Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Language
Publication year range
1.
Braz J Med Biol Res ; 56: e12611, 2023.
Article in English | MEDLINE | ID: mdl-37792778

ABSTRACT

Islet transplantation represents a therapeutic option for type 1 diabetes (T1D). Long-term viability of transplanted islets requires improvement. Mesenchymal stromal cells (MSCs) have been proposed as adjuvants for islet transplantation facilitating grafting and functionality. Stem cell aggregation provides physiological interactions between cells and enhances the in situ concentration of modulators of inflammation and immunity. We established a hanging-drop culture of adult human skin fibroblast-like cells as spheroids, and skin spheroid-derived cells (SphCs) were characterized. We assessed the potential of SphCs in improving islet functionality by cotransplantation with a marginal mass of allogeneic islets in an experimental diabetic mouse model and characterized the secretome of SphCs by mass spectrometry-based proteomics. SphCs were characterized as multipotent progenitors and their coculture with anti-CD3 stimulated mouse splenocytes decreased CD4+ T cell proliferation with skewed cytokine secretion through an increase in the Th2/Th1 ratio profile. SphCs-conditioned media attenuated apoptosis of islets induced by cytokine challenge in vitro and importantly, intratesticular SphCs administration did not show tumorigenicity in immune-deficient mice. Moreover, SphCs improved glycemic control when cotransplanted with a marginal mass of allogeneic islets in a diabetic mouse model without pharmacological immunosuppression. SphCs' protein secretome differed from its paired skin fibroblast-like counterpart in containing 70% of up- and downregulated proteins and biological processes that overall positively influenced islets such as cytoprotection, cellular stress, metabolism, and survival. In summary, SphCs improved the performance of transplanted allogeneic islets in an experimental T1D model, without pharmacological immunosuppression. Future research is warranted to identify SphCs-secreted factors responsible for islets' endurance.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Hematopoietic Stem Cell Transplantation , Islets of Langerhans Transplantation , Islets of Langerhans , Humans , Mice , Animals , Adult , Islets of Langerhans/metabolism , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Experimental/metabolism , Cytokines/metabolism
2.
Braz. j. med. biol. res ; 56: e12611, 2023. graf
Article in English | LILACS-Express | LILACS | ID: biblio-1513883

ABSTRACT

Islet transplantation represents a therapeutic option for type 1 diabetes (T1D). Long-term viability of transplanted islets requires improvement. Mesenchymal stromal cells (MSCs) have been proposed as adjuvants for islet transplantation facilitating grafting and functionality. Stem cell aggregation provides physiological interactions between cells and enhances the in situ concentration of modulators of inflammation and immunity. We established a hanging-drop culture of adult human skin fibroblast-like cells as spheroids, and skin spheroid-derived cells (SphCs) were characterized. We assessed the potential of SphCs in improving islet functionality by cotransplantation with a marginal mass of allogeneic islets in an experimental diabetic mouse model and characterized the secretome of SphCs by mass spectrometry-based proteomics. SphCs were characterized as multipotent progenitors and their coculture with anti-CD3 stimulated mouse splenocytes decreased CD4+ T cell proliferation with skewed cytokine secretion through an increase in the Th2/Th1 ratio profile. SphCs-conditioned media attenuated apoptosis of islets induced by cytokine challenge in vitro and importantly, intratesticular SphCs administration did not show tumorigenicity in immune-deficient mice. Moreover, SphCs improved glycemic control when cotransplanted with a marginal mass of allogeneic islets in a diabetic mouse model without pharmacological immunosuppression. SphCs' protein secretome differed from its paired skin fibroblast-like counterpart in containing 70% of up- and downregulated proteins and biological processes that overall positively influenced islets such as cytoprotection, cellular stress, metabolism, and survival. In summary, SphCs improved the performance of transplanted allogeneic islets in an experimental T1D model, without pharmacological immunosuppression. Future research is warranted to identify SphCs-secreted factors responsible for islets' endurance.

3.
Curr Pharm Biotechnol ; 12(11): 1881-90, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21470131

ABSTRACT

During the past few years, Epithelial-Mesenchymal Transition (EMT) has emerged as one of the most hot spots in clinical research. Its existence in human tumors can form the basis for explaining characteristics of cancer progression and metastasis, as well as certain cases of drug resistance and relapses after treatment. These cellular responses are tightly regulated by intracellular signaling pathways evoked by humoral factors that include growth factors, chemokines and cytokines. Indeed, several gene regulatory programs known to promote EMT during development have recently been discovered to play key roles in cancer progression. A deeper understanding of the cellular and molecular basis of these different programs should aid in both the development of better diagnosis methods, as well as of specific treatments for invasive cancer. In this review we set out to summarize recent novel insights into the molecular players underlying EMT and its relation with cancer progression and metastasis.


Subject(s)
Disease Progression , Epithelial-Mesenchymal Transition , Neoplasms/pathology , Animals , Epithelial-Mesenchymal Transition/immunology , Humans , Matrix Metalloproteinases/metabolism , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/immunology , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Transforming Growth Factor beta/immunology
4.
Diabetologia ; 54(6): 1388-97, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21394492

ABSTRACT

AIMS/HYPOTHESIS: Transplantation of pancreatic islets constitutes a promising alternative treatment for type 1 diabetes. However, it is limited by the shortage of organ donors. Previous results from our laboratory have demonstrated beneficial effects of recombinant human prolactin (rhPRL) treatment on beta cell cultures. We therefore investigated the role of rhPRL action in human beta cell survival, focusing on the molecular mechanisms involved in this process. METHODS: Human pancreatic islets were isolated using an automated method. Islet cultures were pre-treated in the absence or presence of rhPRL and then subjected to serum starvation or cytokine treatment. Beta cells were labelled with Newport green and apoptosis was evaluated using flow cytometry analysis. Levels of BCL2 gene family members were studied by quantitative RT-PCR and western blot. Caspase-8, -9 and -3 activity, as well as nitric oxide production, were evaluated by fluorimetric assays. RESULTS: The proportion of apoptotic beta cells was significantly lowered in the presence of rhPRL under both cell death-induced conditions. We also demonstrated that cytoprotection may involve an increase of BCL2/BAX ratio, as well as inhibition of caspase-8, -9 and -3. CONCLUSIONS/INTERPRETATION: Our study provides relevant evidence for a protective effect of lactogens on human beta cell apoptosis. The results also suggest that the improvement of cell survival may involve, at least in part, inhibition of cell death pathways controlled by the BCL2 gene family members. These findings are highly relevant for improvement of the islet isolation procedure and for clinical islet transplantation.


Subject(s)
Apoptosis/drug effects , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Prolactin/pharmacology , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Adult , Apoptosis/physiology , Caspase Inhibitors , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cytokines/metabolism , Diabetes Mellitus, Type 1/surgery , Humans , Insulin-Secreting Cells/metabolism , Islets of Langerhans Transplantation/methods , Middle Aged , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/physiology , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...