1
|
Gonta R, Schenkel JM, Klein K, Martinez F, Medina MC. Factors influencing white blood cell mobilisation in healthy granulocyte donors. Transfus Med 2025; 35:177-183. [PMID: 39601217 DOI: 10.1111/tme.13115] [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] [Received: 05/10/2024] [Revised: 09/23/2024] [Accepted: 11/10/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Granulocyte transfusions represent a therapeutic option for severely neutropenic patients with bacterial or fungal infections that are otherwise unresponsive to conventional therapy. Prior clinical studies suggest that patients receiving higher granulocyte doses achieve superior outcomes. Consequently, suboptimal donor stimulation and collection leading to lower granulocyte doses likely correlate with worse clinical outcomes. STUDY DESIGN A retrospective analysis was conducted on mobilisation data from 312 granulocyte collections from healthy donors between January 2020 and May 2023. This study was performed in a single blood donor center exclusively supporting a comprehensive cancer center. Donors underwent stimulation with 480 mcg of filgrastim (granulocyte colony stimulating factor [G-CSF]) subcutaneously and 8 mg of dexamethasone orally administered 12 to 14 h before collection. The correlation between donor characteristics (age, gender, body weight (BW), body mass index (BMI), baseline haemoglobin (Hgb), and platelet (PLT) counts) and mobilisation efficiency (Δ WBC, defined as post-mobilisation WBC count-baseline WBC count) was examined to identify factors associated with enhanced mobilisation efficiency. Additionally, the impact of multiple donations on Δ WBC in repeat donors was assessed. RESULTS The median donor age was 43 years (range 18-81), with 224 male and 88 female donors. Female donors exhibited significantly higher baseline PLT counts and post-mobilisation WBC counts. However, donor gender did not significantly affect Δ WBC. A negative correlation was observed between Δ WBC and age (r = -0.235, p = 0.001), with older donors (61-81 years) exhibiting significantly lower mobilisation efficiency. BW and BMI differences had no significant effect on Δ WBC. A positive correlation was identified between baseline PLT count and Δ WBC (r = 0.140, p = 0.014), with females having significantly higher baseline PLT counts (p = 0.0004). No correlation was found between Δ WBC and baseline Hgb (r = 0.004, p = 0.477). Repeat donors showed no statistically significant change in Δ WBC with subsequent donations, with a mean interval of 136.5 days between collections. CONCLUSION Mobilisation efficiency was not impacted by donor BW or BMI suggesting that BW-based G-CSF stimulation is not essential for optimising WBC mobilisation. Rather, a fixed single dose of 480 mcg of G-CSF and 8 mg of dexamethasone was sufficient to mobilise donors, thus reducing the procedural costs and the potential risks for medication-related side effects. The positive correlation found between baseline PLT count and Δ WBC suggests that PLT count could be used as a potential predictor of mobilisation efficiency. Mobilisation response in up to four collections in repeat granulocytes donors was not affected in subsequent donations. However, sample size is a limitation, and more data is needed for a meaningful conclusion of whether frequent granulocyte donations are safe and effective.
Collapse
Affiliation(s)
- Roman Gonta
- Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Pathology and Laboratory Medicine, Moffitt Cancer Center, Florida, USA
| | - Jason M Schenkel
- Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kimberly Klein
- Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Fernando Martinez
- Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mayrin Correa Medina
- Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Pathology and Laboratory Medicine, Moffitt Cancer Center, Florida, USA
| |
Collapse
|
2
|
Chi A, Yang C, Liu J, Zhai Z, Shi X. Reconstructing the Stem Leydig Cell Niche via the Testicular Extracellular Matrix for the Treatment of Testicular Leydig Cell Dysfunction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410808. [PMID: 39555675 PMCID: PMC11727238 DOI: 10.1002/advs.202410808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/24/2024] [Indexed: 11/19/2024]
Abstract
Therapies involving the use of stem Leydig cells (SLCs), as testicular mesenchymal stromal cells, have shown great promise in the treatment of Leydig cell (LC) dysfunction in aging males. However, the outcomes of these therapies are not satisfactory. In this study, it is demonstrated that the aging microenvironment of the testicular interstitium impairs the function of SLCs, leading to poor regeneration of LCs and, consequently, inefficient functional restoration. The study develops a decellularized testicular extracellular matrix (dTECM) hydrogel from young pigs and evaluates its safety and feasibility as a supportive niche for the expansion and differentiation of SLCs. dTECM hydrogel facilitates the steroidogenic differentiation of SLCs into LCs, the primary producers of testosterone. The combination of SLCs with a dTECM hydrogel leads to a significant and sustained increase in testosterone levels, which promotes the restoration of spermatogenesis and fertility in an LC-deficient and aged mouse model. Mechanistically, collagen 1 within the dTECM is identified as a key factor contributing to these effects. Notably, symptoms associated with testosterone deficiency syndrome are significantly alleviated in aged mice. These findings may aid the design of therapeutic interventions for patients with testosterone deficiency in the clinic.
Collapse
Affiliation(s)
- Ani Chi
- National Engineering Research Centre for Tissue Restoration and ReconstructionKey Laboratory of Biomedical Engineering of Guangdong ProvinceSouth China University of TechnologyGuangzhou510640P. R. China
| | - Chao Yang
- National Engineering Research Centre for Tissue Restoration and ReconstructionKey Laboratory of Biomedical Engineering of Guangdong ProvinceSouth China University of TechnologyGuangzhou510640P. R. China
| | - Jie Liu
- National Engineering Research Centre for Tissue Restoration and ReconstructionKey Laboratory of Biomedical Engineering of Guangdong ProvinceSouth China University of TechnologyGuangzhou510640P. R. China
| | - Zhichen Zhai
- National Engineering Research Centre for Tissue Restoration and ReconstructionKey Laboratory of Biomedical Engineering of Guangdong ProvinceSouth China University of TechnologyGuangzhou510640P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong ProvinceSouth China University of TechnologyGuangzhou510006P. R. China
- School of Materials Science and EngineeringSouth China University of TechnologyGuangzhou510640P. R. China
| | - Xuetao Shi
- National Engineering Research Centre for Tissue Restoration and ReconstructionKey Laboratory of Biomedical Engineering of Guangdong ProvinceSouth China University of TechnologyGuangzhou510640P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong ProvinceSouth China University of TechnologyGuangzhou510006P. R. China
- School of Materials Science and EngineeringSouth China University of TechnologyGuangzhou510640P. R. China
| |
Collapse
|
3
|
Seara FAC, Maciel L, Kasai-Brunswick TH, Nascimento JHM, Campos-de-Carvalho AC. Extracellular Vesicles and Cardiac Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1418:33-56. [PMID: 37603271 DOI: 10.1007/978-981-99-1443-2_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Global population aging is a major challenge to health and socioeconomic policies. The prevalence of diseases progressively increases with aging, with cardiovascular disease being the major cause of mortality among elderly people. The allostatic overload imposed by the accumulation of cardiac senescent cells has been suggested to play a pivotal role in the aging-related deterioration of cardiovascular function. Senescent cells exhibit intrinsic disorders and release a senescence-associated secretory phenotype (SASP). Most of these SASP compounds and damaged molecules are released from senescent cells by extracellular vesicles (EVs). Once secreted, these EVs can be readily incorporated by recipient neighboring cells and elicit cellular damage or otherwise can promote extracellular matrix remodeling. This has been associated with the development of cardiac dysfunction, fibrosis, and vascular calcification, among others. The molecular signature of these EVs is highly variable and might provide important information for the development of aging-related biomarkers. Conversely, EVs released by the stem and progenitor cells can exert a rejuvenating effect, raising the possibility of future anti-aging therapies.
Collapse
Affiliation(s)
- Fernando A C Seara
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Physiological Sciences, Institute of Health and Biological Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Brazil
| | - Leonardo Maciel
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Federal University of Rio de Janeiro, Campus Professor Geraldo, Duque de Caxias, Brazil
| | - Tais Hanae Kasai-Brunswick
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Center of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jose H M Nascimento
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
- Laboratory of Cardiac Electrophysiology, Carlos Chagas Filho Institute of Biophysics, Health Sciences Centre, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Antonio C Campos-de-Carvalho
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Center of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Tabibzadeh S. Resolving Geroplasticity to the Balance of Rejuvenins and Geriatrins. Aging Dis 2022; 13:1664-1714. [PMID: 36465174 PMCID: PMC9662275 DOI: 10.14336/ad.2022.0414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/14/2022] [Indexed: 09/29/2024] Open
Abstract
According to the cell centric hypotheses, the deficits that drive aging occur within cells by age dependent progressive damage to organelles, telomeres, biologic signaling pathways, bioinformational molecules, and by exhaustion of stem cells. Here, we amend these hypotheses and propose an eco-centric model for geroplasticity (aging plasticity including aging reversal). According to this model, youth and aging are plastic and require constant maintenance, and, respectively, engage a host of endogenous rejuvenating (rejuvenins) and gero-inducing [geriatrin] factors. Aging in this model is akin to atrophy that occurs as a result of damage or withdrawal of trophic factors. Rejuvenins maintain and geriatrins adversely impact cellular homeostasis, cell fitness, and proliferation, stem cell pools, damage response and repair. Rejuvenins reduce and geriatrins increase the age-related disorders, inflammatory signaling, and senescence and adjust the epigenetic clock. When viewed through this perspective, aging can be successfully reversed by supplementation with rejuvenins and by reducing the levels of geriatrins.
Collapse
Affiliation(s)
- Siamak Tabibzadeh
- Frontiers in Bioscience Research Institute in Aging and Cancer, Irvine, CA 92618, USA
| |
Collapse
|
5
|
Emerging Evidence of the Significance of Thioredoxin-1 in Hematopoietic Stem Cell Aging. Antioxidants (Basel) 2022; 11:antiox11071291. [PMID: 35883782 PMCID: PMC9312246 DOI: 10.3390/antiox11071291] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
The United States is undergoing a demographic shift towards an older population with profound economic, social, and healthcare implications. The number of Americans aged 65 and older will reach 80 million by 2040. The shift will be even more dramatic in the extremes of age, with a projected 400% increase in the population over 85 years old in the next two decades. Understanding the molecular and cellular mechanisms of ageing is crucial to reduce ageing-associated disease and to improve the quality of life for the elderly. In this review, we summarized the changes associated with the ageing of hematopoietic stem cells (HSCs) and what is known about some of the key underlying cellular and molecular pathways. We focus here on the effects of reactive oxygen species and the thioredoxin redox homeostasis system on ageing biology in HSCs and the HSC microenvironment. We present additional data from our lab demonstrating the key role of thioredoxin-1 in regulating HSC ageing.
Collapse
|
6
|
Comparative analysis of mouse embryonic palatal mesenchymal cells isolated by two primary culture methods. Tissue Cell 2022; 76:101783. [DOI: 10.1016/j.tice.2022.101783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/11/2022] [Accepted: 03/11/2022] [Indexed: 11/21/2022]
|
7
|
Hoffman RA, Huang S, Chalasani G, Vallejo AN. Disparate Recruitment and Retention of Plasmacytoid Dendritic Cells to The Small Intestinal Mucosa between Young and Aged Mice. Aging Dis 2021; 12:1183-1196. [PMID: 34341701 PMCID: PMC8279532 DOI: 10.14336/ad.2021.0119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/19/2021] [Indexed: 11/01/2022] Open
Abstract
Plasmacytoid dendritic cells (pDC), a highly specialized class of innate immune cells that serve as rapid sensors of danger signals in circulation or in lymphoid tissue are well studied. However, there remains knowledge gaps about age-dependent changes of pDC function in the intestinal mucosa. Here, we report that under homeostatic conditions, the proportion of pDC expressing C-C chemokine receptor 9 (CCR9) in the intestinal intraepithelial cell (iIEC) population is comparable between young (2-4 months) and aged (18-24 months) mice, but the absolute numbers of iIEC and pDC are significantly lower in aged mice. Employing the classic model of acute endotoxemia induced by lipopolysaccharide (LPS), we found a decrease in the proportion and absolute number of intraepithelial pDC in both young and aged mice despite the LPS-induced increased expression of the chemokine C-C ligand 25 (CCL25), the ligand of CCR9, in the intestinal mucosa of young mice. In adoptive transfer experiments, a significantly lower number of pDC was retained into the intestinal layer of aged host mice after LPS administration. This was associated with recoverable pDC numbers in the intestinal lumen. Furthermore, co-adoptive transfer of young and aged pDC into young hosts also showed significantly lower retention of aged pDC in the epithelial layer compared to the co-transferred young pDC. Collectively, these data show age-associated changes in mucosal CCL25 gene expression and in pDC number. These may underlie the reported inadequate responses to gastrointestinal pathogens during chronologic aging.
Collapse
Affiliation(s)
| | - Sulan Huang
- Department of Health Promotion and Development,
| | | | - Abbe N Vallejo
- Division of Pediatric Rheumatology, Department of Pediatrics, University of Pittsburgh,
- Division of Rheumatology, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA.
| |
Collapse
|
8
|
Prostaglandin E 2 Enhances Aged Hematopoietic Stem Cell Function. Stem Cell Rev Rep 2021; 17:1840-1854. [PMID: 33974233 DOI: 10.1007/s12015-021-10177-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2021] [Indexed: 10/21/2022]
Abstract
Aging of hematopoiesis is associated with increased frequency and clonality of hematopoietic stem cells (HSCs), along with functional compromise and myeloid bias, with donor age being a significant variable in survival after HSC transplantation. No clinical methods currently exist to enhance aged HSC function, and little is known regarding how aging affects molecular responses of HSCs to biological stimuli. Exposure of HSCs from young fish, mice, nonhuman primates, and humans to 16,16-dimethyl prostaglandin E2 (dmPGE2) enhances transplantation, but the effect of dmPGE2 on aged HSCs is unknown. Here we show that ex vivo pulse of bone marrow cells from young adult (3 mo) and aged (25 mo) mice with dmPGE2 prior to serial competitive transplantation significantly enhanced long-term repopulation from aged grafts in primary and secondary transplantation (27 % increase in chimerism) to a similar degree as young grafts (21 % increase in chimerism; both p < 0.05). RNA sequencing of phenotypically-isolated HSCs indicated that the molecular responses to dmPGE2 are similar in young and old, including CREB1 activation and increased cell survival and homeostasis. Common genes within these pathways identified likely key mediators of HSC enhancement by dmPGE2 and age-related signaling differences. HSC expression of the PGE2 receptor EP4, implicated in HSC function, increased with age in both mRNA and surface protein. This work suggests that aging does not alter the major dmPGE2 response pathways in HSCs which mediate enhancement of both young and old HSC function, with significant implications for expanding the therapeutic potential of elderly HSC transplantation.
Collapse
|
9
|
Mendonça de Pontes R, Flores-Montero J, Sanoja-Flores L, Puig N, Pessoa de Magalhães RJ, Corral-Mateos A, Salgado AB, García-Sánchez O, Pérez-Morán J, Mateos MV, Burgos L, Paiva B, te Marvelde J, van der Velden VHJ, Aguilar C, Bárez A, García-Mateo A, Labrador J, Leoz P, Aguilera-Sanz C, Durie B, van Dongen JJM, Maiolino A, Sobral da Costa E, Orfao A. B-Cell Regeneration Profile and Minimal Residual Disease Status in Bone Marrow of Treated Multiple Myeloma Patients. Cancers (Basel) 2021; 13:cancers13071704. [PMID: 33916787 PMCID: PMC8038337 DOI: 10.3390/cancers13071704] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/27/2021] [Accepted: 03/29/2021] [Indexed: 11/16/2022] Open
Abstract
B-cell regeneration during therapy has been considered as a strong prognostic factor in multiple myeloma (MM). However, the effects of therapy and hemodilution in bone marrow (BM) B-cell recovery have not been systematically evaluated during follow-up. MM (n = 177) and adult (≥50y) healthy donor (HD; n = 14) BM samples were studied by next-generation flow (NGF) to simultaneously assess measurable residual disease (MRD) and residual normal B-cell populations. BM hemodilution was detected in 41 out of 177 (23%) patient samples, leading to lower total B-cell, B-cell precursor (BCP) and normal plasma cell (nPC) counts. Among MM BM, decreased percentages (vs. HD) of BCP, transitional/naïve B-cell (TBC/NBC) and nPC populations were observed at diagnosis. BM BCP increased after induction therapy, whereas TBC/NBC counts remained abnormally low. At day+100 postautologous stem cell transplantation, a greater increase in BCP with recovered TBC/NBC cell numbers but persistently low memory B-cell and nPC counts were found. At the end of therapy, complete response (CR) BM samples showed higher CD19- nPC counts vs. non-CR specimens. MRD positivity was associated with higher BCP and nPC percentages. Hemodilution showed a negative impact on BM B-cell distribution. Different BM B-cell regeneration profiles are present in MM at diagnosis and after therapy with no significant association with patient outcome.
Collapse
Affiliation(s)
- Robéria Mendonça de Pontes
- Internal Medicine Postgraduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil; (R.M.d.P.); (A.B.S.); (A.M.); (E.S.d.C.)
- Cytometry Service, Institute of Paediatrics and Puericultura Martagão Gesteira (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-912, Brazil
| | - Juan Flores-Montero
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (J.F.-M.); (A.C.-M.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) (CB16/12/00400, CB16/12/00233, CB16/12/00369, CB16/12/00489 and CB16/12/00480), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (O.G.-S.); (J.P.-M.); (M.-V.M.); (L.B.); (B.P.); (P.L.)
| | - Luzalba Sanoja-Flores
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) (CB16/12/00400, CB16/12/00233, CB16/12/00369, CB16/12/00489 and CB16/12/00480), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (O.G.-S.); (J.P.-M.); (M.-V.M.); (L.B.); (B.P.); (P.L.)
- Institute of Biomedicine of Seville, Department of Hematology, University Hospital Virgen del Rocío of the Consejo Superior de Investigaciones Científicas (CSIC), University of Seville, 41013 Seville, Spain
| | - Noemi Puig
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) (CB16/12/00400, CB16/12/00233, CB16/12/00369, CB16/12/00489 and CB16/12/00480), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (O.G.-S.); (J.P.-M.); (M.-V.M.); (L.B.); (B.P.); (P.L.)
- Service of Hematology, University Hospital of Salamanca (USAL) and IBSAL, 37007 Salamanca, Spain
| | - Roberto J. Pessoa de Magalhães
- Department of Internal Medicine, University Hospital Clementino Fraga Filho, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil;
| | - Alba Corral-Mateos
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (J.F.-M.); (A.C.-M.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) (CB16/12/00400, CB16/12/00233, CB16/12/00369, CB16/12/00489 and CB16/12/00480), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (O.G.-S.); (J.P.-M.); (M.-V.M.); (L.B.); (B.P.); (P.L.)
| | - Anna Beatriz Salgado
- Internal Medicine Postgraduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil; (R.M.d.P.); (A.B.S.); (A.M.); (E.S.d.C.)
| | - Omar García-Sánchez
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) (CB16/12/00400, CB16/12/00233, CB16/12/00369, CB16/12/00489 and CB16/12/00480), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (O.G.-S.); (J.P.-M.); (M.-V.M.); (L.B.); (B.P.); (P.L.)
- Service of Hematology, University Hospital of Salamanca (USAL) and IBSAL, 37007 Salamanca, Spain
| | - José Pérez-Morán
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) (CB16/12/00400, CB16/12/00233, CB16/12/00369, CB16/12/00489 and CB16/12/00480), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (O.G.-S.); (J.P.-M.); (M.-V.M.); (L.B.); (B.P.); (P.L.)
- Service of Hematology, University Hospital of Salamanca (USAL) and IBSAL, 37007 Salamanca, Spain
| | - Maria-Victoria Mateos
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) (CB16/12/00400, CB16/12/00233, CB16/12/00369, CB16/12/00489 and CB16/12/00480), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (O.G.-S.); (J.P.-M.); (M.-V.M.); (L.B.); (B.P.); (P.L.)
- Service of Hematology, University Hospital of Salamanca (USAL) and IBSAL, 37007 Salamanca, Spain
| | - Leire Burgos
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) (CB16/12/00400, CB16/12/00233, CB16/12/00369, CB16/12/00489 and CB16/12/00480), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (O.G.-S.); (J.P.-M.); (M.-V.M.); (L.B.); (B.P.); (P.L.)
- Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Bruno Paiva
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) (CB16/12/00400, CB16/12/00233, CB16/12/00369, CB16/12/00489 and CB16/12/00480), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (O.G.-S.); (J.P.-M.); (M.-V.M.); (L.B.); (B.P.); (P.L.)
- Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Jeroen te Marvelde
- Department of Immunology, Erasmus MC, (EMC) University Medical Center Rotterdam, 3015 GA Rotterdam, The Netherlands; (J.t.M.); (V.H.J.v.d.V.)
| | - Vincent H. J. van der Velden
- Department of Immunology, Erasmus MC, (EMC) University Medical Center Rotterdam, 3015 GA Rotterdam, The Netherlands; (J.t.M.); (V.H.J.v.d.V.)
| | - Carlos Aguilar
- Department of Hematology, Hospital General de Santa Bárbara, 42005 Soria, Spain;
| | - Abelardo Bárez
- Department of Hematology, Complejo Asistencial de Ávila, 05071 Ávila, Spain;
| | | | - Jorge Labrador
- Department of Hematology and Research Unit, Hospital Universitario de Burgos, 09006 Burgos, Spain;
| | - Pilar Leoz
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) (CB16/12/00400, CB16/12/00233, CB16/12/00369, CB16/12/00489 and CB16/12/00480), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (O.G.-S.); (J.P.-M.); (M.-V.M.); (L.B.); (B.P.); (P.L.)
- Service of Hematology, University Hospital of Salamanca (USAL) and IBSAL, 37007 Salamanca, Spain
| | | | - Brian Durie
- Cedars-Sinai Samuel Oschin Cancer Center, Los Angeles, CA 90048, USA;
| | - Jacques J. M. van Dongen
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Correspondence: (J.J.M.v.D.); (A.O.); Tel.: +31-71-526-5128 (J.J.M.v.D.); +34-923-294811 (A.O.)
| | - Angelo Maiolino
- Internal Medicine Postgraduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil; (R.M.d.P.); (A.B.S.); (A.M.); (E.S.d.C.)
- Department of Internal Medicine, University Hospital Clementino Fraga Filho, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil;
- Americas Centro de Oncologia Integrado, Rio de Janeiro 22290-030, Brazil
| | - Elaine Sobral da Costa
- Internal Medicine Postgraduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil; (R.M.d.P.); (A.B.S.); (A.M.); (E.S.d.C.)
- Cytometry Service, Institute of Paediatrics and Puericultura Martagão Gesteira (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-912, Brazil
| | - Alberto Orfao
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (J.F.-M.); (A.C.-M.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) (CB16/12/00400, CB16/12/00233, CB16/12/00369, CB16/12/00489 and CB16/12/00480), Instituto Carlos III, 28029 Madrid, Spain; (L.S.-F.); (N.P.); (O.G.-S.); (J.P.-M.); (M.-V.M.); (L.B.); (B.P.); (P.L.)
- Correspondence: (J.J.M.v.D.); (A.O.); Tel.: +31-71-526-5128 (J.J.M.v.D.); +34-923-294811 (A.O.)
| |
Collapse
|
10
|
Lavazza A, Garasic M. Vampires 2.0? The ethical quandaries of young blood infusion in the quest for eternal life. MEDICINE, HEALTH CARE, AND PHILOSOPHY 2020; 23:421-432. [PMID: 32447568 DOI: 10.1007/s11019-020-09952-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Can transfusions of blood plasma slow down ageing or even rejuvenate people? Recent preclinical studies and experimental tests inspired by the technique known as parabiosis have aroused great media attention, although for now there is no clear evidence of their effectiveness. This line of research and the interest it is triggering testify to the prominent role played by the idea of combating the "natural" ageing process in the scientific and social agenda. While seeking to increase the duration of healthy living time may be considered a duty, it also raises ethical questions about how to pursue this goal. Specifically, therapies and techniques accessible only to a fraction of the population seem destined to exponentially increase social inequality and to produce undesirable consequences. In this article we address the issue precisely in the light of the prospected use of plasma for the rejuvenation of a small elite of people.
Collapse
Affiliation(s)
- Andrea Lavazza
- Centro Universitario Internazionale, via Garbasso 42, 52100, Arezzo, Italy.
| | - Mirko Garasic
- IMT School for Advanced Studies, Lucca, Italy
- Lumsa University, Rome, Italy
| |
Collapse
|
11
|
Häussinger D, Kordes C. Space of Disse: a stem cell niche in the liver. Biol Chem 2020; 401:81-95. [PMID: 31318687 DOI: 10.1515/hsz-2019-0283] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023]
Abstract
Recent evidence indicates that the plasticity of preexisting hepatocytes and bile duct cells is responsible for the appearance of intermediate progenitor cells capable of restoring liver mass after injury without the need of a stem cell compartment. However, mesenchymal stem cells (MSCs) exist in all organs and are associated with blood vessels which represent their perivascular stem cell niche. MSCs are multipotent and can differentiate into several cell types and are known to support regenerative processes by the release of immunomodulatory and trophic factors. In the liver, the space of Disse constitutes a stem cell niche that harbors stellate cells as liver resident MSCs. This perivascular niche is created by extracellular matrix proteins, sinusoidal endothelial cells, liver parenchymal cells and sympathetic nerve endings and establishes a microenvironment that is suitable to maintain stellate cells and to control their fate. The stem cell niche integrity is important for the behavior of stellate cells in the normal, regenerative, aged and diseased liver. The niche character of the space of Disse may further explain why the liver can become an organ of extra-medullar hematopoiesis and why this organ is frequently prone to tumor metastasis.
Collapse
Affiliation(s)
- Dieter Häussinger
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstraße 5, D-40225 Düsseldorf, Germany
| | - Claus Kordes
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstraße 5, D-40225 Düsseldorf, Germany
| |
Collapse
|
12
|
Mobarak H, Heidarpour M, Lolicato F, Nouri M, Rahbarghazi R, Mahdipour M. Physiological impact of extracellular vesicles on female reproductive system; highlights to possible restorative effects on female age-related fertility. Biofactors 2019; 45:293-303. [PMID: 30788863 DOI: 10.1002/biof.1497] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 01/03/2019] [Accepted: 01/28/2019] [Indexed: 12/15/2022]
Abstract
An alternative mechanism of cell-to-cell communication via extracellular vesicles (EVs) has recently raised increasing attention. EVs are spherical structures comprising exosomes and microvesicles, capable of transferring regulatory molecules and genetic information from one cell to another. EVs act as modulators which can alter a wide spectrum of functions at the cellular level in the recipient cells, taking part in a variety of biological processes in both physiological and pathological conditions. Alteration in EVs content, notably exosomes, was reported during cellular senescence and in patients with age-related diseases. Most studies reported regulating the impacts of exosomes on fertility and pregnancy outcomes via their capability in carrying developmental signaling molecules like proteins, RNA cargos, influencing gene expressions, affecting growth, and development of embryos during aging. Alterations in the exosomal content and functions can influence the reproductive performance in human and animals as conveyors of senescence signals from outside of the cells. This review aimed to summarize evidence on the role of EVs on modulating fertility, early embryonic development, maternal-embryo crosstalk for the recognition, and maintenance of pregnancy during maternal aging. Advanced clinical studies are required to strengthen the findings that the benefit of exosomes can be extended to subjects undergoing reproductive aging. © 2019 BioFactors, 45(3):293-303, 2019.
Collapse
Affiliation(s)
- Halimeh Mobarak
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mohammad Heidarpour
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Francesca Lolicato
- Follicle Biology Laboratory, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
13
|
Konieczny J, Arranz L. Updates on Old and Weary Haematopoiesis. Int J Mol Sci 2018; 19:ijms19092567. [PMID: 30158459 PMCID: PMC6163425 DOI: 10.3390/ijms19092567] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/20/2018] [Accepted: 08/26/2018] [Indexed: 12/13/2022] Open
Abstract
Blood formation, or haematopoiesis, originates from haematopoietic stem cells (HSCs), whose functions and maintenance are regulated in both cell- and cell non-autonomous ways. The surroundings of HSCs in the bone marrow create a specific niche or microenvironment where HSCs nest that allows them to retain their unique characteristics and respond rapidly to external stimuli. Ageing is accompanied by reduced regenerative capacity of the organism affecting all systems, due to the progressive decline of stem cell functions. This includes blood and HSCs, which contributes to age-related haematological disorders, anaemia, and immunosenescence, among others. Furthermore, chronological ageing is characterised by myeloid and platelet HSC skewing, inflammageing, and expanded clonal haematopoiesis, which may be the result of the accumulation of preleukaemic lesions in HSCs. Intriguingly, haematological malignancies such as acute myeloid leukaemia have a high incidence among elderly patients, yet not all individuals with clonal haematopoiesis develop leukaemias. Here, we discuss recent work on these aspects, their potential underlying molecular mechanisms, and the first cues linking age-related changes in the HSC niche to poor HSC maintenance. Future work is needed for a better understanding of haematopoiesis during ageing. This field may open new avenues for HSC rejuvenation and therapeutic strategies in the elderly.
Collapse
Affiliation(s)
- Joanna Konieczny
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT, The Arctic University of Norway, 9019 Tromsø, Norway.
| | - Lorena Arranz
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT, The Arctic University of Norway, 9019 Tromsø, Norway.
- Department of Hematology, University Hospital of North Norway, 9019 Tromsø, Norway.
- Young Associate Investigator, Norwegian Center for Molecular Medicine (NCMM), 0349 Oslo, Norway.
| |
Collapse
|
14
|
A virus-acquired host cytokine controls systemic aging by antagonizing apoptosis. PLoS Biol 2018; 16:e2005796. [PMID: 30036358 PMCID: PMC6072105 DOI: 10.1371/journal.pbio.2005796] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 08/02/2018] [Accepted: 07/11/2018] [Indexed: 12/31/2022] Open
Abstract
Aging is characterized by degeneration of unique tissues. However, dissecting the interconnectedness of tissue aging remains a challenge. Here, we employ a muscle-specific DNA damage model in Drosophila to reveal secreted factors that influence systemic aging in distal tissues. Utilizing this model, we uncovered a cytokine—Diedel—that, when secreted from muscle or adipose, can attenuate age-related intestinal tissue degeneration by promoting proliferative homeostasis of stem cells. Diedel is both necessary and sufficient to limit tissue degeneration and regulate lifespan. Secreted homologs of Diedel are also found in viruses, having been acquired from host genomes. Focusing on potential mechanistic overlap between cellular aging and viral-host cell interactions, we found that Diedel is an inhibitor of apoptosis and can act as a systemic rheostat to modulate cell death during aging. These results highlight a key role for secreted antagonists of apoptosis in the systemic coordination of tissue aging. Aging in multicellular organisms is characterized by a progressive decline in the proper function of organs. This deterioration of organ function is a risk factor for many diseases. However, it is unlikely that organs age in isolation, as damage in one organ can presumably impact aging of other organs through either beneficial or detrimental cross-talk. Our work attempts to explore this aspect of aging using fruit flies as a model system. We uncovered that damaged fly muscle can protect against aging in other organs, such as the intestine, through the secretion of a blood-borne factor named Diedel. This blood-borne factor presumably allows damaged organs to communicate with each other during aging. Related factors are also found in certain viruses, which have been hijacked from insect genomes to promote viral spreading during infection. Using this information, we found that viral Diedel inhibits death of infected cells, allowing viruses to spread. Similarly, host (insect) Diedel also blocks cell death in organs during aging, thus limiting deterioration of organ function and extending the organism’s lifespan.
Collapse
|
15
|
Yang M, Teng S, Ma C, Yu Y, Wang P, Yi C. Ascorbic acid inhibits senescence in mesenchymal stem cells through ROS and AKT/mTOR signaling. Cytotechnology 2018; 70:1301-1313. [PMID: 29777434 DOI: 10.1007/s10616-018-0220-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/09/2018] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cell (MSC) aging seriously affects its function in stem cell transplantation for treatment. Extensive studies have focused on how to inhibit senescence in MSCs. However, the mechanism of senescence in MSC was not clear. In this study, we used D-galactose to induce MSC aging. Then we found that the number of aging cells was increased compared with untreated MSCs. We discovered that ascorbic acid could inhibit the production of reactive oxygen species (ROS) and activation of AKT/mTOR signaling in MSCs caused by D-galactose. Especially, when treated together with a ROS scavenger or AKT inhibitor, the senescent cells were obviously decreased in D-galactose-induced MSCs. Taken together, we identify that ascorbic acid owns the potential to inhibit the senescence of MSCs through ROS and Akt/mTOR signaling. Together, our data supports that ascorbic acid can be used to prevent MSCs from senescence, which can enhance the efficiency of stem cell transplantation in the clinic.
Collapse
Affiliation(s)
- Mengkai Yang
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Songsong Teng
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Chunhui Ma
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yinxian Yu
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Peilin Wang
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Chengqing Yi
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
16
|
Abstract
Abstract
Hematopoietic stem cells (HSCs) ensure a balanced production of all blood cells throughout life. As they age, HSCs gradually lose their self-renewal and regenerative potential, whereas the occurrence of cellular derailment strongly increases. Here we review our current understanding of the molecular mechanisms that contribute to HSC aging. We argue that most of the causes that underlie HSC aging result from cell-intrinsic pathways, and reflect on which aspects of the aging process may be reversible. Because many hematological pathologies are strongly age-associated, strategies to intervene in aspects of the stem cell aging process may have significant clinical relevance.
Collapse
|
17
|
Xie H, Zhao S, Liu S. Aging of Human Adult Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1086:105-115. [PMID: 30232755 DOI: 10.1007/978-981-13-1117-8_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
With the continuous development of stem cell research in recent years, it is realized that stem cell aging may be the core issue of organ aging. As an important approach and main content of regenerative medicine, the stem cell research brings great hope to overcome difficult diseases and improve the quality of life for human beings and become the key to solve this issue. Based on this research, the varying characteristics of stem cells in aging could be recognized; the role of stem cells in the organ aging and regeneration will be revealed; the function of stem cells will be controllable and regulatable in tissues and organs; the stem cells from tissues and organs with rapid or slow cell renewal (e.g., liver and neuron) could be continuously observed from the levels of cellular molecules and dynamic complex. With the assistance of systematical research approaches, the function and mechanism studies can be conducted via multi-perspectives and levels during the different stages of organ aging and regeneration. All of the abovementioned requires great efforts to thoroughly understand the basic rule and the way of stem cell regulation in organ aging and regeneration. Final to the end, the dream of antiaging, efficient repair, and organ remodeling could be realized and also can meet the major needs of population health and disease treatment in our country, meaningfully to contribute benefits for the health of human beings.
Collapse
Affiliation(s)
- Han Xie
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Shouliang Zhao
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Shangfeng Liu
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
18
|
Brooks RW, Robbins PD. Treating Age-Related Diseases with Somatic Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1056:29-45. [DOI: 10.1007/978-3-319-74470-4_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
19
|
Conese M, Carbone A, Beccia E, Angiolillo A. The Fountain of Youth: A Tale of Parabiosis, Stem Cells, and Rejuvenation. Open Med (Wars) 2017; 12:376-383. [PMID: 29104943 PMCID: PMC5662775 DOI: 10.1515/med-2017-0053] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 06/19/2017] [Indexed: 01/10/2023] Open
Abstract
Transfusion (or drinking) of blood or of its components has been thought as a rejuvenation method since ancient times. Parabiosis, the procedure of joining two animals so that they share each others blood circulation, has revitalized the concept of blood as a putative drug. Since 2005, a number of papers have reported the anti-ageing effect of heterochronic parabiosis, which is joining an aged mouse to a young partner. The hallmark of aging is the decline of regenerative properties in most tissues, partially attributed to impaired function of stem and progenitor cells. In the parabiosis experiments, it was elegantly shown that factors derived from the young systemic environment are able to activate molecular signaling pathways in hepatic, muscle or neural stem cells of the old parabiont leading to increased tissue regeneration. Eventually, further studies have brought to identify some soluble factors in part responsible for these rejuvenating effects, including the chemokine CCL11, the growth differentiation factor 11, a member of the TGF-β superfamily, and oxytocin. The question about giving whole blood or specific factors in helping rejuvenation is open, as well as the mechanisms of action of these factors, deserving further studies to be translated into the life of (old) human beings.
Collapse
Affiliation(s)
- Massimo Conese
- Biomedical Research Center "E. Altomare", Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, c/o Ospedali Riuniti, Via L. Pinto 1, 71122, Tel.: +39 0881 588014; ;Foggia, Italy
| | - Annalucia Carbone
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Elisa Beccia
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.,Dipartimento di Medicina e Scienze della Salute "V. Tiberio", University of Molise, Campobasso, Italy
| | - Antonella Angiolillo
- Dipartimento di Medicina e Scienze della Salute "V. Tiberio", University of Molise, Campobasso, Italy
| |
Collapse
|
20
|
Sunku CC, Gadi VK, de Laval de Lacoste B, Guthrie KA, Nelson JL. Maternal and fetal microchimerism in granulocytes. CHIMERISM 2017; 1:11-4. [PMID: 21327147 DOI: 10.4161/chim.1.1.13098] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 07/19/2010] [Accepted: 07/21/2010] [Indexed: 12/25/2022]
Abstract
Cell trafficking during pregnancy may result in durable microchimerism, both fetal microchimerism in the mother and maternal microchimerism in her children. Whether microchimerism is continuously replenished has not been well-described. To address this question, we isolated granulocytes, cells with relatively short half-lives, from peripheral blood of healthy women. CD66b-positive cells were isolated by fluorescence activated cell sorting and a panel of polymorphism-specific quantitative pCR assays was employed to investigate fetal and maternal microchimerism. Overall 33% (10/30) of study subjects had at least one source of microchimerism in CD66b(+) cells. Interestingly, maternal microchimerism was more common than fetal microchimerism, 40% vs. 15%, respectively (p = 0.05) and was present at higher levels (p = 0.03). The identification of maternal and fetal origin CD66b(+) cells is strong evidence for an active microchimeric hematopoietic stem and progenitor cell niche. Furthermore, microchimeric CD66b(+) cells could have an impact on innate and adaptive immune responses.
Collapse
|
21
|
Williams KM, Moore AR, Lucas PJ, Wang J, Bare CV, Gress RE. FLT3 ligand regulates thymic precursor cells and hematopoietic stem cells through interactions with CXCR4 and the marrow niche. Exp Hematol 2017; 52:40-49. [PMID: 28552733 DOI: 10.1016/j.exphem.2017.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 05/05/2017] [Accepted: 05/06/2017] [Indexed: 01/07/2023]
Abstract
Impaired immune reconstitution after hematopoietic stem cell transplantation (HSCT) is attributed in part to impaired thymopoiesis. Recent data suggest that precursor input may be a point of regulation for the thymus. We hypothesized that administration of FLT3 ligand (FLT3L) would enhance thymopoiesis after adoptive transfer of aged, FLT3L-treated bone marrow (BM) to aged, Lupron-treated hosts by increasing murine HSC (Lin[minus]Sca1+c-Kit+ [LSK] cells) trafficking and survival. In murine models of aged and young hosts, we show that FLT3L enhances thymopoiesis in aged, Lupron-treated hosts through increased survival and export of LSK cells via CXCR4 regulation. In addition, we elucidate an underlying mechanism of FLT3L action on BM LSK cells-FLT3L drives LSK cells into the stromal niche using Hoescht (Ho) dye perimortem. In summary, we show that FLT3L administration leads to: (1) increased LSK cells and early thymocyte progenitor precursors that can enhance thymopoiesis after transplantation and androgen withdrawal, (2) mobilization of LSK cells through downregulation of CXCR4, (3) enhanced BM stem cell survival associated with Bcl-2 upregulation, and (4) BM stem cell enrichment through increased trafficking to the BM niche. Therefore, we show a mechanism by which FLT3L activity on hematopoeitic and thymic progenitor cells may contribute to thymic recovery. These data have potential clinical relevance to enhance thymic reconstitution after cytoreductive therapy.
Collapse
Affiliation(s)
- Kirsten M Williams
- Children's Research Institute, Children's National Medical Institutes, Washington, DC.
| | - Amber R Moore
- Stanford Immunology, Stanford University School of Medicine, Stanford, CA
| | - Philip J Lucas
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Juin Wang
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Catherine V Bare
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ronald E Gress
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
22
|
Bickford PC, Flowers A, Grimmig B. Aging leads to altered microglial function that reduces brain resiliency increasing vulnerability to neurodegenerative diseases. Exp Gerontol 2017; 94:4-8. [PMID: 28163132 DOI: 10.1016/j.exger.2017.01.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 01/27/2017] [Accepted: 01/31/2017] [Indexed: 11/26/2022]
Abstract
Aging is the primary risk factor for many neurodegenerative diseases. Thus, understanding the basic biological changes that take place with aging that lead to the brain being less resilient to disease progression of neurodegenerative diseases such as Parkinson's disease or Alzheimer's disease or insults to the brain such as stroke or traumatic brain injuries. Clearly this will not cure the disease per se, yet increasing the ability of the brain to respond to injury could improve long term outcomes. The focus of this review is examining changes in microglia with age and possible therapeutic interventions involving the use of polyphenol rich dietary supplements.
Collapse
Affiliation(s)
- Paula C Bickford
- Research Service, James A. Haley Veterans Administration Hospital, 13000 Bruce B Downs Blvd, Tampa, FL, United States; Center of Excellence for Aging and Brain Repair, Dept. of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.
| | - Antwoine Flowers
- Center of Excellence for Aging and Brain Repair, Dept. of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Bethany Grimmig
- Center of Excellence for Aging and Brain Repair, Dept. of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
23
|
Kramer A, Challen GA. The epigenetic basis of hematopoietic stem cell aging. Semin Hematol 2016; 54:19-24. [PMID: 28088983 DOI: 10.1053/j.seminhematol.2016.10.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 10/19/2016] [Accepted: 10/19/2016] [Indexed: 12/12/2022]
Abstract
Highly proliferative tissues such as the gut, skin, and bone marrow lose millions of cells each day to normal attrition and challenge from different biological adversities. To achieve a lifespan beyond the longevity of individual cell types, tissue-specific stem cells sustain these tissues throughout the life of a human. For example, the lifespan of erythrocytes is about 100 days and adults make about two million new erythrocytes every second. A small pool of hematopoietic stem cells (HSCs) in the bone marrow is responsible for the lifetime maintenance of these populations. However, there are changes that occur within the HSC pool during aging. Biologically, these changes manifest as blunted immune responses, decreased bone marrow cellularity, and increased risk of myeloid diseases. Understanding the molecular mechanisms underlying dysfunction of aging HSCs is an important focus of biomedical research. With advances in modern health care, the average age of the general population is ever increasing. If molecular or pharmacological interventions could be discovered that rejuvenate aging HSCs, it could reduce the burden of age related immune system compromise as well as open up new opportunities for treatment of hematological disorders with regenerative therapy.
Collapse
Affiliation(s)
- Ashley Kramer
- Section of Stem Cell Biology, Division of Oncology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO
| | - Grant A Challen
- Section of Stem Cell Biology, Division of Oncology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO; Developmental, Regenerative and Stem Cell Biology Program, Division of Biology and Biomedical Sciences, Washington University in St. Louis School of Medicine, St. Louis, MO.
| |
Collapse
|
24
|
Yao JC, Link DC. Concise Review: The Malignant Hematopoietic Stem Cell Niche. Stem Cells 2016; 35:3-8. [PMID: 27647718 DOI: 10.1002/stem.2487] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/26/2016] [Accepted: 08/17/2016] [Indexed: 12/27/2022]
Abstract
Hematopoietic stem cell (HSC) proliferation, self-renewal, and trafficking are dependent, in part, upon signals generated by stromal cells in the bone marrow. Stromal cells are organized into niches that support specific subsets of hematopoietic progenitors. There is emerging evidence that malignant hematopoietic cells may generate signals that alter the number and/or function of specific stromal cell populations in the bone marrow. At least in some cases, the resulting alterations in the bone marrow microenvironment confer a competitive advantage to the malignant HSC and progenitor cells and/or render them less sensitive to chemotherapy. Targeting these signals represents a promising therapeutic strategy for selected hematopoietic malignancies. In this review, we focus on two questions. How do alterations in bone marrow stromal cells arise in hematopoietic malignancies, and how do they contribute to disease pathogenesis? Stem Cells 2017;35:3-8.
Collapse
Affiliation(s)
- Juo-Chin Yao
- Departments of Medicine and Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Daniel C Link
- Departments of Medicine and Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
25
|
Rawji KS, Mishra MK, Michaels NJ, Rivest S, Stys PK, Yong VW. Immunosenescence of microglia and macrophages: impact on the ageing central nervous system. Brain 2016; 139:653-61. [PMID: 26912633 DOI: 10.1093/brain/awv395] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 11/18/2015] [Indexed: 01/21/2023] Open
Abstract
Ageing of the central nervous system results in a loss of both grey and white matter, leading to cognitive decline. Additional injury to both the grey and white matter is documented in many neurological disorders with ageing, including Alzheimer's disease, traumatic brain and spinal cord injury, stroke, and multiple sclerosis. Accompanying neuronal and glial damage is an inflammatory response consisting of activated macrophages and microglia, innate immune cells demonstrated to be both beneficial and detrimental in neurological repair. This article will propose the following: (i) infiltrating macrophages age differently from central nervous system-intrinsic microglia; (ii) several mechanisms underlie the differential ageing process of these two distinct cell types; and (iii) therapeutic strategies that selectively target these diverse mechanisms may rejuvenate macrophages and microglia for repair in the ageing central nervous system. Most responses of macrophages are diminished with senescence, but activated microglia increase their expression of pro-inflammatory cytokines while diminishing chemotactic and phagocytic activities. The senescence of macrophages and microglia has a negative impact on several neurological diseases, and the mechanisms underlying their age-dependent phenotypic changes vary from extrinsic microenvironmental changes to intrinsic changes in genomic integrity. We discuss the negative effects of age on neurological diseases, examine the response of senescent macrophages and microglia in these conditions, and propose a theoretical framework of therapeutic strategies that target the different mechanisms contributing to the ageing phenotype in these two distinct cell types. Rejuvenation of ageing macrophage/microglia may preserve neurological integrity and promote regeneration in the ageing central nervous system.
Collapse
Affiliation(s)
- Khalil S Rawji
- 1 Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Manoj K Mishra
- 1 Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Nathan J Michaels
- 1 Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Serge Rivest
- 2 Neuroscience Laboratory, CHU de Québec Research Centre, Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec City, Québec G1V 4G2, Canada
| | - Peter K Stys
- 1 Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - V Wee Yong
- 1 Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
26
|
Mitteldorf J. An epigenetic clock controls aging. Biogerontology 2015; 17:257-65. [DOI: 10.1007/s10522-015-9617-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Accepted: 10/07/2015] [Indexed: 12/22/2022]
|
27
|
Kokabu S, Tsuchiya-Hirata S, Fukushima H, Sugiyama G, Lowery JW, Katagiri T, Jimi E. Inhibition of bone morphogenetic protein-induced osteoblast differentiation. J Oral Biosci 2015. [DOI: 10.1016/j.job.2015.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
28
|
Shipounova IN, Petinati NA, Bigildeev AE, Zezina EA, Drize NI, Kuzmina LA, Parovichnikova EN, Savchenko VG. Analysis of results of acute graft-versus-host disease prophylaxis with donor multipotent mesenchymal stromal cells in patients with hemoblastoses after allogeneic bone marrow transplantation. BIOCHEMISTRY (MOSCOW) 2015; 79:1363-70. [PMID: 25716730 DOI: 10.1134/s0006297914120104] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Allogeneic bone marrow transplantation (allo-BMT) is currently the only way to cure many hematoproliferative disorders. However, allo-BMT use is limited by severe complications, the foremost being graft-versus-host disease (GVHD). Due to the lack of efficiency of the existing methods of GVHD prophylaxis, new methods are being actively explored, including the use of donors' multipotent mesenchymal stromal cells (MMSC). In this work, we analyzed the results of acute GVHD (aGVHD) prophylaxis by means of MMSC injections after allo-BMT in patients with hematological malignancies. The study included 77 patients. They were randomized into two groups - those receiving standard prophylaxis of aGVHD and those who were additionally infused with MMSC derived from the bone marrow of hematopoietic stem cell donors. We found that the infusion of MMSC halves the incidence of aGVHD and increases the overall survival of patients. Four of 39 MMSC samples were ineffective for preventing aGVHD. Analysis of individual donor characteristics (gender, age, body mass index) and the MMSC properties of these donors (growth parameters, level of expression of 30 genes involved in proliferation, differentiation, and immunomodulation) revealed no significant difference between the MMSC that were effective or ineffective for preventing aGVHD. We used multiple logistic regression to establish a combination of features that characterize the most suitable MMSC samples for the prevention of aGVHD. A model predicting MMSC sample success for aGVHD prophylaxis was constructed. Significant model parameters were increased relative expression of the FGFR1 gene in combination with reduced expression levels of the PPARG and IGF1 genes. Depending on the chosen margin for probability of successful application of MMSC, this model correctly predicts the outcome of the use of MMSC in 82-94% of cases. The proposed model of prospective evaluation of the effectiveness of MMSC samples will enable prevention of the development of aGVHD in the maximal number of patients.
Collapse
Affiliation(s)
- I N Shipounova
- Hematological Scientific Center, Ministry of Health of Russian Federation, Moscow, 125167, Russia.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Bickford PC, Kaneko Y, Grimmig B, Pappas C, Small B, Sanberg CD, Sanberg PR, Tan J, Douglas Shytle R. Nutraceutical intervention reverses the negative effects of blood from aged rats on stem cells. AGE (DORDRECHT, NETHERLANDS) 2015; 37:103. [PMID: 26410618 PMCID: PMC5005857 DOI: 10.1007/s11357-015-9840-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/15/2015] [Indexed: 06/05/2023]
Abstract
Aging is associated with a decline in function in many of the stem cell niches of the body. An emerging body of literature suggests that one of the reasons for this decline in function is due to cell non-autonomous influences on the niche from the body. For example, studies using the technique of parabiosis have demonstrated a negative influence of blood from aged mice on muscle satellite cells and neurogenesis in young mice. We examined if we could reverse this effect of aged serum on stem cell proliferation by treating aged rats with NT-020, a dietary supplement containing blueberry, green tea, vitamin D3, and carnosine that has been shown to increase neurogenesis in aged rats. Young and aged rats were administered either control NIH-31 diet or one supplemented with NT-020 for 28 days, and serum was collected upon euthanasia. The serum was used in cultures of both rat hippocampal neural progenitor cells (NPCs) and rat bone marrow-derived mesenchymal stem cells (MSCs). Serum from aged rats significantly reduced cell proliferation as measured by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and 5-bromo-2'-deoxyuridine (BrdU) assays in both NPCs and MSCs. Serum from aged rats treated with NT-020 was not different from serum from young rats. Therefore, NT-020 rescued the effect of serum from aged rats to reduce stem cell proliferation.
Collapse
Affiliation(s)
- Paula C Bickford
- Research Service, James A Haley Veterans Affairs Hospital|, Tampa, FL, 33620, USA.
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, MDC-78, USF Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33620, USA.
| | - Yuji Kaneko
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, MDC-78, USF Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33620, USA
| | - Bethany Grimmig
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, MDC-78, USF Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33620, USA
- Department of Molecular Pharmacology and Physiology, USF Morsani College of Medicine, Tampa, FL, 33620, USA
| | | | - Brent Small
- School of Aging Studies, USF, Tampa, FL, 33620, USA
| | | | - Paul R Sanberg
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, MDC-78, USF Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33620, USA
| | - Jun Tan
- Department of Psychiatry, USF Morsani College of Medicine, Tampa, FL, 33620, USA
| | - R Douglas Shytle
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, MDC-78, USF Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33620, USA
| |
Collapse
|
30
|
Kuzmina LA, Petinati NA, Shipounova IN, Sats NV, Bigildeev AE, Zezina EA, Popova MD, Drize NJ, Parovichnikova EN, Savchenko VG. Analysis of multipotent mesenchymal stromal cells used for acute graft-versus-host disease prophylaxis. Eur J Haematol 2015; 96:425-34. [PMID: 26115424 DOI: 10.1111/ejh.12613] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Multipotent mesenchymal stromal cells (MSCs) are used for prophylaxis of acute graft-versus-host disease (aGvHD) after allogeneic hematopoietic cell transplantation (allo-HCT). Not all samples of MSC are efficient for aGvHD prevention. The suitability of MSCs for aGvHD prophylaxis was studied. METHODS MSCs were derived from the bone marrow (BM) of HCT donor and cultivated for no more than three passages. The characteristics of donor BM samples including colony-forming unit fibroblast (CFU-F) concentration, growth parameters of MSCs, and the relative expression levels (REL) of different genes were analyzed. MSCs were injected intravenously precisely at the moment of blood cell reconstitution. RESULTS MSCs infusion induced a significant threefold decrease in aGvHD development and improved overall survival compared with the standard prophylaxis group. In ineffective MSC samples (9.4%), a significant decrease in total cell production and the REL of CSF1, FGFR1, and PDGFRB was observed. In all studied BM samples, the cumulative MSC production and CFU-F concentrations decreased with age. The expression levels of FGFR2, PPARG, and VEGF differed by age. CONCLUSIONS A universal single indicator for the prediction of MSC eligibility for aGvHD prophylaxis was not identified. A multiparameter mathematical model for selecting MSC samples effective for the prevention of aGvHD was proposed.
Collapse
Affiliation(s)
- Larisa A Kuzmina
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russia
| | - Nataliya A Petinati
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russia
| | - Irina N Shipounova
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russia
| | - Natalia V Sats
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russia
| | - Alexey E Bigildeev
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russia
| | - Ekaterina A Zezina
- Department of Molecular Immunology, Faculty of Biology, Moscow State University, Moscow, Russia
| | - Maria D Popova
- Department of Molecular Immunology, Faculty of Biology, Moscow State University, Moscow, Russia
| | - Nina J Drize
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russia
| | - Elena N Parovichnikova
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russia
| | - Valery G Savchenko
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russia
| |
Collapse
|
31
|
Abstract
Stem cells persist in replenishing functional mature cells throughout life by self-renewal and multilineage differentiation. Hematopoietic stem cells (HSCs) are among the best-characterized and understood stem cells, and they are responsible for the life-long production of all lineages of blood cells. HSCs are a heterogeneous population containing lymphoid-biased, myeloid-biased, and balanced subsets. HSCs undergo age-associated phenotypic and functional changes, and the composition of the HSC pool alters with aging. HSCs and their lineage-biased subfractions can be identified and analyzed by flow cytometry based on cell surface makers. Fluorescence-activated cell sorting (FACS) enables the isolation and purification of HSCs that greatly facilitates the mechanistic study of HSCs and their aging process at both cellular and molecular levels. The mouse model has been extensively used in HSC aging study. Bone marrow cells are isolated from young and old mice and stained with fluorescence-conjugated antibodies specific for differentiated and stem cells. HSCs are selected based on the negative expression of lineage markers and positive selection for several sets of stem cell markers. Lineage-biased HSCs can be further distinguished by the level of SLAM/CD150 expression and the extent of Hoechst efflux.
Collapse
Affiliation(s)
- Yi Liu
- Division of Hematology/Bone Marrow Transplantation, Department of Internal Medicine, Markey Cancer Center, University of Kentucky, 800 Rose Street, Lexington, KY, 40536, USA
| | | | | |
Collapse
|
32
|
Abstract
The demonstrated presence in adult tissues of cells with sustained tissue regenerative potential has given rise to the concept of tissue stem cells. Assays to detect and measure such cells indicate that they have enormous proliferative potential and usually an ability to produce all or many of the mature cell types that define the specialized functionality of the tissue. In the hematopoietic system, one or only a few cells can restore lifelong hematopoiesis of the whole organism. To what extent is the maintenance of hematopoietic stem cells required during normal hematopoiesis? How does the constant maintenance of hematopoiesis occur and what is the behavior of the hematopoietic stem cells in the normal organism? How many of the hematopoietic stem cells are created during the development of the organism? How many hematopoietic stem cells are generating more mature progeny at any given moment? What happens to the population of hematopoietic stem cells in aging? This review will attempt to describe the results of recent research which contradict some of the ideas established over the past 30 years about how hematopoiesis is regulated.
Collapse
Affiliation(s)
- Nina Drize
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russian Federation
| | - Nataliya Petinati
- Federal Government Budget Institution National Research Center for Hematology, Ministry of Health, Moscow, Russian Federation
| |
Collapse
|
33
|
Liu HY, Huang CF, Lin TC, Tsai CY, Tina Chen SY, Liu A, Chen WH, Wei HJ, Wang MF, Williams DF, Deng WP. Delayed animal aging through the recovery of stem cell senescence by platelet rich plasma. Biomaterials 2014; 35:9767-9776. [DOI: 10.1016/j.biomaterials.2014.08.034] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 08/21/2014] [Indexed: 11/28/2022]
|
34
|
Hadjiargyrou M, O'Keefe RJ. The convergence of fracture repair and stem cells: interplay of genes, aging, environmental factors and disease. J Bone Miner Res 2014; 29:2307-22. [PMID: 25264148 PMCID: PMC4455538 DOI: 10.1002/jbmr.2373] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 08/11/2014] [Accepted: 09/10/2014] [Indexed: 01/07/2023]
Abstract
The complexity of fracture repair makes it an ideal process for studying the interplay between the molecular, cellular, tissue, and organ level events involved in tissue regeneration. Additionally, as fracture repair recapitulates many of the processes that occur during embryonic development, investigations of fracture repair provide insights regarding skeletal embryogenesis. Specifically, inflammation, signaling, gene expression, cellular proliferation and differentiation, osteogenesis, chondrogenesis, angiogenesis, and remodeling represent the complex array of interdependent biological events that occur during fracture repair. Here we review studies of bone regeneration in genetically modified mouse models, during aging, following environmental exposure, and in the setting of disease that provide insights regarding the role of multipotent cells and their regulation during fracture repair. Complementary animal models and ongoing scientific discoveries define an increasing number of molecular and cellular targets to reduce the morbidity and complications associated with fracture repair. Last, some new and exciting areas of stem cell research such as the contribution of mitochondria function, limb regeneration signaling, and microRNA (miRNA) posttranscriptional regulation are all likely to further contribute to our understanding of fracture repair as an active branch of regenerative medicine.
Collapse
Affiliation(s)
- Michael Hadjiargyrou
- Department of Life Sciences, New York Institute of Technology, Old Westbury, NY, USA
| | | |
Collapse
|
35
|
Khalyavkin AV, Krutko VN. Aging is a simple deprivation syndrome driven by a quasi-programmed preventable and reversible drift of control system set points due to inappropriate organism-environment interaction. BIOCHEMISTRY (MOSCOW) 2014; 79:1133-5. [DOI: 10.1134/s0006297914100150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
36
|
Geiger H, Zheng Y. Regulation of hematopoietic stem cell aging by the small RhoGTPase Cdc42. Exp Cell Res 2014; 329:214-9. [PMID: 25220425 DOI: 10.1016/j.yexcr.2014.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 09/01/2014] [Indexed: 11/16/2022]
Abstract
Aging of stem cells might be the underlying cause of tissue aging in tissue that in the adult heavily rely on stem cell activity, like the blood forming system. Hematopoiesis, the generation of blood forming cells, is sustained by hematopoietic stem cells. In this review article, we introduce the canonical set of phenotypes associated with aged HSCs, focus on the novel aging-associated phenotype apolarity caused by elevated activity of the small RhoGTPase in aged HSCs, discuss the role of Cdc42 in hematopoiesis and describe that pharmacological inhibition of Cdc42 activity in aged HSCs results in functionally young and thus rejuvenated HSCs.
Collapse
Affiliation(s)
- Hartmut Geiger
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children׳s Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA; Institute for Molecular Medicine, Stem Cells and Aging, Ulm University, Ulm 89091, Germany; aging research center, Ulm University, Ulm, Germany.
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children׳s Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
37
|
Abstract
As stem cells (SCs) in adult organs continue to be identified and characterized, it becomes clear that their survival, quiescence, and activation depend on specific signals in their microenvironment, or niche. Although adult SCs of diverse tissues differ by their developmental origin, cycling activity, and regenerative capacity, there appear to be conserved similarities regarding the cellular and molecular components of the SC niche. Interestingly, many organs house both slow-cycling and fast-cycling SC populations, which rely on the coexistence of quiescent and inductive niches for proper regulation. In this review we present a general definition of adult SC niches in the most studied mammalian systems. We further focus on dissecting their cellular organization and on highlighting recently identified key molecular regulators. Finally, we detail the potential involvement of the SC niche in tissue degeneration, with a particular emphasis on aging and cancer.
Collapse
Affiliation(s)
- Amélie Rezza
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Rachel Sennett
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Michael Rendl
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
38
|
Geiger H, Denkinger M, Schirmbeck R. Hematopoietic stem cell aging. Curr Opin Immunol 2014; 29:86-92. [PMID: 24905894 DOI: 10.1016/j.coi.2014.05.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 04/30/2014] [Accepted: 05/02/2014] [Indexed: 02/08/2023]
Abstract
Aging is organized in a hierarchy, in which aging of cells results in aged tissues, ultimately limiting lifespan. For organ systems that also in the adult depend on stem cells for tissue homeostasis like the hematopoietic system that forms immune cells, it is believed that aging of the stem cells strongly contributes to aging-associated dysfunction. In this review, we summarize current aspects on cellular and molecular mechanisms that are associated with aging of hematopoietic stem cells, the role of the stem cell niche for stem cell aging as well as novel and encouraging experimental approaches to attenuate aging of hematopoietic stem cells to target immunosenescence.
Collapse
Affiliation(s)
- Hartmut Geiger
- Institute for Molecular Medicine, Stem Cell and Aging, Ulm University, Ulm, Germany; Aging Research Center, Ulm University, Ulm, Germany; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH, USA.
| | - Michael Denkinger
- AGAPLESION Bethesda Clinic, Geriatric Center Ulm University, Ulm, Germany
| | | |
Collapse
|
39
|
Honoki K, Tsujiuchi T. Senescence bypass in mesenchymal stem cells: a potential pathogenesis and implications of pro-senescence therapy in sarcomas. Expert Rev Anticancer Ther 2014; 13:983-96. [PMID: 23984899 DOI: 10.1586/14737140.2013.820010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cellular senescence is a mechanism that limits the lifespan of somatic cells as the results of replicative proliferation and response to stresses, and that prevents undesired oncogenic changes constituting a barrier against immortalization and tumorigenesis. Mesenchymal stem cells (MSCs) reside in a variety of tissues, and participates in tissue maintenance with their multipotent differentiation ability. MSCs are also considered to be as cells of origin for certain type of sarcomas. We reviewed the mechanisms of cellular senescence in MSCs and hypothesized senescence bypass as the potential pathogenesis for sarcoma development, and proposed the possibility of senescence induction therapy for an alternative treatment strategy against sarcomas, especially cells with the resistance to conventional chemo and radiotherapy including sarcoma stem cells.
Collapse
Affiliation(s)
- Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan.
| | | |
Collapse
|
40
|
Shim J, Gururaja-Rao S, Banerjee U. Nutritional regulation of stem and progenitor cells in Drosophila. Development 2014; 140:4647-56. [PMID: 24255094 DOI: 10.1242/dev.079087] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Stem cells and their progenitors are maintained within a microenvironment, termed the niche, through local cell-cell communication. Systemic signals originating outside the niche also affect stem cell and progenitor behavior. This review summarizes studies that pertain to nutritional effects on stem and progenitor cell maintenance and proliferation in Drosophila. Multiple tissue types are discussed that utilize the insulin-related signaling pathway to convey nutritional information either directly to these progenitors or via other cell types within the niche. The concept of systemic control of these cell types is not limited to Drosophila and may be functional in vertebrate systems, including mammals.
Collapse
Affiliation(s)
- Jiwon Shim
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
41
|
Mazzoccoli G, Tevy MF, Borghesan M, Delle Vergini MR, Vinciguerra M. Caloric restriction and aging stem cells: the stick and the carrot? Exp Gerontol 2013; 50:137-48. [PMID: 24211426 DOI: 10.1016/j.exger.2013.10.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 09/03/2013] [Accepted: 10/28/2013] [Indexed: 12/24/2022]
Abstract
Adult tissue stem cells have the ability to adjust to environmental changes and affect also the proliferation of neighboring cells, with important consequences on tissue maintenance and regeneration. Stem cell renewal and proliferation is strongly regulated during aging of the organism. Caloric restriction is the most powerful anti-aging strategy conserved throughout evolution in the animal kingdom. Recent studies relate the properties of caloric restriction to its ability in reprogramming stem-like cell states and in prolonging the capacity of stem cells to self-renew, proliferate, differentiate, and replace cells in several adult tissues. However this general paradigm presents with exceptions. The scope of this review is to highlight how caloric restriction impacts on diverse stem cell compartments and, by doing so, might differentially delay aging in the tissues of lower and higher organisms.
Collapse
Affiliation(s)
- Gianluigi Mazzoccoli
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Unit, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", S. Giovanni Rotondo, FG, Italy.
| | - Maria Florencia Tevy
- Genomics and Bioinformatics Centre, Major University of Santiago, Santiago, Chile
| | - Michela Borghesan
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Unit, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", S. Giovanni Rotondo, FG, Italy; University College London, Institute for Liver and Digestive Health, Division of Medicine, Royal Free Campus, London, United Kingdom
| | - Maria Rita Delle Vergini
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Unit, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", S. Giovanni Rotondo, FG, Italy
| | - Manlio Vinciguerra
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Unit, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", S. Giovanni Rotondo, FG, Italy; Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy; University College London, Institute for Liver and Digestive Health, Division of Medicine, Royal Free Campus, London, United Kingdom.
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW Hematopoietic stem cells (HSCs) continuously provide mature blood cells during the lifespan of a mammal. The functional decline in hematopoiesis in the elderly, which involves a progressive reduction in the immune response and an increased incidence of myeloid malignancy, is partly linked to HSC aging. Molecular mechanisms of HSC aging remain unclear, hindering rational approaches to slow or reverse the decline of HSC function with age. Identifying conditions under which aged HSCs become equivalent to young stem cells might result in treatments for age-associated imbalances in lymphopoiesis and myelopoiesis and in blood regeneration. RECENT FINDINGS Aging of HSCs has been for a long time thought to be an irreversible process imprinted in stem cells due to the intrinsic nature of HSC aging. Mouse model studies have found that aging is associated with elevated activity of the Rho GTPase Cdc42 in HSCs that is causative for loss of polarity, altered epigenetic modifications and functional deficits of aged HSCs. The work suggests that inhibition of Cdc42 activity in aged HSCs may reverse a number of phenotypes associated with HSC aging. SUMMARY Maintaining the regenerative capacity of organs or organ systems may be a useful way to ensure healthy aging. A defined set of features phenotypically separate young from aged HSCs. Aging of HSCs has been thought to be irreversible. Recent findings support the hypothesis that functional decline of aged HSCs may be reversible by pharmacological intervention of age altered signaling pathways and epigenetic modifications.
Collapse
|
43
|
Long term human reconstitution and immune aging in NOD-Rag (-)-γ chain (-) mice. Immunobiology 2013; 219:131-7. [PMID: 24094417 DOI: 10.1016/j.imbio.2013.08.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 08/20/2013] [Accepted: 08/26/2013] [Indexed: 11/21/2022]
Abstract
Aging of the human immune system is characterized by a gradual loss of immune function and a skewing of hematopoiesis toward the myeloid lineage, a reduction in the lymphocytic lineage, and progressive increases in senescent memory T cells at the expense of naïve T cells. Both the innate and the adaptive branches of the immune system are affected, including neutrophils, macrophages, dendritic cells and lymphocytes. Mice, the most common research model, although inexpensive, do not necessarily reflect the human immune system in terms of its interaction with infectious agents of human origin or environmental factors. This study analyzed whether a human immune system contained within the NOD-Rag (-)-γ chain (-) mouse model could realistically be used to evaluate the development and therapy of aging-related diseases. To that end lightly irradiated NOD-Rag (-)-γ chain (-) mice were injected intra-hepatically on day 1 of life with purified cord blood-derived CD34(+) stem and progenitor cells. Multiple mice were constructed from each cord blood donor. Mice were analyzed quarterly for age-related changes in the hematopoietic and immune systems, and followed for periods up to 18-24 months post-transplant. Flow cytometric analyses were performed for hematopoietic and immune reconstitution. It was observed that NOD-Rag (-)-γ chain (-) mice could be "humanized" long-term using cord blood stem cells, and that some evidence of immune aging occurred during the life of the mice.
Collapse
|
44
|
Khalyavkin AV. Phenoptosis as genetically determined aging influenced by signals from the environment. BIOCHEMISTRY (MOSCOW) 2013; 78:1001-1005. [DOI: 10.1134/s0006297913090058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
45
|
Yang SA, Wang WD, Chen CT, Tseng CY, Chen YN, Hsu HJ. FOXO/Fringe is necessary for maintenance of the germline stem cell niche in response to insulin insufficiency. Dev Biol 2013; 382:124-35. [PMID: 23895933 DOI: 10.1016/j.ydbio.2013.07.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 07/16/2013] [Accepted: 07/19/2013] [Indexed: 12/27/2022]
Abstract
The stem cell niche houses and regulates stem cells by providing both physical contact and local factors that regulate stem cell identity. The stem cell niche also plays a role in integrating niche-local and systemic signals, thereby ensuring that the balance of stem cells meets the needs of the organism. However, it is not clear how these signals are merged within the niche. Nutrient-sensing insulin/FOXO signaling has been previously shown to directly control Notch activation in the Drosophila female germline stem cell (GSC) niche, which maintains the niche and GSC identity. Here, we demonstrate that FOXO directly activates transcription of fringe, a gene encoding a glycosyltransferase that modulates Notch glycosylation. Fringe facilitates Notch inactivation in the GSC niche when insulin signaling is low. We also show that the Notch ligand predominantly involved is GSC niche-derived Delta. These results reveal that FOXO-mediated regulation of fringe links the insulin and Notch signaling pathways in the GSC niche in response to nutrition, and emphasize that stem cells are regulated by complex interactions between niche-local and systemic signals.
Collapse
Affiliation(s)
- Sheng-An Yang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | | | | | | | | | | |
Collapse
|
46
|
Chiba H, Ataka K, Iba K, Nagaishi K, Yamashita T, Fujimiya M. Diabetes impairs the interactions between long-term hematopoietic stem cells and osteopontin-positive cells in the endosteal niche of mouse bone marrow. Am J Physiol Cell Physiol 2013; 305:C693-703. [PMID: 23885062 DOI: 10.1152/ajpcell.00400.2012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hematopoietic stem cells (HSCs) are maintained, and their division/proliferation and quiescence are regulated in the microenvironments, niches, in the bone marrow. Although diabetes is known to induce abnormalities in HSC mobilization and proliferation through chemokine and chemokine receptors, little is known about the interaction between long-term HSCs (LT-HSCs) and osteopontin-positive (OPN) cells in endosteal niche. To examine this interaction, LT-HSCs and OPN cells were isolated from streptozotocin-induced diabetic and nondiabetic mice. In diabetic mice, we observed a reduction in the number of LT-HSCs and OPN cells and impaired expression of Tie2, β-catenin, and N-cadherin on LT-HSCs and β1-integrin, β-catenin, angiopoietin-1, and CXCL12 on OPN cells. In an in vitro coculture system, LT-HSCs isolated from nondiabetic mice exposed to diabetic OPN cells showed abnormal mRNA expression levels of Tie2 and N-cadherin. Conversely, in LT-HSCs derived from diabetic mice exposed to nondiabetic OPN cells, the decreased mRNA expressions of Tie2, β-catenin, and N-cadherin were restored to normal levels. The effects of diabetic or nondiabetic OPN cells on LT-HSCs shown in this coculture system were confirmed by the coinjection of LT-HSCs and OPN cells into bone marrow of irradiated nondiabetic mice. Our results provide new insight into the treatment of diabetes-induced LT-HSC abnormalities and suggest that the replacement of OPN cells may represent a novel treatment strategy.
Collapse
Affiliation(s)
- Hironori Chiba
- Department of Orthopedic Surgery. Sapporo Medical University Faculty of Medicine, Sapporo, Japan, and
| | | | | | | | | | | |
Collapse
|
47
|
Weilner S, Schraml E, Redl H, Grillari-Voglauer R, Grillari J. Secretion of microvesicular miRNAs in cellular and organismal aging. Exp Gerontol 2013; 48:626-33. [PMID: 23283304 PMCID: PMC3695566 DOI: 10.1016/j.exger.2012.11.017] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 11/07/2012] [Accepted: 11/12/2012] [Indexed: 12/15/2022]
Abstract
Changes of factors circulating in the systemic environment during human aging have been investigated for a long time. Only recently however, miRNAs have been found to be secreted into the systemic and tissue environments where they are protected from RNAses by either carrier proteins or by being packaged into microvesicles. These miRNAs are then taken up by recipient cells, changing the cellular behavior by the classical miRNA induced silencing of target mRNAs. The origin of circulating miRNAs, however, is in most instances unclear, but senescent cells emerge as a possible source of such secreted miRNAs. Since differences in the circulating miRNAs have been found in a variety of age-associated diseases, and accumulation of senescent cells in the elderly emerges as a possible detrimental factor in aging, it is well conceivable that these miRNAs might contribute to the functional decline observed during aging of organisms. Therefore, we here give an overview on current knowledge on microvesicular secretion of miRNAs, changes of the systemic and tissue environments during aging of cells and organisms. Finally, we summarize current knowledge on miRNAs that are found to be specific for age-associated diseases.
Collapse
Key Words
- escrt, endosomal sorting complex required for transport
- ilv, intraluminal vesicles
- mirna, microrna
- mrna, messenger rna
- mvb, multivesicular bodies
- msc, mesenchymal stem cell
- pm, plasma membrane
- rab, ras-related in brain
- risc, rna-induced silencing complex
- rrna, ribosomal rna
- sasp, senescence-associated secretory phenotype
- aging
- microrna
- microvesicles
- exosomes
- secretion
- systemic environment
Collapse
Affiliation(s)
- Sylvia Weilner
- BOKU, VIBT, University of Natural Resources and Life Sciences, Department of Biotechnology, Vienna, Austria
- Ludwig Boltzmann Institute for Clinical and Experimental Traumatology, Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Elisabeth Schraml
- BOKU, VIBT, University of Natural Resources and Life Sciences, Department of Biotechnology, Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Clinical and Experimental Traumatology, Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Regina Grillari-Voglauer
- BOKU, VIBT, University of Natural Resources and Life Sciences, Department of Biotechnology, Vienna, Austria
- Evercyte GmbH, Vienna, Austria
| | - Johannes Grillari
- BOKU, VIBT, University of Natural Resources and Life Sciences, Department of Biotechnology, Vienna, Austria
- Evercyte GmbH, Vienna, Austria
| |
Collapse
|
48
|
Abstract
Stem cell niches are special microenvironments that maintain stem cells and control their behavior to ensure tissue homeostasis and regeneration throughout life. The liver has a high regenerative capacity that involves stem/progenitor cells when the proliferation of hepatocytes is impaired. In recent years progress has been made in the identification of potential hepatic stem cell niches. There is evidence that hepatic progenitor cells can originate from niches in the canals of Hering; in addition, the space of Disse may also serve as a stem cell niche during fetal hematopoiesis and constitute a niche for stellate cells in adults.
Collapse
Affiliation(s)
- Claus Kordes
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | |
Collapse
|
49
|
Kucia M, Masternak M, Liu R, Shin DM, Ratajczak J, Mierzejewska K, Spong A, Kopchick JJ, Bartke A, Ratajczak MZ. The negative effect of prolonged somatotrophic/insulin signaling on an adult bone marrow-residing population of pluripotent very small embryonic-like stem cells (VSELs). AGE (DORDRECHT, NETHERLANDS) 2013; 35:315-330. [PMID: 22218782 PMCID: PMC3592960 DOI: 10.1007/s11357-011-9364-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 12/05/2011] [Indexed: 05/31/2023]
Abstract
It is well known that attenuated insulin/insulin-like growth factor signaling (IIS) has a positive effect on longevity in several animal species, including mice. Here, we demonstrate that a population of murine pluripotent very small embryonic-like stem cells (VSELs) that reside in bone marrow (BM) is protected from premature depletion during aging by intrinsic parental gene imprinting mechanisms and the level of circulating insulin-like growth factor-I (IGF-I). Accordingly, an increase in the circulating level of IGF-I, as seen in short-lived bovine growth hormone (bGH)-expressing transgenic mice, which age prematurely, as well as in wild-type animals injected for 2 months with bGH, leads to accelerated depletion of VSELs from bone marrow (BM). In contrast, long-living GHR-null or Ames dwarf mice, which have very low levels of circulating IGF-I, exhibit a significantly higher number of VSELs in BM than their littermates at the same age. However, the number of VSELs in these animals decreases after GH or IGF-I treatment. These changes in the level of plasma-circulating IGF-I corroborate with changes in the genomic imprinting status of crucial genes involved in IIS, such as Igf-2-H19, RasGRF1, and Ig2R. Thus, we propose that a chronic increase in IIS contributes to aging by premature depletion of pluripotent VSELs in adult tissues.
Collapse
Affiliation(s)
- Magda Kucia
- />Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Louisville, KY 40202 USA
- />Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Michal Masternak
- />Burnett School of Biomedical Sciences College of Medicine, Institute of Human Genetics, University of Central Florida, Orlando, FL USA
- />Department of Internal Medicine, School of Medicine, Southern Illinois University, Springfield, IL USA
- />Institute for Human Genetics Polish Academy of Sciences, Poznan, Poland
| | - Riu Liu
- />Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Louisville, KY 40202 USA
| | - Dong-Myung Shin
- />Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Louisville, KY 40202 USA
| | - Janina Ratajczak
- />Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Louisville, KY 40202 USA
- />Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Katarzyna Mierzejewska
- />Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Louisville, KY 40202 USA
| | - Adam Spong
- />Department of Internal Medicine, School of Medicine, Southern Illinois University, Springfield, IL USA
- />Institute for Human Genetics Polish Academy of Sciences, Poznan, Poland
| | - John J. Kopchick
- />Edison Biotechnology Institute and Department of Biomedical Sciences, College of Osteopathic Medicine, Ohio University, Athens, OH USA
| | - Andrzej Bartke
- />Department of Internal Medicine, School of Medicine, Southern Illinois University, Springfield, IL USA
- />Institute for Human Genetics Polish Academy of Sciences, Poznan, Poland
| | - Mariusz Z. Ratajczak
- />Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Louisville, KY 40202 USA
- />Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
50
|
Hao H, Chen G, Liu J, Ti D, Zhao Y, Xu S, Fu X, Han W. Culturing on Wharton's jelly extract delays mesenchymal stem cell senescence through p53 and p16INK4a/pRb pathways. PLoS One 2013; 8:e58314. [PMID: 23516461 PMCID: PMC3596399 DOI: 10.1371/journal.pone.0058314] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 02/01/2013] [Indexed: 12/30/2022] Open
Abstract
Mesenchymal stem cells (MSCs) hold great therapeutic potential. However, MSCs undergo replication senescence during the in vitro expansion process. Wharton's jelly from the human umbilical cord harbors a large number of MSCs. In this study, we hypothesized that Wharton's jelly would be beneficial for in vitro expansion of MSCs. Wharton's jelly extract (WJEs), which is mainly composed of extracellular matrix and cytokines, was prepared as coating substrate. Human MSCs were isolated and cultured on WJE-coated plates. Although the proliferation capacity of cells was not augmented by WJE in early phase culture, adynamic growth in late-phase culture was clearly reduced, suggesting that the replicative senescence of MSCs was efficiently slowed by WJE. This was confirmed by β-galactosidase staining and telomere length measurements of MSCs in late-phase culture. In addition, the decreased differentiation ability of MSCs after long-term culture was largely ameliorated by WJE. Reactive oxygen species (ROS), p53, and p16INK4a/pRb expression increased with passaging. Analysis at the molecular level revealed that WJE-based culture efficiently suppressed the enhancement of intracellular ROS, p53, and p16INK4a/pRb in MSCs. These data demonstrated that WJE provided an ideal microenvironment for MSCs culture expansion in vitro preserved MSC properties by delaying MSCs senescence, and allowed large numbers of MSCs to be obtained for basic research and clinical therapies.
Collapse
Affiliation(s)
- Haojie Hao
- Institute of Basic Medicine Science, Chinese PLA General Hospital, Beijing, China
| | - Guanghui Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Jiejie Liu
- Institute of Basic Medicine Science, Chinese PLA General Hospital, Beijing, China
| | - Dongdong Ti
- Institute of Basic Medicine Science, Chinese PLA General Hospital, Beijing, China
| | - Yali Zhao
- Institute of Basic Medicine Science, Chinese PLA General Hospital, Beijing, China
| | - Shenjun Xu
- Institute of Basic Medicine Science, Chinese PLA General Hospital, Beijing, China
| | - Xiaobing Fu
- Institute of Basic Medicine Science, Chinese PLA General Hospital, Beijing, China
- * E-mail: (WH); (XF)
| | - Weidong Han
- Institute of Basic Medicine Science, Chinese PLA General Hospital, Beijing, China
- * E-mail: (WH); (XF)
| |
Collapse
|