Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Metab ; 3(10): 1342-1356, 2021 10.
Article in English | MEDLINE | ID: mdl-34650272

ABSTRACT

Diet-induced obesity is a major risk factor for metabolic syndrome, diabetes and cardiovascular disease. Here, we show that a 5-d fasting-mimicking diet (FMD), administered every 4 weeks for a period of 2 years, ameliorates the detrimental changes caused by consumption of a high-fat, high-calorie diet (HFCD) in female mice. We demonstrate that monthly FMD cycles inhibit HFCD-mediated obesity by reducing the accumulation of visceral and subcutaneous fat without causing loss of lean body mass. FMD cycles increase cardiac vascularity and function and resistance to cardiotoxins, prevent HFCD-dependent hyperglycaemia, hypercholesterolaemia and hyperleptinaemia and ameliorate impaired glucose and insulin tolerance. The effect of monthly FMD cycles on gene expression associated with mitochondrial metabolism and biogenesis in adipocytes and the sustained ketogenesis in HFCD-fed mice indicate a role for fat cell reprogramming in obesity prevention. These effects of an FMD on adiposity and cardiac ageing could explain the protection from HFCD-dependent early mortality.


Subject(s)
Cardiovascular Diseases/pathology , Diet, High-Fat , Fasting , Longevity , Metabolic Diseases/pathology , Animals , Cardiovascular Diseases/metabolism , Female , Metabolic Diseases/metabolism , Mice
2.
Sci Immunol ; 6(59)2021 05 14.
Article in English | MEDLINE | ID: mdl-33990379

ABSTRACT

Monoamine oxidase A (MAO-A) is an enzyme best known for its function in the brain, where it breaks down neurotransmitters and thereby influences mood and behavior. Small-molecule MAO inhibitors (MAOIs) have been developed and are clinically used for treating depression and other neurological disorders. However, the involvement of MAO-A in antitumor immunity has not been reported. Here, we observed induction of the Maoa gene in tumor-infiltrating immune cells. Maoa knockout mice exhibited enhanced antitumor T cell immunity and suppressed tumor growth. MAOI treatment significantly suppressed tumor growth in preclinical mouse syngeneic and human xenograft tumor models in a T cell-dependent manner. Combining MAOI and anti-PD-1 treatments generated synergistic tumor suppression effects. Clinical data correlation studies associated intratumoral MAOA expression with T cell dysfunction and decreased patient survival in a broad range of cancers. We further demonstrated that MAO-A restrains antitumor T cell immunity through controlling intratumoral T cell autocrine serotonin signaling. Together, these data identify MAO-A as an immune checkpoint and support repurposing MAOI antidepressants for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Immunotherapy , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/immunology , Neoplasms/therapy , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Female , Humans , Mice, Inbred C57BL , Mice, Transgenic , Monoamine Oxidase/genetics , Neoplasms/immunology , Neoplasms/pathology
3.
J Exp Med ; 216(12): 2869-2882, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31628186

ABSTRACT

T cells demand massive energy to combat cancer; however, the metabolic regulators controlling antitumor T cell immunity have just begun to be unveiled. When studying nutrient usage of tumor-infiltrating immune cells in mice, we detected a sharp increase of the expression of a CrT (Slc6a8) gene, which encodes a surface transporter controlling the uptake of creatine into a cell. Using CrT knockout mice, we showed that creatine uptake deficiency severely impaired antitumor T cell immunity. Supplementing creatine to WT mice significantly suppressed tumor growth in multiple mouse tumor models, and the combination of creatine supplementation with a PD-1/PD-L1 blockade treatment showed synergistic tumor suppression efficacy. We further demonstrated that creatine acts as a "molecular battery" conserving bioenergy to power T cell activities. Therefore, our results have identified creatine as an important metabolic regulator controlling antitumor T cell immunity, underscoring the potential of creatine supplementation to improve T cell-based cancer immunotherapies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Creatine/metabolism , Immunomodulation , Neoplasms/immunology , Neoplasms/metabolism , Animals , Antigens, Neoplasm/immunology , Cell Line, Tumor , Creatine/administration & dosage , Creatine/deficiency , Dietary Supplements , Energy Metabolism , Gene Expression Regulation, Neoplastic , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mice, Knockout , Models, Biological , Neoplasms/genetics , Neoplasms/pathology , Tumor Microenvironment
4.
Cell Stem Cell ; 25(4): 542-557.e9, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31495780

ABSTRACT

Invariant natural killer T (iNKT) cells are potent immune cells for targeting cancer; however, their clinical application has been hindered by their low numbers in cancer patients. Here, we developed a proof-of-concept for hematopoietic stem cell-engineered iNKT (HSC-iNKT) cell therapy with the potential to provide therapeutic levels of iNKT cells for a patient's lifetime. Using a human HSC engrafted mouse model and a human iNKT TCR gene engineering approach, we demonstrated the efficient and long-term generation of HSC-iNKT cells in vivo. These HSC-iNKT cells closely resembled endogenous human iNKT cells, could deploy multiple mechanisms to attack tumor cells, and effectively suppressed tumor growth in vivo in multiple human tumor xenograft mouse models. Preclinical safety studies showed no toxicity or tumorigenicity of the HSC-iNKT cell therapy. Collectively, these results demonstrated the feasibility, safety, and cancer therapy potential of the proposed HSC-iNKT cell therapy and laid a foundation for future clinical development.


Subject(s)
Hematopoietic Stem Cells/physiology , Immunotherapy, Adoptive/methods , Natural Killer T-Cells/physiology , Neoplasms/therapy , Animals , Cells, Cultured , Genetic Engineering , Humans , Mice , Mice, SCID , Natural Killer T-Cells/transplantation , Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , Xenograft Model Antitumor Assays
5.
J Cell Physiol ; 233(2): 1202-1212, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28471474

ABSTRACT

Hepatocellular carcinoma (HCC) has a poor outcome. Most HCCs develop in the context of liver fibrosis and cirrhosis caused by chronic inflammation. Short-term fasting approaches enhance the activity of chemotherapy in preclinical cancer models, other than HCC. Multi-tyrosine kinase inhibitor Sorafenib is the mainstay of treatment in HCC. However, its benefit is frequently short-lived. Whether fasting can alleviate liver fibrosis and whether combining fasting with Sorafenib is beneficial remains unknown. A 24 hr fasting (2% serum, 0.1% glucose)-induced changes on human hepatic stellate cells (HSC) LX-2 proliferation/viability/cell cycle were assessed by MTT and flow cytometry. Expression of lypolysaccharide (LPS)-induced activation markers (vimentin, αSMA) was evaluated by qPCR and immunoblotting. Liver fibrosis and inflammation were evaluated in a mouse model of steatohepatitis exposed to cycles of fasting, by histological and biochemical analyses. A 24 hr fasting-induced changes were also analyzed on the proliferation/viability/glucose uptake of human HCC cells exposed to Sorafenib. An expression panel of genes involved in survival, inflammation, and metabolism was examined by qPCR in HCC cells exposed to fasting and/or Sorafenib. Fasting decreased the proliferation and the activation of HSC. Repeated cycles of short term starvation were safe in mice but did not improve fibrosis. Fasting synergized with Sorafenib in hampering HCC cell growth and glucose uptake. Finally, fasting normalized the expression levels of genes which are commonly altered by Sorafenib in HCC cells. Fasting or fasting-mimicking diet diets should be evaluated in preclinical studies as a mean to potentiate the activity of Sorafenib in clinical use.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cell Proliferation/drug effects , Fasting/metabolism , Hepatic Stellate Cells/drug effects , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Glucose/metabolism , Hep G2 Cells , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Lipopolysaccharides/pharmacology , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice, Inbred C57BL , Niacinamide/pharmacology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Sorafenib , Time Factors
7.
PLoS Biol ; 15(3): e2001951, 2017 03.
Article in English | MEDLINE | ID: mdl-28358805

ABSTRACT

Fasting reduces glucose levels and protects mice against chemotoxicity, yet drugs that promote hyperglycemia are widely used in cancer treatment. Here, we show that dexamethasone (Dexa) and rapamycin (Rapa), commonly administered to cancer patients, elevate glucose and sensitize cardiomyocytes and mice to the cancer drug doxorubicin (DXR). Such toxicity can be reversed by reducing circulating glucose levels by fasting or insulin. Furthermore, glucose injections alone reversed the fasting-dependent protection against DXR in mice, indicating that elevated glucose mediates, at least in part, the sensitizing effects of rapamycin and dexamethasone. In yeast, glucose activates protein kinase A (PKA) to accelerate aging by inhibiting transcription factors Msn2/4. Here, we show that fasting or glucose restriction (GR) regulate PKA and AMP-activated protein kinase (AMPK) to protect against DXR in part by activating the mammalian Msn2/4 ortholog early growth response protein 1 (EGR1). Increased expression of the EGR1-regulated cardioprotective peptides atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP) in heart tissue may also contribute to DXR resistance. Our findings suggest the existence of a glucose-PKA pathway that inactivates conserved zinc finger stress-resistance transcription factors to sensitize cells to toxins conserved from yeast to mammals. Our findings also describe a toxic role for drugs widely used in cancer treatment that promote hyperglycemia and identify dietary interventions that reverse these effects.


Subject(s)
Antineoplastic Agents/pharmacology , Dexamethasone/pharmacology , Early Growth Response Protein 1/metabolism , Fasting/metabolism , Glucose/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Atrial Natriuretic Factor/metabolism , Cardiotoxins/toxicity , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoprotection/drug effects , Diet , Female , Hyperglycemia/pathology , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Natriuretic Peptide, Brain/metabolism , Stress, Physiological/drug effects , Time Factors
8.
Sci Transl Med ; 9(377)2017 02 15.
Article in English | MEDLINE | ID: mdl-28202779

ABSTRACT

Calorie restriction or changes in dietary composition can enhance healthy aging, but the inability of most subjects to adhere to chronic and extreme diets, as well as potentially adverse effects, limits their application. We randomized 100 generally healthy participants from the United States into two study arms and tested the effects of a fasting-mimicking diet (FMD)-low in calories, sugars, and protein but high in unsaturated fats-on markers/risk factors associated with aging and age-related diseases. We compared subjects who followed 3 months of an unrestricted diet to subjects who consumed the FMD for 5 consecutive days per month for 3 months. Three FMD cycles reduced body weight, trunk, and total body fat; lowered blood pressure; and decreased insulin-like growth factor 1 (IGF-1). No serious adverse effects were reported. After 3 months, control diet subjects were crossed over to the FMD program, resulting in a total of 71 subjects completing three FMD cycles. A post hoc analysis of subjects from both FMD arms showed that body mass index, blood pressure, fasting glucose, IGF-1, triglycerides, total and low-density lipoprotein cholesterol, and C-reactive protein were more beneficially affected in participants at risk for disease than in subjects who were not at risk. Thus, cycles of a 5-day FMD are safe, feasible, and effective in reducing markers/risk factors for aging and age-related diseases. Larger studies in patients with diagnosed diseases or selected on the basis of risk factors are warranted to confirm the effect of the FMD on disease prevention and treatment.


Subject(s)
Aging/pathology , Biomarkers/metabolism , Cardiovascular Diseases/pathology , Diabetes Mellitus/pathology , Diet , Fasting/physiology , Neoplasms/pathology , Adult , Female , Follow-Up Studies , Humans , Male , Risk Factors
9.
Cancer Cell ; 30(1): 136-146, 2016 07 11.
Article in English | MEDLINE | ID: mdl-27411588

ABSTRACT

Immune-based interventions are promising strategies to achieve long-term cancer-free survival. Fasting was previously shown to differentially sensitize tumors to chemotherapy while protecting normal cells, including hematopoietic stem and immune cells, from its toxic side effects. Here, we show that the combination of chemotherapy and a fasting-mimicking diet (FMD) increases the levels of bone marrow common lymphoid progenitor cells and cytotoxic CD8(+) tumor-infiltrating lymphocytes (TILs), leading to a major delay in breast cancer and melanoma progression. In breast tumors, this effect is partially mediated by the downregulation of the stress-responsive enzyme heme oxygenase-1 (HO-1). These data indicate that FMD cycles combined with chemotherapy can enhance T cell-dependent targeted killing of cancer cells both by stimulating the hematopoietic system and by enhancing CD8(+)-dependent tumor cytotoxicity.


Subject(s)
Breast Neoplasms/diet therapy , Breast Neoplasms/drug therapy , Doxorubicin/administration & dosage , Heme Oxygenase-1/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Breast Neoplasms/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Combined Modality Therapy , Disease Progression , Down-Regulation , Doxorubicin/pharmacology , Fasting , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , MCF-7 Cells , Mice , Neoplasm Transplantation , T-Lymphocytes, Cytotoxic/drug effects
10.
Circ Res ; 118(10): 1612-25, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27174953

ABSTRACT

Recent studies indicate that dietary interventions have the potential to prevent and even treat cardiovascular disease, which is the leading cause of death. Many of these studies have focused on various animal models that are able to recreate one or more conditions or elevate risk factors that characterize the disease. Here, we highlight macronutrient-focused interventions in both mammalian model organisms and humans with emphasis on some of the most relevant and well-established diets known to be associated with cardiovascular disease prevention and treatment. We also discuss more recent dietary interventions in rodents, monkeys, and humans, which affect atherosclerosis and cardiovascular diseases with focus on those that also delay aging.


Subject(s)
Coronary Disease/diet therapy , Diet, Protein-Restricted , Heart/growth & development , Animals , Apolipoproteins E/genetics , Coronary Disease/etiology , Coronary Disease/prevention & control , Humans , Mice , Obesity/complications , Obesity/genetics , Rats
11.
Cell Metab ; 22(1): 86-99, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26094889

ABSTRACT

Prolonged fasting (PF) promotes stress resistance, but its effects on longevity are poorly understood. We show that alternating PF and nutrient-rich medium extended yeast lifespan independently of established pro-longevity genes. In mice, 4 days of a diet that mimics fasting (FMD), developed to minimize the burden of PF, decreased the size of multiple organs/systems, an effect followed upon re-feeding by an elevated number of progenitor and stem cells and regeneration. Bi-monthly FMD cycles started at middle age extended longevity, lowered visceral fat, reduced cancer incidence and skin lesions, rejuvenated the immune system, and retarded bone mineral density loss. In old mice, FMD cycles promoted hippocampal neurogenesis, lowered IGF-1 levels and PKA activity, elevated NeuroD1, and improved cognitive performance. In a pilot clinical trial, three FMD cycles decreased risk factors/biomarkers for aging, diabetes, cardiovascular disease, and cancer without major adverse effects, providing support for the use of FMDs to promote healthspan.


Subject(s)
Cognition , Fasting , Longevity , Abdominal Fat/metabolism , Adult , Aged , Aging , Animals , Body Weight , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/diet therapy , Cardiovascular Diseases/metabolism , Diet , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neoplasms/diet therapy , Neoplasms/metabolism , Neoplasms/pathology , Neurogenesis , Pilot Projects , Psychomotor Performance , Regeneration , Saccharomyces cerevisiae/cytology , Young Adult
12.
J Clin Endocrinol Metab ; 100(7): 2589-96, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25985182

ABSTRACT

CONTEXT: Ecuadorian subjects with GH receptor deficiency (GHRD) have not developed diabetes, despite obesity. OBJECTIVE: We sought to determine the metabolic associations for this phenomenon. DESIGN: Four studies were carried out: 1) glucose, lipid, adipocytokine concentrations; 2) metabolomics evaluation; 3) metabolic responses to a high-calorie meal; and 4) oral glucose tolerance tests. SETTING: Clinical Research Institute in Quito, Ecuador. SUBJECTS: Adults homozygous for the E180 splice mutation of the GH receptor (GHRD) were matched for age, gender, and body mass index with unaffected control relatives (C) as follows: study 1, 27 GHRD and 35 C; study 2, 10 GHRD and 10 C; study 3, seven GHRD and 11 C; and study 4, seven GHRD and seven C. RESULTS: Although GHRD subjects had greater mean percentage body fat than controls, their fasting insulin, 2-hour blood glucose, and triglyceride levels were lower. The indicator of insulin sensitivity, homeostasis model of assessment 2%S, was greater (P < .0001), and the indicator of insulin resistance, homeostasis model of assessment 2-IR, was lower (P = .0025). Metabolomic differences between GHRD and control subjects were consistent with their differing insulin sensitivity, including postprandial decreases of branched-chain amino acids that were more pronounced in controls. High molecular weight and total adiponectin concentrations were greater in GHRD (P = .0004 and P = .0128, respectively), and leptin levels were lower (P = .02). Although approximately 65% the weight of controls, GHRD subjects consumed an identical high-calorie meal; nonetheless, their mean glucose concentrations were lower, with mean insulin levels one-third those of controls. Results of the 2-hour oral glucose tolerance test were similar. MAIN OUTCOME MEASURES: Measures of insulin sensitivity, adipocytokines, and energy metabolites. CONCLUSIONS: Without GH counter-regulation, GHRD is associated with insulin efficiency and obesity. Lower leptin levels, despite higher percentage body fat, suggest that obesity-associated leptin resistance is GH dependent. Elevated adiponectin levels not correlated with percentage body fat indicate that GH signaling is necessary for their typical suppression with obesity.


Subject(s)
Insulin Resistance , Laron Syndrome/complications , Laron Syndrome/metabolism , Obesity/complications , Obesity/metabolism , Adipokines/blood , Adult , Body Mass Index , Carbohydrate Metabolism , Case-Control Studies , Ecuador/epidemiology , Female , Humans , Laron Syndrome/epidemiology , Lipids/blood , Male , Middle Aged , Obesity/epidemiology , Young Adult
13.
Aging Cell ; 12(6): 950-4, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23815295

ABSTRACT

The liver is the only internal human organ capable of natural regeneration of lost tissue, as little as 25% of a liver can regenerate into a whole liver. The process of aging predisposes to hepatic functional and structural impairment and metabolic risk. Therefore, understanding how aging could affect the molecular pathology of liver diseases is particularly important, and few studies to date have tackled this complex process. The most common liver disease, affecting one-third of the overall population, is nonalcoholic fatty liver disease (NAFLD), characterized by an intrahepatic accumulation of lipids. NAFLD can evolve into nonalcoholic steatohepatitis (NASH) in the presence of oxidative stress and inflammation. NASH is a serious risk factor for disabling and deadly liver diseases such as cirrhosis and hepatocellular carcinoma (HCC). Old age seems to favor NAFLD, NASH, and ultimately HCC, in agreement with the inflamm-aging theory, according to which aging accrues inflammation. However, the incidence of HCC drops significantly in the very elderly (individuals aged more than 70) and the relationship between the progression of NAFLD/NASH/HCC and very old age is obscure. In this review, we discuss the literature and we argue that there might be an age window in which the liver becomes resistant to the development of injury; this needs to be studied to understand fully the interaction between age and liver diseases from a therapeutic perspective.


Subject(s)
Aging/pathology , Liver Diseases/physiopathology , Caloric Restriction , Growth Hormone/metabolism , Humans , Inflammation/pathology , Inflammation/physiopathology , Liver/pathology , Liver/physiopathology , Liver Diseases/pathology
14.
PLoS One ; 7(7): e40954, 2012.
Article in English | MEDLINE | ID: mdl-22879864

ABSTRACT

Several drugs and natural compounds are known to be highly neurotoxic, triggering epileptic convulsions or seizures, and causing headaches, agitations, as well as other neuronal symptoms. The neurotoxic effects of some of these compounds, including theophylline and ginkgotoxin, have been traced to their inhibitory activity against human pyridoxal kinase (hPL kinase), resulting in deficiency of the active cofactor form of vitamin B6, pyridoxal 5'-phosphate (PLP). Pyridoxal (PL), an inactive form of vitamin B6 is converted to PLP by PL kinase. PLP is the B6 vitamer required as a cofactor for over 160 enzymatic activities essential in primary and secondary metabolism. We have performed structural and kinetic studies on hPL kinase with several potential inhibitors, including ginkgotoxin and theophylline. The structural studies show ginkgotoxin and theophylline bound at the substrate site, and are involved in similar protein interactions as the natural substrate, PL. Interestingly, the phosphorylated product of ginkgotoxin is also observed bound at the active site. This work provides insights into the molecular basis of hPL kinase inhibition and may provide a working hypothesis to quickly screen or identify neurotoxic drugs as potential hPL kinase inhibitors. Such adverse effects may be prevented by administration of an appropriate form of vitamin B6, or provide clues of how to modify these drugs to help reduce their hPL kinase inhibitory effects.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Neurotoxins/chemistry , Pyridoxal Kinase/antagonists & inhibitors , Pyridoxal Kinase/chemistry , Pyridoxine/analogs & derivatives , Theophylline/chemistry , Crystallography, X-Ray , Humans , Neurotoxins/pharmacology , Pyridoxine/chemistry , Pyridoxine/pharmacology , Theophylline/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...