Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters

Publication year range
1.
Cell ; 180(3): 502-520.e19, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31983537

ABSTRACT

The tumor microenvironment (TME) is critical for tumor progression. However, the establishment and function of the TME remain obscure because of its complex cellular composition. Using a mouse genetic system called mosaic analysis with double markers (MADMs), we delineated TME evolution at single-cell resolution in sonic hedgehog (SHH)-activated medulloblastomas that originate from unipotent granule neuron progenitors in the brain. First, we found that astrocytes within the TME (TuAstrocytes) were trans-differentiated from tumor granule neuron precursors (GNPs), which normally never differentiate into astrocytes. Second, we identified that TME-derived IGF1 promotes tumor progression. Third, we uncovered that insulin-like growth factor 1 (IGF1) is produced by tumor-associated microglia in response to interleukin-4 (IL-4) stimulation. Finally, we found that IL-4 is secreted by TuAstrocytes. Collectively, our studies reveal an evolutionary process that produces a multi-lateral network within the TME of medulloblastoma: a fraction of tumor cells trans-differentiate into TuAstrocytes, which, in turn, produce IL-4 that stimulates microglia to produce IGF1 to promote tumor progression.


Subject(s)
Astrocytes/metabolism , Carcinogenesis/metabolism , Cell Transdifferentiation , Cerebellar Neoplasms/metabolism , Medulloblastoma/metabolism , Paracrine Communication , Animals , Cell Lineage , Cerebellar Neoplasms/pathology , Disease Models, Animal , Female , Hedgehog Proteins/metabolism , Heterografts , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Male , Medulloblastoma/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Tumor Microenvironment
2.
Cancer Immunol Immunother ; 73(7): 128, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38743074

ABSTRACT

The majority of the immune cell population in the tumor microenvironment (TME) consists of tumor-associated macrophages (TAM), which are the main players in coordinating tumor-associated inflammation. TAM has a high plasticity and is divided into two main phenotypes, pro-inflammatory M1 type and anti-inflammatory M2 type, with tumor-suppressive and tumor-promoting functions, respectively. Considering the beneficial effects of M1 macrophages for anti-tumor and the high plasticity of macrophages, the conversion of M2 TAM to M1 TAM is feasible and positive for tumor treatment. This study sought to evaluate whether the glycopeptide derived from simulated digested Codonopsis pilosula extracts could regulate the polarization of M2-like TAM toward the M1 phenotype and the potential regulatory mechanisms. The results showed that after glycopeptide dCP1 treatment, the mRNA relative expression levels of some M2 phenotype marker genes in M2-like TAM in simulated TME were reduced, and the relative expression levels of M1 phenotype marker genes and inflammatory factor genes were increased. Analysis of RNA-Seq of M2-like TAM after glycopeptide dCP1 intervention showed that the gene sets such as glycolysis, which is associated with macrophage polarization in the M1 phenotype, were significantly up-regulated, whereas those of gene sets such as IL-6-JAK-STAT3 pathway, which is associated with polarization in the M2 phenotype, were significantly down-regulated. Moreover, PCA analysis and Pearson's correlation also indicated that M2-like TAM polarized toward the M1 phenotype at the transcriptional level after treatment with the glycopeptide dCP1. Lipid metabolomics was used to further explore the efficacy of the glycopeptide dCP1 in regulating the polarization of M2-like TAM to the M1 phenotype. It was found that the lipid metabolite profiles in dCP1-treated M2-like TAM showed M1 phenotype macrophage lipid metabolism profiles compared with blank M2-like TAM. Analysis of the key differential lipid metabolites revealed that the interconversion between phosphatidylcholine (PC) and diacylglycerol (DG) metabolites may be the central reaction of the glycopeptide dCP1 in regulating the conversion of M2-like TAM to the M1 phenotype. The above results suggest that the glycopeptide dCP1 has the efficacy to regulate the polarization of M2-like TAM to M1 phenotype in simulated TME.


Subject(s)
Codonopsis , Phenotype , Tumor-Associated Macrophages , Animals , Humans , Mice , Glycopeptides/metabolism , Glycopeptides/pharmacology , Macrophage Activation/drug effects , Neoplasms/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/immunology , Tumor Microenvironment/drug effects , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/drug effects
3.
Biochem Biophys Res Commun ; 591: 7-12, 2022 02 05.
Article in English | MEDLINE | ID: mdl-34990904

ABSTRACT

Early-stage gastric cancer (GC) is asymptomatic. How to diagnose the early-stage GC is challenging. The sensitivity and specificity of diagnosing signatures for early-stage patients are still poor. Elastic-net-based analysis was used to identify potential diagnostic signatures of early-stage GC. The expression level of candidate gene was evaluated by immunohistochemistry staining. The potential function of candidate gene was verified by overexpressing in vitro. Consensus genes (including GAMT) were identified using the different strengths of the penalty. Surprisingly, GAMT was still identified even if some multicollinear variables were deleted directly. IHC staining showed that there are no GAMT-positive signals in the cell nuclei of all tumor tissues, while GAMT does express in nuclei of adjacent normal tissue. There are 16.33% positive cell nuclei in paracancerous tissues. In addition, the number of larger-area colonies of overexpression-GAMT group, empty-vector group, and AGS group is 70±29.21, 151.33±15.95, and 111.67±22.03, respectively. Number of larger colonies in group with overexpression of GAMT is significantly less than control groups. Elastic-net-penalty-based workflow is a effective tool to identify diagnostic biomarker for early-stage solid tumor. GAMT has strong potential to be the diagnostic biomarker for the early-stage GC.


Subject(s)
Biomarkers, Tumor/metabolism , Elasticity , Guanidinoacetate N-Methyltransferase/metabolism , Stomach Neoplasms/diagnosis , Cell Line, Tumor , Cell Proliferation , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Staging , Proportional Hazards Models , Reproducibility of Results , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
4.
Biochem Biophys Res Commun ; 630: 41-49, 2022 11 19.
Article in English | MEDLINE | ID: mdl-36137324

ABSTRACT

Hyperuricemia animal models have long been used for evaluating food-derived anti-hyperuricemia compounds. Fructose and potassium oxonate are commonly used for developing hyperuricemia mouse model. Recent research also developed spontaneous hyperuricemia model by uricase knockout (Uox-/-). In this work, we evaluated 3 kinds of models with the same gene background to illustrate the differences between the treatments. Unlike the uric acid levels in potassium oxonate (224.79 ± 33.62 µmol/L) and Uox-/- groups (458.39 ± 38.29 µmol/L), fructose treatment did not lead to higher serum uric acid level (174.93 ± 30.46 µmol/L) comparing to the control group (153.53 ± 40.96 µmol/L). However, abnormal glycometabolism only developed in the fructose and the Uox-/- group. In addition, anemia, inflammasome and severe renal injury occurred in the Uox-/- group. The Uox-/- mice were then treated with puerarin and allopurinol, and found that puerarin could reduce serum uric acid and alleviated the serious renal damage associated with high uric acid. Thus, the Uox-/- mice could be a suitable model for screening and evaluating anti-hyperuricemia compounds.


Subject(s)
Hyperuricemia , Uric Acid , Allopurinol , Animals , Disease Models, Animal , Fructose , Hyperuricemia/drug therapy , Inflammasomes , Mice , Urate Oxidase/genetics
5.
Crit Rev Food Sci Nutr ; 62(17): 4769-4799, 2022.
Article in English | MEDLINE | ID: mdl-33554630

ABSTRACT

Wnt signaling pathway is an evolutionarily conserved pathway that control embryonic development, adult tissue homeostasis, and pathological processes of organisms throughout life. However, dysregulation of the Wnt signaling is associated with the occurrence of chronic diseases. In comparison with the application of chemical drugs as traditional treatment for chronic diseases, dietary agents have unique advantages, such as less side effects, multiple targets, convenience in accessibility and higher acceptability in long-term intervention. In this review, we summarized current progress in manipulating the Wnt signaling using food components and its benefits in managing chronic diseases. The underlying mechanisms of bioactive food components in the management of the disease progression via the Wnt signaling was illustrated. Then, the review focused on the function of dietary pattern (which might act via combination of foods with multiple nutrients or food ingredients) on targeting Wnt signaling at multiple level. The potential caveats and challenges in developing new strategy via modulating Wnt-associated diseases with food-based agents and appropriate dietary pattern are also discussed in detail. This review shed light on the understanding of the regulatory effect of food bioactive components on chronic diseases management through the Wnt signaling, which can be expanded to other specific signaling pathway associated with disease.


Subject(s)
Chronic Disease/therapy , Eating/physiology , Food/classification , Wnt Signaling Pathway/physiology , Adult , Chronic Disease/prevention & control , Humans
6.
Biochem Biophys Res Commun ; 568: 116-123, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34217010

ABSTRACT

Advanced CUBIC clearing method can clear diverse organs and even the entire body of a mouse to enable high-resolution 3D imaging. Advanced CUBIC reagent has often been used due to its low toxicity and easy preparation. However, Advanced CUBIC has a long experimental cycle, which reduces the efficiency of data acquisition. In this study, we first tracked the clarity changes of different organs cleared by Advanced CUBIC and identified the shortest time required for optimal transparency in individual organs. We then introduced ultrasound processing and developed a decolorization cocktail to optimize the clearing efficiency of the Advanced CUBIC method. With the optimized clearing CUBIC-Plus method, we achieved high resolution 3D imaging of mouse organs. Our CUBIC-Plus provides an efficient procedure to clear different organ for 3D imaging at high resolution.


Subject(s)
Fluorescent Antibody Technique/methods , Imaging, Three-Dimensional/methods , Microscopy, Confocal/methods , Animals , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Color , Female , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence/methods , Ultrasonic Waves
7.
Brain Behav Immun ; 96: 227-238, 2021 08.
Article in English | MEDLINE | ID: mdl-34111528

ABSTRACT

Gut microbiota is proven to be involved in the development of beta amyloid (Aß) pathology in Alzheimer's disease (AD). Since there are difficulties in translating microbiota findings based on germ-free mice into clinical practice, here, we used short-term antibiotic cocktail treatment to develop a novel model with a near-germ-free status and without impacting Aß pathology. Three months old APPSWE/PS1ΔE9 mice were fed with antibiotic cocktails for two weeks by gavage to obtain a near "germ-free" status, and then received the donor fecal matter from the 16 months old APPSWE/PS1ΔE9 mice for 7 consecutive days. Fecal pellets were collected prior to antibiotics treatment, following antibiotic exposure, prior to and following fecal microbiota transplantation for gut microbiota analysis. Also, Aß pathology, astrocyte and microglia morphology were further explored. Pre-antibiotic-treated mice successfully allowed engraftment of gut microbiota following 7 consecutive days gavage with aged APPSWE/PS1ΔE9 mice microbiota. Microbiota reconstitution by transplantation was largely attributable to the donor source (e.g. g_Coriobacteriaceae and g_Clostridium) and led to a significant increase in Aß plaques. Surprisingly, astrocyte activation around Aß plaques was suppressed rather than microglia, the well-recognized plaque phagocytic cell type in Aß clearance, following microbiota engraftment. Our findings provide a novel framework for understanding the mechanisms of AD through the gut-brain axis and the translation of gut microbiota manipulation from bench to clinical practice.


Subject(s)
Alzheimer Disease , Microbiota , Alzheimer Disease/therapy , Amyloid beta-Peptides , Animals , Anti-Bacterial Agents , Disease Models, Animal , Fecal Microbiota Transplantation , Mice , Mice, Transgenic
8.
J Chem Inf Model ; 60(1): 17-21, 2020 01 27.
Article in English | MEDLINE | ID: mdl-31851496

ABSTRACT

Taking hydrogen bonds as a basis to explore biomolecular properties and interactions, we constructed the lone-pair electron (LPE) index and a molecular orbital fingerprint based on molecular hybrid orbitals to represent the ability of molecules to form hydrogen bonds. Then, a computational model was constructed to predict molecular interactions. The LPE and orbital fingerprint could effectively predict the biological properties and bioactivities of molecules. This study revealed the significance of hybrid orbitals for understanding cell biochemistry.


Subject(s)
Computer Simulation , Models, Molecular , Electrons , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Lipopolysaccharides/chemistry
9.
Carcinogenesis ; 40(10): 1269-1277, 2019 Oct 16.
Article in English | MEDLINE | ID: mdl-30805585

ABSTRACT

Gastric cancer (GC) has high morbidity and mortality rates worldwide. Abundant literature has reported several individual genes and their related pathways intimately involved in tumor progression. However, little is known about GC progression at the gene network level. Therefore, understanding the underlying mechanisms of pathological transition from early stage to late stage is urgently needed. This study aims to identify potential vital genes and modules involved in the progression of GC. To understand the gene regulatory network of GC progression, we analyzed micro RNAs and messenger RNA s expression profiles by using a couple of bioinformatics tools. miR-205 was identified by differentially expressed analysis and was further confirmed through using multiple kernel learning-based Kronecker regularized least squares. Using weighted gene co-expression network analysis, the gastric cancer progression-related module, which has the highest correlation value with cancer progression, was obtained. Kyoto Encyclopedia of Genes and Genomes pathways and biological processes of the GCPR module genes were related to cell adhesion. Meanwhile, large-scale genes of GCPR module were found to be targeted by miR-205, including two hub genes SORBS1 and LPAR1. In brief, through multiple analytical methods, we found that miR-205 and the GCPR module play critical roles in GC progression. In addition, miR-205 might maintain cell adhesion by regulating SORBS1 and LPAR1. To screen the potential drug candidates, the gene expression profile of the GCPR module was mapped connectivity map (Cmap), and the mTOR inhibitor (Sirolimus) was found to be the most promising candidate. We further confirmed that Sirolimus can suppress cell proliferation of GC cell in vitro.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , MicroRNAs/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcriptome , Apoptosis , Case-Control Studies , Cell Proliferation , Computational Biology , Disease Progression , Follow-Up Studies , Gene Expression Profiling , Humans , Prognosis , Survival Rate , Tumor Cells, Cultured
10.
Langmuir ; 35(10): 3710-3716, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30727729

ABSTRACT

Conventional chemotherapy, because of the high dose to keep the drug above the minimum effective concentration, possesses severe side effects and brings extra pain to patients. A controlled release drug delivery system, which is a bilayer self-assembled nanoparticle (NP) in this study, can solve this problem. Zein, a biodegradable natural protein from corn, was selected for the first layer of the drug encapsulation. The second layer was formed via the reversible ionic hydrogen bonds between zein and folic acid (FA), which was selected because of the two carboxylic acids and one amine group in its simple structure. Doxorubicin (DOX), a popular anticancer drug, was selected as the drug model to form the bilayer drug nanoencapsulation FA-NP-DOX. The in vitro controlled release profile of FA-NP-DOX was obtained. The in vivo pharmacokinetics and anticancer activity of FA-NP-DOX in tumor-xenografted animal models were also conducted. Compared to the zein nanoencapsulation of DOX (NP-DOX) and pure DOX, FA-NP-DOX showed comparable in vitro cytotoxicity but much longer in vitro controlled release time and in vivo circulation time. Both FA-NP-DOX and NP-DOX showed enhanced therapeutical efficiency in vivo than pure DOX. It is concluded that the bilayer self-assembled NP of zein and FA highly prolonged the controlled release and enhanced the therapeutic efficiency of the anticancer drug.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Carriers/chemistry , Lipid Bilayers/chemistry , Nanoparticles/chemistry , Zein/chemistry , Animals , Antibiotics, Antineoplastic/chemistry , Cell Proliferation/drug effects , Disease Models, Animal , Doxorubicin/chemistry , Drug Liberation , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Particle Size , Surface Properties
11.
Development ; 141(24): 4697-709, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25468938

ABSTRACT

The balance between self-renewal and differentiation of adult neural stem cells (aNSCs) is essential for the maintenance of the aNSC reservoir and the continuous supply of new neurons, but how this balance is fine-tuned in the adult brain is not fully understood. Here, we investigate the role of SIRT1, an important metabolic sensor and epigenetic repressor, in regulating adult hippocampal neurogenesis in mice. We found that there was an increase in SIRT1 expression during aNSC differentiation. In Sirt1 knockout (KO) mice, as well as in brain-specific and inducible stem cell-specific conditional KO mice, the proliferation and self-renewal rates of aNSCs in vivo were elevated. Proliferation and self-renewal rates of aNSCs and adult neural progenitor cells (aNPCs) were also elevated in neurospheres derived from Sirt1 KO mice and were suppressed by the SIRT1 agonist resveratrol in neurospheres from wild-type mice. In cultured neurospheres, 2-deoxy-D-glucose-induced metabolic stress suppressed aNSC/aNPC proliferation, and this effect was mediated in part by elevating SIRT1 activity. Microarray and biochemical analysis of neurospheres suggested an inhibitory effect of SIRT1 on Notch signaling in aNSCs/aNPCs. Inhibition of Notch signaling by a γ-secretase inhibitor also largely abolished the increased aNSC/aNPC proliferation caused by Sirt1 deletion. Together, these findings indicate that SIRT1 is an important regulator of aNSC/aNPC self-renewal and a potential mediator of the effect of metabolic changes.


Subject(s)
Adult Stem Cells/physiology , Cell Proliferation/physiology , Dentate Gyrus/cytology , Gene Expression Regulation/physiology , Neural Stem Cells/physiology , Sirtuin 1/metabolism , Adult Stem Cells/metabolism , Animals , Blotting, Western , Bromodeoxyuridine , Cell Proliferation/drug effects , Deoxyglucose/adverse effects , Fluorescence , Gene Expression Regulation/drug effects , Mice , Mice, Knockout , Microarray Analysis , Microscopy, Confocal , Neural Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Sirtuin 1/genetics , Statistics, Nonparametric , Tamoxifen
13.
JAMA ; 322(24): 2444-2445, 2019 12 24.
Article in English | MEDLINE | ID: mdl-31860038
14.
Cancer Pathog Ther ; 2(1): 15-23, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38328712

ABSTRACT

Brain metastases are a leading cause of cancer-related mortality. However, progress in their treatment has been limited over the past decade, due to an incomplete understanding of the underlying biological mechanisms. Employing accurate in vitro and in vivo models to recapitulate the complexities of brain metastasis offers the most promising approach to unravel the intricate cellular and physiological processes involved. Here, we present a comprehensive review of the currently accessible models for studying brain metastasis. We introduce a diverse array of in vitro and in vivo models, including cultured cells using the Transwell system, organoids, microfluidic models, syngeneic models, xenograft models, and genetically engineered models. We have also provided a concise summary of the merits and limitations inherent to each model while identifying the optimal contexts for their effective utilization. This review serves as a comprehensive resource, aiding researchers in making well-informed decisions regarding model selection that align with specific research questions.

15.
CNS Neurosci Ther ; 30(1): e14495, 2024 01.
Article in English | MEDLINE | ID: mdl-37849438

ABSTRACT

AIMS: Glioma is characterized by an immunosuppressed environment and a poor prognosis. The accumulation of Amyloid ß (Aß) leads to an active environment during the early stages of Alzheimer's disease (AD). Aß is also present in glioma tissues; however, the biological and translational implications of Aß in glioma are elusive. METHODS: Immunohistochemical (IHC) staining, Kaplan-Meier (KM) survival analysis and Cox regression analysis on a cohort of 79 patients from our institution were performed to investigate the association between Aß and the malignancy of glioma. Subsequently, the potential of oligomer-Aß42 (OAß42) to inhibit glioma growth was investigated in vivo and in vitro. Immunofluorescence staining and phagocytosis assays were performed to evaluate the activation of microglia. Finally, RNA-seq was utilized to identify the predominant signaling involved in this process and in vitro studies were performed to validate them. RESULTS: A positive correlation between Aß and a favorable prognosis was observed in glioma. Furthermore, OAß42 suppressed glioma growth by enhancing the phagocytic activity of microglia. Insulin-like growth factor 1 (IGF-1) secreted by OAß42-activated microglia was essential in the engulfment process. CONCLUSION: Our study proved an anti-glioma effect of Aß, and microglia could serve as a cellular target for treating glioma with OAß42.


Subject(s)
Alzheimer Disease , Glioma , Humans , Animals , Mice , Amyloid beta-Peptides/metabolism , Microglia , Alzheimer Disease/metabolism , Phagocytosis , Glioma/metabolism , Mice, Transgenic
16.
Mol Nutr Food Res ; : e2400023, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38924315

ABSTRACT

SCOPE: Grifola frondosa has been shown to induce immune modulatory, modulate autophagy, and apoptosis in cancer cells. However, little is known about its potential for managing tumor progression as an adjunct to nutrient restriction. METHODS AND RESULTS: Water extract produces a G. frondosa polysaccharide-protein complex (G. frondosa PPC) of average molecular weight of 46.48 kDa, with glucose (54.8%) as the main constituent. Under serum-restricted conditions, G. frondosa PPC can significantly inhibit MC38 colorectal tumor cell migration in vitro. Under alternate-day fasting condition, G. frondosa PPC can only significantly inhibit the growth of subcutaneous (s.c.) tumor, but is feeble in halting its spread in the intraperitoneal (i.p.) cavity in tumor-bearing mice. Histopathological examination and Raman imaging show a significant increase in lipid content in the tumor microenvironment (TME) tissue of the s.c. tumor-bearing mice. G. frondosa PPC significantly increases C17:0 and C24:0 saturated fatty acids and significantly decreases C16:1 and C18:1 monounsaturated fatty acids in the TME of s.c. tumor-bearing mice compared with the i.p. cavity model. CONCLUSION: G. frondosa PPC significantly inhibits tumor growth in s.c. tumor-bearing mice under intermittent fasting conditions by altering the fatty acid composition of the TME.

17.
Cancer Lett ; : 217076, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38906524

ABSTRACT

Understanding of the metabolic reprogramming has revolutionized our insights into tumor progression and potential treatment. This review concentrates on the aberrant metabolic pathways in cancer cells within the tumor microenvironment (TME). Cancer cells differ from normal cells in their metabolic processing of glucose, amino acids, and lipids in order to adapt to heightened biosynthetic and energy needs. These metabolic shifts, which crucially alter lactic acid, amino acid and lipid metabolism, affect not only tumor cell proliferation but also TME dynamics. This review also explores the reprogramming of various immune cells in the TME. From a therapeutic standpoint, targeting these metabolic alterations represents a novel cancer treatment strategy. This review also discusses approaches targeting the regulation of metabolism of different nutrients in tumor cells and influencing the tumor microenvironment to enhance the immune response. In summary, this review summarizes metabolic reprogramming in cancer and its potential as a target for innovative therapeutic strategies, offering fresh perspectives on cancer treatment.

18.
Adv Sci (Weinh) ; : e2306890, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816931

ABSTRACT

In spite of recent advances in the field of undernutrition, current dietary therapy relying on the supply of high protein high calorie formulas is still plagued with transient recovery of impaired organs resulting in significant relapse of cases. This is partly attributed to the inadequacy of current research models in recapitulating clinical undernutrition for mechanistic exploration. Using 1636 Macaca fascicularis monkeys, a human-relevant criterion for determining undernutrition weight-for-age z-score (WAZ), with a cutoff point of ≤ -1.83 is established as the benchmark for identifying undernourished nonhuman primates (U-NHPs). In U-NHPs, pathological anomalies in multi-organs are revealed. In particular, severe dysregulation of hepatic lipid metabolism characterized by impaired fatty acid oxidation due to mitochondria dysfunction, but unlikely peroxisome disorder, is identified as the anchor metabolic aberration in U-NHPs. Mitochondria dysfunction is typified by reduced mito-number, accumulated long-chain fatty acids, and disruption of OXPHOS complexes. Soy peptide-treated U-NHPs increase in WAZ scores, in addition to attenuated mitochondria dysfunction and restored OXPHOS complex levels. Herein, innovative criteria for identifying U-NHPs are developed, and unknown molecular mechanisms of undernutrition are revealed hitherto, and it is further proved that soypeptide supplementation reprogramed mitochondrial function to re-establish lipid metabolism balance and mitigated undernutrition.

19.
Int J Biol Sci ; 19(13): 4311-4326, 2023.
Article in English | MEDLINE | ID: mdl-37705736

ABSTRACT

Gliomas develop in unique and complicated environments that nourish tumor cells. The tumor microenvironment (TME) of gliomas comprises heterogeneous cells, including brain-resident cells, immune cells, and vascular cells. Reciprocal interactions among these cells are involved in the evolution of the TME. Moreover, the study of attractive therapeutic strategies that target the TME is transitioning from basic research to the clinic. Mouse models are indispensable tools for dissecting the processes and mechanisms leading to TME evolution. In this review, we overview the paradoxical roles of the TME, as well as the recent progress of mouse models in TME research. Finally, we summarize recent advances in TME-targeting therapeutic strategies.


Subject(s)
Glioma , Tumor Microenvironment , Animals , Mice , Tumor Microenvironment/genetics , Glioma/genetics , Disease Models, Animal
20.
BMC Genomics ; 13: 232, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22691069

ABSTRACT

BACKGROUND: The morphogenesis of the cerebral cortex depends on the precise control of gene expression during development. Small non-coding RNAs, including microRNAs and other groups of small RNAs, play profound roles in various physiological and pathological processes via their regulation of gene expression. A systematic analysis of the expression profile of small non-coding RNAs in developing cortical tissues is important for clarifying the gene regulation networks mediating key developmental events during cortical morphogenesis. RESULTS: Global profiling of the small RNA transcriptome was carried out in rat cerebral cortex from E10 till P28 using next-generation sequencing technique. We found an extraordinary degree of developmental stage-specific expression of a large group of microRNAs. A group of novel microRNAs with functional hints were identified, and brain-enriched expression and Dicer-dependent production of high-abundant novel microRNAs were validated. Profound editing of known microRNAs at "seed" sequence and flanking sequence was observed, with much higher editing events detected at late postnatal stages than embryonic stages, suggesting the necessity of microRNA editing for the fine tuning of gene expression during the formation of complicated synaptic connections at postnatal stages. CONCLUSION: Our analysis reveals extensive regulation of microRNAs during cortical development. The dataset described here will be a valuable resource for clarifying new regulatory mechanisms for cortical development and diseases and will greatly contribute to our understanding of the divergence, modification, and function of microRNAs.


Subject(s)
Cerebral Cortex/embryology , Gene Expression Profiling , Gene Expression Regulation, Developmental , MicroRNAs/metabolism , Animals , Cluster Analysis , Rats , Rats, Sprague-Dawley , Sequence Analysis, RNA
SELECTION OF CITATIONS
SEARCH DETAIL