Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
J Geriatr Psychiatry Neurol ; 24(4): 229-34, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22228830

ABSTRACT

UNLABELLED: This study examined the prevalence of irritability and social isolation in veterans with dementia, with and without depression. Participants were diagnosed with dementia and enrolled in a dementia care-coordination and support-service intervention. Participants were interviewed and underwent assessment with the 10-item Center for Epidemiologic Studies Depression scale, a Patient Strain Measure and the Short Blessed Test. In all, of 294 participants completing interviews, 77 (26.2%) were depressed and 107 (36.4%) endorsed irritability; mean social isolation score was 1.59 ± 1.96. Irritability was significantly more likely to be present in depressed versus nondepressed participants (P < .0001), but this relationship was moderated by dementia severity. The mean social isolation score was also significantly more elevated in depressed rather than nondepressed patients (2.82 ± 1.96 vs 1.15 ± 1.76, respectively). CONCLUSIONS: Depressed persons with dementia are significantly more likely to experience irritability and social isolation than those who are not depressed.


Subject(s)
Dementia/complications , Depression/complications , Irritable Mood/physiology , Social Isolation/psychology , Aged , Aged, 80 and over , Cohort Studies , Dementia/epidemiology , Depression/epidemiology , Depression/etiology , Female , Humans , Male , Prevalence , Psychiatric Status Rating Scales , Severity of Illness Index , United States/epidemiology , Veterans/psychology
2.
J Cell Biochem ; 97(2): 217-25, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16288472

ABSTRACT

Heparanase (HPSE-1) is an endo-beta-D-glucuronidase that cleaves heparan sulfate (HS) chains of proteoglycans (HSPG), and its expression has been associated with increased cell growth, invasion, and angiogenesis of tumors as well as with embryogenesis and tissue development. Since metastatic cancer cells express HPSE-1, we have developed an orthotopic brain slice model to study HPSE-1 involvement in brain-metastatic melanoma. This model allows for the characterization of tumor cell invasion at both quantitative and qualitative levels. Brain-metastatic melanoma cells (B16B15b) showed augmenting levels of HPSE-1 protein expression in a time-dependent manner. Secondly, B16B15b cells pre-treated with HPSE-1 showed a significant increase in the number of cells that invaded into the brain tissue. Finally, HPSE-1 exposure-augmented invasion depth in brain sections by brain-metastatic melanoma cells. We concluded that applying this brain slice model can be beneficial to investigate HPSE-1- related in vivo modalities in brain-metastatic melanoma and brain invasion in general. These results also further emphasize the potential relevance of using this model to design therapies for controlling this type of cancer by blocking HPSE-1 functionality.


Subject(s)
Brain Neoplasms/secondary , Glucuronidase/metabolism , Glucuronidase/pharmacology , Melanoma/pathology , Tissue Culture Techniques , Animals , Brain , Brain Neoplasms/pathology , Cell Line, Tumor , Female , Melanoma/enzymology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness
3.
Neoplasia ; 7(3): 253-62, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15799825

ABSTRACT

Cancer metastasis, is a frequent manifestation of malignant melanoma progression. Successful invasion into distant organs by tumor cells must include attachment to microvessel endothelial cells, and degradation of basement membranes and extracellular matrix (ECM). Heparan sulfate proteoglycans (HSPG) are essential and ubiquitous macromolecules associated with the cell surface and ECM of a wide range of cells and tissues. Heparanase (HPSE-1) is an ECM degradative enzyme, which degrades the heparan sulfate (HS) chains of HSPG at specific intrachain sites. To investigate effects of changes in heparanase gene expression in metastatic melanoma cells, we constructed adenoviral vectors containing the full-length human HPSE-1 cDNA in both sense (Ad-S/hep) and antisense orientations (Ad-AS/hep). We found increased HPSE-1 expression and activity in melanoma cell lines following Ad-S/hep infection by Western blot analyses and specific HPSE-1 activity assay. Conversely, HPSE-1 content was significantly inhibited following infection with Ad-AS/Hep. Importantly, HPSE-1 modulation by these adenoviral constructs correlated with invasive cellular properties in vitro and in vivo. Our results suggest that HPSE-1 not only contributes to the invasive phenotype of melanoma cells, but also that the Ad-AS/hep-mediated inhibition of its enzymatic activity can be efficacious in the prevention and treatment of melanoma metastasis.


Subject(s)
Genetic Therapy/methods , Glucuronidase/antagonists & inhibitors , Glucuronidase/genetics , Melanoma/pathology , Oligonucleotides, Antisense/metabolism , Adenoviridae/genetics , Animals , Blotting, Western , Cattle , Cell Line, Tumor , Cell Nucleus/metabolism , DNA, Complementary/metabolism , Heparan Sulfate Proteoglycans/chemistry , Humans , Immunohistochemistry , Kidney/metabolism , Melanoma/metabolism , Melanoma/therapy , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Oligonucleotides, Antisense/chemistry , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
4.
Int J Oncol ; 26(2): 345-52, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15645118

ABSTRACT

Heparanase (HPSE-1) is an endo-beta-D-glucuronidase that cleaves heparan sulfate proteoglycans (HSPG), and its expression has been associated with increased growth, metastasis, and angiogenesis of tumors. Since metastatic melanoma cells express high levels of HSPG and because melanoma tumors grow highly vascularized, we analyzed melanoma tissue specimens for HPSE-1 expression from experimental animals as well as from patients. Laser capture microdissection microscopy was used to extract melanoma cell populations and to isolate them from adjacent tissue. In experimental animals, a 29-fold upregulation of HPSE-1 expression was detected by real-time PCR in metastatic melanoma compared to normal lung tissue. Additionally, immunohistochemistry (IHC) revealed selective HPSE-1 staining in human metastatic melanoma when compared to primary melanoma tumors from the same patient. IHC also showed a marked staining for the enzyme around blood vessels and in vascularized regions. Our results provide evidence demonstrating that HPSE-1 likely plays important roles in regulating the in vivo growth and progression of melanoma. These results further emphasize the importance that therapies designed to block HPSE-1 activity may aid in controlling this type of cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Glucuronidase/biosynthesis , Melanoma/enzymology , Animals , Brain Neoplasms/enzymology , Brain Neoplasms/secondary , Cell Line, Tumor , Cell Proliferation , DNA Primers/chemistry , DNA, Complementary/metabolism , Disease Progression , Female , Humans , Immunohistochemistry , Lasers , Lung/pathology , Melanoma/pathology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms/metabolism , Neovascularization, Pathologic , Polymerase Chain Reaction , RNA/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Up-Regulation
5.
Neuro Oncol ; 6(2): 154-65, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15134630

ABSTRACT

Brain metastasis, which occurs in 20% to 40% of all cancer patients, is an important cause of neoplastic morbidity and mortality. Successful invasion into the brain by tumor cells must include attachment to microvessel endothelial cells, penetration through the blood-brain barrier, and, of relevance, a response to brain survival and growth factors. Neurotrophins (NTs) are important in brain-invasive steps. Human melanoma cell lines express low-affinity NT receptor p75NTR in relation to their brain-metastatic propensity with their invasive properties being regulated by NGF, or nerve growth factor, the prototypic NT. They also express functional TrkC, the putative receptor for the invasion-promoting NT-3. In brain-metastatic melanoma cells, NTs promote invasion by enhancing the production of extracellular matrix (ECM)-degradative enzymes such as heparanase, an enzyme capable of locally destroying both ECM and the basement membrane of the blood-brain barrier. Heparanase is an endo-beta-d-glucuronidase that cleaves heparan sulfate (HS) chains of ECM HS proteoglycans, and it is a unique metastatic determinant because it is the dominant mammalian HS degradative enzyme. Brain-metastatic melanoma cells also produce autocrine/paracrine factors that influence their growth, invasion, and survival in the brain. Synthesis of these factors may serve to regulate NT production by brain cells adjacent to the neoplastic invasion front, such as astrocytes. Increased NT levels have been observed in tumor-adjacent tissues at the invasion front of human brain melanoma. Additionally, astrocytes may contribute to the brain-metastatic specificity of melanoma cells by producing NT-regulated heparanase. Trophic, autocrine, and paracrine growth factors may therefore determine whether metastatic cells can successfully invade, colonize, and grow in the CNS.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Melanoma/metabolism , Melanoma/secondary , Nerve Growth Factors/physiology , Animals , Humans , Receptors, Nerve Growth Factor/physiology
6.
J Cell Biochem ; 91(1): 206-15, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14689592

ABSTRACT

The p75 neurotrophin receptor (p75(NTR)), a common receptor for members of the neurotrophins (NT) family, was previously identified as a molecular determinant of brain metastasis. We have also reported that NT treatment of murine and human brain-metastatic melanoma cells affects their invasive capacities and increases the production of heparanase, an important and unique extracellular matrix (ECM) degradative enzyme. Neurotrophism can be a survival-support mechanism for brain-metastatic cells and a survival assay was devised to mimic the growth limiting conditions of rapidly expanding metastatic tumors prior to neoangiogenesis. We report that p75(NTR) promoted the survival of brain-metastatic melanoma cells but not melanocytes in stress cultures conditions. Secondly, melanoma cells fluorescently sorted for high p75(NTR) expression (p75(NTR-H) cells) had an up to a 15-fold greater survival than those sorted for low p75(NTR) expression (p75(NTR-L) cells). Thirdly, cells overexpressing p75(NTR) associated with the growth fraction and provided these cells with an inherent growth advantage. Finally, we observed an increased survival of sorted p75(NTR-L) cells, dependent upon treatment of NT members whose functional receptors are present on these cells. Together, these results delineate that p75(NTR)-mediated trophic support profoundly affects competitive melanoma-cell survival when the tumor cell microenvironment becomes growth limiting.


Subject(s)
Brain Neoplasms/metabolism , Melanoma/metabolism , Nerve Growth Factors/pharmacology , Receptors, Nerve Growth Factor/metabolism , Skin Neoplasms/metabolism , Animals , Brain Neoplasms/secondary , Cell Survival/drug effects , Cell Survival/physiology , Flow Cytometry , Glucuronidase/metabolism , Humans , Melanocytes/metabolism , Mice , Receptor, Nerve Growth Factor , Signal Transduction/physiology , Tumor Cells, Cultured
7.
Pathol Oncol Res ; 9(3): 147-58, 2003.
Article in English | MEDLINE | ID: mdl-14530807

ABSTRACT

The brain is a unique microenvironment enclosed by the skull and maintaining a highly regulated vascular transport barrier. To metastasize to the brain, malignant tumor cells must attach to microvessel endothelial cells, invade the blood-brain barrier (BBB), and respond to brain survival and growth factors. Neurotrophins (NT) are important in brain invasion because they stimulate this process. In brain-metastatic melanoma cells, NT can promote invasion by enhancing the production of extracellular matrixdegradative enzymes such as heparanase, an enzyme capable of locally destroying both the extracellular matrix and the basement membrane of the BBB. We have examined human and murine melanoma cell lines exhibiting varying abilities to form brain metastases, and have found that they express low-affinity neurotrophin receptor p75NTR in relation to their brain-metastatic potentials. They do not, however, express trkA, the gene encoding the tyrosine kinase receptor TrkA, the high-affinity receptor for nerve growth factor (NGF), the prototypic NT. Presence of functional TrkC, the putative receptor for the invasion-promoting neurotrophin NT-3, was also expressed in these cells. Brain-metastatic melanoma cells can also produce autocrine factors and inhibitors that influence their growth, invasion, and survival in the brain. Synthesis of these factors may influence NT production by brain cells adjacent to the neoplastic invasion front, such as oligodendrocytes and astrocytes. In brain biopsies, we observed increased amounts of NGF and NT-3 in tumor-adjacent tissues at the invasion front of human melanoma tumors. Additionally, we found that astrocytes contribute to the brain-metastatic specificity of melanoma cells by producing NT-regulated heparanase. Trophic, autocrine, and paracrine growth factors may therefore determine whether metastatic cells can successfully invade, colonize, and grow in the central nervous system (CNS).


Subject(s)
Brain Neoplasms/secondary , Melanoma/secondary , Nerve Growth Factors/physiology , Receptors, Nerve Growth Factor/physiology , Animals , Brain Neoplasms/metabolism , Humans , Melanoma/metabolism , Signal Transduction
8.
J Cell Biochem ; 88(5): 865-72, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12616526

ABSTRACT

The various members of the Trk tyrosine kinase family and p75 neurotrophin receptor (p75(NTR)) have been identified as signaling receptors for the structurally related members of the neurotrophins (NT) family. We have previously reported that NT treatment of murine and human brain-metastatic melanoma cells affects their invasive capacities and increases the production of extracellular-matrix degradative enzymes. These cells express aberrant levels of functional p75(NTR) and TrkC, the putative high-affinity receptor for the neurotrophin NT-3. Here we demonstrate that, by using sensitive immune-complex kinase assays in human brain-metastatic (70W) melanoma cells, TrkC receptors associate with a kinase activity exhibiting a dose-dependent susceptibility to inhibition by the purine-analogs 6-thioguanine and 2-aminopurine. The activity of this purine-analog-sensitive kinase (PASK) was induced by NT-3 in a time-dependent fashion, phosphorylating exogenous myelin basic protein (MBP) but not denatured enolase. It is similar to the one reported to relate with p75(NTR) and TrkA receptors and stimulated by the prototypic NT, nerve growth factor. Thus, PASKs may represent unique signaling components common to NT receptors that could engage joint downstream signaling effectors in brain-metastatic melanoma.


Subject(s)
Brain Neoplasms/metabolism , Melanoma/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , 2-Aminopurine/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Neoplasm Metastasis , Neurotrophin 3/pharmacology , Protein Kinases/analysis , Protein Kinases/chemistry , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/chemistry , Receptor, Nerve Growth Factor , Receptor, trkC/chemistry , Receptor, trkC/drug effects , Receptor, trkC/metabolism , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/metabolism , Signal Transduction , Thioguanine/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...