1
|
Afzal A, Thomas N, Warraich Z, Barbay S, Mocco J. Hematopoietic Endothelial Progenitor cells enhance motor function and cortical motor map integrity following cerebral ischemia. Restor Neurol Neurosci 2024; 42:139-149. [PMID: 38820024 DOI: 10.3233/rnn-231378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
Background Hematopoietic stem cells (HSC) are recruited to ischemic areas in the brain and contribute to improved functional outcome in animals. However, little is known regarding the mechanisms of improvement following HSC administration post cerebral ischemia. To better understand how HSC effect post-stroke improvement, we examined the effect of HSC in ameliorating motor impairment and cortical dysfunction following cerebral ischemia. Methods Baseline motor performance of male adult rats was established on validated motor tests. Animals were assigned to one of three experimental cohorts: control, stroke, stroke + HSC. One, three and five weeks following a unilateral stroke all animals were tested on motor skills after which intracortical microstimulation was used to derive maps of forelimb movement representations within the motor cortex ipsilateral to the ischemic injury. Results Stroke + HSC animals significantly outperformed stroke animals on single pellet reaching at weeks 3 and 5 (28±3% and 33±3% versus 11±4% and 17±3%, respectively, p < 0.05 at both time points). Control animals scored 44±1% and 47±1%, respectively. Sunflower seed opening task was significantly improved in the stroke + HSC cohort versus the stroke cohort at week five-post stroke (79±4 and 48±5, respectively, p < 0.05). Furthermore, Stroke + HSC animals had significantly larger forelimb motor maps than animals in the stroke cohort. Overall infarct size did not significantly differ between the two stroked cohorts. Conclusion These data suggest that post stroke treatment of HSC enhances the functional integrity of residual cortical tissue, which in turn supports improved behavioral outcome, despite no observed reduction in infarct size.
Collapse
Affiliation(s)
- Aqeela Afzal
- Department of Neurological Surgery, Vanderbilt University, Nashville, TN, USA
| | - Nagheme Thomas
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
| | | | - Scott Barbay
- Department of Physical Medicine and Rehabilitation, University of Kansas Medical Center, Kansas City, Kansas, KS, USA
| | - J Mocco
- Department of Neurological Surgery, Mount Sinai Health, New York, NY, USA
| |
Collapse
|
2
|
Koeniger T, Bell L, Mifka A, Enders M, Hautmann V, Mekala SR, Kirchner P, Ekici AB, Schulz C, Wörsdörfer P, Mencl S, Kleinschnitz C, Ergün S, Kuerten S. Bone marrow-derived myeloid progenitors in the leptomeninges of adult mice. STEM CELLS (DAYTON, OHIO) 2020; 39:227-239. [PMID: 33270951 DOI: 10.1002/stem.3311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 09/29/2020] [Accepted: 11/17/2020] [Indexed: 11/11/2022]
Abstract
Although the bone marrow contains most hematopoietic activity during adulthood, hematopoietic stem and progenitor cells can be recovered from various extramedullary sites. Cells with hematopoietic progenitor properties have even been reported in the adult brain under steady-state conditions, but their nature and localization remain insufficiently defined. Here, we describe a heterogeneous population of myeloid progenitors in the leptomeninges of adult C57BL/6 mice. This cell pool included common myeloid, granulocyte/macrophage, and megakaryocyte/erythrocyte progenitors. Accordingly, it gave rise to all major myelo-erythroid lineages in clonogenic culture assays. Brain-associated progenitors persisted after tissue perfusion and were partially inaccessible to intravenous antibodies, suggesting their localization behind continuous blood vessel endothelium such as the blood-arachnoid barrier. Flt3Cre lineage tracing and bone marrow transplantation showed that the precursors were derived from adult hematopoietic stem cells and were most likely continuously replaced via cell trafficking. Importantly, their occurrence was tied to the immunologic state of the central nervous system (CNS) and was diminished in the context of neuroinflammation and ischemic stroke. Our findings confirm the presence of myeloid progenitors at the meningeal border of the brain and lay the foundation to unravel their possible functions in CNS surveillance and local immune cell production.
Collapse
Affiliation(s)
- Tobias Koeniger
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Luisa Bell
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Anika Mifka
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Enders
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Valentin Hautmann
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Subba Rao Mekala
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Philipp Kirchner
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Arif B Ekici
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, Ludwig Maximilian University of Munich, Munich, Germany
| | - Philipp Wörsdörfer
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Stine Mencl
- University Hospital Essen, Department of Neurology, University Duisburg-Essen, Essen, Germany
| | - Christoph Kleinschnitz
- University Hospital Essen, Department of Neurology, University Duisburg-Essen, Essen, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany.,Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.,Anatomisches Institut, Neuroanatomie, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| |
Collapse
|
3
|
Pesaresi M, Bonilla-Pons SA, Cosma MP. In vivo somatic cell reprogramming for tissue regeneration: the emerging role of the local microenvironment. Curr Opin Cell Biol 2018; 55:119-128. [PMID: 30071468 DOI: 10.1016/j.ceb.2018.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/01/2018] [Accepted: 07/09/2018] [Indexed: 12/16/2022]
Abstract
The past few years have witnessed an exponential increase of interest in the reprogramming process. This has been motivated by the enthusiasm of unravelling key aspects not only of cell identity and dedifferentiation, but also of the endogenous regenerative capacities of mammalian organs. Here, we present the most recent advances in the field of reprogramming, stressing how they are re-defining the rules of cell fate and plasticity in vivo. Specifically, we focus on the emerging role of the tissue microenvironment, with particular emphasis on tissue damage, inflammation and senescence that can facilitate in vivo reprogramming and regeneration through cell-extrinsic mechanisms.
Collapse
Affiliation(s)
- Martina Pesaresi
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
| | - Sergi A Bonilla-Pons
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain; Universitat de Barcelona (UB), Barcelona, Spain
| | - Maria Pia Cosma
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Dr Aiguader 88, 08003 Barcelona, Spain; ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain.
| |
Collapse
|
4
|
Zhang Y, Zhang H, Lin S, Chen X, Yao Y, Mao X, Shao B, Zhuge Q, Jin K. SDF-1/CXCR7 Chemokine Signaling is Induced in the Peri-Infarct Regions in Patients with Ischemic Stroke. Aging Dis 2018; 9:287-295. [PMID: 29896417 PMCID: PMC5963349 DOI: 10.14336/ad.2017.1112] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 11/12/2017] [Indexed: 12/11/2022] Open
Abstract
Stromal-derived factor-1 (SDF-1, also known as CXCL12) and its receptors CXCR4 and CXCR7 play important roles in brain repair after ischemic stroke, as SDF-1/ CXCR4/CXCR7 chemokine signaling is critical for recruiting stem cells to sites of ischemic injury. Upregulation of SDF-1/CXCR4/CXCR7 chemokine signaling in the ischemic regions has been well-documented in the animal models of ischemic stroke, but not in human ischemic brain. Here, we found that protein expression of SDF-1 and CXCR7, but not CXCR4, were significantly increased in the cortical peri-infarct regions (penumbra) after ischemic stroke in human, compared with adjacent normal tissues and control subjects. Double-label fluorescence immunohistochemistry shows that SDF-1 and CXCR4 proteins were expressed in neuronal cells and astrocytes in the normal brain tissue and peri-infarct regions. CXCR7 protein was also observed in neuronal cells and astrocytes in the normal cortical regions, but predominantly in astrocytes in the penumbra of ischemic brain. Our data suggest that ischemic stroke in human leads to an increase in the expression of SDF-1 and CXCR7, but not CXCR4, in the peri-infarct cerebral cortex. Our findings suggest that chemokine SFD-1 is expressed not only in animal models of stroke, but also in the human brain after an ischemic injury. In addition, unlike animals, CXCR7 may be the primary receptor of SDF-1 in human stroke brain.
Collapse
Affiliation(s)
- Yu Zhang
- 1Department of Neurosurgery, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Hongxia Zhang
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, Texas 76107, USA
| | - Siyang Lin
- 3Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xudong Chen
- 3Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yu Yao
- 4Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - XiaoOu Mao
- 5Buck Institute for Age Research, Novato, California 94945, USA
| | - Bei Shao
- 3Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Qichuan Zhuge
- 1Department of Neurosurgery, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.,3Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Kunlin Jin
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, Texas 76107, USA.,3Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| |
Collapse
|
5
|
Pesaresi M, Bonilla-Pons SA, Simonte G, Sanges D, Di Vicino U, Cosma MP. Endogenous Mobilization of Bone-Marrow Cells Into the Murine Retina Induces Fusion-Mediated Reprogramming of Müller Glia Cells. EBioMedicine 2018. [PMID: 29525572 PMCID: PMC5952225 DOI: 10.1016/j.ebiom.2018.02.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Müller glial cells (MGCs) represent the most plastic cell type found in the retina. Following injury, zebrafish and avian MGCs can efficiently re-enter the cell cycle, proliferate and generate new functional neurons. The regenerative potential of mammalian MGCs, however, is very limited. Here, we showed that N-methyl-d-aspartate (NMDA) damage stimulates murine MGCs to re-enter the cell cycle and de-differentiate back to a progenitor-like stage. These events are dependent on the recruitment of endogenous bone marrow cells (BMCs), which, in turn, is regulated by the stromal cell-derived factor 1 (SDF1)-C-X-C motif chemokine receptor type 4 (CXCR4) pathway. BMCs mobilized into the damaged retina can fuse with resident MGCs, and the resulting hybrids undergo reprogramming followed by re-differentiation into cells expressing markers of ganglion and amacrine neurons. Our findings constitute an important proof-of-principle that mammalian MGCs retain their regenerative potential, and that such potential can be activated via cell fusion with recruited BMCs. In this perspective, our study could contribute to the development of therapeutic strategies based on the enhancement of mammalian endogenous repair capabilities. Endogenous bone marrow cells migrate into NMDA-damaged murine retinae and fuse with retinal Müller glial cells (MGCs). MGCs can be reprogrammed to retinal progenitors to then differentiate into ganglion and amacrine neurons. Modulation of the SDF1/CXCR4 pathway regulates BMC migration, BMC-MGC fusion, and MGC reprogramming.
Retinal degeneration is present in a large and heterogeneous group of debilitating diseases, often not curable. Cell therapy represents an interesting approach to regenerate injured retinal tissue. However, it comes with some hurdles in terms of engraftment and differentiation of the transplanted cells. Here, we reported that murine Müller glia cells can be converted into retinal neurons after fusion with endogenous bone marrow cells. The efficiency of this mechanism can be enhanced by perturbation of the SDF1/CXCR4 signaling pathway. Our study provides an important proof-of-principle that the limited endogenous regeneration capability of mammals can be enhanced by modulation of specific signaling pathways.
Collapse
Affiliation(s)
- Martina Pesaresi
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Sergi A Bonilla-Pons
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain.; Universitat de Barcelona (UB), Barcelona, Spain
| | - Giacoma Simonte
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Daniela Sanges
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Umberto Di Vicino
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Maria Pia Cosma
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain.; ICREA, Barcelona, Spain..
| |
Collapse
|
6
|
Chen C, Chu SF, Liu DD, Zhang Z, Kong LL, Zhou X, Chen NH. Chemokines play complex roles in cerebral ischemia. Neurochem Int 2018. [DOI: 10.1016/j.neuint.2017.06.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
7
|
Goichberg P. Current Understanding of the Pathways Involved in Adult Stem and Progenitor Cell Migration for Tissue Homeostasis and Repair. Stem Cell Rev Rep 2017; 12:421-37. [PMID: 27209167 DOI: 10.1007/s12015-016-9663-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
With the advancements in the field of adult stem and progenitor cells grows the recognition that the motility of primitive cells is a pivotal aspect of their functionality. There is accumulating evidence that the recruitment of tissue-resident and circulating cells is critical for organ homeostasis and effective injury responses, whereas the pathobiology of degenerative diseases, neoplasm and aging, might be rooted in the altered ability of immature cells to migrate. Furthermore, understanding the biological machinery determining the translocation patterns of tissue progenitors is of great relevance for the emerging methodologies for cell-based therapies and regenerative medicine. The present article provides an overview of studies addressing the physiological significance and diverse modes of stem and progenitor cell trafficking in adult mammalian organs, discusses the major microenvironmental cues regulating cell migration, and describes the implementation of live imaging approaches for the exploration of stem cell movement in tissues and the factors dictating the motility of endogenous and transplanted cells with regenerative potential.
Collapse
Affiliation(s)
- Polina Goichberg
- Department Anesthesiology, Perioperative and Pain Medicine, Harvard Medical School, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA.
| |
Collapse
|
8
|
Stem cell therapy for abrogating stroke-induced neuroinflammation and relevant secondary cell death mechanisms. Prog Neurobiol 2017; 158:94-131. [PMID: 28743464 DOI: 10.1016/j.pneurobio.2017.07.004] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/13/2022]
Abstract
Ischemic stroke is a leading cause of death worldwide. A key secondary cell death mechanism mediating neurological damage following the initial episode of ischemic stroke is the upregulation of endogenous neuroinflammatory processes to levels that destroy hypoxic tissue local to the area of insult, induce apoptosis, and initiate a feedback loop of inflammatory cascades that can expand the region of damage. Stem cell therapy has emerged as an experimental treatment for stroke, and accumulating evidence supports the therapeutic efficacy of stem cells to abrogate stroke-induced inflammation. In this review, we investigate clinically relevant stem cell types, such as hematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs), endothelial progenitor cells (EPCs), very small embryonic-like stem cells (VSELs), neural stem cells (NSCs), extraembryonic stem cells, adipose tissue-derived stem cells, breast milk-derived stem cells, menstrual blood-derived stem cells, dental tissue-derived stem cells, induced pluripotent stem cells (iPSCs), teratocarcinoma-derived Ntera2/D1 neuron-like cells (NT2N), c-mycER(TAM) modified NSCs (CTX0E03), and notch-transfected mesenchymal stromal cells (SB623), comparing their potential efficacy to sequester stroke-induced neuroinflammation and their feasibility as translational clinical cell sources. To this end, we highlight that MSCs, with a proven track record of safety and efficacy as a transplantable cell for hematologic diseases, stand as an attractive cell type that confers superior anti-inflammatory effects in stroke both in vitro and in vivo. That stem cells can mount a robust anti-inflammatory action against stroke complements the regenerative processes of cell replacement and neurotrophic factor secretion conventionally ascribed to cell-based therapy in neurological disorders.
Collapse
|
9
|
Niemiro GM, Parel J, Beals J, van Vliet S, Paluska SA, Moore DR, Burd NA, De Lisio M. Kinetics of circulating progenitor cell mobilization during submaximal exercise. J Appl Physiol (1985) 2017; 122:675-682. [DOI: 10.1152/japplphysiol.00936.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/23/2016] [Accepted: 01/08/2017] [Indexed: 12/28/2022] Open
Abstract
Circulating progenitor cells (CPCs) are a heterogeneous population of stem/progenitor cells in peripheral blood that includes hematopoietic stem and progenitor cells (HSPCs and HSCs), endothelial progenitor cells (EPCs), and mesenchymal stem cells (MSCs) that are involved in tissue repair and adaptation. CPC mobilization during exercise remains uncharacterized in young adults. The purpose of this study was to investigate the kinetics of CPC mobilization during and after submaximal treadmill running and their relationship to mobilization factors. Seven men [age = 25.3 ± 2.4 yr, body mass index = 23.5 ± 1.0 kg/m2, peak O2uptake (V̇o2peak) = 60.9 ± 2.74 ml·kg−1·min−1] ran on a treadmill for 60 min at 70% V̇o2peak. Blood sampling occurred before (Pre), during [20 min (20e), 40 min (40e), 60 min (60e)], and after exercise [15 min (15p), 60 min (60p), 120 min (120p)] for quantification of CPCs (CD34+), HSPCs (CD34+/CD45low), HSCs (CD34+/CD45low/CD38−), CD34+MSCs (CD45−/CD34+/CD31−/CD105+), CD34−MSCs (CD45−/CD34−/CD31−/CD105+), and EPCs (CD45−/CD34+/CD31+) via flow cytometry. CPC concentration increased compared with Pre at 20e and 40e (2.7- and 2.4-fold, respectively, P < 0.05). HSPCs and HSCs increased at 20e compared with 60p (2.7- and 2.8-fold, respectively, P < 0.05), whereas EPCs and both MSC populations did not change. CXC chemokine ligand (CXCL) 12 (1.5-fold; P < 0.05) and stem cell factor (1.3-fold; P < 0.05) were increased at 40e and remained elevated postexercise. The peak increase in CPCs was positively correlated to concentration of endothelial cells during exercise with no relationship to CXCL12 and SCF. Our data show the kinetics of progenitor cell mobilization during exercise that could provide insight into cellular mediators of exercise-induced adaptations, and have implication for the use of exercise as an adjuvant therapy for CPC collection in hematopoietic stem cell transplant.NEW & NOTEWORTHY Using a comprehensive evaluation of circulating progenitor cells (CPCs), we show that CPC mobilization during exercise is related to tissue damage, and not plasma concentrations of CXC chemokine ligand 12 and stem cell factor. These data have implications for the use of exercise interventions as adjuvant therapy for CPC mobilization in the context of hematopoietic stem cell transplant and also support the role of mobilized progenitor cells as cellular mediators of systemic adaptations to exercise.
Collapse
Affiliation(s)
- Grace M. Niemiro
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Justin Parel
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Joseph Beals
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Stephan van Vliet
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Scott A. Paluska
- Department of Family Medicine, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Daniel R. Moore
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada; and
| | - Nicholas A. Burd
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Michael De Lisio
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
- School of Human Kinetics, Brain and Mind Institute, Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
10
|
Niemiro GM, Raine LB, Khan NA, Emmons R, Little J, Kramer AF, Hillman CH, De Lisio M. Circulating progenitor cells are positively associated with cognitive function among overweight/obese children. Brain Behav Immun 2016; 57:47-52. [PMID: 27132057 PMCID: PMC7404617 DOI: 10.1016/j.bbi.2016.03.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/11/2016] [Accepted: 03/23/2016] [Indexed: 12/18/2022] Open
Abstract
Recent evidence has indicated that overweight/obese children may experience cognitive and immune dysfunction, but the underlying mechanisms responsible for the association between overweight/obesity, immune dysfunction, and cognition have yet to be established. The present study aimed to identify a novel link between obesity-induced immune system dysregulation and cognition in preadolescent children. A total of 27 male children (age: 8-10years) were recruited and separated by body mass index (BMI) into healthy weight (HW: 5th-84.9th percentile, n=16) and overweight/obese (OW: ⩾85th percentile, n=11) groups. Adiposity was assessed using dual energy X-ray absorptiometry (DXA), and aspects of executive function were assessed using the Woodcock-Johnson III Tests of Cognitive Abilities. Monocyte populations (CD14(+)CD16(-), CD14(+)CD16(+)) with and without expression of chemokine receptor type 2 (CCR2), and circulating progenitor cells (CPCs: CD34(+)CD45(dim)), in peripheral blood were quantified by flow cytometry. CPCs were isolated by flow sorting and cultured for 24h for collection of conditioned media (CM) that was applied to SH-SY5Y neuroblastomas to examine the paracrine effects of CPCs on neurogenesis. OW had significantly higher quantities of both populations of monocytes (CD14(+)CD16(-): 57% increase; CD14(+)CD16(+): 95% increase, both p<0.01), monocytes expressing CCR2 (CD14(+)CD16(-)CCR2(+): 66% increase; CD14(+)CD16(+)CCR2(+): 168% increase, both p<0.01), and CPCs (47% increase, p<0.05) than HW. CPCs were positively correlated with abdominal adiposity in OW, and negatively correlated in HW with a significant difference between correlations (p<0.05). CPC content was positively correlated with executive processes in OW, and negatively correlated in HW with a significant difference in the strength of the correlations between groups (p<0.05 for correlation between OW and HW). Finally, CPC-CM from OW trended to increase neuroblast viability in vitro relative to HW (1.79 fold, p=0.07). These novel findings indicate that increased content of CPCs among OW children may play a role in preventing decrements in cognitive function via paracrine mechanisms.
Collapse
Affiliation(s)
- Grace M. Niemiro
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, United States
| | - Lauren B. Raine
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, United States
| | - Naiman A. Khan
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, United States
| | - Russell Emmons
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, United States
| | - Jonathan Little
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Canada
| | - Arthur F. Kramer
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, United States
| | - Charles H. Hillman
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, United States,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, United States
| | - Michael De Lisio
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, United States.
| |
Collapse
|
11
|
Exercise as an Adjuvant Therapy for Hematopoietic Stem Cell Mobilization. Stem Cells Int 2016; 2016:7131359. [PMID: 27123008 PMCID: PMC4830735 DOI: 10.1155/2016/7131359] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/03/2016] [Accepted: 02/07/2016] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic stem cell transplant (HSCT) using mobilized peripheral blood hematopoietic stem cells (HSPCs) is the only curative strategy for many patients suffering from hematological malignancies. HSPC collection protocols rely on pharmacological agents to mobilize HSPCs to peripheral blood. Limitations including variable donor responses and long dosing protocols merit further investigations into adjuvant therapies to enhance the efficiency of HSPCs collection. Exercise, a safe and feasible intervention in patients undergoing HSCT, has been previously shown to robustly stimulate HSPC mobilization from the bone marrow. Exercise-induced HSPC mobilization is transient limiting its current clinical potential. Thus, a deeper investigation of the mechanisms responsible for exercise-induced HSPC mobilization and the factors responsible for removal of HSPCs from circulation following exercise is warranted. The present review will describe current research on exercise and HSPC mobilization, outline the potential mechanisms responsible for exercise-induced HSPC mobilization, and highlight potential sites for HSPC homing following exercise. We also outline current barriers to the implementation of exercise as an adjuvant therapy for HSPC mobilization and suggest potential strategies to overcome these barriers.
Collapse
|