Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Model Earth Syst Environ ; 9(2): 1633-1649, 2023.
Article in English | MEDLINE | ID: mdl-36341043

ABSTRACT

Soil erosion and sedimentation contribute to deteriorating water quality, adverse alterations in basin hydrology and overall ecosystem biogeochemistry. Thus, understanding soil erosion patterns in catchments is critical for conservation planning. This study was conducted in a peri-urban Inner Murchison Bay (IMB) catchment on the northern shores of Lake Victoria since most soil erosion studies in Sub-Saharan Africa have been focused on rural landscapes. The study sought to identify sediment sources by mapping erosion hotspots using the revised universal soil loss equation (RUSLE) model in appendage with field walks. RUSLE model was built in ArcGIS 10.5 software with factors including: rainfall erosivity, soil erodibility, slope length and steepness, land cover and support practices. The model was run, producing an erosion risk map and field assessments conducted to ground-truth findings and identify other hotspots. The percentage areas for RUSLE modelled erosion rates were: 66.8% for 0-2 t ha-1 year-1; 10.8% for 2-5 t ha-1 year-1; 10.1% for 5-10 t ha-1 year-1; 9% for 10-50 t ha-1 year-1 and 3.3% for 50-100 t ha-1 year-1. Average erosion risk was 7 t ha-1 year-1 and the total watershed erosion risk was 197,400 t year-1, with croplands and steep areas (slope factor > 20) as the major hotspots (> 5 t ha-1 year-1). Field walks revealed exposed soils, marrum (gravel) roads and unlined drainage channels as other sediment sources. This study provided the first assessment of erosion risk in this peri-urban catchment, to serve as a basis for identifying mitigation priorities. It is recommended that tailored soil and water conservation measures be integrated into physical planning, focusing on identified non-conventional hotspots to ameliorate sediment pollution in Lake Victoria.

2.
J Immunol ; 173(3): 2143-50, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15265951

ABSTRACT

The major limiting factor in the successful application of adjuvant therapy for metastatic disease is the lack of adjuvant specificity that leads to severe side effects. Reasoning that T cells of the immune system are highly specific, we generated tumor-specific T cells by genetic modification of mouse primary T cells with a chimeric receptor reactive with the human breast cancer-associated Ag erbB-2. These T cells killed breast cancer cells and secreted IFN-gamma in an Ag-specific manner in vitro. We investigated their use against metastatic breast cancer in mice in an adjuvant setting, and compared their effectiveness with the commonly applied adjuvants doxorubicin, 5-fluorouracil, and herceptin. Mice were inoculated orthotopically with the human erbB-2-expressing spontaneously metastatic mouse breast cancer 4T1.2 in mammary tissue, and the primary tumor was surgically removed 8 days later. Significant metastatic disease was demonstrated in lung and liver at the time of surgery on day 8 with increased tumor burden at later time points. T cell adjuvant treatment of day 8 metastatic disease resulted in dramatic increases in survival of mice, and this survival was significantly greater than that afforded by either doxorubicin, 5-fluorouracil, or herceptin.


Subject(s)
Carcinoma/secondary , Immunotherapy, Adoptive , Mammary Neoplasms, Experimental/therapy , Membrane Proteins/genetics , Neoplasm Proteins/immunology , Receptor, ErbB-2/immunology , Receptors, Antigen, T-Cell/genetics , T-Cell Antigen Receptor Specificity/genetics , T-Lymphocyte Subsets/transplantation , Animals , Antibiotics, Antineoplastic/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Carcinoma/pathology , Carcinoma/surgery , Carcinoma/therapy , Chemotherapy, Adjuvant , Combined Modality Therapy , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm , Female , Fluorouracil/therapeutic use , Genetic Engineering , Humans , Interferon-gamma/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/surgery , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasm Transplantation , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Trastuzumab
3.
Cancer Gene Ther ; 11(5): 371-9, 2004 May.
Article in English | MEDLINE | ID: mdl-15060573

ABSTRACT

T cells engineered to express single-chain antibody receptors that incorporate TCR-zeta and cluster designation (CD)28 signaling domains (scFv-alpha-erbB2-CD28-zeta) can be redirected in vivo to cancer cells that lack triggering costimulatory molecules. To assess the contribution of CD28 signaling to the function of the scFv-CD28-zeta receptor, we expressed a series of mutated scFv-CD28-zeta receptors directed against erbB2. Residues known to be critical for CD28 signaling were mutated from tyrosine to phenylalanine at position 170 or proline to alanine at positions 187 and 190. Primary mouse T cells expressing either of the mutant receptors demonstrated impaired cytokine (IFN-gamma and GM-CSF) production and decreased proliferation after antigen ligation in vitro and decreased antitumor efficacy in vivo compared with T cells expressing the wild-type scFv-CD28-zeta receptor, suggesting a key signaling role for the CD28 component of the scFv-CD28-zeta receptor. Importantly, cell surface expression, binding capacity and cytolytic activity mediated by the scFv-CD28-zeta receptor were not diminished by either mutation. Overall, this study has definitively demonstrated a functional role for the CD28 component of the scFv-CD28-zeta receptor and has shown that incorporation of costimulatory activity in chimeric scFv receptors is a powerful approach for improving adoptive cancer immunotherapy.


Subject(s)
CD28 Antigens/immunology , Immunotherapy, Adoptive , Neoplasms, Experimental/therapy , Receptor, ErbB-2/immunology , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology , Amino Acid Substitution/genetics , Amino Acid Substitution/immunology , Animals , CD28 Antigens/genetics , Cell Line, Tumor , Humans , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Immunotherapy, Adoptive/methods , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/immunology , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , Retroviridae , Transduction, Genetic
4.
J Exp Med ; 199(4): 437-48, 2004 Feb 16.
Article in English | MEDLINE | ID: mdl-14769851

ABSTRACT

Because tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) preferentially induces apoptosis in tumor cells and plays a critical role in tumor surveillance, its receptor is an attractive target for antibody-mediated tumor therapy. Here we report that a monoclonal antibody (mAb) against the mouse TRAIL receptor, DR5, exhibited potent antitumor effects against TRAIL-sensitive tumor cells in vivo by recruiting Fc receptor-expressing innate immune cells, with no apparent systemic toxicity. Administration of the agonistic anti-DR5 mAb also significantly inhibited experimental and spontaneous tumor metastases. Notably, the anti-DR5 mAb-mediated tumor rejection by innate immune cells efficiently evoked tumor-specific T cell immunity that could also eradicate TRAIL-resistant variants. These results suggested that the antibody-based therapy targeting DR5 is an efficient strategy not only to eliminate TRAIL-sensitive tumor cells, but also to induce tumor-specific T cell memory that affords a long-term protection from tumor recurrence.


Subject(s)
Receptors, Tumor Necrosis Factor/immunology , T-Lymphocytes/immunology , Alanine Transaminase/blood , Animals , Apoptosis Regulatory Proteins , Aspartate Aminotransferases/blood , Immunity, Cellular/immunology , Immunotherapy/methods , Mammary Neoplasms, Animal/immunology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , Mice, SCID , Receptors, Fc/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
5.
J Immunol ; 172(2): 757-61, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14707044

ABSTRACT

NKG2D is an activation receptor on NK cells and has been demonstrated as a primary cytotoxicity receptor for mouse NK cells. Primary rejection of class I-deficient RMA-S lymphoma cells expressing the NKG2D ligand, retinoic acid early inducible-1beta, was critically dependent upon NK cell perforin and occurred independently of T cells. NKG2D-triggered NK cell rejection of RMA-S-retinoic acid early inducible-1beta tumor primed a secondary tumor-specific T cell response mediated by both CD4+ and CD8+ T cells in the effector phase. Surprisingly, during the priming phase, CD4+ T cells, but not CD8+ T cells, were also required to generate this secondary T cell immunity; however, T cell priming was independent of Th1 cytokines, such as IFN-gamma and IL-12. These data imply a novel pathway for priming T cell immunity, that is, stimulated upon NK cell-mediated cytotoxicity of NKG2D ligand-expressing tumor cells, dependent upon CD4+ T cells in the primary phase, and independent of conventional Th1-type immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Graft Rejection/immunology , Killer Cells, Natural/immunology , Receptors, Immunologic/physiology , Th1 Cells/immunology , Animals , Cell Line, Tumor , Cytotoxicity, Immunologic/genetics , Immunity, Cellular/genetics , Immunity, Innate/genetics , Immunization, Secondary , Immunologic Memory/genetics , Injections, Subcutaneous , Killer Cells, Natural/transplantation , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K , Neoplasm Transplantation , Receptors, Natural Killer Cell , T-Lymphocytes, Regulatory/immunology
6.
Cancer Res ; 63(1): 207-13, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12517799

ABSTRACT

Recent reports have suggested that the death ligand tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) may partially limit the formation of hepatic metastases of a variety of mouse tumors, and the major source of TRAIL in the liver was shown to be local natural killer cells. We isolated a clone (R331) of the murine renal cancer cell line Renca that was strikingly more sensitive to both Fas and TRAIL death receptor-mediated apoptosis in vitro. R331 grew in tissue culture in vitro at a rate similar to that of the parental Renca cell line but formed larger and more numerous colonies than parental Renca in soft agar. After s.c. implantation, R331 tumors progressed more rapidly than parental Renca tumors. However, R331 formed far fewer lung and liver metastases in wild-type (WT) BALB/c mice. Administration of antibodies that neutralized TRAIL dramatically increased the number of R331 liver metastases. Furthermore, numbers of R331 liver metastases were much greater in TRAIL(-/-) than in WT BALB/c mice. In contrast, no difference was seen in numbers of lung metastases when comparing TRAIL(-/-) and WT mice, suggesting that the antitumor effects of TRAIL in vivo were compartment specific. Transfection of cellular Fas-associated death domain-like interleukin-1beta-converting enzyme inhibitory protein into R331 increased the numbers of liver metastases in BALB/c mice by up to 10-fold, indicating that local TRAIL in the liver was directly mediating tumor cell apoptosis. These organ-specific differences in the endogenous levels of death ligands may apply different selective pressures on the development of liver or lung metastases. Consequently, the efficacy of TRAIL therapy may vary depending on the location of the tumor metastases.


Subject(s)
Apoptosis/physiology , Carcinoma, Renal Cell/secondary , Immunity, Innate/physiology , Kidney Neoplasms/pathology , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Membrane Glycoproteins/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis Regulatory Proteins , Carcinoma, Renal Cell/pathology , Caspase 3 , Caspases/metabolism , Cell Division , Flow Cytometry , Immunity, Innate/genetics , Liver Neoplasms/pathology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...