Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nanoscale ; 16(35): 16500-16509, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39157997

ABSTRACT

Osmium(II) complexes have attractive properties for potential theranostic agents given their anticancer activitiy, their redox potentials favourable for biological transformations within cancer cells and their luminescence in the near infrared (NIR) region. To achieve localised detection and delivery, gold nanoparticles (AuNP) provide an attractive scaffold to attach multiple luminescent agents on a single particle and provide a multimodal platform for detection and loaclaised delivery. We have developed 13 nm and 25 nm AuNP decorated with an osmium complex based on 1,10-phenantholine and surface active bipyridine ligands, OsPhenSS for live cell imaging and singlet oxygen generation, notated as OsPhenSS·AuNP13 and OsPhenSS·AuNP25. The AuNP designs not only allow versatile modalities for localisation of the probe but also water solubility for the osmium metal complex. The osmium decorated nanoparticles OsPhenSS·AuNP13 and OsPhenSS·AuNP25 display characteristic NIR luminescence from the osmium(II) 3MLCT at 785 nm in aqueous solutions with visible excitation. Upon incubation of the nanoparticles in lung cancer and breast carcinoma the luminescence signature of osmium and the gold reflectance reveal localisation in the cytoplasmic and perinuclear compartments. Excitation of the nanoparticles at 552 nm in the presence of a ROS indicator revealed a marked increase in the green fluorescence from the indicator, consistent with photo-induced ROS generation. The detection of singlet oxygen by time-resolved luminescence studies of the osmium and the nanoparticle probes further demonstrates the dual activity of the osmium-based nanoprobes for imaging and therapy. The introduction of gold nanoparticles for carrying osmium imaging probes allows a novel versatile strategy combining detection and localised therapies at the nanoscale.


Subject(s)
Gold , Metal Nanoparticles , Osmium , Singlet Oxygen , Gold/chemistry , Metal Nanoparticles/chemistry , Osmium/chemistry , Humans , Singlet Oxygen/metabolism , Singlet Oxygen/chemistry , Cell Line, Tumor , A549 Cells , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/metabolism
2.
Sci Rep ; 14(1): 12927, 2024 06 05.
Article in English | MEDLINE | ID: mdl-38839833

ABSTRACT

We aimed to characterize the cognitive profile of post-acute COVID-19 syndrome (PACS) patients with cognitive complaints, exploring the influence of biological and psychological factors. Participants with confirmed SARS-CoV-2 infection and cognitive complaints ≥ 8 weeks post-acute phase were included. A comprehensive neuropsychological battery (NPS) and health questionnaires were administered at inclusion and at 1, 3 and 6 months. Blood samples were collected at each visit, MRI scan at baseline and at 6 months, and, optionally, cerebrospinal fluid. Cognitive features were analyzed in relation to clinical, neuroimaging, and biochemical markers at inclusion and follow-up. Forty-nine participants, with a mean time from symptom onset of 10.4 months, showed attention-executive function (69%) and verbal memory (39%) impairment. Apathy (64%), moderate-severe anxiety (57%), and severe fatigue (35%) were prevalent. Visual memory (8%) correlated with total gray matter (GM) and subcortical GM volume. Neuronal damage and inflammation markers were within normal limits. Over time, cognitive test scores, depression, apathy, anxiety scores, MRI indexes, and fluid biomarkers remained stable, although fewer participants (50% vs. 75.5%; p = 0.012) exhibited abnormal cognitive evaluations at follow-up. Altered attention/executive and verbal memory, common in PACS, persisted in most subjects without association with structural abnormalities, elevated cytokines, or neuronal damage markers.


Subject(s)
Biomarkers , COVID-19 , Cognition , Magnetic Resonance Imaging , Neuroimaging , Neuropsychological Tests , Post-Acute COVID-19 Syndrome , Humans , Male , COVID-19/psychology , COVID-19/diagnostic imaging , COVID-19/complications , Female , Biomarkers/blood , Middle Aged , Neuroimaging/methods , Adult , Magnetic Resonance Imaging/methods , SARS-CoV-2/isolation & purification , Aged , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/blood , Anxiety
3.
Struct Dyn ; 10(6): 064101, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37941993

ABSTRACT

Accurate computations of experimental observables are essential for interpreting the high information content held within x-ray spectra. However, for complicated systems this can be difficult, a challenge compounded when dynamics becomes important owing to the large number of calculations required to capture the time-evolving observable. While machine learning architectures have been shown to represent a promising approach for rapidly predicting spectral lineshapes, achieving simultaneously accurate and sufficiently comprehensive training data is challenging. Herein, we introduce Δ-learning for x-ray spectroscopy. Instead of directly learning the structure-spectrum relationship, the Δ-model learns the structure dependent difference between a higher and lower level of theory. Consequently, once developed these models can be used to translate spectral shapes obtained from lower levels of theory to mimic those corresponding to higher levels of theory. Ultimately, this achieves accurate simulations with a much reduced computational burden as only the lower level of theory is computed, while the model can instantaneously transform this to a spectrum equivalent to a higher level of theory. Our present model, demonstrated herein, learns the difference between TDDFT(BLYP) and TDDFT(B3LYP) spectra. Its effectiveness is illustrated using simulations of Rh L3-edge spectra tracking the C-H activation of octane by a cyclopentadienyl rhodium carbonyl complex.

4.
J Neurosci ; 43(5): 863-877, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36549907

ABSTRACT

Lumateperone is indicated for the treatment of schizophrenia in adults and for depressive episodes associated with bipolar I or II disorder (bipolar depression) in adults, as monotherapy and as adjunctive therapy with lithium or valproate (Calabrese et al., 2021). It is currently under evaluation for the treatment of major depressive disorder (www.ClinicalTrials.gov). Lumateperone acts by selectively modulating serotonin, dopamine, and glutamate neurotransmission in the brain. However, other mechanisms could be involved in the actions of lumateperone, and because of the connection between the immune system and psychiatric health, we hypothesized that lumateperone might improve symptoms of depression, at least in part, by normalizing pathologic inflammation. Here, we show that in male and female C57BL/6 mice subjected to an acute immune challenge, lumateperone reduced aberrantly elevated levels of key proinflammatory cytokines (e.g., IL-1ß, IL-6, and TNF-α) in both brain and serum; lumateperone also reduced proinflammatory cytokines in male mice under acute behavioral stress. Further, we demonstrate that lumateperone altered key genes/pathways involved in maintaining tissue integrity and supporting blood-brain barrier function, such as claudin-5 and intercellular adhesion molecule 1. In addition, in acutely stressed male Sprague Dawley rats, lumateperone conferred anxiolytic- and antianhedonic-like properties while enhancing activity in the mammalian target of rapamycin complex 1 pathway in the PFC. Together, our preclinical findings indicate that lumateperone, in addition to its ability to modulate multiple neurotransmitter systems, could also act by reducing the impact of acute inflammatory challenges.SIGNIFICANCE STATEMENT Lumateperone is indicated in adults to treat schizophrenia and depressive episodes associated with bipolar I or II disorder, as monotherapy and adjunctive therapy with lithium or valproate. Because aberrant immune system activity is associated with increased depressive symptoms, the relationship between lumateperone and immune function was studied. Here, lumateperone reduced the levels of proinflammatory cytokines that were increased following an immune challenge or stress in mice. Additionally, lumateperone altered genes and pathways that maintain blood-brain barrier integrity, restored an index of blood-brain barrier function, reduced anxiety-like behavior in rodents, and enhanced mammalian target of rapamycin complex 1 pathway signaling in the PFC. These results highlight the anti-inflammatory actions of lumateperone and describe how lumateperone may reduce immune pathophysiology, which is associated with depressive symptoms.


Subject(s)
Depressive Disorder, Major , Rats , Male , Female , Mice , Animals , Depressive Disorder, Major/metabolism , Lithium , Valproic Acid , Rats, Sprague-Dawley , Mice, Inbred C57BL , Cytokines/metabolism , Inflammation/drug therapy , TOR Serine-Threonine Kinases , Mammals
5.
Diab Vasc Dis Res ; 19(4): 14791641221118626, 2022.
Article in English | MEDLINE | ID: mdl-35975361

ABSTRACT

Insulin receptors are internalized by endothelial cells to facilitate their physiological processes; however, the impact of hyperinsulinemia in brain endothelial cells is not known. Thus, the aim of this study was to elucidate the impact hyperinsulinemia plays on insulin receptor internalization through changes in phosphorylation, as well as the potential impact of protein tyrosine phosphatase 1B (PTP1B). Hippocampal microvessels were isolated from high-fat diet fed mice and assessed for insulin signaling activation, a process known to be involved with receptor internalization. Surface insulin receptors in brain microvascular endothelial cells were labelled to assess the role hyperinsulinemia plays on receptor internalization in response to stimulation, with and without the PTP1B antagonist, Claramine. Our results indicated that insulin receptor levels increased in tandem with decreased receptor signaling in the high-fat diet mouse microvessels. Insulin receptors of cells subjected to hyperinsulinemic treatment demonstrate splice variation towards decreased IR-A mRNA expression and demonstrate a higher membrane-localized proportion. This corresponded with decreased autophosphorylation at sites critical for receptor internalization and signaling. Claramine restored signaling and receptor internalization in cells treated with hyperinsulinemia. In conclusion, hyperinsulinemia impacts brain microvascular endothelial cell insulin receptor signaling and internalization, likely via alternative splicing and increased negative feedback from PTP1B.


Subject(s)
Hyperinsulinism , Receptor, Insulin , Animals , Brain , Endothelial Cells/metabolism , Insulin/metabolism , Mice , Phosphorylation , Receptor, Insulin/genetics , Receptor, Insulin/metabolism
6.
Phys Chem Chem Phys ; 24(16): 9156-9167, 2022 Apr 20.
Article in English | MEDLINE | ID: mdl-35393987

ABSTRACT

X-ray absorption spectroscopy at the L2/3 edge can be used to obtain detailed information about the local electronic and geometric structure of transition metal complexes. By virtue of the dipole selection rules, the transition metal L2/3 edge usually exhibits two distinct spectral regions: (i) the "white line", which is dominated by bound electronic transitions from metal-centred 2p orbitals into unoccupied orbitals with d character; the intensity and shape of this band consequently reflects the d density of states (d-DOS), which is strongly modulated by mixing with ligand orbitals involved in chemical bonding, and (ii) the post-edge, where oscillations encode the local geometric structure around the X-ray absorption site. In this Article, we extend our recently-developed XANESNET deep neural network (DNN) beyond the K-edge to predict X-ray absorption spectra at the Pt L2/3 edge. We demonstrate that XANESNET is able to predict Pt L2/3 -edge X-ray absorption spectra, including both the parts containing electronic and geometric structural information. The performance of our DNN in practical situations is demonstrated by application to two Pt complexes, and by simulating the transient spectrum of a photoexcited dimeric Pt complex. Our discussion includes an analysis of the feature importance in our DNN which demonstrates the role of key features and assists with interpreting the performance of the network.


Subject(s)
Coordination Complexes , Transition Elements , Coordination Complexes/chemistry , Neural Networks, Computer , X-Ray Absorption Spectroscopy , X-Rays
7.
ANZ J Surg ; 92(5): 1190-1195, 2022 05.
Article in English | MEDLINE | ID: mdl-35180330

ABSTRACT

BACKGROUND: Soft tissue lower limb reconstruction often requires free tissue transfer. We investigated whether the target vessels used for micro-vascular anastomosis in the lower limb influences microsurgical outcomes. METHODS: Data from Plastic Surgery Departments of a major tertiary hospital in the United Kingdom (Leeds General Infirmary, LGI) and Australia (Princess Alexandra Hospital, PAH) were retrospectively analysed. Patients who underwent lower limb free flap reconstruction using the posterior (PTA) or anterior tibial artery (ATA) were included. Patient demographics, free flap and microvascular anastomosis details were analysed. Primary outcome was flap failure. Secondary outcome was return to theatre. RESULTS: Two hundred and thirty-four free flaps were included (PAH 115; LGI 119). 60% were muscle flaps. Eighty-one percent of patients were male, with trauma the cause in 82%. PTA was used for microsurgical anastomosis in 70% of cases. Venae comitantes were preferred (96%) for venous anastomosis. PTA group showed a higher proportion of patients with trauma as the mechanism of injury. ATA group was more likely to have an end-to-end arterial anastomosis configuration. Total flap loss was 3.8%. There was no clinically significant difference in flap failure or return to theatre using ATA versus PTA. CONCLUSIONS: Incidence of lower limb free flap failure is low (<5%) and not influenced by use of ATA versus PTA for microsurgical anastomosis. The choice of target vessels for microsurgical reconstruction of the lower limb should be predicated upon factors other than aversion to one or another vessel. If all other microsurgical considerations are equal, the surgeon can exercise personal preference.


Subject(s)
Free Tissue Flaps , Plastic Surgery Procedures , Anastomosis, Surgical/adverse effects , Female , Free Tissue Flaps/blood supply , Humans , Lower Extremity/blood supply , Lower Extremity/surgery , Male , Microsurgery , Postoperative Complications/epidemiology , Plastic Surgery Procedures/adverse effects , Retrospective Studies , Tibial Arteries/surgery , Treatment Outcome
8.
Stem Cell Rev Rep ; 18(4): 1444-1460, 2022 04.
Article in English | MEDLINE | ID: mdl-35013938

ABSTRACT

Mesenchymal stromal cells (MSCs) ameliorate pre-clinical sepsis and sepsis-associated acute kidney injury (SA-AKI) but clinical trials of single-dose MSCs have not indicated robust efficacy. This study investigated immunomodulatory effects of a novel MSC product (CD362-selected human umbilical cord-derived MSCs [hUC-MSCs]) in mouse endotoxemia and polymicrobial sepsis models. Initially, mice received intra-peritoneal (i.p.) lipopolysaccharide (LPS) followed by single i.p. doses of hUC-MSCs or vehicle. Next, mice underwent cecal ligation and puncture (CLP) followed by intravenous (i.v.) doses of hUC-MSCs at 4 h or 4 and 28 h. Analyses included serum/plasma assays of biochemical indices, inflammatory mediators and the AKI biomarker NGAL; multi-color flow cytometry of peritoneal macrophages (LPS) and intra-renal immune cell subpopulations (CLP) and histology/immunohistochemistry of kidney (CLP). At 72 h post-LPS injections, hUC-MSCs reduced serum inflammatory mediators and peritoneal macrophage M1/M2 ratio. Repeated, but not single, hUC-MSC doses administered at 48 h post-CLP resulted in lower serum concentrations of inflammatory mediators, lower plasma NGAL and reversal of sepsis-associated depletion of intra-renal T cell and myeloid cell subpopulations. Hierarchical clustering analysis of all 48-h serum/plasma analytes demonstrated partial co-clustering of repeated-dose hUC-MSC CLP animals with a Sham group but did not reveal a distinct signature of response to therapy. It was concluded that repeated doses of CD362-selected hUC-MSCs are required to modulate systemic and local immune/inflammatory events in polymicrobial sepsis and SA-AKI. Inter-individual variability and lack of effect of single dose MSC administration in the CLP model are consistent with observations to date from early-phase clinical trials.


Subject(s)
Acute Kidney Injury , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Sepsis , Acute Kidney Injury/therapy , Animals , Anti-Inflammatory Agents , Disease Models, Animal , Female , Humans , Inflammation Mediators , Lipocalin-2 , Lipopolysaccharides/pharmacology , Male , Mesenchymal Stem Cell Transplantation/methods , Mice , Sepsis/therapy , Umbilical Cord
9.
J Craniofac Surg ; 32(7): e680-e682, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34705374

ABSTRACT

ABSTRACT: Focal thinning of the calvarial bones unrelated to an underlying systemic disease is uncommon. Of such cases documented in the medical literature, most are bilateral parietal bone thinning, which tends to affect elderly females and results in bilateral symmetric, regularly shaped depressions of the skull. The authors describe 2 cases of unilateral, focal, irregularly shaped calvarial thinning in adolescent males that occurred without an obvious precipitating incident and were unrelated to systemic disease, a known syndrome or previous trauma. The nature and physical parameters of the deformities are demonstrated here and specific historic features such as age of onset and mode of obstetric delivery are explored. The clinical significance and potential pathogenesis of this finding is unclear, though these cases are relevant in highlighting a novel presentation that we henceforth term focal idiopathic calvarial thinning.


Subject(s)
Parietal Bone , Skull , Adolescent , Aged , Female , Humans , Male , Prevalence , Skull/diagnostic imaging
10.
JACS Au ; 1(2): 174-186, 2021 Feb 22.
Article in English | MEDLINE | ID: mdl-33778810

ABSTRACT

Optical microscopy techniques are ideal for live cell imaging for real-time nanoparticle tracking of nanoparticle localization. However, the quantification of nanoparticle uptake is usually evaluated by analytical methods that require cell isolation. Luminescent labeling of gold nanoparticles with transition metal probes yields particles with attractive photophysical properties, enabling cellular tracking using confocal and time-resolved microscopies. In the current study, gold nanoparticles coated with a red-luminescent ruthenium transition metal complex are used to quantify and track particle uptake and localization. Analysis of the red-luminescence signal from particles is used as a metric of cellular uptake, which correlates to total cellular gold and ruthenium content, independently measured and correlated by inductively coupled plasma mass spectrometry. Tracking of the luminescence signal provides evidence of direct diffusion of the nanoparticles across the cytoplasmic membrane with particles observed in the cytoplasm and mitochondria as nonclustered "free" nanoparticles. Electron microscopy and inhibition studies identified macropinocytosis of clusters of particles into endosomes as the major mechanism of uptake. Nanoparticles were tracked inside GFP-tagged cells by following the red-luminescence signal of the ruthenium complex. Tracking of the particles demonstrates their initial location in early endosomes and, later, in lysosomes and autophagosomes. Colocalization was quantified by calculating the Pearson's correlation coefficient between red and green luminescence signals and confirmed by electron microscopy. Accumulation of particles in autophagosomes correlated with biochemical evidence of active autophagy, but there was no evidence of detachment of the luminescent label or breakup of the gold core. Instead, accumulation of particles in autophagosomes caused organelle swelling, breakdown of the surrounding membranes, and endosomal release of the nanoparticles into the cytoplasm. The phenomenon of endosomal release has important consequences for the toxicity, cellular targeting, and therapeutic future applications of gold nanoparticles.

11.
Int J Cancer ; 148(5): 1245-1259, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33152121

ABSTRACT

Tumour stromal cells support tumourigenesis. We report that Syndecan-2 (SDC2) is expressed on a nonepithelial, nonhaematopoietic, nonendothelial stromal cell population within breast cancer tissue. In vitro, syndecan-2 modulated TGFß signalling (SMAD7, PAI-1), migration and immunosuppression of patient-derived tumour-associated stromal cells (TASCs). In an orthotopic immunocompromised breast cancer model, overexpression of syndecan-2 in TASCs significantly enhanced TGFß signalling (SMAD7, PAI-1), tumour growth and metastasis, whereas reducing levels of SDC2 in TASCs attenuated TGFß signalling (SMAD7, PAI-1, CXCR4), tumour growth and metastasis. To explore the potential for therapeutic application, a syndecan-2-peptide was generated that inhibited the migratory and immunosuppressive properties of TASCs in association with reduced expression of TGFß-regulated immunosuppressive genes, such as CXCR4 and PD-L1. Moreover, using an orthotopic syngeneic breast cancer model, overexpression of syndecan-2-peptide in TASCs reduced tumour growth and immunosuppression within the TME. These data provide evidence that targeting stromal syndecan-2 within the TME inhibits tumour growth and metastasis due to decreased TGFß signalling and increased immune control.


Subject(s)
Breast Neoplasms/drug therapy , Immune Evasion , Syndecan-2/antagonists & inhibitors , Animals , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Female , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Signal Transduction/drug effects , Stromal Cells/drug effects , Stromal Cells/physiology , Syndecan-2/physiology , Transforming Growth Factor beta/physiology , Tumor Microenvironment
12.
Mol Ther ; 28(9): 2023-2043, 2020 09 02.
Article in English | MEDLINE | ID: mdl-32531237

ABSTRACT

Mesenchymal stromal cells (MSCs) are a promising therapeutic option for multiple immune diseases/disorders; however, efficacy of MSC treatments can vary significantly. We present a novel licensing strategy to improve the immunosuppressive capacity of MSCs. Licensing murine MSCs with transforming growth factor-ß1 (TGF-ß MSCs) significantly improved their ability to modulate both the phenotype and secretome of inflammatory bone marrow-derived macrophages and significantly increased the numbers of regulatory T lymphocytes following co-culture assays. These TGF-ß MSC-expanded regulatory T lymphocytes also expressed significantly higher levels of PD-L1 and CD73, indicating enhanced suppressive potential. Detailed analysis of T lymphocyte co-cultures revealed modulation of secreted factors, most notably elevated prostaglandin E2 (PGE2). Furthermore, TGF-ß MSCs could significantly prolong rejection-free survival (69.2% acceptance rate compared to 21.4% for unlicensed MSC-treated recipients) in a murine corneal allograft model. Mechanistic studies revealed that (1) therapeutic efficacy of TGF-ß MSCs is Smad2/3-dependent, (2) the enhanced immunosuppressive capacity of TGF-ß MSCs is contact-dependent, and (3) enhanced secretion of PGE2 (via prostaglandin EP4 [E-type prostanoid 4] receptor) by TGF-ß MSCs is the predominant mediator of Treg expansion and T cell activation and is associated with corneal allograft survival. Collectively, we provide compelling evidence for the use of TGF-ß1 licensing as an unconventional strategy for enhancing MSC immunosuppressive capacity.


Subject(s)
Allografts/immunology , Corneal Transplantation/adverse effects , Graft Rejection/immunology , Graft Rejection/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/drug effects , Transforming Growth Factor beta1/pharmacology , Animals , Cells, Cultured , Coculture Techniques/methods , Culture Media, Conditioned , Female , Graft Survival/immunology , Immune Tolerance/drug effects , Lymphocyte Activation/immunology , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Recombinant Proteins/pharmacology , T-Lymphocytes, Regulatory/immunology , Transplantation, Homologous/methods , Treatment Outcome
13.
Cells ; 9(6)2020 06 03.
Article in English | MEDLINE | ID: mdl-32503335

ABSTRACT

Individuals living with type 1 diabetes mellitus may experience an increased risk of long bone fracture. These fractures are often slow to heal, resulting in delayed reunion or non-union. It is reasonable to theorize that the underlying cause of these diabetes-associated osteopathies is faulty repair dynamics as a result of compromised bone marrow progenitor cell function. Here it was hypothesized that the administration of non-diabetic, human adult bone marrow-derived mesenchymal stromal cells (MSCs) would enhance diabetic fracture healing. Human MSCs were locally introduced to femur fractures in streptozotocin-induced diabetic mice, and the quality of de novo bone was assessed eight weeks later. Biodistribution analysis demonstrated that the cells remained in situ for three days following administration. Bone bridging was evident in all animals. However, a large reparative callus was retained, indicating non-union. µCT analysis elucidated comparable callus dimensions, bone mineral density, bone volume/total volume, and volume of mature bone in all groups that received cells as compared to the saline-treated controls. Four-point bending evaluation of flexural strength, flexural modulus, and total energy to re-fracture did not indicate a statistically significant change as a result of cellular administration. An ex vivo lymphocytic proliferation recall assay indicated that the xenogeneic administration of human cells did not result in an immune response by the murine recipient. Due to this dataset, the administration of non-diabetic bone marrow-derived MSCs did not support fracture healing in this pilot study.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/therapy , Fracture Healing , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Adult , Animals , Bone Marrow Cells/cytology , Disease Models, Animal , Humans , Lymphocytes/cytology , Male , Mice, Inbred C57BL , Pilot Projects
14.
Behav Brain Res ; 382: 112454, 2020 03 16.
Article in English | MEDLINE | ID: mdl-31926214

ABSTRACT

Research on the impact of diet and memory has garnered considerable attention while exploring the link between obesity and cognitive impairment. High-fat diet (HFD) rodent models recapitulate the obesity phenotype and subsequent cognitive impairments. While it is known that HFD is associated with sensory impairment, little attention has been given to the potential role these sensory deficits may play in recognition memory testing, one of the most commonly used cognitive tests. Because mice utilize their facial whiskers as their primary sensory apparatus, we modified a common recognition test, the novel object recognition task, by replacing objects with sandpaper grits at ground level, herein referred to as the novel tactile recognition task (NTR). First, we tested whisker-manipulated mice in this task to determine its reliance on intact whiskers. Then, we tested the HFD mouse in the NTR. Finally, to ensure that deficits in the NTR are due to cognitive impairment and not HFD-induced sensory deficiencies, we tested the whisker sensitivity of HFD mice via the corner test. Our results indicate that the NTR is a whisker dependent task, and that HFD mice exhibit tactile recognition memory impairment, not accompanied by whisker sensory deficits.


Subject(s)
Diet, High-Fat , Discrimination, Psychological , Memory , Recognition, Psychology , Touch , Animals , Behavior, Animal , Cognitive Dysfunction/etiology , Male , Mice, Inbred C57BL , Physical Stimulation , Touch Perception , Vibrissae
15.
Neurobiol Dis ; 132: 104580, 2019 12.
Article in English | MEDLINE | ID: mdl-31454547

ABSTRACT

Obesity is a global pandemic associated with macro- and microvascular endothelial dysfunction. Microvascular endothelial dysfunction has recently emerged as a significant risk factor for the development of cognitive impairment. In this review, we present evidence from clinical and preclinical studies supporting a role for obesity in cognitive impairment. Next, we discuss how obesity-related hyperinsulinemia/insulin resistance, systemic inflammation, and gut dysbiosis lead to cognitive impairment through induction of endothelial dysfunction and disruption of the blood brain barrier. Finally, we outline the potential clinical utility of dietary interventions, exercise, and bariatric surgery in circumventing the impacts of obesity on cognitive function.


Subject(s)
Blood-Brain Barrier/physiopathology , Cognitive Dysfunction/etiology , Endothelium, Vascular/physiopathology , Obesity/complications , Animals , Cognitive Dysfunction/physiopathology , Humans , Obesity/physiopathology
16.
Mol Neurodegener ; 14(1): 22, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31182115

ABSTRACT

In order for Alzheimer's disease (AD) to manifest, cells must communicate "pathogenic material" such as proteins, signaling molecules, or genetic material to ensue disease propagation. Small extracellular vesicles are produced via the endocytic pathways and released by nearly all cell types, including neurons. Due to their intrinsic interrelationship with endocytic processes and autophagy, there has been increased interest in studying the role of these neuronally-derived extracellular vesicles (NDEVs) in the propagation of AD. Pathologic cargo associated with AD have been found in a number of studies, and NDEVs have been shown to induce pathogenesis in vivo and in vitro. Exogenous NDEVs are also shown to reduce plaque burden in AD models. Thus, the NDEV has the potential to become a useful biomarker, a pathologic potentiator, and a therapeutic opportunity. While the field of NDEV research in AD is still in its infancy, we review the current literature supporting these three claims.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Neurons/metabolism , tau Proteins/metabolism , Alzheimer Disease/diagnosis , Amyloid beta-Peptides/metabolism , Extracellular Vesicles/metabolism , Humans
17.
Aesthetic Plast Surg ; 42(2): 447-450, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29026955

ABSTRACT

Rib osteomyelitis is an infrequently occurring but important complication of breast implant surgery. Although prosthetic or surgical site infection (SSI) and rib osteomyelitis as separate entities are well described in the literature, only five cases of rib or sternal osteomyelitis related to implant placement have been reported globally. Historically patients who experience this complication have not demonstrated an identifiable prevalence of the traditional risk factors associated with SSI or rib osteomyelitis. This report describes the sequence of clinical manifestations of an unusual case of breast implants complicated by rib osteomyelitis. A 56-year-old female underwent mastectomy and insertion of tissue expanders for bilateral invasive ductal carcinoma following which the tissue expanders became infected in the early postoperative period and were subsequently removed. The patient underwent successful expander insertion and subsequent implant exchange surgery several years later and enjoyed an uncomplicated recovery from this. Following nipple reconstruction more than 12 months after successful implant placement, she presented with Staphylococcus epidermidis bacteremia and a left-sided clinical peri-implant infection. Upon removal of her implant, an intraoperative discovery of rib necrosis/osteomyelitis was made for which she was treated. To provide context, the literature was reviewed for other reported cases of rib osteomyelitis following breast implant surgery. This patient, in combination with others reported in the literature, emphasises the diagnostic difficulties posed by this condition as a result of its low incidence and variable or absent clinical features. LEVEL OF EVIDENCE V: This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .


Subject(s)
Breast Implantation/adverse effects , Breast Implants/adverse effects , Breast Neoplasms/surgery , Osteomyelitis/etiology , Prosthesis-Related Infections/surgery , Ribs/microbiology , Anti-Bacterial Agents/therapeutic use , Australia , Breast Implantation/methods , Breast Neoplasms/pathology , Device Removal , Female , Follow-Up Studies , Humans , Mastectomy/methods , Middle Aged , Osteomyelitis/drug therapy , Osteomyelitis/physiopathology , Prosthesis-Related Infections/diagnosis , Prosthesis-Related Infections/microbiology , Rare Diseases , Ribs/pathology , Risk Assessment , Treatment Outcome
18.
J Infect Prev ; 17(4): 171-176, 2016 Jul.
Article in English | MEDLINE | ID: mdl-28989476

ABSTRACT

BACKGROUND: Environmental contamination has been associated with over half of methicillin-resistant Staphylococcus aureus (MRSA) outbreaks in hospitals. We explored if a hospital-wide environmental and patient cleaning protocol would lower hospital acquired MRSA rates and associated costs. OBJECTIVE: This study evaluates the impact of implementing a hospital-wide environmental and patient cleaning protocol on the rate of MRSA infection and the potential cost benefit of the intervention. METHODS: A retrospective, pre-post interventional study design was used. The intervention comprised a combination of enhanced environmental cleaning of high touch surfaces, daily washing of patients with benzalkonium chloride, and targeted isolation of patients with active infection. The rate of MRSA infection per 1000 patient days (PD) was compared with the rate after the intervention (Steiros Algorithm®) was implemented. A cost-benefit analysis based on the number of MRSA infections avoided was conducted. RESULTS: The MRSA rates decreased by 96% from 3.04 per 1000 PD to 0.11 per 1000 PD (P <0.0001). This reduction in MRSA infections, avoided an estimated $1,655,143 in healthcare costs. DISCUSSION: Implementation of this hospital-wide protocol appears to be associated with a reduction in the rate of MRSA infection and therefore a reduction in associated healthcare costs.

19.
Acta Biomater ; 19: 66-75, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25818949

ABSTRACT

Hyaluronic acid (HA) has received a lot of attention recently as a biomaterial with applications in wound healing, drug delivery, vascular repair and cell and/or gene delivery. Interstitial cystitis (IC) is characterised by an increase in the permeability of the bladder wall urothelium due to loss of the glycosaminoglycan (GAG) layer. The degradation of the urothelium leads to chronic pain and urinary dysfunction. The aetiology of the degradation of the GAG layer in this instance is currently unknown. At a clinical level, GAG replacement therapy using a HA solution is currently utilised as a treatment for IC. However, there is a significant lack of data on the mechanism of action of HA in IC. The current study investigates the mechanistic effect of clinically relevant HA treatment on an in vitro model of IC using urothelial cells, examining cytokine secretion, GAG secretion and trans-epithelial permeability. This study demonstrates that HA can significantly decrease induced cytokine secretion (4-5 fold increase), increase sulphated GAG production (2-fold increase) and without altering tight junction expression, decrease trans-epithelial permeability, suggesting that the HA pathway is a clinical target and potential treatment vector.


Subject(s)
Cystitis, Interstitial/drug therapy , Cystitis, Interstitial/immunology , Hyaluronic Acid/administration & dosage , Interleukin-6/immunology , Interleukin-8/immunology , Urothelium/immunology , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/immunology , Cystitis, Interstitial/pathology , Dose-Response Relationship, Drug , Glycosaminoglycans/immunology , Humans , Materials Testing , Tight Junctions/drug effects , Tight Junctions/immunology , Urothelium/drug effects , Urothelium/pathology
20.
Stem Cell Res Ther ; 5(2): 51, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-25099622

ABSTRACT

Compromised bone-regenerating capability following a long bone fracture is often the result of reduced host bone marrow (BM) progenitor cell numbers and efficacy. Without surgical intervention, these malunions result in mobility restrictions, deformities, and disability. The clinical application of BM-derived mesenchymal stem cells (MSCs) is a feasible, minimally invasive therapeutic option to treat non-union fractures. This review focuses on novel, newly identified cell surface markers in both the mouse and human enabling the isolation and purification of osteogenic progenitor cells as well as their direct and indirect contributions to fracture repair upon administration. Furthermore, clinical success to date is summarized with commentary on autologous versus allogeneic cell sources and the methodology of cell administration. Given our clinical success to date in combination with recent advances in the identification, isolation, and mechanism of action of MSCs, there is a significant opportunity to develop improved technologies for defining therapeutic MSCs and potential to critically inform future clinical strategies for MSC-based bone regeneration.


Subject(s)
Bone Regeneration/physiology , Fractures, Bone/therapy , Mesenchymal Stem Cell Transplantation/methods , Animals , Humans , Mice , Wound Healing/physiology
SELECTION OF CITATIONS
SEARCH DETAIL