Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
2.
Cells ; 8(8)2019 08 16.
Article in English | MEDLINE | ID: mdl-31426355

ABSTRACT

Rheumatoid arthritis (RA) is an immune-mediated inflammatory disease, and Krüppel-like factor 2 (KLF2) regulates immune cell activation and function. Herein, we show that in our experiments 50% global deficiency of KLF2 significantly elevated arthritic inflammation and pathogenesis, osteoclastic differentiation, matrix metalloproteinases (MMPs), and inflammatory cytokines in K/BxN serum-induced mice. The severities of RA pathogenesis, as well as the causative and resultant cellular and molecular factors, were further confirmed in monocyte-specific KLF2 deficient mice. In addition, induction of RA resulted in a decreased level of KLF2 in monocytes isolated from both mice and humans along with higher migration of activated monocytes to the RA sites in humans. Mechanistically, overexpression of KLF2 decreased the level of MMP9; conversely, knockdown of KLF2 increased MMP9 in monocytes along with enrichment of active histone marks and histone acetyltransferases on the MMP9 promoter region. These findings define the critical regulatory role of myeloid KLF2 in RA pathogenesis.


Subject(s)
Arthritis, Rheumatoid/immunology , Inflammation/immunology , Kruppel-Like Transcription Factors/physiology , Matrix Metalloproteinase 9/metabolism , Monocytes/immunology , Animals , Arthritis, Experimental/immunology , Cell Differentiation , Cells, Cultured , Cytokines/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoclasts/metabolism
3.
Sci Rep ; 9(1): 8415, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31182750

ABSTRACT

Despite advances in diabetic wound care, the significant number of amputations that occur every year demands more effective therapeutics. Herein, we offer an aminated polyethersulfone nanofiber-expanded human umbilical cord blood-derived CD34+ cells (henceforth CD34+ cells) effective therapy, tested in cutaneous wounds developed in streptozotocin-induced diabetic NOD/SCID mice. We show that systemic administration of CD34+ cells homed to the wound site and significantly accelerated wound closure. Wound closure was associated with improved re-epithelialization and increased neovascularization; and with decreased sustained pro-inflammatory activity of NF-κB and its downstream effector molecules TNF-α, IL-1ß, and IL-6 at the wound bed. This finding was further supported by the observation of a decreased number of myeloperoxidase positive neutrophils, and concomitantly increased levels of IL-10. In addition, improved granulation tissue formation was observed along with higher collagen deposition and myofibroblasts and decreased expressions of MMP-1. Mechanistically, CD34+ cells reduced the level of MMP-1 expression by inhibiting recruitment of NF-κB to the MMP-1 promoter site in dermal fibroblasts. In summary, we provide evidence of a novel nanofiber-expanded CD34+ stem cell therapeutic development for treating diabetic wounds by defining their cellular and molecular mechanisms.


Subject(s)
Antigens, CD34/metabolism , Diabetes Mellitus, Experimental/pathology , Nanofibers/chemistry , Skin/pathology , Wound Healing , Animals , Cell Line , Cell Proliferation/drug effects , Collagen/metabolism , Dermis/pathology , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fetal Blood/cytology , Fibroblasts/drug effects , Granulation Tissue/pathology , Humans , Inflammation/pathology , Matrix Metalloproteinases/metabolism , Mice, Inbred NOD , Mice, SCID , NF-kappa B/metabolism , Neovascularization, Physiologic/drug effects , Neutrophil Infiltration/drug effects , Streptozocin , Tumor Necrosis Factor-alpha/pharmacology , Wound Healing/drug effects
4.
J Cell Mol Med ; 23(2): 1386-1395, 2019 02.
Article in English | MEDLINE | ID: mdl-30506878

ABSTRACT

Krüppel-like factor 2 (KLF2) critically regulates activation and function of monocyte, which plays important pathogenic role in progressive joint destruction in rheumatoid arthritis (RA). It is yet to be established the molecular basis of KLF2-mediated regulation of monocytes in RA pathogenesis. Herein, we show that a class of compound, HDAC inhibitors (HDACi) induced KLF2 expression in monocytes both in vitro and in vivo. KLF2 level was also elevated in tissues, such as bone marrow, spleen and thymus in mice after infusion of HDACi. Importantly, HDACi significantly reduced osteoclastic differentiation of monocytes with the up-regulation of KLF2 and concomitant down-regulation of matrixmetalloproteinases both in the expression level as well as in the protein level. In addition, HDACi reduced K/BxN serum-induced arthritic inflammation and joint destruction in mice in a dose-dependent manner. Finally, co-immunoprecipitation and overexpression studies confirmed that KLF2 directly interacts with HDAC4 molecule in cells. These findings provide mechanistic evidence of KLF2-mediated regulation of K/BxN serum-induced arthritic inflammation.


Subject(s)
Arthritis, Experimental/prevention & control , Arthritis, Rheumatoid/prevention & control , Histone Deacetylase Inhibitors/pharmacology , Kruppel-Like Transcription Factors/metabolism , Animals , Arthritis, Experimental/etiology , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cell Differentiation , Female , Histone Deacetylases/chemistry , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/metabolism , RAW 264.7 Cells
5.
Angiogenesis ; 20(1): 97-107, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27853962

ABSTRACT

Increased circulating catecholamines have been linked with cardiovascular anomalies as well as with peripheral vascular diseases. Although the roles of epinephrine and norepinephrine have received considerable attention, the role of the other catecholamine, dopamine, has been less studied. Since dopamine is a potent endogenous inhibitor of angiogenesis and as angiogenesis is essential for ischemic healing, we therefore studied the role played by dopamine during ischemic healing using dopamine D2 receptor knockout (KOD2) mice. Although concentration of dopamine and its rate-limiting enzyme, tyrosine hydroxylase, was considerably high in the muscle tissues of wild-type and KOD2 mice with unilateral hind limb ischemia (HLI), recovery was significantly faster in the KOD2 mice compared to the wild-type controls, thereby indicating that peripheral dopamine might have a role in this healing process. In addition, we observed significant differences in post-ischemic angiogenesis between these two groups. Our study further revealed that elevated dopamine independently suppressed activation of local tissue-based renin-angiotensin system (RAS), a critical growth factor system stimulating angiogenesis in ischemia. Angiotensin II (ATII) and its receptor, angiotensin receptor type 1 (AT1R), are the key players in RAS-mediated angiogenesis. Dopamine acting through its D2 receptors in endothelial cells inhibited ATII-mediated angiogenesis by suppressing the expression of AT1R in these cells. This study thus for the first time demonstrates the role played by dopamine in prolonging post-ischemic recovery. Therefore, pharmacological intervention inhibiting the action of dopamine holds promise as future therapeutic strategy for the treatment of HLI and other peripheral arterial diseases.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Dopamine/pharmacology , Endothelial Cells/metabolism , Ischemia/metabolism , Ischemia/pathology , Neovascularization, Physiologic/drug effects , Receptor, Angiotensin, Type 1/metabolism , Wound Healing/drug effects , Angiotensin II/pharmacology , Animals , Endothelial Cells/drug effects , Extremities/blood supply , Mice, Inbred C57BL , Nitric Oxide/biosynthesis , Perfusion , Receptors, Dopamine D2/metabolism , Renin-Angiotensin System/drug effects , Up-Regulation/drug effects
6.
Catheter Cardiovasc Interv ; 85(2): 207-15, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-24905795

ABSTRACT

OBJECTIVES: To report 1- and 2-year clinical outcomes of patients receiving platinum chromium everolimus-eluting stents (PtCr-EES) in the prospective, single-arm PLATINUM small vessel (SV) and long lesion (LL) studies. BACKGROUND: Small vessel diameter and long lesion length are independently associated with increased risk of adverse cardiac events after drug-eluting stent implantation. METHODS: The PLATINUM SV study enrolled 94 patients with coronary artery lesions in vessels ≥2.25 mm to <2.50 mm in diameter and ≤28 mm in length. The PLATINUM LL study enrolled 102 patients with lesions >24 to ≤34 mm long in vessels ≥2.50 to ≤4.25 mm in diameter. The primary endpoint for both studies was target lesion failure (TLF) at 1 year compared to a prespecified performance goal based on outcomes with the TAXUS Express paclitaxel-eluting stent in small vessels and long lesions. RESULTS: One-year TLF rates with the PtCr-EES were significantly (P < 0.001) lower than the predetermined performance goals: 2.4% versus 21.1% in the SV cohort and 3.2% versus 19.4% in the LL cohort. Cumulative rates of TLF to 2 years were 4.7% in the SV cohort and 8.8% in the LL cohort. No myocardial infarction or ARC definite/probable stent thromboses occurred in either cohort through 2-year follow-up. CONCLUSIONS: The clinical efficacy and safety outcomes observed in these small vessel and long lesion cohorts support the use of the PtCr-EES in the treatment of small diameter vessels and long lesions.


Subject(s)
Cardiovascular Agents/administration & dosage , Chromium , Coronary Artery Disease/therapy , Drug-Eluting Stents , Percutaneous Coronary Intervention/instrumentation , Platinum , Sirolimus/analogs & derivatives , Aged , Coronary Artery Disease/diagnosis , Europe , Everolimus , Female , Humans , Japan , Male , Middle Aged , New Zealand , Percutaneous Coronary Intervention/adverse effects , Prospective Studies , Prosthesis Design , Sirolimus/administration & dosage , Time Factors , Treatment Outcome , United States
7.
Methods Mol Biol ; 1213: 209-14, 2014.
Article in English | MEDLINE | ID: mdl-25173385

ABSTRACT

To evaluate therapeutic efficacy and to investigate involved molecular mechanisms of cell-based therapy in osteoporosis, the generation of a clinically relevant model is critically important. Herein, we describe detailed methods in generation of an immune-deficient osteoporotic murine model, and application of human umbilical cord blood-derived stem cells to assess their therapeutic efficacy.


Subject(s)
Cell- and Tissue-Based Therapy , Osteoporosis/therapy , Stem Cell Transplantation , Animals , Antigens, CD34/metabolism , Disease Models, Animal , Female , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Immunocompromised Host , Mice , Osteoporosis/etiology
8.
Biomaterials ; 35(30): 8566-75, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25002260

ABSTRACT

Despite recent advances in cardiovascular medicine, ischemic diseases remain a major cause of morbidity and mortality. Although stem cell-based therapies for the treatment of ischemic diseases show great promise, limited availability of biologically functional stem cells mired the application of stem cell-based therapies. Previously, we reported a PES-nanofiber based ex vivo stem cell expansion technology, which supports expansion of human umbilical cord blood (UCB)-derived CD133(+)/CD34(+) progenitor cells ∼225 fold. Herein, we show that using similar technology and subsequent re-expansion methods, we can achieve ∼5 million-fold yields within 24 days of the initial seeding. Interestingly, stem cell phenotype was preserved during the course of the multiple expansions. The high level of the stem cell homing receptor, CXCR4 was expressed in the primary expansion cells, and was maintained throughout the course of re-expansions. In addition, re-expanded cells preserved their multi-potential differential capabilities in vitro, such as, endothelial and smooth muscle lineages. Moreover, biological functionality of the re-expanded cells was preserved and was confirmed by a murine hind limb ischemia model for revascularization. These cells could also be genetically modified for enhanced vasculogenesis. Immunohistochemical evidences support enhanced expression of angiogenic factors responsible for this enhanced neovascularization. These data further confirms that nanofiber-based ex-vivo expansion technology can generate sufficient numbers of biologically functional stem cells for potential clinical applications.


Subject(s)
Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/physiology , Nanofibers/chemistry , Neovascularization, Physiologic/drug effects , Polymers/pharmacology , Stem Cells/cytology , Sulfones/pharmacology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Immunohistochemistry , Ischemia/pathology , Ischemia/physiopathology , Mice, SCID , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Nanofibers/ultrastructure , Phenotype , Stem Cells/drug effects , Stem Cells/ultrastructure
9.
Biochim Biophys Acta ; 1842(7): 1071-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24726882

ABSTRACT

Many ovarian cancer cells express stress-related molecule MICA/B on their surface that is recognized by Vγ2Vδ2 T cells through their NKG2D receptor, which is transmitted to downstream stress-signaling pathway. However, it is yet to be established how Vγ2Vδ2 T cell-mediated recognition of MICA/B signal is transmitted to downstream stress-related molecules. Identifying targeted molecules would be critical to develop a better therapy for ovarian cancer cells. It is well established that ATM/ATR signal transduction pathways, which is modulated by DNA damage, replication stress, and oxidative stress play central role in stress signaling pathway regulating cell cycle checkpoint and apoptosis. We investigated whether ATM/ATR and its down stream molecules affect Vγ2Vδ2 T cell-mediated cytotoxicity. Herein, we show that ATM/ATR pathway is modulated in ovarian cancer cells in the presence of Vγ2Vδ2 T cells. Furthermore, downregulation of ATM pathway resulted downregulation of MICA, and reduced Vγ2Vδ2 T cell-mediated cytotoxicity. Alternately, stimulating ATM pathway enhanced expression of MICA, and sensitized ovarian cancer cells for cytotoxic lysis by Vγ2Vδ2 T cells. We further show that combining currently approved chemotherapeutic drugs, which induced ATM signal transduction, along with Vγ2Vδ2 T cells enhanced cytotoxicity of resistant ovarian cancer cells. These findings indicate that ATM/ATR pathway plays an important role in tumor recognition, and drugs promoting ATM signaling pathway might be considered as a combination therapy together with Vγ2Vδ2 T cells for effectively treating resistant ovarian cancer cells.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/immunology , Ovarian Neoplasms/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Down-Regulation , Female , Histocompatibility Antigens Class I/metabolism , Humans , Ovarian Neoplasms/drug therapy , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction
10.
Acute Card Care ; 16(2): 74-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24654687

ABSTRACT

BACKGROUND: Intra-aortic balloon pump (IABP) use may be associated with complications; however, in certain patients with ST-elevation myocardial infarction (STEMI) with hemodynamic instability refractory to medical management its use may become necessary. METHODS: 36 STEMI patients with IABP placement for hemodynamic instability after percutaneous coronary intervention were studied. IABP duration ranged from one to seven days (median two days). Based on median time, patients were divided into two groups: IABP duration ≤ 2 days (n = 27) or > 2 days (n = 9). Vascular complications and incidence of bleeding were compared. RESULTS: Mean IABP duration was 1.4 ± 0.5 and 4.1 ± 1.3 days in ≤ 2 day and > 2 day groups, respectively (P < 0.01). Glycoprotein IIb/IIIa inhibitor and anti-coagulation use was not significantly different between groups. Mean duration of anti-coagulation was 1.9 ± 1.2 and 4.5 ± 1.3 days in ≤ 2 day and > 2 day groups, respectively (P < 0.05). Complications (vascular, access site bleeding, gastrointestinal bleeding) were significantly greater in > 2 day group (66%) compared to ≤ 2 day group (18%; P < 0.05). CONCLUSIONS: When an IABP was used for more than two days complications significantly increased. The clinical implications of the study will be strengthened if the findings are confirmed in a prospective study with a larger number of patients.


Subject(s)
Hemorrhage/etiology , Intra-Aortic Balloon Pumping/adverse effects , Myocardial Infarction/surgery , Percutaneous Coronary Intervention , Vascular Diseases/etiology , Aged , Electrocardiography , Female , Hemodynamics , Humans , Male , Middle Aged , Myocardial Infarction/complications , Myocardial Infarction/physiopathology , Risk Factors , Time Factors , Treatment Outcome
11.
J Cell Mol Med ; 18(4): 685-97, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24455991

ABSTRACT

Nanofiber-expanded human umbilical cord blood-derived CD34(+) cell therapy has been shown to have potential applications for peripheral and myocardial ischaemic diseases. However, the efficacies of expanded CD34(+) cell therapy for treating cutaneous wounds and its mechanisms of action have yet to be established. Using an excisional wound model in non-obese diabetic/severe combined immune deficient mice, we show herein that CD34(+) cells accelerate the wound-healing process by enhancing collagen synthesis, and increasing fibroblast cell migration within the wound bed. Concomitantly, reduced levels of matrix metalloproteinase (MMPs) such as MMP1, MMP3, MMP9 and MMP13 were detected in the wound beds of animals treated with CD34(+) cells compared with vehicle-treated controls. CD34(+) cells were found to mediate enhanced migration and proliferation of dermal fibroblast cells in vitro. Moreover, CD34(+) cells secrete collagen in a serum-deprived environment. In mechanistic studies, co-culture of CD34(+) cells with primary skin fibroblasts increased the expression of collagen1A1, a component of type 1 collagen, and decreased the expression of MMP1 in fibroblast cells in the presence of a proteasome inhibitor. Finally, CD34(+) cell-mediated functions were transcriptionally regulated by the c-Jun N-terminal kinases pathway. Collectively, these data provide evidence of therapeutic efficacy and a novel mechanism of nanofiber-expanded CD34(+) cell-mediated accelerated wound healing.


Subject(s)
Antigens, CD34/metabolism , Cell- and Tissue-Based Therapy , Fetal Blood/transplantation , Transplantation, Heterologous , Animals , Fetal Blood/cytology , Humans , Mice , Nanofibers/administration & dosage , Nanofibers/chemistry , Skin/pathology , Wound Closure Techniques , Wound Healing
12.
Stem Cell Res ; 12(1): 275-88, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24321844

ABSTRACT

Nanofiber-expanded human umbilical cord blood-derived CD34+ cell therapy is under consideration for treating peripheral and cardiac ischemia. However, the therapeutic efficacy of nanofiber-expanded human umbilical cord blood-derived (NEHUCB) CD34+ cell therapy for wound healing and its mechanisms are yet to be established. Using an excision wound model in NOD/SCID mice, we show herein that NEHUCB-CD34+ cells home to the wound site and significantly accelerate the wound-healing process compared to vehicle-treated control. Histological analysis reveals that accelerated wound closure is associated with the re-epithelialization and increased angiogenesis. Additionally, NEHUCB-CD34+ cell-therapy decreases expression of pro-inflammatory cytokines, such as TNF-α, IL-1ß, IL-6 and NOS2A in the wound bed, and concomitantly increases expression of IL-10 compared to vehicle-treated control. These findings were recapitulated in vitro using primary dermal fibroblasts and NEHUCB-CD34+ cells. Moreover, NEHUCB-CD34+ cells attenuate NF-κB activation and nuclear translocation in dermal fibroblasts through enhanced secretion of IL-10, which is known to bind to NF-κB and suppress transcriptional activity. Collectively, these data provide novel mechanistic evidence of NEHUCB-CD34+ cell-mediated accelerated wound healing.


Subject(s)
Fetal Blood/cytology , Interleukin-10/metabolism , Skin Diseases/therapy , Stem Cells/cytology , Wound Healing , Animals , Cell- and Tissue-Based Therapy , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Humans , Interleukin-10/genetics , Interleukin-6/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Nanofibers/chemistry , Protein Binding , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Skin Diseases/metabolism , Skin Diseases/pathology , Stem Cell Transplantation , Stem Cells/chemistry , Transcription Factors/metabolism
13.
Front Biosci (Landmark Ed) ; 18(3): 970-81, 2013 06 01.
Article in English | MEDLINE | ID: mdl-23747860

ABSTRACT

Because of their ability for self-renewal and neural differentiation, stem cells are believed to be ideal for cell replacement therapy in Parkinson's disease (PD). Nanofiber-expanded human umbilical cord hematopoietic stem cells (HUHSCs) are advantageous to other stem cells as they provide a source of unlimited stem cell production for clinical application. In this study, we investigated whether 1. nanofiber-expanded HUHSCs are capable of neural differentiation in vitro, and 2. they could improve dopaminergic neuron morphology in the caudate/putamen (CPu) and substantia nigra pars compacta (SNc) of the MPTP-mouse model of PD. When cultured under neural differentiation conditions, nanofiber-expanded HUHSCs were able to undergo neural differentiation in vitro, as determined by gene and protein expression of neural markers such as MAP2, NeuN, HuC, GFAP and Oligo2. Thirty days after a single intracardioventricular injection of HUHSCs to MPTP-mice there was a significant recovery of tyrosine hydroxylase (TH) immunostaining in CPu. There was an increase in the size and staining density of TH+ cells in SNc, while their number was unchanged.


Subject(s)
Dopamine/metabolism , Hematopoietic Stem Cells/cytology , Neurons/metabolism , Parkinsonian Disorders/therapy , Animals , Base Sequence , DNA Primers , Disease Models, Animal , Mice , Polymerase Chain Reaction
14.
Int J Cancer ; 133(9): 2133-44, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23595559

ABSTRACT

Innate immune system has been known to play an important role in inhibiting the malignant transformation, tumor progression and invasion. However, the mechanistic basis remains ambiguous. Despite polyclonality of human γδ T cells, Vγ2Vδ2 T cell subset was shown to recognize and limit the growth of various tumors at various degrees. The differential recognition of the tumor cells by Vγ2Vδ2 T cells are yet to be defined. Our study reveals that γδ T cells limit in vitro growth of most breast tumor cells, such as SkBr7 (HER2+), MCF7 (ER+) and MDA-MB-231 (ER-) by inhibiting their survival and inducing apoptosis, except BrCa-MZ01 (PR+) cells. To investigate detail mechanisms of antineoplastic effects, we found that cell death was associated with the surface expression levels of MICA/B and ICAM1. Molecular signaling analysis demonstrated that inhibition of cell growth by γδ T cells was associated with the lower expression levels of cell survival-related molecules such as AKT, ERK and concomitant upregulation of apoptosis-related molecules, such as PARP, cleaved caspase 3 and tumor suppressor genes PTEN and P53. However, opposite molecular signaling was observed in the resistant cell line after coculture with γδ T cells. In vivo, antineoplastic effects of γδ T cells were also documented, where tumor growth was inhibited due to the downregulation of survival signals, strong induction of apoptotic molecules, disruption of microvasculature and increased infiltration of tumor associated macrophages. These findings reveal that a complex molecular signaling is involved in γδ T cell-mediated antineoplastic effects.


Subject(s)
Apoptosis , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Tumor Microenvironment/immunology , Blotting, Western , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Cell Cycle , Female , Gene Expression Regulation, Neoplastic , Humans , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Tumor Cells, Cultured
15.
Cell Biochem Biophys ; 67(2): 235-45, 2013 Nov.
Article in English | MEDLINE | ID: mdl-22038301

ABSTRACT

Vasculogenesis and angiogenesis are the major forms of blood vessel formation. Angiogenesis is the process where new vessels grow from pre-existing blood vessels, and is very important in the functional recovery of pathological conditions, such as wound healing and ischemic heart diseases. The development of better animal model and imaging technologies in past decades has greatly enriched our understanding on vasculogenesis and angiogenesis processes. Hypoxia turned out to be an important driving force for angiogenesis in various ischemic conditions. It stimulates expression of many growth factors like vascular endothelial growth factor, platelet-derived growth factor, insulin-like growth factor, and fibroblast growth factor, which play critical role in induction of angiogenesis. Other cellular components like monocytes, T cells, neutrophils, and platelets also play significant role in induction and regulation of angiogenesis. Various stem/progenitor cells also being recruited to the ischemic sites play crucial role in the angiogenesis process. Pre-clinical studies showed that stem/progenitor cells with/without combination of growth factors induce neovascularization in the ischemic tissues in various animal models. In this review, we will discuss about the fundamental factors that regulate the angiogenesis process and the use of stem cells as therapeutic regime for the treatment of ischemic diseases.


Subject(s)
Hematopoietic Stem Cells/cytology , Neovascularization, Physiologic , Animals , Cell Hypoxia , Hematopoietic Stem Cells/pathology , Humans , Immunity , Intercellular Signaling Peptides and Proteins/metabolism , Signal Transduction
16.
Acute Card Care ; 14(4): 120-4, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23215746

ABSTRACT

OBJECTIVE: Compare vascular complications and incidence of bleeding of Impella 2.5 and intra-aortic balloon pump (IABP) in high-risk percutaneous coronary interventions (PCI). BACKGROUND: Large arterial sheath size for device insertion is associated with vascular and/or bleeding complications; gastrointestinal bleeding may also occur with anti-coagulation use. METHODS: Patients with an acute coronary syndrome receiving Impella 2.5 or IABP during high-risk PCI were studied (13 Impella; 62 IABP). Vascular complications and incidence of bleeding were compared. RESULTS: Post-procedure hematocrit was similar between groups. Blood transfusion occurred in 38.4% and 32.2% of patients in the Impella and IABP groups, respectively (P = NS); 65.3%, 30.7% and 3.8% of bleeding were due to vascular access site/procedure related, gastrointestinal and genitourinary, respectively. There was no statistical significant difference in vascular complications between the Impella and IABP groups (15.3% and 6.4% of patients, respectively); mesenteric ischemia (n = 1) and aortic rupture (n = 1) were only in the IABP group. In-hospital and one-year mortality were not statistically significant between groups. CONCLUSION: Impella can be used as safely as IABP during high-risk PCI with similar vascular and bleeding complications. Importantly, approximately one third of bleeding was from the gastrointestinal system warranting careful prophylactic measures and monitoring.


Subject(s)
Acute Coronary Syndrome/therapy , Heart-Assist Devices/adverse effects , Hemorrhage/etiology , Intra-Aortic Balloon Pumping/adverse effects , Acute Coronary Syndrome/mortality , Aged , Blood Transfusion/statistics & numerical data , Coronary Stenosis , Female , Gastrointestinal Hemorrhage/etiology , Hematocrit , Hemodynamics , Hospital Mortality , Humans , Male , Middle Aged , Retrospective Studies
17.
PLoS One ; 7(6): e39365, 2012.
Article in English | MEDLINE | ID: mdl-22724005

ABSTRACT

BACKGROUND: Osteoporosis is a bone disorder associated with loss of bone mineral density and micro architecture. A balance of osteoblasts and osteoclasts activities maintains bone homeostasis. Increased bone loss due to increased osteoclast and decreased osteoblast activities is considered as an underlying cause of osteoporosis. METHODS AND FINDINGS: The cures for osteoporosis are limited, consequently the potential of CD34+ cell therapies is currently being considered. We developed a nanofiber-based expansion technology to obtain adequate numbers of CD34(+) cells isolated from human umbilical cord blood, for therapeutic applications. Herein, we show that CD34(+) cells could be differentiated into osteoblastic lineage, in vitro. Systemically delivered CD34(+) cells home to the bone marrow and significantly improve bone deposition, bone mineral density and bone micro-architecture in osteoporotic mice. The elevated levels of osteocalcin, IL-10, GM-CSF, and decreased levels of MCP-1 in serum parallel the improvements in bone micro-architecture. Furthermore, CD34(+) cells improved osteoblast activity and concurrently impaired osteoclast differentiation, maturation and functionality. CONCLUSIONS: These findings demonstrate a novel approach utilizing nanofiber-expanded CD34(+) cells as a therapeutic application for the treatment of osteoporosis.


Subject(s)
Antigens, CD34 , Cord Blood Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Osteoblasts/cytology , Osteoclasts/cytology , Osteoporosis/therapy , Animals , Antigens, CD34/metabolism , Bone Marrow/metabolism , Bone and Bones/ultrastructure , Calcification, Physiologic , Cell Culture Techniques , Cell Differentiation , Cytokines/blood , Disease Models, Animal , Female , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Osteoblasts/metabolism , Osteocalcin/blood , Osteoclasts/metabolism , Osteogenesis , Osteoporosis/metabolism
18.
J Mol Cell Cardiol ; 52(5): 958-61, 2012 May.
Article in English | MEDLINE | ID: mdl-22269791

ABSTRACT

UNLABELLED: CD39 (ectonucleoside triphosphate diphosphohydrolase-1; ENTPD-1) rapidly hydrolyzes ATP and ADP to AMP; AMP is hydrolyzed by ecto-5'-nucleotidase (CD73) to adenosine, an anti-thrombotic and cardiovascular protective mediator. While expression of human CD39 in a murine model of myocardial ischemia/reperfusion (I/R) injury confers cardiac protection, the translational therapeutic potential of these findings requires further testing in a large animal model. To determine if transgenic expression of CD39 reduces infarct size in a swine model of myocardial ischemia/reperfusion injury, transgenic pigs expressing human CD39 (hCD39) were generated via somatic cell nuclear transfer and characterized. Expression of hC39 in cardiac tissue was confirmed by immunoblot and immunohistochemistry. Myocardial I/R injury was induced by intracoronary balloon inflation in the left anterior descending (LAD) artery for 60 min followed by 3 hours of reperfusion. The ischemic area was delineated by perfusion with 5% phthalo blue and the myocardial infarct size was determined by triphenyl tetrazolium chloride (TTC) staining. During ischemia, the rate-pressure product was significantly lower in control versus hCD39-Tg swine. Following reperfusion, compared to littermate control swine, hCD39-Tg animals displayed a significant reduction in infarct size (hCD39-Tg: 17.2 ± 4.3% vs. CONTROL: 44.7 ± 5.2%, P=0.0025). Our findings demonstrate for the first time that the findings in transgenic mouse models translate to large animal transgenic models and validate the potential to translate CD39 into the clinical arena to attenuate human myocardial ischemia/reperfusion injury.


Subject(s)
Antigens, CD/biosynthesis , Apyrase/biosynthesis , Myocardial Reperfusion Injury/metabolism , Swine/genetics , Animals , Animals, Genetically Modified , Antigens, CD/genetics , Apyrase/genetics , Blood Pressure , Coronary Vessels/pathology , Heart Rate , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Ischemia/metabolism , Ischemia/pathology , Myocardial Reperfusion Injury/pathology , Promoter Regions, Genetic , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
19.
Recent Pat Biotechnol ; 5(1): 40-53, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21517745

ABSTRACT

Maintenance of hematopoietic stem cells (HSCs) pool depends on fine balance between self-renewal and differentiation of HSCs. HSCs normally reside within the bone marrow niche of an adult mammal. The embryonic development of HSCs is a complex process that involves the migration of developing HSCs in multiple anatomical sites. Throughout the process, developing HSCs receive internal (transcriptional program) and external (HSC niche) signals, which direct them to maintain balance between self-renewal and differentiation, also to generate a pool of HSCs. In physiological condition HSCs differentiate into all mature cell types present in the blood. However, in pathological condition they may differentiate into non-hematological cells according to the need of the body. It was shown that HSCs can transdifferentiate into cell types that do not belong to the hematopoietic system suggests a complete paradigm shift of the hierarchical hematopoietic tree. This review describes the developmental origins and regulation of HSCs focusing on developmental signals that induce the adult hematopoietic stem cell program, as these informations are very critical for manipulating conditions for expansion of HSCs in ex vivo condition. This review also states clinical application and related patents using HSC.


Subject(s)
Hematopoietic Stem Cells/cytology , Animals , Cell Differentiation , Cell Proliferation , Embryonic Development , Hematopoietic Stem Cell Transplantation , Humans , Patents as Topic
20.
Front Biosci (Elite Ed) ; 3(2): 506-14, 2011 01 01.
Article in English | MEDLINE | ID: mdl-21196330

ABSTRACT

The Phase I clinical study was designed to assess the safety and feasibility of a dose escalating intracoronary infusion of autologous bone marrow (BM)-derived CD133+ stem cell therapy to the patients with chronic total occlusion (CTO) and ischemia. Nine patients were received CD133+ cells into epicardial vessels supplying collateral flow to areas of viable ischemic myocardium in the distribution of the CTO. There were no major adverse cardiac events (MACE), revascularization, re-admission to the hospital secondary to angina, or acute myocardial infarction (AMI) for the 24-month period following cellular infusion. In addition, there were no periprocedural infusion-related complications including malignant arrhythmias, loss of normal coronary blood flow or acute neurologic events. Cardiac enzymes were negative in all patients. There was an improvement in the degree of ischemic myocardium, which was accompanied by a trend towards reduction in anginal symptoms. Intracoronary infusion of autologous CD133+ marrow-derived cells is safe and feasible. Cellular therapy with CD133+ cells to reduce anginal symptoms and to improve ischemia in patients with CTO awaits clinical investigation in Phase II/III trials.


Subject(s)
Angina Pectoris/therapy , Antigens, CD/metabolism , Cell- and Tissue-Based Therapy/methods , Coronary Occlusion/therapy , Glycoproteins/metabolism , Ischemia/therapy , Peptides/metabolism , Stem Cell Transplantation/methods , Stem Cells/metabolism , AC133 Antigen , Adult , Angina Pectoris/etiology , Coronary Occlusion/complications , Humans , Ischemia/complications , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...