Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 408
Filter
Add more filters

Publication year range
1.
Cell ; 187(15): 4095-4112.e21, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38885650

ABSTRACT

The growth of antimicrobial resistance (AMR) highlights an urgent need to identify bacterial pathogenic functions that may be targets for clinical intervention. Although severe infections profoundly alter host metabolism, prior studies have largely ignored microbial metabolism in this context. Here, we describe an iterative, comparative metabolomics pipeline to uncover microbial metabolic features in the complex setting of a host and apply it to investigate gram-negative bloodstream infection (BSI) in patients. We find elevated levels of bacterially derived acetylated polyamines during BSI and discover the enzyme responsible for their production (SpeG). Blocking SpeG activity reduces bacterial proliferation and slows pathogenesis. Reduction of SpeG activity also enhances bacterial membrane permeability and increases intracellular antibiotic accumulation, allowing us to overcome AMR in culture and in vivo. This study highlights how tools to study pathogen metabolism in the natural context of infection can reveal and prioritize therapeutic strategies for addressing challenging infections.


Subject(s)
Metabolomics , Polyamines , Humans , Animals , Polyamines/metabolism , Mice , Bacteremia/microbiology , Bacteremia/metabolism , Bacteremia/drug therapy , Anti-Bacterial Agents/pharmacology , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/metabolism , Female
2.
Cell ; 187(19): 5413-5430.e29, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39163861

ABSTRACT

Bacterial vaginosis (BV), a common syndrome characterized by Lactobacillus-deficient vaginal microbiota, is associated with adverse health outcomes. BV often recurs after standard antibiotic therapy in part because antibiotics promote microbiota dominance by Lactobacillus iners instead of Lactobacillus crispatus, which has more beneficial health associations. Strategies to promote L. crispatus and inhibit L. iners are thus needed. We show that oleic acid (OA) and similar long-chain fatty acids simultaneously inhibit L. iners and enhance L. crispatus growth. These phenotypes require OA-inducible genes conserved in L. crispatus and related lactobacilli, including an oleate hydratase (ohyA) and putative fatty acid efflux pump (farE). FarE mediates OA resistance, while OhyA is robustly active in the vaginal microbiota and enhances bacterial fitness by biochemically sequestering OA in a derivative form only ohyA-harboring organisms can exploit. OA promotes L. crispatus dominance more effectively than antibiotics in an in vitro BV model, suggesting a metabolite-based treatment approach.


Subject(s)
Fatty Acids , Lactobacillus , Vagina , Vaginosis, Bacterial , Vaginosis, Bacterial/drug therapy , Vaginosis, Bacterial/microbiology , Female , Humans , Vagina/microbiology , Lactobacillus/metabolism , Fatty Acids/metabolism , Oleic Acid/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Lactobacillus crispatus/metabolism , Microbiota/drug effects , Bacterial Proteins/metabolism
3.
Cell ; 186(4): 732-747.e16, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36803603

ABSTRACT

Hematopoietic stem cells (HSCs) have a number of unique physiologic adaptations that enable lifelong maintenance of blood cell production, including a highly regulated rate of protein synthesis. Yet, the precise vulnerabilities that arise from such adaptations have not been fully characterized. Here, inspired by a bone marrow failure disorder due to the loss of the histone deubiquitinase MYSM1, characterized by selectively disadvantaged HSCs, we show how reduced protein synthesis in HSCs results in increased ferroptosis. HSC maintenance can be fully rescued by blocking ferroptosis, despite no alteration in protein synthesis rates. Importantly, this selective vulnerability to ferroptosis not only underlies HSC loss in MYSM1 deficiency but also characterizes a broader liability of human HSCs. Increasing protein synthesis rates via MYSM1 overexpression makes HSCs less susceptible to ferroptosis, more broadly illustrating the selective vulnerabilities that arise in somatic stem cell populations as a result of physiologic adaptations.


Subject(s)
Ferroptosis , Hematopoietic Stem Cells , Humans , Endopeptidases/metabolism , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Trans-Activators/metabolism , Ubiquitin-Specific Proteases/metabolism
4.
Cell ; 185(26): 4921-4936.e15, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36563663

ABSTRACT

The perinatal period represents a critical window for cognitive and immune system development, promoted by maternal and infant gut microbiomes and their metabolites. Here, we tracked the co-development of microbiomes and metabolomes from late pregnancy to 1 year of age using longitudinal multi-omics data from a cohort of 70 mother-infant dyads. We discovered large-scale mother-to-infant interspecies transfer of mobile genetic elements, frequently involving genes associated with diet-related adaptations. Infant gut metabolomes were less diverse than maternal but featured hundreds of unique metabolites and microbe-metabolite associations not detected in mothers. Metabolomes and serum cytokine signatures of infants who received regular-but not extensively hydrolyzed-formula were distinct from those of exclusively breastfed infants. Taken together, our integrative analysis expands the concept of vertical transmission of the gut microbiome and provides original insights into the development of maternal and infant microbiomes and metabolomes during late pregnancy and early life.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Female , Humans , Infant , Pregnancy , Gastrointestinal Microbiome/genetics , Microbiota/genetics , Mothers , Breast Feeding , Feces , Interspersed Repetitive Sequences
5.
Cell ; 185(23): 4280-4297.e12, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36323316

ABSTRACT

The gut microbiome has an important role in infant health and development. We characterized the fecal microbiome and metabolome of 222 young children in Dhaka, Bangladesh during the first two years of life. A distinct Bifidobacterium longum clade expanded with introduction of solid foods and harbored enzymes for utilizing both breast milk and solid food substrates. The clade was highly prevalent in Bangladesh, present globally (at lower prevalence), and correlated with many other gut taxa and metabolites, indicating an important role in gut ecology. We also found that the B. longum clades and associated metabolites were implicated in childhood diarrhea and early growth, including positive associations between growth measures and B. longum subsp. infantis, indolelactate and N-acetylglutamate. Our data demonstrate geographic, cultural, seasonal, and ecological heterogeneity that should be accounted for when identifying microbiome factors implicated in and potentially benefiting infant development.


Subject(s)
Bifidobacterium longum , Infant , Child , Female , Humans , Child, Preschool , Bifidobacterium longum/metabolism , Bifidobacterium/metabolism , Weaning , Oligosaccharides/metabolism , Bangladesh , Milk, Human , Feces/microbiology
6.
Cell ; 177(2): 315-325.e14, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30929905

ABSTRACT

Transmission of malaria parasites occurs when a female Anopheles mosquito feeds on an infected host to acquire nutrients for egg development. How parasites are affected by oogenetic processes, principally orchestrated by the steroid hormone 20-hydroxyecdysone (20E), remains largely unknown. Here we show that Plasmodium falciparum development is intimately but not competitively linked to processes shaping Anopheles gambiae reproduction. We unveil a 20E-mediated positive correlation between egg and oocyst numbers; impairing oogenesis by multiple 20E manipulations decreases parasite intensities. These manipulations, however, accelerate Plasmodium growth rates, allowing sporozoites to become infectious sooner. Parasites exploit mosquito lipids for faster growth, but they do so without further affecting egg development. These results suggest that P. falciparum has adopted a non-competitive evolutionary strategy of resource exploitation to optimize transmission while minimizing fitness costs to its mosquito vector. Our findings have profound implications for currently proposed control strategies aimed at suppressing mosquito populations.


Subject(s)
Ecdysterone/metabolism , Host-Parasite Interactions/physiology , Malaria, Falciparum/parasitology , Animals , Anopheles/parasitology , Culicidae , Ecdysterone/physiology , Female , HEK293 Cells , Humans , Insect Vectors , Malaria/parasitology , Mice , Mosquito Vectors , NIH 3T3 Cells , Oogenesis/physiology , Plasmodium/metabolism , Plasmodium falciparum , Sporozoites , Steroids/metabolism
7.
Cell ; 179(7): 1483-1498.e22, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31813625

ABSTRACT

Metabolism has been shown to control peripheral immunity, but little is known about its role in central nervous system (CNS) inflammation. Through a combination of proteomic, metabolomic, transcriptomic, and perturbation studies, we found that sphingolipid metabolism in astrocytes triggers the interaction of the C2 domain in cytosolic phospholipase A2 (cPLA2) with the CARD domain in mitochondrial antiviral signaling protein (MAVS), boosting NF-κB-driven transcriptional programs that promote CNS inflammation in experimental autoimmune encephalomyelitis (EAE) and, potentially, multiple sclerosis. cPLA2 recruitment to MAVS also disrupts MAVS-hexokinase 2 (HK2) interactions, decreasing HK enzymatic activity and the production of lactate involved in the metabolic support of neurons. Miglustat, a drug used to treat Gaucher and Niemann-Pick disease, suppresses astrocyte pathogenic activities and ameliorates EAE. Collectively, these findings define a novel immunometabolic mechanism that drives pro-inflammatory astrocyte activities, outlines a new role for MAVS in CNS inflammation, and identifies candidate targets for therapeutic intervention.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Astrocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Phospholipases A2, Secretory/metabolism , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/pharmacology , 1-Deoxynojirimycin/therapeutic use , Adaptor Proteins, Signal Transducing/genetics , Animals , Astrocytes/drug effects , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Hexokinase/metabolism , Humans , Lactic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phospholipases A2, Secretory/genetics
8.
Cell ; 170(1): 199-212.e20, 2017 Jun 29.
Article in English | MEDLINE | ID: mdl-28666119

ABSTRACT

Type 2 diabetes (T2D) affects Latinos at twice the rate seen in populations of European descent. We recently identified a risk haplotype spanning SLC16A11 that explains ∼20% of the increased T2D prevalence in Mexico. Here, through genetic fine-mapping, we define a set of tightly linked variants likely to contain the causal allele(s). We show that variants on the T2D-associated haplotype have two distinct effects: (1) decreasing SLC16A11 expression in liver and (2) disrupting a key interaction with basigin, thereby reducing cell-surface localization. Both independent mechanisms reduce SLC16A11 function and suggest SLC16A11 is the causal gene at this locus. To gain insight into how SLC16A11 disruption impacts T2D risk, we demonstrate that SLC16A11 is a proton-coupled monocarboxylate transporter and that genetic perturbation of SLC16A11 induces changes in fatty acid and lipid metabolism that are associated with increased T2D risk. Our findings suggest that increasing SLC16A11 function could be therapeutically beneficial for T2D. VIDEO ABSTRACT.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Basigin/metabolism , Cell Membrane/metabolism , Chromosomes, Human, Pair 17/metabolism , Gene Knockdown Techniques , Haplotypes , Hepatocytes/metabolism , Heterozygote , Histone Code , Humans , Liver/metabolism , Models, Molecular , Monocarboxylic Acid Transporters/chemistry
9.
Cell ; 164(5): 884-95, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26919427

ABSTRACT

Ischemic preconditioning is the phenomenon whereby brief periods of sublethal ischemia protect against a subsequent, more prolonged, ischemic insult. In remote ischemic preconditioning (RIPC), ischemia to one organ protects others organs at a distance. We created mouse models to ask if inhibition of the alpha-ketoglutarate (αKG)-dependent dioxygenase Egln1, which senses oxygen and regulates the hypoxia-inducible factor (HIF) transcription factor, could suffice to mediate local and remote ischemic preconditioning. Using somatic gene deletion and a pharmacological inhibitor, we found that inhibiting Egln1 systemically or in skeletal muscles protects mice against myocardial ischemia-reperfusion (I/R) injury. Parabiosis experiments confirmed that RIPC in this latter model was mediated by a secreted factor. Egln1 loss causes accumulation of circulating αKG, which drives hepatic production and secretion of kynurenic acid (KYNA) that is necessary and sufficient to mediate cardiac ischemic protection in this setting.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Ischemic Preconditioning , Ketoglutaric Acids/metabolism , Animals , Ischemia/prevention & control , Kynurenic Acid/metabolism , Liver/metabolism , Mice , Models, Animal , Myocardial Reperfusion Injury/prevention & control , Parabiosis
10.
Cell ; 167(1): 171-186.e15, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27641501

ABSTRACT

While acute myeloid leukemia (AML) comprises many disparate genetic subtypes, one shared hallmark is the arrest of leukemic myeloblasts at an immature and self-renewing stage of development. Therapies that overcome differentiation arrest represent a powerful treatment strategy. We leveraged the observation that the majority of AML, despite their genetically heterogeneity, share in the expression of HoxA9, a gene normally downregulated during myeloid differentiation. Using a conditional HoxA9 model system, we performed a high-throughput phenotypic screen and defined compounds that overcame differentiation blockade. Target identification led to the unanticipated discovery that inhibition of the enzyme dihydroorotate dehydrogenase (DHODH) enables myeloid differentiation in human and mouse AML models. In vivo, DHODH inhibitors reduced leukemic cell burden, decreased levels of leukemia-initiating cells, and improved survival. These data demonstrate the role of DHODH as a metabolic regulator of differentiation and point to its inhibition as a strategy for overcoming differentiation blockade in AML.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Molecular Targeted Therapy , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Cell Differentiation , Dihydroorotate Dehydrogenase , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , High-Throughput Screening Assays , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Myeloid Cells/pathology , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Pyrimidines/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/isolation & purification , Small Molecule Libraries/therapeutic use , Xenograft Model Antitumor Assays
11.
Cell ; 163(6): 1413-27, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26607793

ABSTRACT

Th17 cells play a critical role in host defense against extracellular pathogens and tissue homeostasis but can induce autoimmunity. The mechanisms implicated in balancing "pathogenic" and "non-pathogenic" Th17 cell states remain largely unknown. We used single-cell RNA-seq to identify CD5L/AIM as a regulator expressed in non-pathogenic, but not in pathogenic Th17 cells. Although CD5L does not affect Th17 differentiation, it is a functional switch that regulates the pathogenicity of Th17 cells. Loss of CD5L converts non-pathogenic Th17 cells into pathogenic cells that induce autoimmunity. CD5L mediates this effect by modulating the intracellular lipidome, altering fatty acid composition and restricting cholesterol biosynthesis and, thus, ligand availability for Rorγt, the master transcription factor of Th17 cells. Our study identifies CD5L as a critical regulator of the Th17 cell functional state and highlights the importance of lipid metabolism in balancing immune protection and disease induced by T cells.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Lipid Metabolism , Receptors, Immunologic/metabolism , Th17 Cells/pathology , Animals , Cell Differentiation , Central Nervous System/pathology , Cholesterol/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/immunology , Fatty Acids, Unsaturated/metabolism , Humans , Lymph Nodes/pathology , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Receptors, Scavenger , Single-Cell Analysis , Th17 Cells/immunology
12.
Nature ; 626(7998): 419-426, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38052229

ABSTRACT

Determining the structure and phenotypic context of molecules detected in untargeted metabolomics experiments remains challenging. Here we present reverse metabolomics as a discovery strategy, whereby tandem mass spectrometry spectra acquired from newly synthesized compounds are searched for in public metabolomics datasets to uncover phenotypic associations. To demonstrate the concept, we broadly synthesized and explored multiple classes of metabolites in humans, including N-acyl amides, fatty acid esters of hydroxy fatty acids, bile acid esters and conjugated bile acids. Using repository-scale analysis1,2, we discovered that some conjugated bile acids are associated with inflammatory bowel disease (IBD). Validation using four distinct human IBD cohorts showed that cholic acids conjugated to Glu, Ile/Leu, Phe, Thr, Trp or Tyr are increased in Crohn's disease. Several of these compounds and related structures affected pathways associated with IBD, such as interferon-γ production in CD4+ T cells3 and agonism of the pregnane X receptor4. Culture of bacteria belonging to the Bifidobacterium, Clostridium and Enterococcus genera produced these bile amidates. Because searching repositories with tandem mass spectrometry spectra has only recently become possible, this reverse metabolomics approach can now be used as a general strategy to discover other molecules from human and animal ecosystems.


Subject(s)
Amides , Bile Acids and Salts , Esters , Fatty Acids , Metabolomics , Animals , Humans , Bifidobacterium/metabolism , Bile Acids and Salts/chemistry , Bile Acids and Salts/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Clostridium/metabolism , Cohort Studies , Crohn Disease/metabolism , Enterococcus/metabolism , Esters/chemistry , Esters/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , Inflammatory Bowel Diseases/metabolism , Metabolomics/methods , Phenotype , Pregnane X Receptor/metabolism , Reproducibility of Results , Tandem Mass Spectrometry , Amides/chemistry , Amides/metabolism
13.
Nature ; 633(8031): 878-886, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39294375

ABSTRACT

Persistent colonization and outgrowth of potentially pathogenic organisms in the intestine can result from long-term antibiotic use or inflammatory conditions, and may perpetuate dysregulated immunity and tissue damage1,2. Gram-negative Enterobacteriaceae gut pathobionts are particularly recalcitrant to conventional antibiotic treatment3,4, although an emerging body of evidence suggests that manipulation of the commensal microbiota may be a practical alternative therapeutic strategy5-7. Here we isolated and down-selected commensal bacterial consortia from stool samples from healthy humans that could strongly and specifically suppress intestinal Enterobacteriaceae. One of the elaborated consortia, comprising 18 commensal strains, effectively controlled ecological niches by regulating gluconate availability, thereby re-establishing colonization resistance and alleviating Klebsiella- and Escherichia-driven intestinal inflammation in mice. Harnessing these activities in the form of live bacterial therapies may represent a promising solution to combat the growing threat of proinflammatory, antimicrobial-resistant Enterobacteriaceae infection.


Subject(s)
Enterobacteriaceae , Feces , Gastrointestinal Microbiome , Klebsiella , Symbiosis , Animals , Mice , Humans , Female , Klebsiella/isolation & purification , Feces/microbiology , Male , Enterobacteriaceae Infections/microbiology , Inflammation/microbiology , Inflammation/immunology , Intestines/microbiology , Mice, Inbred C57BL
14.
Cell ; 158(6): 1309-1323, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25215489

ABSTRACT

The balance between oxidative and nonoxidative glucose metabolism is essential for a number of pathophysiological processes. By deleting enzymes that affect aerobic glycolysis with different potencies, we examine how modulating glucose metabolism specifically affects hematopoietic and leukemic cell populations. We find that a deficiency in the M2 pyruvate kinase isoform (PKM2) reduces the levels of metabolic intermediates important for biosynthesis and impairs progenitor function without perturbing hematopoietic stem cells (HSCs), whereas lactate dehydrogenase A (LDHA) deletion significantly inhibits the function of both HSCs and progenitors during hematopoiesis. In contrast, leukemia initiation by transforming alleles putatively affecting either HSCs or progenitors is inhibited in the absence of either PKM2 or LDHA, indicating that the cell-state-specific responses to metabolic manipulation in hematopoiesis do not apply to the setting of leukemia. This finding suggests that fine-tuning the level of glycolysis may be explored therapeutically for treating leukemia while preserving HSC function.


Subject(s)
Glycolysis , Hematopoiesis , Leukemia/metabolism , Animals , Gene Deletion , Hematopoietic Stem Cells/metabolism , Humans , Isoenzymes/metabolism , L-Lactate Dehydrogenase/metabolism , Lactate Dehydrogenase 5 , Mice , Mice, Congenic , Mice, Inbred C57BL , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism
15.
Cell ; 156(1-2): 317-331, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24439385

ABSTRACT

Ferroptosis is a form of nonapoptotic cell death for which key regulators remain unknown. We sought a common mediator for the lethality of 12 ferroptosis-inducing small molecules. We used targeted metabolomic profiling to discover that depletion of glutathione causes inactivation of glutathione peroxidases (GPXs) in response to one class of compounds and a chemoproteomics strategy to discover that GPX4 is directly inhibited by a second class of compounds. GPX4 overexpression and knockdown modulated the lethality of 12 ferroptosis inducers, but not of 11 compounds with other lethal mechanisms. In addition, two representative ferroptosis inducers prevented tumor growth in xenograft mouse tumor models. Sensitivity profiling in 177 cancer cell lines revealed that diffuse large B cell lymphomas and renal cell carcinomas are particularly susceptible to GPX4-regulated ferroptosis. Thus, GPX4 is an essential regulator of ferroptotic cancer cell death.


Subject(s)
Carbolines/pharmacology , Cell Death/drug effects , Glutathione Peroxidase/antagonists & inhibitors , Piperazines/pharmacology , Animals , Carcinoma, Renal Cell/drug therapy , Cell Line, Tumor , Gene Knockdown Techniques , Glutathione/metabolism , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Heterografts , Humans , Lymphoma, B-Cell/drug therapy , Mice , Neoplasm Transplantation , Neoplasms/drug therapy , Phospholipid Hydroperoxide Glutathione Peroxidase
16.
Nature ; 616(7956): 339-347, 2023 04.
Article in English | MEDLINE | ID: mdl-36991126

ABSTRACT

There is a need to develop effective therapies for pancreatic ductal adenocarcinoma (PDA), a highly lethal malignancy with increasing incidence1 and poor prognosis2. Although targeting tumour metabolism has been the focus of intense investigation for more than a decade, tumour metabolic plasticity and high risk of toxicity have limited this anticancer strategy3,4. Here we use genetic and pharmacological approaches in human and mouse in vitro and in vivo models to show that PDA has a distinct dependence on de novo ornithine synthesis from glutamine. We find that this process, which is mediated through ornithine aminotransferase (OAT), supports polyamine synthesis and is required for tumour growth. This directional OAT activity is usually largely restricted to infancy and contrasts with the reliance of most adult normal tissues and other cancer types on arginine-derived ornithine for polyamine synthesis5,6. This dependency associates with arginine depletion in the PDA tumour microenvironment and is driven by mutant KRAS. Activated KRAS induces the expression of OAT and polyamine synthesis enzymes, leading to alterations in the transcriptome and open chromatin landscape in PDA tumour cells. The distinct dependence of PDA, but not normal tissue, on OAT-mediated de novo ornithine synthesis provides an attractive therapeutic window for treating patients with pancreatic cancer with minimal toxicity.


Subject(s)
Ornithine-Oxo-Acid Transaminase , Pancreatic Neoplasms , Polyamines , Animals , Humans , Mice , Arginine/deficiency , Arginine/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Ornithine/biosynthesis , Ornithine/metabolism , Ornithine-Oxo-Acid Transaminase/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Polyamines/metabolism , Tumor Microenvironment
17.
Cell ; 155(2): 397-409, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24120138

ABSTRACT

The pyruvate kinase M2 isoform (PKM2) is expressed in cancer and plays a role in regulating anabolic metabolism. To determine whether PKM2 is required for tumor formation or growth, we generated mice with a conditional allele that abolishes PKM2 expression without disrupting PKM1 expression. PKM2 deletion accelerated mammary tumor formation in a Brca1-loss-driven model of breast cancer. PKM2 null tumors displayed heterogeneous PKM1 expression, with PKM1 found in nonproliferating tumor cells and no detectable pyruvate kinase expression in proliferating cells. This suggests that PKM2 is not necessary for tumor cell proliferation and implies that the inactive state of PKM2 is associated with the proliferating cell population within tumors, whereas nonproliferating tumor cells require active pyruvate kinase. Consistent with these findings, variable PKM2 expression and heterozygous PKM2 mutations are found in human tumors. These data suggest that regulation of PKM2 activity supports the different metabolic requirements of proliferating and nonproliferating tumor cells.


Subject(s)
Breast Neoplasms/metabolism , Gene Deletion , Mammary Neoplasms, Experimental/metabolism , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Animals , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Exons , Female , Gene Knockout Techniques , Heterografts , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Mutagenesis , Mutation , Neoplasm Metastasis , Neoplasm Transplantation , RNA Splicing
18.
Nature ; 603(7903): 907-912, 2022 03.
Article in English | MEDLINE | ID: mdl-35296854

ABSTRACT

The microbiota modulates gut immune homeostasis. Bacteria influence the development and function of host immune cells, including T helper cells expressing interleukin-17A (TH17 cells). We previously reported that the bile acid metabolite 3-oxolithocholic acid (3-oxoLCA) inhibits TH17 cell differentiation1. Although it was suggested that gut-residing bacteria produce 3-oxoLCA, the identity of such bacteria was unknown, and it was unclear whether 3-oxoLCA and other immunomodulatory bile acids are associated with inflammatory pathologies in humans. Here we identify human gut bacteria and corresponding enzymes that convert the secondary bile acid lithocholic acid into 3-oxoLCA as well as the abundant gut metabolite isolithocholic acid (isoLCA). Similar to 3-oxoLCA, isoLCA suppressed TH17 cell differentiation by inhibiting retinoic acid receptor-related orphan nuclear receptor-γt, a key TH17-cell-promoting transcription factor. The levels of both 3-oxoLCA and isoLCA and the 3α-hydroxysteroid dehydrogenase genes that are required for their biosynthesis were significantly reduced in patients with inflammatory bowel disease. Moreover, the levels of these bile acids were inversely correlated with the expression of TH17-cell-associated genes. Overall, our data suggest that bacterially produced bile acids inhibit TH17 cell function, an activity that may be relevant to the pathophysiology of inflammatory disorders such as inflammatory bowel disease.


Subject(s)
Bacteria , Bile Acids and Salts , Inflammatory Bowel Diseases , Bacteria/metabolism , Cell Differentiation , Gastrointestinal Tract/microbiology , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Interleukin-17 , Lithocholic Acid/metabolism , Lithocholic Acid/pharmacology , Th17 Cells
19.
Mol Cell ; 73(5): 1001-1014.e8, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30527540

ABSTRACT

In Parkinson's disease (PD), α-synuclein (αS) pathologically impacts the brain, a highly lipid-rich organ. We investigated how alterations in αS or lipid/fatty acid homeostasis affect each other. Lipidomic profiling of human αS-expressing yeast revealed increases in oleic acid (OA, 18:1), diglycerides, and triglycerides. These findings were recapitulated in rodent and human neuronal models of αS dyshomeostasis (overexpression; patient-derived triplication or E46K mutation; E46K mice). Preventing lipid droplet formation or augmenting OA increased αS yeast toxicity; suppressing the OA-generating enzyme stearoyl-CoA-desaturase (SCD) was protective. Genetic or pharmacological SCD inhibition ameliorated toxicity in αS-overexpressing rat neurons. In a C. elegans model, SCD knockout prevented αS-induced dopaminergic degeneration. Conversely, we observed detrimental effects of OA on αS homeostasis: in human neural cells, excess OA caused αS inclusion formation, which was reversed by SCD inhibition. Thus, monounsaturated fatty acid metabolism is pivotal for αS-induced neurotoxicity, and inhibiting SCD represents a novel PD therapeutic approach.


Subject(s)
Antiparkinson Agents/pharmacology , Drug Discovery/methods , Enzyme Inhibitors/pharmacology , Lipid Metabolism/drug effects , Metabolomics/methods , Neurons/drug effects , Parkinson Disease/drug therapy , Stearoyl-CoA Desaturase/antagonists & inhibitors , alpha-Synuclein/toxicity , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , Cell Line , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Diglycerides/metabolism , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/enzymology , Dopaminergic Neurons/pathology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/enzymology , Induced Pluripotent Stem Cells/pathology , Lipid Droplets/drug effects , Lipid Droplets/enzymology , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Nerve Degeneration , Neural Stem Cells/drug effects , Neural Stem Cells/enzymology , Neural Stem Cells/pathology , Neurons/enzymology , Neurons/pathology , Oleic Acid/metabolism , Parkinson Disease/enzymology , Parkinson Disease/genetics , Parkinson Disease/pathology , Rats, Sprague-Dawley , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Stearoyl-CoA Desaturase/metabolism , Triglycerides/metabolism , alpha-Synuclein/genetics
20.
Proc Natl Acad Sci U S A ; 121(28): e2318691121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38968121

ABSTRACT

Dietary lipids play an essential role in regulating the function of the gut microbiota and gastrointestinal tract, and these luminal interactions contribute to mediating host metabolism. Palmitic Acid Hydroxy Stearic Acids (PAHSAs) are a family of lipids with antidiabetic and anti-inflammatory properties, but whether the gut microbiota contributes to their beneficial effects on host metabolism is unknown. Here, we report that treating chow-fed female and male germ-free (GF) mice with PAHSAs improves glucose tolerance, but these effects are lost upon high fat diet (HFD) feeding. However, transfer of feces from PAHSA-treated, but not vehicle-treated, chow-fed conventional mice increases insulin sensitivity in HFD-fed GF mice. Thus, the gut microbiota is necessary for, and can transmit, the insulin-sensitizing effects of PAHSAs in HFD-fed GF male mice. Analyses of the cecal metagenome and lipidome of PAHSA-treated mice identified multiple lipid species that associate with the gut commensal Bacteroides thetaiotaomicron (Bt) and with insulin sensitivity resulting from PAHSA treatment. Supplementing live, and to some degree, heat-killed Bt to HFD-fed female mice prevented weight gain, reduced adiposity, improved glucose tolerance, fortified the colonic mucus barrier and reduced systemic inflammation compared to HFD-fed controls. These effects were not observed in HFD-fed male mice. Furthermore, ovariectomy partially reversed the beneficial Bt effects on host metabolism, indicating a role for sex hormones in mediating the Bt probiotic effects. Altogether, these studies highlight the fact that PAHSAs can modulate the gut microbiota and that the microbiota is necessary for the beneficial metabolic effects of PAHSAs in HFD-fed mice.


Subject(s)
Diet, High-Fat , Gastrointestinal Microbiome , Insulin Resistance , Obesity , Animals , Male , Female , Mice , Gastrointestinal Microbiome/drug effects , Obesity/metabolism , Obesity/microbiology , Obesity/etiology , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Stearic Acids/metabolism , Palmitic Acid/metabolism , Feces/microbiology , Mice, Obese
SELECTION OF CITATIONS
SEARCH DETAIL