14 research outputs found

    Molecular and Cellular Mediators of the Gut-Liver Axis in the Progression of Liver Diseases

    Get PDF
    The gut-liver axis covers the bidirectional communication between the gut and the liver, and thus includes signals from liver-to-gut (e.g., bile acids, immunoglobulins) and from gut-to-liver (e.g., nutrients, microbiota-derived products, and recirculating bile acids). In a healthy individual, liver homeostasis is tightly controlled by the mostly tolerogenic liver resident macrophages, the Kupffer cells, capturing the gut-derived antigens from the blood circulation. However, disturbances of the gut-liver axis have been associated to the progression of varying chronic liver diseases, such as non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, and primary sclerosing cholangitis. Notably, changes of the gut microbiome, or intestinal dysbiosis, combined with increased intestinal permeability, leads to the translocation of gut-derived bacteria or their metabolites into the portal vein. In the context of concomitant or subsequent liver inflammation, the liver is then infiltrated by responsive immune cells (e.g., monocytes, neutrophils, lymphoid, or dendritic cells), and microbiota-derived products may provoke or exacerbate innate immune responses, hence perpetuating liver inflammation and fibrosis, and potentiating the risks of developing cirrhosis. Similarly, food derived antigens, bile acids, danger-, and pathogen-associated molecular patterns are able to reshape the liver immune microenvironment. Immune cell intracellular signaling components, such as inflammasome activation, toll-like receptor or nucleotide-binding oligomerization domain-like receptors signaling, are potent targets of interest for the modulation of the immune response. This review describes the current understanding of the cellular landscape and molecular pathways involved in the gut-liver axis and implicated in chronic liver disease progression. We also provide an overview of innovative therapeutic approaches and current clinical trials aiming at targeting the gut-liver axis for the treatment of patients with chronic liver and/or intestinal diseases

    Deciphering the Immune Microenvironment on A Single Archival Formalin-Fixed Paraffin-Embedded Tissue Section by An Immediately Implementable Multiplex Fluorescence Immunostaining Protocol

    Get PDF
    Technological breakthroughs have fundamentally changed our understanding on the complexity of the tumor microenvironment at the single-cell level. Characterizing the immune cell composition in relation to spatial distribution and histological changes may provide important diagnostic and therapeutic information. Immunostaining on formalin-fixed paraffin-embedded (FFPE) tissue samples represents a widespread and simple procedure, allowing the visualization of cellular distribution and processes, on preserved tissue structure. Recent advances in microscopy and molecular biology have made multiplexing accessible, yet technically challenging. We herein describe a novel, simple and cost-effective method for a reproducible and highly flexible multiplex immunostaining on archived FFPE tissue samples, which we optimized for solid organs (e.g., liver, intestine, lung, kidney) from mice and humans. Our protocol requires limited specific equipment and reagents, making multiplexing (>12 antibodies) immediately implementable to any histology laboratory routinely performing immunostaining. Using this method on single sections and combining it with automated whole-slide image analysis, we characterize the hepatic immune microenvironment in preclinical mouse models of liver fibrosis, steatohepatitis and hepatocellular carcinoma (HCC) and on human-patient samples with chronic liver diseases. The data provide useful insights into tissue organization and immune-parenchymal cell-to-cell interactions. It also highlights the profound macrophage heterogeneity in liver across premalignant conditions and HCC

    RĂ©gulation de l'expression membranaire du transporteur de phospholipides biliaires ABCB4 : effet de mutations

    No full text
    ABCB4 is exclusively expressed at the canalicular membrane of hepatocytes where its function is to translocate phosphatidylcholine (PC) into bile. Variations in ABCB4 gene sequence are associated with several chronic and progressive liver diseases. The most severe is PFIC3 which develops early in childhood and most often requires liver transplantation. Less severe diseases are the intrahepatic cholestasis of pregnancy and the low phospholipid- associated cholelithiasis syndrome which occur in young adults. Up to now, about 500 disease-causing ABCB4 variants have been reported. A challenge is to find pharmacological treatments for the severe forms of the diseases. We have studied the effect of five disease-causing variations that reside in the highly conserved motifs of ABC transporters, involved in ATP binding. Using three-dimension structural modeling and in vitro studies, we showed that the five mutants were normally processed and targeted to the plasma membrane, whereas their PC secretion activity was dramatically decreased. PC secretion activity of the mutants was rescued by the clinically approved CFTR potentiator ivacaftor (VX-770). These results pave the way for personalized therapy in ABCB4-related diseases.The second part of my project was aimed at investigating the potential role of two ABCB4 partners, the kinase MRCKalpha and its effector the myosin light chain II (MLCII) in the expression and function of ABCB4. We found that downregulation of both partners didn’t affect the canalicular localization of ABCB4 but led to a reduction of its endocytosis. Our results open new insights into the mechanisms underlying the regulation of ABCB4 expression and function.ABCB4 est exprimĂ© Ă  la membrane canaliculaire des hĂ©patocytes oĂč il sĂ©crĂšte un composant majeur de la bile : la phosphatidylcholine. Plus de 500 mutations d’ABCB4 sont associĂ©es Ă  des maladies biliaires. La pathologie la plus sĂ©vĂšre est la PFIC3, qui se dĂ©veloppe tĂŽt dans l’enfance et progresse rapidement vers l’insuffisance hĂ©patique. La transplantation hĂ©patique reste la seule thĂ©rapie efficace. Le dĂ©veloppement d’alternatives reprĂ©sente donc un enjeu majeur. Cette thĂšse s’intĂ©resse Ă  l’effet de cinq mutations dĂ©crites chez des patients et situĂ©es dans les sites de liaison Ă  l’ATP d’ABCB4. En combinant la modĂ©lisation 3D avec des Ă©tudes in vitro, nous avons montrĂ© que ces mutations sont responsables d’un dĂ©faut d’activitĂ© d’ABCB4. Nous avons mis en Ă©vidence que le VX-770, un mĂ©dicament approuvĂ© en clinique pour traiter les patients atteints de mucoviscidose, restaure l’activitĂ© des cinq mutants. Ces travaux ouvrent des perspectives de repositionnement du VX-770 pour le traitement ciblĂ© de patients atteints de pathologies biliaires liĂ©es Ă  ABCB4. La seconde partie de cette thĂšse consiste Ă  Ă©tudier le rĂŽle de l’interaction du domaine N-terminal d’ABCB4 avec la kinase MRCKalpha. L’inhibition ou l’extinction de cette kinase montrent que MRCKalpha joue un rĂŽle dans la rĂ©gulation de l’expression membranaire d’ABCB4 en contrĂŽlant son internalisation depuis la membrane plasmique. En inhibant la protĂ©ine MLC2, effecteur de MRCKa, nous avons ensuite montrĂ© que l’effet de MRCKalpha sur l’expression d’ABCB4 passe par MLC2, qui est un partenaire du domaine linker d’ABCB4. Notre travail montre un rĂŽle commun de ces deux partenaires dans la rĂ©gulation de l’internalisation d’ABCB4

    Expression and Function of the Biliary Phospholipids Transporter ABCB4 : Effect of Disease-Causing Mutations

    No full text
    ABCB4 est exprimĂ© Ă  la membrane canaliculaire des hĂ©patocytes oĂč il sĂ©crĂšte un composant majeur de la bile : la phosphatidylcholine. Plus de 500 mutations d’ABCB4 sont associĂ©es Ă  des maladies biliaires. La pathologie la plus sĂ©vĂšre est la PFIC3, qui se dĂ©veloppe tĂŽt dans l’enfance et progresse rapidement vers l’insuffisance hĂ©patique. La transplantation hĂ©patique reste la seule thĂ©rapie efficace. Le dĂ©veloppement d’alternatives reprĂ©sente donc un enjeu majeur. Cette thĂšse s’intĂ©resse Ă  l’effet de cinq mutations dĂ©crites chez des patients et situĂ©es dans les sites de liaison Ă  l’ATP d’ABCB4. En combinant la modĂ©lisation 3D avec des Ă©tudes in vitro, nous avons montrĂ© que ces mutations sont responsables d’un dĂ©faut d’activitĂ© d’ABCB4. Nous avons mis en Ă©vidence que le VX-770, un mĂ©dicament approuvĂ© en clinique pour traiter les patients atteints de mucoviscidose, restaure l’activitĂ© des cinq mutants. Ces travaux ouvrent des perspectives de repositionnement du VX-770 pour le traitement ciblĂ© de patients atteints de pathologies biliaires liĂ©es Ă  ABCB4. La seconde partie de cette thĂšse consiste Ă  Ă©tudier le rĂŽle de l’interaction du domaine N-terminal d’ABCB4 avec la kinase MRCKalpha. L’inhibition ou l’extinction de cette kinase montrent que MRCKalpha joue un rĂŽle dans la rĂ©gulation de l’expression membranaire d’ABCB4 en contrĂŽlant son internalisation depuis la membrane plasmique. En inhibant la protĂ©ine MLC2, effecteur de MRCKa, nous avons ensuite montrĂ© que l’effet de MRCKalpha sur l’expression d’ABCB4 passe par MLC2, qui est un partenaire du domaine linker d’ABCB4. Notre travail montre un rĂŽle commun de ces deux partenaires dans la rĂ©gulation de l’internalisation d’ABCB4.ABCB4 is exclusively expressed at the canalicular membrane of hepatocytes where its function is to translocate phosphatidylcholine (PC) into bile. Variations in ABCB4 gene sequence are associated with several chronic and progressive liver diseases. The most severe is PFIC3 which develops early in childhood and most often requires liver transplantation. Less severe diseases are the intrahepatic cholestasis of pregnancy and the low phospholipid- associated cholelithiasis syndrome which occur in young adults. Up to now, about 500 disease-causing ABCB4 variants have been reported. A challenge is to find pharmacological treatments for the severe forms of the diseases. We have studied the effect of five disease-causing variations that reside in the highly conserved motifs of ABC transporters, involved in ATP binding. Using three-dimension structural modeling and in vitro studies, we showed that the five mutants were normally processed and targeted to the plasma membrane, whereas their PC secretion activity was dramatically decreased. PC secretion activity of the mutants was rescued by the clinically approved CFTR potentiator ivacaftor (VX-770). These results pave the way for personalized therapy in ABCB4-related diseases.The second part of my project was aimed at investigating the potential role of two ABCB4 partners, the kinase MRCKalpha and its effector the myosin light chain II (MLCII) in the expression and function of ABCB4. We found that downregulation of both partners didn’t affect the canalicular localization of ABCB4 but led to a reduction of its endocytosis. Our results open new insights into the mechanisms underlying the regulation of ABCB4 expression and function

    Ferroptosis in Cancer Immunotherapy—Implications for Hepatocellular Carcinoma

    No full text
    Ferroptosis is a recently recognized iron-dependent form of non-apoptotic regulated cell death (RCD) characterized by lipid peroxide accumulation to lethal levels. Cancer cells, which show an increased iron dependency to enable rapid growth, seem vulnerable to ferroptosis. There is also increasing evidence that ferroptosis might be immunogenic and therefore could synergize with immunotherapies. Hepatocellular carcinoma (HCC) is the most common primary liver tumor with a low survival rate due to frequent recurrence and limited efficacy of conventional chemotherapies, illustrating the urgent need for novel drug approaches or combinatorial strategies. Immunotherapy is a new treatment approach for advanced HCC patients. In this setting, ferroptosis inducers may have substantial clinical potential. However, there are still many questions to answer before the mystery of ferroptosis is fully unveiled. This review discusses the existing studies and our current understanding regarding the molecular mechanisms of ferroptosis with the goal of enhancing response to immunotherapy of liver cancer. In addition, challenges and opportunities in clinical applications of potential candidates for ferroptosis-driven therapeutic strategies will be summarized. Unraveling the role of ferroptosis in the immune response could benefit the development of promising anti-cancer therapies that overcome drug resistance and prevent tumor metastasis

    Molecular Regulation of Canalicular ABC Transporters

    No full text
    International audienceThe ATP-binding cassette (ABC) transporters expressed at the canalicular membrane of hepatocytes mediate the secretion of several compounds into the bile canaliculi and therefore play a key role in bile secretion. Among these transporters, ABCB11 secretes bile acids, ABCB4 translocates phosphatidylcholine and ABCG5/G8 is responsible for cholesterol secretion, while ABCB1 and ABCC2 transport a variety of drugs and other compounds. The dysfunction of these transporters leads to severe, rare, evolutionary biliary diseases. The development of new therapies for patients with these diseases requires a deep understanding of the biology of these transporters. In this review, we report the current knowledge regarding the regulation of canalicular ABC transporters' folding, trafficking, membrane stability and function, and we highlight the role of molecular partners in these regulating mechanisms

    Ivacaftor-Mediated Potentiation of ABCB4 Missense Mutations Affecting Critical Motifs of the NBDs: Repositioning Perspectives for Hepatobiliary Diseases

    No full text
    ABCB4 (ATP-binding cassette subfamily B member 4) is a hepatocanalicular floppase involved in biliary phosphatidylcholine (PC) secretion. Variations in the ABCB4 gene give rise to several biliary diseases, including progressive familial intrahepatic cholestasis type 3 (PFIC3), an autosomal recessive disease that can be lethal in the absence of liver transplantation. In this study, we investigated the effect and potential rescue of ten ABCB4 missense variations in NBD1:NBD2 homologous positions (Y403H/Y1043H, K435M/K1075M, E558K/E1200A, D564G/D1206G and H589Y/H1231Y) all localized at the conserved and functionally critical motifs of ABC transporters, six of which are mutated in patients. By combining structure analysis and in vitro studies, we found that all ten mutants were normally processed and localized at the canalicular membrane of HepG2 cells, but showed dramatically impaired PC transport activity that was significantly rescued by treatment with the clinically approved CFTR potentiator ivacaftor. Our results provide evidence that functional ABCB4 mutations are rescued by ivacaftor, paving the way for the repositioning of this potentiator for the treatment of selected patients with PFIC3 caused by mutations in the ATP-binding sites of ABCB4

    MRCK-Alpha and Its Effector Myosin II Regulatory Light Chain Bind ABCB4 and Regulate Its Membrane Expression

    No full text
    ABCB4, is an adenosine triphosphate-binding cassette (ABC) transporter localized at the canalicular membrane of hepatocytes, where it mediates phosphatidylcholine secretion into bile. Gene variations of ABCB4 cause different types of liver diseases, including progressive familial intrahepatic cholestasis type 3 (PFIC3). The molecular mechanisms underlying the trafficking of ABCB4 to and from the canalicular membrane are still unknown. We identified the serine/threonine kinase Myotonic dystrophy kinase-related Cdc42-binding kinase isoform α (MRCKα) as a novel partner of ABCB4. The role of MRCKα was explored, either by expression of dominant negative mutant or by gene silencing using the specific RNAi and CRISPR-cas9 strategy in cell models. The expression of a dominant-negative mutant of MRCKα and MRCKα inhibition by chelerythrine both caused a significant increase in ABCB4 steady-state expression in primary human hepatocytes and HEK-293 cells. RNA interference and CRISPR-Cas9 knockout of MRCKα also caused a significant increase in the amount of ABCB4 protein expression. We demonstrated that the effect of MRCKα was mediated by its downstream effector, the myosin II regulatory light chain (MRLC), which was shown to also bind ABCB4. Our findings provide evidence that MRCKα and MRLC bind to ABCB4 and regulate its cell surface expression

    RAB10 Interacts with ABCB4 and Regulates Its Intracellular Traffic

    No full text
    International audienceABCB4 (ATP-binding cassette subfamily B member 4) is an ABC transporter expressed at the canalicular membrane of hepatocytes where it ensures phosphatidylcholine secretion into bile. Genetic variations of ABCB4 are associated with several rare cholestatic diseases. The available treatments are not efficient for a significant proportion of patients with ABCB4-related diseases and liver transplantation is often required. The development of novel therapies requires a deep understanding of the molecular mechanisms regulating ABCB4 expression, intracellular traffic, and function. Using an immunoprecipitation approach combined with mass spectrometry analyses, we have identified the small GTPase RAB10 as a novel molecular partner of ABCB4. Our results indicate that the overexpression of wild type RAB10 or its dominant-active mutant significantly increases the amount of ABCB4 at the plasma membrane expression and its phosphatidylcholine floppase function. Contrariwise, RAB10 silencing induces the intracellular retention of ABCB4 and then indirectly diminishes its secretory function. Taken together, our findings suggest that RAB10 regulates the plasma membrane targeting of ABCB4 and consequently its capacity to mediate phosphatidylcholine secretion
    corecore