Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Int J Radiat Biol ; : 1-11, 2022 Apr 22.
Article in English | MEDLINE | ID: mdl-35394402

ABSTRACT

Purpose: This study aimed to evaluate the radiation-induced direct and bystander (BYS) responses of mesenchymal stem cells (MSCs) and to characterize these cells radiobiologically.Methods and materials: MSCs were irradiated (IR) and parameters related to DNA damage and cellular signaling were verified in a dose range from 0.5 to 15 Gy; also a transwell insert co-culture system was used to study medium-mediated BYS effects.Results: The main effects on directly IR cells were seen at doses higher than 6 Gy: induction of cell death, cell cycle arrest, upregulation of p21, and alteration of redox status. Irrespective of a specific dose, induction of micronuclei formation, H2AX phosphorylation, and decreased Akt expression also occurred. Thus, mTOR expression, cell senescence, nitric oxide generation, and calcium levels, in general were not significantly modulated by radiation. Data from the linear-quadratic model showed a high alpha/beta ratio, which is consistent with a more exponential survival curve. BYS effects from the unirradiated MSCs placed into companion wells with the directly IR cells, were not observed.Conclusions: The results can be interpreted as a positive outcome, meaning that the radiation damage is restricted to the directed IR MSCs not leading to off-target cell responses.

2.
Physiol Rep ; 9(9): e14863, 2021 05.
Article in English | MEDLINE | ID: mdl-33991464

ABSTRACT

Intermittent hypoxia (IH) is a feature of obstructive sleep apnea (OSA), a condition highly associated with hypertension-related cardiovascular diseases. Repeated episodes of IH contribute to imbalance of angiogenic growth factors in the hypertrophic heart, which is key in the progression of cardiovascular complications. In particular, the interaction between vascular endothelial growth factor (VEGF) and the kallikrein-kinin system (KKS) is essential for promoting angiogenesis. However, researchers have yet to investigate experimental models of IH that reproduce OSA, myocardial angiogenesis, and expression of KKS components. We examined temporal changes in cardiac angiogenesis in a mouse IH model. Adult male C57BI/6 J mice were implanted with Matrigel plugs and subjected to IH for 1-5 weeks with subsequent weekly histological evaluation of vascularization. Expression of VEGF and KKS components was also evaluated. After 3 weeks, in vivo myocardial angiogenesis and capillary density were decreased, accompanied by a late increase of VEGF and its type 2 receptor. Furthermore, IH increased left ventricular myocardium expression of the B2 bradykinin receptor, while reducing mRNA levels of B1 receptor. These results suggest that in IH, an unexpected response of the VEGF and KKS systems could explain the reduced capillary density and impaired angiogenesis in the hypoxic heart, with potential implications in hypertrophic heart malfunction.


Subject(s)
Cardiomegaly/metabolism , Hypoxia/metabolism , Kinins/metabolism , Myocardium/metabolism , Neovascularization, Physiologic , Sleep Apnea, Obstructive/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Capillaries/metabolism , Capillaries/physiology , Cardiomegaly/complications , Coronary Vessels/metabolism , Coronary Vessels/physiology , Hypoxia/complications , Kallikreins/genetics , Kallikreins/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Bradykinin/genetics , Receptors, Bradykinin/metabolism , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Sleep Apnea, Obstructive/complications , Vascular Endothelial Growth Factor A/genetics
3.
Stem Cell Rev Rep ; 17(3): 968-980, 2021 06.
Article in English | MEDLINE | ID: mdl-33206285

ABSTRACT

Ionizing radiation (IR) is an important medical tool. Despite the effects associated with high-dose radiation during or after treatment, as well as in accidental exposures, the direct or indirect effect of low-dose IR in cells remain poorly documented. IR can affect the tissue microenvironment, including mesenchymal stem cells (MSCs), which have high regenerative and immunomodulatory capacities. This study aimed to investigate the effect of low-dose IR in association with the inflammatory stimuli of TNF-α on the immunomodulatory capacity of MSCs. MSCs were irradiated with a low-dose IR, stimulated with TNF-α, and cultivated in a bystander system with murine spleen cells. The results showed that TNF-R1 is expressed in MSCs and is not affected, even in irradiated MSCs. However, irradiated MSCs produced reduced amounts of IL-6 and increased amounts of IL-10. The levels of PGE2 and NO• in MSCs were also increased when stimulated with TNF-α. Furthermore, conditioned media from irradiated MSCs reduced the proliferation of bystander lymphocytes and reduced the metabolic activity of macrophages. In addition, conditioned media from irradiated MSCs modulated the profile of cytokines in bystander spleen cells (lymphocytes and macrophages), reducing inflammatory and increasing anti-inflammatory cytokines, also increasing Treg cells. In conclusion, low-dose IR in association with an inflammatory stimulus affects the immunomodulatory properties of MSCs. In this way, the immunosuppressive capability of MSCs can be explored for several disease treatments where IR usually part of the context of the treatment. However, a complete understanding of the mechanisms underlying these interactions need further investigation. Graphical Abstract.


Subject(s)
Mesenchymal Stem Cells , Animals , Culture Media, Conditioned/pharmacology , Mice , Tumor Necrosis Factor-alpha
4.
Nutr Res Rev ; 31(2): 267-280, 2018 12.
Article in English | MEDLINE | ID: mdl-29983125

ABSTRACT

Micronutrients are indispensable for adequate metabolism, such as biochemical function and cell production. The production of blood cells is named haematopoiesis and this process is highly consuming due to the rapid turnover of the haematopoietic system and consequent demand for nutrients. It is well established that micronutrients are relevant to blood cell production, although some of the mechanisms of how micronutrients modulate haematopoiesis remain unknown. The aim of the present review is to summarise the effect of Fe, Mn, Ca, Mg, Na, K, Co, iodine, P, Se, Cu, Li and Zn on haematopoiesis. This review deals specifically with the physiological requirements of selected micronutrients to haematopoiesis, showing various studies related to the physiological requirements, deficiency or excess of these minerals on haematopoiesis. The literature selected includes studies in animal models and human subjects. In circumstances where these minerals have not been studied for a given condition, no information was used. All the selected minerals have an important role in haematopoiesis by influencing the quality and quantity of blood cell production. In addition, it is highly recommended that the established nutrition recommendations for these minerals be followed, because cases of excess or deficient mineral intake can affect the haematopoiesis process.


Subject(s)
Blood Cells/metabolism , Hematopoiesis/drug effects , Minerals/pharmacology , Nutritional Requirements , Trace Elements/pharmacology , Animals , Deficiency Diseases/complications , Humans , Nutritional Status
5.
J Nutr Biochem ; 55: 200-208, 2018 05.
Article in English | MEDLINE | ID: mdl-29554498

ABSTRACT

Magnesium (Mg2+) is a mineral with the ability to influence cell proliferation and to modulate inflammatory/immune responses, due to its anti-inflammatory properties. In addition, mesenchymal stem cells (MSCs) modulate the function of all major immune cell populations. Knowing that, the current work aimed to investigate the effects of Mg2+ enrichment, and its influence on the immunomodulatory capacity of MSCs. Murine C3H/10T1/2 MSCs were cultivated in media with different concentrations of Mg2+ (0, 1, 3 and 5 mM), in order to evaluate the effects of Mg2+ on MSC immunomodulatory properties, cell proliferation rates, expression of NFκB and STAT-3, production of IL-1ß, IL-6, TGF-ß, IL-10, PGE2 and NO, and TRPM7 expression. The results showed that TRPM7 is expressed in MSCs, but Mg2+, in the way that cells were cultivated, did not affect TRPM7 expression. Additionally, there was no difference in the intracellular concentration of Mg2+. Mg2+, especially at 5 mM, raised proliferation rates of MSCs, and modulated immune responses by decreasing levels of IL-1ß and IL-6, and by increasing levels of IL-10 and PGE2 in cells stimulated with LPS or TNF-α. In addition, MSCs cultured in 5 mM Mg2+ expressed lower levels of pNFκB/NFκB and higher levels of pSTAT-3/STAT-3. Furthermore, conditioned media from MSCs reduced lymphocyte and macrophage proliferation, but Mg2+ did not affect this parameter. In addition, conditioned media from MSCs cultured at 5 mM of Mg2+ modulated the production profile of cytokines, especially of IL-1ß and IL-6 in macrophages. In conclusion, Mg2+ is able to modulate some immunoregulatory properties of MSCs.


Subject(s)
Cytokines/metabolism , Magnesium/physiology , Mesenchymal Stem Cells/immunology , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cytokines/immunology , Dinoprostone/metabolism , Immunomodulation , Lymphocytes/cytology , Lymphocytes/drug effects , Macrophages/cytology , Macrophages/metabolism , Magnesium/pharmacology , Mesenchymal Stem Cells/drug effects , Mice , NF-kappa B/metabolism , Nitric Oxide/metabolism , STAT3 Transcription Factor/metabolism , TRPM Cation Channels/metabolism
6.
Nutrition ; 48: 96-104, 2018 04.
Article in English | MEDLINE | ID: mdl-29469028

ABSTRACT

OBJECTIVE: Dietary restriction (DR) is a nutritional intervention that exerts profound effects on biochemical and immunologic parameters, modulating some inflammatory properties. Glutamine (GLN) is a conditionally essential amino acid that can modulate inflammatory properties. However, there is a lack of data evaluating the effects of DR and GLN supplementation, especially in relation to inflammatory cytokine production and the expression of transcription factors such as nuclear factor (NF)-κB. METHODS: We subjected 3-mo-old male Balb/c mice to DR by reducing their food intake by 30%. DR animals lost weight and showed reduced levels of serum triacylglycerols, glucose, cholesterol, and calcium as well as a reduction in bone density. Additionally, blood, peritoneal, and spleen cellularity were reduced, lowering the number of peritoneal F4/80- and CD86-positive cells and the total number of splenic CD4- and CD8-positive cells. RESULTS: The production of interleukin (IL)-10 and the expression of NF-κB in splenic cells were not affected by DR or by GLN supplementation. However, peritoneal macrophages from DR animals showed reduced IL-12 and tumor necrosis factor-α production and increased IL-10 production with reduced phosphorylation of NF-κB expression. Additionally, GLN was able to modulate cytokine production by peritoneal cells from the control group, although no effects were observed in cells from the DR group. CONCLUSION: DR induces biochemical and immunologic changes, in particular by reducing IL-12 and tumor necrosis factor-α production by macrophages and clearly upregulating IL-10 production, whereas GLN supplementation did not modify these parameters in cells from DR animals.


Subject(s)
Caloric Restriction/adverse effects , Dietary Supplements , Glutamine/pharmacology , Animals , Interleukin-10/metabolism , Interleukin-12/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Phosphorylation , Spleen/cytology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
7.
Inflammopharmacology ; 26(3): 829-838, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29052795

ABSTRACT

The immune system is essential for the control and elimination of infections, and macrophages are cells that act as important players in orchestrating the various parts of the inflammatory/immune response. Amino acids play important role in mediating functionality of the inflammatory response, especially mediating macrophages functions and cytokines production. We investigated the influence of glutamine, taurine and their association on the modulation of inflammatory pathway markers in macrophages. The RAW 264.7 macrophage cell line was cultivated in the presence of glutamine and taurine and proliferation rates, cell viability, cell cycle phases, IL-1α, IL-6, IL-10 and TNF-α as well as H2O2 production and the expression of the transcription factor, NFκB, and its inhibitor, IκBα, were evaluated. Our results showed an increase in viable cells and increased proliferation rates of cells treated with glutamine concentrations over 2 mM, as well as cells treated with both glutamine and taurine. The cell cycle showed a higher percentage of cells in the phases S, G2 and M when they were treated with 2 or 10 mM glutamine, or with glutamine and taurine in cells stimulated with lipopolysaccharide. The pNFκB/NFκB showed reduced ratio expression when cells were treated with 10 mM of glutamine or with glutamine in association with taurine. These conditions also resulted in reduced TNF-α, IL-1α and H2O2 production, and higher production of IL-10. These findings demonstrate that glutamine and taurine are able to modulate macrophages inflammatory pathways, and that taurine can potentiate the effects of glutamine, illustrating their immunomodulatory properties.


Subject(s)
Glutamine/pharmacology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/metabolism , Taurine/pharmacology , Animals , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Synergism , Inflammation Mediators/immunology , Macrophages/immunology , Mice , RAW 264.7 Cells
8.
Clin Nutr ; 36(4): 1149-1157, 2017 08.
Article in English | MEDLINE | ID: mdl-27623433

ABSTRACT

Tissues that require a great supply of nutrients and possess high metabolic demands, such as lympho-hemopoietics tissues, are the first to be affected by protein malnutrition (PM). Thus, PM directly affects hemopoiesis and the production and function of immune cells. Consequently, malnourished individuals are more susceptible to infections. Mesenchymal stem cells (MSCs) have immunomodulatory properties and are important in the formation of lympho-hemopoietic stroma. Since an adequate supply of nutrients is essential to sustain stroma formation, which is mainly constituted of MSCs and differentiated cells originated from them, this study investigated whether PM would influence some biological and immunomodulatory aspects of MSCs. Two-month-old Balb/c mice were divided into control and malnourished groups receiving normoproteic or hypoproteic diets, respectively (12% and 2% of protein) for 28 days. MSCs obtained from control (MSCct) and malnourished (MSCmaln) animals were characterized. In addition, the proliferation rate and cell cycle protein expression were determined, but no differences in these parameters were observed. In order to evaluate whether PM affects the immunomodulatory properties of MSCs, the expression of NFκB and STAT-3, and the production of IL-1α, IL-1ß, IL-6, IL-10, TGF-ß and TNF-α by MSCs were assessed. MSCmaln expressed lower levels of NF-κB and the production of IL-1ß, IL-6 and TGF-ß was significantly influenced by PM. Furthermore, MSCct and MSCmaln culture supernatants affected lymphocyte and macrophage proliferation. However, MSCmaln did not reduce the production of IFN-γ nor stimulate the production of IL-10 in lymphocytes in the same manner as observed in MSCct. Overall, this study implied that PM modifies immunosuppressive properties of MSCs.


Subject(s)
Bone Marrow Cells/pathology , Gene Expression Regulation , Immunomodulation , Mesenchymal Stem Cells/pathology , Protein Deficiency/pathology , Protein-Energy Malnutrition/pathology , Stem Cells/pathology , Adaptive Immunity , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Proliferation , Cells, Cultured , Culture Media, Conditioned/metabolism , Cytokines/genetics , Cytokines/metabolism , Dietary Proteins , Immunity, Innate , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Mice, Inbred BALB C , Protein Deficiency/immunology , Protein Deficiency/metabolism , Protein-Energy Malnutrition/immunology , Protein-Energy Malnutrition/metabolism , Stem Cells/immunology , Stem Cells/metabolism
9.
Peptides ; 85: 41-45, 2016 11.
Article in English | MEDLINE | ID: mdl-27614284

ABSTRACT

Due to the cytotoxic effect of antimicrobial peptides (AMP) against several microorganism and tumor cells has been proposed their association with the immune system. However, just a few reports have shown this relationship. In this study, mice were treated with gomesin, a ß-hairpin AMP that exhibit high cytotoxicity against bacterial and tumor cells. Different effects in the immune system were observed, such as, decrease of CD3+ in T lymphocytes (Control: 17.7±1.4%; Gomesin: 7.67±1.2%) and in hematopoietic progenitors and increase of hematopoietic stem cell (Control: 0.046±0.004%; Gomesin: 0.067±0.003%), B220+ B lymphocytes (Control: 38.63±1.5%; Gomesin: 47.83±0.48%), and Mac-1+F4/80+ macrophages (Control: 11.76±3.4%; Gomesin: 27.13±4.0%). Additionally, macrophage increase was accompanied by an increase of macrophage phagocytosis (Control 20.85±1.53; Gomesin 31.32±1 Geometric mean), interleukin 6 (Control: 47.24±1.9ng/mL; Gomesin: 138.68±33.68ng/mL) and monocyte chemoattractant protein-1 (Control: 0.872±0.093ng/mL; Gomesin: 1.83±0.067ng/mL). Thus, this report showed immunomodulatory activity of gomesin in the immune system of mice.


Subject(s)
Antimicrobial Cationic Peptides/immunology , Cell Differentiation/genetics , Macrophage Activation/genetics , Myeloid Cells/metabolism , Animals , Antimicrobial Cationic Peptides/administration & dosage , Immune System/metabolism , Immunomodulation/genetics , Macrophage Activation/immunology , Mice , Monocytes/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
10.
Inflammation ; 39(6): 1883-1891, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27565164

ABSTRACT

Protein malnutrition (PM) is a major public health problem in developing countries, affecting the inflammatory response and increasing susceptibility to opportunistic infections. For this reason, an adequate nutritional intervention can improve the quality of life of patients. Glutamine (GLN) is a nonessential amino acid, but can be considered "conditionally essential" for macrophage function in stress situations, in which it plays a role in the improvement of the inflammatory response. Concerning this issue, in the current study, it was of interest to evaluate some biological aspects of peritoneal cells from a protein malnutrition (PM) mouse model challenged with lipopolysaccharide (LPS) and treated intravenously with GLN. Two-month-old male Balb/c mice were subjected to a low-protein diet (2 % protein) and stimulated intravenously with LPS 1 h prior to the injection of 0.75 mg/kg GLN. Malnourished animals showed a reduced number of total peritoneal cells. Malnourished animals stimulated with LPS or LPS plus GLN did not show differences in peritoneal cell counts; however, the control group showed increased cellularity after LPS stimulus, which was reversed after GLN injection. Further, in the animals from both groups stimulated with LPS, GLN decreased the circulating levels of TNF-α and the levels of TNF-α produced by peritoneal cells; additionally, GLN decreased the IL-10 circulating levels in the malnourished animals stimulated with LPS. In addition, peritoneal cells of the control and malnourished groups stimulated with LPS showed a negative modulation of the NFkB signaling pathway after GLN injection. In conclusion, this study shows that GLN has the capacity to reduce TNF-α synthesis as well as to act as a negative regulator of NFkB phosphorylation, leading to a positive outcome in the control of TNF-α production.


Subject(s)
Glutamine/administration & dosage , Protein-Energy Malnutrition/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Disease Models, Animal , Glutamine/therapeutic use , Interleukin-10/blood , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Peritoneum/cytology , Phosphorylation/drug effects , Protein-Energy Malnutrition/drug therapy , Tumor Necrosis Factor-alpha/analysis
11.
Toxicon ; 120: 22-8, 2016 Sep 15.
Article in English | MEDLINE | ID: mdl-27452928

ABSTRACT

Tityus serrulatus is the scorpion specie responsible for the majority of scorpion sting accidents in Brazil. Symptoms of envenomation by Tityus serrulatus range from local pain to severe systemic reactions such as cardiac dysfunction and pulmonary edema. Thus, this study has evaluated the participation of bronchial epithelial cells in the pulmonary effects of Tityus serrulatus scorpion venom (Tsv). Human bronchial epithelial cell line BEAS-2B were utilized as a model target and were incubated with Tsv (10 or 50 µg/mL) for 1, 3, 6 and 24 h. Effects on cellular response of venom-induce cytotoxicity were examined including cell viability, cell integrity, cell morphology, apoptosis/necrosis as well as cell activation through the release of pro-inflammatory cytokines IL-1ß, IL-6 and IL-8. Tsv caused a decrease in cell viability at 10 and 50 µg/mL, which was confirmed by lactate dehydrogenase (LDH) measurement. Flow cytometry analyses revealed necrosis as the main cell death pathway caused by Tsv. Furthermore, Tsv induced the release of IL-1ß, IL-6 and IL-8. Altogether, these results demonstrate that Tsv induces cytotoxic effects on bronchial epithelial cells, involving necrosis and release of pro-inflammatory cytokines, suggesting that bronchial epithelial cells may play a role in the pulmonary injury caused by Tsv.


Subject(s)
Bronchi/drug effects , Cytokines/biosynthesis , Scorpion Venoms/toxicity , Animals , Apoptosis/drug effects , Bronchi/cytology , Bronchi/metabolism , Cell Line , Cell Survival/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , In Vitro Techniques , Necrosis , Scorpions
12.
J Cell Physiol ; 231(6): 1313-20, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26517012

ABSTRACT

B lymphocytes are immune cells crucial for the maintenance and viability of the humoral response. Sleep is an essential event for the maintenance and integrity of all systems, including the immune system (IS). Thus, sleep deprivation (SD) causes problems in metabolism and homeostasis in many cell systems, including the IS. In this study, our goal was to determine changes in B lymphocytes from the bone marrow (BM) and spleen after SD. Three-month-old male Swiss mice were used. These mice were sleep deprived through the modified multiple platform method for different periods (24, 48, and 72 h), whereas another group was allowed to sleep for 24 h after 72 h of SD (rebound group) and a third group was allowed to sleep normally during the entire experiment. After this, the spleen and BM were collected, and cell analyses were performed. The numbers of B lymphocytes in the BM and spleen were reduced by SD. Additionally, reductions in the percentage of lymphocyte progenitors and their ability to form colonies were observed. Moreover, an increase in the death of B lymphocytes from the BM and spleen was associated with an increase in oxidative stress indicators, such as DCFH-DA, CAT, and mitochondrial SOD. Rebound was not able to reverse most of the alterations elicited by SD. The reduction in B lymphocytes and their progenitors by cell death, with a concomitant increase in oxidative stress, showed that SD promoted a failure in B lymphopoiesis.


Subject(s)
B-Lymphocytes/immunology , Bone Marrow/immunology , Lymphopoiesis , Precursor Cells, B-Lymphoid/immunology , Sleep Deprivation/immunology , Spleen/immunology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Biomarkers/metabolism , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Death , Disease Models, Animal , Lymphocyte Count , Male , Mice , Oxidative Stress , Precursor Cells, B-Lymphoid/metabolism , Precursor Cells, B-Lymphoid/pathology , Sleep Deprivation/metabolism , Sleep Deprivation/pathology , Spleen/metabolism , Spleen/pathology , Time Factors
13.
Front Biosci (Elite Ed) ; 8(1): 100-12, 2016 01 01.
Article in English | MEDLINE | ID: mdl-26709649

ABSTRACT

Erythropoiesis is a complex process that starts in the course of embryo formation and it is maintained throughout the life of an organism. During the fetal development, erythropoiesis arises from different body sites and erythroblast maturation occurs in the fetal liver. After birth, erythropoiesis and erythroblast maturation take place exclusively in the bone marrow, generating a lifetime reservoir of red blood cells (RBCs), which are responsible for transporting oxygen through the bloodstream to tissues and organs. Several transcription factors and cytokines, such as GATA-1, GATA-2, FOG-1 and erythropoietin (EPO), constitute an elaborated molecular network that regulates erythropoiesis as they are involved in the differentiation and maturation of RBCs. The profound understanding of erythropoiesis is fundamental to avoid, treat or even soften the effects of erythropoietic clinical disorders and may be useful to improve patients' well-being.


Subject(s)
Erythropoiesis , Vertebrates/physiology , Age Factors , Animals , Erythropoietin/physiology , Fetus/physiology , Hemoglobins/physiology , Iron/metabolism
14.
J Cell Biochem ; 116(7): 1334-40, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25735790

ABSTRACT

Several studies have shown the important actions of cytokine leptin that regulates food intake and energy expenditure. Additionally, the ability to modulate hematopoiesis has also been demonstrated. Previous reports have shown that some synthetic sequences of leptin molecules can activate leptin receptor. Herein, decapeptides encompassing amino acids from positions 98 to 122 of the leptin molecule were constructed to evaluate their effects on hematopoiesis. Among them, the synthetic peptide Lep(110-119)-NH2 (LEP F) was the only peptide that possessed the ability to increase the percentage of hematopoietic stem cells (HSC). Moreover, LEP F also produced an increase of granulocyte/macrophage colony-forming units and activated leptin receptor. Furthermore, LEP F also improves the grafting of HSC in bone marrow, but did not accelerate the recovery of bone marrow after ablation with 5-fluorouracil. These results show that LEP F is a positive modulator of the in vivo expansion of HSC and could be useful in bone marrow transplantation.


Subject(s)
Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Leptin/administration & dosage , Peptide Fragments/administration & dosage , Receptors, Leptin/metabolism , Animals , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Injections, Intraperitoneal , Janus Kinase 2/metabolism , Leptin/metabolism , Leptin/pharmacology , Mice , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Phosphorylation/drug effects
15.
Stem Cells ; 32(11): 2949-60, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24964894

ABSTRACT

There are a growing number of reports showing the influence of redox modulation in cellular signaling. Although the regulation of hematopoiesis by reactive oxygen species (ROS) and reactive nitrogen species (RNS) has been described, their direct participation in the differentiation of hematopoietic stem cells (HSCs) remains unclear. In this work, the direct role of nitric oxide (NO(•)), a RNS, in the modulation of hematopoiesis was investigated using two sources of NO(•) , one produced by endothelial cells stimulated with carbachol in vitro and another using the NO(•)-donor S-nitroso-N-acetyl-D,L-penicillamine (SNAP) in vivo. Two main NO(•) effects were observed: proliferation of HSCs-especially of the short-term HSCs-and its commitment and terminal differentiation to the myeloid lineage. NO(•)-induced proliferation was characterized by the increase in the number of cycling HSCs and hematopoietic progenitor cells positive to BrdU and Ki-67, upregulation of Notch-1, Cx43, PECAM-1, CaR, ERK1/2, Akt, p38, PKC, and c-Myc. NO(•)-induced HSCs differentiation was characterized by the increase in granulocytic-macrophage progenitors, granulocyte-macrophage colony forming units, mature myeloid cells, upregulation of PU.1, and C/EBPα genes concomitantly to the downregulation of GATA-3 and Ikz-3 genes, activation of Stat5 and downregulation of the other analyzed proteins mentioned above. Also, redox status modulation differed between proliferation and differentiation responses, which is likely associated with the transition of the proliferative to differentiation status. Our findings provide evidence of the role of NO(•) in inducing HSCs proliferation and myeloid differentiation involving multiple signaling.


Subject(s)
Bone Marrow Cells/metabolism , Cell Lineage , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Nitric Oxide/metabolism , Animals , Cell Proliferation/physiology , Gene Expression/physiology , Hematopoietic Stem Cells/cytology , Mice , Oxidation-Reduction , Reactive Oxygen Species/metabolism
16.
Cancer Cell Int ; 13(1): 123, 2013 Dec 23.
Article in English | MEDLINE | ID: mdl-24365069

ABSTRACT

Over the last few years, studies have suggested that oxidative stress plays a role in the regulation of hematopoietic cell homeostasis. In particular, the effects of hydrogen peroxide (H2O2) range from hematopoietic cell proliferation to cell death, depending on its concentration in the intracellular milieu. In this work, we evaluated the effects of an oxidative environment on normal and leukemic hematopoietic cells by stimulating normal human (umbilical cord blood) and murine (bone marrow) hematopoietic cells, as well as human myeloid leukemic cells (HL-60 lineage), upon H2O2 stimulus. Total cell populations and primitive subsets were evaluated for each cell type. H2O2 stimulus induces HL-60 cell death, whereas the viability of human and murine normal cells was not affected. The effects of H2O2 stimulus on hematopoietic stem/progenitor cell subsets were examined and the normal primitive cells were found to be unaffected; however, the percentage of leukemic stem cells (LSC) increased in response to H2O2, while clonogenic ability of these cells to generate myeloid clones was inhibited. In addition, H2O2 stimulus caused a decrease in the levels of p-AKT in HL-60 cells, which most likely mediates the observed decrease of viability. In summary, we found that at low concentrations, H2O2 preferentially affects both the LSC subset and total HL-60 cells without damage normal cells.

17.
Peptides ; 50: 24-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24090593

ABSTRACT

Leptin is a cytokine that regulates food intake, energy expenditure and hematopoiesis. Based on the tridimensional structure of the human leptin molecule, six fragments have been synthesized, (Ac-Lep23-47-NH2, [LEP1]; Ac-Lep48-71-NH2, [LEP2]; Ac-Lep72-88-NH2, [LEP3]; Ac-Lep92-115-NH2, [LEP4], Ac-[Ser(117)]-Lep116-140-NH2, [LEP5] and Ac-Lep141-164-NH2, [LEP6]), and their effects on hematopoiesis were evaluated. The mice were treated with 1mg/kg LEP5 for 3 days. The mature and primitive hematopoietic populations were quantified. We observed that the mature populations from the bone marrow and spleen were not affected by LEP5. However, the peptide caused at least a two-fold increase in the number of hematopoietic stem cells, the most primitive population of the bone marrow. Additionally, the number of granulocyte/macrophage colony-forming units produced by bone marrow cells in methylcellulose also increased by 40% after treatment with LEP5, and the leptin receptor was activated. These results show that the leptin fragment LEP5 is a positive modulator of the in vivo expansion of hematopoietic stem cells.


Subject(s)
Bone Marrow Cells/drug effects , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Leptin/pharmacology , Peptide Fragments/pharmacology , Spleen/drug effects , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Proliferation/drug effects , Gene Expression/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Peptide Fragments/chemical synthesis , Receptors, Leptin/agonists , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Spleen/cytology , Spleen/metabolism
18.
Exp Biol Med (Maywood) ; 238(4): 375-84, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23760003

ABSTRACT

It is well established that the excessive consumption of a high-fat diet (HFD) results in overweight, obesity and an increase in leptin concentrations, which triggers a chronic inflammatory condition that is associated with a high white blood cell count. Two-month-old male Wistar rats were fed a control (CON) diet or an HFD for 12 weeks. After this period, hemogram, myelogram and biochemical parameters were evaluated along with the cell cycle and the percentage of CD34(+) cells in the bone marrow as well as cell proliferation and differentiation assays and the production of stem cell factor, interleukin 3 (IL-3), granulocyte colony-stimulating factor (G-CSF) and granulocyte macrophage colony-stimulating factor (GM-CSF). The HFD animals exhibited leukocytosis and neutrophilia with increased C-reactive protein, leptin, cholesterol and triglyceride concentrations. In the HFD group, the bone marrow revealed myeloid hyperplasia, especially of the granulocytic compartment with a higher percentage of CD34(+) cells and a higher percentage of cells in the G2/S/M cell cycle phases. In addition, the HFD bone marrow cells had a higher capacity to proliferate and differentiate into granulocytic cells in an in vitro system and a higher capacity to produce IL-3 and G-CSF. These data led us to infer that the HFD induces leukocytosis and neutrophilia suggesting alterations in hematopoiesis system modulation.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Diet, High-Fat/adverse effects , Dietary Fats/pharmacology , Granulocyte Colony-Stimulating Factor/metabolism , Interleukin-3/metabolism , Leukocytosis/chemically induced , Animals , Bone Marrow Cells/drug effects , C-Reactive Protein/metabolism , Cells, Cultured , Cholesterol/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hyperplasia/chemically induced , Hyperplasia/metabolism , Hyperplasia/pathology , In Vitro Techniques , Leptin/metabolism , Leukocytosis/metabolism , Leukocytosis/pathology , Male , Models, Animal , Neutrophils/drug effects , Neutrophils/pathology , Rats , Rats, Wistar , Stem Cell Factor/metabolism , Triglycerides/metabolism
19.
J Biol Chem ; 288(19): 13641-54, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23511635

ABSTRACT

BACKGROUND: Kallikreins play a pivotal role in establishing prostate cancer. RESULTS: In contrast to the classical Kunitz plant inhibitor SbTI, the recombinant kallikrein inhibitor (rBbKIm) led to prostate cancer cell death, whereas fibroblast viability was not affected. CONCLUSION: rBbKIm shows selective cytotoxic effect and angiogenesis inhibition against prostate cancer cells. SIGNIFICANCE: New actions of rBbKIm may contribute to understanding the mechanisms of prostate cancer. Prostate cancer is the most common type of cancer, and kallikreins play an important role in the establishment of this disease. rBbKIm is the recombinant Bauhinia bauhinioides kallikreins inhibitor that was modified to include the RGD/RGE motifs of the inhibitor BrTI from Bauhinia rufa. This work reports the effects of rBbKIm on DU145 and PC3 prostate cancer cell lines. rBbKIm inhibited the cell viability of DU145 and PC3 cells but did not affect the viability of fibroblasts. rBbKIm caused an arrest of the PC3 cell cycle at the G0/G1 and G2/M phases but did not affect the DU145 cell cycle, although rBbKIm triggers apoptosis and cytochrome c release into the cytosol of both cell types. The differences in caspase activation were observed because rBbKIm treatment promoted activation of caspase-3 in DU145 cells, whereas caspase-9 but not caspase-3 was activated in PC3 cells. Because angiogenesis is important to the development of a tumor, the effect of rBbKIm in this process was also analyzed, and an inhibition of 49% was observed in in vitro endothelial cell capillary-like tube network formation. In summary, we demonstrated that different properties of the protease inhibitor rBbKIm may be explored for investigating the androgen-independent prostate cancer cell lines PC3 and DU145.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Survival/drug effects , Kallikreins/antagonists & inhibitors , Plant Proteins/pharmacology , Apoptosis/drug effects , Calcium Signaling , Caspase 3 , Caspase 9/metabolism , Cell Adhesion/drug effects , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Movement , Cytochromes c/metabolism , Fibroblasts/drug effects , Fibroblasts/physiology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Hydrophobic and Hydrophilic Interactions , Lipopolysaccharides/pharmacology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Prostatic Neoplasms , Recombinant Proteins/pharmacology , Trypsin Inhibitor, Kunitz Soybean/pharmacology
20.
Front Biosci (Elite Ed) ; 5(1): 130-40, 2013 01 01.
Article in English | MEDLINE | ID: mdl-23276976

ABSTRACT

Leukemias are the most common malignancy of childhood and have the highest mortality among aging people. Leukemias are a group of blood disorders characterized by an accumulation of leukemic cells in the peripheral blood of patients as a result of disturbances in proliferation and differentiation. Refractory leukemia remains the most common therapeutic challenge. In recent years, the presence of a cancer stem cell population in leukemias has been proposed as a cause for the refractory phenomenon. Insights into the cellular and molecular features of leukemia led to a new point of view in the choice of novel therapeutic agents. New agents for the treatment of this disease should selectively target leukemia stem cells or exhibit higher cytotoxic effects in cancer cells than in normal cells. A special interest is focused on anticancer agents from biological and natural sources that can be used in the treatment of leukemia. This review discusses the characteristics of some of these potential new agents.


Subject(s)
Antimicrobial Cationic Peptides/therapeutic use , Biological Factors/therapeutic use , Free Radicals/metabolism , Hematopoiesis/physiology , Hematopoietic System/metabolism , Leukemia/drug therapy , Receptors, Purinergic P2/therapeutic use , Signal Transduction/physiology , Depsipeptides , Humans , Indoles , Receptors, Purinergic P2/metabolism , Shikimic Acid/analogs & derivatives , Sirolimus , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...