1
|
Lagorgette L, Bogdanova DA, Belotserkovskaya EV, Garrido C, Demidov ON. PP2C phosphatases-terminators of suicidal thoughts. Cell Death Dis 2024; 15:919. [PMID: 39702569 DOI: 10.1038/s41419-024-07269-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/16/2024] [Accepted: 11/27/2024] [Indexed: 12/21/2024]
Abstract
Cell death and related signaling pathways are essential during development and in various physiological and pathological conditions. Post-translational modifications such as ubiquitination and phosphorylation play an important role in these signaling pathways. The involvement of kinases - enzymes that catalyze protein phosphorylation - in cell death signaling has been extensively studied. On the other hand, not many studies have been devoted to analyzing the role in cell death of phosphatases, enzymes involved in the removal of phosphorylated residues added to proteins by kinases. Obviously, the two opposite reactions, phosphorylation and dephosphorylation, are equally important in the regulation of protein functions and subsequently in the execution of the cell death program. Here, we have summarized recent work on the involvement of serine-threonine PP2C phosphatases in cell death pathways, senescence and autophagy, focusing in particular on the most studied phosphatase PPM1D (PP2Cδ) as an example of the regulatory role of PP2Cs in cell death. The review should help to draw attention to the importance of PP2C family phosphatases in cell death checkpoints and to discover new targets for drug development.
Collapse
Affiliation(s)
- Lisa Lagorgette
- INSERM, UMR 1231, Laboratoire d'Excellence LipSTIC and « Equipe labellisée par la Ligue Nationale contre le Cancer », University of Burgundy, Dijon, France
- University of Burgundy, Faculty of Medicine and Pharmacy, Dijon, France
| | - Daria A Bogdanova
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius University of Science and Technology, Sochi, Russia
- Institute of Cytology RAS, St. Petersburg, Russia
| | | | - Carmen Garrido
- INSERM, UMR 1231, Laboratoire d'Excellence LipSTIC and « Equipe labellisée par la Ligue Nationale contre le Cancer », University of Burgundy, Dijon, France
- University of Burgundy, Faculty of Medicine and Pharmacy, Dijon, France
- Center for Cancer Georges-François Leclerc, Dijon, France
| | - Oleg N Demidov
- INSERM, UMR 1231, Laboratoire d'Excellence LipSTIC and « Equipe labellisée par la Ligue Nationale contre le Cancer », University of Burgundy, Dijon, France.
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius University of Science and Technology, Sochi, Russia.
- Institute of Cytology RAS, St. Petersburg, Russia.
| |
Collapse
|
2
|
Sukhtankar DD, Fung JJ, Kim MN, Cayton T, Chiou V, Caculitan NG, Zalicki P, Kim S, Jo Y, Kim S, Lee JM, Choi J, Mun S, Chin A, Jang Y, Lee JY, Kim G, Kim EH, Huh WK, Jeong JY, Seen DS, Cardarelli PM. GPC-100, a novel CXCR4 antagonist, improves in vivo hematopoietic cell mobilization when combined with propranolol. PLoS One 2023; 18:e0287863. [PMID: 37878624 PMCID: PMC10599528 DOI: 10.1371/journal.pone.0287863] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/27/2023] [Indexed: 10/27/2023] Open
Abstract
Autologous Stem Cell Transplant (ASCT) is increasingly used to treat hematological malignancies. A key requisite for ASCT is mobilization of hematopoietic stem cells into peripheral blood, where they are collected by apheresis and stored for later transplantation. However, success is often hindered by poor mobilization due to factors including prior treatments. The combination of G-CSF and GPC-100, a small molecule antagonist of CXCR4, showed potential in a multiple myeloma clinical trial for sufficient and rapid collection of CD34+ stem cells, compared to the historical results from the standards of care, G-CSF alone or G-CSF with plerixafor, also a CXCR4 antagonist. In the present study, we show that GPC-100 has high affinity towards the chemokine receptor CXCR4, and it potently inhibits β-arrestin recruitment, calcium flux and cell migration mediated by its ligand CXCL12. Proximity Ligation Assay revealed that in native cell systems with endogenous receptor expression, CXCR4 co-localizes with the beta-2 adrenergic receptor (β2AR). Co-treatment with CXCL12 and the β2AR agonist epinephrine synergistically increases β-arrestin recruitment to CXCR4 and calcium flux. This increase is blocked by the co-treatment with GPC-100 and propranolol, a non-selective beta-adrenergic blocker, indicating a functional synergy. In mice, GPC-100 mobilized more white blood cells into peripheral blood compared to plerixafor. GPC-100 induced mobilization was further amplified by propranolol pretreatment and was comparable to mobilization by G-CSF. Addition of propranolol to the G-CSF and GPC-100 combination resulted in greater stem cell mobilization than the G-CSF and plerixafor combination. Together, our studies suggest that the combination of GPC-100 and propranolol is a novel strategy for stem cell mobilization and support the current clinical trial in multiple myeloma registered as NCT05561751 at www.clinicaltrials.gov.
Collapse
Affiliation(s)
- Devki D. Sukhtankar
- GPCR Therapeutics USA, Inc., Redwood City, California, United States of America
| | - Juan José Fung
- GPCR Therapeutics USA, Inc., Redwood City, California, United States of America
| | - Mi-na Kim
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - Thomas Cayton
- GPCR Therapeutics USA, Inc., Redwood City, California, United States of America
| | - Valerie Chiou
- GPCR Therapeutics USA, Inc., Redwood City, California, United States of America
| | - Niña G. Caculitan
- GPCR Therapeutics USA, Inc., Redwood City, California, United States of America
| | - Piotr Zalicki
- GPCR Therapeutics USA, Inc., Redwood City, California, United States of America
| | - Sujeong Kim
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - Yoonjung Jo
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - SoHui Kim
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - Jae Min Lee
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - Junhee Choi
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | | | - Ashley Chin
- GPCR Therapeutics USA, Inc., Redwood City, California, United States of America
| | - Yongdae Jang
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - Ji Yeong Lee
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - Gowoon Kim
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - Eun Hee Kim
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | - Won-Ki Huh
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
- Institute of Microbiology, Seoul National University, Seoul, Republic of Korea
| | - Jae-Yeon Jeong
- GPCR Therapeutics Inc., Gwanak-gu, Seoul, Republic of Korea
| | | | - Pina M. Cardarelli
- GPCR Therapeutics USA, Inc., Redwood City, California, United States of America
| |
Collapse
|
3
|
Kröpfl JM, Beltrami FG, Gruber HJ, Stelzer I, Spengler CM. Exercise-Induced Circulating Hematopoietic Stem and Progenitor Cells in Well-Trained Subjects. Front Physiol 2020; 11:308. [PMID: 32457637 PMCID: PMC7220991 DOI: 10.3389/fphys.2020.00308] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 03/19/2020] [Indexed: 12/18/2022] Open
Abstract
It has been proposed that exercise-induced systemic oxidative stress increases circulating hematopoietic stem and progenitor cell (HPC) number in active participants, while HPC clonogenicity is reduced post-exercise. However, HPCs could be protected against exercise-induced reactive oxygen species in a trained state. Therefore, we characterized the acute exercise-induced HPC profile of well-trained participants including cell number, clonogenicity, and clearance. Twenty-one healthy, well-trained participants-12 runners, 9 cyclists; age 30.0 (4.3) years-performed a strenuous acute exercise session consisting of 4 bouts of 4-min high-intensity with 3-min low-intensity in-between, which is known to elicit oxidative stress. Average power/speed of intense phases was 85% of the peak achieved in a previous incremental test. Before and 10 min after exercise, CD34+/45dim cell number and clonogenicity, total oxidative (TOC), and antioxidative (TAC) capacities, as well as CD31 expression on detected HPCs were investigated. TOC significantly decreased from 0.093 (0.059) nmol/l to 0.083 (0.052) nmol/l post-exercise (p = 0.044). Although HPC proportions significantly declined below baseline (from 0.103 (0.037)% to 0.079 (0.028)% of mononuclear cells, p < 0.001), HPC concentrations increased post-exercise [2.10 (0.75) cells/μl to 2.46 (0.98) cells/μl, p = 0.002] without interaction between exercise modalities, while HPC clonogenicity was unaffected. Relating HPC concentrations and clonogenicity to exercise session specific (anti-) oxidative parameters, no association was found. CD31 median fluorescent intensity expression on detected HPCs was diminished post-exercise [from 1,675.9 (661.0) to 1,527.1 (558.9), p = 0.023] and positively correlated with TOC (r rm = 0.60, p = 0.005). These results suggest that acute exercise-reduced oxidative stress influences HPC clearance but not mobilization in well-trained participants. Furthermore, a well-trained state protected HPCs' clonogenicity from post-exercise decline.
Collapse
Affiliation(s)
- Julia M Kröpfl
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Fernando G Beltrami
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Hans-Jürgen Gruber
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Ingeborg Stelzer
- Institute of Medical and Chemical Laboratory Diagnostics, LKH Hochsteiermark, Leoben, Austria
| | - Christina M Spengler
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
4
|
Zhang WB, Liu YQ, Zhang X, Lin L, Yin SL. The role of β-adrenergic receptors and p38MAPK signaling pathways in physiological processes of cardiosphere-derived cells. J Cell Biochem 2017; 119:1204-1214. [PMID: 28722223 DOI: 10.1002/jcb.26292] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 07/18/2017] [Indexed: 01/21/2023]
Abstract
The effects of β adrenergic receptors (β-ARs) and p38 mitogen-activated protein kinases (MAPK) pathways on cardiosphere-derived cells (CDCs) are largely unknown. This study aimed to investigate the roles of β-ARs and p38MAPK pathways on the proliferation, apoptosis, and differentiation capacity of CDCs. The CDCs were treated with β1-AR blocker (Met group), β2-AR antagonist (ICI group), and p38MAPK inhibitor (SB group), non-selective β-AR blocker (PRO group), and β-AR agonist (ISO group). The viability, apoptotic rate and differentiation status of CDCs were determined by MST-1 assay, flow cytometery, and Western blot, respectively. The CDCs viability significantly reduced in ICI group (all P < 0.05), and SB group had a significant high viability after 48 h treatment (P < 0.05). Compared with control group, all treated groups had a low apoptotic rate. After treatment for 72 h, ISO treatment elevated the expression of Nkx2.5, and could partially or fully attenuate the inhibitory effects of β-AR antagonists and/or p38MAPK inhibitor. A similar overall trend of protein expression levels among all groups could be observed between protein pairs of cTnT and β1-AR as well as c-Kit and β2-AR, respectively. These results suggested that β-ARs and p38MAPK signaling pathways play crucial roles in the proliferation and differentiation of CDCs. Our findings should be helpful for better understanding the molecular mechanism underlying the physiological processes of CDCs.
Collapse
Affiliation(s)
- Wen-Bo Zhang
- Department of Cardiac Surgery Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yun-Qi Liu
- Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xi Zhang
- Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lin Lin
- Department of Cardiac Surgery Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Sheng-Li Yin
- Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
5
|
Gallina D, Palazzo I, Steffenson L, Todd L, Fischer AJ. Wnt/β-catenin-signaling and the formation of Müller glia-derived progenitors in the chick retina. Dev Neurobiol 2015; 76:983-1002. [PMID: 26663639 DOI: 10.1002/dneu.22370] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 11/25/2015] [Accepted: 12/02/2015] [Indexed: 11/12/2022]
Abstract
Müller glia can be stimulated to de-differentiate, proliferate, and form Müller glia-derived progenitor cells (MGPCs) that are capable of producing retinal neurons. The signaling pathways that influence the de-differentiation of mature Müller glia and proliferation of MGPCs may include the Wnt-pathway. The purpose of this study was to investigate how Wnt-signaling influences the formation of MGPCs in the chick retina in vivo. In NMDA-damaged retinas where MGPCs are known to form, we find dynamic changes in retinal levels of potential readouts of Wnt-signaling, including dkk1, dkk3, axin2, c-myc, tcf-1, and cd44. We find accumulations of nuclear β-catenin in MGPCs that peaks at 3 days and rapidly declines by 5 days after NMDA-treatment. Inhibition of Wnt-signaling with XAV939 in damaged retinas suppressed the formation of MGPCs, increased expression of ascl1a and decreased hes5, but had no effect upon the differentiation of progeny produced by MGPCs. Activation of Wnt-signaling, with GSK3β-inhibitors, in the absence of retinal damage, failed to stimulate the formation of MGPCs, whereas activation of Wnt-signaling in damaged retinas stimulated the formation of MGPCs. In the absence of retinal damage, FGF2/MAPK-signaling stimulated the formation of MGPCs by activating a signaling network that includes Wnt/β-catenin. In FGF2-treated retinas, inhibition of Wnt-signaling reduced numbers of proliferating MGPCs, whereas activation of Wnt-signaling failed to influence the formation of proliferating MGPCs. Our findings indicate that Wnt-signaling is part of a network initiated by FGF2/MAPK or retinal damage, and activation of canonical Wnt-signaling is required for the formation of proliferating MGPCs. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 76: 983-1002, 2016.
Collapse
Affiliation(s)
- Donika Gallina
- Department of Neuroscience, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Isabella Palazzo
- Department of Neuroscience, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Lillia Steffenson
- Department of Neuroscience, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Levi Todd
- Department of Neuroscience, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Andy J Fischer
- Department of Neuroscience, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
6
|
Adrenergic DNA damage of embryonic pluripotent cells via β2 receptor signalling. Sci Rep 2015; 5:15950. [PMID: 26516061 PMCID: PMC4626766 DOI: 10.1038/srep15950] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 10/02/2015] [Indexed: 12/16/2022] Open
Abstract
Embryonic pluripotent cells are sensitive to genotoxicity though they need more stringent genome integrity to avoid compromising multiple cell lineages and subsequent generations. However it remains unknown whether the cells are susceptible to adrenergic stress which can induce somatic cell genome lesion. We have revealed that adrenergic stress mediators cause DNA damage of the cells through the β2 adrenergic receptor/adenylate cyclase/cAMP/PKA signalling pathway involving an induction of intracellular reactive oxygen species (ROS) accumulation. The adrenergic stress agonists adrenaline, noradrenaline, and isoprenaline caused DNA damage and apoptosis of embryonic stem (ES) cells and embryonal carcinoma stem cells. The effects were mimicked by β2 receptor-coupled signalling molecules and abrogated by selective blockade of β2 receptors and inhibition of the receptor signalling pathway. RNA interference targeting β2 receptors of ES cells conferred the cells the ability to resist the DNA damage and apoptosis. In addition, adrenergic stimulation caused a consistent accumulation of ROS in the cells and the effect was abrogated by β2 receptor blockade; quenching of ROS reversed the induced DNA damage. This finding will improve the understanding of the stem cell regulatory physiology/pathophysiology in an adrenergic receptor subtype signalling mechanism.
Collapse
|
7
|
Chronic restraint stress after injury and shock is associated with persistent anemia despite prolonged elevation in erythropoietin levels. J Trauma Acute Care Surg 2015; 79:91-6; discussion 96-7. [PMID: 26091320 DOI: 10.1097/ta.0000000000000686] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Following severe traumatic injury, critically ill patients have a prolonged hypercatacholamine state that is associated with bone marrow (BM) dysfunction and persistent anemia. However, current animal models of injury and shock result in a transient anemia. Daily restraint stress (chronic stress [CS]) has been shown to increase catecholamines. We hypothesize that adding CS following injury or injury and shock in rats will prolong the hypercatecholaminemia and prolong the initial anemia, despite elevated erythropoietin (EPO) levels. METHODS Male Sprague-Dawley rats (n = 6-8 per group) underwent lung contusion (LC) or combined LC/hemorrhagic shock (LCHS) followed by 6 days of CS. CS consisted of a 2-hour restraint period interrupted with repositioning and alarms every 30 minutes. At 7 days, urine was assessed for norepinephrine (NE) levels, blood for EPO and hemoglobin (Hgb), and BM for erythroid progenitor growth. RESULTS Animals undergoing LC or combined LCHS predictably recovered by Day 7; urine NE, EPO, and Hgb levels were normal. The addition of CS to LC and LCHS models was associated with a significant elevation in NE on Day 6. The addition of CS to LC led to a persistent 20% to 25% decrease in the growth of BM hematopoietic progenitor cells. These findings were further exaggerated when CS was added following LCHS, resulting in a 20%q to 40% reduction in BM erythroid progenitor colony growth and a 20% decrease in Hgb when compared with LCHS alone. CONCLUSION Exposing injured animals to CS results in prolonged elevation of NE and EPO, which is associated with worsening BM erythroid function and persistent anemia. Chronic restraint stress following injury and shock provides a clinically relevant model to further evaluate persistent injury-associated anemia seen in critically ill trauma patients. Furthermore, alleviating CS after severe injury is a potential therapeutic target to improve BM dysfunction and anemia.
Collapse
|
8
|
Vasin MV, Ushakov IB. Comparative efficacy and the window of radioprotection for adrenergic and serotoninergic agents and aminothiols in experiments with small and large animals. JOURNAL OF RADIATION RESEARCH 2015; 56:1-10. [PMID: 25312329 PMCID: PMC4572585 DOI: 10.1093/jrr/rru087] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 08/25/2014] [Accepted: 09/05/2014] [Indexed: 06/04/2023]
Abstract
This review gives a comparative evaluation of the radioprotective properties and the therapeutic index (TI) of radioprotectors from various pharmacological group in experiments on both small and large animals. It presents a hypothesis explaining the decrease in the TI of cystamine and 5-methoxytryptamine (mexamine), and the retention of that of α1-adrenomimetic indralin, and also compares the effects on large and small animals. The considerable differences in the therapeutic indices of catecholamines, serotonin and cystamine are a consequence of specific features of their mechanisms of radioprotective action. Radioprotectors acting via receptor mediation tend to provide a more expanded window of protection. The reduction in the TI of cystamine in larger animals, such as dogs, may be caused by the greater increase in toxicity of aminothiols in relation to the decrease in their optimal doses for radioprotective effect in going from mice to dogs, which is a consequence of the slower metabolic processes in larger animals. The somatogenic phase of intoxication by cystamine is significantly longer than the duration of its radioprotective effect, and increases with irradiation. The decrease in the radioprotective effect and the TI of mexamine in experiments with dogs may be caused by their lower sensitivity to the acute hypoxia induced by the mexamine. This is because of lower gradient in oxygen tension between tissue cells and blood capillaries under acute hypoxia that is determined by lower initial oxygen consumption in a large animal as compared with a small animal. Indralin likely provides optimal radioprotective effects and a higher TI for large animals via the increased specificity of its adrenergic effect on tissue respiration, which supports the development of acute hypoxia in the radiosensitive tissues of large animals. The stimulatory effect of indralin on early post-irradiation haematopoietic recovery cannot provide a high level of radioprotective action for large animals, but it may promote recovery.
Collapse
Affiliation(s)
- Mikhail V Vasin
- State Scientific Center Russian Federation - Institute of Biomedical Problems, Russian Academy of Science, 76a Khoroshovskoe schuss, Moscow123007, Russia
| | - Igor B Ushakov
- State Scientific Center Russian Federation - Institute of Biomedical Problems, Russian Academy of Science, 76a Khoroshovskoe schuss, Moscow123007, Russia
| |
Collapse
|
9
|
Stelzer I, Kröpfl JM, Fuchs R, Pekovits K, Mangge H, Raggam RB, Gruber HJ, Prüller F, Hofmann P, Truschnig-Wilders M, Obermayer-Pietsch B, Haushofer AC, Kessler HH, Mächler P. Ultra-endurance exercise induces stress and inflammation and affects circulating hematopoietic progenitor cell function. Scand J Med Sci Sports 2014; 25:e442-50. [PMID: 25438993 DOI: 10.1111/sms.12347] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2014] [Indexed: 01/18/2023]
Abstract
Although amateur sports have become increasingly competitive within recent decades, there are as yet few studies on the possible health risks for athletes. This study aims to determine the impact of ultra-endurance exercise-induced stress on the number and function of circulating hematopoietic progenitor cells (CPCs) and hematological, inflammatory, clinical, metabolic, and stress parameters in moderately trained amateur athletes. Following ultra-endurance exercise, there were significant increases in leukocytes, platelets, interleukin-6, fibrinogen, tissue enzymes, blood lactate, serum cortisol, and matrix metalloproteinase-9. Ultra-endurance exercise did not influence the number of CPCs but resulted in a highly significant decline of CPC functionality after the competition. Furthermore, Epstein-Barr virus was seen to be reactivated in one of seven athletes. The link between exercise-induced stress and decline of CPC functionality is supported by a negative correlation between cortisol and CPC function. We conclude that ultra-endurance exercise induces metabolic stress and an inflammatory response that affects not only mature hematopoietic cells but also the function of the immature hematopoietic stem and progenitor cell fraction, which make up the immune system and provide for regeneration.
Collapse
Affiliation(s)
- I Stelzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - J M Kröpfl
- Institute of Human Movement Sciences and Sport, Exercise Physiology Lab, ETH Zurich, Zurich, Switzerland.,Institute of Biophysics, Medical University of Graz, Graz, Austria
| | - R Fuchs
- Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria
| | - K Pekovits
- Department of Ophthalmology, Medical University of Graz, Graz, Austria
| | - H Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.,BioTechMed-Graz, Graz, Austria
| | - R B Raggam
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - H-J Gruber
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - F Prüller
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - P Hofmann
- Institute of Sports Science, Karl-Franzens-University of Graz, Graz, Austria
| | - M Truschnig-Wilders
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - B Obermayer-Pietsch
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - A C Haushofer
- Institute of Medical and Chemical Laboratory Diagnostics Wels-Grieskirchen, Wels-Grieskirchen, Austria
| | - H H Kessler
- Research Unit Molecular Diagnostics, IHMEM, Medical University of Graz, Graz, Austria
| | - P Mächler
- Center for Cardiac Rehabilitation, SKA-PVA St. Radegund, Graz, Austria
| |
Collapse
|
10
|
Kröpfl JM, Stelzer I, Mangge H, Pekovits K, Fuchs R, Allard N, Schinagl L, Hofmann P, Dohr G, Wallner-Liebmann S, Domej W, Müller W. Exercise-induced norepinephrine decreases circulating hematopoietic stem and progenitor cell colony-forming capacity. PLoS One 2014; 9:e106120. [PMID: 25180783 PMCID: PMC4152172 DOI: 10.1371/journal.pone.0106120] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 07/28/2014] [Indexed: 11/18/2022] Open
Abstract
A recent study showed that ergometry increased circulating hematopoietic stem and progenitor cell (CPC) numbers, but reduced hematopoietic colony forming capacity/functionality under normoxia and normobaric hypoxia. Herein we investigated whether an exercise-induced elevated plasma free/bound norepinephrine (NE) concentration could be responsible for directly influencing CPC functionality. Venous blood was taken from ten healthy male subjects (25.3+/-4.4 yrs) before and 4 times after ergometry under normoxia and normobaric hypoxia (FiO2<0.15). The circulating hematopoietic stem and progenitor cell numbers were correlated with free/bound NE, free/bound epinephrine (EPI), cortisol (Co) and interleukin-6 (IL-6). Additionally, the influence of exercise-induced NE and blood lactate (La) on CPC functionality was analyzed in a randomly selected group of subjects (n = 6) in vitro under normoxia by secondary colony-forming unit granulocyte macrophage assays. Concentrations of free NE, EPI, Co and IL-6 were significantly increased post-exercise under normoxia/hypoxia. Ergometry-induced free NE concentrations found in vivo showed a significant impairment of CPC functionality in vitro under normoxia. Thus, ergometry-induced free NE was thought to trigger CPC mobilization 10 minutes post-exercise, but as previously shown impairs CPC proliferative capacity/functionality at the same time. The obtained results suggest that an ergometry-induced free NE concentration has a direct negative effect on CPC functionality. Cortisol may further influence CPC dynamics and functionality.
Collapse
Affiliation(s)
- Julia M. Kröpfl
- Institute of Human Movement Sciences and Sport, Exercise Physiology Lab, ETH Zurich, Zurich, Switzerland
- Institute of Biophysics, Medical University of Graz, Graz, Austria
| | - Ingeborg Stelzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Research Unit on Lifestyle and Inflammation-associated Risk Biomarkers, Medical University of Graz, Graz, Austria
- Institute for Pathophysiology and Immunology, Medical University of Graz, Graz, Austria
| | - Harald Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Research Unit on Lifestyle and Inflammation-associated Risk Biomarkers, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Karl-Franzens University & Technical University & Medical University of Graz, Graz, Austria
| | - Karin Pekovits
- Department of Ophthalmology, Medical University of Graz, Graz, Austria
| | - Robert Fuchs
- Institute for Pathophysiology and Immunology, Medical University of Graz, Graz, Austria
| | - Nathalie Allard
- Institute for Pathophysiology and Immunology, Medical University of Graz, Graz, Austria
| | - Lukas Schinagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Research Unit on Lifestyle and Inflammation-associated Risk Biomarkers, Medical University of Graz, Graz, Austria
- Institute for Pathophysiology and Immunology, Medical University of Graz, Graz, Austria
| | - Peter Hofmann
- Institute of Sports Science, Karl-Franzens University of Graz, Graz, Austria
| | - Gottfried Dohr
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | | | - Wolfgang Domej
- Department of Pulmonology, Medical University of Graz, Graz, Austria
| | - Wolfram Müller
- Institute of Biophysics, Medical University of Graz, Graz, Austria
| |
Collapse
|
11
|
Fischer AJ, Zelinka C, Gallina D, Scott MA, Todd L. Reactive microglia and macrophage facilitate the formation of Müller glia-derived retinal progenitors. Glia 2014; 62:1608-28. [PMID: 24916856 DOI: 10.1002/glia.22703] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/11/2014] [Accepted: 05/16/2014] [Indexed: 12/12/2022]
Abstract
In retinas where Müller glia have been stimulated to become progenitor cells, reactive microglia are always present. Thus, we investigated how the activation or ablation of microglia/macrophage influences the formation of Müller glia-derived progenitor cells (MGPCs) in the retina in vivo. Intraocular injections of the Interleukin-6 (IL6) stimulated the reactivity of microglia/macrophage, whereas other types of retinal glia appear largely unaffected. In acutely damaged retinas where all of the retinal microglia/macrophage were ablated, the formation of proliferating MGPCs was greatly diminished. With the microglia ablated in damaged retinas, levels of Notch and related genes were unchanged or increased, whereas levels of ascl1a, TNFα, IL1β, complement component 3 (C3) and C3a receptor were significantly reduced. In the absence of retinal damage, the combination of insulin and Fibroblast growth factor 2 (FGF2) failed to stimulate the formation of MGPCs when the microglia/macrophage were ablated. In addition, intraocular injections of IL6 and FGF2 stimulated the formation of MGPCs in the absence of retinal damage, and this generation of MGPCs was blocked when the microglia/macrophage were absent. We conclude that the activation of microglia and/or infiltrating macrophage contributes to the formation of proliferating MGPCs, and these effects may be mediated by components of the complement system and inflammatory cytokines.
Collapse
Affiliation(s)
- Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio
| | | | | | | | | |
Collapse
|
12
|
Abstract
The intra-uterine environment provides the first regulatory connection for the developing fetus and shapes its physiological responses in preparation for postnatal life. Psychological stress acts as a programming determinant by setting functional parameters to abnormal levels, thus inducing postnatal maladaptation. The effects of prenatal maternal stress (PNMS) on the developing immune system have been documented mostly through animal studies, but inconsistent results and methodological differences have hampered the complete understanding of these findings. As the immune system follows a similar ontogenic pattern in all mammals, a translational framework based on the developmental windows of vulnerability proposed by immunotoxicology studies was created to integrate these findings. The objective of this review is to examine the available literature on PNMS and immune function in the offspring through the above framework and gain a better understanding of these results by elucidating the moderating influence of the stressor type, timing and duration, and the offspring species, sex and age at assessment. The evaluation of the literature through this framework showed that the effects of PNMS are parameter specific: the moderating effects of timing in gestation were relevant for lymphocyte population numbers, Natural Killer cell function and mitogen-induced proliferation. The presence of an important and directional sexual dimorphism was evident and the influence of the type or duration of PNMS paralleled that of stress in non-pregnant animals. In conclusion, PNMS is a relevant factor in the programming of immune function. Its consequences may be related to disorders with an important immune component such as allergies.
Collapse
Affiliation(s)
- Franz Veru
- Department of Psychiatry, McGill University , Montreal, QC , Canada and
| | | | | | | |
Collapse
|
13
|
Urao N, Ushio-Fukai M. Redox regulation of stem/progenitor cells and bone marrow niche. Free Radic Biol Med 2013; 54:26-39. [PMID: 23085514 PMCID: PMC3637653 DOI: 10.1016/j.freeradbiomed.2012.10.532] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 10/02/2012] [Accepted: 10/05/2012] [Indexed: 01/19/2023]
Abstract
Bone marrow (BM)-derived stem and progenitor cell functions including self-renewal, differentiation, survival, migration, proliferation, and mobilization are regulated by unique cell-intrinsic and -extrinsic signals provided by their microenvironment, also termed the "niche." Reactive oxygen species (ROS), especially hydrogen peroxide (H(2)O(2)), play important roles in regulating stem and progenitor cell functions in various physiologic and pathologic responses. The low level of H(2)O(2) in quiescent hematopoietic stem cells (HSCs) contributes to maintaining their "stemness," whereas a higher level of H(2)O(2) within HSCs or their niche promotes differentiation, proliferation, migration, and survival of HSCs or stem/progenitor cells. Major sources of ROS are NADPH oxidase and mitochondria. In response to ischemic injury, ROS derived from NADPH oxidase are increased in the BM microenvironment, which is required for hypoxia and hypoxia-inducible factor-1α expression and expansion throughout the BM. This, in turn, promotes progenitor cell expansion and mobilization from BM, leading to reparative neovascularization and tissue repair. In pathophysiological states such as aging, atherosclerosis, heart failure, hypertension, and diabetes, excess amounts of ROS create an inflammatory and oxidative microenvironment, which induces cell damage and apoptosis of stem and progenitor cells. Understanding the molecular mechanisms of how ROS regulate the functions of stem and progenitor cells and their niche in physiological and pathological conditions will lead to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Norifumi Urao
- Department of Pharmacology, Center for Lung and Vascular Biology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | |
Collapse
|
14
|
Kroepfl JM, Pekovits K, Stelzer I, Fuchs R, Zelzer S, Hofmann P, Sedlmayr P, Dohr G, Wallner-Liebmann S, Domej W, Mueller W. Exercise increases the frequency of circulating hematopoietic progenitor cells, but reduces hematopoietic colony-forming capacity. Stem Cells Dev 2012; 21:2915-25. [PMID: 22616638 DOI: 10.1089/scd.2012.0017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Circulating hematopoietic progenitor cells (CPCs) may be triggered by physical exercise and/or normobaric hypoxia from the bone marrow. The aim of the study was to investigate the influence of physical exercise and normobaric hypoxia on CPC number and functionality in the peripheral blood as well as the involvement of oxidative stress parameters as possibly active agents. Ten healthy male subjects (25.3±4.4 years) underwent a standardized cycle incremental exercise test protocol (40 W+20 W/min) under either normoxic (FiO2 ∼0.21) or hypoxic conditions (FiO2<0.15, equals 3,500 m, 3 h xposure) within a time span of at least 1 week. Blood was drawn from the cubital vein before and 10, 30, 60, and 120 min after exercise. The number of CPCs in the peripheral blood was analyzed by flow cytometry (CD34/CD45-positive cells). The functionality of cells present was addressed by secondary colony-forming unit-granulocyte macrophage (CFU-GM) assays. To determine a possible correlation between the mobilization of CPCs and reactive oxygen species, parameters for oxidative stress such as malondialdehyde (MDA) and myeloperoxidase (MPO) were obtained. Data showed a significant increase of CPC release under normoxic as well as hypoxic conditions after 10 min of recovery (P<0.01). Most interestingly, although CD34+/CD45dim cells increased in number, the proliferative capacity of CPCs decreased significantly 10 min after cessation of exercise (P<0.05). A positive correlation between CPCs and MDA/MPO levels turned out to be significant for both normoxic and hypoxic conditions (P<0.05/P<0.01). Hypoxia did not provoke an additional effect. Although the CPC frequency increased, the functionality of CPCs decreased significantly after exercise, possibly due to the influence of increased oxidative stress levels.
Collapse
Affiliation(s)
- Julia Maria Kroepfl
- Human Performance Research Graz (HPRGraz), Karl-Franzens-University and Medical University of Graz, Graz, Austria.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Diaz-Montero CM, Wang Y, Shao L, Feng W, Zidan AA, Pazoles CJ, Montero AJ, Zhou D. The glutathione disulfide mimetic NOV-002 inhibits cyclophosphamide-induced hematopoietic and immune suppression by reducing oxidative stress. Free Radic Biol Med 2012; 52:1560-1568. [PMID: 22343421 PMCID: PMC3341494 DOI: 10.1016/j.freeradbiomed.2012.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 01/20/2012] [Accepted: 02/04/2012] [Indexed: 12/22/2022]
Abstract
The oxidized glutathione mimetic NOV-002 is a unique anti-tumor agent that not only has the ability to inhibit tumor cell proliferation, survival, and invasion, but in some settings can also ameliorate cytotoxic chemotherapy-induced hematopoietic and immune suppression. However, the mechanisms by which NOV-002 protects the hematopoietic and immune systems against the cytotoxic effects of chemotherapy are not known. Therefore, in this study we investigated the mechanisms of action of NOV-002 using a mouse model in which hematopoietic and immune suppression was induced by cyclophosphamide (CTX) treatment. We found that NOV-002 treatment in a clinically comparable dose regimen attenuated CTX-induced reduction in bone marrow hematopoietic stem and progenitor cells (HSPCs) and reversed the immunosuppressive activity of myeloid-derived suppressor cells (MDSCs), which led to a significant improvement in hematopoietic and immune functions. These effects of NOV-002 may be attributable to its ability to modulate cellular redox. This suggestion is supported by the finding that NOV-002 treatment upregulated the expression of superoxide dismutase 3 and glutathione peroxidase 2 in HSPCs, inhibited CTX-induced increases in reactive oxygen species production in HSPCs and MDSCs, and attenuated CTX-induced reduction of the ratio of reduced glutathione to oxidized glutathione in splenocytes. These findings provide a better understanding of the mechanisms whereby NOV-002 modulates chemotherapy-induced myelosuppression and immune dysfunction and a stronger rationale for clinical utilization of NOV-002 to reduce chemotherapy-induced hematopoietic and immune suppression.
Collapse
Affiliation(s)
| | - Yong Wang
- Department of Pathology, Medical University of South Carolina, Charleston, SC 29425
| | - Lijian Shao
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - Wei Feng
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - Abdel-Aziz Zidan
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136
| | | | - Alberto J. Montero
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Daohong Zhou
- Department of Pathology, Medical University of South Carolina, Charleston, SC 29425
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| |
Collapse
|
16
|
Abstract
MSCs (mesenchymal stem cells) are planned foruse in regenerative medicine to offset age-dependent alterations. However, MSCs are affected by replicative senescence associated with decreasing proliferation potential, telomere shortening and DNA damage during in vitro propagation. To monitor in vitro senescence, we have assessed the integrity of DNA by the alkaline comet assay. For optimization of the comet assay we have enhanced the stability of comet slides in liquid and minimized the background noise of the method by improving adhesion of agarose gels on the comet slides and concentrating cells on a defined small area on the slides. The modifications of the slide preparation increase the overall efficiency and reproducibility of the comet assay and minimize the image capture and storage. DNA damage of human MSCs during in vitro cultivation increased with time, as assessed by the comet assay, which therefore offers a fast and easy screening tool in future efforts to minimize replicative senescence of MSCs in vitro.
Collapse
|
17
|
Fuchs R, Schraml E, Leitinger G, Stelzer I, Allard N, Haas HS, Schauenstein K, Sadjak A. α1-adrenergic drugs modulate differentiation and cell death of human erythroleukemia cells through non adrenergic mechanism. Exp Cell Res 2011; 317:2239-51. [DOI: 10.1016/j.yexcr.2011.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 07/01/2011] [Accepted: 07/06/2011] [Indexed: 11/28/2022]
|
18
|
Fuchs R, Schraml E, Leitinger G, Letofsky-Papst I, Stelzer I, Haas HS, Schauenstein K, Sadjak A. α1-adrenergic drugs exhibit affinity to a thapsigargin-sensitive binding site and interfere with the intracellular Ca2+ homeostasis in human erythroleukemia cells. Exp Cell Res 2011; 317:2969-80. [PMID: 21851819 DOI: 10.1016/j.yexcr.2011.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 08/02/2011] [Indexed: 01/10/2023]
Abstract
Even though the erythroleukemia cell lines K562 and HEL do not express α1-adrenoceptors, some α1-adrenergic drugs influence both survival and differentiation of these cell lines. Since Ca2+ is closely related to cellular homeostasis, we examined the capacity of α1-adrenergic drugs to modulate the intracellular Ca2+ content in K562 cells. Because of morphological alterations of mitochondria following α1-adrenergic agonist treatment, we also scrutinized mitochondrial functions. In order to visualize the non-adrenoceptor binding site(s) of α1-adrenergic drugs in erythroleukemia cells, we evaluated the application of the fluorescent α1-adrenergic antagonist BODIPY® FL-Prazosin. We discovered that the α1-adrenergic agonists naphazoline, oxymetazoline and also the α1-adrenergic antagonist benoxathian are able to raise the intracellular Ca2+-content in K562 cells. Furthermore, we demonstrate that naphazoline treatment induces ROS-formation as well as an increase in Δψm in K562 cells. Using BODIPY® FL-Prazosin we were able to visualize the non-adrenoceptor binding site(s) of α1-adrenergic drugs in erythroleukemia cells. Interestingly, the SERCA-inhibitor thapsigargin appears to interfere with the binding of BODIPY® FL-Prazosin. Our data suggest that the effects of α1-adrenergic drugs on erythroleukemia cells are mediated by a thapsigargin sensitive binding site, which controls the fate of erythroleukemia cells towards differentiation, senescence and cell death through modulation of intracellular Ca2+.
Collapse
Affiliation(s)
- Robert Fuchs
- Institute of Pathophysiology and Immunology, Center of Molecular Medicine, Medical University of Graz, Heinrichstrasse 31A, 8010 Graz, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Elhassan IO, Hannoush EJ, Sifri ZC, Jones E, Alzate WD, Rameshwar P, Livingston DH, Mohr AM. Beta-blockade prevents hematopoietic progenitor cell suppression after hemorrhagic shock. Surg Infect (Larchmt) 2011; 12:273-8. [PMID: 21790478 PMCID: PMC3159105 DOI: 10.1089/sur.2010.043] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Severe injury is accompanied by sympathetic stimulation that induces bone marrow (BM) dysfunction by both suppression of hematopoietic progenitor cell (HPC) growth and loss of cells via HPC mobilization to the peripheral circulation and sites of injury. Previous work demonstrated that beta-blockade (BB) given prior to tissue injury both reduces HPC mobilization and restores HPC colony growth within the BM. This study examined the effect and timing of BB on BM function in a hemorrhagic shock (HS) model. METHODS Male Sprague-Dawley rats underwent HS via blood withdrawal, maintaining the mean arterial blood pressure at 30-40 mm Hg for 45 min, after which the extracted blood was reinfused. Propranolol (10 mg/kg) was given either prior to or immediately after HS. Blood pressure, heart rate, BM cellularity, and death were recorded. Bone marrow HPC growth was assessed by counting colony-forming unit-granulocyte-, erythrocyte-, monocyte-, megakaryocyte (CFU-GEMM), burst-forming unit-erythroid (BFU-E), and colony-forming unit-erythroid (CFU-E) cells. RESULTS Administration of BB prior to injury restored HPC growth to that of naïve animals (CFU-GEMM 59 ± 11 vs. 61 ± 4, BFU-E 68 ± 9 vs. 73 ± 3, and CFU-E 81 ± 35 vs. 78 ± 14 colonies/plate). Beta-blockade given after HS increased the growth of CFU-GEMM, BFU-E, and CFU-E significantly and improved BM cellularity compared with HS alone. The mortality rate was not increased in the groups receiving BB. CONCLUSION Administration of propranolol either prior to injury or immediately after resuscitation significantly reduced post-shock BM suppression. After HS, BB may improve BM cellularity by decreasing HPC mobilization. Therefore, the early use of BB post-injury may play an important role in attenuating the BM dysfunction accompanying HS.
Collapse
Affiliation(s)
- Ihab O. Elhassan
- Department of Surgery, Division of Trauma, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, New Jersey
| | - Edward J. Hannoush
- Department of Surgery, Division of Trauma, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, New Jersey
| | - Ziad C. Sifri
- Department of Surgery, Division of Trauma, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, New Jersey
| | - Eyone Jones
- Department of Surgery, Division of Trauma, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, New Jersey
| | - Walter D. Alzate
- Department of Surgery, Division of Trauma, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, New Jersey
| | - Pranela Rameshwar
- Department of Medicine-Hematology, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, New Jersey
| | - David H. Livingston
- Department of Surgery, Division of Trauma, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, New Jersey
| | - Alicia M. Mohr
- Department of Surgery, Division of Trauma, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, New Jersey
| |
Collapse
|
20
|
Does beta blockade postinjury prevent bone marrow suppression? ACTA ACUST UNITED AC 2011; 70:1043-9; discussion 1049-50. [PMID: 21610422 DOI: 10.1097/ta.0b013e3182169326] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Trauma-induced hypercatecholaminemia negatively impacts bone marrow (BM) function by suppressing BM hematopoietic progenitor cell (HPC) growth and increasing HPC egress to injured tissue. Beta blockade (BB) given before tissue injury alone has been shown to reduce both HPC mobilization and restore HPC colony growth within the BM. In a clinically relevant model, this study examines the effect of BB given after both tissue injury and hemorrhagic shock (HS). METHODS Male Sprague-Dawley rats underwent lung contusion (LC) with a blast wave percussion. HS was achieved after LC by maintaining the mean arterial blood pressure 30 mm Hg to 35 mm Hg for 45 minutes. Propranolol (10 mg/kg) was given once the mean arterial blood pressure>80 mm Hg and subsequent doses were given daily (LC/HS/BB). One-day and 7-day postinjury, analysis of BM and lung tissue for the growth of HPCs, hematologic parameters, and histology of lung injury were performed. RESULTS LC/HS significantly worsens BM CFU-E growth suppression (15±8 vs. 35±2) and increases CFU-E growth in injured tissue when compared with LC at 1 day and 7 days (33±5 vs. 22±9). The use of BB after LC/HS ameliorated BM suppression, the degree of anemia and HPC growth in the injured lung at 1 day and 7 days postinjury. Lung injury score shows that there was no worsening of lung healing with BB (LC/HS/BB 3.2±2 vs. LC/HS 3.8±0.8). CONCLUSION In an injury and shock model, administration of propranolol immediately after resuscitation significantly reduced BM suppression, and the protective effect is maintained at 7 days with daily BB. Although BB appears to improve BM function by decreasing HPC mobilization to injured tissue, there was no worsening of lung healing. Therefore, the use of propranolol after trauma and resuscitation may minimize long-term BM suppression after injury with no adverse impact on healing.
Collapse
|
21
|
Acharya A, Das I, Chandhok D, Saha T. Redox regulation in cancer: a double-edged sword with therapeutic potential. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2010; 3:23-34. [PMID: 20716925 PMCID: PMC2835886 DOI: 10.4161/oxim.3.1.10095] [Citation(s) in RCA: 355] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Oxidative stress, implicated in the etiology of cancer, results from an imbalance in the production of reactive oxygen species (ROS) and cell’s own antioxidant defenses. ROS deregulate the redox homeostasis and promote tumor formation by initiating an aberrant induction of signaling networks that cause tumorigenesis. Ultraviolet (UV) exposures, γ-radiation and other environmental carcinogens generate ROS in the cells, which can exert apoptosis in the tumors, thereby killing the malignant cells or induce the progression of the cancer growth by blocking cellular defense system. Cancer stem cells take the advantage of the aberrant redox system and spontaneously proliferate. Oxidative stress and gene-environment interactions play a significant role in the development of breast, prostate, pancreatic and colon cancer. Prolonged lifetime exposure to estrogen is associated with several kinds of DNA damage. Oxidative stress and estrogen receptor-associated proliferative changes are suggested to play important roles in estrogen-induced breast carcinogenesis. BRCA1, a tumor suppressor against hormone responsive cancers such as breast and prostate cancer, plays a significant role in inhibiting ROS and estrogen mediated DNA damage; thereby regulate the redox homeostasis of the cells. Several transcription factors and tumor suppressors are involved during stress response such as Nrf2, NFκB and BRCA1. A promising strategy for targeting redox status of the cells is to use readily available natural substances from vegetables, fruits, herbs and spices. Many of the phytochemicals have already been identified to have chemopreventive potential, capable of intervening in carcinogenesis.
Collapse
Affiliation(s)
- Asha Acharya
- Lombardi Comprehensive Cancer Center, Pre Clinical Science, Washington DC, USA.
| | | | | | | |
Collapse
|
22
|
Lajevic MD, Suleiman S, Cohen RL, Chambers DA. Activation of p38 mitogen-activated protein kinase by norepinephrine in T-lineage cells. Immunology 2010; 132:197-208. [PMID: 21039464 DOI: 10.1111/j.1365-2567.2010.03354.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The catecholamine norepinephrine (NE) stimulates T lymphocytes through a beta-adrenergic receptor (βAR)/adenylyl cyclase (AC)/cyclic AMP (cAMP)/protein kinase A (PKA) pathway, leading to altered cell responsiveness and apoptosis. p38 Mitogen-activated protein kinase (MAPK), a major intracellular signalling mediator for cellular and environmental stressors, is involved in the production of immune modulators and in the regulation of T-cell development, survival and death. In these studies we investigated the relationship among NE signalling, p38 MAPK activity and T-cell death. We showed that NE stimulation of BALB/c mouse thymocytes and S49 thymoma cells selectively increases the dual phosphorylation and activity of p38α MAPK. p38 MAPK activation involves the βAR, Gs protein, AC, cAMP and PKA, as determined through the use of a βAR antagonist, activators of AC and cAMP, and S49 clonal mutants deficient in Gs and PKA. Dual phosphorylation of p38 MAPK is also dependent on its own catalytic activity. Inhibition of p38 MAPK activity revealed its involvement in cAMP-mediated activating transcription factor-2 (ATF-2) phosphorylation, Fas ligand messenger RNA (mRNA) up-regulation, and cell death. These results identify a mechanism through which NE stimulation of the βAR/Gs/PKA pathway activates p38 MAPK, which can be potentiated by autophosphorylation, and leads to changes in T-cell dynamics, in part through the regulation of Fas ligand mRNA expression.
Collapse
Affiliation(s)
- Melissa D Lajevic
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
23
|
Lee JS, Lee MO, Moon BH, Shim SH, Fornace AJ, Cha HJ. Senescent growth arrest in mesenchymal stem cells is bypassed by Wip1-mediated downregulation of intrinsic stress signaling pathways. Stem Cells 2010; 27:1963-75. [PMID: 19544416 DOI: 10.1002/stem.121] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Human mesenchymal stem cells (hMSCs) have been widely studied as a source of primary adult stem cells for cell therapy because of their multidifferentiation potential; however, the growth arrest (also known as "premature senescence") often found in hMSCs cultured in vitro has been a major obstacle to the in-depth characterization of these cells. In addition, the inability to maintain constant cell growth hampers the development of additional genetic modifications aimed at achieving desired levels of differentiation to specific tissues; however, the molecular mechanisms that govern this phenomenon remain unclear, with the exception of a few studies demonstrating that induction of p16INK4a is responsible for this senescence-like event. Here, we observed that the premature growth arrest in hMSCs occurs in parallel with the induction of p16INK4a, following abrogation of inhibitory phosphorylation of retinoblastoma protein. These stress responses were concurrent with increased formation of reactive oxygen species (ROSs) from mitochondria and increased p38 mitogen-activated protein kinase (MAPK) activity. The introduction of Wip1 (wild-type p53 inducible phosphatase-1), a well-studied stress modulator, significantly lowered p16INK4a expression and led to p38 MAPK inactivation, although it failed to affect the levels of ROSs. Moreover, the suppression of stress responses by Wip1 apparently extended the life span of hMSCs, compared with control conditions, while maintaining their multilineage differentiation potential. Based on these results, we suggest that senescent growth arrest in hMSCs may result from activation of stress signaling pathways and consequent onset of stress responses, due in part to ROS production during prolonged in vitro culture.
Collapse
Affiliation(s)
- Ji-Seon Lee
- Stem Cell Research Laboratory, CHA Stem Cell Institute, Seoul, Korea
| | | | | | | | | | | |
Collapse
|