1
|
Zhang Z, He T, Gu H, Zhao Y, Tang S, Han K, Hu Y, Wang H, Yu H. Single-cell RNA sequencing identifies the expression of hemoglobin in chondrocyte cell subpopulations in osteoarthritis. BMC Mol Cell Biol 2024; 25:28. [PMID: 39736555 DOI: 10.1186/s12860-024-00519-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/02/2024] [Indexed: 01/01/2025] Open
Abstract
In recent years, chondrocytes have been found to contain hemoglobin, which might be an alternative strategy for adapting to the hypoxic environment, while the potential mechanisms of that is still unclear. Here, we report the expression characteristics and potential associated pathways of hemoglobin in chondrocytes using single-cell RNA sequencing (scRNA-seq). We downloaded data of normal people and patients with osteoarthritis (OA) from the Gene Expression Omnibus (GEO) database and cells are unbiased clustered based on gene expression pattern. We determined the expression levels of hemoglobin in various chondrocyte subpopulations. Meanwhile, we further explored the difference in the enriched signaling pathways and the cell-cell interaction in chondrocytes of the hemoglobin high-expression and low-expression groups. Specifically, we found that SPP1 was closely associated with the expression of hemoglobin in OA progression. Our findings provide new insights into the distribution characteristics of hemoglobin in chondrocytes and provide potential clues to the underlying role of hemoglobin in OA and the mechanisms related to that, providing potential new ideas for the treatment of OA.
Collapse
Affiliation(s)
- Zhihao Zhang
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Ting He
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hongwen Gu
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Yuanhang Zhao
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Shilei Tang
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Kangen Han
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Yin Hu
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Hongwei Wang
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China.
| | - Hailong Yu
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China.
| |
Collapse
|
2
|
Netti V, Cocca MA, Cutrera N, Molina Ponce T, Ford P, Di Giusto G, Capurro C. Osteopontin Regulates AQP4 Expression by TRPV4 Activation in Müller Cells: Implications for Retinal Homeostasis. Mol Neurobiol 2024:10.1007/s12035-024-04595-6. [PMID: 39485629 DOI: 10.1007/s12035-024-04595-6] [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: 03/25/2024] [Accepted: 10/25/2024] [Indexed: 11/03/2024]
Abstract
During the intense neuronal activity in the retina, Müller cells are exposed to a hypotonic environment and activate a regulatory volume decrease (RVD) response, which depends on Aquaporin-4 (AQP4) and the calcium channel Transient Receptor Potential Vanilloid 4 (TRPV4). It was reported that Osteopontin (OPN), a cytokine and component of the extracellular matrix (ECM), may modulate the RVD of Müller cells. In other cell types, OPN participates in cell survival and migration, which Müller cells undergo to maintain retinal homeostasis. Therefore, the aim of this work was to study the putative crosstalk of OPN with AQP4 and/or TRPV4 in the main functions of Müller cells: RVD, morphology maintenance and migration. We used a human Müller cell line (MIO-M1) exposed to OPN and evaluated cell volume and osmotic permeability (Pf) during an osmotic swelling, AQP4 expression, cell morphology and migration. We observed that OPN induced a reduced Pf and RVD by downregulating AQP4 expression, which was prevented by TRPV4 inhibition. OPN also induced significant changes in cell morphology with an increased number of cytoplasmic projections. Finally, OPN reduced the migration of Müller cells, being this effect dependent on TRPV4. We propose that OPN affects water permeability and cell volume regulation of Müller cells by activating TRPV4 to reduce AQP4 expression. This represents a novel mechanism of regulation of water permeability by the ECM in Müller cells. Additionally, OPN-induced changes in morphology and migration of Müller cells may have an impact on retinal physiology.
Collapse
Affiliation(s)
- Vanina Netti
- Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas, Laboratorio de Biomembranas, Universidad de Buenos Aires, Buenos Aires, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Paraguay 2155, 7Th Floor (1121), Ciudad Autónoma de Buenos Aires, Argentina.
| | - María Azul Cocca
- Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas, Laboratorio de Biomembranas, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Paraguay 2155, 7Th Floor (1121), Ciudad Autónoma de Buenos Aires, Argentina
| | - Nicolás Cutrera
- Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas, Laboratorio de Biomembranas, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Paraguay 2155, 7Th Floor (1121), Ciudad Autónoma de Buenos Aires, Argentina
| | - Tomás Molina Ponce
- Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas, Laboratorio de Biomembranas, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Paraguay 2155, 7Th Floor (1121), Ciudad Autónoma de Buenos Aires, Argentina
| | - Paula Ford
- Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas, Laboratorio de Biomembranas, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Paraguay 2155, 7Th Floor (1121), Ciudad Autónoma de Buenos Aires, Argentina
| | - Gisela Di Giusto
- Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas, Laboratorio de Biomembranas, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Paraguay 2155, 7Th Floor (1121), Ciudad Autónoma de Buenos Aires, Argentina
| | - Claudia Capurro
- Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas, Laboratorio de Biomembranas, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Paraguay 2155, 7Th Floor (1121), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
3
|
Zhang Y, Zhang J, Lesani P, Lu Z, Zreiqat H. Osteopontin Rejuvenates Senescent Adipose-Derived Stem Cells and Restores their Bone Tissue Regenerative Function. Stem Cell Rev Rep 2024; 20:1106-1120. [PMID: 38472643 PMCID: PMC11087332 DOI: 10.1007/s12015-024-10707-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 03/14/2024]
Abstract
The regenerative function of stem cells is compromised when the proportion of senescent stem cells increases with ageing advance. Therefore, combating stem cell senescence is of great importance for stem cell-based tissue engineering in the elderly, but remains largely unexplored. Osteopontin (OPN), a glycosylated phosphoprotein, is one of the key extracellular matrix molecules in bone tissue. OPN activates various signalling pathways and modulates cellular activities, including cell senescence. However, the role of OPN in stem cell senescence remains largely unknown. This study aims to investigate if OPN modulates cell senescence and bone regenerative function in human adipose-derived mesenchymal stem cells (ASCs), and to determine the underlying mechanisms. We first developed a senescent ASC model using serial passaging until passage 10 (P10), in which senescent cells were characterised by reduced proliferation and osteogenic differentiation capacity compared to P4 ASCs. The conditioned medium from P10 ASCs exhibited a diminished trophic effect on human osteoblasts (HOBs), compared to that from P4 ASCs. P10 ASCs on OPN-coated surface showed rejuvenated phenotype and enhanced osteogenic differentiation. The conditioned medium from P10 ASCs on OPN-coating improved trophic effects on HOBs. OPN regulated the morphology of senescent ASCs, transforming them from a more rounded and flattened cell shape to an elongated shape with a smaller area. These findings demonstrated the effects of OPN in restoring senescent ASCs functions, possibly through a mechanism that involves the modulation of cell morphology, indicating that OPN might hold a great potential for rejuvenating senescent stem cells and could potentially open a new venue for regenerating bone tissue in age-related diseases.
Collapse
Affiliation(s)
- Yiran Zhang
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Darlington, NSW, 2006, Australia
| | - Junni Zhang
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Darlington, NSW, 2006, Australia
| | - Pooria Lesani
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Darlington, NSW, 2006, Australia
| | - Zufu Lu
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Darlington, NSW, 2006, Australia.
| | - Hala Zreiqat
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Darlington, NSW, 2006, Australia.
| |
Collapse
|
4
|
HIF-1 directly induces TET3 expression to enhance 5-hmC density and induce erythroid gene expression in hypoxia. Blood Adv 2021; 4:3053-3062. [PMID: 32634239 DOI: 10.1182/bloodadvances.2020001535] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/02/2020] [Indexed: 12/20/2022] Open
Abstract
In mammalian cells, cytosines found within cytosine guanine dinucleotides can be methylated to 5-methylcytosine (5-mC) by DNA methyltransferases and further oxidized by the Ten-eleven translocation dioxygenase (TET) enzymes to 5-hydroxymethylcytosine (5-hmC). We have previously shown that hematopoietic stem and progenitor cells (HSPCs) with TET2 mutations have aberrant 5-hmC distribution and less erythroid differentiation potential. However, these experiments were performed under standard tissue culture conditions with 21% oxygen (O2), whereas HSPCs in human bone marrow reside in ∼1% O2. Therefore, to model human erythropoiesis more accurately, we compared 5-hmC distribution and gene expression in hypoxic vs normoxic conditions. Despite TET enzymes having limited O2 as a substrate in hypoxia, 5-hmC peaks were more numerous and pronounced than in normoxia. Among the TET genes, TET3 was upregulated specifically in hypoxia. We identified 2 HIF-1 binding sites in TET3 by chromatin immunoprecipitation of HIF-1α followed by sequencing, and TET3 upregulation was abrogated with deletion of both sites, indicating that TET3 is a direct HIF-1 target. Finally, we showed that loss of one or both of these HIF-1 binding sites in K562 cells disrupted erythroid differentiation in hypoxia and lowered cell viability. This work provides a molecular link between O2 availability, epigenetic modification of chromatin, and erythroid differentiation.
Collapse
|
5
|
Fu X, Yao M, Ye C, Fang T, Wu R. Osteopontin Regulates Endometrial Stromal Cell Migration in Endometriosis through the PI3K Pathway : Osteopontin Regulates Endometrial Cell Migration in Endometriosis. Reprod Sci 2020; 28:435-446. [PMID: 32909189 PMCID: PMC7808973 DOI: 10.1007/s43032-020-00301-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/17/2020] [Indexed: 01/29/2023]
Abstract
Endometriosis is generally characterized as a tumor-like disease because of its potential for distant metastasis and local tissue invasion, while whether osteopontin (OPN) plays a role in the pathogenesis of endometriosis has not been thoroughly investigated. We investigated the expression of OPN, urokinase plasminogen activator (uPA), phosphatidylinositol 3 kinase (PI3K), and phospho-PI3 kinase (p-PI3K) in endometrial stromal cells (ESCs). The serum concentration of OPN was determined by enzyme-linked immunosorbent assays (ELISA). OPN was downregulated to explore the corresponding change of uPA, p-PI3K, F-actin, and α-tubulin. The expression of OPN, uPA, PI3K, and p-PI3K was evaluated by western blot and quantitative real-time PCR (RT-qPCR) and the expression of F-actin and α-tubulin was confirmed by immunofluorescence assay. The proliferation and migration abilities of ESCs were investigated by CCK8, transwell, and wound scratch assays. Endometrial OPN, p-PI3K, and uPA expressions and serum OPN levels were increased in patients with endometriosis compared with the control. The expressions of p-PI3K, uPA, and α-tubulin were decreased by siRNA-OPN interference in ectopic ESCs. Activation and inhibition of the PI3K pathway apparently upregulate and downregulate uPA expression. Knockdown of OPN and inhibition of the PI3K pathway remarkably inhibited cell migration in ectopic ESCs. Meanwhile, activation of the PI3K pathway promoted the migration ability of ectopic ESCs. OPN may regulate the expression of uPA through the PI3K signal pathway to affect the migration ability of ESCs, indicating that OPN, uPA, and the PI3K pathway may be potential targets for interrupting development of endometriosis.
Collapse
Affiliation(s)
- Xiaoxia Fu
- Department of Obstetrics and Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang Province, People's Republic of China
| | - Mengyun Yao
- Institute of Burn Research, South-West Hospital, State Key Lab of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Third Military Medical University, Chongqing, China
| | - Chaoshuang Ye
- Department of Obstetrics and Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang Province, People's Republic of China
| | - Tao Fang
- Department of Obstetrics and Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang Province, People's Republic of China
| | - Ruijin Wu
- Department of Obstetrics and Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang Province, People's Republic of China.
| |
Collapse
|
6
|
Yamamiya M, Tanabe S, Muramatsu R. Microglia promote the proliferation of neural precursor cells by secreting osteopontin. Biochem Biophys Res Commun 2019; 513:841-845. [PMID: 31003770 DOI: 10.1016/j.bbrc.2019.04.076] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 04/10/2019] [Indexed: 02/07/2023]
Abstract
Microglia are central nervous system-resident immune cells that play a crucial role in brain development by interacting with neural precursor cells (NPCs). It has been reported that microglia regulate the number of NPC by phagocytosis, inducing apoptosis, and promoting proliferation. Microglia surrounding the subventricular zone express osteopontin (OPN) during brain development. The present study investigated the role of microglia in proliferation of NPCs in vitro, and identified the OPN receptor critical for proliferation of NPCs. Microglia co-cultured with NPCs in the presence of an OPN-neutralizing antibody resulted in OPN inhibition and reduced microglia-induced proliferation of NPCs. NPCs express integrin αvβ3, which has been identified as an OPN receptor. Cilengitide, an inhibitor of integrin αvβ3, also inhibited microglia-induced proliferation of NPCs. These results suggest that microglia promote the proliferation of NPCs via OPN-integrin αvβ3 signaling.
Collapse
Affiliation(s)
- Miwako Yamamiya
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Shogo Tanabe
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan.
| | - Rieko Muramatsu
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan.
| |
Collapse
|
7
|
Calvez M, Hseeh G, Benzer S, Brown AM. Osteopontin counters human immunodeficiency virus type 1-induced impairment of neurite growth through mammalian target of rapamycin and beta-integrin signaling pathways. J Neurovirol 2019; 25:384-396. [PMID: 30758811 PMCID: PMC6647884 DOI: 10.1007/s13365-019-00729-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/16/2019] [Accepted: 01/25/2019] [Indexed: 02/07/2023]
Abstract
Despite the fact that human immunodeficiency virus type 1 (HIV-1) does not enter or replicate in neurons, its infection of a subset of resident brain glia cells (microglia and astrocytes) induces via disparate mechanisms, dysregulation of glutamate metabolism, neurotoxicity, and inflammation. Antiretroviral therapies suppress viral load, but cellular activation and release of proinflammatory factors, some of which is likely related to viral reservoirs, continue to promote a microenvironment that is injurious to neurons. However, the molecular mechanisms remain to be identified. Osteopontin (OPN) is a proinflammatory cytokine-like, extracellular matrix protein that is elevated within the brain and CSF in several neurodegenerative disorders, including HIV-associated cognitive disorder. However, the impact of elevated OPN on neuronal integrity and function in HIV-infected individuals who exhibit cognitive dysfunction remains unknown. In this study, using a neuronal cell line and primary cultures of cortical rat neurons, we identify the mammalian target of rapamycin pathway involvement in a signaling interaction between OPN-β1-integrins and the HIV-1 envelope glycoprotein, which stimulates neurite growth. These findings link for the first time HIV X4-envelope receptor engagement and osteopontin-mediated signaling through β1-integrin receptors to the mTOR pathway and alterations in the cytoskeleton of cortical neurons.
Collapse
Affiliation(s)
- Mathilde Calvez
- Department of Biology, Ecole Normale Superieure de Lyon, Lyon, France
| | - George Hseeh
- Department of Neurology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 6-119, Baltimore, MD, 21287, USA
| | - Simon Benzer
- Department of Neurology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 6-119, Baltimore, MD, 21287, USA
| | - Amanda M Brown
- Department of Neurology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 6-119, Baltimore, MD, 21287, USA.
| |
Collapse
|
8
|
Carvalho MS, Cabral JM, da Silva CL, Vashishth D. Synergistic effect of extracellularly supplemented osteopontin and osteocalcin on stem cell proliferation, osteogenic differentiation, and angiogenic properties. J Cell Biochem 2018; 120:6555-6569. [PMID: 30362184 DOI: 10.1002/jcb.27948] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 10/02/2018] [Indexed: 12/17/2022]
Abstract
A high demand for functional bone grafts is being observed worldwide, especially due to the increased life expectancy. Osteoinductive components should be incorporated into functional bone grafts, accelerating cell recruitment, cell proliferation, angiogenesis, and new bone formation at a defect site. Noncollagenous bone matrix proteins, especially osteopontin (OPN) and osteocalcin (OC), have been reported to regulate some physiological process, such as cell migration and bone mineralization. However, the effects of OPN and OC on cell proliferation, osteogenic differentiation, mineralization, and angiogenesis are still undefined. Therefore, we assessed the exogenous effect of OPN and OC supplementation on human bone marrow mesenchymal stem/stromal cells (hBM MSC) proliferation and osteogenic differentiation. OPN dose-dependently increased the proliferation of hBM MSC, as well as improved the angiogenic properties of human umbilical vein endothelial cells (HUVEC) by increasing the capillary-like tube formation in vitro. On the other hand, OC enhanced the differentiation of hBM MSC into osteoblasts and demonstrated an increase in extracellular calcium levels and alkaline phosphatase activity, as well as higher messenger RNA levels of mature osteogenic markers osteopontin and osteocalcin. In vivo assessment of OC/OPN-enhanced scaffolds in a critical-sized defect rabbit long-bone model revealed no infection, while new bone was being formed. Taken together, these results suggest that OC and OPN stimulate bone regeneration by inducing stem cell proliferation, osteogenesis and by enhancing angiogenic properties. The synergistic effect of OC and OPN observed in this study can be applied as an attractive strategy for bone regeneration therapeutics by targeting different vital cellular processes.
Collapse
Affiliation(s)
- Marta S Carvalho
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York.,Department of Bioengineering, iBB - Institute of Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim Ms Cabral
- Department of Bioengineering, iBB - Institute of Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering, iBB - Institute of Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Deepak Vashishth
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
9
|
Macrophage Cytokines Enhance Cell Proliferation of Normal Prostate Epithelial Cells through Activation of ERK and Akt. Sci Rep 2018; 8:7718. [PMID: 29769604 PMCID: PMC5955920 DOI: 10.1038/s41598-018-26143-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 04/10/2018] [Indexed: 12/20/2022] Open
Abstract
Macrophage infiltrations (inflammation) are associated with prostate disorders such as prostatitis, prostatic hyperplasia and prostate cancer. All prostate disorders have elevated cell proliferation, and are initiated from normal prostate epithelial cells. To date, the mechanism of how macrophages regulate normal prostate epithelial cell proliferation remains largely unknown. Using a 3D co-culture system, we here show that Raw 264.7 macrophages increased cell proliferation of normal prostate epithelial PZ-HPV-7 cells. In addition, these Raw 264.7 macrophages expressed higher levels of Ym1 and CD206. We further identify macrophage-secreted cytokines including CCL3, IL-1ra, osteopontin, M-CSF1 and GDNF as mediators for potentiating PZ-HPV-7 cell proliferation in 3D. All these cytokines differentially activated ERK and Akt. Blockade of both kinases through their inhibitors hindered macrophage-induced cell proliferation of PZ-HPV-7 cells. Hence, our data provide mechanistic insight of how inflammation may contribute to development of prostatic diseases at a very early stage through augment of cell proliferation of normal prostate epithelial cells.
Collapse
|
10
|
Zhang J, Yang G, Zhu Y, Peng X, Li T, Liu L. Relationship of Cx43 regulation of vascular permeability to osteopontin-tight junction protein pathway after sepsis in rats. Am J Physiol Regul Integr Comp Physiol 2018; 314:R1-R11. [PMID: 28978514 DOI: 10.1152/ajpregu.00443.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Our previous study demonstrated that connexin (Cx)43 participated in the regulation of vascular permeability in severe sepsis. Osteopontin (OPN) has been demonstrated to participate in the occurrence of atherosclerosis, inflammation, as well as the adhesion and migration of cells. It is not clear whether OPN is involved in Cx43 regulating vascular permeability after sepsis and if it is related to tight-junction proteins. with the use of cecal ligation and puncture (CLP)-induced septic rats and lipopolysaccharide (LPS)-treated pulmonary vein vascular endothelial cells (VECs), the role of zona occuldens 1 (ZO-1) and claudin-5 in Cx43 regulation of vascular permeability and its relationship to OPN were investigated in the present study. The results showed that the expression of ZO-1 and claudin-5 in pulmonary vein were decreased in CLP rats and LPS-treated pulmonary vein VECs. Cx43-overexpressed lentivirus induced the degradation of ZO-1 and claudin-5, while Cx43 RNAi lentivirus abrogated the degradation of ZO-1 and claudin-5 induced by LPS. The vascular permeability and expression of OPN in pulmonary veins were significantly increased in CLP rats and LPS-treated pulmonary vein VECs. Silencing OPN by OPN RNAi lentivirus inhibited the vascular hyperpermeability induced by LPS. Overexpressed Cx43 lentivirus increased the expression of OPN and vascular permeability and downregulated the expression of ZO-1 and claudin-5 in pulmonary vein VECs. Silencing OPN by OPN RNAi lentivirus inhibited the effects of Cx43-overexpressed lentivirus on downregulation of ZO-1 and claudin-5 and vascular hyperpermeability in pulmonary vein VECs. Transfection of specific double-stranded RNA targeting to β-catenin and T-cell factor-4 (Tcf-4) abolished the upregulation of OPN induced by Cx43 overexpression. These results suggest that OPN participates in the regulation of vascular permeability by Cx43 after sepsis. Cx43 upregulation of OPN is via the Tcf-4/β-catenin transcription pathway; OPN increases vascular permeability by downregulating the expression of the tight junction proteins ZO-1 and claudin-5.
Collapse
Affiliation(s)
- Jie Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Guangming Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Yu Zhu
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Xiaoyong Peng
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Liangming Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
11
|
Zhu X, Hu T, Ho MH, Wang Y, Yu M, Patel N, Pi W, Choi JH, Xu H, Ganapathy V, Kutlar F, Kutlar A, Tuan D. Hydroxyurea differentially modulates activator and repressors of γ-globin gene in erythroblasts of responsive and non-responsive patients with sickle cell disease in correlation with Index of Hydroxyurea Responsiveness. Haematologica 2017; 102:1995-2004. [PMID: 28971909 PMCID: PMC5709098 DOI: 10.3324/haematol.2017.175646] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/29/2017] [Indexed: 01/24/2023] Open
Abstract
Hydroxyurea (HU), the first of two drugs approved by the US Food and Drug Administration for treating patients with sickle cell disease (SCD), produces anti-sickling effect by re-activating fetal γ-globin gene to enhance production of fetal hemoglobin. However, approximately 30% of the patients do not respond to HU therapy. The molecular basis of non-responsiveness to HU is not clearly understood. To address this question, we examined HU-induced changes in the RNA and protein levels of transcription factors NF-Y, GATA-1, -2, BCL11A, TR4, MYB and NF-E4 that assemble the γ-globin promoter complex and regulate transcription of γ-globin gene. In erythroblasts cultured from peripheral blood CD34+ cells of patients with SCD, we found that HU-induced changes in the protein but not the RNA levels of activator GATA-2 and repressors GATA-1, BCL11A and TR4 correlated with HU-induced changes in fetal hemoglobin (HbF) levels in the peripheral blood of HU high and low responders. However, HU did not significantly induce changes in the protein or RNA levels of activators NF-Y and NF-E4. Based on HU-induced changes in the protein levels of GATA-2, -1 and BCL11A, we calculated an Index of Hydroxyurea Responsiveness (IndexHU-3). Compared to the HU-induced fold changes in the individual transcription factor protein levels, the numerical values of IndexHU-3 statistically correlated best with the HU-induced peripheral blood HbF levels of the patients. Thus, IndexHU-3 can serve as an appropriate indicator for inherent HU responsiveness of patients with SCD.
Collapse
Affiliation(s)
- Xingguo Zhu
- Department of Biochemistry and Molecular Biology, Augusta University, GA, USA
| | - Tianxiang Hu
- Department of Biochemistry and Molecular Biology, Augusta University, GA, USA
| | - Meng Hsuan Ho
- Department of Biochemistry and Molecular Biology, Augusta University, GA, USA.,School of Dentistry, Meharry Medical College, Nashville, TN, USA
| | - Yongchao Wang
- Department of Biochemistry and Molecular Biology, Augusta University, GA, USA.,Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Miao Yu
- Georgia Cancer Research Center, Augusta University, GA, USA
| | - Niren Patel
- Division of Hematology/Oncology, Augusta University, GA, USA
| | - Wenhu Pi
- Department of Biochemistry and Molecular Biology, Augusta University, GA, USA.,Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jeong-Hyeon Choi
- Georgia Cancer Research Center, Augusta University, GA, USA.,Department of Biostatistics, Augusta University, GA, USA
| | - Hongyan Xu
- Department of Biostatistics, Augusta University, GA, USA
| | - Vadivel Ganapathy
- Department of Biochemistry and Molecular Biology, Augusta University, GA, USA.,Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Ferdane Kutlar
- Division of Hematology/Oncology, Augusta University, GA, USA
| | - Abdullah Kutlar
- Division of Hematology/Oncology, Augusta University, GA, USA
| | - Dorothy Tuan
- Department of Biochemistry and Molecular Biology, Augusta University, GA, USA
| |
Collapse
|
12
|
Herdl S, Huebner H, Volkert G, Marek I, Menendez-Castro C, Noegel SC, Ruebner M, Rascher W, Hartner A, Fahlbusch FB. Integrin α8 Is Abundant in Human, Rat, and Mouse Trophoblasts. Reprod Sci 2017; 24:1426-1437. [PMID: 28136130 DOI: 10.1177/1933719116689597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Integrins exert regulatory functions in placentogenesis. Null mutation of certain integrin α subunits leads to placental defects with subsequent fetal growth restriction or embryonic lethality in mice. So far, the placental role of α8 integrin remains to be determined. METHODS Localization of α8 integrin and its ligands, fibronectin (FN) and osteopontin (OPN), was studied by immunohistochemistry in human, rat, and mouse placenta. The vascularization of the placental labyrinth layer of α8 integrin-deficient mice was determined by CD31 staining. In humans, α8 integrin expression was assessed via real-time polymerase chain reaction in healthy placentas, in the placental pathologies such as intrauterine growth restriction (IUGR), preeclampsia, and HELLP-syndrome (hemolysis, elevated liver enzymes, low platelet count), as well as in primary extravillous trophoblasts (EVT) and villous trophoblasts. RESULTS In humans, α8 integrin was detected in first and third trimester syncytiotrophoblast and EVT. Although OPN showed the same localization, FN was observed in EVT only. No expressional changes in α8 integrin were detected in the placental pathologies studied. Rodent placenta showed α8 integrin expression in giant cells and in the labyrinth layer. The localization of OPN and FN, however, showed species-specific differences. Knockout of α8 integrin in mice did not cause IUGR, despite some reduction in labyrinth layer vascularization. CONCLUSION α8 Integrin is expressed in functional placental compartments among its ligands, OPN and/or FN, across species. Although this may point to a regulatory role in trophoblast function, our data from α8 integrin-deficient mice indicated only mild placental pathology. Thus, the lack of placental α8 integrin seems to be largely compensated for.
Collapse
Affiliation(s)
- Sebastian Herdl
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Hanna Huebner
- 2 Department of Gynaecology and Obstetrics, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Gudrun Volkert
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Ines Marek
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Carlos Menendez-Castro
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Stephanie C Noegel
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Ruebner
- 2 Department of Gynaecology and Obstetrics, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Wolfgang Rascher
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Andrea Hartner
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Fabian B Fahlbusch
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
13
|
Noulin F. Malaria modeling: In vitro stem cells vs in vivo models. World J Stem Cells 2016; 8:88-100. [PMID: 27022439 PMCID: PMC4807312 DOI: 10.4252/wjsc.v8.i3.88] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 12/07/2015] [Accepted: 01/29/2016] [Indexed: 02/06/2023] Open
Abstract
The recent development of stem cell research and the possibility of generating cells that can be stably and permanently modified in their genome open a broad horizon in the world of in vitro modeling. The malaria field is gaining new opportunities from this important breakthrough and novel tools were adapted and opened new frontiers for malaria research. In addition to the new in vitro systems, in recent years there were also significant advances in the development of new animal models that allows studying the entire cell cycle of human malaria. In this paper, we review the different protocols available to study human Plasmodium species either by using stem cell or alternative animal models.
Collapse
|
14
|
Rosario GX, Stewart CL. The Multifaceted Actions of Leukaemia Inhibitory Factor in Mediating Uterine Receptivity and Embryo Implantation. Am J Reprod Immunol 2016; 75:246-55. [PMID: 26817565 DOI: 10.1111/aji.12474] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 12/07/2015] [Indexed: 01/08/2023] Open
Abstract
Embryo implantation is mediated by the combined actions of the ovarian hormones E2 and P4 on the uterus. In addition, the pro-inflammatory cytokine, leukaemia inhibitory factor (LIF), plays a pivotal role in regulating uterine receptivity. LIF is expressed in the endometrial glands and has a robust action on the uterine luminal epithelium (LE). In mice, LIF is induced by nidatory E2 and functions to convert the LE from a non-receptive to an embryo-responsive state. LIF mediates its actions by activating the JAK-STAT pathway specifically in the LE. Activation of JAK-STAT pathway results in the induction of many additional pathways, including some 40 + transcription factors, many of which initiate a cascade of changes affecting epithelial polarity, epithelial-mesenchymal interactions, angiogenesis, stromal cell decidualization, and inhibiting cell proliferation. This review discusses the role of LIF and the recent analysis of its action on the uterine LE in regulating endometrial receptivity and implantation.
Collapse
Affiliation(s)
- Gracy Xavier Rosario
- Developmental and Regenerative Biology, Institute of Medical Biology, Singapore City, Singapore
| | - Colin L Stewart
- Developmental and Regenerative Biology, Institute of Medical Biology, Singapore City, Singapore
| |
Collapse
|
15
|
Reduced DOCK4 expression leads to erythroid dysplasia in myelodysplastic syndromes. Proc Natl Acad Sci U S A 2015; 112:E6359-68. [PMID: 26578796 DOI: 10.1073/pnas.1516394112] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Anemia is the predominant clinical manifestation of myelodysplastic syndromes (MDS). Loss or deletion of chromosome 7 is commonly seen in MDS and leads to a poor prognosis. However, the identity of functionally relevant, dysplasia-causing, genes on 7q remains unclear. Dedicator of cytokinesis 4 (DOCK4) is a GTPase exchange factor, and its gene maps to the commonly deleted 7q region. We demonstrate that DOCK4 is underexpressed in MDS bone marrow samples and that the reduced expression is associated with decreased overall survival in patients. We show that depletion of DOCK4 levels leads to erythroid cells with dysplastic morphology both in vivo and in vitro. We established a novel single-cell assay to quantify disrupted F-actin filament network in erythroblasts and demonstrate that reduced expression of DOCK4 leads to disruption of the actin filaments, resulting in erythroid dysplasia that phenocopies the red blood cell (RBC) defects seen in samples from MDS patients. Reexpression of DOCK4 in -7q MDS patient erythroblasts resulted in significant erythropoietic improvements. Mechanisms underlying F-actin disruption revealed that DOCK4 knockdown reduces ras-related C3 botulinum toxin substrate 1 (RAC1) GTPase activation, leading to increased phosphorylation of the actin-stabilizing protein ADDUCIN in MDS samples. These data identify DOCK4 as a putative 7q gene whose reduced expression can lead to erythroid dysplasia.
Collapse
|
16
|
Dorn DC, Dorn A. Stem cell autotomy and niche interaction in different systems. World J Stem Cells 2015; 7:922-944. [PMID: 26240680 PMCID: PMC4515436 DOI: 10.4252/wjsc.v7.i6.922] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 05/27/2015] [Indexed: 02/06/2023] Open
Abstract
The best known cases of cell autotomy are the formation of erythrocytes and thrombocytes (platelets) from progenitor cells that reside in special niches. Recently, autotomy of stem cells and its enigmatic interaction with the niche has been reported from male germline stem cells (GSCs) in several insect species. First described in lepidopterans, the silkmoth, followed by the gipsy moth and consecutively in hemipterans, foremost the milkweed bug. In both, moths and the milkweed bug, GSCs form finger-like projections toward the niche, the apical cells (homologs of the hub cells in Drosophila). Whereas in the milkweed bug the projection terminals remain at the surface of the niche cells, in the gipsy moth they protrude deeply into the singular niche cell. In both cases, the projections undergo serial retrograde fragmentation with progressing signs of autophagy. In the gipsy moth, the autotomized vesicles are phagocytized and digested by the niche cell. In the milkweed bug the autotomized vesicles accumulate at the niche surface and disintegrate. Autotomy and sprouting of new projections appears to occur continuously. The significance of the GSC-niche interactions, however, remains enigmatic. Our concept on the signaling relationship between stem cell-niche in general and GSC and niche (hub cells and cyst stem cells) in particular has been greatly shaped by Drosophila melanogaster. In comparing the interactions of GSCs with their niche in Drosophila with those in species exhibiting GSC autotomy it is obvious that additional or alternative modes of stem cell-niche communication exist. Thus, essential signaling pathways, including niche-stem cell adhesion (E-cadherin) and the direction of asymmetrical GSC division - as they were found in Drosophila - can hardly be translated into the systems where GSC autotomy was reported. It is shown here that the serial autotomy of GSC projections shows remarkable similarities with Wallerian axonal destruction, developmental axon pruning and dying-back degeneration in neurodegenerative diseases. Especially the hypothesis of an existing evolutionary conserved “autodestruction program” in axons that might also be active in GSC projections appears attractive. Investigations on the underlying signaling pathways have to be carried out. There are two other well known cases of programmed cell autotomy: the enucleation of erythroblasts in the process of erythrocyte maturation and the segregation of thousands of thrombocytes (platelets) from one megakaryocyte. Both progenitor cell types - erythroblasts and megakaryocytes - are associated with a niche in the bone marrow, erythroblasts with a macrophage, which they surround, and the megakaryocytes with the endothelial cells of sinusoids and their extracellular matrix. Although the regulatory mechanisms may be specific in each case, there is one aspect that connects all described processes of programmed cell autotomy and neuronal autodestruction: apoptotic pathways play always a prominent role. Studies on the role of male GSC autotomy in stem cell-niche interaction have just started but are expected to reveal hitherto unknown ways of signal exchange. Spermatogenesis in mammals advance our understanding of insect spermatogenesis. Mammal and insect spermatogenesis share some broad principles, but a comparison of the signaling pathways is difficult. We have intimate knowledge from Drosophila, but of almost no other insect, and we have only limited knowledge from mammals. The discovery of stem cell autotomy as part of the interaction with the niche promises new general insights into the complicated stem cell-niche interdependence.
Collapse
|
17
|
Rabenstein M, Hucklenbroich J, Willuweit A, Ladwig A, Fink GR, Schroeter M, Langen KJ, Rueger MA. Osteopontin mediates survival, proliferation and migration of neural stem cells through the chemokine receptor CXCR4. Stem Cell Res Ther 2015; 6:99. [PMID: 25998490 PMCID: PMC4464234 DOI: 10.1186/s13287-015-0098-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 04/23/2015] [Accepted: 05/13/2015] [Indexed: 12/13/2022] Open
Abstract
Introduction Osteopontin (OPN) is a phosphoglycoprotein with important roles in tissue homeostasis, wound healing, immune regulation, and stress responses. It is expressed constitutively in the brain and upregulated during neuroinflammatory responses; for example, after focal cerebral ischemia. To date, its effects on neural stem cells (NSC) remain to be elucidated and are, accordingly, the subject of this study. Method Primary fetal rat NSC were cultured as homogenous monolayers and treated with different concentrations of OPN. Fundamental properties of NSC were assessed following OPN exposure, including proliferative activity, survival under oxidative stress, migration, and differentiation potential. To elucidate a putative action of OPN via the CXC chemokine receptor type 4 (CXCR4), the latter was blocked with AMD3100. To investigate effects of OPN on endogenous NSC in vivo, recombinant OPN was injected into the brain of healthy adult rats as well as rats subjected to focal cerebral ischemia. Effects of OPN on NSC proliferation and neurogenesis in the subventricular zone were studied immunohistochemically. Results OPN dose-dependently increased the number of NSC in vitro. As hypothesized, this effect was mediated through CXCR4. The increase in NSC number was due to both enhanced cell proliferation and increased survival, and was confirmed in vivo. Additionally, OPN dose-dependently stimulated the migration of NSC via CXCR4. Moreover, in the presence of OPN, differentiation of NSC led to a significant increase in neurogenesis both in vitro as well as in vivo after cerebral ischemia. Conclusion Data show positive effects of OPN on survival, proliferation, migration, and neuronal differentiation of NSC. At least in part these effects were mediated via CXCR4. Results suggest that OPN is a promising substance for the targeted activation of NSC in future experimental therapies for neurological disorders such as stroke. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0098-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Monika Rabenstein
- Department of Neurology, University Hospital of Cologne, Cologne, Germany.
| | - Joerg Hucklenbroich
- Department of Neurology, University Hospital of Cologne, Cologne, Germany. .,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Leo-Brandt-Straße, 52425, Juelich, Germany.
| | - Antje Willuweit
- Medical Imaging Physics, Institute of Neuroscience and Medicine (INM-4), Research Centre Juelich, Juelich, Germany.
| | - Anne Ladwig
- Department of Neurology, University Hospital of Cologne, Cologne, Germany.
| | - Gereon Rudolf Fink
- Department of Neurology, University Hospital of Cologne, Cologne, Germany. .,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Leo-Brandt-Straße, 52425, Juelich, Germany.
| | - Michael Schroeter
- Department of Neurology, University Hospital of Cologne, Cologne, Germany. .,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Leo-Brandt-Straße, 52425, Juelich, Germany.
| | - Karl-Josef Langen
- Medical Imaging Physics, Institute of Neuroscience and Medicine (INM-4), Research Centre Juelich, Juelich, Germany.
| | - Maria Adele Rueger
- Department of Neurology, University Hospital of Cologne, Cologne, Germany. .,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Leo-Brandt-Straße, 52425, Juelich, Germany.
| |
Collapse
|
18
|
Risso A, Ciana A, Achilli C, Antonutto G, Minetti G. Neocytolysis: none, one or many? A reappraisal and future perspectives. Front Physiol 2014; 5:54. [PMID: 24592241 PMCID: PMC3924315 DOI: 10.3389/fphys.2014.00054] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/28/2014] [Indexed: 12/11/2022] Open
Abstract
Neocytolysis is the hypothesis formulated to explain experimental evidence of selective lysis of young red blood cells (RBCs) (neocytes) associated with decreased plasma levels of erythropoietin (EPO). In humans, it appears to take place whenever a fast RBC mass reduction is required, i.e., in astronauts during the first days of spaceflight under weightlessness, where a fast reduction in plasma volume and increase in haematocrit occur. EPO plasma levels then decline and a decrease in RBC mass takes place, apparently because of the selective lysis of the youngest, recently generated RBCs (neocytes). The same process seems to occur in people descending to sea level after acclimatization at high altitude. After descent, the polycythaemia developed at high altitude must be abrogated, and a rapid reduction in the number of circulating RBCs is obtained by a decrease in EPO synthesis and the lysis of what seem to be young RBCs. In vivo, neocytolysis seems to be abolished by EPO administration. More recent research has ascribed to neocytolysis the RBC destruction that occurs under such disparate pathophysiologic conditions as nephropathy, severe obstructive pulmonary disease, blood doping, and even malaria anaemia. According to the theory, EPO's central role would be not only to stimulate the production of new RBCs in conditions of anaemia, as maintained by the orthodox view, but also that of a cytoprotective factor for circulating young RBCs. Why neocytes are specifically destroyed and how is this related to decreased EPO levels has not yet been elucidated. Changes in membrane molecules of young RBCs isolated from astronauts or mountain climbers upon return to normal conditions seem to indicate a higher susceptibility of neocytes to ingestion by macrophages. By limiting the context to space missions and high altitude expeditions, this review will address unresolved and critical issues that in our opinion have not been sufficiently highlighted in previous works.
Collapse
Affiliation(s)
- Angela Risso
- Department of Agricultural and Environmental Sciences, University of Udine Udine, Italy
| | - Annarita Ciana
- Laboratories of Biochemistry, Department of Biology and Biotechnology, University of Pavia Pavia, Italy
| | - Cesare Achilli
- Laboratories of Biochemistry, Department of Biology and Biotechnology, University of Pavia Pavia, Italy
| | - Guglielmo Antonutto
- Department of Medical and Biological Sciences, University of Udine Udine, Italy
| | - Giampaolo Minetti
- Laboratories of Biochemistry, Department of Biology and Biotechnology, University of Pavia Pavia, Italy
| |
Collapse
|
19
|
Sui Z, Nowak RB, Bacconi A, Kim NE, Liu H, Li J, Wickrema A, An XL, Fowler VM. Tropomodulin3-null mice are embryonic lethal with anemia due to impaired erythroid terminal differentiation in the fetal liver. Blood 2014; 123:758-67. [PMID: 24159174 PMCID: PMC3907761 DOI: 10.1182/blood-2013-03-492710] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 10/11/2013] [Indexed: 11/20/2022] Open
Abstract
Tropomodulin (Tmod) is a protein that binds and caps the pointed ends of actin filaments in erythroid and nonerythoid cell types. Targeted deletion of mouse tropomodulin3 (Tmod3) leads to embryonic lethality at E14.5-E18.5, with anemia due to defects in definitive erythropoiesis in the fetal liver. Erythroid burst-forming unit and colony-forming unit numbers are greatly reduced, indicating defects in progenitor populations. Flow cytometry of fetal liver erythroblasts shows that late-stage populations are also decreased, including reduced percentages of enucleated cells. Annexin V staining indicates increased apoptosis of Tmod3(-/-) erythroblasts, and cell-cycle analysis reveals that there are more Ter119(hi) cells in S-phase in Tmod3(-/-) embryos. Notably, enucleating Tmod3(-/-) erythroblasts are still in the process of proliferation, suggesting impaired cell-cycle exit during terminal differentiation. Tmod3(-/-) late erythroblasts often exhibit multilobular nuclear morphologies and aberrant F-actin assembly during enucleation. Furthermore, native erythroblastic island formation was impaired in Tmod3(-/-) fetal livers, with Tmod3 required in both erythroblasts and macrophages. In conclusion, disruption of Tmod3 leads to impaired definitive erythropoiesis due to reduced progenitors, impaired erythroblastic island formation, and defective erythroblast cell-cycle progression and enucleation. Tmod3-mediated actin remodeling may be required for erythroblast-macrophage adhesion, coordination of cell cycle with differentiation, and F-actin assembly and remodeling during erythroblast enucleation.
Collapse
Affiliation(s)
- Zhenhua Sui
- The Scripps Research Institute, La Jolla, CA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Madzo J, Liu H, Rodriguez A, Vasanthakumar A, Sundaravel S, Caces DBD, Looney TJ, Zhang L, Lepore JB, Macrae T, Duszynski R, Shih AH, Song CX, Yu M, Yu Y, Grossman R, Raumann B, Verma A, He C, Levine RL, Lavelle D, Lahn BT, Wickrema A, Godley LA. Hydroxymethylation at gene regulatory regions directs stem/early progenitor cell commitment during erythropoiesis. Cell Rep 2013; 6:231-244. [PMID: 24373966 DOI: 10.1016/j.celrep.2013.11.044] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 08/21/2013] [Accepted: 11/26/2013] [Indexed: 01/28/2023] Open
Abstract
Hematopoietic stem cell differentiation involves the silencing of self-renewal genes and induction of a specific transcriptional program. Identification of multiple covalent cytosine modifications raises the question of how these derivatized bases influence stem cell commitment. Using a replicative primary human hematopoietic stem/progenitor cell differentiation system, we demonstrate dynamic changes of 5-hydroxymethylcytosine (5-hmC) during stem cell commitment and differentiation to the erythroid lineage. Genomic loci that maintain or gain 5-hmC density throughout erythroid differentiation contain binding sites for erythroid transcription factors and several factors not previously recognized as erythroid-specific factors. The functional importance of 5-hmC was demonstrated by impaired erythroid differentiation, with augmentation of myeloid potential, and disrupted 5-hmC patterning in leukemia patient-derived CD34+ stem/early progenitor cells with TET methylcytosine dioxygenase 2 (TET2) mutations. Thus, chemical conjugation and affinity purification of 5-hmC-enriched sequences followed by sequencing serve as resources for deciphering functional implications for gene expression during stem cell commitment and differentiation along a particular lineage.
Collapse
Affiliation(s)
- Jozef Madzo
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Hui Liu
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Alexis Rodriguez
- Center for Research Informatics, The University of Chicago, Chicago, IL 60637, USA
| | - Aparna Vasanthakumar
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Sriram Sundaravel
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Donne Bennett D Caces
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Timothy J Looney
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA
| | - Li Zhang
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA
| | - Janet B Lepore
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Trisha Macrae
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Robert Duszynski
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Alan H Shih
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Chun-Xiao Song
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Miao Yu
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Yiting Yu
- Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Robert Grossman
- Center for Research Informatics, The University of Chicago, Chicago, IL 60637, USA
| | - Brigitte Raumann
- Center for Research Informatics, The University of Chicago, Chicago, IL 60637, USA
| | - Amit Verma
- Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Chuan He
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Ross L Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Don Lavelle
- Department of Medicine, University of Illinois, Chicago, Chicago, IL 60612, USA
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Bruce T Lahn
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA
| | - Amittha Wickrema
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Lucy A Godley
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
21
|
Mehrotra S, Sharma B, Joshi S, Kroczynska B, Majchrzak B, Stein BL, McMahon B, Altman JK, Licht JD, Baker DP, Eklund EA, Wickrema A, Verma A, Fish EN, Platanias LC. Essential role for the Mnk pathway in the inhibitory effects of type I interferons on myeloproliferative neoplasm (MPN) precursors. J Biol Chem 2013; 288:23814-22. [PMID: 23814052 DOI: 10.1074/jbc.m113.476192] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The mechanisms of generation of the antineoplastic effects of interferons (IFNs) in malignant hematopoietic cells remain to be precisely defined. We examined the activation of type I IFN-dependent signaling pathways in malignant cells transformed by Jak2V617F, a critical pathogenic mutation in myeloproliferative neoplasms (MPNs). Our studies demonstrate that during engagement of the type I IFN receptor (IFNAR), there is activation of Jak-Stat pathways and also engagement of Mnk kinases. Activation of Mnk kinases is regulated by the Mek/Erk pathway and is required for the generation of IFN-induced growth inhibitory responses, but Mnk kinase activation does not modulate IFN-regulated Jak-Stat signals. We demonstrate that for type I IFNs to exert suppressive effects in malignant hematopoietic progenitors from patients with polycythemia vera, induction of Mnk kinase activity is required, as evidenced by studies involving pharmacological inhibition of Mnk or siRNA-mediated Mnk knockdown. Altogether, these findings provide evidence for key and essential roles of the Mnk kinase pathway in the generation of the antineoplastic effects of type I IFNs in Jak2V617F-dependent MPNs.
Collapse
Affiliation(s)
- Swarna Mehrotra
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Schordan S, Grisk O, Schordan E, Miehe B, Rumpel E, Endlich K, Giebel J, Endlich N. OPN deficiency results in severe glomerulosclerosis in uninephrectomized mice. Am J Physiol Renal Physiol 2013; 304:F1458-70. [PMID: 23552865 DOI: 10.1152/ajprenal.00615.2012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Osteopontin (OPN) expression has been reported to be elevated in experimental models of renal injury such as arterial hypertension or diabetic nephropathy finally leading to focal segmental glomerulosclerosis (FSGS). FSGS is characterized by glomerular matrix deposition and loss or damage of podocytes that represent the main constituents of the glomerular filtration barrier. To evaluate the role of OPN in the kidney we investigated WT and OPN knockout mice (OPN-/-) without treatment, after uninephrectomy (UNX), as well as after UNX and desoxycorticosterone acetate (DOCA)-salt treatment with respect to urine parameters, glomerular morphology, and expression of podocyte markers. OPN-/- mice showed normal urine parameters while a thickening of the glomerular basement membrane was evident. Intriguingly, following UNX, OPN-/- mice exhibited prominent FSGS, proteinuria, and glomerular matrix deposition. Electron microscopy revealed bulgings of the glomerular basement membrane and occasionally an effacement of podocytes. After UNX and DOCA-salt treatment, severe glomerular lesions as well as proteinuria and albuminuria were seen in WT and OPN-/- mice. Moreover, we found a reduction of specific markers such as Wilm's tumor-1, podocin, and synaptopodin in both experimental groups indicating a loss of podocytes. Podocyte damage was accompanied by increased number of Ki-67-positive cells in the parietal epithelium of Bowman's capsule. We conclude that OPN plays a crucial role in adaptation of podocytes following renal ablation and is renoprotective when glomerular mechanical load is increased.
Collapse
Affiliation(s)
- Sandra Schordan
- Department of Anatomy and Cell Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Yu Y, Mo Y, Ebenezer D, Bhattacharyya S, Liu H, Sundaravel S, Giricz O, Wontakal S, Cartier J, Caces B, Artz A, Nischal S, Bhagat T, Bathon K, Maqbool S, Gligich O, Suzuki M, Steidl U, Godley L, Skoultchi A, Greally J, Wickrema A, Verma A. High resolution methylome analysis reveals widespread functional hypomethylation during adult human erythropoiesis. J Biol Chem 2013; 288:8805-14. [PMID: 23306203 DOI: 10.1074/jbc.m112.423756] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Differentiation of hematopoietic stem cells to red cells requires coordinated expression of numerous erythroid genes and is characterized by nuclear condensation and extrusion during terminal development. To understand the regulatory mechanisms governing these widespread phenotypic changes, we conducted a high resolution methylomic and transcriptomic analysis of six major stages of human erythroid differentiation. We observed widespread epigenetic differences between early and late stages of erythropoiesis with progressive loss of methylation being the dominant change during differentiation. Gene bodies, intergenic regions, and CpG shores were preferentially demethylated during erythropoiesis. Epigenetic changes at transcription factor binding sites correlated significantly with changes in gene expression and were enriched for binding motifs for SCL, MYB, GATA, and other factors not previously implicated in erythropoiesis. Demethylation at gene promoters was associated with increased expression of genes, whereas epigenetic changes at gene bodies correlated inversely with gene expression. Important gene networks encoding erythrocyte membrane proteins, surface receptors, and heme synthesis proteins were found to be regulated by DNA methylation. Furthermore, integrative analysis enabled us to identify novel, potential regulatory areas of the genome as evident by epigenetic changes in a predicted PU.1 binding site in intron 1 of the GATA1 gene. This intronic site was found to be conserved across species and was validated to be a novel PU.1 binding site by quantitative ChIP in erythroid cells. Altogether, our study provides a comprehensive analysis of methylomic and transcriptomic changes during erythroid differentiation and demonstrates that human terminal erythropoiesis is surprisingly associated with hypomethylation of the genome.
Collapse
Affiliation(s)
- Yiting Yu
- Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Zhu X, Wang Y, Pi W, Liu H, Wickrema A, Tuan D. NF-Y recruits both transcription activator and repressor to modulate tissue- and developmental stage-specific expression of human γ-globin gene. PLoS One 2012; 7:e47175. [PMID: 23071749 PMCID: PMC3468502 DOI: 10.1371/journal.pone.0047175] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 09/10/2012] [Indexed: 11/19/2022] Open
Abstract
The human embryonic, fetal and adult β-like globin genes provide a paradigm for tissue- and developmental stage-specific gene regulation. The fetal γ-globin gene is expressed in fetal erythroid cells but is repressed in adult erythroid cells. The molecular mechanism underlying this transcriptional switch during erythroid development is not completely understood. Here, we used a combination of in vitro and in vivo assays to dissect the molecular assemblies of the active and the repressed proximal γ-globin promoter complexes in K562 human erythroleukemia cell line and primary human fetal and adult erythroid cells. We found that the proximal γ-globin promoter complex is assembled by a developmentally regulated, general transcription activator NF-Y bound strongly at the tandem CCAAT motifs near the TATA box. NF-Y recruits to neighboring DNA motifs the developmentally regulated, erythroid transcription activator GATA-2 and general repressor BCL11A, which in turn recruit erythroid repressor GATA-1 and general repressor COUP-TFII to form respectively the NF-Y/GATA-2 transcription activator hub and the BCL11A/COUP-TFII/GATA-1 transcription repressor hub. Both the activator and the repressor hubs are present in both the active and the repressed γ-globin promoter complexes in fetal and adult erythroid cells. Through changes in their levels and respective interactions with the co-activators and co-repressors during erythroid development, the activator and the repressor hubs modulate erythroid- and developmental stage-specific transcription of γ-globin gene.
Collapse
Affiliation(s)
- Xingguo Zhu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia and College of Graduate Studies, Georgia Health Sciences University, Augusta, Georgia, United States of America
| | - Yongchao Wang
- Department of Biochemistry and Molecular Biology, Medical College of Georgia and College of Graduate Studies, Georgia Health Sciences University, Augusta, Georgia, United States of America
| | - Wenhu Pi
- Department of Biochemistry and Molecular Biology, Medical College of Georgia and College of Graduate Studies, Georgia Health Sciences University, Augusta, Georgia, United States of America
| | - Hui Liu
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Amittha Wickrema
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Dorothy Tuan
- Department of Biochemistry and Molecular Biology, Medical College of Georgia and College of Graduate Studies, Georgia Health Sciences University, Augusta, Georgia, United States of America
| |
Collapse
|
25
|
Flatt JF, Musa RH, Ayob Y, Hassan A, Asidin N, Yahya NM, Mathlouthi R, Thornton N, Anstee DJ, Bruce LJ. Study of the D-- phenotype reveals erythrocyte membrane alterations in the absence of RHCE. Br J Haematol 2012; 158:262-273. [PMID: 22571328 DOI: 10.1111/j.1365-2141.2012.09149.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 03/27/2012] [Indexed: 02/04/2023]
Abstract
Red cells with the D-- phenotype do not express the RHCE protein because of mutations in both alleles of the RHCE gene. At present, little is known of the effect this has on the normal function of erythrocytes. In this study a group of five families belonging to a nomadic tribe in Malaysia were identified as carriers of the D-- haplotype. Analysis of homozygous individuals' genomic DNA showed two separate novel mutations. In four of the families, RHCE exons 1, 9 and 10 were present, while the 5th family possessed RHCE exons 1-3 and 10. Analysis of cDNA revealed hybrid transcripts, suggesting a gene conversion event with RHD, consistent with previously reported D-- mutations. Immunoblotting analysis of D-- erythrocyte membrane proteins found that Rh-associated glycoprotein (RHAG) migrates with altered electrophoretic mobility on sodium dodecyl sulphate polyacrylamide gel electrophoresis, consistent with increased glycosylation. Total amounts of Rh polypeptide in D-- membranes were comparable with controls, indicating that the exalted D antigen displayed by D-- red cells may be associated with altered surface epitope presentation. The adhesion molecules CD44 and CD47 are significantly reduced in D--. Together these results suggest that absence of RHCE polypeptide alters the structure and packing of the band 3/Rh macrocomplex.
Collapse
Affiliation(s)
- Joanna F Flatt
- Bristol Institute for Transfusion Sciences, N.H.S. Blood and Transplant, Filton, Bristol, UK
| | - Rozi H Musa
- Immunohaematology Division, National Blood Centre, Kuala Lumpur, Malaysia
| | - Yasmin Ayob
- Immunohaematology Division, National Blood Centre, Kuala Lumpur, Malaysia
| | - Afifah Hassan
- Immunohaematology Division, National Blood Centre, Kuala Lumpur, Malaysia
| | - Norhanim Asidin
- Immunohaematology Division, National Blood Centre, Kuala Lumpur, Malaysia
| | - Nurul M Yahya
- Immunohaematology Division, National Blood Centre, Kuala Lumpur, Malaysia
| | - Rosalind Mathlouthi
- International Blood Group Reference Laboratory, N.H.S. Blood and Transplant, Filton, Bristol, UK
| | - Nicole Thornton
- International Blood Group Reference Laboratory, N.H.S. Blood and Transplant, Filton, Bristol, UK
| | - David J Anstee
- Bristol Institute for Transfusion Sciences, N.H.S. Blood and Transplant, Filton, Bristol, UK
| | - Lesley J Bruce
- Bristol Institute for Transfusion Sciences, N.H.S. Blood and Transplant, Filton, Bristol, UK
| |
Collapse
|
26
|
Keerthivasan G, Liu H, Gump JM, Dowdy SF, Wickrema A, Crispino JD. A novel role for survivin in erythroblast enucleation. Haematologica 2012; 97:1471-9. [PMID: 22491741 DOI: 10.3324/haematol.2011.061093] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Nucleus free red blood cells are unique to mammals. During their terminal stage of differentiation, mammalian erythroblasts exit the cell cycle and enucleate. We previously found that survivin, a member of the chromosomal passenger complex that is required for cytokinesis, is highly expressed in late non-dividing cells. The role of survivin in enucleating erythroblasts is not known. DESIGN AND METHODS In order to identify the role of survivin in these late erythroblasts, we performed proteomic analysis on survivin-bound protein complexes purified from murine erythroleukemia cells. Various molecular and cell biological techniques were used to confirm the presence and function of this novel complex. Furthermore, we used survivin(fl/fl) mice to study the effect of loss of survivin in enucleating erythroblasts. RESULTS We found that survivin failed to co-localize with its known partners' inner centromere protein or Aurora-B in enucleating erythroblasts but rather exists in a multi-protein complex with epidermal growth factor receptor substrate15 and clathrin, two proteins that mediate endocytic vesicle trafficking. As evidence for a direct role of this latter complex in enucleation, we found that knockdown of the genes reduced the efficiency of enucleation of primary human erythroblasts. We also observed that loss of survivin in murine erythroblasts inhibited enucleation and that survivin-deficient cells harbored smaller cytoplasmic vacuoles. Interestingly, vacuolin-1, a small molecule that induces vacuole fusion, rescued the defective enucleation caused by survivin deficiency. CONCLUSIONS This study identified a novel role for survivin in erythroblast enucleation through previously unknown protein partners.
Collapse
Affiliation(s)
- Ganesan Keerthivasan
- Division of Hematology/Oncology Northwestern University, 303 East Superior Street, Lurie 5-113 Chicago, IL, USA
| | | | | | | | | | | |
Collapse
|
27
|
Keerthivasan G, Wickrema A, Crispino JD. Erythroblast enucleation. Stem Cells Int 2011; 2011:139851. [PMID: 22007239 PMCID: PMC3189604 DOI: 10.4061/2011/139851] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 08/10/2011] [Indexed: 12/22/2022] Open
Abstract
Even though the production of orthochromatic erythroblasts can be scaled up to fulfill clinical requirements, enucleation remains one of the critical rate-limiting steps in the production of transfusable red blood cells. Mammalian erythrocytes extrude their nucleus prior to entering circulation, likely to impart flexibility and improve the ability to traverse through capillaries that are half the size of erythrocytes. Recently, there have been many advances in our understanding of the mechanisms underlying mammalian erythrocyte enucleation. This review summarizes these advances, discusses the possible future directions in the field, and evaluates the prospects for improved ex vivo production of red blood cells.
Collapse
Affiliation(s)
- Ganesan Keerthivasan
- Division of Hematology/Oncology, Northwestern University, Chicago, IL 60611, USA
| | | | | |
Collapse
|
28
|
P. falciparum modulates erythroblast cell gene expression in signaling and erythrocyte production pathways. PLoS One 2011; 6:e19307. [PMID: 21573240 PMCID: PMC3087761 DOI: 10.1371/journal.pone.0019307] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 03/28/2011] [Indexed: 01/18/2023] Open
Abstract
Global, genomic responses of erythrocytes to infectious agents have been difficult to measure because these cells are e-nucleated. We have previously demonstrated that in vitro matured, nucleated erythroblast cells at the orthochromatic stage can be efficiently infected by the human malaria parasite Plasmodium falciparum. We now show that infection of orthochromatic cells induces change in 609 host genes. 592 of these transcripts are up-regulated and associated with metabolic and chaperone pathways unique to P. falciparum infection, as well as a wide range of signaling pathways that are also induced in related apicomplexan infections of mouse hepatocytes or human fibroblast cells. Our data additionally show that polychromatophilic cells, which precede the orthochromatic stage and are not infected when co-cultured with P. falciparum, up-regulate a small set of genes, at least two of which are associated with pathways of hematopoiesis and/or erythroid cell development. These data support the idea that P. falciparum affects erythropoiesis at multiple stages during erythroblast differentiation. Further P. falciparum may modulate gene expression in bystander erythroblasts and thus influence pathways of erythrocyte development. This study provides a benchmark of the host erythroblast cell response to infection by P. falciparum.
Collapse
|
29
|
OPN promotes survival of activated T cells by up-regulating CD44 in patients with oral lichen planus. Clin Immunol 2011; 138:291-8. [DOI: 10.1016/j.clim.2010.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Revised: 12/05/2010] [Accepted: 12/10/2010] [Indexed: 01/06/2023]
|
30
|
Abstract
Enucleation of mammalian erythroblasts is a process whose mechanism is largely undefined. The prevailing model suggests that nuclear extrusion occurs via asymmetric cytokinesis. To test this hypothesis, we treated primary erythroblasts with inhibitors of cytokinesis, including blebbistatin, hesperadin, and nocodazole, and then assayed for enucleation. Although these agents inhibited cell-cycle progression and subsequent enucleation when added early in culture, they failed to block enucleation proper when added to postmitotic cells. These results suggest that contraction of the actomyosin ring is not essential for nuclear expulsion. Next, by ultrastructural examination of primary erythroblasts, we observed an accumulation of vacuoles in the cytoplasm proximal to the extruding nucleus. This finding led us to hypothesize that vesicle trafficking contributes to erythroblast enucleation. Here, we show that chemical inhibitors of vesicle trafficking block enucleation of primary erythroblasts without affecting differentiation, cell division, or apoptosis. Moreover, knock-down of clathrin inhibited the enucleation of late erythroblasts. In contrast, vacuolin-1, a small molecule that induces vacuole formation, increased the percentage of enucleated cells. Together, these results illustrate that vesicle trafficking, specifically the formation, movement, and subsequent coalescence of vacuoles at the junction of the nucleus and the cytoplasm, is a critical component of mammalian erythroblast enucleation.
Collapse
|
31
|
Kodippili GC, Spector J, Kang GE, Liu H, Wickrema A, Ritchie K, Low PS. Analysis of the kinetics of band 3 diffusion in human erythroblasts during assembly of the erythrocyte membrane skeleton. Br J Haematol 2010; 150:592-600. [PMID: 20553270 DOI: 10.1111/j.1365-2141.2010.08268.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
During definitive erythropoiesis, erythroid precursors undergo differentiation through multiple nucleated states to an enucleated reticulocyte, which loses its residual RNA/organelles to become a mature erythrocyte. Over the course of these transformations, continuous changes in membrane proteins occur, including shifts in protein abundance, rates of expression, isoform prominence, states of phosphorylation, and stability. In an effort to understand when assembly of membrane proteins into an architecture characteristic of the mature erythrocyte occurs, we quantitated the lateral diffusion of the most abundant membrane protein, band 3 (AE1), during each stage of erythropoiesis using single particle tracking. Analysis of the lateral trajectories of individual band 3 molecules revealed a gradual reduction in mobility of the anion transporter as erythroblasts differentiated. Evidence for this progressive immobilization included a gradual decline in diffusion coefficients as determined at a video acquisition rate of 120 frames/s and a decrease in the percentage of compartment sizes >100 nm. Because complete acquisition of the properties of band 3 seen in mature erythrocytes is not observed until circulating erythrocytes are formed, we suggest that membrane maturation involves a gradual and cooperative assembly process that is not triggered by the synthesis of any single protein.
Collapse
|
32
|
Viñas JL, Sola A, Jung M, Mastora C, Vinuesa E, Pi F, Hotter G. Inhibitory action of Wnt target gene osteopontin on mitochondrial cytochrome c release determines renal ischemic resistance. Am J Physiol Renal Physiol 2010; 299:F234-42. [PMID: 20392802 DOI: 10.1152/ajprenal.00687.2009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Certain determinants of ischemic resistance in the Brown Norway rat strain have been proposed, but no studies to date have focused on the role of the Wnt pathway in the ischemic resistance mechanism. We performed a comparative genomic study in Brown Norway vs. Sprague-Dawley rats. Selective manipulations of the Wnt pathway in vivo and in vitro allowed us to study whether the action of the Wnt pathway on apoptosis through the regulation of osteopontin was critical to the maintenance of inherent ischemic resistance mechanisms. The results revealed a major gene upregulation of the Wnt family in Brown Norway rats after renal ischemia-reperfusion. Manipulation of the Wnt signaling cascade by selective antibodies increased mitochondrial cytochrome c release and caspase 3 activity. The antiapoptotic role of Wnt was mediated by osteopontin, a direct Wnt target gene. Osteopontin was reduced by Wnt antibody administration in vivo, and osteopontin gene silencing in vitro significantly increased mitochondrial cytochrome c release. The overexpression of Wnt pathway genes detected in Brown Norway rats is critical in the maintenance of their inherent ischemic resistance. Activation of the Wnt signaling cascade reduces mitochondrial cytochrome c release and caspase 3 activity through the action of osteopontin.
Collapse
Affiliation(s)
- Jose Luis Viñas
- Centro de Investigaciones Biomédicas en Red de Bioingeniería, Biomateriales y Nanomedicina, Barcelona, Spain.
| | | | | | | | | | | | | |
Collapse
|
33
|
Congote LF, Sadvakassova G, Dobocan MC, Difalco MR, Kriazhev L. Biological activities and molecular interactions of the C-terminal residue of thrombospondin-4, an epitome of acidic amphipathic peptides. Peptides 2010; 31:723-35. [PMID: 20006665 DOI: 10.1016/j.peptides.2009.12.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 12/04/2009] [Accepted: 12/07/2009] [Indexed: 11/17/2022]
Abstract
C21, the C-terminal residue of thrombospondin-4 (TSP-4), was identified as a peptide growth factor during an investigation concerning erythropoietin-dependent, erythroid stimulating factors of endothelial origin. It is active in cultures of several human hematopoietic stem cells, skin fibroblasts and kidney epithelial cells and stimulates red cell formation in anemic mice. A method of affinity chromatography in the presence of high concentrations of Triton X-100, previously developed for identifying proteins associated with the TSP-1 receptor CD47, was utilized for the detection of C21 binding molecules and their detergent-resistant, associated partners. These experiments helped to delineate two different mechanisms of C21 action, which are compatible with its cell proliferating activity. As a cell matrix peptide, C21 binds to the osteopontin receptor CD44 and could act as an osteopontin antagonist, preventing the inhibition of primitive hematopoietic stem cell proliferation. TSP-1, another matrix protein, binds to C21 and could indirectly act as an antagonist, by shunting C21-CD44 interactions. The second mechanism is a direct effect of C21 on cell proliferation. The extremely rapid internalization and nuclear localization of the peptide could be explained by CD44-mediated internalization, followed by a microtubule-mediated transport towards the nucleus, or, eventually, direct membrane insertion. These alternative hypotheses are supported by previously observed membrane insertion of similar synthetic and viral acidic amphipathic peptides, the presence of microtubule-associated protein 1B (MAP1B) and dynactin in the triton-soluble complexes associated with C21 and the presence in such complexes of dual compartment proteins for nuclei and plasma membranes, such as MAP1B, AHNAK and CD44.
Collapse
Affiliation(s)
- Luis F Congote
- Endocrine Laboratory, McGill University Health Centre, 687 Avenue des Pins, Ouest, Montreal, Canada H3A 1A1.
| | | | | | | | | |
Collapse
|
34
|
Sadvakassova G, Dobocan MC, Congote LF. Osteopontin and the C-terminal peptide of thrombospondin-4 compete for CD44 binding and have opposite effects on CD133+ cell colony formation. BMC Res Notes 2009; 2:215. [PMID: 19852812 PMCID: PMC2771039 DOI: 10.1186/1756-0500-2-215] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Accepted: 10/23/2009] [Indexed: 11/10/2022] Open
Abstract
Background C21, the C-terminal peptide of thrombospondin-4, has growth promoting activity and was discovered as one of several erythropoietin-dependent endothelial proteins. C21 stimulates red cell formation in anemic mice and is a growth factor for CD34+ and CD36+ hematopoietic cells, skin fibroblasts and kidney epithelial cells. ROD1 has been identified as an intracellular mediator. Nothing is known about the existence of putative C21 receptors on plasma membranes of target cells. Findings We analyzed the nature of C21-binding proteins in cell lysates of skin fibroblasts using C21 affinity columns. The membrane receptor CD44 was identified as C21-binding protein by mass spectrometry. We were unable to demonstrate any direct involvement of CD44 on cell growth or the effect of C21 on cell proliferation. A soluble form of CD44 was synthesized in insect cells and purified from culture supernatants with a combination of PVDF filtration in the presence of ammonium sulphate and HPLC. Both osteopontin and hyaluronic acid competitively displaced Biotin-C21 binding to CD44. In a colony-forming assay using primitive CD133+ hematopoietic stem cells from cord blood, osteopontin and C21 had opposite effects and C21 reduced the inhibitory action of osteopontin. Conclusion CD44 is a C21-binding membrane protein. We could not demonstrate an involvement of CD44 in the proliferative action of C21. Nevertheless, based on the antagonism of C21 and osteopontin in hematopoietic precursors, we speculate that C21 could indirectly have a major impact on hematopoietic stem cell proliferation, by hindering osteopontin membrane binding at the level of the bone marrow niche.
Collapse
Affiliation(s)
- Gulzhakhan Sadvakassova
- Endocrine Laboratory, McGill University Health Centre, 687 avenue des pins, ouest, Montreal, Canada.
| | | | | |
Collapse
|
35
|
Haldar K, Mohandas N. Malaria, erythrocytic infection, and anemia. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2009; 2009:87-93. [PMID: 20008186 PMCID: PMC2933134 DOI: 10.1182/asheducation-2009.1.87] [Citation(s) in RCA: 137] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Malaria is a major world health problem. It results from infection of parasites belonging to the genus Plasmodium. Plasmodium falciparum and Plasmodium vivax cause the major human malarias, with P falciparum being the more virulent. During their blood stages of infection, both P falciparum and P vivax induce anemia. Severe malarial anemia caused by P falciparum is responsible for approximately a third of the deaths associated with disease. Malarial anemia appears to be multi-factorial. It involves increased removal of circulating erythrocytes as well as decreased production of erythrocytes in the bone marrow. The molecular mechanisms underlying malarial anemia are largely unknown. Over the last five years, malaria parasite ligands have been investigated for their remodeling of erythrocytes and possible roles in destruction of mature erythrocytes. Polymorphisms in cytokines have been associated with susceptibility to severe malarial anemia: these cytokines and malaria "toxins" likely function by perturbing erythropoiesis. Finally a number of co-infections increase susceptibility to malarial anemia, likely because they exacerbate inflammation caused by malaria. Because of the complexities involved, the study of severe malarial anemia may need a "systems approach" to yield comprehensive understanding of defects in both erythropoiesis and immunity associated with disease. New and emerging tools such as (i) mathematical modeling of the dynamics of host control of malarial infection, (ii) ex vivo perfusion of human spleen to measure both infected and uninfected erythrocyte retention, and (iii) in vitro development of erythroid progenitors to dissect responsiveness to cytokine imbalance or malaria toxins, may be especially useful to develop integrated mechanistic insights and therapies to control this major and fatal disease pathology.
Collapse
Affiliation(s)
- Kasturi Haldar
- Center for Rare and Neglected Diseases, University of Notre Dame, South Bend, IN, USA.
| | | |
Collapse
|
36
|
Abstract
Vasomotor effects in human skin induced by vibration of low amplitude (10-25 microns) and high frequency (150-250 Hz) have been studied by using skin temperature changes as an approximative measure of variations in skin blood flow. In all tested areas of the body surface, including the face, low-amplitude high-frequency vibration regularly induces vasodilatation. The spatial distribution of the temperature changes induced from different sites of stimulation was studied by infrared thermography. The latencies of the temperature changes, determined by thermistor recordings, were found to vary with site of stimulation and stimulus parameters. The increase in temperature to a given stimulus is greater the lower the prevalent skin temperature, i.e. the increase in blood flow is larger the greater the initial vasomotor tone. The results are in accordance with the view that the vasodilatation is due to a reflex inhibition of pre-existent vasomotor tone in the skin by the afferent inflow from vibration-sensitive mechanoreceptors. High-amplitude vibration (100-200 microns), performed in a few comparative experiments, caused vasoconstriction.
Collapse
|