Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.161
Filter
Add more filters

Publication year range
1.
Cell ; 177(3): 683-696.e18, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30929902

ABSTRACT

Microbiota and intestinal epithelium restrict pathogen growth by rapid nutrient consumption. We investigated how pathogens circumvent this obstacle to colonize the host. Utilizing enteropathogenic E. coli (EPEC), we show that host-attached bacteria obtain nutrients from infected host cell in a process we termed host nutrient extraction (HNE). We identified an inner-membrane protein complex, henceforth termed CORE, as necessary and sufficient for HNE. The CORE is a key component of the EPEC injectisome, however, here we show that it supports the formation of an alternative structure, composed of membranous nanotubes, protruding from the EPEC surface to directly contact the host. The injectisome and flagellum are evolutionarily related, both containing conserved COREs. Remarkably, CORE complexes of diverse ancestries, including distant flagellar COREs, could rescue HNE capacity of EPEC lacking its native CORE. Our results support the notion that HNE is a widespread virulence strategy, enabling pathogens to thrive in competitive niches.


Subject(s)
Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Proteins/metabolism , Nutrients/metabolism , Amino Acids/metabolism , Bacterial Adhesion/physiology , Enteropathogenic Escherichia coli/growth & development , Enteropathogenic Escherichia coli/metabolism , Fluoresceins/metabolism , HeLa Cells , Humans , Membrane Proteins/metabolism , Microscopy, Electron, Scanning , Microscopy, Fluorescence
2.
Cell ; 178(6): 1299-1312.e29, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31474368

ABSTRACT

Metformin is the first-line therapy for treating type 2 diabetes and a promising anti-aging drug. We set out to address the fundamental question of how gut microbes and nutrition, key regulators of host physiology, affect the effects of metformin. Combining two tractable genetic models, the bacterium E. coli and the nematode C. elegans, we developed a high-throughput four-way screen to define the underlying host-microbe-drug-nutrient interactions. We show that microbes integrate cues from metformin and the diet through the phosphotransferase signaling pathway that converges on the transcriptional regulator Crp. A detailed experimental characterization of metformin effects downstream of Crp in combination with metabolic modeling of the microbiota in metformin-treated type 2 diabetic patients predicts the production of microbial agmatine, a regulator of metformin effects on host lipid metabolism and lifespan. Our high-throughput screening platform paves the way for identifying exploitable drug-nutrient-microbiome interactions to improve host health and longevity through targeted microbiome therapies. VIDEO ABSTRACT.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Gastrointestinal Microbiome/drug effects , Host Microbial Interactions/drug effects , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Agmatine/metabolism , Animals , Caenorhabditis elegans/microbiology , Cyclic AMP Receptor Protein , Escherichia coli/drug effects , Escherichia coli/genetics , Humans , Hypoglycemic Agents/pharmacology , Lipid Metabolism/drug effects , Longevity/drug effects , Metformin/pharmacology , Nutrients/metabolism
3.
Annu Rev Cell Dev Biol ; 35: 453-475, 2019 10 06.
Article in English | MEDLINE | ID: mdl-31283377

ABSTRACT

Macroautophagy is an intracellular degradation system that delivers diverse cytoplasmic materials to lysosomes via autophagosomes. Recent advances have enabled identification of several selective autophagy substrates and receptors, greatly expanding our understanding of the cellular functions of autophagy. In this review, we describe the diverse cellular functions of macroautophagy, including its essential contribution to metabolic adaptation and cellular homeostasis. We also discuss emerging findings on the mechanisms and functions of various types of selective autophagy.


Subject(s)
Autophagosomes/metabolism , Autophagy/genetics , Endoplasmic Reticulum/metabolism , Lysosomes/metabolism , Mitochondria/metabolism , Animals , Autophagosomes/enzymology , Autophagosomes/microbiology , Autophagy/physiology , Endoplasmic Reticulum/physiology , Homeostasis/genetics , Homeostasis/physiology , Humans , Lysosomes/pathology , Mitochondria/pathology , Nutrients/deficiency , Nutrients/metabolism , Peroxisomes/metabolism , Peroxisomes/physiology
4.
Nat Immunol ; 21(7): 790-801, 2020 07.
Article in English | MEDLINE | ID: mdl-32424361

ABSTRACT

Plasmodium parasite-specific antibodies are critical for protection against malaria, yet the development of long-lived and effective humoral immunity against Plasmodium takes many years and multiple rounds of infection and cure. Here, we report that the rapid development of short-lived plasmablasts during experimental malaria unexpectedly hindered parasite control by impeding germinal center responses. Metabolic hyperactivity of plasmablasts resulted in nutrient deprivation of the germinal center reaction, limiting the generation of memory B cell and long-lived plasma cell responses. Therapeutic administration of a single amino acid to experimentally infected mice was sufficient to overcome the metabolic constraints imposed by plasmablasts and enhanced parasite clearance and the formation of protective humoral immune memory responses. Thus, our studies not only challenge the current model describing the role and function of blood-stage Plasmodium-induced plasmablasts but they also reveal new targets and strategies to improve anti-Plasmodium humoral immunity.


Subject(s)
Immunity, Humoral , Malaria/immunology , Plasma Cells/metabolism , Plasmodium falciparum/immunology , Adolescent , Adult , Amino Acids/administration & dosage , Amino Acids/metabolism , Animals , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Antibodies, Protozoan/metabolism , Antimalarials/administration & dosage , DNA, Protozoan/isolation & purification , Disease Models, Animal , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , Host-Parasite Interactions/immunology , Humans , Malaria/blood , Malaria/drug therapy , Malaria/parasitology , Mice , Mice, Transgenic , Middle Aged , Nutrients/metabolism , Plasma Cells/immunology , Plasma Cells/parasitology , Plasmodium falciparum/genetics , Plasmodium falciparum/isolation & purification , Proof of Concept Study , Young Adult
5.
Nature ; 626(7997): 111-118, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38297171

ABSTRACT

The recovery of top predators is thought to have cascading effects on vegetated ecosystems and their geomorphology1,2, but the evidence for this remains correlational and intensely debated3,4. Here we combine observational and experimental data to reveal that recolonization of sea otters in a US estuary generates a trophic cascade that facilitates coastal wetland plant biomass and suppresses the erosion of marsh edges-a process that otherwise leads to the severe loss of habitats and ecosystem services5,6. Monitoring of the Elkhorn Slough estuary over several decades suggested top-down control in the system, because the erosion of salt marsh edges has generally slowed with increasing sea otter abundance, despite the consistently increasing physical stress in the system (that is, nutrient loading, sea-level rise and tidal scour7-9). Predator-exclusion experiments in five marsh creeks revealed that sea otters suppress the abundance of burrowing crabs, a top-down effect that cascades to both increase marsh edge strength and reduce marsh erosion. Multi-creek surveys comparing marsh creeks pre- and post-sea otter colonization confirmed the presence of an interaction between the keystone sea otter, burrowing crabs and marsh creeks, demonstrating the spatial generality of predator control of ecosystem edge processes: densities of burrowing crabs and edge erosion have declined markedly in creeks that have high levels of sea otter recolonization. These results show that trophic downgrading could be a strong but underappreciated contributor to the loss of coastal wetlands, and suggest that restoring top predators can help to re-establish geomorphic stability.


Subject(s)
Brachyura , Estuaries , Otters , Predatory Behavior , Soil Erosion , Wetlands , Animals , Biomass , Brachyura/physiology , Otters/physiology , United States , Plants , Sea Level Rise , Tidal Waves , Nutrients/metabolism , Food Chain
6.
Nature ; 621(7978): 330-335, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37587345

ABSTRACT

Projected responses of ocean net primary productivity to climate change are highly uncertain1. Models suggest that the climate sensitivity of phytoplankton nutrient limitation in the low-latitude Pacific Ocean plays a crucial role1-3, but this is poorly constrained by observations4. Here we show that changes in physical forcing drove coherent fluctuations in the strength of equatorial Pacific iron limitation through multiple El Niño/Southern Oscillation (ENSO) cycles, but that this was overestimated twofold by a state-of-the-art climate model. Our assessment was enabled by first using a combination of field nutrient-addition experiments, proteomics and above-water hyperspectral radiometry to show that phytoplankton physiological responses to iron limitation led to approximately threefold changes in chlorophyll-normalized phytoplankton fluorescence. We then exploited the >18-year satellite fluorescence record to quantify climate-induced nutrient limitation variability. Such synoptic constraints provide a powerful approach for benchmarking the realism of model projections of net primary productivity to climate changes.


Subject(s)
Climate Models , El Nino-Southern Oscillation , Iron , Chlorophyll/metabolism , Climate Change , Fluorescence , Iron/metabolism , Nutrients/metabolism , Pacific Ocean , Phytoplankton/metabolism , Proteomics , Radiometry , Satellite Imagery
7.
Nature ; 619(7968): 102-111, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37258676

ABSTRACT

The stability and resilience of the Earth system and human well-being are inseparably linked1-3, yet their interdependencies are generally under-recognized; consequently, they are often treated independently4,5. Here, we use modelling and literature assessment to quantify safe and just Earth system boundaries (ESBs) for climate, the biosphere, water and nutrient cycles, and aerosols at global and subglobal scales. We propose ESBs for maintaining the resilience and stability of the Earth system (safe ESBs) and minimizing exposure to significant harm to humans from Earth system change (a necessary but not sufficient condition for justice)4. The stricter of the safe or just boundaries sets the integrated safe and just ESB. Our findings show that justice considerations constrain the integrated ESBs more than safety considerations for climate and atmospheric aerosol loading. Seven of eight globally quantified safe and just ESBs and at least two regional safe and just ESBs in over half of global land area are already exceeded. We propose that our assessment provides a quantitative foundation for safeguarding the global commons for all people now and into the future.


Subject(s)
Climate Change , Earth, Planet , Environmental Justice , Internationality , Safety , Humans , Aerosols/metabolism , Climate , Water/metabolism , Nutrients/metabolism , Safety/legislation & jurisprudence , Safety/standards
8.
Nature ; 621(7977): 171-178, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37648867

ABSTRACT

Triacylglycerols (TAGs) are the main source of stored energy in the body, providing an important substrate pool for mitochondrial beta-oxidation. Imbalances in the amount of TAGs are associated with obesity, cardiac disease and various other pathologies1,2. In humans, TAGs are synthesized from excess, coenzyme A-conjugated fatty acids by diacylglycerol O-acyltransferases (DGAT1 and DGAT2)3. In other organisms, this activity is complemented by additional enzymes4, but whether such alternative pathways exist in humans remains unknown. Here we disrupt the DGAT pathway in haploid human cells and use iterative genetics to reveal an unrelated TAG-synthesizing system composed of a protein we called DIESL (also known as TMEM68, an acyltransferase of previously unknown function) and its regulator TMX1. Mechanistically, TMX1 binds to and controls DIESL at the endoplasmic reticulum, and loss of TMX1 leads to the unconstrained formation of DIESL-dependent lipid droplets. DIESL is an autonomous TAG synthase, and expression of human DIESL in Escherichia coli endows this organism with the ability to synthesize TAG. Although both DIESL and the DGATs function as diacylglycerol acyltransferases, they contribute to the cellular TAG pool under specific conditions. Functionally, DIESL synthesizes TAG at the expense of membrane phospholipids and maintains mitochondrial function during periods of extracellular lipid starvation. In mice, DIESL deficiency impedes rapid postnatal growth and affects energy homeostasis during changes in nutrient availability. We have therefore identified an alternative TAG biosynthetic pathway driven by DIESL under potent control by TMX1.


Subject(s)
Acyltransferases , Triglycerides , Animals , Humans , Mice , Acyltransferases/metabolism , Coenzyme A/metabolism , Diacylglycerol O-Acyltransferase/metabolism , Escherichia coli/metabolism , Homeostasis , Triglycerides/biosynthesis , Energy Metabolism , Nutrients/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism
9.
Nature ; 623(7985): 167-174, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37757899

ABSTRACT

During nutrient stress, macroautophagy degrades cellular macromolecules, thereby providing biosynthetic building blocks while simultaneously remodelling the proteome1,2. Although the machinery responsible for initiation of macroautophagy has been well characterized3,4, our understanding of the extent to which individual proteins, protein complexes and organelles are selected for autophagic degradation, and the underlying targeting mechanisms, is limited. Here we use orthogonal proteomic strategies to provide a spatial proteome census of autophagic cargo during nutrient stress in mammalian cells. We find that macroautophagy has selectivity for recycling membrane-bound organelles (principally Golgi and endoplasmic reticulum). Through autophagic cargo prioritization, we identify a complex of membrane-embedded proteins, YIPF3 and YIPF4, as receptors for Golgiphagy. During nutrient stress, YIPF3 and YIPF4 interact with ATG8 proteins through LIR motifs and are mobilized into autophagosomes that traffic to lysosomes in a process that requires the canonical autophagic machinery. Cells lacking YIPF3 or YIPF4 are selectively defective in elimination of a specific cohort of Golgi membrane proteins during nutrient stress. Moreover, YIPF3 and YIPF4 play an analogous role in Golgi remodelling during programmed conversion of stem cells to the neuronal lineage in vitro. Collectively, the findings of this study reveal prioritization of membrane protein cargo during nutrient-stress-dependent proteome remodelling and identify a Golgi remodelling pathway that requires membrane-embedded receptors.


Subject(s)
Autophagy , Golgi Apparatus , Membrane Proteins , Nutrients , Proteome , Animals , Autophagy/physiology , Autophagy-Related Protein 8 Family/metabolism , Endoplasmic Reticulum , Golgi Apparatus/metabolism , Mammals/metabolism , Membrane Proteins/metabolism , Nutrients/metabolism , Proteome/metabolism , Proteomics
10.
Nature ; 620(7972): 163-171, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37495694

ABSTRACT

An outstanding mystery in biology is why some species, such as the axolotl, can regenerate tissues whereas mammals cannot1. Here, we demonstrate that rapid activation of protein synthesis is a unique feature of the injury response critical for limb regeneration in the axolotl (Ambystoma mexicanum). By applying polysome sequencing, we identify hundreds of transcripts, including antioxidants and ribosome components that are selectively activated at the level of translation from pre-existing messenger RNAs in response to injury. By contrast, protein synthesis is not activated in response to non-regenerative digit amputation in the mouse. We identify the mTORC1 pathway as a key upstream signal that mediates tissue regeneration and translational control in the axolotl. We discover unique expansions in mTOR protein sequence among urodele amphibians. By engineering an axolotl mTOR (axmTOR) in human cells, we show that these changes create a hypersensitive kinase that allows axolotls to maintain this pathway in a highly labile state primed for rapid activation. This change renders axolotl mTOR more sensitive to nutrient sensing, and inhibition of amino acid transport is sufficient to inhibit tissue regeneration. Together, these findings highlight the unanticipated impact of the translatome on orchestrating the early steps of wound healing in a highly regenerative species and provide a missing link in our understanding of vertebrate regenerative potential.


Subject(s)
Ambystoma mexicanum , Biological Evolution , Protein Biosynthesis , Regeneration , TOR Serine-Threonine Kinases , Animals , Humans , Mice , Ambystoma mexicanum/physiology , Amino Acid Sequence , Extremities/physiology , Regeneration/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , TOR Serine-Threonine Kinases/metabolism , Wound Healing , Mechanistic Target of Rapamycin Complex 1/metabolism , Species Specificity , Antioxidants/metabolism , Nutrients/metabolism , Polyribosomes/genetics , Polyribosomes/metabolism
11.
Mol Cell ; 81(18): 3731-3748, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34547236

ABSTRACT

Nutrient supply and demand delineate cell behavior in health and disease. Mammalian cells have developed multiple strategies to secure the necessary nutrients that fuel their metabolic needs. This is more evident upon disruption of homeostasis in conditions such as cancer, when cells display high proliferation rates in energetically challenging conditions where nutritional sources may be scarce. Here, we summarize the main routes of nutrient acquisition that fuel mammalian cells and their implications in tumorigenesis. We argue that the molecular mechanisms of nutrient acquisition not only tip the balance between nutrient supply and demand but also determine cell behavior upon nutrient limitation and energetic stress and contribute to nutrient partitioning and metabolic coordination between different cell types in inflamed or tumorigenic environments.


Subject(s)
Membrane Transport Proteins/metabolism , Neoplasms/metabolism , Nutrients/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Biological Transport/physiology , Carcinogenesis/metabolism , Cell Membrane/metabolism , Homeostasis/physiology , Humans , Solute Carrier Proteins/metabolism
12.
Nature ; 611(7935): 301-305, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36323777

ABSTRACT

Enrichment of nutrients and loss of herbivores are assumed to cause a loss of plant diversity in grassland ecosystems because they increase plant cover, which leads to a decrease of light in the understory1-3. Empirical tests of the role of competition for light in natural systems are based on indirect evidence, and have been a topic of debate for the last 40 years. Here we show that experimentally restoring light to understory plants in a natural grassland mitigates the loss of plant diversity that is caused by either nutrient enrichment or the absence of mammalian herbivores. The initial effect of light addition on restoring diversity under fertilization was transitory and outweighed by the greater effect of herbivory on light levels, indicating that herbivory is a major factor that controls diversity, partly through light. Our results provide direct experimental evidence, in a natural system, that competition for light is a key mechanism that contributes to the loss of biodiversity after cessation of mammalian herbivory. Our findings also show that the effects of herbivores can outpace the effects of fertilization on competition for light. Management practices that target maintaining grazing by native or domestic herbivores could therefore have applications in protecting biodiversity in grassland ecosystems, because they alleviate competition for light in the understory.


Subject(s)
Biodiversity , Herbivory , Light , Plants , Animals , Grassland , Mammals/physiology , Nutrients/metabolism , Plants/classification , Plants/metabolism , Plants/radiation effects , Fertilizers
13.
Nature ; 607(7919): 610-616, 2022 07.
Article in English | MEDLINE | ID: mdl-35831510

ABSTRACT

Mechanistic target of rapamycin complex 1 (mTORC1) controls growth by regulating anabolic and catabolic processes in response to environmental cues, including nutrients1,2. Amino acids signal to mTORC1 through the Rag GTPases, which are regulated by several protein complexes, including GATOR1 and GATOR2. GATOR2, which has five components (WDR24, MIOS, WDR59, SEH1L and SEC13), is required for amino acids to activate mTORC1 and interacts with the leucine and arginine sensors SESN2 and CASTOR1, respectively3-5. Despite this central role in nutrient sensing, GATOR2 remains mysterious as its subunit stoichiometry, biochemical function and structure are unknown. Here we used cryo-electron microscopy to determine the three-dimensional structure of the human GATOR2 complex. We found that GATOR2 adopts a large (1.1 MDa), two-fold symmetric, cage-like architecture, supported by an octagonal scaffold and decorated with eight pairs of WD40 ß-propellers. The scaffold contains two WDR24, four MIOS and two WDR59 subunits circularized via two distinct types of junction involving non-catalytic RING domains and α-solenoids. Integration of SEH1L and SEC13 into the scaffold through ß-propeller blade donation stabilizes the GATOR2 complex and reveals an evolutionary relationship to the nuclear pore and membrane-coating complexes6. The scaffold orients the WD40 ß-propeller dimers, which mediate interactions with SESN2, CASTOR1 and GATOR1. Our work reveals the structure of an essential component of the nutrient-sensing machinery and provides a foundation for understanding the function of GATOR2 within the mTORC1 pathway.


Subject(s)
Amino Acids , Cryoelectron Microscopy , Multiprotein Complexes , Nutrients , Protein Subunits , Humans , Amino Acids/metabolism , Arginine , Carrier Proteins , Leucine , Mechanistic Target of Rapamycin Complex 1/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Nutrients/metabolism , Protein Domains , Protein Subunits/chemistry , Protein Subunits/metabolism , Proteins
14.
Nature ; 611(7934): 81-87, 2022 11.
Article in English | MEDLINE | ID: mdl-36224391

ABSTRACT

To meet the growing food demand while addressing the multiple challenges of exacerbating phosphorus (P) pollution and depleting P rock reserves1-15, P use efficiency (PUE, the ratio of productive P output to P input in a defined system) in crop production needs to be improved. Although many efforts have been devoted to improving nutrient management practices on farms, few studies have examined the historical trajectories of PUE and their socioeconomic and agronomic drivers on a national scale1,2,6,7,11,16,17. Here we present a database of the P budget (the input and output of the crop production system) and PUE by country and by crop type for 1961-2019, and examine the substantial contribution of several drivers for PUE, such as economic development stages and crop portfolios. To address the P management challenges, we found that global PUE in crop production must increase to 68-81%, and recent trends indicate some meaningful progress towards this goal. However, P management challenges and opportunities in croplands vary widely among countries.


Subject(s)
Crop Production , Crops, Agricultural , Phosphorus , Sustainable Development , Crop Production/methods , Crop Production/trends , Crops, Agricultural/classification , Crops, Agricultural/metabolism , Farms , Nutrients/metabolism , Phosphorus/metabolism , Sustainable Development/trends , Internationality , Socioeconomic Factors , Databases, Factual
15.
Nature ; 608(7922): 374-380, 2022 08.
Article in English | MEDLINE | ID: mdl-35831501

ABSTRACT

Food and water are rewarding in part because they satisfy our internal needs1,2. Dopaminergic neurons in the ventral tegmental area (VTA) are activated by gustatory rewards3-5, but how animals learn to associate these oral cues with the delayed physiological effects of ingestion is unknown. Here we show that individual dopaminergic neurons in the VTA respond to detection of nutrients or water at specific stages of ingestion. A major subset of dopaminergic neurons tracks changes in systemic hydration that occur tens of minutes after thirsty mice drink water, whereas different dopaminergic neurons respond to nutrients in the gastrointestinal tract. We show that information about fluid balance is transmitted to the VTA by a hypothalamic pathway and then re-routed to downstream circuits that track the oral, gastrointestinal and post-absorptive stages of ingestion. To investigate the function of these signals, we used a paradigm in which a fluid's oral and post-absorptive effects can be independently manipulated and temporally separated. We show that mice rapidly learn to prefer one fluid over another based solely on its rehydrating ability and that this post-ingestive learning is prevented if dopaminergic neurons in the VTA are selectively silenced after consumption. These findings reveal that the midbrain dopamine system contains subsystems that track different modalities and stages of ingestion, on timescales from seconds to tens of minutes, and that this information is used to drive learning about the consequences of ingestion.


Subject(s)
Dopamine , Dopaminergic Neurons , Hypothalamus , Neural Pathways , Nutrients , Organism Hydration Status , Ventral Tegmental Area , Animals , Cues , Digestion , Dopamine/metabolism , Dopaminergic Neurons/physiology , Eating , Gastrointestinal Tract/metabolism , Hypothalamus/cytology , Hypothalamus/physiology , Mesencephalon/cytology , Mesencephalon/physiology , Mice , Nutrients/metabolism , Organism Hydration Status/drug effects , Reward , Time Factors , Ventral Tegmental Area/cytology , Ventral Tegmental Area/physiology , Water/metabolism , Water/pharmacology , Water-Electrolyte Balance
16.
Genes Dev ; 34(19-20): 1345-1358, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32912902

ABSTRACT

The uptake of macromolecules and cellular debris through macropinocytosis has emerged as an important nutrient acquisition strategy of cancer cells. Genetic alterations commonly found in human cancers (e.g. mutations in KRAS or loss of PTEN) have been shown to increase macropinocytosis. To identify additional effectors that enable cell growth dependent on the uptake of extracellular proteins, pancreatic ductal adenocarcinoma (PDA) cells were selected for growth in medium where extracellular albumin was the obligate source of the essential amino acid leucine. Analysis of global changes in chromatin availability and gene expression revealed that PDA cells selected under these conditions exhibited elevated activity of the transcriptional activators Yap/Taz. Knockout of Yap/Taz prevented growth of PDA cells in leucine-deficient medium, but not in complete medium. Furthermore, constitutively active forms of Yap or Taz were sufficient to stimulate macropinocytosis of extracellular protein. In addition to promoting the uptake of plasma proteins, Yap/Taz also promoted the scavenging of apoptotic cell bodies and necrotic debris by PDA cells. The Yap/Taz transcriptional target Axl was found to be essential for cell growth dependent on the uptake of dead cells and cell debris. Together, these studies suggest that the Hippo pathway effectors Yap and Taz are important transcriptional regulators of endocytic nutrient uptake.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Nutrients/metabolism , Pinocytosis/physiology , Transcription Factors/metabolism , Acyltransferases , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Extracellular Space/metabolism , Humans , Mice , YAP-Signaling Proteins
17.
Development ; 151(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38577841

ABSTRACT

Enteroendocrine cells (EECs) are crucial for sensing ingested nutrients and regulating feeding behavior. How gut microbiota regulate the nutrient-sensing EEC activity is unclear. Our transcriptomic analysis demonstrates that commensal microbiota colonization significantly increases the expression of many genes associated with mitochondrial function. Using new methods to image EEC cytoplasmic and mitochondrial Ca2+ activity in live zebrafish, our data revealed that it is dynamically regulated during the EEC development process. Mature EECs display an increased mitochondrial-to-cytoplasmic Ca2+ ratio. Mitochondria are evenly distributed in the cytoplasm of immature EECs. As EECs mature, their mitochondria are highly localized at the basal membrane where EEC vesicle secretion occurs. Conventionalized (CV) EECs, but not germ-free (GF) EECs, exhibit spontaneous low-amplitude Ca2+ fluctuation. The mitochondrial-to-cytoplasmic Ca2+ ratio is significantly higher in CV EECs. Nutrient stimulants, such as fatty acid, increase cytoplasmic Ca2+ in a subset of EECs and promote a sustained mitochondrial Ca2+ and ATP increase. However, the nutrient-induced EEC mitochondrial activation is nearly abolished in GF zebrafish. Together, our study reveals that commensal microbiota are crucial in supporting EEC mitochondrial function and maturation.


Subject(s)
Calcium , Enteroendocrine Cells , Gastrointestinal Microbiome , Mitochondria , Zebrafish , Animals , Zebrafish/microbiology , Enteroendocrine Cells/metabolism , Mitochondria/metabolism , Gastrointestinal Microbiome/physiology , Calcium/metabolism , Nutrients/metabolism , Adenosine Triphosphate/metabolism
18.
Plant Cell ; 36(5): 1504-1523, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38163641

ABSTRACT

As an essential nutrient element, phosphorus (P) is primarily acquired and translocated as inorganic phosphate (Pi) by plant roots. Pi is often sequestered in the soil and becomes limited for plant growth. Plants have developed a sophisticated array of adaptive responses, termed P starvation responses, to cope with P deficiency by improving its external acquisition and internal utilization. Over the past 2 to 3 decades, remarkable progress has been made toward understanding how plants sense and respond to changing environmental P. This review provides an overview of the molecular mechanisms that regulate or coordinate P starvation responses, emphasizing P transport, sensing, and signaling. We present the major players and regulators responsible for Pi uptake and translocation. We then introduce how P is perceived at the root tip, how systemic P signaling is operated, and the mechanisms by which the intracellular P status is sensed and conveyed. Additionally, the recent exciting findings about the influence of P on plant-microbe interactions are highlighted. Finally, the challenges and prospects concerning the interplay between P and other nutrients and strategies to enhance P utilization efficiency are discussed. Insights obtained from this knowledge may guide future research endeavors in sustainable agriculture.


Subject(s)
Phosphorus , Plants , Signal Transduction , Phosphorus/metabolism , Biological Transport , Plants/metabolism , Plant Roots/metabolism , Phosphates/metabolism , Nutrients/metabolism
19.
Nature ; 595(7865): 58-65, 2021 07.
Article in English | MEDLINE | ID: mdl-34194019

ABSTRACT

The natural world provides many examples of multiphase transport and reaction processes that have been optimized by evolution. These phenomena take place at multiple length and time scales and typically include gas-liquid-solid interfaces and capillary phenomena in porous media1,2. Many biological and living systems have evolved to optimize fluidic transport. However, living things are exceptionally complex and very difficult to replicate3-5, and human-made microfluidic devices (which are typically planar and enclosed) are highly limited for multiphase process engineering6-8. Here we introduce the concept of cellular fluidics: a platform of unit-cell-based, three-dimensional structures-enabled by emerging 3D printing methods9,10-for the deterministic control of multiphase flow, transport and reaction processes. We show that flow in these structures can be 'programmed' through architected design of cell type, size and relative density. We demonstrate gas-liquid transport processes such as transpiration and absorption, using evaporative cooling and CO2 capture as examples. We design and demonstrate preferential liquid and gas transport pathways in three-dimensional cellular fluidic devices with capillary-driven and actively pumped liquid flow, and present examples of selective metallization of pre-programmed patterns. Our results show that the design and fabrication of architected cellular materials, coupled with analytical and numerical predictions of steady-state and dynamic behaviour of multiphase interfaces, provide deterministic control of fluidic transport in three dimensions. Cellular fluidics may transform the design space for spatial and temporal control of multiphase transport and reaction processes.


Subject(s)
Cells/metabolism , Microfluidics/instrumentation , Microfluidics/methods , Absorption, Physicochemical , Carbon Dioxide/metabolism , Gases/metabolism , Nutrients/metabolism , Oxygen/metabolism , Plant Transpiration , Videodisc Recording , Water/metabolism
20.
Nature ; 593(7858): 282-288, 2021 05.
Article in English | MEDLINE | ID: mdl-33828302

ABSTRACT

Cancer cells characteristically consume glucose through Warburg metabolism1, a process that forms the basis of tumour imaging by positron emission tomography (PET). Tumour-infiltrating immune cells also rely on glucose, and impaired immune cell metabolism in the tumour microenvironment (TME) contributes to immune evasion by tumour cells2-4. However, whether the metabolism of immune cells is dysregulated in the TME by cell-intrinsic programs or by competition with cancer cells for limited nutrients remains unclear. Here we used PET tracers to measure the access to and uptake of glucose and glutamine by specific cell subsets in the TME. Notably, myeloid cells had the greatest capacity to take up intratumoral glucose, followed by T cells and cancer cells, across a range of cancer models. By contrast, cancer cells showed the highest uptake of glutamine. This distinct nutrient partitioning was programmed in a cell-intrinsic manner through mTORC1 signalling and the expression of genes related to the metabolism of glucose and glutamine. Inhibiting glutamine uptake enhanced glucose uptake across tumour-resident cell types, showing that glutamine metabolism suppresses glucose uptake without glucose being a limiting factor in the TME. Thus, cell-intrinsic programs drive the preferential acquisition of glucose and glutamine by immune and cancer cells, respectively. Cell-selective partitioning of these nutrients could be exploited to develop therapies and imaging strategies to enhance or monitor the metabolic programs and activities of specific cell populations in the TME.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Nutrients/metabolism , Tumor Microenvironment , Animals , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Female , Glucose/metabolism , Glutamine/metabolism , Humans , Lipid Metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasms/immunology , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL