Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
Cells ; 12(11)2023 05 30.
Article in English | MEDLINE | ID: mdl-37296634

ABSTRACT

Previous studies from our lab demonstrated that the crosstalk between brain-metastasizing melanoma cells and microglia, the macrophage-like cells of the central nervous system, fuels progression to metastasis. In the present study, an in-depth investigation of melanoma-microglia interactions elucidated a pro-metastatic molecular mechanism that drives a vicious melanoma-brain-metastasis cycle. We employed RNA-Sequencing, HTG miRNA whole transcriptome assay, and reverse phase protein arrays (RPPA) to analyze the impact of melanoma-microglia interactions on sustainability and progression of four different human brain-metastasizing melanoma cell lines. Microglia cells exposed to melanoma-derived IL-6 exhibited upregulated levels of STAT3 phosphorylation and SOCS3 expression, which, in turn, promoted melanoma cell viability and metastatic potential. IL-6/STAT3 pathway inhibitors diminished the pro-metastatic functions of microglia and reduced melanoma progression. SOCS3 overexpression in microglia cells evoked microglial support in melanoma brain metastasis by increasing melanoma cell migration and proliferation. Different melanomas exhibited heterogeneity in their microglia-activating capacity as well as in their response to microglia-derived signals. In spite of this reality and based on the results of the present study, we concluded that the activation of the IL-6/STAT3/SOCS3 pathway in microglia is a major mechanism by which reciprocal melanoma-microglia signaling engineers the interacting microglia to reinforce the progression of melanoma brain metastasis. This mechanism may operate differently in different melanomas.


Subject(s)
Brain Neoplasms , Melanoma , Humans , Microglia/metabolism , Interleukin-6/metabolism , Signal Transduction , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Melanoma/pathology , Brain Neoplasms/metabolism , Brain/metabolism , STAT3 Transcription Factor/metabolism
2.
Mol Oncol ; 15(5): 1376-1390, 2021 05.
Article in English | MEDLINE | ID: mdl-33274599

ABSTRACT

Previous studies indicated that microglia cells upregulate the expression of aldolase C (ALDOC) in melanoma cells. The present study using brain-metastasizing variants from three human melanomas explores the functional role of ALDOC in the formation and maintenance of melanoma brain metastasis (MBM). ALDOC overexpression impacted differentially the malignant phenotype of these three variants. In the first variant, ALDOC overexpression promoted cell viability, adhesion to and transmigration through a layer of brain endothelial cells, and amplified brain micrometastasis formation. The cross-talk between this MBM variant and microglia cells promoted the proliferation and migration of the latter cells. In sharp contrast, ALDOC overexpression in the second brain-metastasizing melanoma variant reduced or did not affect the same malignancy features. In the third melanoma variant, ALDOC overexpression augmented certain characteristics of malignancy and reduced others. The analysis of biological functions and disease pathways in the ALDOC overexpressing variants clearly indicated that ALDOC induced the expression of tumor progression promoting genes in the first variant and antitumor progression properties in the second variant. Overall, these results accentuate the complex microenvironment interactions between microglia cells and MBM, and the functional impact of intertumor heterogeneity. Since intertumor heterogeneity imposes a challenge in the planning of cancer treatment, we propose to employ the functional response of tumors with an identical histology, to a particular drug or the molecular signature of this response, as a predictive indicator of response/nonresponse to this drug.


Subject(s)
Brain Neoplasms/secondary , Fructose-Bisphosphate Aldolase/physiology , Melanoma/pathology , Tumor Microenvironment/physiology , Animals , Biological Variation, Population/genetics , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Survival/genetics , Fructose-Bisphosphate Aldolase/genetics , HEK293 Cells , Humans , Male , Melanoma/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Phenotype , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tumor Microenvironment/genetics
3.
Cells ; 9(7)2020 07 13.
Article in English | MEDLINE | ID: mdl-32668704

ABSTRACT

Granulocyte-monocyte colony stimulating factor (GM-CSF) is used as an adjuvant in various clinical and preclinical studies with contradictory results. These were attributed to opposing effects of GM-CSF on the immune or myeloid systems of the treated patients or to lack of optimal dosing regimens. The results of the present study point to inter-tumor heterogeneity as a possible mechanism accounting for the contrasting responses to GM-CSF incorporating therapies. Employing xenograft models of human melanomas in nude mice developed in our lab, we detected differential functional responses of melanomas from different patients to GM-CSF both in vitro as well as in vivo. Whereas cells of one melanoma acquired pro metastatic features following exposure to GM-CSF, cells from another melanoma either did not respond or became less malignant. We propose that inter-melanoma heterogeneity as manifested by differential responses of melanoma cells (and perhaps also of other tumor) to GM-CSF may be developed into a predictive marker providing a tool to segregate melanoma patients who will benefit from GM-CSF therapy from those who will not.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Melanoma/pathology , Skin Neoplasms/pathology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Brain/pathology , Cell Line, Tumor , Cellular Microenvironment/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Interleukin-1alpha/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Solubility , Transendothelial and Transepithelial Migration/drug effects , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL