140 research outputs found

    Effects of dietary ingredients and feed additives on the health and production of European sea bass (Dicentrarchus labrax) for applications in aquaculture

    Get PDF
    Experiment one revealed fishmeal (FM) replacement with soy protein concentrate (SPC) alone, and in combination with pea protein concentrate (PPC) and saponins (S) modulated the intestinal bacterial communities of D. labrax, increasing the presence of lactic acid bacteria. Intestinal histology revealed significantly reduced goblet cell’s (GC’s) in fish fed the SPC+S, epithelial microvilli densities (MD) in fish fed the SPC+PPC, SPC+PPC+S and SPC+S after two weeks feeding. Significant reductions in GC’s and intraepithelial leukocytes (IEL’s) in fish fed the SPC+S, and MD’s in fish fed the SPC+S and SPC+PPC+S after four weeks feeding, relative to fish fed the FM control. Furthermore, fish fed all plant based diets appeared to exhibit a loss of membrane integrity at the microvilli tips, most pronounced in fish fed the SPC+S diet. These results suggest a sub-acute enteritis response in the posterior intestine of D. labrax, which was deemed to be most pronounced in fish fed the SPC+S diets. Experiment two utilised the SPC+S diet as a sub-optimal basal diet to assess the potential of the probiotic Bacillus subtilis and the prebiotic Previda®, individually and in combination, in alleviating the enteritis-like effects induced by this diet, observed in the first experiment. Microbiological analyses revealed B. subtilis modulated the allochthonous bacterial communities. Fish fed the combination of B. subtilis and Previda® (synbiotic) diet exhibited a significantly increased intestinal perimeter ratio, compared to fish fed the basal. Significantly elevated GC’s in fish fed the probiotic and synbiotic treatments, and significantly elevated epithelial MD’s, and intestinal absorptive surface index in fish fed the probiotic diet was observed, relative to fed fish the basal. The loss of membrane integrity induced by the basal diet, was reduced in fish fed the probiotic, prebiotic and synbiotic diets. The intestinal gene expression of the pro-inflammatory cytokines IL-1β and TNFα was significantly up-regulated in fish fed all experimental diets, relative to fish fed the basal. The intestinal gene expression of HSP70, CASP3 and PCNA was significantly down-regulated in fish fed the probiotic, prebiotic and synbiotic relative to fish fed the basal. At the end of the experiment intestinal samples were exposed to one of four treatments [1. PBS (control), 2. B. subtilis, 3. Vibrio anguillarum and 4. B. subtilis + V. anguillarum], ex vivo, to determine if the feed additives could mitigate enteric pathogen damage. All feed additives revealed the potential to reduce the morphological damage caused by the pathogen. Experiment three assessed B. subtilis and the phytobiotic Next Enhance 150® on the growth and health of D. labrax. B. subtilis modulated the allochthonous bacterial communities and reduced the presence of some potential pathogens. The intestinal gene expression of HSP70, CASP3, PCNA and CAL was significantly down-regulation in fish fed the probiotic diet relative to fish fed the control. Significantly elevated IEL’s were observed in fish fed the probiotic and Next Enhance 150® diets relative to fish fed the control. Growth performance was remained unaffected. The present research demonstrates that dietary B. subtilis modulates the allochthonous bacterial communities, as well as, improving the intestinal morphology and localised immunity in European sea bass. Dietary Previda® and Next Enhance 150® were also observed to confer beneficial effects on the gut health of this species. No detrimental effects were observed as a consequence of any of the feed additives used in the present research.Novus International, Society for Applied Microbiolog

    Gut Associated Lymphoid Tissue (GALT) primary cells and stable cell lines as predictive models for intestinal health in rainbow trout (Oncorhynchus mykiss)

    Get PDF
    Funding This work was funded by the University of Aberdeen. Acknowledgments This work was funded by Skretting AI and the University of Aberdeen. Technical support by Dr Dawn Shewring was greatly appreciated. We also thank Dr Tiehui Wang, Scottish Fish Immunology Research Centre for providing the rIL-1β.Peer reviewedPublisher PD

    Procedures and accuracy estimates for beta-beat correction in the LHC

    Get PDF
    The LHC aperture imposes a tight tolerance of 20% on the maximum acceptable beta-beat in the machine. An accurate knowledge of the transfer functions for the individually powered insertion quadrupoles and techniques to compensate beta-beat are key prerequisites for successful operation with high intensity beams. We perform realistic simulations to identify quadrupole errors in LHC and explore possible ways of correction to minimize beta-beat below the 20% level

    Feed, Microbiota, and Gut Immunity: Using the Zebrafish Model to Understand Fish Health

    Get PDF
    Aquafeed companies aim to provide solutions to the various challenges related to nutrition and health in aquaculture. Solutions to promote feed efficiency and growth, as well as improving the fish health or protect the fish gut from inflammation may include dietary additives such as prebiotics and probiotics. The general assumption is that feed additives can alter the fish microbiota which, in turn, interacts with the host immune system. However, the exact mechanisms by which feed influences host-microbe-immune interactions in fish still remain largely unexplored. Zebrafish rapidly have become a well-recognized animal model to study host-microbe-immune interactions because of the diverse set of research tools available for these small cyprinids. Genome editing technologies can create specific gene-deficient zebrafish that may contribute to our understanding of immune functions. Zebrafish larvae are optically transparent, which allows for in vivo imaging of specific (immune) cell populations in whole transgenic organisms. Germ-free individuals can be reared to study host-microbe interactions. Altogether, these unique zebrafish features may help shed light on the mechanisms by which feed influences host-microbe-immune interactions and ultimately fish health. In this review, we first describe the anatomy and function of the zebrafish gut: the main surface where feed influences host-microbe-immune interactions. Then, we further describe what is currently known about the molecular pathways that underlie this interaction in the zebrafish gut. Finally, we summarize and critically review most of the recent research on prebiotics and probiotics in relation to alterations of zebrafish microbiota and immune responses. We discuss the advantages and disadvantages of the zebrafish as an animal model for other fish species to study feed effects on host-microbe-immune interactions.</p

    NCI First International Workshop on the Biology, Prevention, and Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation: Report from the Committee on Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation

    Get PDF
    Relapse is a major cause of treatment failure after allogeneic hematopoietic stem cell transplantation (alloHSCT). Treatment options for relapse have been inadequate, and the majority of patients ultimately die of their disease. There is no standard approach to treating relapse after alloHSCT. Withdrawal of immune suppression and donor lymphocyte infusions are commonly used for all diseases; although these interventions are remarkably effective for relapsed chronic myelogenous leukemia, they have limited efficacy in other hematologic malignancies. Conventional and novel chemotherapy, monoclonal antibody therapy, targeted therapies, and second transplants have been utilized in a variety of relapsed diseases, but reports on these therapies are generally anecdotal and retrospective. As such, there is an immediate need for well-designed, disease-specific trials for treatment of relapse after alloHSCT. This report summarizes current treatment options under investigation for relapse after alloHSCT in a disease-specific manner. In addition, recommendations are provided for specific areas of research necessary in the treatment of relapse after alloHSCT

    Comparison between antithymocyte globulin and alemtuzumab and the possible impact of KIR-ligand mismatch after dose-reduced conditioning and unrelated stem cell transplantation in patients with multiple myeloma

    Get PDF
    We compared antithymocyte globulin (ATG) with alemtuzumab in 73 patients with multiple myeloma, who underwent reduced conditioning with melphalan/fludarabine, followed by allogeneic stem cell transplantation from human leucocyte antigen-matched or -mismatched unrelated donors. The ATG group had more prior high-dose chemotherapies (P &lt; 0.001), while bone marrow was used more as the stem cell source in the alemtuzumab group (P &lt; 0.001). Alemtuzumab resulted in faster engraftment of leucocytes (P = 0.03) and platelets (P = 0.02) and in a lower incidence of acute graft versus host disease (GvHD) grades II-IV (24% vs. 47%, P = 0.06). More cytomegalovirus (CMV) seropositive patients in the alemtuzumab group experienced CMV reactivation (100% vs. 47%, P = 0.001). The cumulative incidence of treatment-related mortality at 2 years was 26% [95% confidence interval (CI) = 12-37%] for ATG vs. 28% (95% CI = 15-55%) for alemtuzumab, P = 0.7. There was no significant difference in the estimated 2-year overall and progression-free survival between ATG and alemtuzumab: 54% (95% CI: 39-75%) vs. 45% (95% CI: 28-73%) and 30% (95% CI: 16-55%) vs. 36% (95% CI: 20-62%) respectively. In multivariate analysis, treatment with alemtuzumab had a higher risk for relapse (hazard ratio: 2.37; P = 0.05) while killer immunoglobulin-like receptor (KIR)-ligand mismatch was protective for relapse (P &lt; 0.0001). We conclude that alemtuzumab produced less acute GvHD, but higher probability of relapse. The data implicated a major role of KIR-ligand mismatched transplantation in multiple myeloma

    Dual targeting of CD19 and CD22 with Bicistronic CAR-T cells in Patients with Relapsed/Refractory Large B Cell Lymphoma

    Get PDF
    Relapse following CD19-directed chimeric antigen receptor T-cells (CAR-T) for relapsed/refractory large B-cell lymphoma (r/r LBCL) is commonly ascribed to antigen loss or CAR-T exhaustion. Multi-antigen targeting and PD-1 blockade are rational approaches to prevent relapse. Here, we test CD19/22 dual-targeting CAR-T (AUTO3) plus pembrolizumab in r/r LBCL as inpatient or outpatient therapy (NCT03289455, https://clinicaltrials.gov/ct2/show/NCT03289455). Endpoints include toxicity (primary) and response rates (secondary). AUTO3 was manufactured for 62 patients using autologous leukapheresis, modified with a bicistronic transgene. 52 patients received AUTO3 (7/52,50x106; 45/52,150-450x106) and 48/52 received pembrolizumab. Median age was 59 years (range,27-83) and 46/52 had stage III/IV disease. Median follow-up was 21.6 months (range,15.1-51.3) at last data cut (Feb 28, 2022). AUTO3 was safe: grade 1-2 and grade 3 CRS affected 18/52 (34.6%) and 1/52 (1.9%) patients, neurotoxicity arose in 4 patients (2/4, grade 3-4), HLH affected 2 patients, and no Pembrolizumab-associated autoimmune sequalae were observed. On this basis, outpatient administration was tested in 20 patients, saving a median of 14 hospital days/patient. AUTO3 was effective: overall response rates were 66% (48.9%, CR; 17%, PR). For patients with CR, median DOR was not reached, with 54.4% (CI: 32.8, 71.7) projected to remain progression-free beyond 12 months after onset of remission. DOR for all responding patients was 8.3 months (95% CI: 3.0, NE) with 42.6% projected to remain progression-free beyond 12 months after onset of remission. Overall, AUTO3 +/- pembrolizumab for r/r LBCL was safe, lending itself to outpatient administration, and delivered durable remissions in 54.4% of complete responders, associated with robust CAR-T expansion. Neither dual-targeting CAR-T nor pembrolizumab prevented relapse in a significant proportion of patients, and future developments include next-generation-AUTO3, engineered for superior expansion/persistence in vivo, and selection of CAR binders active at low antigen densities

    Outcomes after alemtuzumab-containing reduced-intensity allogeneic transplantation regimen for relapsed and refractory non-Hodgkin lymphoma

    Get PDF
    18-60 months) , the actuarial overall survival (OS) rates at 3 years were 34% for HG-NHL, 60% for MCL, and 73% for LG-NHL (P &lt; .001). The 100-day and 3-year transplant-related mortality (TRM) rates for patients with LG-NHL were 2% and 11%, respectively, and were better (P ‫؍‬ .01) than they were for patients with HG-NHL (27% and 38%, respectively). The actuarial current progression-free survival (PFS) rate at 3 years, including the rate for patients who achieved remission after donor lymphocyte infusion (DLI) for progression, was 65% for LG-NHL, 50% for MCL, and 34% for HG-NHL (P ‫؍‬ .002)

    Targeting the T cell receptor β-chain constant region for immunotherapy of T cell malignancies

    Get PDF
    Mature T cell cancers are typically aggressive, treatment resistant and associated with poor prognosis. Clinical application of immunotherapeutic approaches has been limited by a lack of target antigens that discriminate malignant from healthy (normal) T cells. Unlike B cell depletion, pan–T cell aplasia is prohibitively toxic. We report a new targeting strategy based on the mutually exclusive expression of T cell receptor β-chain constant domains 1 and 2 (TRBC1 and TRBC2). We identify an antibody with unique TRBC1 specificity and use it to demonstrate that normal and virus-specific T cell populations contain both TRBC1+ and TRBC2+ compartments, whereas malignancies are restricted to only one. As proof of concept for anti-TRBC immunotherapy, we developed anti-TRBC1 chimeric antigen receptor (CAR) T cells, which recognized and killed normal and malignant TRBC1+, but not TRBC2+, T cells in vitro and in a disseminated mouse model of leukemia. Unlike nonselective approaches targeting the entire T cell population, TRBC-targeted immunotherapy could eradicate a T cell malignancy while preserving sufficient normal T cells to maintain cellular immunity

    Transduction of SIV-Specific TCR Genes into Rhesus Macaque CD8+ T Cells Conveys the Ability to Suppress SIV Replication

    Get PDF
    The SIV/rhesus macaque model for HIV/AIDS is a powerful system for examining the contribution of T cells in the control of AIDS viruses. To better our understanding of CD8(+) T-cell control of SIV replication in CD4(+) T cells, we asked whether TCRs isolated from rhesus macaque CD8(+) T-cell clones that exhibited varying abilities to suppress SIV replication could convey their suppressive properties to CD8(+) T cells obtained from an uninfected/unvaccinated animal.We transferred SIV-specific TCR genes isolated from rhesus macaque CD8(+) T-cell clones with varying abilities to suppress SIV replication in vitro into CD8(+) T cells obtained from an uninfected animal by retroviral transduction. After sorting and expansion, transduced CD8(+) T-cell lines were obtained that specifically bound their cognate SIV tetramer. These cell lines displayed appropriate effector function and specificity, expressing intracellular IFNγ upon peptide stimulation. Importantly, the SIV suppression properties of the transduced cell lines mirrored those of the original TCR donor clones: cell lines expressing TCRs transferred from highly suppressive clones effectively reduced wild-type SIV replication, while expression of a non-suppressing TCR failed to reduce the spread of virus. However, all TCRs were able to suppress the replication of an SIV mutant that did not downregulate MHC-I, recapitulating the properties of their donor clones.Our results show that antigen-specific SIV suppression can be transferred between allogenic T cells simply by TCR gene transfer. This advance provides a platform for examining the contributions of TCRs versus the intrinsic effector characteristics of T-cell clones in virus suppression. Additionally, this approach can be applied to develop non-human primate models to evaluate adoptive T-cell transfer therapy for AIDS and other diseases
    corecore