Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Cardiopulm Rehabil Prev ; 37(1): 39-48, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27676463

ABSTRACT

PURPOSE: Hopelessness is associated with increased adverse events and decreased survival in patients with coronary heart disease (CHD). The purpose of this research was to examine the effect of regular home- and hospital-based cardiac rehabilitation (CR) exercise on hopelessness levels in patients with CHD, hypothesizing that increased exercise in either setting would lead to decreased state hopelessness. METHODS: A descriptive longitudinal design was used at a large teaching hospital in Michigan. A total of 324 patients provided data during hospitalization and at least 1 followup time point (3, 8, and 12 months). RESULTS: The patients had persistent, modest levels of state and trait hopelessness across all time points. Among home exercisers with moderate to severe state hopelessness at baseline, both mean state (P = .002) and trait (P = .02) hopelessness were reduced at later time points compared with those who quit or did not start exercise. Multivariable models showed that when individuals had moderate to severe baseline state hopelessness, home exercise remained associated with decreases in state hopelessness compared with no exercise, even after adjusting for hospital exercise, depression, and demographic variables. CONCLUSIONS: Exercise may be effective in reducing moderate to severe hopelessness in patients with CHD. Moderate to severe baseline state hopelessness was a predictor of attrition in this cohort, especially for home exercisers, but this was mediated in hospital-based programs. Further research is needed to determine how hopeless individuals can be encouraged to exercise and whether home- or a hospital-based CR exercise is superior in impacting hopelessness.


Subject(s)
Cardiac Rehabilitation/methods , Coronary Disease/rehabilitation , Exercise Therapy/methods , Home Care Services/statistics & numerical data , Hope , Hospitalization/statistics & numerical data , Adult , Aged , Aged, 80 and over , Cardiac Rehabilitation/psychology , Cardiac Rehabilitation/statistics & numerical data , Coronary Disease/psychology , Exercise Therapy/psychology , Exercise Therapy/statistics & numerical data , Female , Follow-Up Studies , Humans , Longitudinal Studies , Male , Michigan , Middle Aged , Treatment Outcome
2.
West J Nurs Res ; 36(4): 552-70, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24122739

ABSTRACT

Hopelessness is predictive in the development of coronary heart disease (CHD) and can persist in patients after a CHD event, adversely affecting recovery. Hopelessness may represent a temporary response (state) or a chronic outlook (trait). Common hopelessness measures fail to differentiate state from trait hopelessness, a potentially important differentiation for treatment. The State-Trait Hopelessness Scale (STHS) was developed and pilot tested with two groups of college students (n = 39 and 190) and patients with CHD (n = 44). The instrument was then used with 520 patients, confirming reliability (Cronbach's α) for the State (.88) and Trait (.91) subscales and concurrent and predictive validity. Separate exploratory factor analyses showed two factors (hopelessness present or hopelessness absent) for the State and Trait subscales, accounting for 58.9% and 57.3% of variance, respectively. These findings support future use of the tool in clinical settings and in intervention studies focused on hopelessness.


Subject(s)
Coronary Disease/psychology , Hope , Humans , Pilot Projects
3.
PLoS One ; 8(8): e72749, 2013.
Article in English | MEDLINE | ID: mdl-24023642

ABSTRACT

BACKGROUND: Cellular therapy is a promising therapeutic strategy for malignant diseases. The efficacy of this therapy can be limited by poor infiltration of the tumor by immune effector cells. In particular, NK cell infiltration is often reduced relative to T cells. A novel class of fusion proteins was designed to enhance the recruitment of specific leukocyte subsets based on their expression of a given chemokine receptor. The proteins are composed of an N-terminal chemokine head, the mucin domain taken from the membrane-anchored chemokine CX3CL1, and a C-terminal glycosylphosphatidylinositol (GPI) membrane anchor replacing the normal transmembrane domain allowing integration of the proteins into cell membranes when injected into a solid tumor. The mucin domain in conjunction with the chemokine head acts to specifically recruit leukocytes expressing the corresponding chemokine receptor. METHODOLOGY/PRINCIPAL FINDINGS: A fusion protein comprising a CXCL10 chemokine head (CXCL10-mucin-GPI) was used for proof of concept for this approach and expressed constitutively in Chinese Hamster Ovary cells. FPLC was used to purify proteins. The recombinant proteins efficiently integrated into cell membranes in a process dependent upon the GPI anchor and were able to activate the CXCR3 receptor on lymphocytes. Endothelial cells incubated with CXCL10-mucin-GPI efficiently recruited NK cells in vitro under conditions of physiologic flow, which was shown to be dependent on the presence of the mucin domain. Experiments conducted in vivo using established tumors in mice suggested a positive effect of CXCL10-mucin-GPI on the recruitment of NK cells. CONCLUSIONS: The results suggest enhanced recruitment of NK cells by CXCL10-mucin-GPI. This class of fusion proteins represents a novel adjuvant in cellular immunotherapy. The underlying concept of a chemokine head fused to the mucin domain and a GPI anchor signal sequence may be expanded into a broader family of reagents that will allow targeted recruitment of cells in various settings.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Chemokine CXCL10/therapeutic use , Glycosylphosphatidylinositols/therapeutic use , Immunotherapy , Killer Cells, Natural/immunology , Neoplasms/therapy , Recombinant Fusion Proteins/therapeutic use , Animals , CHO Cells , Cell Adhesion , Cell Line , Cell Membrane/metabolism , Cricetinae , Cricetulus , Endocytosis , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Glycosylphosphatidylinositols/isolation & purification , Humans , Mice , Mice, Inbred C57BL , Mucins/metabolism , Neoplasm Transplantation , Neoplasms/immunology , Neoplasms/pathology , Protein Engineering , Recombinant Fusion Proteins/isolation & purification , Rheology
4.
J Invest Dermatol ; 133(3): 803-811, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23096710

ABSTRACT

Tissue inhibitors of metalloproteinases exhibit diverse physiological/biological functions including moderation of the proteolytic processing of growth factors and turnover of extracellular matrix. These various biological activities are linked in part to the stoichiometry of tissue inhibitor of metalloprotein/matrix metalloprotein (TIMP/MMP)/surface protein interactions. TIMP-1, a secreted protein, can be detected on the cell surface only through its interaction with surface-bound proteins. Proteins anchored by glycosylphosphatidylinositol (GPI), when purified and added to cells or tissues, are efficiently incorporated into their surface membranes. A GPI anchor was fused to TIMP-1 to focus defined concentrations of the inhibitory protein independently on the surface of primary dermal fibroblast cells. Exogenously added recombinant TIMP-1-GPI effectively inserted into the cell membrane of fibroblasts blocked the secretion of MMPs and markedly altered the stoichiometry of MMP association with the cell surface. TIMP-1-GPI treatment resulted in inhibition of fibroblast-reduced proliferation, and transiently reduced expression of fibrosis-associated genes. These effects were dose dependent. Treated cells also showed a more proapoptotic phenotype based on apoptotic assays and western blot analysis for apoptosis-associated protein expression. GPI-anchored TIMP-1 may represent a more effective version of the protein for use in therapeutic approaches to help control fibrosis and scar formation.


Subject(s)
Cicatrix/physiopathology , Dermis/pathology , Fibroblasts/drug effects , GPI-Linked Proteins/pharmacology , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Apoptosis , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Humans , In Vitro Techniques , Matrix Metalloproteinases/metabolism , Tumor Necrosis Factor-alpha/pharmacology
5.
Proc Natl Acad Sci U S A ; 109(6): E309-16, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22223664

ABSTRACT

Tissue inhibitor of metalloproteinases 1 (TIMP-1) is a matrix metalloproteinase (MMP)-independent regulator of growth and apoptosis in various cell types. The receptors and signaling pathways that are involved in the growth factor activities of TIMP-1, however, remain controversial. RNA interference of TIMP-1 has revealed that endogenous TIMP-1 suppresses the proliferation, metabolic activity, and osteogenic differentiation capacity of human mesenchymal stem cells (hMSCs). The knockdown of TIMP-1 in hMSCs activated the Wnt/ß-catenin signaling pathway as indicated by the increased stability and nuclear localization of ß-catenin in TIMP-1-deficient hMSCs. Moreover, TIMP-1 knockdown cells exhibited enhanced ß-catenin transcriptional activity, determined by Wnt/ß-catenin target gene expression analysis and a luciferase-based ß-catenin-activated reporter assay. An analysis of a mutant form of TIMP-1 that cannot inhibit MMP indicated that the effect of TIMP-1 on ß-catenin signaling is MMP independent. Furthermore, the binding of CD63 to TIMP-1 on the surface of hMSCs is essential for the TIMP-1-mediated effects on Wnt/ß-catenin signaling. An array analysis of microRNAs (miRNAs) and transfection studies with specific miRNA inhibitors and mimics showed that let-7f miRNA is crucial for the regulation of ß-catenin activity and osteogenic differentiation by TIMP-1. Let-7f was up-regulated in TIMP-1-depleted hMSCs and demonstrably reduced axin 2, an antagonist of ß-catenin stability. Our results demonstrate that TIMP-1 is a direct regulator of hMSC functions and reveal a regulatory network in which let-7f modulates Wnt/ß-catenin activity.


Subject(s)
Mesenchymal Stem Cells/enzymology , MicroRNAs/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Wnt Signaling Pathway , Cell Differentiation , Cell Line , Cell Proliferation , Gene Knockdown Techniques , Humans , Mesenchymal Stem Cells/cytology , Osteogenesis , Protein Binding , Tetraspanin 30/metabolism , beta Catenin/metabolism
6.
Biol Chem ; 393(12): 1463-70, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23667903

ABSTRACT

Tissue inhibitor of metalloproteinase 1 (TIMP-1) controls matrix metalloproteinase activity through 1:1stoichiometric binding. Human TIMP-1 fused to a glycosylphosphatidylinositol(GPI) anchor (TIMP-1 - GPI) shifts the activity of TIMP-1 from the extracellular matrix to the cell surface. TIMP-1 - GPI treated renal cell carcinoma cells show increased apoptosis and reduced proliferation.Transcriptomic profiling and regulatory pathway mapping were used to identify the potential mechanisms driving these effects. Significant changes in the DNA binding inhibitors, TGF- ß 1/SMAD and BMP pathways resulted from TIMP-1 - GPI treatment. These events were linked to reduced TGF- ß 1 signaling mediated by inhibition of proteolytic processing of latent TGF- ß 1 by TIMP-1 - GPI.


Subject(s)
Carcinoma, Renal Cell/therapy , Glycosylphosphatidylinositols/therapeutic use , Kidney Neoplasms/therapy , Recombinant Fusion Proteins/therapeutic use , Tissue Inhibitor of Metalloproteinase-1/therapeutic use , Transforming Growth Factor beta1/metabolism , Apoptosis , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glycosylphosphatidylinositols/genetics , Humans , Kidney/cytology , Kidney/metabolism , Kidney/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Matrix Metalloproteinases/metabolism , Oligonucleotide Array Sequence Analysis , Recombinant Fusion Proteins/genetics , Signal Transduction , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism
7.
Am J Pathol ; 179(1): 436-51, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21703422

ABSTRACT

Tissue dendritic cells (DCs) may influence the progression of renal cell carcinoma (RCC) by regulating the functional capacity of antitumor effector cells. DCs and their interaction with T cells were analyzed in human RCC and control kidney tissues. The frequency of CD209(+) DCs in RCCs was found to be associated with an unfavorable T(H)1 cell balance in the tissue and advanced tumor stages. The CD209(+) DCs in RCC were unusual because most of them co-expressed macrophage markers (CD14, CD163). The phenotype of these enriched-in-renal-carcinoma DCs (ercDCs) could be reiterated in vitro by carcinoma-secreted factors (CXCL8/IL-8, IL-6, and vascular endothelial growth factor). ErcDCs resembled conventional DCs in costimulatory molecule expression and antigen cross-presentation. They did not suppress cognate cytotoxic T-lymphocyte function and did not cause CD3ζ down-regulation, FOXP3 induction, or T-cell apoptosis in situ or in vitro; thus, they are different from classic myeloid-derived suppressor cells. ErcDCs secreted high levels of metalloproteinase 9 and used T-cell crosstalk to increase tumor-promoting tumor necrosis factor α and reduce chemokines relevant for T(H)1-polarized lymphocyte recruitment. This modulation of the tumor environment exerted by ercDCs suggests an immunologic mechanism by which tumor control can fail without involving cytotoxic T-lymphocyte inhibition. Pharmacologic targeting of the deviated DC differentiation could improve the efficacy of immunotherapy against RCC.


Subject(s)
Carcinoma, Renal Cell/immunology , Dendritic Cells/immunology , Kidney Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Apoptosis , Blotting, Western , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Case-Control Studies , Cell Differentiation , Cell Movement , Cell Proliferation , Chemokines/metabolism , Cross-Priming , Cytokines/metabolism , Dendritic Cells/metabolism , Dendritic Cells/pathology , Endocytosis , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Interleukin-6/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Middle Aged , Phagocytosis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology , Tumor Cells, Cultured
8.
Biol Chem ; 390(9): 893-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19558322

ABSTRACT

Exogenous application of recombinant TIMP-1 protein modified by addition of a glycosylphosphatidylinositol (GPI) anchor allows efficient insertion of the fusion protein into cell membranes. This 'cell surface engineering' leads to changes in the proteolytic environment. TIMP-1-GPI shows enhanced as well as novel in vitro biological activities including suppression of proliferation, reduced migration, and inhibition of invasion of the colon carcinoma cell line SW480. Treatment of SW480 tumors implanted in Rag (-/-) common gamma chain (-/-) C57BL/6 mice with peritumorally applied TIMP-1-GPI, control rhTIMP-1 protein, or vehicle shows that TIMP-1-GPI leads to a significant reduction in tumor growth.


Subject(s)
Colonic Neoplasms/drug therapy , DNA-Binding Proteins/physiology , Glycosylphosphatidylinositols/therapeutic use , Tissue Inhibitor of Metalloproteinase-1/therapeutic use , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , Glycosylphosphatidylinositols/chemistry , Humans , Matrix Metalloproteinase 7/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Tissue Inhibitor of Metalloproteinase-1/chemistry
9.
Cancer Immunol Immunother ; 58(3): 361-71, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18618109

ABSTRACT

Resistance to apoptosis is a prominent feature of malignant melanoma. Hyperthermic therapy can be an effective adjuvant treatment for some tumors including melanoma. We developed a fusion protein based on the tissue inhibitor of matrix metalloproteinase-1 linked to a glycosylphosphatidylinositol anchor (TIMP-1-GPI). The TIMP-1-GPI-fusion protein shows unique properties. Exogenous administration of TIMP-1-GPI can result in transient morphological changes to treated cells including modulation of proliferation and decreased resistance to apoptosis. The effect of TIMP-1-GPI on the biology of melanoma in the context of a defined hyperthermic dose was evaluated in vitro. Clonogenic assays were used to measure cell survival. Gelatinase zymography determined secretion of MMP-2 and MMP-9. Monoclonal antibody against FAS/CD95 was applied to induce apoptosis. The expression of pro- and anti-apoptotic proteins and the secretion of immunoregulatory cytokines were then evaluated using Western blot and ELISA. TIMP-1-GPI combined with a sub-lethal hyperthermic treatment (41.8 degrees C for 2 h) suppressed tumor cell growth capacity as measured by clonogenic assay. The co-treatment also significantly suppressed tumor cell proliferation, enhanced FAS receptor surface expression increased tumor cell susceptibility to FAS-mediated killing. The increased sensitivity to FAS-induced apoptosis was linked to alterations in the apoptotic mediators Bcl-2, Bax, Bcl-XL and Apaf-1. The agent works in concert with sub-lethal hyperthermic treatment to render melanoma cells sensitive to FAS killing. The targeted delivery of TIMP-1-GPI to tumor environments in the context of regional hyperthermic therapy could be optimized through the use of thermosensitive liposomes.


Subject(s)
Apoptosis , Glycosylphosphatidylinositols/metabolism , Hyperthermia, Induced , Melanoma/therapy , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Tissue Inhibitor of Metalloproteinase-1/metabolism , fas Receptor/metabolism , Cell Proliferation , Cell Separation , Cell Survival , Flow Cytometry , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...