Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Methods Mol Biol ; 2650: 77-88, 2023.
Article in English | MEDLINE | ID: mdl-37310625

ABSTRACT

The equilibrium between stem cell self-renewal and differentiation followed by proper lineage specification of progenitor cells is considered imperative for maintaining intestinal homeostasis. In the hierarchical model, intestinal differentiation is defined by the stepwise acquisition of lineage-specific mature cell features, where Notch signaling and lateral inhibition instructively regulate the cell-fate decisions. Recent studies reveal a broadly permissive intestinal chromatin underlies the lineage plasticity and adaptation to diet mediated by Notch transcriptional program. Here, we review the conventional understanding of Notch programming in intestinal differentiation and describe how new data from epigenetic and transcriptional analyses may refine or revise the current view. We provide instructions on sample preparation and data analysis and explain how to use ChIP-seq and scRNA-seq in combination of lineage tracing assay to determine the dynamics of Notch program and intestinal differentiation in the context of dietary and metabolic regulation of cell-fate decisions.


Subject(s)
Acclimatization , Epigenomics , Biological Assay , Cell Differentiation/genetics , Epigenesis, Genetic
2.
Methods Mol Biol ; 2650: 123-132, 2023.
Article in English | MEDLINE | ID: mdl-37310628

ABSTRACT

Organoid cultures have been developed to model intestinal stem cell (ISC) function in self-renewal and differentiation. Upon differentiation, the first fate decision for ISC and early progenitors to make is between secretory (Paneth cell, goblet cell, enteroendocrine cell, or tuft cell) and absorptive (enterocyte and M cell) lineages. Using genetic and pharmacological approaches, in vivo studies in the past decade have revealed that Notch signaling functions as a binary switch for the secretory vs. absorptive lineage decision in adult intestine. Recent breakthroughs in organoid-based assays enable real-time observation of smaller-scale and higher-throughput experiments in vitro, which have begun contributing to new understandings of mechanistic principles underlying intestinal differentiation. In this chapter, we summarize the in vivo and in vitro tools for modulating Notch signaling and assess its impact on intestinal cell fate. We also provide example protocols of how to use intestinal organoids as functional assays to study Notch activity in intestinal lineage decisions.


Subject(s)
Enterocytes , Intestines , Adult , Humans , Enteroendocrine Cells , Biological Assay , Organoids
3.
Res Sq ; 2023 Jan 10.
Article in English | MEDLINE | ID: mdl-36711807

ABSTRACT

For more than a century, fasting regimens have improved health, lifespan, and tissue regeneration in diverse organisms, including humans. However, how fasting and post-fast refeeding impact adult stem cells and tumour formation has yet to be explored in depth. Here, we demonstrate that post-fast refeeding increases intestinal stem cell (ISC) proliferation and tumour formation: Post-fast refeeding augments the regenerative capacity of Lgr5+ intestinal stem cells (ISCs), and loss of the tumour suppressor Apc in ISCs under post-fast refeeding leads to a higher tumour incidence in the small intestine and colon than in the fasted or ad libitum (AL) fed states. This demonstrates that post-fast refeeding is a distinct state. Mechanistically, we discovered that robust induction of mTORC1 in post-fast-refed ISCs increases protein synthesis via polyamine metabolism to drive these changes, as inhibition of mTORC1, polyamine metabolite production, or protein synthesis abrogates the regenerative or tumourigenic effects of post-fast refeeding. Thus, fast-refeeding cycles must be carefully considered when planning diet-based strategies for regeneration without increasing cancer risk, as post-fast refeeding leads to a burst not only in stem cell-driven regeneration but also in tumourigenicity.

4.
Cell Rep ; 37(8): 110044, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34818540

ABSTRACT

ß-hydroxybutyrate (ß-OHB) is an essential metabolic energy source during fasting and functions as a chromatin regulator by lysine ß-hydroxybutyrylation (Kbhb) modification of the core histones H3 and H4. We report that Kbhb on histone H3 (H3K9bhb) is enriched at proximal promoters of critical gene subsets associated with lipolytic and ketogenic metabolic pathways in small intestine (SI) crypts during fasting. Similar Kbhb enrichment is observed in Lgr5+ stem cell-enriched epithelial spheroids treated with ß-OHB in vitro. Combinatorial chromatin state analysis reveals that H3K9bhb is associated with active chromatin states and that fasting enriches for an H3K9bhb-H3K27ac signature at active metabolic gene promoters and distal enhancer elements. Intestinal knockout of Hmgcs2 results in marked loss of H3K9bhb-associated loci, suggesting that local production of ß-OHB is responsible for chromatin reprogramming within the SI crypt. We conclude that modulation of H3K9bhb in SI crypts is a key gene regulatory event in response to fasting.


Subject(s)
3-Hydroxybutyric Acid/metabolism , Fasting/metabolism , Histones/metabolism , Acetylation , Animals , Chromatin/metabolism , Fasting/physiology , Female , Gene Expression/genetics , Gene Expression Regulation/genetics , Intestine, Small/metabolism , Ketone Bodies/metabolism , Lysine/analogs & derivatives , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Regulatory Sequences, Nucleic Acid/genetics
5.
Sci Adv ; 7(46): eabk0271, 2021 Nov 12.
Article in English | MEDLINE | ID: mdl-34767451

ABSTRACT

Stem cells are remarkably small. Whether small size is important for stem cell function is unknown. We find that hematopoietic stem cells (HSCs) enlarge under conditions known to decrease stem cell function. This decreased fitness of large HSCs is due to reduced proliferation and was accompanied by altered metabolism. Preventing HSC enlargement or reducing large HSCs in size averts the loss of stem cell potential under conditions causing stem cell exhaustion. Last, we show that murine and human HSCs enlarge during aging. Preventing this age-dependent enlargement improves HSC function. We conclude that small cell size is important for stem cell function in vivo and propose that stem cell enlargement contributes to their functional decline during aging.

6.
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
7.
Int J Mol Sci ; 22(7)2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33808324

ABSTRACT

Calcium (Ca2+) is an important mediator of multicellular homeostasis and is involved in several diseases. The interplay among the kidney, bone, intestine, and parathyroid gland in Ca2+ homeostasis is strictly modulated by numerous hormones and signaling pathways. The calcium-sensing receptor (CaSR) is a G protein-coupled receptor, that is expressed in calcitropic tissues such as the parathyroid gland and the kidney, plays a pivotal role in Ca2+ regulation. CaSR is important for renal Ca2+, as a mutation in this receptor leads to hypercalciuria and calcium nephrolithiasis. In addition, CaSR is also widely expressed in the vascular system, including vascular endothelial cells (VECs) and vascular smooth muscle cells (VSMCs) and participates in the process of vascular calcification. Aberrant Ca2+ sensing by the kidney and VSMCs, owing to altered CaSR expression or function, is associated with the formation of nephrolithiasis and vascular calcification. Based on emerging epidemiological evidence, patients with nephrolithiasis have a higher risk of vascular calcification, but the exact mechanism linking the two conditions is unclear. However, a dysregulation in Ca2+ homeostasis and dysfunction in CaSR might be the connection between the two. This review summarizes renal calcium handling and calcium signaling in the vascular system, with a special focus on the link between nephrolithiasis and vascular calcification.


Subject(s)
Calcium Signaling/physiology , Nephrolithiasis/metabolism , Vascular Calcification/metabolism , Animals , Calcium/metabolism , Endothelial Cells/metabolism , Humans , Hypercalciuria/genetics , Hypercalciuria/metabolism , Hypercalciuria/physiopathology , Kidney/metabolism , Kidney Calculi/metabolism , Myocytes, Smooth Muscle/metabolism , Nephrolithiasis/physiopathology , Receptors, Calcium-Sensing/genetics , Vascular Calcification/genetics , Vascular Calcification/physiopathology
8.
Cell Stem Cell ; 28(4): 600-602, 2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33798421

ABSTRACT

COVID-19 has unfortunately halted lab work, conferences, and in-person networking, which is especially detrimental to researchers just starting their labs. Through social media and our reviewer networks, we met some early-career stem cell investigators impacted by the closures. Here, they introduce themselves and their research to our readers.


Subject(s)
Research Personnel , Stem Cells , COVID-19 , Humans
9.
Cell Stem Cell ; 28(3): 370-373, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33667357

ABSTRACT

In this forum piece, we review progress in exploiting diet and nutrition for enhancing tissue regeneration with a particular emphasis on how dietary composition and diet-induced physiology influence adult stem cell biology.


Subject(s)
Diet
10.
Trends Endocrinol Metab ; 32(1): 20-35, 2021 01.
Article in English | MEDLINE | ID: mdl-33277157

ABSTRACT

Food and nutrition have a profound impact on organismal health and diseases, and tissue-specific adult stem cells play a crucial role in coordinating tissue maintenance by responding to dietary cues. Emerging evidence indicates that adult intestinal stem cells (ISCs) actively adjust their fate decisions in response to diets and nutritional states to drive intestinal adaptation. Here, we review the signaling mechanisms mediating the dietary responses imposed by caloric intake and nutritional composition (i.e., macronutrients and micronutrients), fasting-feeding patterns, diet-induced growth factors, and microbiota on ISCs and their relevance to the beginnings of intestinal tumors.


Subject(s)
Intestines/cytology , Stem Cells/cytology , Stem Cells/metabolism , Animals , Carcinogenesis , Colonic Neoplasms/metabolism , Homeostasis/physiology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism
11.
Methods Mol Biol ; 2171: 53-64, 2020.
Article in English | MEDLINE | ID: mdl-32705635

ABSTRACT

This protocol describes a multipronged approach that we have created to determine the transcriptional induction of fatty acid oxidation (FAO) genes in Lgr5high intestinal stem cells and a subsequent metabolomics-based approach for assessing fatty acid utilization in the mammalian intestinal crypt. More specifically, we describe methods for crypt isolation followed by a FACS-based purification of stem and progenitor populations and RNA-sequencing analysis. Using this workflow, we can determine both basal gene expression profiles of key metabolic genes as well as corresponding changes in response to altered metabolic states, such as fasting. Subsequently, we describe a complementary metabolomics-based approach that we have developed to assess fatty acid uptake and utilization in the crypt using 13C stable isotope tracing. Combining these approaches, one can gain a better understanding of substrate utilization and the preceding transcriptional changes that accommodate these reactions in physiologic states of low carbohydrate utilization or during overabundance of dietary lipids.


Subject(s)
Lipid Metabolism/physiology , Stem Cells/cytology , Stem Cells/metabolism , Animals , Fatty Acids/metabolism , Flow Cytometry , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiology , Lipid Metabolism/genetics , Metabolomics/methods , Oxidation-Reduction , Stem Cells/physiology , Transcription, Genetic/genetics
12.
Cell Rep ; 31(4): 107565, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32348758

ABSTRACT

The small intestine is responsible for nutrient absorption and one of the most important interfaces between the environment and the body. During aging, changes of the epithelium lead to food malabsorption and reduced barrier function, thus increasing disease risk. The drivers of these alterations remain poorly understood. Here, we compare the proteomes of intestinal crypts from mice across different anatomical regions and ages. We find that aging alters epithelial immunity, metabolism, and cell proliferation and is accompanied by region-dependent skewing in the cellular composition of the epithelium. Of note, short-term dietary restriction followed by refeeding partially restores the epithelium by promoting stem cell differentiation toward the secretory lineage. We identify Hmgcs2 (3-hydroxy-3-methylglutaryl-coenzyme A [CoA] synthetase 2), the rate-limiting enzyme for ketogenesis, as a modulator of stem cell differentiation that responds to dietary changes, and we provide an atlas of region- and age-dependent proteome changes of the small intestine.


Subject(s)
Diet Therapy/methods , Proteomics/methods , Age Factors , Animals , Humans , Intestinal Mucosa/metabolism , Mice
13.
Cell ; 178(5): 1115-1131.e15, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442404

ABSTRACT

Little is known about how metabolites couple tissue-specific stem cell function with physiology. Here we show that, in the mammalian small intestine, the expression of Hmgcs2 (3-hydroxy-3-methylglutaryl-CoA synthetase 2), the gene encoding the rate-limiting enzyme in the production of ketone bodies, including beta-hydroxybutyrate (ßOHB), distinguishes self-renewing Lgr5+ stem cells (ISCs) from differentiated cell types. Hmgcs2 loss depletes ßOHB levels in Lgr5+ ISCs and skews their differentiation toward secretory cell fates, which can be rescued by exogenous ßOHB and class I histone deacetylase (HDAC) inhibitor treatment. Mechanistically, ßOHB acts by inhibiting HDACs to reinforce Notch signaling, instructing ISC self-renewal and lineage decisions. Notably, although a high-fat ketogenic diet elevates ISC function and post-injury regeneration through ßOHB-mediated Notch signaling, a glucose-supplemented diet has the opposite effects. These findings reveal how control of ßOHB-activated signaling in ISCs by diet helps to fine-tune stem cell adaptation in homeostasis and injury.


Subject(s)
Diet, High-Fat , Ketone Bodies/metabolism , Stem Cells/metabolism , 3-Hydroxybutyric Acid/blood , 3-Hydroxybutyric Acid/pharmacology , Aged, 80 and over , Animals , Cell Differentiation/drug effects , Cell Self Renewal , Female , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxymethylglutaryl-CoA Synthase/deficiency , Hydroxymethylglutaryl-CoA Synthase/genetics , Hydroxymethylglutaryl-CoA Synthase/metabolism , Intestines/cytology , Intestines/pathology , Male , Mice , Mice, Knockout , Receptors, G-Protein-Coupled/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Stem Cells/cytology , Young Adult
14.
Cell Stem Cell ; 22(5): 769-778.e4, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29727683

ABSTRACT

Diet has a profound effect on tissue regeneration in diverse organisms, and low caloric states such as intermittent fasting have beneficial effects on organismal health and age-associated loss of tissue function. The role of adult stem and progenitor cells in responding to short-term fasting and whether such responses improve regeneration are not well studied. Here we show that a 24 hr fast augments intestinal stem cell (ISC) function in young and aged mice by inducing a fatty acid oxidation (FAO) program and that pharmacological activation of this program mimics many effects of fasting. Acute genetic disruption of Cpt1a, the rate-limiting enzyme in FAO, abrogates ISC-enhancing effects of fasting, but long-term Cpt1a deletion decreases ISC numbers and function, implicating a role for FAO in ISC maintenance. These findings highlight a role for FAO in mediating pro-regenerative effects of fasting in intestinal biology, and they may represent a viable strategy for enhancing intestinal regeneration.


Subject(s)
Aging , Fasting/metabolism , Fatty Acids/metabolism , Homeostasis , Intestines/cytology , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cells, Cultured , Mice , Mice, Inbred Strains , Oxidation-Reduction
15.
Cell Stem Cell ; 22(2): 141-143, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29395049

ABSTRACT

In this issue of Cell Stem Cell, Wang et al. (2018) identify a novel link between Lpcat3-mediated phospholipid remodeling (the Lands cycle) and cholesterol biosynthesis that modulates intestinal stem cell proliferation and tumorigenesis. Notably, inhibition of cholesterol biosynthesis dampens many of the Lpcat3-deficiency-mediated effects in the intestine.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase , Carcinogenesis , Cholesterol , Humans , Intestines , Stem Cells
16.
Cell ; 168(5): 775-788.e12, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28235195

ABSTRACT

Stem-cell-based therapies can potentially reverse organ dysfunction and diseases, but the removal of impaired tissue and activation of a program leading to organ regeneration pose major challenges. In mice, a 4-day fasting mimicking diet (FMD) induces a stepwise expression of Sox17 and Pdx-1, followed by Ngn3-driven generation of insulin-producing ß cells, resembling that observed during pancreatic development. FMD cycles restore insulin secretion and glucose homeostasis in both type 2 and type 1 diabetes mouse models. In human type 1 diabetes pancreatic islets, fasting conditions reduce PKA and mTOR activity and induce Sox2 and Ngn3 expression and insulin production. The effects of the FMD are reversed by IGF-1 treatment and recapitulated by PKA and mTOR inhibition. These results indicate that a FMD promotes the reprogramming of pancreatic cells to restore insulin generation in islets from T1D patients and reverse both T1D and T2D phenotypes in mouse models. PAPERCLIP.


Subject(s)
Diabetes Mellitus, Type 1/diet therapy , Diabetes Mellitus, Type 2/diet therapy , Fasting , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Diet , Glucose Tolerance Test , Humans , In Vitro Techniques , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans , Mice , Nerve Tissue Proteins/genetics , Pancreas/cytology , Pancreas/metabolism , Signal Transduction , Transcriptome
17.
Sci Transl Med ; 9(377)2017 02 15.
Article in English | MEDLINE | ID: mdl-28202779

ABSTRACT

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


Subject(s)
Aging/pathology , Biomarkers/metabolism , Cardiovascular Diseases/pathology , Diabetes Mellitus/pathology , Diet , Fasting/physiology , Neoplasms/pathology , Adult , Female , Follow-Up Studies , Humans , Male , Risk Factors
18.
Nat Med ; 23(1): 14-15, 2017 01 06.
Article in English | MEDLINE | ID: mdl-28060803
19.
Cancer Cell ; 30(1): 136-146, 2016 07 11.
Article in English | MEDLINE | ID: mdl-27411588

ABSTRACT

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


Subject(s)
Breast Neoplasms/diet therapy , Breast Neoplasms/drug therapy , Doxorubicin/administration & dosage , Heme Oxygenase-1/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Breast Neoplasms/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Combined Modality Therapy , Disease Progression , Down-Regulation , Doxorubicin/pharmacology , Fasting , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , MCF-7 Cells , Mice , Neoplasm Transplantation , T-Lymphocytes, Cytotoxic/drug effects
20.
BMC Cancer ; 16: 360, 2016 06 10.
Article in English | MEDLINE | ID: mdl-27282289

ABSTRACT

BACKGROUND: Short-term starvation prior to chemotherapy administration protects mice against toxicity. We undertook dose-escalation of fasting prior to platinum-based chemotherapy to determine safety and feasibility in cancer patients. METHODS: 3 cohorts fasted before chemotherapy for 24, 48 and 72 h (divided as 48 pre-chemo and 24 post-chemo) and recorded all calories consumed. Feasibility was defined as ≥ 3/6 subjects in each cohort consuming ≤ 200 kcal per 24 h during the fast period without excess toxicity. Oxidative stress was evaluated in leukocytes using the COMET assay. Insulin, glucose, ketones, insulin-like growth factor-1 (IGF-1) and IGF binding proteins (IGFBPs) were measured as biomarkers of the fasting state. RESULTS: The median age of our 20 subjects was 61, and 85 % were women. Feasibility criteria were met. Fasting-related toxicities were limited to ≤ grade 2, most commonly fatigue, headache, and dizziness. The COMET assay indicated reduced DNA damage in leukocytes from subjects who fasted for ≥48 h (p = 0.08). There was a non-significant trend toward less grade 3 or 4 neutropenia in the 48 and 72 h cohorts compared to 24 h cohort (p = 0.17). IGF-1 levels decreased by 30, 33 and 8 % in the 24, 48 and 72 h fasting cohorts respectively after the first fasting period. CONCLUSION: Fasting for 72 h around chemotherapy administration is safe and feasible for cancer patients. Biomarkers such as IGF-1 may facilitate assessment of differences in chemotherapy toxicity in subgroups achieving the physiologic fasting state. An onging randomized trial is studying the effect of 72 h of fasting. TRIAL REGISTRATION: NCT00936364 , registered propectively on July 9, 2009.


Subject(s)
Fasting/blood , Insulin-Like Growth Factor I/metabolism , Neoplasms/drug therapy , Platinum/adverse effects , Adult , Aged , Biomarkers/blood , Biomarkers/metabolism , Feasibility Studies , Female , Humans , Male , Middle Aged , Neoplasms/blood , Oxidative Stress , Platinum/therapeutic use , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...