Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
2.
Perit Dial Int ; 44(1): 56-65, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37592841

ABSTRACT

BACKGROUND: Patients with end-stage kidney disease (ESKD) require dialysis or transplantation for their survival. There are few experimental animal models mimicking the human situation in which the animals are dependent on dialysis for their survival. We developed a peritoneal dialysis (PD) system for rats to enable long-term treatment under controlled conditions. METHOD: Rats were chemically nephrectomised using orellanine to render them uremic. Two studies were performed, the first with highly uremic rats on PD for 5 days, and the other with moderately uremic rats on PD for 21 days. Blood and dialysate samples were collected repeatedly from the first study and solute concentrations analysed. Based on these values, dialysis parameters were calculated together with generation rates allowing for kinetic modelling of the effects of PD. In the second study, the general conditions of the rats were evaluated during a longer dialysis period. RESULTS: For rats with estimated glomerular filtration rate (GFR) 5-10% of normal (moderately uremic rats), five daily PD cycles kept the rats in good condition for 3 weeks. For highly uremic rats (GFR below 3% of normal), more extensive dialysis is needed to maintain homeostasis and our simulations show that a six daily and four nightly PD cycles should be needed to keep the rats in good condition. CONCLUSION: In conclusion, the PD system described in this study can be used for long-term studies of PD on uremic dialysis-dependent rats mimicking the human setting. To maintain whole body homeostasis of highly uremic rats, intense PD is needed during both day and night.


Subject(s)
Kidney Failure, Chronic , Peritoneal Dialysis , Humans , Rats , Animals , Kidney Failure, Chronic/therapy , Renal Dialysis , Disease Models, Animal
3.
Front Immunol ; 14: 1256404, 2023.
Article in English | MEDLINE | ID: mdl-37908353

ABSTRACT

Introduction: Natural killer (NK) cells are a key component of the innate immune system, involved in defending the host against virus-infected cells and tumor immunosurveillance. Under in vitro culture conditions, IL-12/15/18 can induce a memory-like phenotype in NK cells. These cytokine-induced memory-like (CIML) NK cells possess desirable characteristics for immunotherapies, including a longer lifespan and increased cytotoxicity. Methods: In this study, NK cells were isolated from peripheral blood of healthy donors and stimulated with IL-12/15/18 to induce a memory-like phenotype or with IL-15 alone as a control. After seven days of culture, multiparametric flow cytometry analysis was performed to evaluate the phenotypic and functional profiles of CIML and control NK cells. Results: Our results showed a significantly higher expression of CD25, CD69, NKG2D, NKp30, NKp44, NKp46, TACTILE, and Granzyme B in CIML NK cells compared to control NK cells. In contrast, KIR2D expression was significantly lower in CIML NK cells than in control NK cells. Moreover, functional experiments demonstrated that CIML NK cells displayed enhanced degranulation capacity and increased intracellular IFN-γ production against the target cell line K562. Interestingly, the degranulation capacity of CIML NK cells was positively correlated with the expression of the activating receptors NKp46 and NKp30, as well as with the inhibitory receptor TACTILE. Discussion: In conclusion, this study provides a deep phenotypic characterization of in vitro-expanded CIML NK cells. Moreover, the correlations found between NK cell receptors and degranulation capacity of CIML NK cells allowed the identification of several biomarkers that could be useful in clinical settings.


Subject(s)
Cytokines , Killer Cells, Natural , Cytokines/metabolism , Receptors, Natural Killer Cell/metabolism , Flow Cytometry , Interleukin-12/metabolism
4.
Cancers (Basel) ; 12(8)2020 Aug 05.
Article in English | MEDLINE | ID: mdl-32764229

ABSTRACT

Background: Acute myeloid leukemia (AML) remains a major clinical challenge due to poor overall survival, which is even more dramatic in elderly patients. TIGIT, an inhibitory receptor that interacts with CD155 and CD112 molecules, is considered as a checkpoint in T and NK cell activation. This receptor shares ligands with the co-stimulatory receptor DNAM-1 and with TACTILE. The aim of this work was to analyze the expression of DNAM-1, TIGIT and TACTILE in NK cells and T cell subsets in AML patients. Methods: We have studied 36 patients at the time of diagnosis of AML and 20 healthy volunteers. The expression of DNAM-1, TIGIT and TACTILE in NK cells and T cells, according to the expression of CD3 and CD56, was performed by flow cytometry. Results: NK cells, CD56- T cells and CD56+ T (NKT-like) cells from AML patients presented a reduced expression of DNAM-1 compared with healthy volunteers. An increased expression of TIGIT was observed in mainstream CD56- T cells. No differences were observed in the expression of TACTILE. Simplified presentation of incredibly complex evaluations (SPICE) analysis of the co-expression of DNAM-1, TIGIT and TACTILE showed an increase in NK and T cells lacking DNAM-1 and co-expressing TIGIT and TACTILE. Low percentages of DNAM-1-TIGIT+TACTILE+ NK cells and DNAM-1- TIGIT+TACTILE+ CD56- T cells were associated with a better survival of AML patients. Conclusions: The expression of DNAM-1 is reduced in NK cells and in CD4+ and CD8+ T cells from AML patients compared with those from healthy volunteers. An increased percentage of NK and T cells lacking DNAM-1 and co-expressing TIGIT and TACTILE is associated with patient survival, supporting the role of TIGIT as a novel candidate for checkpoint blockade.

5.
Cancer Immunol Immunother ; 69(5): 879-899, 2020 May.
Article in English | MEDLINE | ID: mdl-32130453

ABSTRACT

A better understanding of the complex interactions between the immune system and tumour cells from different origins has opened the possibility to design novel procedures of antitumoral immunotherapy. One of these novel approaches is based on the use of autologous or allogeneic natural killer (NK) cells to treat cancer. In the last decade, different strategies to activate NK cells and their use in adoptive NK cell-based therapy have been established. Although NK cells are often considered as a uniform cell population, several phenotypic and functionally distinct NK cells subsets exist in healthy individuals, that are differentially affected by ageing or by apparently innocuous viruses such as cytomegalovirus (CMV). In addition, further alterations in the expression of activating and inhibitory receptors are found in NK cells from cancer patients, likely because of their interaction with tumour cells. Thus, NK cells represent a promising strategy for adoptive immunotherapy of cancer already tested in phase 1/2 clinical trials. However, the existence of NK cell subpopulations expressing different patterns of activating and inhibitory receptors and different functional capacities, that can be found to be altered not only in cancer patients but also in healthy individuals stratified by age or CMV infection, makes necessary a personalized definition of the procedures used in the selection, expansion, and activation of the relevant NK cell subsets to be successfully used in NK cell-based immunotherapy.


Subject(s)
Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Neoplasms/immunology , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Humans , Immunotherapy, Adoptive/trends , Killer Cells, Natural/transplantation , Lymphocyte Subsets/transplantation , Neoplasms/therapy , Transplantation, Autologous/methods , Transplantation, Autologous/trends , Transplantation, Homologous/methods , Transplantation, Homologous/trends
6.
J Lipid Res ; 61(2): 178-191, 2020 02.
Article in English | MEDLINE | ID: mdl-31857389

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) are emerging as leading causes of liver disease worldwide and have been recognized as one of the major unmet medical needs of the 21st century. Our recent translational studies in mouse models, human cell lines, and well-characterized patient cohorts have identified serine/threonine kinase (STK)25 as a protein that coats intrahepatocellular lipid droplets (LDs) and critically regulates liver lipid homeostasis and progression of NAFLD/NASH. Here, we studied the mechanism-of-action of STK25 in steatotic liver by relative quantification of the hepatic LD-associated phosphoproteome from high-fat diet-fed Stk25 knockout mice compared with their wild-type littermates. We observed a total of 131 proteins and 60 phosphoproteins that were differentially represented in STK25-deficient livers. Most notably, a number of proteins involved in peroxisomal function, ubiquitination-mediated proteolysis, and antioxidant defense were coordinately regulated in Stk25-/- versus wild-type livers. We confirmed attenuated peroxisomal biogenesis and protection against oxidative and ER stress in STK25-deficient human liver cells, demonstrating the hepatocyte-autonomous manner of STK25's action. In summary, our results suggest that regulation of peroxisomal function and metabolic stress response may be important molecular mechanisms by which STK25 controls the development and progression of NAFLD/NASH.


Subject(s)
Fatty Liver/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lipid Droplets/enzymology , Peroxisomes/metabolism , Protein Serine-Threonine Kinases/metabolism , Stress, Physiological , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/deficiency
7.
Cancers (Basel) ; 11(6)2019 Jun 23.
Article in English | MEDLINE | ID: mdl-31234588

ABSTRACT

Natural killer (NK) cells are lymphocytes of the innate immune response characterized by their role in the destruction of tumor cells. Activation of NK cells depend on a fine balance between activating and inhibitory signals mediated by different receptors. In recent years, a family of paired receptors that interact with ligands of the Nectin/Nectin-like (Necl) family has attracted great interest. Two of these ligands, Necl-5 (usually termed CD155 or PVR) and Nectin-2 (CD112), frequently expressed on different types of tumor cells, are recognized by a group of receptors expressed on T and NK cells that exert opposite functions after interacting with their ligands. These receptors include DNAM-1 (CD226), TIGIT, TACTILE (CD96) and the recently described PVRIG. Whereas activation through DNAM-1 after recognition of CD155 or CD112 enhances NK cell-mediated cytotoxicity against a wide range of tumor cells, TIGIT recognition of these ligands exerts an inhibitory effect on NK cells by diminishing IFN-γ production, as well as NK cell-mediated cytotoxicity. PVRIG has also been identified as an inhibitory receptor that recognizes CD112 but not CD155. However, little is known about the role of TACTILE as modulator of immune responses in humans. TACTILE control of tumor growth and metastases has been reported in murine models, and it has been suggested that it negatively regulates the anti-tumor functions mediated by DNAM-1. In NK cells from patients with solid cancer and leukemia, it has been observed a decreased expression of DNAM-1 that may shift the balance in favor to the inhibitory receptors TIGIT or PVRIG, further contributing to the diminished NK cell-mediated cytotoxic capacity observed in these patients. Analysis of DNAM-1, TIGIT, TACTILE and PVRIG on human NK cells from solid cancer or leukemia patients will clarify the role of these receptors in cancer surveillance. Overall, it can be speculated that in cancer patients the TIGIT/PVRIG pathways are upregulated and represent novel targets for checkpoint blockade immunotherapy.

8.
Cancer Immunol Immunother ; 68(5): 861-870, 2019 May.
Article in English | MEDLINE | ID: mdl-30953117

ABSTRACT

The incidence of some types of tumours has increased progressively in recent years and is expected to continue growing in the coming years due in part to the aging of the population. The design of new therapies based on natural killer (NK) cells opens new possibilities especially for the treatment of elderly patients who are particularly susceptible to the toxicity of conventional chemotherapy treatments. In recent years, the potential use of NK cells in cancer immunotherapy has been of great interest thanks to advances in the study of NK cell biology. The identification of key points (checkpoints) in the activation of NK cells that can be regulated by monoclonal antibodies has allowed the design of new therapeutic strategies based on NK cells. However, there are still limitations for its use and the first clinical trials blocking KIR inhibitory receptors have shown little efficacy by inhibiting the maturation of NK cells. Blockade of other inhibitory receptors such as TIGIT, TIM3, LAG3 and PD1 may represent novel strategies to increase NK function in cancer patients. Altogether, the identification of NK cell and tumour cell markers of resistance or susceptibility to the action of NK cells will contribute to identifying those patients that will most likely benefit from NK cell-based immunotherapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Costimulatory and Inhibitory T-Cell Receptors/immunology , Immunotherapy/methods , Killer Cells, Natural/immunology , Neoplasms/therapy , Animals , Humans , Immunomodulation , Neoplasms/immunology
9.
Cell Mol Gastroenterol Hepatol ; 7(3): 597-618, 2019.
Article in English | MEDLINE | ID: mdl-30576769

ABSTRACT

BACKGROUND & AIMS: Nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) are emerging as leading causes of liver disease worldwide. Currently, no specific pharmacologic therapy is available for NAFLD/NASH, which has been recognized as one of the major unmet medical needs of the 21st century. Our recent studies in genetic mouse models, human cell lines, and well-characterized patient cohorts have identified serine/threonine protein kinase (STK)25 as a critical regulator of hepatic lipid partitioning and NAFLD/NASH. Here, we studied the metabolic benefit of liver-specific STK25 inhibitors on NAFLD development and progression in a mouse model of diet-induced obesity. METHODS: We developed a hepatocyte-specific triantennary N-acetylgalactosamine (GalNAc)-conjugated antisense oligonucleotide (ASO) targeting Stk25 and evaluated its effect on NAFLD features in mice after chronic exposure to dietary lipids. RESULTS: We found that systemic administration of hepatocyte-targeting GalNAc-Stk25 ASO in obese mice effectively ameliorated steatosis, inflammatory infiltration, hepatic stellate cell activation, nutritional fibrosis, and hepatocellular damage in the liver compared with mice treated with GalNAc-conjugated nontargeting ASO, without any systemic toxicity or local tolerability concerns. We also observed protection against high-fat-diet-induced hepatic oxidative stress and improved mitochondrial function with Stk25 ASO treatment in mice. Moreover, GalNAc-Stk25 ASO suppressed lipogenic gene expression and acetyl-CoA carboxylase protein abundance in the liver, providing insight into the molecular mechanisms underlying repression of hepatic steatosis. CONCLUSIONS: This study provides in vivo nonclinical proof-of-principle for the metabolic benefit of liver-specific inhibition of STK25 in the context of obesity and warrants future investigations to address the therapeutic potential of GalNAc-Stk25 ASO in the prevention and treatment of NAFLD.


Subject(s)
Hepatocytes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Non-alcoholic Fatty Liver Disease/therapy , Oligonucleotides, Antisense/pharmacology , Protein Serine-Threonine Kinases/metabolism , Acetyl-CoA Carboxylase/metabolism , Acetylglucosamine/metabolism , Animals , Apoptosis/drug effects , Body Weight/drug effects , Diet, High-Fat , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Lipogenesis/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Obese , Mitochondria/drug effects , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/blood , Organ Size/drug effects , Oxidative Stress/drug effects , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Arterioscler Thromb Vasc Biol ; 38(8): 1723-1737, 2018 08.
Article in English | MEDLINE | ID: mdl-29930001

ABSTRACT

Objective- Recent cohort studies have shown that nonalcoholic fatty liver disease (NAFLD), and especially nonalcoholic steatohepatitis (NASH), associate with atherosclerosis and cardiovascular disease, independently of conventional cardiometabolic risk factors. However, the mechanisms underlying the pathophysiological link between NAFLD/NASH and cardiovascular disease still remain unclear. Our previous studies have identified STK25 (serine/threonine protein kinase 25) as a critical determinant in ectopic lipid storage, meta-inflammation, and progression of NAFLD/NASH. The aim of this study was to assess whether STK25 is also one of the mediators in the complex molecular network controlling the cardiovascular disease risk. Approach and Results- Atherosclerosis was induced in Stk25 knockout and transgenic mice, and their wild-type littermates, by gene transfer of gain-of-function mutant of PCSK9 (proprotein convertase subtilisin/kexin type 9), which induces the downregulation of hepatic LDLR (low-density lipoprotein receptor), combined with an atherogenic western-type diet. We found that Stk25-/- mice displayed reduced atherosclerosis lesion area as well as decreased lipid accumulation, macrophage infiltration, collagen formation, and oxidative stress in aortic lesions compared with wild-type littermates, independently from alterations in dyslipidemia. Reciprocally, Stk25 transgenic mice presented aggravated plaque formation and maturation compared with wild-type littermates despite similar levels of fasting plasma cholesterol. We also found that STK25 protein was expressed in all layers of the aorta, suggesting a possible direct role in cardiovascular disease. Conclusions- This study provides the first evidence that STK25 plays a critical role in regulation of cardiovascular disease risk and suggests that pharmacological inhibition of STK25 function may provide new possibilities for prevention/treatment of atherosclerosis.


Subject(s)
Aorta/enzymology , Aortic Diseases/enzymology , Atherosclerosis/enzymology , Hypercholesterolemia/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Plaque, Atherosclerotic , Protein Serine-Threonine Kinases/metabolism , Animals , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Diet, High-Fat , Disease Models, Animal , Disease Progression , Gene Transfer Techniques , Hypercholesterolemia/genetics , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Liver/enzymology , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Mutation , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Proprotein Convertase 9/genetics , Proprotein Convertase 9/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction
11.
J Endocrinol ; 238(3): 187-202, 2018 09.
Article in English | MEDLINE | ID: mdl-29794231

ABSTRACT

Whole-body energy homeostasis at over-nutrition critically depends on how well adipose tissue remodels in response to excess calories. We recently identified serine/threonine protein kinase (STK)25 as a critical regulator of ectopic lipid storage in non-adipose tissue and systemic insulin resistance in the context of nutritional stress. Here, we investigated the role of STK25 in regulation of adipose tissue dysfunction in mice challenged with a high-fat diet. We found that overexpression of STK25 in high-fat-fed mice resulted in impaired mitochondrial function and aggravated hypertrophy, inflammatory infiltration and fibrosis in adipose depots. Reciprocally, Stk25-knockout mice displayed improved mitochondrial function and were protected against diet-induced excessive fat storage, meta-inflammation and fibrosis in brown and white adipose tissues. Furthermore, in rodent HIB-1B cell line, STK25 depletion resulted in enhanced mitochondrial activity and consequently, reduced lipid droplet size, demonstrating an autonomous action for STK25 within adipocytes. In summary, we provide the first evidence for a key function of STK25 in controlling the metabolic balance of lipid utilization vs lipid storage in brown and white adipose depots, suggesting that repression of STK25 activity offers a potential strategy for establishing healthier adipose tissue in the context of chronic exposure to dietary lipids.


Subject(s)
Adipose Tissue/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Lipid Metabolism/genetics , Oxidative Stress/genetics , Protein Serine-Threonine Kinases/physiology , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Cells, Cultured , Diet, High-Fat , Intracellular Signaling Peptides and Proteins/genetics , Lipid Droplets/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics
12.
Hepatol Commun ; 2(1): 69-83, 2018 01.
Article in English | MEDLINE | ID: mdl-29404514

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) contributes to the pathogenesis of type 2 diabetes and cardiovascular disease, and patients with nonalcoholic steatohepatitis (NASH) are also at risk of developing cirrhosis, liver failure, and hepatocellular carcinoma. To date, no specific therapy exists for NAFLD/NASH, which has been recognized as one of the major unmet medical needs of the twenty-first century. We recently identified serine/threonine protein kinase (STK)25 as a critical regulator of energy homeostasis and NAFLD progression. Here, we investigated the effect of antisense oligonucleotides (ASOs) targeting Stk25 on the metabolic and molecular phenotype of mice after chronic exposure to dietary lipids. We found that Stk25 ASOs efficiently reversed high-fat diet-induced systemic hyperglycemia and hyperinsulinemia, improved whole-body glucose tolerance and insulin sensitivity, and ameliorated liver steatosis, inflammatory infiltration, apoptosis, hepatic stellate cell activation, and nutritional fibrosis in obese mice. Moreover, Stk25 ASOs suppressed the abundance of liver acetyl-coenzyme A carboxylase (ACC) protein, a key regulator of both lipid oxidation and synthesis, revealing the likely mechanism underlying repression of hepatic fat accumulation by ASO treatment. We also found that STK25 protein levels correlate significantly and positively with NASH development in human liver biopsies, and several common nonlinked single-nucleotide polymorphisms in the human STK25 gene are associated with altered liver fat, supporting a critical role of STK25 in the pathogenesis of NAFLD in humans. Conclusion: Preclinical validation for the metabolic benefit of pharmacologically inhibiting STK25 in the context of obesity is provided. Therapeutic intervention aimed at reducing STK25 function may provide a new strategy for the treatment of patients with NAFLD, type 2 diabetes, and related complex metabolic diseases. (Hepatology Communications 2018;2:69-83).

13.
Front Immunol ; 8: 931, 2017.
Article in English | MEDLINE | ID: mdl-28824651

ABSTRACT

Despite recent progress in the therapeutic approach of malignant hemopathies, their prognoses remain frequently poor. Immunotherapy could open a new window of great interest in this setting. Natural killer (NK) cells constitute an important area of research for hematologic malignancies, because this subpopulation is able to kill target cells spontaneously without previous sensitization, representing a novel tool in the treatment of them. Abnormal NK cytolytic function is observed in several hematological malignancies, including acute myeloid leukemia (AML) and myelodysplastic syndromes. Several mechanisms are involved in this abnormal function, such as decreased expression of activating receptors, increased expression of inhibitory receptors or defective expression of NK cell ligands on target cells. New immunotherapies are focused in identifying factors that could increase the expression of these activating receptors, to counteract inhibitory receptors expression, and therefore, to improve the NK cell cytotoxic capacities against tumor cells. In this work, we analyze the effect of interleukin (IL)-15 on the expression of NK cell-activating receptors that play a crucial role in the lysis of blasts from AML patients. Our results showed that IL-15 increased the surface expression of NKp30 on NK cells from healthy donors and AML patients with the consequent improvement of NK cell cytotoxicity. Besides, the upregulation of NKp30 induced by IL-15 is associated with an improvement of NK-mediated myeloid dendritic cells (DCs) maturation. NK cells cultured with IL-15 showed an upregulation of NKp30, which is associated with an increase anti-tumor activity and with an improved maturation of immature DCs. In our in vitro model, IL-15 exerted a great activating stimulus that could be used as novel immunotherapy in AML patients.

14.
J Endocrinol ; 234(1): 15-27, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28442507

ABSTRACT

Characterising the molecular networks that negatively regulate pancreatic ß-cell function is essential for understanding the underlying pathogenesis and developing new treatment strategies for type 2 diabetes. We recently identified serine/threonine protein kinase 25 (STK25) as a critical regulator of ectopic fat storage, meta-inflammation, and fibrosis in liver and skeletal muscle. Here, we assessed the role of STK25 in control of progression of non-alcoholic fatty pancreas disease in the context of chronic exposure to dietary lipids in mice. We found that overexpression of STK25 in high-fat-fed transgenic mice aggravated diet-induced lipid storage in the pancreas compared with that of wild-type controls, which was accompanied by exacerbated pancreatic inflammatory cell infiltration, stellate cell activation, fibrosis and apoptosis. Pancreas of Stk25 transgenic mice also displayed a marked decrease in islet ß/α-cell ratio and alteration in the islet architecture with an increased presence of α-cells within the islet core, whereas islet size remained similar between genotypes. After a continued challenge with a high-fat diet, lower levels of fasting plasma insulin and C-peptide, and higher levels of plasma leptin, were detected in Stk25 transgenic vs wild-type mice. Furthermore, the glucose-stimulated insulin secretion was impaired in high-fat-fed Stk25 transgenic mice during glucose tolerance test, in spite of higher net change in blood glucose concentrations compared with wild-type controls, suggesting islet ß-cell dysfunction. In summary, this study unravels a role for STK25 in determining the susceptibility to diet-induced non-alcoholic fatty pancreas disease in mice in connection to obesity. Our findings highlight STK25 as a potential drug target for metabolic disease.


Subject(s)
Adipose Tissue/metabolism , Diet, High-Fat , Intracellular Signaling Peptides and Proteins/physiology , Pancreatic Diseases/physiopathology , Protein Serine-Threonine Kinases/physiology , Animals , Blood Glucose/analysis , C-Peptide/blood , Gene Expression , Glucose Tolerance Test , Inflammation/pathology , Insulin/blood , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology , Intracellular Signaling Peptides and Proteins/genetics , Islets of Langerhans/pathology , Islets of Langerhans/physiopathology , Leptin/blood , Lipid Metabolism , Mice , Mice, Transgenic , Obesity/metabolism , Pancreatic Diseases/pathology , Protein Serine-Threonine Kinases/genetics
15.
Diabetologia ; 60(3): 553-567, 2017 03.
Article in English | MEDLINE | ID: mdl-27981357

ABSTRACT

AIMS/HYPOTHESIS: Understanding the molecular networks controlling ectopic lipid deposition and insulin responsiveness in skeletal muscle is essential for developing new strategies to treat type 2 diabetes. We recently identified serine/threonine protein kinase 25 (STK25) as a critical regulator of liver steatosis, hepatic lipid metabolism and whole body glucose and insulin homeostasis. Here, we assessed the role of STK25 in control of ectopic fat storage and insulin responsiveness in skeletal muscle. METHODS: Skeletal muscle morphology was studied by histological examination, exercise performance and insulin sensitivity were assessed by treadmill running and euglycaemic-hyperinsulinaemic clamp, respectively, and muscle lipid metabolism was analysed by ex vivo assays in Stk25 transgenic and wild-type mice fed a high-fat diet. Lipid accumulation and mitochondrial function were also studied in rodent myoblasts overexpressing STK25. Global quantitative phosphoproteomics was performed in skeletal muscle of Stk25 transgenic and wild-type mice fed a high-fat diet to identify potential downstream mediators of STK25 action. RESULTS: We found that overexpression of STK25 in transgenic mice fed a high-fat diet increases intramyocellular lipid accumulation, impairs skeletal muscle mitochondrial function and sarcomeric ultrastructure, and induces perimysial and endomysial fibrosis, thereby reducing endurance exercise capacity and muscle insulin sensitivity. Furthermore, we observed enhanced lipid accumulation and impaired mitochondrial function in rodent myoblasts overexpressing STK25, demonstrating an autonomous action for STK25 within cells. Global phosphoproteomic analysis revealed alterations in the total abundance and phosphorylation status of different target proteins located predominantly to mitochondria and sarcomeric contractile elements in Stk25 transgenic vs wild-type muscle, respectively, providing a possible molecular mechanism for the observed phenotype. CONCLUSIONS/INTERPRETATION: STK25 emerges as a new regulator of the complex interplay between lipid storage, mitochondrial energetics and insulin action in skeletal muscle, highlighting the potential of STK25 antagonists for type 2 diabetes treatment.


Subject(s)
Insulin Resistance/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Lipid Metabolism/physiology , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Blotting, Western , Chromatography, Liquid , Diet, High-Fat , Insulin Resistance/genetics , Intracellular Signaling Peptides and Proteins/genetics , Lipid Metabolism/genetics , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/pathology , Protein Serine-Threonine Kinases/genetics , Proteomics , Rats , Real-Time Polymerase Chain Reaction
16.
Cancer Immunol Immunother ; 66(2): 233-245, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27530271

ABSTRACT

Cancer is primarily considered a disease of old age. Immunosenescence refers to the age-associated changes in the immune system, and its contribution to the increased risk of cancer in old individuals has been discussed for many years. Natural killer (NK) cells are cytotoxic innate immune cells specialized in defence against tumour and virus-infected cells. NK cell cytotoxicity is the result of a fine balance between activating and inhibitory receptors. Several activating receptors have been identified that recognize different ligands frequently found over-expressed on tumour cells or virus-infected cells. The most important NK cell inhibitory receptors interact with major histocompatibility complex class I molecules expressed on almost all nucleated cells preventing NK cell-mediated lysis of healthy cells. NK cell immunosenescence is characterized by a redistribution of NK cell subsets, a diminished expression of several activating receptors and lower per-cell cytotoxicity. Altered expression of activating receptors has also been described in young and elderly cancer patients probably due to chronic exposure to ligands on tumour cells. Thus, the effect of both age and cancer may act synergistically to diminish NK cell-mediated tumour immunosurveillance. Different strategies harnessing the power of NK cells to target tumour cells have been designed including adoptive therapy with autologous or allogeneic expanded NK cells. In addition, checkpoint blockade of inhibitory receptors and the use of agonist antibodies to stimulate activating receptors are emerging areas of research. In this context, the effect of immunosenescence should be considered to improve the efficiency of cancer immunotherapy.


Subject(s)
Immunosenescence/immunology , Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Neoplasms/immunology , Neoplasms/therapy , Animals , Humans , Ligands
17.
FASEB J ; 30(10): 3628-3643, 2016 10.
Article in English | MEDLINE | ID: mdl-27421788

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, and 10-20% of patients with NAFLD progress to nonalcoholic steatohepatitis (NASH) with a high risk of cirrhosis, liver failure, and hepatocellular carcinoma. Despite its high medical importance, the molecular mechanisms controlling progression from simple liver steatosis to NASH remain elusive. We recently identified serine/threonine protein kinase (STK)25 as a critical regulator of ectopic lipid deposition, systemic glucose, and insulin homeostasis. To elucidate the role of STK25 in the development of NASH, we challenged Stk25-knockout and transgenic mice with a methionine and choline-deficient (MCD) diet. We show that Stk25-/- mice are protected against MCD-diet-induced NASH, as evidenced by repressed liver steatosis, oxidative damage, inflammation, and fibrosis, whereas Stk25 transgenic mice developed a more severe NASH phenotype, compared with corresponding wild-type littermates. Consistently, NASH features were suppressed in STK25-deficient human hepatocytes cultured in MCD medium, and reciprocally enhanced in STK25-overexpressing cells. We also found a significant positive correlation in human liver biopsies between STK25 expression and NASH development. The study provides evidence for multiple roles of STK25 in NASH pathogenesis and future investigations to address the potential therapeutic relevance of pharmacological STK25 inhibitors in prevention and treatment of NASH are warranted.-Amrutkar, M., Chursa, U., Kern, M., Nuñez-Durán, E., Ståhlman, M., Sütt, S., Borén, J., Johansson, B. R., Marschall, H.-U., Blüher, M., Mahlapuu, M. STK25 is a critical determinant in nonalcoholic steatohepatitis.


Subject(s)
Choline Deficiency/metabolism , Hepatocytes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Choline Deficiency/complications , Disease Models, Animal , Lipid Metabolism/genetics , Mice, Transgenic , Triglycerides/metabolism
18.
Diabetologia ; 59(2): 341-53, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26553096

ABSTRACT

AIMS/HYPOTHESIS: Type 2 diabetes is closely associated with pathological lipid accumulation in the liver, which is suggested to actively contribute to the development of insulin resistance. We recently identified serine/threonine protein kinase 25 (STK25) as a regulator of liver steatosis, whole-body glucose tolerance and insulin sensitivity in a mouse model system. The aim of this study was to assess the role of STK25 in the control of lipid metabolism in human liver. METHODS: Intracellular fat deposition, lipid metabolism and insulin sensitivity were studied in immortalised human hepatocytes (IHHs) and HepG2 hepatocellular carcinoma cells in which STK25 was overexpressed or knocked down by small interfering RNA. The association between STK25 mRNA expression in human liver biopsies and hepatic fat content was analysed. RESULTS: Overexpression of STK25 in IHH and HepG2 cells enhanced lipid deposition by suppressing ß-oxidation and triacylglycerol (TAG) secretion, while increasing lipid synthesis. Conversely, knockdown of STK25 attenuated lipid accumulation by stimulating ß-oxidation and TAG secretion, while inhibiting lipid synthesis. Furthermore, TAG hydrolase activity was repressed in hepatocytes overexpressing STK25 and reciprocally increased in cells with STK25 knockdown. Insulin sensitivity was reduced in STK25-overexpressing cells and enhanced in STK25-deficient hepatocytes. We also found a statistically significant positive correlation between STK25 mRNA expression in human liver biopsies and hepatic fat content. CONCLUSIONS/INTERPRETATION: Our data suggest that STK25 regulates lipid partitioning in human liver cells by controlling TAG synthesis as well as lipolytic activity and thereby NEFA release from lipid droplets for ß-oxidation and TAG secretion. Our findings highlight STK25 as a potential drug target for the prevention and treatment of type 2 diabetes.


Subject(s)
Hepatocytes/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Lipid Metabolism/genetics , Liver/metabolism , Protein Serine-Threonine Kinases/physiology , Adiposity , Animals , Biological Transport/genetics , Cells, Cultured , Female , Hep G2 Cells , Humans , Lipid Mobilization/genetics , Male , Mice , Mice, Knockout , Triglycerides/metabolism
19.
Cancer Immunol Immunother ; 65(4): 453-63, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26059279

ABSTRACT

Several age-associated changes in natural killer (NK) cell phenotype have been reported that contribute to the defective NK cell response observed in elderly patients. A remodelling of the NK cell compartment occurs in the elderly with a reduction in the output of immature CD56(bright) cells and an accumulation of highly differentiated CD56(dim) NK cells. Acute myeloid leukaemia (AML) is generally a disease of older adults. NK cells in AML patients show diminished expression of several activating receptors that contribute to impaired NK cell function and, in consequence, to AML blast escape from NK cell immunosurveillance. In AML patients, phenotypic changes in NK cells have been correlated with disease progression and survival. NK cell-based immunotherapy has emerged as a possibility for the treatment of AML patients. The understanding of age-associated alterations in NK cells is therefore necessary to define adequate therapeutic strategies in older AML patients.


Subject(s)
Aging/immunology , Immunosenescence , Killer Cells, Natural/immunology , Leukemia, Myeloid/immunology , Acute Disease , Aged , CD56 Antigen/immunology , CD56 Antigen/metabolism , Humans , Immunotherapy, Adoptive/methods , Killer Cells, Natural/metabolism , Killer Cells, Natural/transplantation , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/therapy , Models, Immunological
20.
Diabetes ; 64(8): 2791-804, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25845663

ABSTRACT

Understanding the molecular networks controlling ectopic lipid deposition, glucose tolerance, and insulin sensitivity is essential to identifying new pharmacological approaches to treat type 2 diabetes. We recently identified serine/threonine protein kinase 25 (STK25) as a negative regulator of glucose and insulin homeostasis based on observations in myoblasts with acute depletion of STK25 and in STK25-overexpressing transgenic mice. Here, we challenged Stk25 knockout mice and wild-type littermates with a high-fat diet and showed that STK25 deficiency suppressed development of hyperglycemia and hyperinsulinemia, improved systemic glucose tolerance, reduced hepatic gluconeogenesis, and increased insulin sensitivity. Stk25(-/-) mice were protected from diet-induced liver steatosis accompanied by decreased protein levels of acetyl-CoA carboxylase, a key regulator of both lipid oxidation and synthesis. Lipid accumulation in Stk25(-/-) skeletal muscle was reduced, and expression of enzymes controlling the muscle oxidative capacity (Cpt1, Acox1, Cs, Cycs, Ucp3) and glucose metabolism (Glut1, Glut4, Hk2) was increased. These data are consistent with our previous study of STK25 knockdown in myoblasts and reciprocal to the metabolic phenotype of Stk25 transgenic mice, reinforcing the validity of the results. The findings suggest that STK25 deficiency protects against the metabolic consequences of chronic exposure to dietary lipids and highlight the potential of STK25 antagonists for the treatment of type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat , Gluconeogenesis/genetics , Insulin Resistance/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Acetyl-CoA Carboxylase/metabolism , Animals , Blood Glucose/metabolism , Body Composition/genetics , Body Weight/genetics , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/genetics , Fatty Liver/genetics , Fatty Liver/metabolism , Glucose Tolerance Test , Hyperglycemia/genetics , Hyperglycemia/metabolism , Hyperinsulinism/genetics , Hyperinsulinism/metabolism , Insulin/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Male , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...