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










Publication year range
1.
Front Oncol ; 12: 994333, 2022.
Article in English | MEDLINE | ID: mdl-36212401

ABSTRACT

Designing studies of immunotherapy is limited due to a lack of pre-clinical models that reliably predict effective immunotherapy responses. To address this gap, we developed humanized mouse models of colorectal cancer (CRC) incorporating patient-derived xenografts (PDX) with human peripheral blood mononuclear cells (PBMC). Humanized mice with CRC PDXs were generated via engraftment of autologous (isolated from the same patients as the PDXs) or allogeneic (isolated from healthy donors) PBMCs. Human T cells were detected in mouse blood, tissues, and infiltrated the implanted PDXs. The inclusion of anti-PD-1 therapy revealed that tumor responses in autologous but not allogeneic models were more comparable to that of patients. An overall non-specific graft-vs-tumor effect occurred in allogeneic models and negatively correlated with that seen in patients. In contrast, autologous humanized mice more accurately correlated with treatment outcomes by engaging pre-existing tumor specific T-cell populations. As autologous T cells appear to be the major drivers of tumor response thus, autologous humanized mice may serve as models at predicting treatment outcomes in pre-clinical settings for therapies reliant on pre-existing tumor specific T-cell populations.

2.
Cancer Res ; 82(18): 3335-3344, 2022 Sep 16.
Article in English | MEDLINE | ID: mdl-35913398

ABSTRACT

KRAS and NRAS mutations occur in 45% of colorectal cancers, with combined MAPK pathway and CDK4/6 inhibition identified as a potential therapeutic strategy. In the current study, this combinatorial treatment approach was evaluated in a co-clinical trial in patient-derived xenografts (PDX), and safety was established in a clinical trial of binimetinib and palbociclib in patients with metastatic colorectal cancer with RAS mutations. Across 18 PDX models undergoing dual inhibition of MEK and CDK4/6, 60% of tumors regressed, meeting the co-clinical trial primary endpoint. Prolonged duration of response occurred predominantly in TP53 wild-type models. Clinical evaluation of binimetinib and palbociclib in a safety lead-in confirmed safety and provided preliminary evidence of activity. Prolonged treatment in PDX models resulted in feedback activation of receptor tyrosine kinases and acquired resistance, which was reversed with a SHP2 inhibitor. These results highlight the clinical potential of this combination in colorectal cancer, along with the utility of PDX-based co-clinical trial platforms for drug development. SIGNIFICANCE: This co-clinical trial of combined MEK-CDK4/6 inhibition in RAS mutant colorectal cancer demonstrates therapeutic efficacy in patient-derived xenografts and safety in patients, identifies biomarkers of response, and uncovers targetable mechanisms of resistance.


Subject(s)
Colorectal Neoplasms , Protein Kinase Inhibitors , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinase 4 , Humans , Mitogen-Activated Protein Kinase Kinases , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Tyrosine/genetics , Xenograft Model Antitumor Assays
3.
Front Oncol ; 11: 643815, 2021.
Article in English | MEDLINE | ID: mdl-34367949

ABSTRACT

Both platelets and the liver play important roles in the processes of coagulation and innate immunity. Platelet responses at the site of an injury are rapid; their immediate activation and structural changes minimize the loss of blood. The majority of coagulation proteins are produced by the liver-a multifunctional organ that also plays a critical role in many processes: removal of toxins and metabolism of fats, proteins, carbohydrates, and drugs. Chronic inflammation, trauma, or other causes of irreversible damage to the liver can dysregulate these pathways leading to organ and systemic abnormalities. In some cases, platelet-to-lymphocyte ratios can also be a predictor of disease outcome. An example is cirrhosis, which increases the risk of bleeding and prothrombotic events followed by activation of platelets. Along with a triggered coagulation cascade, the platelets increase the risk of pro-thrombotic events and contribute to cancer progression and metastasis. This progression and the resulting tissue destruction is physiologically comparable to a persistent, chronic wound. Various cancers, including colorectal cancer, have been associated with increased thrombocytosis, platelet activation, platelet-storage granule release, and thrombosis; anti-platelet agents can reduce cancer risk and progression. However, in cancer patients with pre-existing liver disease who are undergoing chemotherapy, the risk of thrombotic events becomes challenging to manage due to their inherent risk for bleeding. Chemotherapy, also known to induce damage to the liver, further increases the frequency of thrombotic events. Depending on individual patient risks, these factors acting together can disrupt the fragile balance between pro- and anti-coagulant processes, heightening liver thrombogenesis, and possibly providing a niche for circulating tumor cells to adhere to-thus promoting both liver metastasis and cancer-cell survival following treatment (that is, with minimal residual disease in the liver).

4.
Clin Cancer Res ; 27(11): 3039-3049, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33811152

ABSTRACT

PURPOSE: Despite the prognostic importance of immune infiltrate in colorectal cancer, immunotherapy has demonstrated limited clinical activity in refractory metastatic proficient mismatch-repair (pMMR) colorectal cancer. This study explores combining anti-CTLA-4 and an anti-PD-L1 therapy in the preoperative management of resectable colorectal cancer liver metastases with the intent to improve immune responses in this disease setting. PATIENTS AND METHODS: Patients with resectable colorectal cancer liver-only metastases received one dose of tremelimumab and durvalumab preoperatively followed by single-agent durvalumab postoperatively. Primary objectives were to determine feasibility and safety. RESULTS: A total of 24 patients were enrolled between November 2016 and November 2019. Twenty-three patients received treatment [21 pMMR and 2 deficient mismatch-repair (dMMR)] and subsequently 17 (74%; 95% CI: 53%-88%) underwent surgical resection. Grade 3/4 treatment-related immune toxicity and postoperative grade 3/4 toxicity were seen in 5/23 (22%; 95% CI: 10%-44%) and 2/17 (12%; 95% CI: 2%-38%) patients. The median relapse-free survival (RFS) was 9.7 (95% CI: 8.1-17.8) months, and overall survival was 24.5 (95% CI: 16.5-28.4) months. Four patients demonstrated complete pathologic response, two dMMR patients and two POLE mutation patients. Pre- and post-tumor tissue analysis by flow cytometry, immunofluorescence, and RNA sequencing revealed similar levels of T-cell infiltration, but did demonstrate evidence of CD8+ and CD4+ activation posttreatment. An increase in B-cell transcriptome signature and B-cell density was present in posttreatment samples from patients with prolonged RFS. CONCLUSIONS: This study demonstrates the safety of neoadjuvant combination tremelimumab and durvalumab prior to colorectal cancer liver resection. Evidence for T- and B-cell activation following this therapy was seen in pMMR metastatic colorectal cancer.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal/administration & dosage , Colorectal Neoplasms/therapy , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Neoadjuvant Therapy/methods , Adult , Aged , Colorectal Neoplasms/pathology , Digestive System Surgical Procedures/methods , Feasibility Studies , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Perioperative Period , Pilot Projects , Safety , Treatment Outcome
5.
BMC Cancer ; 20(1): 871, 2020 Sep 10.
Article in English | MEDLINE | ID: mdl-32912193

ABSTRACT

BACKGROUND: Non-steroidal anti-inflammatory drugs (NSAIDs) such as aspirin and sulindac are effective for colorectal cancer prevention in humans and some animal models, but concerns over gastro-intestinal (GI) ulceration and bleeding limit their potential for chemopreventive use in broader populations. Recently, the combination of aspirin with a phospholipid, packaged as PL-ASA, was shown to reduce GI toxicity in a small clinical trial. However, these studies were done for relatively short periods of time. Since prolonged, regular use is needed for chemopreventive benefit, it is important to know whether GI safety is maintained over longer use periods and whether cancer prevention efficacy is preserved when an NSAID is combined with a phospholipid. METHODS: As a first step to answering these questions, we treated seven to eight-week-old, male and female C57B/6 Apcmin/+ mice with the NSAID sulindac, with and without phosphatidylcholine (PC) for 3-weeks. At the end of the treatment period, we evaluated polyp burden, gastric toxicity, urinary prostaglandins (as a marker of sulindac target engagement), and blood chemistries. RESULTS: Both sulindac and sulindac-PC treatments resulted in significantly reduced polyp burden, and decreased urinary prostaglandins, but sulindac-PC treatment also resulted in the reduction of gastric lesions compared to sulindac alone. CONCLUSIONS: Together these data provide pre-clinical support for combining NSAIDs with a phospholipid, such as phosphatidylcholine to reduce GI toxicity while maintaining chemopreventive efficacy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colonic Polyps/drug therapy , Colorectal Neoplasms/drug therapy , Sulindac/pharmacology , Adenomatous Polyposis Coli Protein/genetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colonic Polyps/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Models, Animal , Humans , Mice , Phospholipids/pharmacology
6.
Mol Cell Biochem ; 459(1-2): 151-156, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31172369

ABSTRACT

L-Cysteine (LC) is an essential precursor of GSH biosynthesis. GSH is a major physiological antioxidant, and its depletion increases oxidative stress. Diabetes is associated with lower blood levels of LC and GSH. The mechanisms leading to a decrease in LC in diabetes are not entirely known. This study reports a significant decrease in LC in human monocytes exposed to high glucose (HG) concentrations as well as in the blood of type 2 diabetic rats. Thus, a significant decrease in the level of LC in response to exposure to HG supports the assertion that uncontrolled hyperglycemia contributes to a reduction of blood levels of LC and GSH seen in diabetic patients. Increased requirement of LC to replace GSH needed to scavenge excess ROS generated by hyperglycemia can result in lower levels of LC and GSH. Animal and human studies report that LC supplementation improves GSH biosynthesis and is beneficial in lowering oxidative stress and insulin resistance. This suggests that hyperglycemia has a direct role in the impairment of LC and GSH homeostasis in diabetes.


Subject(s)
Cysteine/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Glutathione/metabolism , Hyperglycemia/metabolism , Monocytes/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Humans , Hyperglycemia/pathology , Monocytes/pathology , Rats , Rats, Zucker , U937 Cells
7.
Cancer Metastasis Rev ; 37(2-3): 439-454, 2018 09.
Article in English | MEDLINE | ID: mdl-30112590

ABSTRACT

Platelets can serve as "first responders" in cancer and metastasis. This is partly due to bioactive lipid metabolism that drives both platelet and cancer biology. The two primary eicosanoid metabolites that maintain platelet rapid response homeostasis are prostacyclin made by endothelial cells that inhibits platelet function, which is counterbalanced by thromboxane produced by platelets during activation, aggregation, and platelet recruitment. Both of these arachidonic acid metabolites are inherently unstable due to their chemical structure. Tumor cells by contrast predominantly make more chemically stable prostaglandin E2, which is the primary bioactive lipid associated with inflammation and oncogenesis. Pharmacological, clinical, and epidemiologic studies demonstrate that non-steroidal anti-inflammatory drugs (NSAIDs), which target cyclooxygenases, can help prevent cancer. Much of the molecular and biological impact of these drugs is generally accepted in the field. Cyclooxygenases catalyze the rate-limiting production of substrate used by all synthase molecules, including those that produce prostaglandins along with prostacyclin and thromboxane. Additional eicosanoid metabolites include lipoxygenases, leukotrienes, and resolvins that can also influence platelets, inflammation, and carcinogenesis. Our knowledge base and technology are now progressing toward identifying newer molecular and cellular interactions that are leading to revealing additional targets. This review endeavors to summarize new developments in the field.


Subject(s)
Blood Platelets/metabolism , Lipid Metabolism , Neoplasms/etiology , Neoplasms/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biomarkers , Blood Platelets/drug effects , Epoprostenol/metabolism , Glucose/metabolism , Humans , Immunomodulation , Inflammation/complications , Inflammation/etiology , Inflammation/metabolism , Lipid Metabolism/drug effects , Lipoxygenase/metabolism , Metabolic Networks and Pathways/drug effects , Mixed Function Oxygenases/metabolism , Neoplasms/pathology , Neoplasms/prevention & control , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/metabolism , Receptors, Prostaglandin/metabolism , Thromboxane-A Synthase/antagonists & inhibitors , Thromboxane-A Synthase/metabolism
8.
Antioxid Redox Signal ; 29(17): 1792-1807, 2018 12 10.
Article in English | MEDLINE | ID: mdl-30160165

ABSTRACT

AIMS: 25-Hydroxyvitamin D [25(OH)VD] deficiency/inadequacy is a major public health issue affecting more than 1 billion people worldwide. A convincing association exists between low levels of circulating 25(OH)VD and the poor health outcomes associated with chronic diseases. However, high supraphysiological doses of VD are needed to achieve the required 25(OH)VD levels in the blood, because many subjects respond poorly to supplementation. RESULTS: This study reports a link between 25(OH)VD deficiency and a reduction in glutathione (GSH) in obese adolescents. The improvement in GSH status that results from cosupplementation with VD and l-cysteine (LC; a GSH precursor) significantly reduced oxidative stress in a mouse model of 25(OH)VD deficiency. It also positively upregulated VD regulatory genes (VDBP/VD-25-hydroxylase/VDR) in the liver and glucose metabolism genes (PGC-1α/VDR/GLUT-4) in muscle, boosted 25(OH)VD, and reduced inflammation and insulin resistance (IR) levels in the blood compared with supplementation with VD alone. In vitro GSH deficiency caused increased oxidative stress and downregulation of VDBP/VD-25-hydroxylase/VDR and upregulation of CYP24a1 in hepatocytes and downregulation of PGC-1α/VDR/GLUT-4 in myotubes. This study demonstrates that improvement in the GSH status exerts beneficial effects on the blood levels of 25(OH)VD, as well as on the inflammation and IR in a VD-deficient mouse model. Thus, the VD supplements widely consumed by the public are unlikely to be successful unless the GSH status is also corrected. INNOVATION: These studies demonstrate a previously undiscovered mechanism by which GSH status positively upregulates the bioavailability of 25(OH)VD. CONCLUSION: Supplementation with a combination of VD and LC or GSH precursor, rather than supplementation with VD alone, is beneficial and helps achieve more successful VD supplementation. Antioxid. Redox Signal. 00, 000-000.


Subject(s)
Glucose/metabolism , Glutathione/pharmacology , Inflammation/drug therapy , Oxidative Stress/drug effects , Vitamin D Deficiency/drug therapy , Vitamin D/blood , Vitamin D/metabolism , Adolescent , Animals , Cells, Cultured , Disease Models, Animal , Female , Humans , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Vitamin D/analogs & derivatives , Vitamin D Deficiency/blood
9.
Front Oncol ; 8: 107, 2018.
Article in English | MEDLINE | ID: mdl-29732316

ABSTRACT

The role of platelets in cancer progression has been well recognized in the field of cancer biology. Emerging studies are elaborating further the additional roles and added extent that platelets play in promoting tumorigenesis. Platelets release factors that support tumor growth and also form heterotypic aggregates with tumor cells, which can provide an immune-evasive advantage. Their most critical role may be the inhibition of immune cell function that can negatively impact the body's ability in preventing tumor establishment and growth. This review summarizes the importance of platelets in tumor progression, therapeutic response, survival, and finally the notion of immunotherapy modulation being likely to benefit from the inclusion of platelet inhibitors.

10.
Cancer Metastasis Rev ; 36(2): 235-248, 2017 06.
Article in English | MEDLINE | ID: mdl-28667367

ABSTRACT

Platelets are cytoplasmic fragments generated by megakaryocytes in the bone marrow and do not possess a nucleus. They contribute to the "Circulome" consisting of all circulating cells, factors and macromolecules such as cfDNA. Their primary function is to recognize vascular lesions and initiate thrombus formation that ceases bleeding. This distinctive characteristic of platelets also contributes to cancer and its progression. The ability of platelets to recognize and interact with other cells and neighboring platelets enables them to interact with tumor cells in the circulation. Receptor recognition and factor mediated crosstalk between tumor cells and platelets stimulate platelet activation, release of factors, and aggregation that promotes tumor cell survival and cancer progression. This review describes platelet: (i) contributions to the "Circulome" (ii) their importance as diagnostic tools in predicting cancer risk and (iii) therapies targeting platelet activation in inhibiting tumor progression and metastasis.


Subject(s)
Blood Platelets/metabolism , Blood Platelets/pathology , Colorectal Neoplasms/blood , Colorectal Neoplasms/pathology , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Animals , Colorectal Neoplasms/genetics , DNA, Neoplasm/blood , Humans , RNA, Neoplasm/blood
11.
Cancer Metastasis Rev ; 36(2): 273-288, 2017 06.
Article in English | MEDLINE | ID: mdl-28681242

ABSTRACT

The consensus molecular subtypes (CMS) in colorectal cancer (CRC) represent distinct molecular subcategories of disease as reflected by comprehensive molecular profiling. The four CMS subtypes represent unique biology. CMS1 represents high immune infiltration. CMS2 demonstrates upregulation of canonical pathways such as WNT signaling. Widespread metabolic changes are seen in CMS3. CMS4 represents a mesenchymal phenotype with hallmark features including complement activation, matrix remodeling, angiogenesis, epithelial-mesechymal transition (EMT), integrin upregulation and stromal infiltration. In contrast to this new paradigm, a number of observations regarding CRC remain disconnected. Cancers are associated with thrombocytosis. Venous thromboembolic events are more likely in malignancy and may signify worse prognosis. Aspirin, an anti-platelet agent, has been linked in large observational studies to decrease incidence of adenocarcinoma and less advanced presentations of cancer, in particular CRC. Inflammatory bowel disease is a risk factor for CRC. Gross markers to recognize the immunothrombotic link such as the platelet to lymphocyte ratio are associated with poorer outcomes in many cancers. Platelets are increasingly recognized for their dual roles in coordinating the immune response in addition to hemostasis. Here, we explore how these different but related observations coalesce. Platelets, as first responders to pathogens and injury, form the link between hemostasis and immunity. We outline how platelets contribute to tumorigenesis and how some disconnected ideas may be linked through inflammation. CMS4 through its shared mechanisms has predicted platelet activation as a hallmark feature. We demonstrate a platelet gene expression signature that predicts platelet presence within CMS4 tumors.


Subject(s)
Blood Platelets/physiology , Colorectal Neoplasms/blood , Animals , Blood Platelets/pathology , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Humans , Transcriptome
13.
Free Radic Biol Med ; 95: 268-77, 2016 06.
Article in English | MEDLINE | ID: mdl-27036365

ABSTRACT

Diets that boost ketone production are increasingly used for treating several neurological disorders. Elevation in ketones in most cases is considered favorable, as they provide energy and are efficient in fueling the body's energy needs. Despite all the benefits from ketones, the above normal elevation in the concentration of ketones in the circulation tend to illicit various pathological complications by activating injurious pathways leading to cellular damage. Recent literature demonstrates a plausible link between elevated levels of circulating ketones and oxidative stress, linking hyperketonemia to innumerable morbid conditions. Ketone bodies are produced by the oxidation of fatty acids in the liver as a source of alternative energy that generally occurs in glucose limiting conditions. Regulation of ketogenesis and ketolysis plays an important role in dictating ketone concentrations in the blood. Hyperketonemia is a condition with elevated blood levels of acetoacetate, 3-ß-hydroxybutyrate, and acetone. Several physiological and pathological triggers, such as fasting, ketogenic diet, and diabetes cause an accumulation and elevation of circulating ketones. Complications of the brain, kidney, liver, and microvasculature were found to be elevated in diabetic patients who had elevated ketones compared to those diabetics with normal ketone levels. This review summarizes the mechanisms by which hyperketonemia and ketoacidosis cause an increase in redox imbalance and thereby increase the risk of morbidity and mortality in patients.


Subject(s)
Diabetes Mellitus, Type 1/blood , Ketone Bodies/metabolism , Ketosis/blood , Oxidative Stress/genetics , Acetoacetates/blood , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Energy Metabolism/genetics , Glucose/metabolism , Humans , Ketones/metabolism , Ketosis/pathology , Risk Factors
14.
J Steroid Biochem Mol Biol ; 159: 94-101, 2016 May.
Article in English | MEDLINE | ID: mdl-26949104

ABSTRACT

BACKGROUND: There is a significantly higher incidence of cardiovascular disease (CVD) among type 1 diabetic (T1D) patients than among non-diabetic subjects. T1D is associated with hyperketonemia, a condition with elevated blood levels of ketones, in addition to hyperglycemia. The biochemical mechanism by which vitamin D (VD) may reduce the risk of CVD is not known. This study examines whether VD can be beneficial in reducing hyperketonemia (acetoacetate, AA) induced oxidative stress in endothelial cells. METHODS: HUVEC were pretreated with 1,25(OH)2D3, and later exposed to the ketone body acetoacetate. RESULTS: The increases in ROS production, ICAM-1 expression, MCP-1 secretion, and monocyte adhesion in HUVEC treated with AA were significantly reduced following treatment with 1,25(OH)2D3. Interestingly, an increase in glutathione (GSH) levels was also observed with 1,25(OH)2D3 in ketone treated cells. The effects of 1,25(OH)2D3 on GSH, ROS, and monocyte-endothelial adhesion were prevented in GCLC knockdown HUVEC. This suggests that 1,25(OH)2D3 inhibits ROS, MCP-1, ICAM-1, and adherence of monocytes mediated by the upregulation of GCLC and GSH. CONCLUSION: This study provides evidence for the biochemical mechanism through which VD supplementation may reduce the excess monocyte adhesion to endothelium and inflammation associated with T1D.


Subject(s)
Calcitriol/pharmacology , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Ketosis/metabolism , Oxidative Stress/drug effects , Acetoacetates/pharmacology , Cell Adhesion , Cell Line , Cytokines/metabolism , Drug Evaluation, Preclinical , Glutamate-Cysteine Ligase/genetics , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Monocytes/drug effects , Monocytes/metabolism , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Up-Regulation
15.
Mol Nutr Food Res ; 60(5): 1090-8, 2016 05.
Article in English | MEDLINE | ID: mdl-26778482

ABSTRACT

SCOPE: Vitamin D binding protein (VDBP) status has an effect on and can potentially improve the status of 25(OH) vitamin D and increase the metabolic actions of 25(OH) vitamin D under physiological and pathological conditions. Diabetes is associated with lower levels of glutathione (GSH) and 25(OH) vitamin D. This study examined the hypothesis that upregulation of GSH will also upregulate blood levels of VDBP and 25(OH) vitamin D in type 2 diabetic rats. METHODS AND RESULTS: L-cysteine (LC) supplementation was used to upregulate GSH status in a FL83B hepatocyte cell culture model and in vivo using Zucker diabetic fatty (ZDF) rats. Results show that LC supplementation upregulates both protein and mRNA expression of VDBP and vitamin D receptor (VDR) and GSH status in hepatocytes exposed to high glucose, and that GSH deficiency, induced by glutamate cysteine ligase knockdown, resulted in the downregulation of GSH, VDBP, and VDR and an increase in oxidative stress levels in hepatocytes. In vivo, LC supplementation increased GSH and protein and mRNA expression of VDBP and vitamin D 25-hydroxylase (CYP2R1) in the liver, and simultaneously resulted in elevated blood levels of LC and GSH, as well as increases in VDBP and 25(OH) vitamin D levels, and decreased inflammatory biomarkers in ZDF rats compared with those in placebo-supplemented ZDF rats consuming a similar diet. CONCLUSION: LC supplementation may provide a novel approach by which to raise blood levels of VDBP and 25(OH) vitamin D in type 2 diabetes.


Subject(s)
Cysteine/pharmacology , Glutathione/blood , Hepatocytes/drug effects , Liver/drug effects , Vitamin D-Binding Protein/blood , Vitamin D/analogs & derivatives , Animals , Biomarkers/blood , Cell Line , Culture Media/chemistry , Diabetes Mellitus, Experimental , Dietary Supplements , Gene Expression Regulation , Glucose/analysis , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Glutathione/deficiency , Hepatocytes/metabolism , Liver/metabolism , Male , Mice , Oxidative Stress/drug effects , Rats , Rats, Zucker , Up-Regulation , Vitamin D/blood , Vitamin D-Binding Protein/genetics
16.
Cell Physiol Biochem ; 37(6): 2160-70, 2015.
Article in English | MEDLINE | ID: mdl-26606728

ABSTRACT

BACKGROUND/AIMS: Type 1 diabetic (T1D) patients have a higher incidence of liver disease. T1D patients frequently experience elevated plasma ketone levels along with hyperglycemia. However, no study has examined whether hyperketonemia per se has any role in excess liver damage in T1D. This study investigates the hypothesis that hyperketonemia can induce oxidative stress and cellular dysfunction. METHODS: STZ treated diabetic rats, FL83B hepatocytes, and GCLC knocked down (GSH deficient) hepatocytes were used. RESULTS: The blood levels of ALT and AST, biomarkers of liver damage, and ketones were elevated in T1D rats. An increase in NOX4 and ROS along with a reduction in GSH and GCLC levels was observed in T1D rat livers in comparison to those seen in non-diabetic control or type 2 diabetic rats. MCP-1 and ICAM-1 were also elevated in T1D rat livers and ketone treated hepatocytes. Macrophage markers CCR2 and CD11A that interact with MCP-1, and ICAM-1 respectively, were also elevated in the T1D liver, indicating macrophage infiltration. Additionally, activated macrophages increased hepatocyte damage with ketone treatment, which was similar to that seen in GCLC knockdown hepatocytes without ketones. CONCLUSION: Hyperketonemia per se can induce macrophage mediated damage to hepatocytes and the liver, caused by GSH depletion and oxidative stress up regulation in T1D.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Hepatocytes/metabolism , Ketones/blood , Liver/metabolism , Oxidative Stress , Animals , Diabetes Mellitus, Type 1/pathology , Glutathione/metabolism , Hepatocytes/pathology , Intercellular Adhesion Molecule-1/metabolism , Liver/enzymology , Liver/pathology , Male , NADPH Oxidase 4 , NADPH Oxidases/metabolism , Rats , Rats, Sprague-Dawley , Rats, Zucker , Reactive Oxygen Species/metabolism
17.
Cell Physiol Biochem ; 35(1): 364-73, 2015.
Article in English | MEDLINE | ID: mdl-25591777

ABSTRACT

BACKGROUND/AIMS: The incidence of developing microvascular dysfunction is significantly higher in type 1 diabetic (T1D) patients. Hyperketonemia (acetoacetate, ß-hydroxybutyrate) is frequently found along with hyperglycemia in T1D. Whether hyperketonemia per se contributes to the excess oxidative stress and cellular injury observed in T1D is not known. METHODS: HUVEC were treated with ketones in the presence or absence of high glucose for 24 h. NOX4 siRNA was used to specifically knockdown NOX4 expression in HUVEC. RESULTS: Ketones alone or in combination with high glucose treatment cause a significant increase in oxidative stress, ICAM-1, and monocyte adhesivity to HUVEC. Using an antisense approach, we show that ketone induced increases in ROS, ICAM-1 expression, and monocyte adhesion in endothelial cells were prevented in NOX4 knockdown cells. CONCLUSION: This study reports that elevated levels of ketones upregulate NOX, contributing to increased oxidative stress, ICAM-1 levels, and cellular dysfunction. This provides a novel biochemical mechanism that elucidates the role of hyperketonemia in the excess cellular injury in T1D. New drugs targeting inhibition of NOX seems promising in preventing higher risk of complications associated with T1D.


Subject(s)
Acetoacetates/toxicity , Intercellular Adhesion Molecule-1/metabolism , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Up-Regulation/drug effects , 3-Hydroxybutyric Acid/toxicity , Cell Adhesion/drug effects , Cell Line , Cell Survival/drug effects , Glucose/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Monocytes/cytology , Monocytes/metabolism , NADPH Oxidase 4 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism
18.
Mol Cell Biochem ; 391(1-2): 251-6, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24627243

ABSTRACT

Type 1 diabetic (T1D) patients are hyperglycemic and also show elevated blood levels of ketone bodies, particularly acetoacetate (AA) and ß-hydroxybutyrate (BHB). T1D patients have a greater risk of developing endothelial dysfunction and cardiovascular disease (CVD). Supplementation with cysteine-rich milk proteins has been shown to be beneficial in improving various biomarkers of endothelial dysfunction and CVD. This study examines whether L-cysteine (LC) per se prevents monocyte adhesion to endothelial cells, a critical step in endothelial dysfunction. Human umbilical vein endothelial cells and THP-1 monocytes were pretreated with and without LC (500 µM) for 2 h and then exposed to ketones (AA or BHB, 0-4 mM) and/or high glucose (HG) (25 mM) for 24 h. This study shows that LC reduces HG and ketone-induced ROS production, ICAM-1 expression, and the adhesion of monocytes to endothelial cells. This study provides a biochemical mechanism by which milk protein supplementation can be beneficial in preventing the excess endothelial dysfunction and CVD seen in diabetic patients.


Subject(s)
Cysteine/pharmacology , Endothelial Cells/cytology , Glucose/pharmacology , Ketones/pharmacology , Monocytes/cytology , Reactive Oxygen Species/metabolism , Cell Adhesion/drug effects , Dietary Supplements , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Models, Biological , Monocytes/drug effects , Monocytes/metabolism
19.
Can J Physiol Pharmacol ; 90(12): 1642-6, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23210443

ABSTRACT

Type 1 diabetic patients have hyperketonemia, elevated levels of pro-inflammatory and oxidative stress markers, and a higher incidence of vascular disease. This study examines the hypothesis that hyperketonemia increases reactive oxygen species (ROS) and is in part responsible for increased expression of adhesion molecules in monocytes. THP-1 monocytes were treated with acetoacetate (AA) or ß-hydroxybutyrate (BHB) (0-10 mmol/L) for 24 h. Results show that AA, but not BHB, increases ROS production in monocytes. Pretreatment of monocytes with N-acetylcysteine (NAC) inhibited AA-induced ROS production. AA treatment induced upregulation of LFA-1 and pretreatment of monocytes with NAC or an inhibitor to p38 MAPK inhibited this upregulation in monocytes. This suggests that physiological concentrations of AA can contribute to increased ROS and activation of p38 MAPK, which may be responsible for AA-induced upregulation of LFA-1 in monocytes. Thus, hyperketonemia contributes to the risk for cardiovascular disease in type 1 diabetes.


Subject(s)
Ketosis/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Monocytes/metabolism , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , 3-Hydroxybutyric Acid/pharmacology , Acetoacetates/pharmacology , Acetylcysteine/pharmacology , Cardiovascular Diseases/enzymology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cells, Cultured , Humans , Ketosis/enzymology , Ketosis/genetics , Lymphocyte Function-Associated Antigen-1/genetics , MAP Kinase Signaling System , Monocytes/enzymology , Oxidative Stress/drug effects , Oxidative Stress/genetics , Up-Regulation , p38 Mitogen-Activated Protein Kinases/genetics
20.
BMC Res Notes ; 4: 13, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21255428

ABSTRACT

BACKGROUND: The S. cerevisiae mating type switch model of double-strand break (DSB) repair, utilizing the HO endonuclease, is one of the best studied systems for both Homologous Recombination Repair (HRR) and direct ends-joining repair (Non-Homologous Ends Joining - NHEJ). We have recently transposed that system to a mammalian cell culture model taking advantage of an adenovirus expressing HO and an integrated genomic target. This made it possible to compare directly the mechanism of repair between yeast and mammalian cells for the same type of induced DSB. Studies of DSB repair have emphasized commonality of features, proteins and machineries between organisms, and differences when conservation is not found. Two proteins that stand out that differ between yeast and mammalian cells are DNA-PK, a protein kinase that is activated by the presence of DSBs, and Artemis, a nuclease whose activity is modulated by DNA-PK and ATM. In this report we describe how these two proteins may be involved in a specific pattern of ends-processing at the DSB, particularly in the context of heterochromatin. FINDINGS: We previously published that the repair of the HO-induced DSB was generally accurate and occurred by simple rejoining of the cohesive 3'-overhangs generated by HO. During continuous passage of those cells in the absence of puromycin selection, the locus appears to have become more heterochromatic and silenced by displaying several features. 1) The site had become less accessible to cleavage by the HO endonuclease; 2) the expression of the puro mRNA, which confers resistance to puromycin, had become reduced; 3) occupancy of nucleosomes at the site (ChIP for histone H3) was increased, an indicator for more condensed chromatin. After reselection of these cells by addition of puromycin, many of these features were reversed. However, even the reselected cells were not identical in the pattern of cleavage and repair as the cells when originally created. Specifically, the pattern of repair revealed discrete deletions at the DSB that indicated unit losses of nucleosomes (or other protein complexes) before religation, represented by a ladder of PCR products reminiscent of an internucleosomal cleavage that is typically observed during apoptosis. This pattern of cleavage suggested to us that perhaps, Artemis, a protein that is believed to generate the internucleosomal fragments during apoptosis and in DSB repair, was involved in that specific pattern of ends-processing. Preliminary evidence indicates that this may be the case, since knock-down of Artemis with siRNA eliminated the laddering pattern and revealed instead an extensive exonucleolytic processing of the ends before religation. CONCLUSIONS: e have generated a system in mammalian cells where the absence of positive selection resulted in chromatin remodeling at the target locus that recapitulates many of the features of the mating-type switching system in yeast. Specifically, just as for yeast HML and HMR, the locus had become transcriptionally repressed; accessibility to cleavage by the HO endonuclease was reduced; and processing of the ends was drastically changed. The switch was from high-fidelity religation of the cohesive ends, to a pattern of release of internucleosomal fragments, perhaps in search of micro-homology stretches for ligation. This is consistent with reports that the involvement of ATM, DNA-PK and Artemis in DSB repair is largely focused to heterochromatic regions, and not required for the majority of IR-induced DSB repair foci in euchromatin.

SELECTION OF CITATIONS
SEARCH DETAIL
...