Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 13(5): e0196598, 2018.
Article in English | MEDLINE | ID: mdl-29742113

ABSTRACT

Quinoline-3-carboxamides (Q compounds) are immunomodulatory compounds that have shown efficacy both in autoimmune disease and cancer. We have in here investigated the impact of one such compound, paquinimod, on the development of diabetes in the NOD mouse model for type I diabetes (T1D). In cohorts of NOD mice treated with paquinimod between weeks 10 to 20 of age and followed up until 40 weeks of age, we observed dose-dependent reduction in incidence of disease as well as delayed onset of disease. Further, in contrast to untreated controls, the majority of NOD mice treated from 15 weeks of age did not develop diabetes at 30 weeks of age. Importantly, these mice displayed significantly less insulitis, which correlated with selectively reduced number of splenic macrophages and splenic Ly6Chi inflammatory monocytes at end point as compared to untreated controls. Collectively, these results demonstrate that paquinimod treatment can significantly inhibit progression of insulitis to T1D in the NOD mouse. We propose that the effect of paquinimod on disease progression may be related to the reduced number of these myeloid cell populations. Our finding also indicates that this compound could be a candidate for clinical development towards diabetes therapy in humans.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Immunosuppressive Agents/therapeutic use , Quinolines/therapeutic use , Animals , Diabetes Mellitus, Type 1/pathology , Female , Glycosuria/prevention & control , Immunosuppressive Agents/pharmacology , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , Macrophages/drug effects , Mice , Mice, Inbred NOD , Monocytes/drug effects , Myeloid Cells/drug effects , Quinolines/pharmacology
2.
Biochem Biophys Res Commun ; 500(3): 564-568, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29679568

ABSTRACT

We show here that increased S100A8 and S100A9 protein expression is induced in spleen of animals with active inflammation or with inoculated tumors. In tumor bearing animals an increased expression was also detected in the lung. To further analyze the induced proteins, we performed chemical cross-linking followed by Western blotting. We observed in protein extracts from spleen that both S100A8/S100A9 heterodimers as well as S100A9 homodimers were formed, both after tumor and inflammatory challenge. The cellular source for S100A9 homodimers were CD11b+GR1+ cells. S100A9 homodimers were also secreted into the extracellular space. Lastly, in the spleen from normal and tumor bearing animals cells expressing relatively higher levels of S100A9 compared to S100A8 could be observed by immunohistochemistry. Taken together, these data show that the biologically potent dimeric form of S100A9 is induced in vivo in situations of tumor burden or inflammatory challenge.


Subject(s)
Calgranulin B/metabolism , Protein Multimerization , Animals , Calgranulin A/metabolism , Extracellular Space/metabolism , Inflammation/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/metabolism , Spleen/metabolism
4.
BMC Cancer ; 16: 440, 2016 07 11.
Article in English | MEDLINE | ID: mdl-27400708

ABSTRACT

BACKGROUND: Previous work has demonstrated immunomodulatory, anti-tumor, anti-metastatic and anti-angiogenic effects of the small molecule quinoline-3-carboxamide tasquinimod in pre-clinical cancer models. To better understand the anti-tumor effects of tasquinimod in transplantable tumor models, we have evaluated the impact of the compound both on recruitment of myeloid cells to tumor tissue and on tumor-induced myeloid cell expansion as these cells are known to promote tumor development. METHODS: Mice bearing subcutaneous 4 T1 mammary carcinoma tumors were treated with tasquinimod in the drinking water. A BrdU-based flow cytometry assay was utilized to assess the impact of short-term tasquinimod treatment on myeloid cell recruitment to tumors. Additionally, long-term treatment was performed to study the anti-tumor effect of tasquinimod as well as its effects on splenic myeloid cells and their progenitors. Myeloid cell populations were also immune-depleted by in vivo antibody treatment. RESULTS: Short-term tasquinimod treatment did not influence the proliferation of splenic Ly6C(hi) and Ly6G(hi) cells, but instead reduced the influx of Ly6C(hi) cells to the tumor. Treatment with tasquinimod for various periods of time after tumor inoculation revealed that the anti-tumor effect of this compound mainly operated during the first few days of tumor growth. Similar to tasquinimod treatment, antibody-mediated depletion of Ly6C(hi) cells within that same time frame, caused reduced tumor growth, thereby confirming a significant role for these cells in tumor development. Additionally, long-term tasquinimod treatment reduced the splenomegaly and expansion of splenic myeloid cells during a later phase of tumor development. In this phase, tasquinimod normalized the tumor-induced alterations in myeloerythroid progenitor cells in the spleen but had only limited impact on the same populations in the bone marrow. CONCLUSIONS: Our results indicate that tasquinimod treatment reduces tumor growth by operating early after tumor inoculation and that this effect is at least partially caused by reduced recruitment of Ly6C(hi) cells to tumor tissue. Long-term treatment also reduces the number of splenic myeloid cells and myeloerythroid progenitors, but these effects did not influence established rapidly growing tumors.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinogenesis/drug effects , Cell Proliferation/drug effects , Myeloid Cells/drug effects , Quinolines/pharmacology , Quinolones/pharmacology , Administration, Oral , Angiogenesis Inhibitors/administration & dosage , Animals , Antigens, Ly/metabolism , CD11b Antigen/metabolism , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/metabolism , Myeloid Cells/pathology , Myeloid Progenitor Cells/drug effects , Myelopoiesis/drug effects , Quinolines/administration & dosage , Quinolones/administration & dosage , Spleen/cytology , Spleen/drug effects , Xenograft Model Antitumor Assays
5.
Int J Dermatol ; 54(7): e254-60, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26108267

ABSTRACT

BACKGROUND: Fumaric acid esters such as dimethyl fumarate (DMF) have proven to be effective in the treatment of psoriasis. OBJECTIVES: In view of the role of Th17 in the pathogenesis of psoriasis, the present study was conducted to investigate the effects of DMF on Th1, Th2, and Th17 responses in patients. METHODS: Peripheral blood mononuclear cells (PBMCs) were isolated from psoriasis patients and healthy individuals and were cultured in the presence or absence of phytohemagglutinin and DMF. The cell supernatants were removed to measure cytokine secretion, and the lymphocytes were used for real-time polymerase chain reaction to establish gene expression. RESULTS: An increase in gene expression of interferon-γ (IFN-γ), as a marker for Th1 activity, and interleukin-17 (IL-17), granulocyte macrophage colony-stimulating factor (GM-CSF) and IL-22 representing the Th17 subset in the PBMCs of patients in comparison with those of control subjects was observed. Culture of PBMCs from psoriasis patients and controls in the presence of DMF decreased IFN-γ and increased IL-4 gene expression in both groups. Treatment with DMF could significantly decrease IL-17, GM-CSF, and IL-22 mRNA levels in the PBMCs of patients. Decreased release of IFN-γ and GM-CSF cytokine secretion after DMF treatment was also observed in PBMC cultures of patients and controls. CONCLUSIONS: These data show the effectiveness of DMF in modulating Th17 cells in addition to Th1/Th2 cells and reflect one of the underlying mechanisms of action of DMF in psoriasis. These findings may also support the possible benefits of using fumarate in the treatment of other autoimmune diseases in the pathogeneses of which Th1 and Th17 cells play major roles.


Subject(s)
Cytokines/genetics , Dermatologic Agents/pharmacology , Dimethyl Fumarate/pharmacology , Gene Expression/drug effects , Psoriasis/immunology , T-Lymphocytes, Helper-Inducer/drug effects , Adult , Case-Control Studies , Cells, Cultured , Cytokines/metabolism , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Interleukins/genetics , Interleukins/metabolism , Male , Middle Aged , Psoriasis/drug therapy , RNA, Messenger/analysis , Th1 Cells/drug effects , Th17 Cells/drug effects , Th2 Cells/drug effects , Young Adult , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL
...