Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
Elife ; 132024 May 24.
Article in English | MEDLINE | ID: mdl-38787918

ABSTRACT

The tumor microenvironment is a determinant of cancer progression and therapeutic efficacy, with nutrient availability playing an important role. Although it is established that the local abundance of specific nutrients defines the metabolic parameters for tumor growth, the factors guiding nutrient availability in tumor compared to normal tissue and blood remain poorly understood. To define these factors in renal cell carcinoma (RCC), we performed quantitative metabolomic and comprehensive lipidomic analyses of tumor interstitial fluid (TIF), adjacent normal kidney interstitial fluid (KIF), and plasma samples collected from patients. TIF nutrient composition closely resembles KIF, suggesting that tissue-specific factors unrelated to the presence of cancer exert a stronger influence on nutrient levels than tumor-driven alterations. Notably, select metabolite changes consistent with known features of RCC metabolism are found in RCC TIF, while glucose levels in TIF are not depleted to levels that are lower than those found in KIF. These findings inform tissue nutrient dynamics in RCC, highlighting a dominant role of non-cancer-driven tissue factors in shaping nutrient availability in these tumors.


Cancer cells convert nutrients into energy differently compared to healthy cells. This difference in metabolism allows them to grow and divide more quickly and sometimes to migrate to different areas of the body. The environment around cancer cells ­ known as the tumor microenvironment ­ contains a variety of different cells and blood vessels, which are bathed in interstitial fluid. This microenvironment provides nutrients for the cancer cells to metabolize, and therefore influences how well a tumor grows and how it might respond to treatment. Recent advances with techniques such as mass spectrometry, which can measure the chemical composition of a substance, have allowed scientists to measure nutrient levels in the tumor microenvironments of mice. However, it has been more difficult to conduct such studies in humans, as well as to compare the tumor microenvironment to the healthy tissue the tumors arose from. Abbott, Ali, Reinfeld et al. aimed to fill this gap in knowledge by using mass spectrometry to measure the nutrient levels in the tumor microenvironment of 55 patients undergoing surgery to remove kidney tumors. Comparing the type and levels of nutrients in the tumor interstitial fluid, the neighboring healthy kidney and the blood showed that nutrients in the tumor and healthy kidney were more similar to each other than those in the blood. For example, both the tumor and healthy kidney interstitial fluids contained less glucose than the blood. However, the difference between nutrient composition in the tumor and healthy kidney interstitial fluids was insignificant, suggesting that the healthy kidney and its tumor share a similar environment. Taken together, the findings indicate that kidney cancer cells must adapt to the nutrients available in the kidney, rather than changing what nutrients are available in the tissue. Future studies will be required to investigate whether this finding also applies to other types of cancer. A better understanding of how cancer cells adapt to their environments may aid the development of drugs that aim to disrupt the metabolism of tumors.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Metabolome , Nutrients , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Humans , Kidney Neoplasms/metabolism , Nutrients/metabolism , Metabolomics/methods , Tumor Microenvironment , Extracellular Fluid/metabolism , Female , Male , Lipidomics
2.
bioRxiv ; 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38187626

ABSTRACT

The tumor microenvironment is a determinant of cancer progression and therapeutic efficacy, with nutrient availability playing an important role. Although it is established that the local abundance of specific nutrients defines the metabolic parameters for tumor growth, the factors guiding nutrient availability in tumor compared to normal tissue and blood remain poorly understood. To define these factors in renal cell carcinoma (RCC), we performed quantitative metabolomic and comprehensive lipidomic analyses of tumor interstitial fluid (TIF), adjacent normal kidney interstitial fluid (KIF), and plasma samples collected from patients. TIF nutrient composition closely resembles KIF, suggesting that tissue-specific factors unrelated to the presence of cancer exert a stronger influence on nutrient levels than tumor-driven alterations. Notably, select metabolite changes consistent with known features of RCC metabolism are found in RCC TIF, while glucose levels in TIF are not depleted to levels that are lower than those found in KIF. These findings inform tissue nutrient dynamics in RCC, highlighting a dominant role of non-cancer driven tissue factors in shaping nutrient availability in these tumors.

3.
Nat Metab ; 5(12): 2131-2147, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37957387

ABSTRACT

Glutamine is a critical metabolite for rapidly proliferating cells as it is used for the synthesis of key metabolites necessary for cell growth and proliferation. Glutamine metabolism has been proposed as a therapeutic target in cancer and several chemical inhibitors are in development or in clinical trials. How cells subsist when glutamine is limiting is poorly understood. Here, using an unbiased screen, we identify ALDH18A1, which encodes P5CS, the rate-limiting enzyme in the proline biosynthetic pathway, as a gene that cells can downregulate in response to glutamine starvation. Notably, P5CS downregulation promotes de novo glutamine synthesis, highlighting a previously unrecognized metabolic plasticity of cancer cells. The glutamate conserved from reducing proline synthesis allows cells to produce the key metabolites necessary for cell survival and proliferation under glutamine-restricted conditions. Our findings reveal an adaptive pathway that cancer cells acquire under nutrient stress, identifying proline biosynthesis as a previously unrecognized major consumer of glutamate, a pathway that could be exploited for developing effective metabolism-driven anticancer therapies.


Subject(s)
Glutamine , Neoplasms , Humans , Glutamine/metabolism , Cell Proliferation , Proline , Glutamates
4.
Cell Chem Biol ; 30(9): 1156-1168.e7, 2023 09 21.
Article in English | MEDLINE | ID: mdl-37689063

ABSTRACT

A challenge for screening new anticancer drugs is that efficacy in cell culture models is not always predictive of efficacy in patients. One limitation of standard cell culture is a reliance on non-physiological nutrient levels, which can influence cell metabolism and drug sensitivity. A general assessment of how physiological nutrients affect cancer cell response to small molecule therapies is lacking. To address this, we developed a serum-derived culture medium that supports the proliferation of diverse cancer cell lines and is amenable to high-throughput screening. We screened several small molecule libraries and found that compounds targeting metabolic enzymes were differentially effective in standard compared to serum-derived medium. We exploited the differences in nutrient levels between each medium to understand why medium conditions affected the response of cells to some compounds, illustrating how this approach can be used to screen potential therapeutics and understand how their efficacy is modified by available nutrients.


Subject(s)
Cell Culture Techniques , High-Throughput Screening Assays , Humans , Cell Line , Small Molecule Libraries/pharmacology
5.
Phys Med Rehabil Clin N Am ; 34(3): 513-522, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37419528

ABSTRACT

Patients who are hospitalized due to COVID-19 are predisposed to requiring acute inpatient rehabilitation. Multiple factors have posed challenges to inpatient rehabilitation during the COVID-19 pandemic, such as staff shortages, restrictions with therapy, and barriers to discharge. Despite these challenges, data have shown that inpatient rehabilitation plays a key role in functional gains for this patient population. There remains a need for more data on the current challenges that are faced in the inpatient rehabilitation setting, as well as better understanding of long-term functional outcomes following COVID-19.


Subject(s)
COVID-19 , Humans , Inpatients , Pandemics
6.
bioRxiv ; 2023 Feb 27.
Article in English | MEDLINE | ID: mdl-36909640

ABSTRACT

A challenge for screening new candidate drugs to treat cancer is that efficacy in cell culture models is not always predictive of efficacy in patients. One limitation of standard cell culture is a reliance on non-physiological nutrient levels to propagate cells. Which nutrients are available can influence how cancer cells use metabolism to proliferate and impact sensitivity to some drugs, but a general assessment of how physiological nutrients affect cancer cell response to small molecule therapies is lacking. To enable screening of compounds to determine how the nutrient environment impacts drug efficacy, we developed a serum-derived culture medium that supports the proliferation of diverse cancer cell lines and is amenable to high-throughput screening. We used this system to screen several small molecule libraries and found that compounds targeting metabolic enzymes were enriched as having differential efficacy in standard compared to serum-derived medium. We exploited the differences in nutrient levels between each medium to understand why medium conditions affected the response of cells to some compounds, illustrating how this approach can be used to screen potential therapeutics and understand how their efficacy is modified by available nutrients.

7.
Elife ; 122023 02 09.
Article in English | MEDLINE | ID: mdl-36756948

ABSTRACT

Methylation is a widely occurring modification that requires the methyl donor S-adenosylmethionine (SAM) and acts in regulation of gene expression and other processes. SAM is synthesized from methionine, which is imported or generated through the 1-carbon cycle (1 CC). Alterations in 1 CC function have clear effects on lifespan and stress responses, but the wide distribution of this modification has made identification of specific mechanistic links difficult. Exploiting a dynamic stress-induced transcription model, we find that two SAM synthases in Caenorhabditis elegans, SAMS-1 and SAMS-4, contribute differently to modification of H3K4me3, gene expression and survival. We find that sams-4 enhances H3K4me3 in heat shocked animals lacking sams-1, however, sams-1 cannot compensate for sams-4, which is required to survive heat stress. This suggests that the regulatory functions of SAM depend on its enzymatic source and that provisioning of SAM may be an important regulatory step linking 1 CC function to phenotypes in aging and stress.


Subject(s)
Histones , S-Adenosylmethionine , Animals , S-Adenosylmethionine/metabolism , Histones/metabolism , Caenorhabditis elegans/physiology , Heat-Shock Response , Gene Expression
8.
J Exp Med ; 220(1)2023 01 02.
Article in English | MEDLINE | ID: mdl-36219197

ABSTRACT

Variants in the triggering receptor expressed on myeloid cells 2 (TREM2) gene are associated with increased risk for late-onset AD. Genetic loss of or decreased TREM2 function impairs the microglial response to amyloid-ß (Aß) plaques, resulting in more diffuse Aß plaques and increased peri-plaque neuritic dystrophy and AD-tau seeding. Thus, microglia and TREM2 are at a critical intersection of Aß and tau pathologies in AD. Since genetically decreasing TREM2 function increases Aß-induced tau seeding, we hypothesized that chronically increasing TREM2 signaling would decrease amyloid-induced tau-seeding and spreading. Using a mouse model of amyloidosis in which AD-tau is injected into the brain to induce Aß-dependent tau seeding/spreading, we found that chronic administration of an activating TREM2 antibody increases peri-plaque microglial activation but surprisingly increases peri-plaque NP-tau pathology and neuritic dystrophy, without altering Aß plaque burden. Our data suggest that sustained microglial activation through TREM2 that does not result in strong amyloid removal may exacerbate Aß-induced tau pathology, which may have important clinical implications.


Subject(s)
Alzheimer Disease , Amyloidosis , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid , Amyloid beta-Peptides , Amyloidosis/pathology , Animals , Disease Models, Animal , Microglia/pathology , Plaque, Amyloid/pathology
9.
Nature ; 609(7929): 1005-1011, 2022 09.
Article in English | MEDLINE | ID: mdl-36131016

ABSTRACT

Lysosomes have many roles, including degrading macromolecules and signalling to the nucleus1. Lysosomal dysfunction occurs in various human conditions, such as common neurodegenerative diseases and monogenic lysosomal storage disorders (LSDs)2-4. For most LSDs, the causal genes have been identified but, in some, the function of the implicated gene is unknown, in part because lysosomes occupy a small fraction of the cellular volume so that changes in lysosomal contents are difficult to detect. Here we develop the LysoTag mouse for the tissue-specific isolation of intact lysosomes that are compatible with the multimodal profiling of their contents. We used the LysoTag mouse to study CLN3, a lysosomal transmembrane protein with an unknown function. In children, the loss of CLN3 causes juvenile neuronal ceroid lipofuscinosis (Batten disease), a lethal neurodegenerative LSD. Untargeted metabolite profiling of lysosomes from the brains of mice lacking CLN3 revealed a massive accumulation of glycerophosphodiesters (GPDs)-the end products of glycerophospholipid catabolism. GPDs also accumulate in the lysosomes of CLN3-deficient cultured cells and we show that CLN3 is required for their lysosomal egress. Loss of CLN3 also disrupts glycerophospholipid catabolism in the lysosome. Finally, we found elevated levels of glycerophosphoinositol in the cerebrospinal fluid of patients with Batten disease, suggesting the potential use of glycerophosphoinositol as a disease biomarker. Our results show that CLN3 is required for the lysosomal clearance of GPDs and reveal Batten disease as a neurodegenerative LSD with a defect in glycerophospholipid metabolism.


Subject(s)
Esters , Glycerophospholipids , Inositol Phosphates , Lysosomes , Membrane Glycoproteins , Molecular Chaperones , Animals , Biomarkers/cerebrospinal fluid , Biomarkers/metabolism , Child , Esters/metabolism , Glycerophospholipids/cerebrospinal fluid , Glycerophospholipids/metabolism , Humans , Inositol Phosphates/cerebrospinal fluid , Inositol Phosphates/metabolism , Lysosomal Storage Diseases/cerebrospinal fluid , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/metabolism , Lysosomes/metabolism , Lysosomes/pathology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Neuronal Ceroid-Lipofuscinoses/cerebrospinal fluid , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/metabolism
10.
Nat Cell Biol ; 24(8): 1252-1264, 2022 08.
Article in English | MEDLINE | ID: mdl-35927450

ABSTRACT

Nucleotide metabolism supports RNA synthesis and DNA replication to enable cell growth and division. Nucleotide depletion can inhibit cell growth and proliferation, but how cells sense and respond to changes in the relative levels of individual nucleotides is unclear. Moreover, the nucleotide requirement for biomass production changes over the course of the cell cycle, and how cells coordinate differential nucleotide demands with cell cycle progression is not well understood. Here we find that excess levels of individual nucleotides can inhibit proliferation by disrupting the relative levels of nucleotide bases needed for DNA replication and impeding DNA replication. The resulting purine and pyrimidine imbalances are not sensed by canonical growth regulatory pathways like mTORC1, Akt and AMPK signalling cascades, causing excessive cell growth despite inhibited proliferation. Instead, cells rely on replication stress signalling to survive during, and recover from, nucleotide imbalance during S phase. We find that ATR-dependent replication stress signalling is activated during unperturbed S phases and promotes nucleotide availability to support DNA replication. Together, these data reveal that imbalanced nucleotide levels are not detected until S phase, rendering cells reliant on replication stress signalling to cope with this metabolic problem and disrupting the coordination of cell growth and division.


Subject(s)
DNA Replication , Nucleotides , Cell Cycle/genetics , Cell Division , DNA Replication/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Nucleotides/genetics , Nucleotides/metabolism , S Phase
11.
Cell Rep ; 40(7): 111187, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35977507

ABSTRACT

Dietary protein restriction (PR) has rapid effects on metabolism including improved glucose and lipid homeostasis, via multiple mechanisms. Here, we investigate responses of fecal microbiome, hepatic transcriptome, and hepatic metabolome to six diets with protein from 18% to 0% of energy in mice. PR alters fecal microbial composition, but metabolic effects are not transferable via fecal transplantation. Hepatic transcriptome and metabolome are significantly altered in diets with lower than 10% energy from protein. Changes upon PR correlate with calorie restriction but with a larger magnitude and specific changes in amino acid (AA) metabolism. PR increases steady-state aspartate, serine, and glutamate and decreases glucose and gluconeogenic intermediates. 13C6 glucose and glycerol tracing reveal increased fractional enrichment in aspartate, serine, and glutamate. Changes remain intact in hepatic ATF4 knockout mice. Together, this demonstrates an ATF4-independent shift in gluconeogenic substrate utilization toward specific AAs, with compensation from glycerol to promote a protein-sparing response.


Subject(s)
Glucose , Glycerol , Animals , Aspartic Acid/metabolism , Dietary Proteins/metabolism , Gluconeogenesis , Glucose/metabolism , Glutamic Acid/metabolism , Glycerol/metabolism , Liver/metabolism , Mice , Serine/metabolism
12.
Science ; 377(6601): 47-56, 2022 07.
Article in English | MEDLINE | ID: mdl-35771919

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) kinase controls growth in response to nutrients, including the amino acid leucine. In cultured cells, mTORC1 senses leucine through the leucine-binding Sestrin proteins, but the physiological functions and distribution of Sestrin-mediated leucine sensing in mammals are unknown. We find that mice lacking Sestrin1 and Sestrin2 cannot inhibit mTORC1 upon dietary leucine deprivation and suffer a rapid loss of white adipose tissue (WAT) and muscle. The WAT loss is driven by aberrant mTORC1 activity and fibroblast growth factor 21 (FGF21) production in the liver. Sestrin expression in the liver lobule is zonated, accounting for zone-specific regulation of mTORC1 activity and FGF21 induction by leucine. These results establish the mammalian Sestrins as physiological leucine sensors and reveal a spatial organization to nutrient sensing by the mTORC1 pathway.


Subject(s)
Diet , Leucine , Liver , Mechanistic Target of Rapamycin Complex 1 , Sestrins , Adipose Tissue, White/enzymology , Animals , Leucine/metabolism , Liver/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Sestrins/metabolism , Signal Transduction
14.
Blood ; 139(4): 538-553, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34624079

ABSTRACT

Burkitt lymphoma (BL) is an aggressive lymphoma type that is currently treated by intensive chemoimmunotherapy. Despite the favorable clinical outcome for most patients with BL, chemotherapy-related toxicity and disease relapse remain major clinical challenges, emphasizing the need for innovative therapies. Using genome-scale CRISPR-Cas9 screens, we identified B-cell receptor (BCR) signaling, specific transcriptional regulators, and one-carbon metabolism as vulnerabilities in BL. We focused on serine hydroxymethyltransferase 2 (SHMT2), a key enzyme in one-carbon metabolism. Inhibition of SHMT2 by either knockdown or pharmacological compounds induced anti-BL effects in vitro and in vivo. Mechanistically, SHMT2 inhibition led to a significant reduction of intracellular glycine and formate levels, which inhibited the mTOR pathway and thereby triggered autophagic degradation of the oncogenic transcription factor TCF3. Consequently, this led to a collapse of tonic BCR signaling, which is controlled by TCF3 and is essential for BL cell survival. In terms of clinical translation, we also identified drugs such as methotrexate that synergized with SHMT inhibitors. Overall, our study has uncovered the dependency landscape in BL, identified and validated SHMT2 as a drug target, and revealed a mechanistic link between SHMT2 and the transcriptional master regulator TCF3, opening up new perspectives for innovative therapies.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/metabolism , Glycine Hydroxymethyltransferase/antagonists & inhibitors , Glycine Hydroxymethyltransferase/metabolism , Animals , Burkitt Lymphoma/genetics , Cell Line, Tumor , Cell Survival/drug effects , Drug Discovery , Formates/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Glycine/metabolism , Glycine Hydroxymethyltransferase/genetics , Humans , Mice , Molecular Targeted Therapy , Proteolysis/drug effects
15.
Leukemia ; 36(2): 348-360, 2022 02.
Article in English | MEDLINE | ID: mdl-34341479

ABSTRACT

Despite progress in the treatment of acute lymphoblastic leukemia (ALL), T-cell ALL (T-ALL) has limited treatment options, particularly in the setting of relapsed/refractory disease. Using an unbiased genome-scale CRISPR-Cas9 screen we sought to identify pathway dependencies for T-ALL which could be harnessed for therapy development. Disruption of the one-carbon folate, purine and pyrimidine pathways scored as the top metabolic pathways required for T-ALL proliferation. We used a recently developed inhibitor of SHMT1 and SHMT2, RZ-2994, to characterize the effect of inhibiting these enzymes of the one-carbon folate pathway in T-ALL and found that T-ALL cell lines were differentially sensitive to RZ-2994, with the drug inducing a S/G2 cell cycle arrest. The effects of SHMT1/2 inhibition were rescued by formate supplementation. Loss of both SHMT1 and SHMT2 was necessary for impaired growth and cell cycle arrest, with suppression of both SHMT1 and SHMT2 inhibiting leukemia progression in vivo. RZ-2994 also decreased leukemia burden in vivo and remained effective in the setting of methotrexate resistance in vitro. This study highlights the significance of the one-carbon folate pathway in T-ALL and supports further development of SHMT inhibitors for treatment of T-ALL and other cancers.


Subject(s)
CRISPR-Cas Systems , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Folic Acid/metabolism , Glycine Hydroxymethyltransferase/antagonists & inhibitors , Methotrexate/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Cell Cycle , Cell Proliferation , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Science ; 374(6572): 1227-1237, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34855504

ABSTRACT

For electrons to continuously enter and flow through the mitochondrial electron transport chain (ETC), they must ultimately land on a terminal electron acceptor (TEA), which is known to be oxygen in mammals. Paradoxically, we find that complex I and dihydroorotate dehydrogenase (DHODH) can still deposit electrons into the ETC when oxygen reduction is impeded. Cells lacking oxygen reduction accumulate ubiquinol, driving the succinate dehydrogenase (SDH) complex in reverse to enable electron deposition onto fumarate. Upon inhibition of oxygen reduction, fumarate reduction sustains DHODH and complex I activities. Mouse tissues display varying capacities to use fumarate as a TEA, most of which net reverse the SDH complex under hypoxia. Thus, we delineate a circuit of electron flow in the mammalian ETC that maintains mitochondrial functions under oxygen limitation.


Subject(s)
Electron Transport , Electrons , Fumarates/metabolism , Animals , Cell Hypoxia , Cell Line , Cell Line, Tumor , Dihydroorotate Dehydrogenase/metabolism , Electron Transport Complex I/metabolism , Electron Transport Complex III/metabolism , Electron Transport Complex IV/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Oxidation-Reduction , Oxygen/metabolism , Succinate Dehydrogenase/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/metabolism
17.
Nat Metab ; 3(11): 1500-1511, 2021 11.
Article in English | MEDLINE | ID: mdl-34799701

ABSTRACT

Folate metabolism can be an effective target for cancer treatment. However, standard cell culture conditions utilize folic acid, a non-physiological folate source for most tissues. We find that the enzyme that couples folate and methionine metabolic cycles, methionine synthase, is required for cancer cell proliferation and tumour growth when 5-methyl tetrahydrofolate (THF), the major folate found in circulation, is the extracellular folate source. In such physiological conditions, methionine synthase incorporates 5-methyl THF into the folate cycle to maintain intracellular levels of the folates needed for nucleotide production. 5-methyl THF can sustain intracellular folate metabolism in the absence of folic acid. Therefore, cells exposed to 5-methyl THF are more resistant to methotrexate, an antifolate drug that specifically blocks folic acid incorporation into the folate cycle. Together, these data argue that the environmental folate source has a profound effect on folate metabolism, determining how both folate cycle enzymes and antifolate drugs impact proliferation.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Neoplasms/metabolism , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/genetics , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm/genetics , Folic Acid/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Methotrexate/pharmacology , Neoplasms/etiology , Neoplasms/pathology , Tetrahydrofolates/metabolism
18.
Curr Protoc ; 1(9): e245, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34516047

ABSTRACT

Studies in various tissues have revealed a central role of metabolic pathways in regulating adult stem cell function in tissue regeneration and tumor initiation. The unique metabolic dependences or preferences of adult stem cells, therefore, are emerging as a new category of therapeutic target. Recently, advanced methods including high-resolution metabolomics, proteomics, and transcriptomics have been developed to address the growing interest in stem cell metabolism. A practical framework integrating the omics analyses is needed to systematically perform metabolic characterization in a cell-type-specific manner. Here, we leverage recent advances in transcriptomics and proteomics research to identify cell-type-specific metabolic features by reconstructing cell identity using genes and the encoded enzymes involved in major metabolic pathways. We provide protocols for cell isolation, transcriptome and proteome analyses, and metabolite profiling and measurement. The workflow for mapping cell-type-specific metabolic signatures presented here, although initially developed for intestinal crypt cells, can be easily implemented for cell populations in other tissues, and is highly compatible with most public datasets. © 2021 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Intestinal crypt isolation and cell population purification Basic Protocol 2: Transcriptome analyses for cell-type-specific metabolic gene expression Basic Protocol 3: Proteome analyses for cell-type-specific metabolic enzyme levels Basic Protocol 4: Metabolite profiling and measurement.


Subject(s)
Proteome , Transcriptome , Gene Expression Profiling , Metabolic Networks and Pathways/genetics , Proteome/genetics , Proteomics
19.
Cell Rep ; 35(10): 109212, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34107251

ABSTRACT

Obesity is an established risk factor for cancer in many tissues. In the mammalian intestine, a pro-obesity high-fat diet (HFD) promotes regeneration and tumorigenesis by enhancing intestinal stem cell (ISC) numbers, proliferation, and function. Although PPAR (peroxisome proliferator-activated receptor) nuclear receptor activity has been proposed to facilitate these effects, their exact role is unclear. Here we find that, in loss-of-function in vivo models, PPARα and PPARδ contribute to the HFD response in ISCs. Mechanistically, both PPARs do so by robustly inducing a downstream fatty acid oxidation (FAO) metabolic program. Pharmacologic and genetic disruption of CPT1A (the rate-controlling enzyme of mitochondrial FAO) blunts the HFD phenotype in ISCs. Furthermore, inhibition of CPT1A dampens the pro-tumorigenic consequences of a HFD on early tumor incidence and progression. These findings demonstrate that inhibition of a HFD-activated FAO program creates a therapeutic opportunity to counter the effects of a HFD on ISCs and intestinal tumorigenesis.


Subject(s)
Carcinogenesis/pathology , Diet, High-Fat/adverse effects , Fatty Acids/metabolism , Intestines/pathology , Obesity/physiopathology , PPAR alpha/metabolism , Stem Cells/metabolism , Animals , Humans , Mice , Oxidation-Reduction
20.
CA Cancer J Clin ; 71(4): 333-358, 2021 07.
Article in English | MEDLINE | ID: mdl-33982817

ABSTRACT

Cancer has myriad effects on metabolism that include both rewiring of intracellular metabolism to enable cancer cells to proliferate inappropriately and adapt to the tumor microenvironment, and changes in normal tissue metabolism. With the recognition that fluorodeoxyglucose-positron emission tomography imaging is an important tool for the management of many cancers, other metabolites in biological samples have been in the spotlight for cancer diagnosis, monitoring, and therapy. Metabolomics is the global analysis of small molecule metabolites that like other -omics technologies can provide critical information about the cancer state that are otherwise not apparent. Here, the authors review how cancer and cancer therapies interact with metabolism at the cellular and systemic levels. An overview of metabolomics is provided with a focus on currently available technologies and how they have been applied in the clinical and translational research setting. The authors also discuss how metabolomics could be further leveraged in the future to improve the management of patients with cancer.


Subject(s)
Metabolomics , Neoplasms/metabolism , Biomedical Research , Humans , Medical Oncology , Molecular Targeted Therapy , Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...