Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Front Bioeng Biotechnol ; 11: 1258753, 2023.
Article in English | MEDLINE | ID: mdl-38033821

ABSTRACT

Many preclinical studies have shown that birth-associated tissues, cells and their secreted factors, otherwise known as perinatal derivatives (PnD), possess various biological properties that make them suitable therapeutic candidates for the treatment of numerous pathological conditions. Nevertheless, in the field of PnD research, there is a lack of critical evaluation of the PnD standardization process: from preparation to in vitro testing, an issue that may ultimately delay clinical translation. In this paper, we present the PnD e-questionnaire developed to assess the current state of the art of methods used in the published literature for the procurement, isolation, culturing preservation and characterization of PnD in vitro. Furthermore, we also propose a consensus for the scientific community on the minimal criteria that should be reported to facilitate standardization, reproducibility and transparency of data in PnD research. Lastly, based on the data from the PnD e-questionnaire, we recommend to provide adequate information on the characterization of the PnD. The PnD e-questionnaire is now freely available to the scientific community in order to guide researchers on the minimal criteria that should be clearly reported in their manuscripts. This review is a collaborative effort from the COST SPRINT action (CA17116), which aims to guide future research to facilitate the translation of basic research findings on PnD into clinical practice.

2.
Cells ; 12(3)2023 02 03.
Article in English | MEDLINE | ID: mdl-36766851

ABSTRACT

In this Special Issue of Cells, entitled "Immunomodulation by Mesenchymal Stem Cells 2020", you can find five excellent papers on the role of mesenchymal stem/stromal cells (MSCs) in immunomodulation, which also includes regenerative processes, such as wound healing [...].


Subject(s)
Mesenchymal Stem Cells , Wound Healing
3.
Front Immunol ; 13: 1045168, 2022.
Article in English | MEDLINE | ID: mdl-36466922

ABSTRACT

Hematopoietic allogeneic stem cell transplantation (allo-SCT) is a curative option for patients with hematological malignancies. However, due to disparities in major and minor histocompatibility antigens between donor and recipient, severe inflammatory complications can occur, among which chronic graft-versus-host disease (cGVHD) can be life-threatening. A classical therapeutic approach to the prevention and treatment of cGVHD has been broad immunosuppression, but more recently adjuvant immunotherapies have been tested. This review summarizes and discusses immunomodulatory approaches with T cells, including chimeric antigen receptor (CAR) and regulatory T cells, with natural killer (NK) cells and innate lymphoid cells (ILCs), and finally with mesenchymal stromal cells (MSC) and extracellular vesicles thereof. Clinical studies and pre-clinical research results are presented likewise.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Humans , Graft vs Host Disease/prevention & control , Immunity, Innate , Cell- and Tissue-Based Therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Killer Cells, Natural
4.
Front Bioeng Biotechnol ; 10: 981061, 2022.
Article in English | MEDLINE | ID: mdl-36185431

ABSTRACT

Perinatal tissues, mainly the placenta and umbilical cord, contain a variety of different somatic stem and progenitor cell types, including those of the hematopoietic system, multipotent mesenchymal stromal cells (MSCs), epithelial cells and amnion epithelial cells. Several of these perinatal derivatives (PnDs), as well as their secreted products, have been reported to exert immunomodulatory therapeutic and regenerative functions in a variety of pre-clinical disease models. Following experience with MSCs and their extracellular vesicle (EV) products, successful clinical translation of PnDs will require robust functional assays that are predictive for the relevant therapeutic potency. Using the examples of T cell and monocyte/macrophage assays, we here discuss several assay relevant parameters for assessing the immunomodulatory activities of PnDs. Furthermore, we highlight the need to correlate the in vitro assay results with preclinical or clinical outcomes in order to ensure valid predictions about the in vivo potency of therapeutic PnD cells/products in individual disease settings.

5.
Front Bioeng Biotechnol ; 10: 958669, 2022.
Article in English | MEDLINE | ID: mdl-36312547

ABSTRACT

Perinatal derivatives or PnDs refer to tissues, cells and secretomes from perinatal, or birth-associated tissues. In the past 2 decades PnDs have been highly investigated for their multimodal mechanisms of action that have been exploited in various disease settings, including in different cancers and infections. Indeed, there is growing evidence that PnDs possess anticancer and antimicrobial activities, but an urgent issue that needs to be addressed is the reproducible evaluation of efficacy, both in vitro and in vivo. Herein we present the most commonly used functional assays for the assessment of antitumor and antimicrobial properties of PnDs, and we discuss their advantages and disadvantages in assessing the functionality. This review is part of a quadrinomial series on functional assays for the validation of PnDs spanning biological functions such as immunomodulation, anticancer and antimicrobial, wound healing, and regeneration.

6.
Front Bioeng Biotechnol ; 10: 965006, 2022.
Article in English | MEDLINE | ID: mdl-35992360

ABSTRACT

Perinatal derivatives (PnD) are birth-associated tissues, such as placenta, umbilical cord, amniotic and chorionic membrane, and thereof-derived cells as well as secretomes. PnD play an increasing therapeutic role with beneficial effects on the treatment of various diseases. The aim of this review is to elucidate the modes of action of non-hematopoietic PnD on inflammation, angiogenesis and wound healing. We describe the source and type of PnD with a special focus on their effects on inflammation and immune response, on vascular function as well as on cutaneous and oral wound healing, which is a complex process that comprises hemostasis, inflammation, proliferation (including epithelialization, angiogenesis), and remodeling. We further evaluate the different in vitro assays currently used for assessing selected functional and therapeutic PnD properties. This review is a joint effort from the COST SPRINT Action (CA17116) with the intention to promote PnD into the clinics. It is part of a quadrinomial series on functional assays for validation of PnD, spanning biological functions, such as immunomodulation, anti-microbial/anti-cancer activities, anti-inflammation, wound healing, angiogenesis, and regeneration.

7.
Front Immunol ; 12: 676756, 2021.
Article in English | MEDLINE | ID: mdl-33995421

ABSTRACT

Chronic graft-versus-host disease (cGvHD) is an immune mediated late complication of allogeneic hematopoietic stem cell transplantation (alloHSCT). Discovery of adequate biomarkers could identify high-risk patients and provide an effective pre-emptive intervention or early modification of therapeutic strategy, thus reducing prevalence and severity of the disease among long-term survivors of alloHSCT. Inflammation, endothelial injury, and endothelial dysfunction are involved in cGvHD development. Altered levels of acute phase reactants have shown a strong correlation with the activity of several immune mediated disorders and are routinely used in clinical practice. Since elevated von Willebrand factor (VWF) and factor VIII (FVIII) levels have been described as acute phase reactants that may indicate endothelial dysfunction and inflammation in different settings, including chronic autoimmune diseases, they could serve as potential candidate biomarkers of cGvHD. In this review we focused on reported data regarding VWF and FVIII as well as other markers of inflammation and endothelial dysfunction, evaluating their potential role in cGvHD.


Subject(s)
Acute-Phase Proteins/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Factor VIII/metabolism , Graft vs Host Disease/blood , von Willebrand Factor/metabolism , Biomarkers/blood , Chronic Disease , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Hemostasis/immunology , Humans , Inflammation/blood , Transplantation, Homologous/adverse effects
8.
Front Bioeng Biotechnol ; 8: 610544, 2020.
Article in English | MEDLINE | ID: mdl-33392174

ABSTRACT

Progress in the understanding of the biology of perinatal tissues has contributed to the breakthrough revelation of the therapeutic effects of perinatal derivatives (PnD), namely birth-associated tissues, cells, and secreted factors. The significant knowledge acquired in the past two decades, along with the increasing interest in perinatal derivatives, fuels an urgent need for the precise identification of PnD and the establishment of updated consensus criteria policies for their characterization. The aim of this review is not to go into detail on preclinical or clinical trials, but rather we address specific issues that are relevant for the definition/characterization of perinatal cells, starting from an understanding of the development of the human placenta, its structure, and the different cell populations that can be isolated from the different perinatal tissues. We describe where the cells are located within the placenta and their cell morphology and phenotype. We also propose nomenclature for the cell populations and derivatives discussed herein. This review is a joint effort from the COST SPRINT Action (CA17116), which broadly aims at approaching consensus for different aspects of PnD research, such as providing inputs for future standards for the processing and in vitro characterization and clinical application of PnD.

9.
Exp Dermatol ; 28(11): 1298-1308, 2019 11.
Article in English | MEDLINE | ID: mdl-31487753

ABSTRACT

BACKGROUND: Atopic dermatitis (AD) is a complex skin disease involving causative effects from both intrinsic and extrinsic sources. Murine models of the disease often fall short in one of these components and, as a result, do not fully encapsulate these disease mechanisms. OBJECTIVE: We aimed to determine whether the protease-activated receptor 2 over-expressor mouse (PAR2OE) with topical house dust mite (HDM) application is a more comprehensive and clinically representative AD model. METHODS: Following HDM extract application to PAR2OE mice and controls, AD clinical scoring, itching behaviour, skin morphology and structure, barrier function, immune cell infiltration and inflammatory markers were assessed. Skin morphology was analysed using haematoxylin and eosin staining, and barrier function was assessed by transepidermal water loss measurements. Immune infiltrate was characterised by histological and immunofluorescence staining. Finally, an assessment of AD-related gene expression was performed using quantitative RT-PCR. RESULTS: PAR2OE mice treated with HDM displays all the characteristic clinical symptoms including erythema, dryness and oedema, skin morphology, itch and inflammation typically seen in patients with AD. There is a significant influx of mast cells (P < .01) and eosinophils (P < .0001) into the dermis of these mice. Furthermore, the PAR2OE + HDM mice exhibit similar expression patterns of key differentially expressed genes as seen in human AD. CONCLUSION: The PAR2OE + HDM mouse presents with a classic AD pathophysiology and is a valuable model in terms of reproducibility and overall disease representation to study the condition and potential therapeutic approaches.


Subject(s)
Dermatitis, Atopic/etiology , Disease Models, Animal , Pyroglyphidae/immunology , Receptor, PAR-2/physiology , Animals , Dermatitis, Atopic/pathology , Skin/immunology , Skin/pathology
10.
J Biomed Mater Res A ; 102(4): 958-66, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23650018

ABSTRACT

A cardiac patch is a construct devised in regenerative medicine to replace necrotic heart tissue after myocardial infarctions. The cardiac patch consists of a scaffold seeded with stem cells. To identify the best scaffold for cardiac patch construction we compared polyurethane, Collagen Cell Carriers, ePTFE, and ePTFE SSP1-RGD regarding their receptiveness to seeding with mesenchymal stem cells isolated from umbilical cord tissue. Seeding was tested at an array of cell seeding densities. The bioartificial patches were cultured for up to 35 days and evaluated by scanning electron microscopy, microscopy of histological stains, fluorescence microscopy, and mitochondrial assays. Polyurethane was the only biomaterial which resulted in an organized multilayer (seeding density: 0.750 × 10(6) cells/cm(2)). Cultured over 35 days at this seeding density the mitochondrial activity of the cells on polyurethane patches continually increased. There was no decrease in the E Modulus of polyurethane once seeded with cells. Seeding of CCC could only be realized at a low seeding density and both ePTFE and ePTFE SSP1-RGD were found to be unreceptive to seeding. Of the tested scaffolds polyurethane thus crystallized as the most appropriate for seeding with mesenchymal stem cells in the framework of myocardial tissue engineering.


Subject(s)
Materials Testing , Myocardium/metabolism , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Biomechanical Phenomena/drug effects , Collagen/pharmacology , Elastic Modulus/drug effects , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Microscopy, Electron, Scanning , Mitochondria/drug effects , Mitochondria/metabolism , Polytetrafluoroethylene/pharmacology , Polyurethanes/pharmacology , Propidium/metabolism , Staining and Labeling
11.
Int J Mol Cell Med ; 1(3): 119-32, 2012.
Article in English | MEDLINE | ID: mdl-24551768

ABSTRACT

Heart failure is one of the leading causes of death worldwide. End stage disease often requires heart transplantation, which is hampered by donor organ shortage. Tissue engineering represents a promising alternative approach for cardiac repair. For the generation of artificial heart muscle tissue several cell types, scaffold materials and bioreactor designs are under investigation. In this review, the use of mesenchymal stem cells derived from human umbilical cord tissue (UCMSC) for cardiac tissue engineering will be discussed.

12.
Int Immunol ; 23(8): 485-92, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21652515

ABSTRACT

Numerous data indicate that CD4+CD25+FoxP3+ regulatory T cells (Treg cells) can attenuate alloresponses of conventional T lymphocytes against professional antigen-presenting cells and thus qualify for clinical use in various transplant settings. However, it is unknown whether Treg cells also influence T cell-endothelial cell interactions. CD8+ PBMC (CD8+ PBMC, CTL) from healthy human donors were stimulated for 7 days with an allogeneic microvascular endothelial cell line (CDC/EU. HMEC-1, an immortalized human microvascular endothelial cell line, further referred to as HMEC) and additional endothelial cell types and analysed for their lytic activity against these target cells in the presence or absence of Treg cells. Addition of Treg cells (1:1:1) to the CTL/HMEC co-cultures in the efferent immune phase (day -1 prior to the assay) led to an increased cytotoxicity against HMEC. In contrast, Treg cells alone did not lyse HMEC. Treg cell-mediated enhancement of CTL activity was endothelial cell specific since lysis of HLA-matched Epstein-Barr virus-transformed B lymphoblastoid cells (B-LCL) was not influenced by the addition of Treg cells. Further analysis of CD28-positive and CD28-negative CTL sub-populations revealed that only the CD28-negative CTL showed an increased activity against HMEC after Treg cell co-culture. Although there is no doubt about the potential therapeutic efficacy of Treg cells to ameliorate outcome of allogeneic transplants, the endothelium might require additional protective interventions to prevent endothelial cell type-specific alloreactivity.


Subject(s)
Endothelium, Vascular/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Antigen-Presenting Cells/immunology , CD28 Antigens/immunology , CD28 Antigens/metabolism , Cell Communication/immunology , Cell Line , Cytotoxicity, Immunologic/immunology , Endothelial Cells/immunology , Endothelium, Vascular/metabolism , Forkhead Transcription Factors/metabolism , HLA Antigens/genetics , HLA Antigens/immunology , Humans , Immunity, Cellular/genetics , Immunity, Cellular/immunology , Immunophenotyping , T-Lymphocytes, Regulatory/metabolism
13.
J Funct Biomater ; 2(3): 107-18, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-24956300

ABSTRACT

After myocardial infarction, the implantation of stem cell seeded scaffolds on the ischemic zone represents a promising strategy for restoration of heart function. However, mechanical integrity and functionality of tissue engineered constructs need to be determined prior to implantation. Therefore, in this study a novel pulsatile bioreactor mimicking the myocardial contraction was developed to analyze the behavior of mesenchymal stem cells derived from umbilical cord tissue (UCMSC) colonized on titanium-coated polytetrafluorethylene scaffolds to friction stress. The design of the bioreactor enables a simple handling and defined mechanical forces on three seeded scaffolds at physiological conditions. The compact system made of acrylic glass, Teflon®, silicone, and stainless steel allows the comparison of different media, cells and scaffolds. The bioreactor can be gas sterilized and actuated in a standard incubator. Macroscopic observations and pressure-measurements showed a uniformly sinusoidal pulsation, indicating that the bioreactor performed well. Preliminary experiments to determine the adherence rate and morphology of UCMSC after mechanical loadings showed an almost confluent cellular coating without damage on the cell surface. In summary, the bioreactor is an adequate tool for the mechanical stress of seeded scaffolds and offers dynamic stimuli for pre-conditioning of cardiac tissue engineered constructs in vitro.

14.
PLoS One ; 5(12): e15339, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-21179573

ABSTRACT

BACKGROUND: Apart from the platelet/endothelial cell adhesion molecule 1 (PECAM-1, CD31), endoglin (CD105) and a positive factor VIII-related antigen staining, human primary and immortalized macro- and microvascular endothelial cells (ECs) differ in their cell surface expression of activating and inhibitory ligands for natural killer (NK) cells. Here we comparatively study the effects of irradiation on the phenotype of ECs and their interaction with resting and activated NK cells. METHODOLOGY/PRINCIPAL FINDINGS: Primary macrovascular human umbilical vein endothelial cells (HUVECs) only express UL16 binding protein 2 (ULBP2) and the major histocompatibility complex (MHC) class I chain-related protein MIC-A (MIC-A) as activating signals for NK cells, whereas the corresponding immortalized EA.hy926 EC cell line additionally present ULBP3, membrane heat shock protein 70 (Hsp70), intercellular adhesion molecule ICAM-1 (CD54) and HLA-E. Apart from MIC-B, the immortalized human microvascular endothelial cell line HMEC, resembles the phenotype of EA.hy926. Surprisingly, primary HUVECs are more sensitive to Hsp70 peptide (TKD) plus IL-2 (TKD/IL-2)-activated NK cells than their immortalized EC counterpatrs. This finding is most likely due to the absence of the inhibitory ligand HLA-E, since the activating ligands are shared among the ECs. The co-culture of HUVECs with activated NK cells induces ICAM-1 (CD54) and HLA-E expression on the former which drops to the initial low levels (below 5%) when NK cells are removed. Sublethal irradiation of HUVECs induces similar but less pronounced effects on HUVECs. Along with these findings, irradiation also induces HLA-E expression on macrovascular ECs and this correlates with an increased resistance to killing by activated NK cells. Irradiation had no effect on HLA-E expression on microvascular ECs and the sensitivity of these cells to NK cells remained unaffected. CONCLUSION/SIGNIFICANCE: These data emphasize that an irradiation-induced, transient up-regulation of HLA-E on macrovascular ECs might confer protection against NK cell-mediated vascular injury.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/radiation effects , Gene Expression Regulation , HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/radiation effects , Up-Regulation , Antigens, CD/biosynthesis , Cells, Cultured , Endoglin , Humans , Microcirculation , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Phenotype , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Receptors, Cell Surface/biosynthesis , von Willebrand Factor/metabolism , HLA-E Antigens
15.
J Immunol Methods ; 363(1): 80-9, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21035451

ABSTRACT

Mesenchymal stem cells (MSCs) are fibroblast-like multipotent stem cells that can differentiate into cell types of mesenchymal origin. Because of their immune properties and differentiation, potential MSCs are discussed for the use in tissue regeneration and tolerance induction in transplant medicine. This cell type can easily be obtained from the umbilical cord tissue (UCMSC) without medical intervention. Standard culture conditions include fetal bovine serum (FBS) which may not be approved for clinical settings. Here, we analyzed the phenotypic and functional properties of UCMSC under xeno-free (XF, containing GMP-certified human serum) and serum-free (SF) culture conditions in comparison with standard UCMSC cultures. Phenotypically, UCMSC showed no differences in the expression of mesenchymal markers or differentiation capacity. Functionally, XF and SF-cultured UCMSC have comparable adipogenic, osteogenic, and endothelial differentiation potential. Interestingly, the UCMSC-mediated suppression of T cell proliferation in an allogeneic mixed lymphocyte reaction (MLR) is more effective in XF and SF media than in standard FBS-containing cultures. Regarding the mechanism of action of MLR suppression, transwell experiments revealed that in neither UCMSC culture a direct cell-cell contact is necessary for inhibiting T cell proliferation, and that the major effector molecule is prostaglandin E2 (PGE2). Taken together, GMP-compliant growth media qualify for long-term cultures of UCMSC which is important for a future clinical study design in regenerative and transplant medicine.


Subject(s)
Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Umbilical Cord/cytology , Adipogenesis/drug effects , Animals , Cattle , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Culture Media, Serum-Free/pharmacology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Osteogenesis/drug effects , Serum , Umbilical Cord/metabolism
16.
Biomed Mater ; 5(6): 065004, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20924136

ABSTRACT

Umbilical cord tissue comprises an attractive new source for mesenchymal stem cells. Umbilical cord tissue-derived mesenchymal stem cells (UCMSC) exhibit self-renewal, multipotency and immunological naivity, and they can be obtained without medical intervention. The transfer of UCMSC to the ischemic region of the heart may have a favorable impact on tissue regeneration. Benefit from typical cell delivery by injection to the infarcted area is often limited due to poor cell retention and survival. Another route of administration is to use populated scaffolds implanted into the infarcted zone. In this paper, the seeding efficiency of UCMSC on uncoated and titanium-coated expanded polytetrafluoroethylene (ePTFE) scaffolds with different surface structures was determined. Dualmesh (DM) offers a corduroy-like surface in contrast to the comparatively planar surface of cardiovascular patch (CVP). The investigation of adherence, viability and proliferation of UCMSC demonstrates that titanium-coated scaffolds are superior to uncoated scaffolds, independent of the surface structure. Microscopic images reveal spherical UCMSC seeded on uncoated scaffolds. In contrast, UCMSC on titanium-coated scaffolds display their characteristic spindle-shaped morphology and a homogeneous coverage of CVP. In summary, titanium coating of clinically approved CVP enhances the retention of UCMSC and thus offers a potential cell delivery system for the repair of the damaged myocardium.


Subject(s)
Coated Materials, Biocompatible/chemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Polytetrafluoroethylene/chemistry , Tissue Scaffolds , Umbilical Cord/cytology , Cell Adhesion , Cell Proliferation , Cell Size , Cell Survival , Cells, Cultured , Equipment Design , Equipment Failure Analysis , Humans , Materials Testing , Umbilical Cord/physiology
17.
Biol Blood Marrow Transplant ; 15(5): 537-46, 2009 May.
Article in English | MEDLINE | ID: mdl-19361745

ABSTRACT

This study evaluated the relative impact of the intensity of the conditioning regimen and the alloreactivity in the endothelial dysfunction occurring after allogeneic hematopoietic stem cell transplantation (allo-HSCT). It involved a comparative analysis of the effect of incubating human umbilical vein endothelial cells (ECs) with serum samples from patients receiving autologous HSCT (auto-HSCT) or unrelated donor allo-HSCT. In both groups, blood samples were collected through a central line before conditioning (Pre), before transplantation (day 0), and at days 7, 14, and 21 after transplantation. Changes in the expression of EC receptors and adhesion proteins, adhesion of leukocytes and platelets under flow, and signaling pathways were analyzed. Endothelial activation and damage were observed in both groups, but with differing patterns. All markers of endothelial dysfunction demonstrated a progressive increase from day Pre to day 14 in the auto-HSCT group and exhibited 2 peaks of maximal expression (at days 0 and 21) in the allo-HSCT group. Both treatments induced a proinflammatory state (ie, expression of adhesion receptors, leukocyte adhesion, and p38 MAPK activation) and cell proliferation (ie, morphology and activation of ErK42/44). Prothrombotic changes (ie, von Willebrand factor expression and platelet adhesion) predominated after allo-HSCT, and a proapoptotic tendency (ie, activation of SAPK/JNK) was seen only in this group. These findings indicate that endothelial activation and damage after HSCT also occur in the autologous setting and affect macrovascular ECs. After the initial damage induced by the conditioning regimen, other factors, such as granulocyte colony-stimulating factor (G-CSF) toxicity, engraftment, and alloreactivity, may contribute to the endothelial damage seen during HSCT. Further studies are needed to explore the association between this endothelial damage and the vascular complications associated with HSCT.


Subject(s)
Endothelial Cells/pathology , Endothelium, Vascular/pathology , Hematopoietic Stem Cell Transplantation/adverse effects , Adult , Aged , Biomarkers/blood , Blood Cells/pathology , Cell Adhesion , Cells, Cultured , Endothelial Cells/metabolism , Female , Humans , Inflammation , Male , Middle Aged , Signal Transduction , Thrombophilia , Transplantation Conditioning/adverse effects , Transplantation Conditioning/methods , Transplantation, Homologous
18.
Biol Blood Marrow Transplant ; 14(10): 1180-1189, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18804049

ABSTRACT

Aside from major and minor histocompatibility antigens, genetic polymorphisms of various donor and host genes have been found to be risk factors for graft-versus-host disease and transplantation-related mortality (TRM). The heme oxygenase I (HO-I) protein has been implicated in regulating inflammatory response and has been described as a "protective gene" in solid organ transplantation. In humans, the promoter region displays length polymorphism due to a variable number of GT repeats. Individuals exhibiting 29 or fewer GT repeats express higher levels of HO-I on cellular stress compared with individuals with 30 or more GT repeats. We retrospectively analyzed length polymorphisms of 92 donor-host pairs undergoing allogeneic stem cell transplantation. Our findings demonstrate that mainly donor polymorphism leading to high expression of HO-1 (<30 GT repeats) on stress signals is associated with reduced overall survival, and that TRM is significantly increased in this group. This reduction in survival was most prominent when unrelated donors were used. Polymorphisms of the recipient HO-1 genes did not influence posttransplantation outcomes. We conclude that HO-1 polymorphism represents a new genetic risk factor for TRM and overall survival.


Subject(s)
Hematopoietic Stem Cell Transplantation/mortality , Heme Oxygenase-1/genetics , Polymorphism, Genetic , Promoter Regions, Genetic/genetics , Dinucleotide Repeats , Humans , Retrospective Studies , Survival Rate , Tissue Donors , Transplantation, Homologous
19.
Cancer Biol Ther ; 6(5): 686-90, 2007 May.
Article in English | MEDLINE | ID: mdl-17495522

ABSTRACT

Defibrotide (DF) is a polydisperse mixture of 90% single-stranded oligonucleotides with anti-thrombotic and anti-apoptotic functions. DF is used in the treatment of endothelial complications in the course of allogeneic stem cell transplantation. Recent preclinical evidence suggests that DF might also have anti-neoplastic properties. In the present study we hypothesized that DF might inhibit tumors via an anti-angiogenic effect. The anti-angiogenic potential of DF was tested in vitro using human microvascular endothelial cells forming vessel structures across a layer of dermal fibroblasts. Our results show that pharmacologic DF concentrations (100 mug/ml) significantly reduced vessel formation in this assay. Similarly, DF blocked sprouting from cultured rat aortic rings. In vivo, angiogenesis in a human gastric tumor (TMK1) implanted in dorsal skin-fold chambers (in nude mice) was inhibited by i.v. application of 450 mg/kg DF. Notably, due to its short half-life, DF was most effective when given on a daily basis. Although the precise mechanism of DF remains to be elucidated, initial Western blots show that DF reduces phosphorylation-activation of p70S6 kinase, which is a key target in the PI3K/Akt/mTOR signaling pathway linked to endothelial cell and pericyte proliferation and activation. However, in vitro data suggest that DF acts independently of vascular endothelial growth factor. Taken together, our data suggest that while DF is known for its endothelium-protecting function in SCT, it also inhibits formation of new blood vessels, and thus should be considered for further testing as an adjuvant anti-cancer agent, either alone, or in combination with other drugs.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelium, Vascular/drug effects , Platelet Aggregation Inhibitors/pharmacology , Polydeoxyribonucleotides/pharmacology , Stomach Neoplasms/blood supply , Stomach Neoplasms/drug therapy , Adenocarcinoma/blood supply , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Animals , Aorta/physiology , Aptamers, Nucleotide/pharmacology , Biological Assay , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Collagen/metabolism , Drug Combinations , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Laminin/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/physiology , Phosphorylation/drug effects , Proteoglycans/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/metabolism , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/metabolism , Vascular Endothelial Growth Factor A/metabolism
20.
Perfusion ; 21(3): 133-7, 2006 May.
Article in English | MEDLINE | ID: mdl-16817285

ABSTRACT

Endothelial activation is considered a key process in the development of a whole body inflammatory response secondary to cardiopulmonary bypass (CPB). Increased levels of a multitude of soluble mediators have been described as being released during and after cardiac surgery. Circulating endothelial cells have recently been established as a novel marker of endothelial damage in a variety of vascular disorders. Blood samples from 20 patients undergoing elective coronary artery bypass surgery were obtained preoperatively and 1, 6, 12, 24, and 48 h after termination of CPB. Control samples were obtained from ten healthy volunteers. Circulating endothelial cells (CEC) were isolated with immunomagnetic anti-CD146-coated Dynabeads, and counted in a Nageotte chamber. Low numbers of CEC were observed in healthy control volunteers (12 +/- 6 cells/mL; median: 9 cells/mL). CEC numbers were already significantly elevated in all patients before CPB, and there was a further significant increase after weaning from CPB (maximum increase at 6 h after CPB: 73 +/- 30 cells/mL; range: 30-153 cells/mL, p < 0.001). The number of CEC provides further and direct evidence that CPB is associated with a pronounced endothelial injury and/or damage. CEC appear to be most useful markers for vascular endothelial activation because they are specific, stable, and circulating components of injured vessel wall.


Subject(s)
Angina Pectoris/blood , Cardiopulmonary Bypass , Endothelial Cells , Endothelium, Vascular/injuries , Aged , Cardiopulmonary Bypass/adverse effects , Cell Separation , Elective Surgical Procedures/adverse effects , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Female , Humans , Intraoperative Period , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...