45 research outputs found

    Albendazole reduces hepatic inflammation and endoplasmic reticulum-stress in a mouse model of chronic Echinococcus multilocularis infection.

    Get PDF
    BACKGROUND Echinococcus multilocularis causes alveolar echinococcosis (AE), a rising zoonotic disease in the northern hemisphere. Treatment of this fatal disease is limited to chemotherapy using benzimidazoles and surgical intervention, with frequent disease recurrence in cases without radical surgery. Elucidating the molecular mechanisms underlying E. multilocularis infections and host-parasite interactions ultimately aids developing novel therapeutic options. This study explored an involvement of unfolded protein response (UPR) and endoplasmic reticulum-stress (ERS) during E. multilocularis infection in mice. METHODS E. multilocularis- and mock-infected C57BL/6 mice were subdivided into vehicle, albendazole (ABZ) and anti-programmed death ligand 1 (αPD-L1) treated groups. To mimic a chronic infection, treatments of mice started six weeks post i.p. infection and continued for another eight weeks. Liver tissue was then collected to examine inflammatory cytokines and the expression of UPR- and ERS-related genes. RESULTS E. multilocularis infection led to an upregulation of UPR- and ERS-related proteins in the liver, including ATF6, CHOP, GRP78, ERp72, H6PD and calreticulin, whilst PERK and its target eIF2α were not affected, and IRE1α and ATF4 were downregulated. ABZ treatment in E. multilocularis infected mice reversed, or at least tended to reverse, these protein expression changes to levels seen in mock-infected mice. Furthermore, ABZ treatment reversed the elevated levels of interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α and interferon (IFN)-γ in the liver of infected mice. Similar to ABZ, αPD-L1 immune-treatment tended to reverse the increased CHOP and decreased ATF4 and IRE1α expression levels. CONCLUSIONS AND SIGNIFICANCE AE caused chronic inflammation, UPR activation and ERS in mice. The E. multilocularis-induced inflammation and consecutive ERS was ameliorated by ABZ and αPD-L1 treatment, indicating their effectiveness to inhibit parasite proliferation and downregulate its activity status. Neither ABZ nor αPD-L1 themselves affected UPR in control mice. Further research is needed to elucidate the link between inflammation, UPR and ERS, and if these pathways offer potential for improved therapies of patients with AE

    Impact on Bile Acid Concentrations by Alveolar Echinococcosis and Treatment with Albendazole in Mice.

    Get PDF
    Alveolar echinococcosis (AE) caused by Echinococcus multilocularis is a chronic, progressive liver disease widely distributed in the Northern Hemisphere. The main treatment options include surgical interventions and chemotherapy with benzimidazole albendazole (ABZ). To improve the current diagnosis and therapy of AE, further investigations into parasite-host interactions are needed. This study used liquid chromatography-tandem mass spectrometry (LC-MS/MS) to assess serum and liver tissue bile acid profiles in the i.p. chronic E. multilocularis-infected mouse model and evaluated the effects of the anthelmintic drug ABZ. Additionally, hepatic mRNA and protein expression of enzymes and transporters regulating bile acid concentrations were analyzed. AE significantly decreased unconjugated bile acids in serum and liver tissue. Taurine-conjugated bile salts were unchanged or increased in the serum and unchanged or decreased in the liver. Ratios of unconjugated to taurine-conjugated metabolites are proposed as useful serum markers of AE. The expression of the bile acid synthesis enzymes cytochrome P450 (CYP) 7A1 and aldo-keto reductase (AKR) 1D1 tended to decrease or were decreased in mice with AE, along with decreased expression of the bile acid transporters Na+/taurocholate cotransporting polypeptide (NTCP) and bile salt efflux pump (BSEP). Importantly, treatment with ABZ partially or completely reversed the effects induced by E. multilocularis infection. ABZ itself had no effect on the bile acid profiles and the expression of relevant enzymes and transporters. Further research is needed to uncover the exact mechanism of the AE-induced changes in bile acid homeostasis and to test whether serum bile acids and ratios thereof can serve as biomarkers of AE and for monitoring therapeutic efficacy

    Disclosure of Diagnosis and Prognosis to Cancer Patients in Traditional Societies: A Qualitative Assessment from Lebanon

    Get PDF
    Background: The issue of when, how, and whether to disclose full information about cancer diagnosis and prognosis to patients is still debated in some parts of the world, including Lebanon. Despite formal academic emphasis on a larger autonomy for Lebanese patients in deciding the course of their disease, there has been no apparent impact on either clinical practices nor public expectations.  The topic of full disclosure is rarely if ever discussed in open fora, or in mass media channels in Lebanon. Subjects and Method: Seven key stakeholders were identified and interviewed regarding obstacles to spelling out clear guidelines within our national context. The interviews were transcribed and subsequently analyzed for recurrent patterns and concepts.Results: Senior oncologists interviewed generally favored gradual disclosure and most perceived a changing trend among both patients and physicians towards more disclosure. They also agreed on a need for the formal training of residents and fellows to better communicate bad news to patients. All the interviewed physicians attested to the benefits of candid disclosure in terms of patient psychology and overall wellbeing. They also mentioned that psychological services, which may facilitate the disclosure process, are greatly under-utilized in oncology. Lawyers highlighted the vagueness of the current Lebanese legislation regarding the obligation of truthful disclosure in comparison to laws in developed countries and the implications on patient autonomy. Conclusion: The study identified the need for improvements at various levels, including interventions to modify the expectations of the Lebanese public regarding cancer disclosure and to clarify existing legislative texts.Keywords: Ethics; Legislation; Middle-East; DisclosureCorrespondence: James Feghali. Faculty of Medicine, American University of Beirut (AUB), Lebanon, 1101 North Calvert Street, 610, Baltimore, Maryland, 21202. E-mail: [email protected]. Telephone: +1-(267)-595-9995.Journal of Epidemiology and Public Health (2019), 4(2): 109-116https://doi.org/10.26911/jepublichealth.2019.04.02.0

    Innate and adaptive immune responses following PD-L1 blockade in treating chronic murine alveolar echinococcosis.

    Get PDF
    BACKGROUND Programmed death-1 (PD-1) and programmed death ligand-1 (PD-L1) immune checkpoint blockade is efficacious in certain cancer therapies. OBJECTIVES The present study aimed to provide a picture about the development of innate and adaptive immune responses upon PD-L1 blockade in treating chronic murine AE. METHODS Immune treatment started at 6 weeks post E. multilocularis-infection, and was maintained for 8 weeks with twice per week anti-PD-L1 administration (intraperitoneal). The study included an outgroup-control with mice perorally medicated with albendazole five days/week, and another one with both treatments combined. Assessment of treatment efficacy was based on determining parasite weight, innate and adaptive immune cell profiles, histopathology, and liver tissue cytokine levels. RESULTS/CONCLUSIONS Findings showed that the parasite load was significantly reduced in response to PD-L1 blockade, and this blockade a) contributed to T cell activity by increasing CD4+ /CD8+ effector T cells, and decreasing Tregs; b) had the capacity to re-store DCs and Kupffer cells/Macrophages; c) suppressed NKT and NK cells; and thus d) lead to an improved control of E. multilocularis infection in mice. This study suggests that the PD-L1 pathway plays an important role by regulating adaptive and innate immune cells against E. multilocularis infection, with significant modulation of tissue inflammation

    Characteristics and comparative clinical outcomes of prisoner versus non-prisoner populations hospitalized with COVID-19

    Get PDF
    Prisons in the United States have become a hotbed for spreading COVID-19 among incarcerated individuals. COVID-19 cases among prisoners are on the rise, with more than 143,000 confirmed cases to date. However, there is paucity of data addressing clinical outcomes and mortality in prisoners hospitalized with COVID-19. An observational study of all patients hospitalized with COVID-19 between March 10 and May 10, 2020 at two Henry Ford Health System hospitals in Michigan. Clinical outcomes were compared amongst hospitalized prisoners and non-prisoner patients. The primary outcomes were intubation rates, in-hospital mortality, and 30-day mortality. Multivariable logistic regression and Cox-regression models were used to investigate primary outcomes. Of the 706 hospitalized COVID-19 patients (mean age 66.7 ± 16.1 years, 57% males, and 44% black), 108 were prisoners and 598 were non-prisoners. Compared to non-prisoners, prisoners were more likely to present with fever, tachypnea, hypoxemia, and markedly elevated inflammatory markers. Prisoners were more commonly admitted to the intensive care unit (ICU) (26.9% vs. 18.7%), required vasopressors (24.1% vs. 9.9%), and intubated (25.0% vs. 15.2%). Prisoners had higher unadjusted inpatient mortality (29.6% vs. 20.1%) and 30-day mortality (34.3% vs. 24.6%). In the adjusted models, prisoner status was associated with higher in-hospital death (odds ratio, 2.32; 95% confidence interval (CI), 1.33 to 4.05) and 30-day mortality (hazard ratio, 2.00; 95% CI, 1.33 to 3.00). In this cohort of hospitalized COVID-19 patients, prisoner status was associated with more severe clinical presentation, higher rates of ICU admissions, vasopressors requirement, intubation, in-hospital mortality, and 30-day mortality

    Loss of claudin-3 impairs hepatic metabolism, biliary barrier function and cell proliferation in the murine liver.

    Get PDF
    BACKGROUND & AIMS Tight junctions in the liver are essential to maintain the blood-biliary-barrier, however the functional contribution of individual tight junction proteins to barrier- and metabolic homeostasis remains largely unexplored. Here, we describe the cell type specific expression of tight junction genes in the murine liver, and explore the regulation and functional importance of the transmembrane protein claudin-3 in liver metabolism, barrier function and cell proliferation. METHODS The cell type specific expression of hepatic tight junction genes is described using our mouse liver single cell sequencing dataset. Differential gene expression in Cldn3-/- and Cldn3+/+ livers was assessed in young and aged mice by RNA-seq and hepatic tissue was analysed for lipid content and bile acid composition. A surgical model of partial hepatectomy (PHx) was used to induce liver cell proliferation. RESULTS Claudin-3 is a highly expressed tight junction protein found in the liver and is expressed predominantly in hepatocytes and cholangiocytes. The histology of Cldn3-/- livers showed no overt phenotype, and the canalicular tight junctions appeared intact. Nevertheless, by RNAseq we detected a downregulation of metabolic pathways in the livers of Cldn3-/- young and aged mice as well as a decrease in lipid content and a weakened biliary-barrier for primary bile acids, such as TCA, TCDCA and TMCA. Coinciding with defects in the biliary barrier and lower lipid metabolism, there was a diminished hepatocyte proliferative response in Cldn3-/- mice following PHx. CONCLUSION Our data shows that in the liver, claudin-3 is necessary to maintain metabolic homeostasis, retention of bile acids, and optimal hepatocyte proliferation during liver regeneration

    Human and Mouse CD8+CD25+FOXP3+ Regulatory T Cells at Steady State and during Interleukin-2 Therapy

    Get PDF
    International audienceIn addition to CD4+ regulatory T cells (Tregs), CD8+ suppressor T cells are emerging as an important subset of regulatory T cells. Diverse populations of CD8+ T cells with suppressive activities have been described. Among them, a small population of CD8+CD25+FOXP3+ T cells is found both in mice and humans. In contrast to thymic-derived CD4+CD25+FOXP3+ Tregs, their origin and their role in the pathophysiology of autoimmune diseases (AIDs) are less understood. We report here the number, phenotype, and function of CD8+ Tregs cells in mice and humans, at the steady state and in response to low-dose interleukin-2 (IL-2). CD8+ Tregs represent approximately 0.4 and 0.1% of peripheral blood T cells in healthy humans and mice, respectively. In mice, their frequencies are quite similar in lymph nodes (LNs) and the spleen, but two to threefold higher in Peyer patches and mesenteric LNs. CD8+ Tregs express low levels of CD127. CD8+ Tregs express more activation or proliferation markers such as CTLA-4, ICOS, and Ki-67 than other CD8+ T cells. In vitro, they suppress effector T cell proliferation as well as or even better than CD4+ Tregs. Owing to constitutive expression of CD25, CD8+ Tregs are 20- to 40-fold more sensitive to in vitro IL-2 stimulation than CD8+ effector T cells, but 2–4 times less than CD4+ Tregs. Nevertheless, low-dose IL-2 dramatically expands and activates CD8+ Tregs even more than CD4+ Tregs, in mice and humans. Further studies are warranted to fully appreciate the clinical relevance of CD8+ Tregs in AIDs and the efficacy of IL-2 treatment
    corecore