Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
J Proteome Res ; 23(4): 1433-1442, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38488493

ABSTRACT

MDR3 (multidrug resistance 3) deficiency in humans (MDR2 in mice) causes progressive familial intrahepatic cholestasis type 3 (PFIC3). PFIC3 is a lethal disease characterized by an early onset of intrahepatic cholestasis progressing to liver cirrhosis, a preneoplastic condition, putting individuals at risk of hepatocellular carcinoma (HCC). Hepatocyte-like organoids from MDR2-deficient mice (MDR2KO) were used in this work to study the molecular alterations caused by the deficiency of this transporter. Proteomic analysis by mass spectrometry allowed characterization of 279 proteins that were differentially expressed in MDR2KO compared with wild-type organoids. Functional enrichment analysis indicated alterations in three main cellular functions: (1) interaction with the extracellular matrix, (2) remodeling intermediary metabolism, and (3) cell proliferation and differentiation. The affected cellular processes were validated by orthogonal molecular biology techniques. Our results point to molecular mechanisms associated with PFIC3 that may drive the progression to liver cirrhosis and HCC and suggest proteins and cellular processes that could be targeted for the development of early detection strategies for these severe liver diseases.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B , Carcinoma, Hepatocellular , Cholestasis, Intrahepatic , Cholestasis , Liver Neoplasms , Animals , Humans , Mice , ATP Binding Cassette Transporter, Subfamily B/deficiency , Carcinoma, Hepatocellular/pathology , Cholestasis/genetics , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice, Knockout , Proteomics
2.
Sci Adv ; 10(4): eadk9394, 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38266080

ABSTRACT

The smooth and precise transition from totipotency to pluripotency is a key process in embryonic development, generating pluripotent stem cells capable of forming all cell types. While endogenous retroviruses (ERVs) are essential for early development, their precise roles in this transition remains mysterious. Using cutting-edge genetic and biochemical techniques in mice, we identify MERVL-gag, a retroviral protein, as a crucial modulator of pluripotent factors OCT4 and SOX2 during lineage specification. MERVL-gag tightly operates with URI, a prefoldin protein that concurs with pluripotency bias in mouse blastomeres, and which is indeed required for totipotency-to-pluripotency transition. Accordingly, URI loss promotes a stable totipotent-like state and embryo arrest at 2C stage. Mechanistically, URI binds and shields OCT4 and SOX2 from proteasome degradation, while MERVL-gag displaces URI from pluripotent factor interaction, causing their degradation. Our findings reveal the symbiotic coevolution of ERVs with their host cells to ensure the smooth and timely progression of early embryo development.


Subject(s)
Endogenous Retroviruses , Pluripotent Stem Cells , Female , Pregnancy , Animals , Mice , Endogenous Retroviruses/genetics , Embryo, Mammalian , Embryonic Development/genetics
3.
iScience ; 26(7): 107010, 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37332670

ABSTRACT

Cold therapy has been used for centuries, from Julius Caesar to Mohandas Gandhi, as a potent therapeutic approach. However, it has been largely forgotten in modern medicine. This review explores the history of cold therapy and its potential application as a therapeutic strategy against various diseases, including cancer. We examine the different techniques of cold exposure and the use of other therapeutical approaches, such as cryoablation, cryotherapy, cryoimmunotherapy, cryothalectomy, and delivery of cryogen agents. While clinical trials using cold therapy for cancer treatment are still limited, recent research shows promising results in experimental animal cancer models. This area of research is becoming increasingly significant and warrants further investigation.

4.
Trends Mol Med ; 29(7): 489-511, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37263858

ABSTRACT

In recent years dietary interventions have become a promising tool in cancer treatment and have demonstrated a powerful ability to alter metabolism and tumor growth, development, and therapeutic response. However, because the mechanisms underlying dietary therapeutics are poorly understood, they are frequently ignored as a potential line of treatment for cancer. We discuss the proposed mechanisms behind the anticancer effects of various diets and their development for clinical use. This review aims to provide researchers and clinicians in the field of oncology with a complete overview of the contemporary landscape of nutritional interventions and precision nutrition as cancer therapeutics, and offers a perspective on the steps necessary to establish nutritional interventions as a standard line of treatment.


Subject(s)
Caloric Restriction , Neoplasms , Humans , Diet , Neoplasms/therapy
5.
iScience ; 26(4): 106511, 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37091251

ABSTRACT

In their latest article, Seki and colleagues investigate the potential role of cold as a therapeutical option to treat various cancer types, including even clinically untreatable cancers such as pancreatic cancers. The authors suggest that cold exposure may have a tumor-suppressive effect mediated by the activation of brown adipose tissue (BAT), in charge of dissipating heat through non-shivering thermogenesis. In this regard, circulating blood glucose is decreased, restricting the tumor glucose uptake, which is redistributed, favoring BAT uptake to fuel thermogenesis.1.

6.
Trends Cancer ; 9(2): 140-157, 2023 02.
Article in English | MEDLINE | ID: mdl-36347768

ABSTRACT

The liver is the largest organ of the mammalian body and has the remarkable ability to fully regenerate in order to maintain tissue homeostasis. The adult liver consists of hexagonal lobules, each with a central vein surrounded by six portal triads localized in the lobule border containing distinct parenchymal and nonparenchymal cells. Because the liver is continuously exposed to diverse stress signals, several sophisticated regenerative processes exist to restore its functional status following impairment. However, these stress signals can affect the liver's capacity to regenerate and may lead to the development of hepatocellular carcinoma (HCC), one of the most aggressive liver cancers. Here, we review the mechanisms of hepatic regeneration and their potential to influence HCC development.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Adult , Animals , Humans , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Hepatocytes/pathology , Liver Regeneration , Mammals
7.
J Exp Med ; 219(11)2022 11 07.
Article in English | MEDLINE | ID: mdl-36098959

ABSTRACT

Intestinal epithelium regenerates rapidly through proliferation of intestinal stem cells (ISCs), orchestrated by potent mitogens secreted within the crypt niche. However, mechanisms regulating these mitogenic factors remain largely unknown. Here, we demonstrate that transit-amplifying (TA) cells, marked by unconventional prefoldin RPB5 interactor (URI), control R-spondin production to guide ISC proliferation. Genetic intestinal URI ablation in mice injures TA cells, reducing their survival capacity, leading to an inflamed tissue and subsequently decreasing R-spondin levels, thereby causing ISC quiescence and disruption of intestinal structure. R-spondin supplementation or restoration of R-spondin levels via cell death inhibition by c-MYC elimination or the suppression of inflammation reinstates ISC proliferation in URI-depleted mice. However, selective c-MYC and p53 suppression are required to fully restore TA cell survival and differentiation capacity and preserve complete intestinal architecture. Our data reveal an unexpected role of TA cells, which represent a signaling platform instrumental for controlling inflammatory cues and R-spondin production, essential for maintaining ISC proliferation and tissue regeneration.


Subject(s)
Intestinal Mucosa , Intestines , Animals , Cell Proliferation , Intestinal Mucosa/metabolism , Mice , Signal Transduction , Stem Cells
8.
J Hepatol ; 76(4): 850-861, 2022 04.
Article in English | MEDLINE | ID: mdl-34958836

ABSTRACT

BACKGROUND & AIMS: Owing to the lack of genetic animal models that adequately recreate key clinical characteristics of cirrhosis, the molecular pathogenesis of cirrhosis has been poorly characterized, and treatments remain limited. Hence, we aimed to better elucidate the pathological mechanisms of cirrhosis using a novel murine model. METHODS: We report on the first murine genetic model mimicking human cirrhosis induced by hepatocyte-specific elimination of microspherule protein 1 (MCRS1), a member of non-specific lethal (NSL) and INO80 chromatin-modifier complexes. Using this genetic tool with other mouse models, cell culture and human samples, combined with quantitative proteomics, single nuclei/cell RNA sequencing and chromatin immunoprecipitation assays, we investigated mechanisms of cirrhosis. RESULTS: MCRS1 loss in mouse hepatocytes modulates the expression of bile acid (BA) transporters - with a pronounced downregulation of Na+-taurocholate cotransporting polypeptide (NTCP) - concentrating BAs in sinusoids and thereby activating hepatic stellate cells (HSCs) via the farnesoid X receptor (FXR), which is predominantly expressed in human and mouse HSCs. Consistently, re-expression of NTCP in mice reduces cirrhosis, and genetic ablation of FXR in HSCs suppresses fibrotic marks in mice and in vitro cell culture. Mechanistically, deletion of a putative SANT domain from MCRS1 evicts histone deacetylase 1 from its histone H3 anchoring sites, increasing histone acetylation of BA transporter genes, modulating their expression and perturbing BA flow. Accordingly, human cirrhosis displays decreased nuclear MCRS1 and NTCP expression. CONCLUSIONS: Our data reveal a previously unrecognized function of MCRS1 as a critical histone acetylation regulator, maintaining gene expression and liver homeostasis. MCRS1 loss induces acetylation of BA transporter genes, perturbation of BA flow, and consequently, FXR activation in HSCs. This axis represents a central and universal signaling event in cirrhosis, which has significant implications for cirrhosis treatment. LAY SUMMARY: By genetic ablation of MCRS1 in mouse hepatocytes, we generate the first genetic mouse model of cirrhosis that recapitulates human features. Herein, we demonstrate that the activation of the bile acid/FXR axis in liver fibroblasts is key in cirrhosis development.


Subject(s)
Histones , RNA-Binding Proteins , Receptors, Cytoplasmic and Nuclear , Acetylation , Animals , Bile Acids and Salts/metabolism , Carrier Proteins , Histones/metabolism , Liver/pathology , Liver Cirrhosis/pathology , Membrane Glycoproteins , Mice , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism
9.
iScience ; 24(11): 103273, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34761191

ABSTRACT

Prefoldins (PFDNs) are evolutionary conserved co-chaperones, initially discovered in archaea but universally present in eukaryotes. PFDNs are prevalently organized into hetero-hexameric complexes. Although they have been overlooked since their discovery and their functions remain elusive, several reports indicate they act as co-chaperones escorting misfolded or non-native proteins to group II chaperonins. Unlike the eukaryotic PFDNs which interact with cytoskeletal components, the archaeal PFDNs can bind and stabilize a wide range of substrates, possibly due to their great structural diversity. The discovery of the unconventional RPB5 interactor (URI) PFDN-like complex (UPC) suggests that PFDNs have versatile functions and are required for different cellular processes, including an important role in cancer. Here, we summarize their functions across different species. Moreover, a comprehensive analysis of PFDNs genomic alterations across cancer types by using large-scale cancer genomic data indicates that PFDNs are a new class of non-mutated proteins significantly overexpressed in some cancer types.

10.
Cancers (Basel) ; 13(20)2021 Oct 09.
Article in English | MEDLINE | ID: mdl-34680192

ABSTRACT

Because the liver is considered to be one of the most important metabolic organs in the body, it is continuously exposed to damaging environmental agents. Upon damage, several complex cellular and molecular mechanisms in charge of liver recovery and regeneration are activated to prevent the failure of the organ. When liver injury becomes chronic, the regenerative response goes awry and impairs the liver function, consequently leading to cirrhosis, a liver disorder that can cause patient death. Cirrhosis has a disrupted liver architecture and zonation, along with the presence of fibrosis and parenchymal nodules, known as regenerative nodules (RNs). Inflammatory cues contribute to the cirrhotic process in response to chronic damaging agents. Cirrhosis can progress to HCC, the most common and one of the most lethal liver cancers with unmet medical needs. Considering the essential role of inflammatory pathways in the development of cirrhosis, further understanding of the relationship between immune cells and the activation of RNs and fibrosis would guide the design of innovative therapeutic strategies to ameliorate the survival of cirrhotic and HCC patients. In this review, we will summarize the inflammatory mechanisms implicated in the development of cirrhosis.

11.
STAR Protoc ; 2(3): 100631, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34258592

ABSTRACT

Chromosomal instability (CIN), a type of genomic instability, favors changes in chromosome number and structure and it is associated with the progression and initiation of multiple diseases, including cancer. Therefore, CIN identification and analysis represents a useful tool for cancer diagnosis and treatment. Here, we report an optimized molecular cytogenetic protocol to detect CIN in formalin-fixed, paraffin-embedded mouse and human tissues, using fluorescent in situ hybridization to visualize and quantify chromosomal alterations such as amplifications, deletions, and translocations. For complete information on the generation and use of this protocol, please refer to Brandt et al. (2018).


Subject(s)
Chromosomal Instability , In Situ Hybridization, Fluorescence/methods , Interphase , Animals , Chromosomes, Artificial, Bacterial , DNA/genetics , Electrophoresis, Agar Gel , Mice , Paraffin Embedding
12.
Commun Biol ; 4(1): 747, 2021 06 16.
Article in English | MEDLINE | ID: mdl-34135460

ABSTRACT

Tumour recurrence is a serious impediment to cancer treatment, but the mechanisms involved are poorly understood. The most frequently used anti-tumour therapies-chemotherapy and radiotherapy-target highly proliferative cancer cells. However non- or slow-proliferative dormant cancer cells can persist after treatment, eventually causing tumour relapse. Whereas the reversible growth arrest mechanism allows quiescent cells to re-enter the cell cycle, senescent cells are largely thought to be irreversibly arrested, and may instead contribute to tumour growth and relapse through paracrine signalling mechanisms. Thus, due to the differences in their growth arrest mechanism, metabolic features, plasticity and adaptation to their respective tumour microenvironment, dormant-senescent and -quiescent cancer cells could have different but complementary roles in fuelling tumour growth. In this review article, we discuss the implication of dormant cancer cells in tumour relapse and the need to understand how quiescent and senescent cells, respectively, may play a part in this process.


Subject(s)
Cell Plasticity/physiology , Cell Proliferation/physiology , Cellular Senescence/physiology , Neoplasm Recurrence, Local/pathology , Neoplasms/pathology , Cell Cycle/physiology , Humans , Neoplasms/therapy , Signal Transduction , Tumor Microenvironment/physiology
13.
Nat Metab ; 3(4): 496-512, 2021 04.
Article in English | MEDLINE | ID: mdl-33859430

ABSTRACT

Overnutrition causes obesity, a global health problem without any effective therapy. Obesity is characterized by low-grade inflammation, which predisposes individuals to metabolic syndrome via unknown mechanisms. Here, we demonstrate that abolishing the interleukin-17A (IL-17A) axis in mice by inhibition of RORγt-mediated IL-17A production by digoxin, or by ubiquitous deletion of IL-17 receptor A (Il17ra), suppresses diet-induced obesity (DIO) and metabolic disorders, and promotes adipose-tissue browning, thermogenesis and energy expenditure. Genetic ablation of Il17ra specifically in adipocytes is sufficient to completely prevent DIO and metabolic dysfunction in mice. IL-17A produced in response to DIO induces PPARγ phosphorylation at Ser273 in adipocytes in a CDK5-dependent manner, thereby modifying expression of diabetogenic and obesity genes, which correlates with IL-17A signalling in white adipose tissues of individuals with morbid obesity. These findings reveal an unanticipated role for IL-17A in adipocyte biology, in which its direct action pathogenically reprograms adipocytes, promoting DIO and metabolic syndrome. Targeting the IL-17A axis could be an efficient antiobesity strategy.


Subject(s)
Adipocytes/drug effects , Interleukin-17/antagonists & inhibitors , Metabolic Diseases/prevention & control , Obesity/prevention & control , Adipose Tissue, Brown/physiology , Animals , Cyclin-Dependent Kinase 5/metabolism , Diet , Diet, High-Fat , Digoxin/pharmacology , Energy Metabolism/physiology , Feces/chemistry , Gene Deletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Overnutrition , PPAR gamma/metabolism , Phosphorylation , Thermogenesis/physiology
14.
Trends Cancer ; 7(1): 29-36, 2021 01.
Article in English | MEDLINE | ID: mdl-32917550

ABSTRACT

The liver is one of the major metabolic organs in the body, susceptible to injury caused by various factors. In response to injury, sophisticated mechanisms are engaged to repair and regenerate the damaged liver, preventing its failure. When the damage is chronic, regeneration goes awry, impairing liver function and causing cirrhosis. Hence, cirrhosis may rather be a protective response to injury, where wound-healing processes are set to primarily repair the damaged liver. Although cirrhosis is clinically considered a risk factor for hepatocellular carcinoma (HCC), comprehensive population-based studies demonstrate a very modest incidence, refuting the idea that cirrhosis progresses to HCC. Here, we discuss and shed light on the provocative question of whether cirrhosis predisposes to HCC.


Subject(s)
Carcinoma, Hepatocellular/epidemiology , Liver Cirrhosis/epidemiology , Liver Neoplasms/epidemiology , Liver Regeneration/genetics , Liver/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Disease Progression , Genetic Predisposition to Disease , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Transgenic , Protective Factors , Risk Factors
15.
Cell Rep Med ; 1(7): 100125, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33205075

ABSTRACT

Enteroviruses are suspected to contribute to insulin-producing ß cell loss and hyperglycemia-induced diabetes. However, mechanisms are not fully defined. Here, we show that coxsackievirus B type 4 (CVB4) infection in human islet-engrafted mice and in rat insulinoma cells displays loss of unconventional prefoldin RPB5 interactor (URI) and PDX1, affecting ß cell function and identity. Genetic URI ablation in the mouse pancreas causes PDX1 depletion in ß cells. Importantly, diabetic PDX1 heterozygous mice overexpressing URI in ß cells are more glucose tolerant. Mechanistically, URI loss triggers estrogen receptor nuclear translocation leading to DNA methyltransferase 1 (DNMT1) expression, which induces Pdx1 promoter hypermethylation and silencing. Consequently, demethylating agent procainamide-mediated DNMT1 inhibition reinstates PDX1 expression and protects against diabetes in pancreatic URI-depleted mice . Finally, the ß cells of human diabetes patients show correlations between viral protein 1 and URI, PDX1, and DNMT1 levels. URI and DNMT1 expression and PDX1 silencing provide a causal link between enterovirus infection and diabetes.


Subject(s)
Capsid Proteins/genetics , Coxsackievirus Infections/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Diabetes Mellitus, Type 2/genetics , Enterovirus B, Human/genetics , Homeodomain Proteins/genetics , Repressor Proteins/genetics , Trans-Activators/genetics , Animals , Capsid Proteins/metabolism , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/pathology , Coxsackievirus Infections/virology , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/virology , Disease Models, Animal , Enterovirus B, Human/metabolism , Enterovirus B, Human/pathogenicity , Female , Gene Expression Regulation , Glucose/metabolism , Glucose/pharmacology , Homeodomain Proteins/metabolism , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/transplantation , Male , Mice , Mice, Transgenic , Procainamide/pharmacology , Rats , Repressor Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Transplantation, Heterologous
16.
Immunity ; 52(2): 342-356.e6, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32023490

ABSTRACT

Interleukin-17A (IL-17A) is a major mediator of tissue inflammation in many autoimmune diseases. Anti-IL-17A is an effective treatment for psoriasis and is showing promise in clinical trials in multiple sclerosis. In this study, we find that IL-17A-defective mice or mice treated with anti-IL-17A at induction of experimental autoimmune encephalomyelitis (EAE) are resistant to disease and have defective priming of IL-17-secreting γδ T (γδT17) cells and Th17 cells. However, T cells from Il17a-/- mice induce EAE in wild-type mice following in vitro culture with autoantigen, IL-1ß, and IL-23. Furthermore, treatment with IL-1ß or IL-17A at induction of EAE restores disease in Il17a-/- mice. Importantly, mobilization of IL-1ß-producing neutrophils and inflammatory monocytes and activation of γδT17 cells is reduced in Il17a-/- mice. Our findings demonstrate that a key function of IL-17A in central nervous system (CNS) autoimmunity is to recruit IL-1ß-secreting myeloid cells that prime pathogenic γδT17 and Th17 cells.


Subject(s)
Autoimmunity/immunology , Interleukin-17/immunology , Interleukin-1beta/metabolism , Intraepithelial Lymphocytes/immunology , Myeloid Cells/immunology , Th17 Cells/immunology , Animals , Autoantigens/immunology , Autoimmunity/genetics , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/antagonists & inhibitors , Interleukin-17/deficiency , Interleukin-17/metabolism , Interleukin-1beta/immunology , Interleukin-23/immunology , Interleukin-23/metabolism , Intraepithelial Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Myeloid Cells/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Th17 Cells/metabolism
17.
iScience ; 21: 168-187, 2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31669832

ABSTRACT

The intestinal epithelium is a very dynamic tissue under a high regenerative pressure, which makes it susceptible to malignant transformation. Proper integration of various cell signaling pathways and a balanced cross talk between different cell types composing the organ are required to maintain intestinal homeostasis. Dysregulation of this balance can lead to colorectal cancer (CRC). Here, we review important insights into molecular and cellular mechanisms of CRC. We discuss how perturbation in complex regulatory networks, including the Wnt, Notch, BMP, and Hedgehog pathways; and how variations in inflammatory signaling, nutrients, and microbiota can affect intestinal homeostasis contributing to the malignant transformation of intestinal cells.

18.
Science ; 364(6443)2019 05 31.
Article in English | MEDLINE | ID: mdl-31147493

ABSTRACT

Ionizing radiation (IR) can cause gastrointestinal syndrome (GIS), a lethal disorder, by means of unknown mechanisms. We show that high-dose irradiation increases unconventional prefoldin RPB5 interactor (URI) levels in mouse intestinal crypt, but organ regeneration correlates with URI reductions. URI overexpression in intestine protects mice from radiation-induced GIS, whereas halving URI expression sensitizes mice to IR. URI specifically inhibits ß-catenin in stem cell-like label-retaining (LR) cells, which are essential for organ regeneration after IR. URI reduction activates ß-catenin-induced c-MYC expression, causing proliferation of and DNA damage to LR cells, rendering them radiosensitive. Therefore, URI labels LR cells which promote tissue regeneration in response to high-dose irradiation, and c-MYC inhibitors could be countermeasures for humans at risk of developing GIS.


Subject(s)
Gastrointestinal Diseases/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/radiation effects , Radiation Injuries/metabolism , Radiation Tolerance , Regeneration , Repressor Proteins/metabolism , Animals , Gastrointestinal Diseases/genetics , Gene Knock-In Techniques , Intestinal Mucosa/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Radiation Injuries/genetics , Radiation, Ionizing , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Repressor Proteins/genetics , beta Catenin/metabolism
19.
Adv Exp Med Biol ; 1106: 95-108, 2018.
Article in English | MEDLINE | ID: mdl-30484155

ABSTRACT

Almost 15 years ago, the URI prefoldin-like complex was discovered by Krek and colleagues in immunoprecipitation experiments conducted in mammalian cells with the aim of identifying new binding partners of the E3 ubiquitin-protein ligase S-phase kinase-associated protein 2 (SKP2) (Gstaiger et al. Science 302(5648):1208-1212, 2003). The URI prefoldin-like complex is a heterohexameric chaperone complex comprising two α and four ß subunits (α2ß4). The α subunits are URI and STAP1, while the ß subunits are PFDN2, PFDN6, and PFDN4r, one of which is probably present in duplicate. Elucidating the roles and functions of these components in vitro and in vivo will help to clarify the mechanistic behavior of what appears to be a remarkably important cellular machine.


Subject(s)
Molecular Chaperones/chemistry , Animals , Immunoprecipitation
20.
EMBO Mol Med ; 10(7)2018 07.
Article in English | MEDLINE | ID: mdl-29907597

ABSTRACT

The protein kinase p38α plays a key role in cell homeostasis, and p38α signaling in intestinal epithelial cells protects against colitis-induced tumorigenesis. However, little is known on the contribution of p38α signaling in intestinal stromal cells. Here, we show that myeloid cell-specific downregulation of p38α protects mice against inflammation-associated colon tumorigenesis. The reduced tumorigenesis correlates with impaired detection in the colon of crucial chemokines for immune cell recruitment. We identify insulin-like growth factor-1 (IGF-1) as a novel mediator of the p38α pathway in macrophages. Moreover, using genetic and pharmacological approaches, we confirm the implication of IGF-1 produced by myeloid cells in colon inflammation and tumorigenesis. We also show a correlation between IGF-1 pathway activation and the infiltration of myeloid cells with active p38α in colon samples from patients with ulcerative colitis or colon cancer. Altogether, our results uncover an important role for myeloid IGF-1 downstream of p38α in colitis-associated tumorigenesis and suggest the interest in evaluating IGF-1 therapies for inflammation-associated intestinal diseases, taking into consideration IGF-1 signaling and immune cell infiltration in patient biopsies.


Subject(s)
Carcinogenesis/metabolism , Colitis/complications , Colonic Neoplasms/etiology , Insulin-Like Growth Factor I/metabolism , Intestinal Mucosa/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , Myeloid Cells/metabolism , Animals , Carcinogenesis/immunology , Chemokines/metabolism , Colitis/immunology , Colitis/metabolism , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Down-Regulation , Female , Humans , Intestines , Male , Mice , Mice, Inbred C57BL , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...