN3, FOXO1, and FOXP1. In silico expression analyses of these FOX gene candidates in HL patient samples supported the cell line data. Chromosomal analyses demonstrated an amplification of the FOXC1 locus at 6p25 and a gain of the FOXR2 locus at Xp11, indicting genomic aberrations for their upregulation. Comparative expression profiling and ensuing stimulation experiments revealed implementation of the TGFβ- and WNT-signaling pathways in deregulation of FOXD1 and FOXN3. Functional analysis of FOXP1 implicated miR9 and miR34a as upstream regulators and PAX5, TCF3, and RAG2 as downstream targets. A similar exercise for FOXC1 revealed repression of MSX1 and activation of IPO7, both mediating inhibition of the B-cell specific homeobox gene ZHX2. Taken together, our data show that aberrantly expressed FOX genes and their downstream targets are involved in the pathogenesis of HL via deregulation of B-cell differentiation and may represent useful diagnostic markers and/or therapeutic targets.
Collapse
Affiliation(s)
- Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | | | | | | | | |
Collapse
|
26
|
Vukasovic A, Grbesa D, Nikuseva Martic T, Kusec V, Miskovic B, Serman A, Soken N, Serman L. Glycosylation pattern and axin expression in normal and IUGR placentae. J Matern Fetal Neonatal Med 2014; 28:558-63. [PMID: 24846767 DOI: 10.3109/14767058.2014.926326] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the protein glycosylation pattern and AXIN1 protein expression in human placentae of normal pregnancies and compare them with placentae of pregnancies complicated with intrauterine growth restriction (IUGR). METHODS A total of 38 placentae (17 placentae of IUGR fetuses from singleton pregnancies and gestational age-matched 21 control placentae from normal singleton pregnancies) were collected from the Clinical Hospital Sveti Duh, Department of Gynecology and Obstetrics, Zagreb, Croatia. Gestational age was determined according to the last menstrual period (LMP) and by ultrasound measurements. Expression of glycoproteins was measured by Western blotting with SNA, UEA-I, PHA-E and DBA lectins as probes whereas expression of AXIN1 was determined by immunohistochemistry. RESULTS Comparison of detected sugars revealed differences in protein glycosylation between normal and IUGR placentae. Higher expression of AXIN1 protein located mostly in the cytoplasm of syncytiotrophoblast and to a lesser extent in its nuclei was found in IUGR placentae. CONCLUSION Results of our study suggest that changes in glycoprotein content may contribute to restricted placenta growth and development. Higher expression of AXIN1 protein in IUGR placentae indicates a role of Wnt/β-catenin signaling pathway in pathology of placental development.
Collapse
|
27
|
Kulpa DA, Brehm JH, Fromentin R, Cooper A, Cooper C, Ahlers J, Chomont N, Sékaly RP. The immunological synapse: the gateway to the HIV reservoir. Immunol Rev 2014; 254:305-25. [PMID: 23772628 PMCID: PMC3707302 DOI: 10.1111/imr.12080] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
A major challenge in the development of a cure for human immunodeficiency virus (HIV) has been the incomplete understanding of the basic mechanisms underlying HIV persistence during antiretroviral therapy. It is now realized that the establishment of a latently infected reservoir refractory to immune system recognition has thus far hindered eradication efforts. Recent investigation into the innate immune response has shed light on signaling pathways downstream of the immunological synapse critical for T-cell activation and establishment of T-cell memory. This has led to the understanding that the cell-to-cell contacts observed in an immunological synapse that involve the CD4+ T cell and antigen-presenting cell or T-cell–T-cell interactions enhance efficient viral spread and facilitate the induction and maintenance of latency in HIV-infected memory T cells. This review focuses on recent work characterizing the immunological synapse and the signaling pathways involved in T-cell activation and gene regulation in the context of HIV persistence.
Collapse
Affiliation(s)
- Deanna A Kulpa
- Division of Infectious Diseases, Vaccine and Gene Therapy Institute-Florida (VGTI-FL), Port Saint Lucie, FL 34987, USA
| | | | | | | | | | | | | | | |
Collapse
|