Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Immunol Methods ; 450: 50-57, 2017 11.
Article in English | MEDLINE | ID: mdl-28757372

ABSTRACT

Two novel magnetic agarose bead based assays have been developed to measure complement component C5 interaction with C3b and the Factor I Modules (FIMs) of C7. One innovation was to couple C3b onto the magnetic agarose bead using the alternative pathway C3 convertase, which resulted in a linkage of the ligand by a covalent ester bond. A second innovation was to employ nickel ion charged N,N,N'-tris(carboxymethyl)ethylene-diamine-magnetic agarose to capture recombinantly prepared C7 FIMs that were expressed with an oligo-histidine linker followed by an acidic domain that provided a spacer enabling the C7 modules exposure to C5. Detection was brought about by peroxidase coupled to C5. Both assays exhibited adequate statistics suitable for screening. As examples of the utility of these new methods, we chose to examine influence of natural products on C5 interaction. Fucoidan and ß-glucans were observed to inhibit C3b-C5 interaction, and dextran sulfate was similarly active; however, rosmarinic acid had no measurable effect. In contrast only ß-glucans from two species of macrofungi were able to interfere with interaction of C5 with the FIMs of C7.


Subject(s)
Complement Activation , Complement C3b/immunology , Complement C5/immunology , Complement C7/immunology , Immunologic Techniques , Magnetics , Complement Activation/drug effects , Complement C3-C5 Convertases/metabolism , Complement C3b/metabolism , Complement C5/metabolism , Complement C7/metabolism , Complement Hemolytic Activity Assay , Complement Inactivating Agents/pharmacology , Dextran Sulfate/pharmacology , Hemolysis , Humans , Iron/chemistry , Polysaccharides/pharmacology , Protein Binding , Sepharose/chemistry , beta-Glucans/pharmacology
2.
Protein Expr Purif ; 121: 118-24, 2016 May.
Article in English | MEDLINE | ID: mdl-26826315

ABSTRACT

A method is reported to purify Fibulin-1 from human plasma resulting in a 36% recovery. The steps involve removal of the cryoglobulin and the vitamin K dependent proteins followed by polyethylene glycol and ammonium sulfate precipitations, DEAE-Sephadex column chromatography and finally Factor H-Sepharose affinity purification. The procedure is designed to be integrated into an overall scheme for the isolation of over 30 plasma proteins from a single batch of human plasma. Results from mass spectroscopy, SDS-PAGE, and Western blotting indicate that human plasma Fibulin-1 is a single chain of the largest isotype. Functional binding assays demonstrated calcium ion dependent interaction of Fibulin-1 for fibrinogen, fibronectin, and Factor H. The procedure described is the first to our knowledge that enables a large scale purification of Fibulin-1 from human plasma.


Subject(s)
Calcium-Binding Proteins/blood , Chromatography, Affinity , Complement Factor H/chemistry , Mass Spectrometry , Ammonium Sulfate/chemistry , Calcium-Binding Proteins/chemistry , Fibrinogen/chemistry , Fibronectins/chemistry , Humans , Sepharose/chemistry
3.
World J Stem Cells ; 7(8): 1090-108, 2015 Sep 26.
Article in English | MEDLINE | ID: mdl-26435769

ABSTRACT

The complement pathway is best known for its role in immune surveillance and inflammation. However, its ability of opsonizing and removing not only pathogens, but also necrotic and apoptotic cells, is a phylogenetically ancient means of initiating tissue repair. The means and mechanisms of complement-mediated tissue repair are discussed in this review. There is increasing evidence that complement activation contributes to tissue repair at several levels. These range from the chemo-attraction of stem and progenitor cells to areas of complement activation, to increased survival of various cell types in the presence of split products of complement, and to the production of trophic factors by cells activated by the anaphylatoxins C3a and C5a. This repair aspect of complement biology has not found sufficient appreciation until recently. The following will examine this aspect of complement biology with an emphasis on the anaphylatoxins C3a and C5a.

4.
Adv Cancer Res ; 123: 149-89, 2014.
Article in English | MEDLINE | ID: mdl-25081529

ABSTRACT

The fate of both endogenous and transplanted stem cells is dependent on the functional status of the regulatory local microenvironment, which is compromised by disease and therapeutic intervention. The glycosaminoglycan hyaluronan (HA) is a critical component of the hematopoietic microenvironment. We summarize recent advances in our understanding of the role of HA in regulating mesenchymal stem cells, osteoblasts, fibroblasts, macrophages, and endothelium in bone marrow (BM) and their crosstalk within the hematopoietic microenvironment. HA not only determines the volume, hydration, and microfluidics of the BM interstitial space, but also, via interactions with specific receptors, regulates multiple cell functions including differentiation, migration, and production of regulatory factors. The effects of HA are dependent on the polymer size and are influenced by the formation of complexes with other molecules. In healthy BM, HA synthases and hyaluronidases form a molecular network that maintains extracellular HA levels within a discrete physiological window, but HA homeostasis is often perturbed in pathological conditions, including hematological malignancies. Recent studies have suggested that HA synthases may have functions beyond HA production and contribute to the intracellular regulatory machinery. We discuss a possible role for HA synthases, intracellular and extracellular HA in the malignant BM microenvironment, and resistance to therapy.


Subject(s)
Bone Marrow Cells/metabolism , Hematopoietic Stem Cells/metabolism , Hyaluronic Acid/physiology , Leukemia/metabolism , Animals , Aorta/pathology , Bone Marrow/metabolism , Bone Marrow Cells/cytology , Cell Differentiation , Cell Movement , Fibroblasts/cytology , Glucuronosyltransferase/metabolism , Homeostasis , Humans , Hyaluronan Synthases , Hyaluronic Acid/chemistry , Macrophages/cytology , Mice , Mice, Knockout , Muscle, Smooth/cytology , Osteoblasts/cytology , Osteoclasts/cytology , Polymers/chemistry , Protein Binding , Stem Cells/cytology , Time Factors
5.
Stem Cells Transl Med ; 3(2): 229-40, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24396034

ABSTRACT

Human neural stem cells (hNSCs) hold great potential for treatment of a wide variety of neurodegenerative and neurotraumatic conditions. Heretofore, administration has been through intracranial injection or implantation of cells. Because neural stem cells are capable of migrating to the injured brain from the intravascular space, it seemed feasible to administer them intravenously if their ability to circumvent the blood-brain barrier was enhanced. In the present studies, we found that interactions of hNSCs in vitro on the luminal surface of human umbilical vein endothelial cells was enhanced following enforced expression of cutaneous lymphocyte antigen on cell surface moieties by incubation of hNSCs with fucosyltransferase VI and GDP-fucose (fhNSCs). Interestingly, ex vivo fucosylation of hNSCs not only did not improve the cells homing into the brain injured by stroke following intravenous administration but also increased mortality of rats compared with the nonfucosylated hNSC group. Efforts to explain these unexpected findings using a three-dimensional flow chamber device revealed that transmigration of fhNSCs (under conditions of physiological shear stress) mediated by stromal cell-derived factor 1α was significantly decreased compared with controls. Further analysis revealed that hNSCs poorly withstand physiological shear stress, and their ability is further decreased following fucosylation. In addition, fhNSCs demonstrated a higher frequency of cellular aggregate formation as well as a tendency for removal of fucose from the cell surface. In summary, our findings suggest that the behavior of hNSCs in circulation is different from that observed with other cell types and that, at least for stroke, intravenous administration is a suboptimal route, even when the in vitro rolling ability of hNSCs is optimized by enforced fucosylation.


Subject(s)
Blood-Brain Barrier/cytology , Endothelial Cells/cytology , Neural Stem Cells/cytology , Neural Stem Cells/transplantation , Stroke/therapy , Veins/cytology , Animals , Cell Communication , Cell Movement/physiology , Cell Survival/physiology , Diffusion Chambers, Culture , Disease Models, Animal , Endothelial Cells/physiology , Female , Fucose/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Injections, Intravenous , Neural Stem Cells/physiology , Rats , Rats, Sprague-Dawley , Stress, Mechanical , Stroke/pathology , Veins/physiology
6.
J Biol Chem ; 287(30): 25419-33, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22654110

ABSTRACT

The contribution of hyaluronan (HA) to the regulatory network of the hematopoietic microenvironment was studied using knock-out mice of three hyaluronan synthase genes (Has1, Has2, and Has3). The number of hematopoietic progenitors was decreased in bone marrow and increased in extramedullary sites of Prx1-Cre;Has2(flox/flox);Has1(-/-);Has3(-/-) triple knock-out (tKO) mice as compared with wild type (WT) and Has1(-/-);Has3(-/-) double knock-out (dKO) mice. In line with this observation, decreased hematopoietic activity was observed in long term bone marrow cultures (LTBMC) from tKO mice, whereas the formation of the adherent layer and generation of hematopoietic cells in WT and dKO cultures was not different. 4-Methylumbelliferone (4MU) was used to pharmacologically inhibit the production of HA in LTBMC. Treatment with 4MU inhibited HA synthesis, decreased expression of HAS2 and HAS3, and eliminated hematopoiesis in LTBMC, and this effect was alleviated by the addition of exogenous HA. Exogenous HA also augmented the cell motility in LTBMC, which correlated with the HA-stimulated production of chemokines and growth factors. Conditioned media from HA-induced LTBMC enhanced the chemotaxis of hematopoietic stem/progenitor cells (HSPC) in response to SDF-1. Exposure of endothelial cells to 4MU decreased their ability to support HSPC rolling and adhesion. In addition, migration of transplanted HSPC into the marrow of 4MU-pretreated mice was lower than in untreated mice. Collectively, the results suggest that HA depletion reduces the ability of the microenvironment to support HSPC, and confirm a role for HA as a necessary regulatory element in the structure of the hematopoietic microenvironment.


Subject(s)
Bone Marrow/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Hyaluronic Acid/metabolism , Stem Cell Niche/physiology , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cells, Cultured , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Chemotaxis/drug effects , Chemotaxis/physiology , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Humans , Hyaluronan Synthases , Hyaluronic Acid/genetics , Hymecromone/analogs & derivatives , Hymecromone/pharmacology , Mice , Mice, Knockout , Stem Cell Niche/drug effects
7.
J Biol Chem ; 287(13): 10210-10222, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22267737

ABSTRACT

The complement membrane attack complex (MAC) is formed by the sequential assembly of C5b with four homologous proteins as follows: one copy each of C6, C7, and C8 and 12-14 copies of C9. Together these form a lytic pore in bacterial membranes. C6 through C9 comprise a MAC-perforin domain flanked by 4-9 "auxiliary" domains. Here, we report the crystal structure of C6, the first and longest of the pore proteins to be recruited by C5b. Comparisons with the structures of the C8αßγ heterodimer and perforin show that the central domain of C6 adopts a "closed" (perforin-like) state that is distinct from the "open" conformations in C8. We further show that C6, C8α, and C8ß contain three homologous subdomains ("upper," "lower," and "regulatory") related by rotations about two hinge points. In C6, the regulatory segment includes four auxiliary domains that stabilize the closed conformation, inhibiting release of membrane-inserting elements. In C8ß, rotation of the regulatory segment is linked to an opening of the central ß-sheet of its clockwise partner, C8α. Based on these observations, we propose a model for initiation and unidirectional propagation of the MAC in which the auxiliary domains play key roles: in the assembly of the C5b-8 initiation complex; in driving and regulating the opening of the ß-sheet of the MAC-performin domain of each new recruit as it adds to the growing pore; and in stabilizing the final pore. Our model of the assembled pore resembles those of the cholesterol-dependent cytolysins but is distinct from that recently proposed for perforin.


Subject(s)
Complement C6/chemistry , Complement Membrane Attack Complex , Models, Biological , Models, Molecular , Complement C6/metabolism , Complement System Proteins/chemistry , Complement System Proteins/metabolism , Crystallography, X-Ray , Humans , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship
8.
Immunology ; 135(4): 287-98, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22117697

ABSTRACT

The observation that human monocytes cultured in the presence of the chemokine CCL18 showed increased survival, led us to profile cytokine expression in CCL18-stimulated versus control cultures. CCL18 caused significantly increased expression of chemokines (CXCL8, CCL2, CCL3 and CCL22), interleukin-10 (IL-10) and platelet-derived growth factor, but no up-regulation of M1 cytokines IL-1ß or IL-12. CCL18-stimulated monocytes matured into cells with morphological resemblance to IL-4-stimulated macrophages, and expressed the monocyte marker CD14 as well the M2 macrophage markers CD206 and 15-lipoxygenase, but no mature dendritic cell markers (CD80, CD83 or CD86). Functionally, CCL18-stimulated macrophages showed a high capacity for unspecific phagocytosis and for pinocytosis, which was not associated with an oxidative burst. These findings suggest that CCL18-activated macrophages stand at the cross-roads between inflammation and its resolution. The chemokines that are produced in response to CCL18 are angiogenic and attract various leucocyte populations, which sustain inflammation. However, the capacity of these cells to remove cellular debris without causing oxidative damage and the production of the anti-inflammatory IL-10 will initiate termination of the inflammatory response. In summary, CCL18 induces an M2 spectrum macrophage phenotype in the absence of IL-4.


Subject(s)
Cell Differentiation/drug effects , Chemokines, CC/pharmacology , Macrophages/immunology , Monocytes/cytology , Animals , Cells, Cultured , Chemokines/metabolism , Chemokines, CC/immunology , Guinea Pigs , Humans , Interleukin-10/metabolism , Macrophages/cytology , Mice , Monocytes/drug effects , Monocytes/immunology , Phagocytosis
9.
Front Biosci (Landmark Ed) ; 16(6): 2271-88, 2011 06 01.
Article in English | MEDLINE | ID: mdl-21622176

ABSTRACT

Mesenchymal stem cells (MSC) are multipotent stem cells that hold promise for an expanding list of therapeutic uses, not only due to their ability to differentiate into all connective tissues including bone, fat and cartilage, but additionally due to their trophic and anti-inflammatory effects which contribute to healing and tissue regeneration. Ongoing research is starting to illuminate important aspects of the microenvironmental niche, which supports MSC self-renewal. In this review, we summarize recent findings on cellular structures and molecular pathways that are involved in regulation of MSC self-renewal versus differentiation, and in retention of MSCs within the niche versus mobilization and recruitment to sites of injury. In addition, the contribution of MSCs to the structure and function of hematopoietic and cancerous niches is discussed.


Subject(s)
Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Animals , Cell Differentiation , Cell Proliferation , Cytokines/physiology , Extracellular Matrix/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans , Intercellular Signaling Peptides and Proteins/physiology , Models, Biological , Neoplasms/etiology , Neoplasms/pathology , Neoplasms/therapy , Signal Transduction , Wound Healing/physiology , Wounds and Injuries/pathology , Wounds and Injuries/physiopathology
10.
Protein Expr Purif ; 79(1): 72-80, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21550406

ABSTRACT

An expression method has been developed to produce soluble cationic polypeptides in Escherichia coli while avoiding inclusion body deposition. For this technique the recombinant product is linked through a thrombin or factor Xa susceptible bond to the amino-terminal domain of the precursor of eosinophil major basic protein (MBP). This N-terminal domain is strongly acidic and is apparently able to shield eosinophils from the potentially injurious activities of MBP. It was reasoned that constructs of this acidic domain with small heterologous cationic proteins expressed in E. coli could result in soluble expression while preventing trafficking and packaging into insoluble inclusion bodies. This has been demonstrated using four examples: complement C5a, CCL18, fibroblast growth factor-ß, and leukemia inhibitory factor, whose isoelectric points range from 8.93 to 9.59. Further general applicability of this technique has been shown by using two different expression systems, one which encodes an amino-terminal oligo-histidine leash, and another that codes for an amino-terminal glutathione-S-transferase. Thus the utility of coupling MAP to cationic polypeptides for the purpose of soluble heterologous protein expression in E. coli has been demonstrated.


Subject(s)
Cloning, Molecular/methods , Eosinophil Major Basic Protein/genetics , Escherichia coli/genetics , Recombinant Fusion Proteins/genetics , Chemokines, CC/genetics , Chemokines, CC/isolation & purification , Complement C5a/genetics , Complement C5a/isolation & purification , Eosinophil Major Basic Protein/isolation & purification , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/isolation & purification , Gene Expression , HEK293 Cells , Humans , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/isolation & purification , Recombinant Fusion Proteins/isolation & purification , Solubility
11.
Mol Cancer Ther ; 9(11): 3024-32, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20833754

ABSTRACT

Bone marrow hypoplasia and pancytopenia are among the most undesirable sequelae of chemotherapy for the treatment of cancer. We recently showed that hyaluronan (HA) facilitates hematopoietic recovery in tumor-free animals receiving chemotherapeutic agents. However, following a chemotherapeutic regimen in tumor-bearing animals, it is possible that residual tumor cells might respond to systemic injections of HA. Thus, in this study, we investigated the effect of HA on the regrowth of residual tumor cells following chemotherapy. As a model, we used the HCT-8 human colon carcinoma cell line, which expresses the HA receptor CD44, binds exogenous HA, and is susceptible to a chemotherapy protocol containing irinotecan and 5-fluorouracil in a human/mouse xenograft model. HCT-8 cells were implanted in severe combined immunodeficient mice, followed by irinotecan/5-fluorouracil treatment. After three rounds of chemotherapy, residual tumors were allowed to regrow in the presence or absence of HA. The dynamics of tumor regrowth in the group treated with HA was slower compared with the control group. By week 5 after tumor implantation, the difference in the size of regrown tumors was statistically significant and correlated with lower proliferation and higher apoptosis in HA-treated tumors as compared with controls. This finding provides evidence that HA treatment does not stimulate but delays the growth of residual cancer cells, which is an important parameter in establishing whether the use of HA can enhance current chemotherapeutic strategies.


Subject(s)
Carcinoma/drug therapy , Colonic Neoplasms/drug therapy , Hyaluronic Acid/pharmacology , Neoplasm Recurrence, Local/prevention & control , Animals , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chemotherapy, Adjuvant , Colonic Neoplasms/pathology , Cytostatic Agents/administration & dosage , Cytostatic Agents/pharmacology , Drug Administration Schedule , Female , Humans , Hyaluronic Acid/administration & dosage , Mice , Mice, SCID , Models, Biological , Xenograft Model Antitumor Assays
12.
J Stem Cells ; 5(1): 9-21, 2010.
Article in English | MEDLINE | ID: mdl-20861924

ABSTRACT

Hyaluronan (HA) is an important component of the microenvironment in bone marrow, but its role in regulation of the development of hematopoietic cells is not well understood. To address the role of HA in regulation of human embryonic stem cell (hESC) differentiation into the hematopoietic lineage, we screened for genes encoding components of the HA pathway. Using gene arrays, we found that HA synthases and HA receptors are expressed in both undifferentiated and differentiating hESCs. Enzymatic degradation of HA resulted in decreased numbers of hematopoietic progenitors and lower numbers of CD45+ cells generated in HA-deprived embryoid bodies (EBs). In addition, deprivation of HA resulted in the inhibition of generation of CD31+ cells, stromal fibroblast-like cells and contracting myocytes in EBs. RT-PCR and immunocytochemistry revealed that HA deprivation did not influence the dynamics of OCT4 expression, but decreased the expression of BRY, an early mesoderm marker, and BMP2, a later mesoderm marker in differentiating EBs. In addition, the endoderm markers α-FP and SOX17 were decreased, whereas the expression of the ectoderm markers GFAP and FGF5 was higher in HA-deprived cultures. Our findings indicate that endogenously produced HA contributes to the network that regulates the differentiation of hESC and the generation of mesodermal lineage in general and hematopoietic cells specifically.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Hematopoietic System/cytology , Hyaluronic Acid/physiology , Biomarkers/metabolism , Cell Lineage , Cells, Cultured , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Developmental , Hematopoietic System/drug effects , Hematopoietic System/metabolism , Humans , Immunoenzyme Techniques , Mesoderm/cytology , Mesoderm/drug effects , Mesoderm/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
Eur J Immunol ; 39(11): 3181-94, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19750480

ABSTRACT

LL-37, derived from human cathelicidin, stimulates immune responses in neutrophils. Although FPR2 and P2X7 were proposed as LL-37 receptors, we have shown that among 21 neutrophil receptors only CXCR2 was down-regulated by LL-37. LL-37 functions similarly to CXCR2-specific chemokines CXCL1 and CXCL7 in terms of receptor down-regulation and intracellular calcium mobilization on freshly isolated neutrophils. Neutrophils pretreated with CXCL8, a chemokine that binds both CXCR1/2, completely blocked the calcium mobilization in response to LL-37, while LL-37 also partially inhibited (125)I-CXCL8 binding to neutrophils. SB225002, a selective CXCR2 antagonist, blocked LL-37-induced calcium mobilization and migration of neutrophils. LL-37 stimulates calcium mobilization in CXCR2-transfected HEK293 cells, CXCR2(+) THP-1 cells and monocytes, but not in CXCR1-transfected HEK293 cells. WKYMVm peptide (ligand for FPR2) does not block LL-37-stimulated calcium flux in either THP-1 (FPR2(-)) or monocytes (FPR2(high)), further confirming the specificity of LL-37 for CXCR2 and not FPR2. Among all ligands tested (ATP, BzATP, WKYMVm, CXCL1, and LL-37), only LL-37 stimulated migration of monocytes (CXCR2(+) and FPR2(+)) and migration was inhibited by the CXCR2 inhibitor SB225002. Moreover, CXCR2 but not CXCR1 was internalized in LL-37-treated neutrophils. Thus, our data provide evidence that LL-37 may act as a functional ligand for CXCR2 on human neutrophils.


Subject(s)
Antimicrobial Cationic Peptides/immunology , Neutrophils/immunology , Receptors, Interleukin-8B/immunology , Antimicrobial Cationic Peptides/metabolism , Calcium/metabolism , Cell Separation , Down-Regulation , Flow Cytometry , Humans , Ligands , Microscopy, Confocal , Neutrophil Infiltration/immunology , Neutrophils/metabolism , Receptors, Interleukin-8B/metabolism , Cathelicidins
14.
J Immunol ; 182(6): 3827-36, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19265162

ABSTRACT

Mesenchymal stem cells (MSCs) have a great potential for tissue repair, especially if they can be delivered efficiently to sites of tissue injury. Since complement activation occurs whenever there is tissue damage, the effects of the complement activation products C3a and C5a on MSCs were examined. Both C3a and C5a were chemoattractants for human bone marrow-derived MSCs, which expressed both the C3a receptor (C3aR) and the C5a receptor (C5aR; CD88) on the cell surface. Specific C3aR and C5aR inhibitors blocked the chemotactic response, as did pertussis toxin, indicating that the response was mediated by the known anaphylatoxin receptors in a G(i) activation-dependent fashion. While C5a causes strong and prolonged activation of various signaling pathways in many different cell types, the response observed with C3a is generally transient and weak. However, we show herein that in MSCs both C3a and C5a caused prolonged and robust ERK1/2 and Akt phosphorylation. Phospho-ERK1/2 was translocated to the nucleus in both C3a and C5a-stimulated MSCs, which was associated with subsequent phosphorylation of the transcription factor Elk, which could not be detected in other cell types stimulated with C3a. More surprisingly, the C3aR itself was translocated to the nucleus in C3a-stimulated MSCs, especially at low cell densities. Since nuclear activation/translocation of G protein-coupled receptors has been shown to induce long-term effects, this novel observation implies that C3a exerts far-reaching consequences on MSC biology. These results suggest that the anaphylatoxins C3a and C5a present in injured tissues contribute to the recruitment of MSCs and regulation of their behavior.


Subject(s)
Chemotactic Factors/physiology , Complement C3a/physiology , Complement C5a/physiology , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/immunology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Cell Line , Cells, Cultured , Chemotaxis, Leukocyte/immunology , Humans , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Anaphylatoxin C5a , Receptors, Complement/biosynthesis , Receptors, Complement/genetics , Time Factors
15.
J Stem Cells ; 4(3): 147-60, 2009.
Article in English | MEDLINE | ID: mdl-20232600

ABSTRACT

Embryonic stem cells (ESCs) offer a powerful in vitro model to study mechanisms implicated in cell fate decision. Developmental pathways by which pluripotent ESCs become committed to specific lineages are reflected in dynamic changes of signaling and transcriptional programs. However, the mechanisms that govern the regulatory intracellular networks underlying lineage fate decisions and differentiation programs remain poorly understood and differ significantly in different species. In this review we analyze the current understanding of the signaling mechanisms and transcriptional regulation of differentiation of murine and human ESCs into the mesoderm.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Mesenchymal Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Animals , Humans , Signal Transduction/genetics , Transcription Factors/metabolism
16.
J Stem Cells ; 4(4): 191-202, 2009.
Article in English | MEDLINE | ID: mdl-20720593

ABSTRACT

Hyaluronan (HA) is expressed by cells in bone marrow where it contributes to the regulation of hematopoietic homeostasis. In this study, we have demonstrated that exogenous low molecular weight HA (LMW HA) polymers mobilize leukocytes, but not hematopoietic progenitor cells, to peripheral blood within a 3 hour time period following HA administration. Mobilization of leukocytes correlated with increased extracellular MMP-9 concentrations induced by LMW HA, but not high molecular weight (HMW) HA. In contrast, HMW HA up-regulated TIMP-1 expression in bone marrow cells. In vitro, HMW HA did not influence SDF-1 - mediated chemotaxis of hematopoietic progenitors, whereas LMW HA polymers demonstrated inhibitory activity. These findings suggest that the effects of HA on cell motility depend on the size of the HA polymers and on the type of target cells.


Subject(s)
Bone Marrow Cells/drug effects , Cell Movement/drug effects , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/drug effects , Hyaluronic Acid/pharmacology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Movement/physiology , Cells, Cultured , Glucuronosyltransferase/metabolism , Hematopoietic Stem Cells/physiology , Hyaluronan Receptors/metabolism , Hyaluronan Synthases , Hyaluronic Acid/chemistry , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C
17.
J Stem Cells ; 4(4): 203-15, 2009.
Article in English | MEDLINE | ID: mdl-20720594

ABSTRACT

The efficient migration of mesenchymal stem cells (MSCs) to diseased tissues is required for the fulfillment of their regenerative potential. Recruitment of circulating cells into the damaged tissues is regulated by a complex network, which includes the non-neural cholinergic system. We found that human MSCs (hMSCs) express nicotinic acetylcholine receptor subunits alpha 7, beta 2 and beta 4. The receptor agonist nicotine caused calcium (Ca(++)) influx into hMSCs suggesting that the calcium ion channel alpha 7 homopolymer mediated this response. While high concentrations of nicotine (10(5)M) induced hMSC apoptosis, physiological concentrations (10(7)M) did not interfere with cell survival. At non-toxic concentrations, nicotine increased spontaneous migration of hMSCs, whereas chemotaxis of hMSCs toward C3a and bFGF in vitro and migration of intravenously infusion hMSCs into bone marrow and spleen in vivo were inhibited. The antagonist for the alpha 7 homopolymer, bungarotoxin, blocked the inhibitory effect of nicotine on chemotactic factor-induced migration of hMSCs. These findings reveal an involvement of the non-neural cholinergic system in regulation of hMSC migration.


Subject(s)
Cell Movement/physiology , Mesenchymal Stem Cells/physiology , Protein Subunits/metabolism , Receptors, Nicotinic/metabolism , Animals , Calcium/metabolism , Cell Movement/drug effects , Cells, Cultured , Chemokines/pharmacology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred NOD , Mice, SCID , Nicotine/metabolism , Nicotine/pharmacology , Nicotinic Agonists/metabolism , Nicotinic Agonists/pharmacology , Protein Subunits/genetics , Receptors, Nicotinic/genetics , alpha7 Nicotinic Acetylcholine Receptor
18.
Methods ; 45(2): 159-67, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18593612

ABSTRACT

This review is focused on methods that are used to derive hematopoietic cells from embryonic stem cells (ESCs). One of the strategies that have been recently used to achieve this goal is an approach of mimicking the hematopoietic niche in vitro by using hematopoiesis-supportive feeder cells, cocktails of soluble hematopoietic growth factors and a variety of matrices. While there is clear evidence that it is possible to derive hematopoietic stem cells (HSCs) and subsequently committed hematopoietic progenitors and mature cells from ESCs, there remains the need to address multiple issues including the efficiency of HSCs derivation in vitro and their proper functionality.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Animals , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cell Line/metabolism , Cell Lineage/genetics , Cell Separation/methods , Culture Media/pharmacology , Culture Media/standards , Diffusion Chambers, Culture , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/physiology , Hematopoiesis , Hematopoietic System/physiology , Humans , Mice , Stromal Cells/cytology , Stromal Cells/drug effects
19.
Immunology ; 125(2): 252-62, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18397265

ABSTRACT

Chemokines orchestrate the organization of leucocyte recruitment during inflammation and homeostasis. Despite growing knowledge of chemokine receptors, some orphan chemokine receptors are still not characterized. The gene CCRL2 encodes such a receptor that exists in two splice variants, CRAM-A and CRAM-B. Here, we report that CRAM is expressed by human peripheral blood and bone marrow B cells, and by different B-cell lines dependent on the B-cell maturation stage. Intriguingly, CRAM surface expression on the pre-B-cell lines Nalm6 and G2 is specifically upregulated in response to the inflammatory chemokine CCL5 (RANTES), a chemokine that is well known to play an important role in modulating immune responses. Although Nalm6 cells do not express any of the known CCL5 binding receptors, extracellular signal-regulated kinases 1 and 2 (ERK1/2) are phosphorylated upon CCL5 stimulation, suggesting a direct effect of CCL5 through the CRAM receptor. However, no calcium mobilization or migratory responses upon CCL5 stimulation are induced in B-cell lines or in transfected cells. Also, ERK1/2 phosphorylation cannot be inhibited by pertussis toxin, suggesting that CRAM does not couple to Gi proteins. Our results describe the expression of a novel, non-classical chemokine receptor on B cells that is potentially involved in immunomodulatory functions together with CCL5.


Subject(s)
B-Lymphocytes/immunology , Chemokine CCL5/immunology , Receptors, CCR/metabolism , B-Lymphocytes/ultrastructure , Calcium Signaling/immunology , Cell Differentiation/immunology , Cells, Cultured , Chemotaxis, Leukocyte/immunology , Humans , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/immunology , Phosphorylation , Protein Isoforms/immunology , Signal Transduction/immunology , Stress Fibers/immunology
20.
Breast Cancer Res Treat ; 110(2): 211-22, 2008 Jul.
Article in English | MEDLINE | ID: mdl-17687643

ABSTRACT

The chemokine receptor CXCR4 is functionally expressed on the cell surface of various cancer cells, and plays a role in cell proliferation and migration of these cells. Specifically, in breast cancer cells the CXCR4/CXCL12 axis has been implicated in cell migration in vitro and in metastasis in vivo, but the underlying signaling mechanisms are incompletely understood. The xenograft-derived MDA-MB-231 breast cancer cell line (231mfp), which was shown previously to grow more aggressively than the parent cells, showed increased CXCR4 expression at the mRNA, total protein and cell surface expression level. This correlated with an enhanced response to CXCL12, specifically in augmented and prolonged Akt activation in a G(i), Src family kinase and PI-3 kinase dependent fashion. 231mfp cells migrated towards CXCL12--in contrast to the parent cell line--and this chemotaxis was blocked by inhibition of G(i), Src family kinases, PI-3 kinase and interestingly, Akt itself, as could be shown with two pharmacological inhibitors, a dominant negative Akt construct and with Akt shRNA. Collectively, we have demonstrated that prolonged Akt activation is an important signaling pathway for breast cancer cells expressing CXCR4 and is necessary for CXCL12-dependent cell migration.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Nucleus/metabolism , Chemokine CXCL12/metabolism , Chemotaxis , Enzyme Activation , Humans , Matrix Metalloproteinases/metabolism , Models, Biological , Pertussis Toxin/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Receptors, CXCR4/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...