Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Sci Transl Med ; 15(714): eadg8656, 2023 09 20.
Article in English | MEDLINE | ID: mdl-37729432

ABSTRACT

Human diseases may be modeled in animals to allow preclinical assessment of putative new clinical interventions. Recent, highly publicized failures of large clinical trials called into question the rigor, design, and value of preclinical assessment. We established the Stroke Preclinical Assessment Network (SPAN) to design and implement a randomized, controlled, blinded, multi-laboratory trial for the rigorous assessment of candidate stroke treatments combined with intravascular thrombectomy. Efficacy and futility boundaries in a multi-arm multi-stage statistical design aimed to exclude from further study highly effective or futile interventions after each of four sequential stages. Six independent research laboratories performed a standard focal cerebral ischemic insult in five animal models that included equal numbers of males and females: young mice, young rats, aging mice, mice with diet-induced obesity, and spontaneously hypertensive rats. The laboratories adhered to a common protocol and efficiently enrolled 2615 animals with full data completion and comprehensive animal tracking. SPAN successfully implemented treatment masking, randomization, prerandomization inclusion and exclusion criteria, and blinded assessment of outcomes. The SPAN design and infrastructure provide an effective approach that could be used in similar preclinical, multi-laboratory studies in other disease areas and should help improve reproducibility in translational science.


Subject(s)
Ischemic Stroke , Stroke , Female , Humans , Male , Rats , Animals , Mice , Rodentia , Laboratories , Reproducibility of Results , Stroke/therapy
3.
Stroke ; 54(2): 620-631, 2023 02.
Article in English | MEDLINE | ID: mdl-36601951

ABSTRACT

The Stroke Preclinical Assessment Network (SPAN) is a multicenter preclinical trial platform using rodent models of transient focal cerebral ischemia to address translational failure in experimental stroke. In addition to centralized randomization and blinding and large samples, SPAN aimed to introduce heterogeneity to simulate the heterogeneity embodied in clinical trials for robust conclusions. Here, we report the heterogeneity introduced by allowing the 6 SPAN laboratories to vary most of the biological and experimental model variables and the impact of this heterogeneity on middle cerebral artery occlusion (MCAo) performance. We included the modified intention-to-treat population of the control mouse cohort of the first SPAN trial (n=421) and examined the biological and procedural independent variables and their covariance. We then determined their impact on the dependent variables cerebral blood flow drop during MCAo, time to achieve MCAo, and total anesthesia duration using multivariable analyses. We found heterogeneity in biological and procedural independent variables introduced mainly by the site. Consequently, all dependent variables also showed heterogeneity among the sites. Multivariable analyses with the site as a random effect variable revealed filament choice as an independent predictor of cerebral blood flow drop after MCAo. Comorbidity, sex, use of laser Doppler flow to monitor cerebral blood flow, days after trial onset, and maintaining anesthesia throughout the MCAo emerged as independent predictors of time to MCAo. Total anesthesia duration was predicted by most independent variables. We present with high granularity the heterogeneity introduced by the biological and model selections by the testing sites in the first trial of cerebroprotection in rodent transient filament MCAo by SPAN. Rather than trying to homogenize all variables across all sites, we embraced the heterogeneity to better approximate clinical trials. Awareness of the heterogeneity, its sources, and how it impacts the study performance may further improve the study design and statistical modeling for future multicenter preclinical trials.


Subject(s)
Ischemic Attack, Transient , Stroke , Mice , Animals , Disease Models, Animal , Infarction, Middle Cerebral Artery , Research Design , Cerebrovascular Circulation/physiology , Multicenter Studies as Topic
4.
Stroke ; 53(5): 1802-1812, 2022 05.
Article in English | MEDLINE | ID: mdl-35354299

ABSTRACT

Cerebral ischemia and reperfusion initiate cellular events in brain that lead to neurological disability. Investigating these cellular events provides ample targets for developing new treatments. Despite considerable work, no such therapy has translated into successful stroke treatment. Among other issues-such as incomplete mechanistic knowledge and faulty clinical trial design-a key contributor to prior translational failures may be insufficient scientific rigor during preclinical assessment: nonblinded outcome assessment; missing randomization; inappropriate sample sizes; and preclinical assessments in young male animals that ignore relevant biological variables, such as age, sex, and relevant comorbid diseases. Promising results are rarely replicated in multiple laboratories. We sought to address some of these issues with rigorous assessment of candidate treatments across 6 independent research laboratories. The Stroke Preclinical Assessment Network (SPAN) implements state-of-the-art experimental design to test the hypothesis that rigorous preclinical assessment can successfully reduce or eliminate common sources of bias in choosing treatments for evaluation in clinical studies. SPAN is a randomized, placebo-controlled, blinded, multilaboratory trial using a multi-arm multi-stage protocol to select one or more putative stroke treatments with an implied high likelihood of success in human clinical stroke trials. The first stage of SPAN implemented procedural standardization and experimental rigor. All participating research laboratories performed middle cerebral artery occlusion surgery adhering to a common protocol and rapidly enrolled 913 mice in the first of 4 planned stages with excellent protocol adherence, remarkable data completion and low rates of subject loss. SPAN stage 1 successfully implemented treatment masking, randomization, prerandomization inclusion/exclusion criteria, and blinded assessment to exclude bias. Our data suggest that a large, multilaboratory, preclinical assessment effort to reduce known sources of bias is feasible and practical. Subsequent SPAN stages will evaluate candidate treatments for potential success in future stroke clinical trials using aged animals and animals with comorbid conditions.


Subject(s)
Brain Ischemia , Stroke , Aged , Animals , Brain , Brain Ischemia/therapy , Feasibility Studies , Humans , Infarction, Middle Cerebral Artery/therapy , Male , Mice , Stroke/therapy
7.
Ital J Pediatr ; 46(1): 83, 2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32527281

ABSTRACT

BACKGROUND: Data about acute poisoning in Italian pediatric patients are obsolete or absent. This study would partially fill this exiting gap and compare the scene with others around the world. METHODS: A retrospective evaluation was performed on a 2012-2017 data registry of the Children's Emergency Department at the Regina Margherita Hospital of Turin, where 1030 children under age 14 were accepted with a diagnosis of acute intoxication. RESULTS: The median age of the patients was 2.2 years (IQR 2.3) and 55% were male. Events occurred mostly in children aged 1-4 years (n = 751, 72.9%). Six hundred and eight patients (59%) were exposed to Nonpharmaceutical agents, the household cleaning products being the more frequent (n = 298, 49%). Exposure to Pharmaceuticals were 422 (41%); the most common Pharmaceuticals were analgesics (n = 88, 20.8%), psychotropics (n = 77, 18.2%) and cardiovascular (n = 53, 12.6%) drugs. The 85% of the intoxications occurred accidentally, the 10.6% as therapeutic error, the 2.3% as suicide attempts and the 1.5% for recreational purposes. No patient died. CONCLUSIONS: Despite acute poisoning being a relevant problem in pediatric emergency, our results would seem to paint a less worrying picture if compared to other countries, mainly when considering the children hospitalized in the pediatric intensive care unit and the number of deaths. Nevertheless, our study might represent a tool for public health authorities to program incisive interventions.


Subject(s)
Emergency Service, Hospital , Poisoning/epidemiology , Adolescent , Age Distribution , Child , Child, Preschool , Female , Hospitalization , Humans , Infant , Infant, Newborn , Italy , Male , Poisoning/diagnosis , Poisoning/therapy , Registries , Retrospective Studies , Sex Distribution
8.
Transl Stroke Res ; 9(3): 258-266, 2018 06.
Article in English | MEDLINE | ID: mdl-29633156

ABSTRACT

The first annual Stroke Translational Research Advancement Workshop (STRAW), entitled "Uncovering the Rosetta Stone: Key Elements in Translating Stroke Therapeutics from Pre-Clinical to Clinical" was held at the University of Kentucky on October 4-5, 2017. This workshop was organized by the Center for Advanced Translational Stroke Science. The workshop consisted of 2 days of activities. These included three presentations establishing the areas of research in stroke therapeutics, discussing the routes for translation from bench to bedside, and identifying successes and failures in the field. On day 2, grant funding opportunities and goals for the National Institute for Neurological Diseases and Stroke were presented. In addition, the meeting also included break-out sessions designed to connect researchers in areas of stroke, and to foster potential collaborations. Finally, the meeting concluded with an open discussion among attendees led by a panel of experts.


Subject(s)
Brain Ischemia/therapy , Congresses as Topic , Disease Models, Animal , Stroke/therapy , Translational Research, Biomedical , Animals , Humans , Translational Research, Biomedical/economics , Translational Research, Biomedical/methods , Translational Research, Biomedical/organization & administration
10.
Brain Res Bull ; 135: 47-52, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28923306

ABSTRACT

Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system characterized by recurrent and progressive demyelination/remyelination cycles, neuroinflammation, oligodendrocyte loss, and axonal pathology. Baicalein isolated from the roots of Scutellaria baicalensis has been shown to exert anti-inflammatory and antioxidant effects. The cuprizone model is an established mouse model of MS and causes demyelination and motor dysfunction and induces neuroinflammation, such as glial activation and pro-inflammatory cytokine production. To determine whether Baicalein attenuates cuprizone-induced demyelination, we administrated Baicalein to cuprizone-exposed mice. Baicalein attenuated weight loss (P<0.05) and motor dysfunction (P<0.05) in the cuprizone model mice. Baicalein treatment effectively suppressed the demyelination (P<0.01) and gene expressions of CNP (P<0.05) and MBP (P<0.05). Baicalein treatment also inhibited the cuprizone-induced increase in Iba1-positive microglia (P<0.001), GFAP-positive astrocytes (P<0.001), and the gene expressions of CD11b (P<0.01), GFAP (P<0.05), TNFα (P<0.05), IL-1ß (P<0.05), and iNOS (p<0.01). We found that Baicalein treatment attenuated cuprizone-induced demyelination, glial activation, pro-inflammatory cytokine expression, and motor dysfunction. Our results suggest that Baicalein may be a useful therapeutic agent in demyelinating diseases to suppress neuroinflammation.


Subject(s)
Demyelinating Diseases/drug therapy , Flavanones/metabolism , Animals , Astrocytes/drug effects , Cuprizone/metabolism , Cuprizone/pharmacology , Cytokines/metabolism , Demyelinating Diseases/chemically induced , Disease Models, Animal , Flavanones/pharmacology , Flavonoids/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Multiple Sclerosis/drug therapy , Myelin Sheath/pathology , Neuroimmunomodulation/drug effects , Oligodendroglia/drug effects , Tumor Necrosis Factor-alpha/metabolism
14.
Cell Mol Neurobiol ; 36(2): 281-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27095366

ABSTRACT

The World Health Organization reports that 47.5 million people are affected by dementia worldwide. With aging populations and 7.7 million new cases each year, the burden of illness due to dementia approaches crisis proportions. Despite significant advances in our understanding of the biology of Alzheimer's disease (AD), the leading dementia diagnosis, the actual causes of dementia in affected individuals are unknown except for rare fully penetrant genetic forms. Evidence from epidemiology and pathology studies indicates that damage to the vascular system is associated with an increased risk of many types of dementia. Both Alzheimer's pathology and cerebrovascular disease increase with age. How AD affects small blood vessel function and how vascular dysfunction contributes to the molecular pathology of Alzheimer's are areas of intense research. The science of vascular contributions to cognitive impairment and dementia (VCID) integrates diverse aspects of biology and incorporates the roles of multiple cell types that support the function of neural tissue. Because of the proven ability to prevent and treat cardiovascular disease and hypertension with population benefits for heart and stroke outcomes, it is proposed that understanding and targeting the biological mechanisms of VCID can have a similarly positive impact on public health.


Subject(s)
Cognitive Dysfunction/pathology , Dementia, Vascular/pathology , Research , Animals , Cost of Illness , Dementia, Vascular/diagnosis , Humans , Models, Biological
15.
Alzheimers Dement ; 11(6): 710-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25510382

ABSTRACT

Scientific evidence continues to demonstrate the linkage of vascular contributions to cognitive impairment and dementia such as Alzheimer's disease. In December, 2013, the Alzheimer's Association, with scientific input from the National Institute of Neurological Disorders and Stroke and the National Heart, Lung and Blood Institute from the National Institutes of Health, convened scientific experts to discuss the research gaps in our understanding of how vascular factors contribute to Alzheimer's disease and related dementia. This manuscript summarizes the meeting and the resultant discussion, including an outline of next steps needed to move this area of research forward.


Subject(s)
Cerebrovascular Disorders/physiopathology , Cognition Disorders/physiopathology , Dementia/physiopathology , Animals , Brain/blood supply , Brain/physiopathology , Congresses as Topic , Humans , National Heart, Lung, and Blood Institute (U.S.) , National Institute of Neurological Disorders and Stroke (U.S.) , Societies , United States
17.
Pediatr Nephrol ; 29(9): 1545-51, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24687448

ABSTRACT

BACKGROUND: Henoch-Schönlein purpura (HSP) nephritis and primary IgA nephropathy (pIgAN) present with glomerular IgA deposits, but differ with regard to clinical features. The suspected involvement of different immune system pathways is largely unknown. METHODS: This study was aimed at investigating some of the immunological features including Toll-like receptors (TLR), proteasome (PS)/immunoproteasome (iPS) switch, and the regulatory T cell system (Treg/Th17 cells) in 63 children with HSP with/without renal involvement and in 25 with pIgAN. Real-time PRC (Taqman) was used to quantify mRNA levels in peripheral blood mononuclear cells (PBMC). RESULTS: The expression of mRNAs encoding for TLR4 in both HSP and pIgAN was higher than in controls (HC) and in both diseases FoxP3mRNA and TGF-ß1mRNA expression was significantly lower than in HC. A switch from PS to iPS (LMP2/ß1) was detected only in PBMC of HSP and it correlated with the level of TLR2mRNA, which was selectively increased only in children with HSP. CONCLUSION: Children with HSP and pIgAN present with similar signs of engagement of the innate immunity and regulatory T cell depression. The increased immunoproteasome switch, which correlated with TLR2 activation, may suggest an innate immunity pathway peculiar to HSP vasculitic presentation. This research area also deserves further investigation for possible therapeutic applications.


Subject(s)
Glomerulonephritis, IGA/immunology , IgA Vasculitis/immunology , Proteasome Endopeptidase Complex/immunology , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptors/immunology , Child , Female , Humans , Male , Real-Time Polymerase Chain Reaction
18.
Article in English | MEDLINE | ID: mdl-24095587

ABSTRACT

Inflammation is a physiological response to exogenous and endogenous stimuli and, together with demyelination and immune system activation, is one of the key features of multiple sclerosis (MS). Arachidonic acid (AA) metabolism by cyclooxygenase (COX) and lipoxygenase (LO) enzymes leads to the production of proinflammatory eicosanoids, and stimulates cytokine production and activation of microglia and astrocytes, thereby contributing to MS pathology. Current therapies target the immune system but do not specifically target AA-related inflammatory pathway. Corticosteroids and non-steroidal anti-inflammatory drugs (NSAIDs) are frequently associated with immunomodulatory therapies to treat flu-like adverse effects. Few clinical and mounting preclinical data in MS show that AA metabolism contributes to immune system activation, demyelination and motor disabilities, and administration of NSAIDs reduces these symptoms. The beneficial effect of NSAIDs seems to be a prerogative of COX-2 selective inhibitors and suggests that NSAIDs selective for COX-2 may be more effective than mixed COX-1/2 inhibitors.


Subject(s)
Brain/metabolism , Cyclooxygenase 2/metabolism , Eicosanoids/biosynthesis , Multiple Sclerosis/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arachidonic Acid/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Brain/drug effects , Brain/pathology , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Eicosanoids/antagonists & inhibitors , Humans , Immunity, Innate , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lipoxygenase/metabolism , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology
19.
J Neurochem ; 127(3): 378-93, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23919613

ABSTRACT

Docosahexaenoic acid (22:6n-3) is the major brain n-3 polyunsaturated fatty acid and it is possible that docosahexaenoic acid is anti-inflammatory in the brain as it is known to be in other tissues. Using a combination of models including the fat-1 transgenic mouse, chronic dietary n-3 polyunsaturated fatty acid modulation in transgenic and wild-type mice, and acute direct brain infusion, we demonstrated that unesterified docosahexaenoic acid attenuates neuroinflammation initiated by intracerebroventricular lipopolysaccharide. Hippocampal neuroinflammation was assessed by gene expression and immunohistochemistry. Furthermore, docosahexaenoic acid protected against lipopolysaccharide-induced neuronal loss. Acute intracerebroventricular infusion of unesterified docosahexaenoic acid or its 12/15-lipoxygenase product and precursor to protectins and resolvins, 17S-hydroperoxy-docosahexaenoic acid, mimics anti-neuroinflammatory aspects of chronically increased unesterified docosahexaenoic acid. LC-MS/MS revealed that neuroprotectin D1 and several other docosahexaenoic acid-derived specialized pro-resolving mediators are present in the hippocampus. Acute intracerebroventricular infusion of 17S-hydroperoxy-docosahexaenoic acid increases hippocampal neuroprotectin D1 levels concomitant to attenuating neuroinflammation. These results show that unesterified docosahexaenoic acid is protective in a lipopolysaccharide-initiated mouse model of acute neuroinflammation, at least in part, via its conversion to specialized pro-resolving mediators; these docosahexaenoic acid stores may provide novel targets for the prevention and treatment(s) of neurological disorders with a neuroinflammatory component. Our study shows that chronically increased brain unesterified DHA levels, but not solely phospholipid DHA levels, attenuate neuroinflammation. Similar attenuations occur with acute increases in brain unesterified DHA or 17S-HpDHA levels, highlighting the importance of an available pool of precursor unesterified DHA for the production of enzymatically derived specialized pro-resolving mediators that are critical in the regulation of neuroinflammation.


Subject(s)
Anti-Inflammatory Agents , Docosahexaenoic Acids/pharmacology , Inflammation/pathology , Inflammation/prevention & control , Neuroprotective Agents , Animals , Carbon Radioisotopes , Diet , Docosahexaenoic Acids/metabolism , Fatty Acids, Nonesterified/metabolism , Fatty Acids, Omega-3/pharmacology , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Immunohistochemistry , Infusion Pumps, Implantable , Injections, Intraventricular , Lipopolysaccharides/pharmacology , Mass Spectrometry , Mice , Mice, Inbred C57BL , Phospholipids/metabolism
20.
J Neurochem ; 124(1): 59-68, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23083210

ABSTRACT

Several epidemiological and preclinical studies suggest that non-steroidal anti-inflammatory drugs (NSAIDs), which inhibit cyclooxygenase (COX), reduce the risk of Alzheimer's disease (AD) and can lower ß-amyloid (Aß) production and inhibit neuroinflammation. However, follow-up clinical trials, mostly using selective cyclooxygenase (COX)-2 inhibitors, failed to show any beneficial effect in AD patients with mild to severe cognitive deficits. Recent data indicated that COX-1, classically viewed as the homeostatic isoform, is localized in microglia and is actively involved in brain injury induced by pro-inflammatory stimuli including Aß, lipopolysaccharide, and interleukins. We hypothesized that neuroinflammation is critical for disease progression and selective COX-1 inhibition, rather than COX-2 inhibition, can reduce neuroinflammation and AD pathology. Here, we show that treatment of 20-month-old triple transgenic AD (3 × Tg-AD) mice with the COX-1 selective inhibitor SC-560 improved spatial learning and memory, and reduced amyloid deposits and tau hyperphosphorylation. SC-560 also reduced glial activation and brain expression of inflammatory markers in 3 × Tg-AD mice, and switched the activated microglia phenotype promoting their phagocytic ability. The present findings are the first to demonstrate that selective COX-1 inhibition reduces neuroinflammation, neuropathology, and improves cognitive function in 3 × Tg-AD mice. Thus, selective COX-1 inhibition should be further investigated as a potential therapeutic approach for AD.


Subject(s)
Alzheimer Disease/complications , Amyloidogenic Proteins/metabolism , Cyclooxygenase Inhibitors/therapeutic use , Memory Disorders/drug therapy , Memory Disorders/etiology , Pyrazoles/therapeutic use , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Mutation/genetics , Phagocytes/drug effects , Phosphorylation/drug effects , Presenilin-1/genetics , tau Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...