Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
J Physiol ; 602(4): 737-757, 2024 Feb.
Article En | MEDLINE | ID: mdl-38345534

Paraoxonase 3 (PON3) is expressed in the aldosterone-sensitive distal nephron, where filtered Na+ is reabsorbed mainly via the epithelial Na+ channel (ENaC) and Na+ -coupled co-transporters. We previously showed that PON3 negatively regulates ENaC through a chaperone mechanism. The present study aimed to determine the physiological role of PON3 in renal Na+ and K+ homeostasis. Pon3 knockout (KO) mice had higher amiloride-induced natriuresis and lower plasma [K+ ] at baseline. Single channel recordings in split-open tubules showed that the number of active channels per patch was significantly higher in KO mice, resulting in a higher channel activity in the absence of PON3. Although whole kidney abundance of ENaC subunits was not altered in Pon3 KOs, ENaC gamma subunit was more apically distributed within the connecting tubules and cortical collecting ducts of Pon3 KO kidneys. Additionally, small interfering RNA-mediated knockdown of PON3 in cultured mouse cortical collecting duct cells led to an increased surface abundance of ENaC gamma subunit. As a result of lower plasma [K+ ], sodium chloride co-transporter phosphorylation was enhanced in the KO kidneys, a phenotype that was corrected by a high K+ diet. Finally, PON3 expression was upregulated in mouse kidneys under dietary K+ restriction, potentially providing a mechanism to dampen ENaC activity and associated K+ secretion. Taken together, our results show that PON3 has a role in renal Na+ and K+ homeostasis through regulating ENaC functional expression in the distal nephron. KEY POINTS: Paraoxonase 3 (PON3) is expressed in the distal nephron of mouse kidneys and functions as a molecular chaperone to reduce epithelial Na+ channel (ENaC) expression and activity in heterologous expression systems. We examined the physiological role of PON3 in renal Na+ and K+ handling using a Pon3 knockout (KO) mouse model. At baseline, Pon3 KO mice had lower blood [K+ ], more functional ENaC in connecting tubules/cortical collecting ducts, higher amiloride-induced natriuresis, and enhanced sodium chloride co-transporter (NCC) phosphorylation. Upon challenge with a high K+ diet, Pon3 KO mice had normalized blood [K+ ] and -NCC phosphorylation but lower circulating aldosterone levels compared to their littermate controls. Kidney PON3 abundance was altered in mice under dietary K+ loading or K+ restriction, providing a potential mechanism for regulating ENaC functional expression and renal Na+ and K+ homeostasis in the distal nephron.


Amiloride , Symporters , Mice , Animals , Amiloride/pharmacology , Aryldialkylphosphatase/metabolism , Epithelial Sodium Channels/metabolism , Aldosterone/metabolism , Sodium Chloride/metabolism , Sodium/metabolism , Nephrons/metabolism
2.
ACS Synth Biol ; 12(12): 3608-3622, 2023 Dec 15.
Article En | MEDLINE | ID: mdl-38010428

Small, single-domain protein scaffolds are compelling sources of molecular binding ligands with the potential for efficient physiological transport, modularity, and manufacturing. Yet, mini-proteins require a balance between biophysical robustness and diversity to enable new functions. We tested the developability and evolvability of millions of variants of 43 designed libraries of synthetic 40-amino acid ßαßß proteins with diversified sheet, loop, or helix paratopes. We discovered a scaffold library that yielded hundreds of binders to seven targets while exhibiting high stability and soluble expression. Binder discovery yielded 6-122 nM affinities without affinity maturation and Tms averaging ≥78 °C. Broader ßαßß libraries exhibited varied developability and evolvability. Sheet paratopes were the most consistently developable, and framework 1 was the most evolvable. Paratope evolvability was dependent on target, though several libraries were evolvable across many targets while exhibiting high stability and soluble expression. Select ßαßß proteins are strong starting points for engineering performant binders.


Peptide Library , Proteins , Ligands , Proteins/genetics , Proteins/chemistry
3.
Commun Biol ; 6(1): 962, 2023 09 21.
Article En | MEDLINE | ID: mdl-37735514

BRAF mutations occur early in serrated colorectal cancers, but their long-term influence on tissue homeostasis is poorly characterized. We investigated the impact of short-term (3 days) and long-term (6 months) expression of BrafV600E in the intestinal tissue of an inducible mouse model. We show that BrafV600E perturbs the homeostasis of intestinal epithelial cells, with impaired differentiation of enterocytes emerging after prolonged expression of the oncogene. Moreover, BrafV600E leads to a persistent transcriptional reprogramming with enrichment of numerous gene signatures indicative of proliferation and tumorigenesis, and signatures suggestive of metabolic rewiring. We focused on the top-ranking cholesterol biosynthesis signature and confirmed its increased expression in human serrated lesions. Functionally, the cholesterol lowering drug atorvastatin prevents the establishment of intestinal crypt hyperplasia in BrafV600E-mutant mice. Overall, our work unveils the long-term impact of BrafV600E expression in intestinal tissue and suggests that colorectal cancers with mutations in BRAF might be prevented by statins.


Colorectal Neoplasms , Proto-Oncogene Proteins B-raf , Animals , Humans , Mice , Cholesterol , Colorectal Neoplasms/genetics , Lipid Metabolism , Proto-Oncogene Proteins B-raf/genetics , Transcriptional Activation
4.
bioRxiv ; 2023 Feb 11.
Article En | MEDLINE | ID: mdl-36798186

Cells rely on antioxidants to survive. The most abundant antioxidant is glutathione (GSH). The synthesis of GSH is non-redundantly controlled by the glutamate-cysteine ligase catalytic subunit (GCLC). GSH imbalance is implicated in many diseases, but the requirement for GSH in adult tissues is unclear. To interrogate this, we developed a series of in vivo models to induce Gclc deletion in adult animals. We find that GSH is essential to lipid abundance in vivo. GSH levels are reported to be highest in liver tissue, which is also a hub for lipid production. While the loss of GSH did not cause liver failure, it decreased lipogenic enzyme expression, circulating triglyceride levels, and fat stores. Mechanistically, we found that GSH promotes lipid abundance by repressing NRF2, a transcription factor induced by oxidative stress. These studies identify GSH as a fulcrum in the liver's balance of redox buffering and triglyceride production.

5.
Physiol Rep ; 11(1): e15554, 2023 01.
Article En | MEDLINE | ID: mdl-36636010

The epithelial Na+ channel (ENaC) is traditionally composed of three subunits, although non-canonical expression has been found in various tissues including the vasculature, brain, lung, and dendritic cells of the immune system. Studies of ENaC structure and function have largely relied on heterologous expression systems, often with epitope-tagged channel subunits. Relevant in vivo physiological studies have used ENaC inhibitors, mice with global or tissue specific knockout of subunits, and anti-ENaC subunit antibodies generated by investigators or by commercial sources. Availability of well-characterized, specific antibodies is imperative as we move forward in understanding the role of ENaC in non-epithelial tissues where expression, subunit organization, and electrophysiological characteristics may differ from epithelial tissues. We report that a commonly used commercial anti-α subunit antibody recognizes an intense non-specific band on mouse whole kidney and lung immunoblots, which migrates adjacent to a less intense, aldosterone-induced full length α-subunit. This antibody localizes to the basolateral membrane of aquaporin 2 negative cells in kidney medulla. We validated antibodies against the ß- and γ-subunits from the same commercial source. Our work illustrates the importance of validation studies when using popular, commercially available anti-ENaC antibodies.


Epithelial Sodium Channels , Kidney , Mice , Animals , Epithelial Sodium Channels/metabolism , Kidney/metabolism , Sodium/metabolism , Epithelium/metabolism , Kidney Medulla/metabolism
6.
Ann Rheum Dis ; 82(1): 154-160, 2023 01.
Article En | MEDLINE | ID: mdl-35961761

BACKGROUND: Treatment options in patients with enthesitis-related arthritis (ERA) and juvenile psoriatic arthritis (JPsA) are currently limited. This trial aimed to demonstrate the efficacy and safety of secukinumab in patients with active ERA and JPsA with inadequate response to conventional therapy. METHODS: In this randomised, double-blind, placebo-controlled, treatment-withdrawal, phase 3 trial, biologic-naïve patients (aged 2 to <18 years) with active disease were treated with open-label subcutaneous secukinumab (75/150 mg in patients <50/≥50 kg) in treatment period (TP) 1 up to week 12, and juvenile idiopathic arthritis (JIA) American College of Rheumatology 30 responders at week 12 were randomised 1:1 to secukinumab or placebo up to 100 weeks. Patients who flared in TP2 immediately entered open-label secukinumab TP3 that lasted up to week 104. Primary endpoint was time to disease flare in TP2. RESULTS: A total of 86 patients (median age, 14 years) entered open-label secukinumab in TP1. In TP2, responders (ERA, 44/52; JPsA, 31/34) received secukinumab or placebo. The study met its primary end point and demonstrated a statistically significant longer time to disease flare in TP2 for ERA and JPsA with secukinumab versus placebo (27% vs 55%, HR, 0.28; 95% CI 0.13 to 0.63; p<0.001). Exposure-adjusted incidence rates (per 100 patient-years (PY), 95% CI) for total patients were 290.7/100 PY (230.2 to 362.3) for adverse events and 8.2/100 PY (4.1 to 14.6) for serious adverse events in the overall JIA population. CONCLUSIONS: Secukinumab demonstrated significantly longer time to disease flare than placebo in children with ERA and JPsA with a consistent safety profile with the adult indications of psoriatic arthritis and axial spondyloarthritis. TRIAL REGISTRATION NUMBER: NCT03031782.


Antirheumatic Agents , Arthritis, Juvenile , Arthritis, Psoriatic , Adult , Child , Humans , Adolescent , Arthritis, Juvenile/drug therapy , Antirheumatic Agents/adverse effects , Symptom Flare Up , Treatment Outcome , Arthritis, Psoriatic/drug therapy , Arthritis, Psoriatic/chemically induced , Double-Blind Method
7.
Angew Chem Int Ed Engl ; 61(19): e202116175, 2022 May 02.
Article En | MEDLINE | ID: mdl-35285999

Here we calculate T1 →S0 transition energies in nine phosphorescent iridium complexes using the iterative qubit coupled cluster (iQCC) method to determine if quantum simulations have any advantages over classical methods. These simulations would require a gate-based quantum computer with at least 72 fully-connected logical qubits. Since such devices do not yet exist, we demonstrate the iQCC method using a purpose-built quantum simulator on classical hardware. The results are compared to a selection of common DFT functionals, ab initio methods, and empirical data. iQCC is found to match the accuracy of the best DFT functionals, but with a better correlation coefficient, demonstrating that it is better at predicting the structure-property relationship. Results indicate that the iQCC method has the required accuracy to design organometallic complexes when deployed on emerging quantum hardware and sets an industrially relevant target for demonstrating quantum advantage.

8.
Rheumatol Ther ; 9(2): 753-762, 2022 Apr.
Article En | MEDLINE | ID: mdl-35044647

INTRODUCTION: Systemic juvenile idiopathic arthritis (sJIA) and adult-onset Still's disease (AOSD) represent pediatric and adult variants of the Still's disease continuum. To determine whether clinical outcomes between patients with sJIA and AOSD were similar, Bayesian and population model-based analyses were conducted on endpoints from studies of canakinumab in both patient populations. The objective was to further support the efficacy of canakinumab in patients with AOSD. METHODS: A Bayesian analysis of endpoints from a study of canakinumab in AOSD was conducted borrowing information from five pooled sJIA studies using a robust meta-analytic predictive (MAP) approach. Similarity of clinical outcomes across populations was fulfilled if the AOSD study posterior median fell within the 95% predicted credible interval for the outcome of interest, based on the pooled sJIA data. Population model-based analyses (pharmacokinetic [PK] and PK/pharmacodynamic [PKPD]) were conducted to compare the pharmacokinetics and exposure-response relationships between populations. RESULTS: The AOSD study posterior medians for adapted American College of Rheumatology (ACR)30 response, continuous adapted ACR response, number of active joints, C-reactive protein, and absence of fever were within the 95% credible interval for the prediction of the MAP analysis from the pooled sJIA data, supporting the similarity in outcomes between patient populations. PK analysis demonstrated comparable exposure across sJIA age groups and patients with AOSD. PKPD relationships were consistent across patient populations. Analyses indicated that no therapeutic benefit can be expected from a dose increase in patients with AOSD. CONCLUSION: The analyses presented support the similarity of clinical outcomes following treatment with canakinumab in patients with sJIA and AOSD.

9.
JAMA ; 326(3): 230-239, 2021 07 20.
Article En | MEDLINE | ID: mdl-34283183

Importance: Effective treatments for patients with severe COVID-19 are needed. Objective: To evaluate the efficacy of canakinumab, an anti-interleukin-1ß antibody, in patients hospitalized with severe COVID-19. Design, Setting, and Participants: This randomized, double-blind, placebo-controlled phase 3 trial was conducted at 39 hospitals in Europe and the United States. A total of 454 hospitalized patients with COVID-19 pneumonia, hypoxia (not requiring invasive mechanical ventilation [IMV]), and systemic hyperinflammation defined by increased blood concentrations of C-reactive protein or ferritin were enrolled between April 30 and August 17, 2020, with the last assessment of the primary end point on September 22, 2020. Intervention: Patients were randomly assigned 1:1 to receive a single intravenous infusion of canakinumab (450 mg for body weight of 40-<60 kg, 600 mg for 60-80 kg, and 750 mg for >80 kg; n = 227) or placebo (n = 227). Main Outcomes and Measures: The primary outcome was survival without IMV from day 3 to day 29. Secondary outcomes were COVID-19-related mortality, measurements of biomarkers of systemic hyperinflammation, and safety evaluations. Results: Among 454 patients who were randomized (median age, 59 years; 187 women [41.2%]), 417 (91.9%) completed day 29 of the trial. Between days 3 and 29, 198 of 223 patients (88.8%) survived without requiring IMV in the canakinumab group and 191 of 223 (85.7%) in the placebo group, with a rate difference of 3.1% (95% CI, -3.1% to 9.3%) and an odds ratio of 1.39 (95% CI, 0.76 to 2.54; P = .29). COVID-19-related mortality occurred in 11 of 223 patients (4.9%) in the canakinumab group vs 16 of 222 (7.2%) in the placebo group, with a rate difference of -2.3% (95% CI, -6.7% to 2.2%) and an odds ratio of 0.67 (95% CI, 0.30 to 1.50). Serious adverse events were observed in 36 of 225 patients (16%) treated with canakinumab vs 46 of 223 (20.6%) who received placebo. Conclusions and Relevance: Among patients hospitalized with severe COVID-19, treatment with canakinumab, compared with placebo, did not significantly increase the likelihood of survival without IMV at day 29. Trial Registration: ClinicalTrials.gov Identifier: NCT04362813.


Antibodies, Monoclonal, Humanized/therapeutic use , COVID-19 Drug Treatment , Interleukin-1beta/antagonists & inhibitors , Respiration, Artificial/statistics & numerical data , Aged , Antibodies, Monoclonal, Humanized/adverse effects , C-Reactive Protein/analysis , COVID-19/mortality , COVID-19/therapy , Combined Modality Therapy , Double-Blind Method , Female , Ferritins/blood , Fibrin Fibrinogen Degradation Products/analysis , Hospitalization , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Survival Rate , Treatment Outcome
10.
Cancer Immunol Immunother ; 70(12): 3525-3540, 2021 Dec.
Article En | MEDLINE | ID: mdl-33903974

Immune checkpoint inhibitors (ICIs) have emerged as promising therapies for the treatment of cancer. However, existing ICIs, namely PD-(L)1 and CTLA-4 inhibitors, generate durable responses only in a subset of patients. TIGIT is a co-inhibitory receptor and member of the DNAM-1 family of immune modulating proteins. We evaluated the prevalence of TIGIT and its cognate ligand, PVR (CD155), in human cancers by assessing their expression in a large set of solid tumors. TIGIT is expressed on CD4+ and CD8+ TILs and is upregulated in tumors compared to normal tissues. PVR is expressed on tumor cells and tumor-associated macrophages from multiple solid tumors. We explored the therapeutic potential of targeting TIGIT by generating COM902, a fully human anti-TIGIT hinge-stabilized IgG4 monoclonal antibody that binds specifically to human, cynomolgus monkey, and mouse TIGIT, and disrupts the binding of TIGIT with PVR. COM902, either alone or in combination with a PVRIG (COM701) or PD-1 inhibitor, enhances antigen-specific human T cell responses in-vitro. In-vivo, a mouse chimeric version of COM902 in combination with an anti-PVRIG or anti-PD-L1 antibody inhibited tumor growth and increased survival in two syngeneic mouse tumor models. In summary, COM902 enhances anti-tumor immune responses and is a promising candidate for the treatment of advanced malignancies.


Antibodies, Monoclonal/immunology , B7-H1 Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Receptors, Cell Surface/immunology , Receptors, Immunologic/immunology , Signal Transduction/immunology , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Female , Humans , Immunoglobulin G/immunology , Immunotherapy/methods , Jurkat Cells , Macaca fascicularis , Mice , Mice, Inbred BALB C
11.
Cancer Discov ; 11(5): 1040-1051, 2021 05.
Article En | MEDLINE | ID: mdl-33687987

Therapeutic antibodies targeting the CTLA4/PD-1 pathways have revolutionized cancer immunotherapy by eliciting durable remission in patients with cancer. However, relapse following early response, attributable to primary and adaptive resistance, is frequently observed. Additional immunomodulatory pathways are being studied in patients with primary or acquired resistance to CTLA4 or PD-1 blockade. The DNAM1 axis is a potent coregulator of innate and adaptive immunity whose other components include the immunoglobulin receptors TIGIT, PVRIG, and CD96, and their nectin and nectin-like ligands. We review the basic biology and therapeutic relevance of this family, which has begun to show promise in cancer clinical trials. SIGNIFICANCE: Recent studies have outlined the immuno-oncologic ascendancy of coinhibitory receptors in the DNAM1 axis such as TIGIT and PVRIG and, to a lesser extent, CD96. Biological elucidation backed by ongoing clinical trials of single-agent therapy directed against TIGIT or PVRIG is beginning to provide the rationale for testing combination regimens of DNAM1 axis blockers in conjunction with anti-PD-1/PD-L1 agents.


Antibodies, Monoclonal/therapeutic use , Antigens, Differentiation, T-Lymphocyte/metabolism , Neoplasms/drug therapy , Receptors, Immunologic/metabolism , Humans , Immunotherapy
12.
Haematologica ; 106(12): 3115-3124, 2021 12 01.
Article En | MEDLINE | ID: mdl-33147937

This study explored the novel immune checkpoint poliovirus receptor-related immunoglobulin domain-containing (PVRIG) in acute myeloid leukemia (AML). We showed that AML patient blasts consistently expressed the PVRIG ligand (poliovirus receptor-related 2, PVRL2). Furthermore, PVRIG blockade significantly enhanced NK cell killing of PVRL2+, poliovirus receptor (PVR)lo AML cell lines, and significantly increased NK cell activation and degranulation in the context of patient primary AML blasts. However, in AML patient bone marrow, NK cell PVRIG expression levels were not increased. To understand how PVRIG blockade might potentially be exploited therapeutically, we investigated the biology of PVRIG and revealed that NK cell activation resulted in reduced PVRIG expression on the cell surface. This occurred whether NK cells were activated by tumour cell recognition, cytokines (IL-2 and IL-12) or activating receptor stimulation (CD16 and NKp46). PVRIG was present at higher levels in the cytoplasm than on the cell surface, particularly on CD56bright NK cells, which further increased cytoplasmic PVRIG levels following IL-2 and IL-12 activation. PVRIG was continually transported to the cell surface via the endoplasmic reticulum (ER) and Golgi in both unstimulated and activated NK cells. Taken together, our findings suggest that anti- PVRIG blocking antibody functions by binding to surface-bound PVRIG, which undergoes rapid turnover in both unstimulated and activated NK cells. We conclude that the PVRIGPVRL2 immune checkpoint axis can feasibly be targeted with PVRIG blocking antibody for NK-mediated immunotherapy of PVRL2+ AML.


Immune Checkpoint Proteins , Killer Cells, Natural , Leukemia, Myeloid, Acute , Receptors, Cell Surface , Humans , Immunotherapy , Lymphocyte Activation , Receptors, Natural Killer Cell
13.
Ir J Med Sci ; 188(4): 1251-1259, 2019 Nov.
Article En | MEDLINE | ID: mdl-30924006

AIMS: Indacaterol/glycopyrronium (IND/GLY) 110/50 µg is a once-daily (o.d.) fixed-dose combination of long-acting ß2-agonist/long-acting muscarinic antagonist approved in over 90 countries, including Ireland, for the management of COPD. The present study was conducted to evaluate health status of COPD patients, initiated on IND/GLY 110/50 µg o.d., using the Clinical COPD Questionnaire (CCQ) tool in a real-world primary care setting in Ireland. METHODS: This was a real-world, prospective, open-label study. COPD patients aged > 40 years and with a smoking history of > 10 pack-years were included and switched to once-daily IND/GLY 110/50 µg. Enrolment of patients into the study occurred only after the decision had been made by the physician to prescribe IND/GLY 110/50 µg. Data were collected at baseline and Week 26. Health status was assessed using the validated CCQ. RESULTS: A total of 200 patients were included in study. The mean CCQ total score decreased from 2.36 at baseline to 1.44 at Week 26 (Δ, 0.92; P < 0.0005). Of the 156 patients who completed study, 113 (72.4%) achieved minimum clinically important difference in CCQ total score with IND/GLY 110/50 µg. CCQ domain scores also decreased during the study. Improvement in health status was observed across all GOLD groups and irrespective of prior COPD treatment. Adverse events were reported by 20% of patients with COPD exacerbation/infected COPD being the most common AE, reported by 11 patients. CONCLUSIONS: In real-life clinical practice in Ireland, IND/GLY 110/50 µg o.d. demonstrated statistically significant and clinically important improvement in health status in patients with COPD.


Adrenergic beta-2 Receptor Agonists/administration & dosage , Glycopyrrolate/administration & dosage , Indans/administration & dosage , Muscarinic Antagonists/administration & dosage , Pulmonary Disease, Chronic Obstructive/drug therapy , Quinolones/administration & dosage , Adult , Aged , Aged, 80 and over , Bronchodilator Agents/administration & dosage , Drug Combinations , Female , Health Status , Humans , Ireland , Male , Middle Aged , Prospective Studies , Severity of Illness Index , Surveys and Questionnaires , Treatment Outcome
14.
Cancer Immunol Res ; 7(2): 257-268, 2019 02.
Article En | MEDLINE | ID: mdl-30659054

Although checkpoint inhibitors that block CTLA-4 and PD-1 have improved cancer immunotherapies, targeting additional checkpoint receptors may be required to broaden patient response to immunotherapy. PVRIG is a coinhibitory receptor of the DNAM/TIGIT/CD96 nectin family that binds to PVRL2. We report that antagonism of PVRIG and TIGIT, but not CD96, increased CD8+ T-cell cytokine production and cytotoxic activity. The inhibitory effect of PVRL2 was mediated by PVRIG and not TIGIT, demonstrating that the PVRIG-PVRL2 pathway is a nonredundant signaling node. A combination of PVRIG blockade with TIGIT or PD-1 blockade further increased T-cell activation. In human tumors, PVRIG expression on T cells was increased relative to normal tissue and trended with TIGIT and PD-1 expression. Tumor cells coexpressing PVR and PVRL2 were observed in multiple tumor types, with highest coexpression in endometrial cancers. Tumor cells expressing either PVR or PVRL2 were also present in numbers that varied with the cancer type, with ovarian cancers having the highest percentage of PVR-PVRL2+ tumor cells and colorectal cancers having the highest percentage of PVR+PVRL2- cells. To demonstrate a role of PVRIG and TIGIT on tumor-derived T cells, we examined the effect of PVRIG and TIGIT blockade on human tumor-infiltrating lymphocytes. For some donors, blockade of PVRIG increased T-cell function, an effect enhanced by combination with TIGIT or PD-1 blockade. In summary, we demonstrate that PVRIG and PVRL2 are expressed in human cancers and the PVRIG-PVRL2 and TIGIT-PVR pathways are nonredundant inhibitory signaling pathways.See related article on p. 244.


CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Nectins/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Receptors, Cell Surface/metabolism , Animals , Gene Expression Regulation, Neoplastic , Humans , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Neoplasms/genetics , Neoplasms/pathology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Signal Transduction
15.
Eur J Immunol ; 46(9): 2111-20, 2016 09.
Article En | MEDLINE | ID: mdl-27485474

The adult human liver is enriched with natural killer (NK) cells, accounting for 30-50% of hepatic lymphocytes, which include tissue-resident hepatic NK-cell subpopulations, distinct from peripheral blood NK cells. In murine liver, a subset of liver-resident hepatic NK cells have altered expression of the two highly related T-box transcription factors, T-bet and eomesodermin (Eomes). Here, we investigate the heterogeneity of T-bet and Eomes expression in NK cells from healthy adult human liver with a view to identifying human liver-resident populations. Hepatic NK cells were isolated from donor liver perfusates and biopsies obtained during orthotopic liver transplantation (N = 28). Hepatic CD56(bright) NK cells were Eomes(hi) T-bet(lo) , a phenotype virtually absent from peripheral blood. These NK cells express the chemokine receptor CXCR6 (chemokine (C-X-C motif) receptor 6), a marker of tissue residency, which is absent from hepatic CD56(dim) and blood NK cells. Compared to blood populations, these hepatic CD56(bright) NK cells have increased expression of activatory receptors (NKp44, NKp46, and NKG2D). They show reduced ability to produce IFN-γ but enhanced degranulation in response to challenge with target cells. This functionally distinct population of hepatic NK cells constitutes 20-30% of the total hepatic lymphocyte repertoire and represents a tissue-resident immune cell population adapted to the tolerogenic liver microenvironment.


CD56 Antigen/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Liver/immunology , T-Box Domain Proteins/metabolism , Adult , Biomarkers , Cell Degranulation/immunology , Cytotoxicity, Immunologic , Gene Expression , Humans , Immune Tolerance , Immunophenotyping , Interferon-gamma/biosynthesis , Liver/cytology , Liver/metabolism , Liver Transplantation , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Phenotype , T-Box Domain Proteins/genetics
16.
Stem Cells Dev ; 25(7): 530-41, 2016 Apr 01.
Article En | MEDLINE | ID: mdl-26879149

Poor myeloid engraftment remains a barrier to experimental use of humanized mice. Focusing primarily on peripheral blood cells, we compared the engraftment profile of NOD-scid-IL2Rγc(-/-) (NSG) mice with that of NSG mice transgenic for human membrane stem cell factor (hu-mSCF mice), NSG mice transgenic for human interleukin (IL)-3, granulocyte-macrophage-colony stimulating factor (GM-CSF), and stem cell factor (SGM3 mice). hu-mSCF and SGM3 mice showed enhanced engraftment of human leukocytes compared to NSG mice, and this was reflected in the number of human neutrophils and monocytes present in these strains. Importantly, discrete classical, intermediate, and nonclassical monocyte populations were identifiable in the blood of NSG and hu-mSCF mice, while the nonclassical population was absent in the blood of SGM3 mice. Granulocyte-colony stimulating factor (GCSF) treatment increased the number of blood monocytes in NSG and hu-mSCF mice, and neutrophils in NSG and SGM3 mice; however, this effect appeared to be at least partially dependent on the stem cell donor used to engraft the mice. Furthermore, GCSF treatment resulted in a preferential expansion of nonclassical monocytes in both NSG and hu-mSCF mice. Human tubulointerstitial CD11c(+) cells were present in the kidneys of hu-mSCF mice, while monocytes and neutrophils were identified in the liver of all strains. Bone marrow-derived macrophages prepared from NSG mice were most effective at phagocytosing polystyrene beads. In conclusion, hu-mSCF mice provide the best environment for the generation of human myeloid cells, with GCSF treatment further enhancing peripheral blood human monocyte cell numbers in this strain.


Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Myeloid Cells/transplantation , Transgenes , Animals , Cells, Cultured , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-3/genetics , Interleukin-3/metabolism , Leukocytes/cytology , Mice , Mice, Inbred NOD , Myeloid Cells/cytology , Myeloid Cells/drug effects , Neutrophils/cytology , Stem Cell Factor/genetics , Stem Cell Factor/metabolism
17.
J Med Microbiol ; 59(Pt 8): 898-903, 2010 Aug.
Article En | MEDLINE | ID: mdl-20466838

Campylobacter jejuni is a major causative agent of diarrhoeal disease worldwide in the human population. In contrast, heavy colonization of poultry typically does not lead to disease and colonized chickens are a major source of Campylobacter infections in humans. Previously, we have shown that chicken (but not human) intestinal mucus inhibits C. jejuni internalization. In this study, we test the hypothesis that chicken mucin, the main component of mucus, is responsible for this inhibition of C. jejuni virulence. Purified chicken intestinal mucin attenuated C. jejuni binding and internalization into HCT-8 cells depending on the site of origin of the mucin (large intestine>small intestine>caecum). C. jejuni invasion of HCT-8 cells was preferentially inhibited compared to bacterial binding to cells. Exposure of the mucin to sodium metaperiodate recovered bacterial invasion levels, suggesting a glycan-mediated effect. However, fucosidase or sialidase pre-treatment of mucin failed to abrogate the inhibition of C. jejuni pathogenicity. In conclusion, differences in the composition of chicken and human intestinal mucin may contribute to the differential outcome of Campylobacter infection of these hosts.


Bacterial Adhesion , Campylobacter jejuni/pathogenicity , Mucins/immunology , Animals , Cell Line , Chickens , Epithelial Cells/microbiology , Humans , Mucins/isolation & purification , Neuraminidase/metabolism , Periodic Acid/metabolism , Polysaccharides/metabolism , Species Specificity , Virulence , alpha-L-Fucosidase/metabolism
18.
Nurs Stand ; 25(3): 33, 2010 Sep 22.
Article En | MEDLINE | ID: mdl-28076048

Further to Christian Duffin's interview with Cathy Warwick, general secretary of the Royal College of Midwives (career development September 8), I fear for the future of maternity services.

...