Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Nat Rev Cancer ; 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649750
2.
Cell Rep Med ; 5(1): 101351, 2024 01 16.
Article in English | MEDLINE | ID: mdl-38134930

ABSTRACT

Multiple sclerosis is a chronic inflammatory disease of the central nervous system. Whereas T cells are likely the main drivers of disease development, the striking efficacy of B cell-depleting therapies (BCDTs) underscore B cells' involvement in disease progression. How B cells contribute to multiple sclerosis (MS) pathogenesis-and consequently the precise mechanism of action of BCDTs-remains elusive. Here, we analyze the impact of BCDTs on the immune landscape in patients with MS using high-dimensional single-cell immunophenotyping. Algorithm-guided analysis reveals a decrease in circulating T follicular helper-like (Tfh-like) cells alongside increases in CD27 expression in memory T helper cells and Tfh-like cells. Elevated CD27 indicates disrupted CD27/CD70 signaling, as sustained CD27 activation in T cells leads to its cleavage. Immunohistological analysis shows CD70-expressing B cells at MS lesion sites. These results suggest that the efficacy of BCDTs may partly hinge upon the disruption of Th cell and B cell interactions.


Subject(s)
Multiple Sclerosis , Humans , Multiple Sclerosis/pathology , B-Lymphocytes , T-Lymphocytes, Helper-Inducer , Signal Transduction , Immunophenotyping
3.
Cell ; 187(1): 149-165.e23, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38134933

ABSTRACT

Deciphering the cell-state transitions underlying immune adaptation across time is fundamental for advancing biology. Empirical in vivo genomic technologies that capture cellular dynamics are currently lacking. We present Zman-seq, a single-cell technology recording transcriptomic dynamics across time by introducing time stamps into circulating immune cells, tracking them in tissues for days. Applying Zman-seq resolved cell-state and molecular trajectories of the dysfunctional immune microenvironment in glioblastoma. Within 24 hours of tumor infiltration, cytotoxic natural killer cells transitioned to a dysfunctional program regulated by TGFB1 signaling. Infiltrating monocytes differentiated into immunosuppressive macrophages, characterized by the upregulation of suppressive myeloid checkpoints Trem2, Il18bp, and Arg1, over 36 to 48 hours. Treatment with an antagonistic anti-TREM2 antibody reshaped the tumor microenvironment by redirecting the monocyte trajectory toward pro-inflammatory macrophages. Zman-seq is a broadly applicable technology, enabling empirical measurements of differentiation trajectories, which can enhance the development of more efficacious immunotherapies.


Subject(s)
Glioblastoma , Humans , Gene Expression Profiling , Glioblastoma/pathology , Immunotherapy , Killer Cells, Natural , Macrophages , Tumor Microenvironment , Single-Cell Analysis
4.
Nat Neurosci ; 26(10): 1701-1712, 2023 10.
Article in English | MEDLINE | ID: mdl-37749256

ABSTRACT

Interleukin-12 (IL-12) is a potent driver of type 1 immunity. Paradoxically, in autoimmune conditions, including of the CNS, IL-12 reduces inflammation. The underlying mechanism behind these opposing properties and the involved cellular players remain elusive. Here we map IL-12 receptor (IL-12R) expression to NK and T cells as well as neurons and oligodendrocytes. Conditionally ablating the IL-12R across these cell types in adult mice and assessing their susceptibility to experimental autoimmune encephalomyelitis revealed that the neuroprotective role of IL-12 is mediated by neuroectoderm-derived cells, specifically neurons, and not immune cells. In human brain tissue from donors with multiple sclerosis, we observe an IL-12R distribution comparable to mice, suggesting similar mechanisms in mice and humans. Combining flow cytometry, bulk and single-nucleus RNA sequencing, we reveal an IL-12-induced neuroprotective tissue adaption preventing early neurodegeneration and sustaining trophic factor release during neuroinflammation, thereby maintaining CNS integrity in mice.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Interleukin-12 , Neuroprotection , Adult , Animals , Humans , Mice , Central Nervous System , Mice, Inbred C57BL , Neuroinflammatory Diseases , Neurons/metabolism
5.
Sci Transl Med ; 15(697): eadf2281, 2023 05 24.
Article in English | MEDLINE | ID: mdl-37224228

ABSTRACT

Glioblastoma is the most aggressive primary brain tumor with an unmet need for more effective therapies. Here, we investigated combination therapies based on L19TNF, an antibody-cytokine fusion protein based on tumor necrosis factor that selectively localizes to cancer neovasculature. Using immunocompetent orthotopic glioma mouse models, we identified strong anti-glioma activity of L19TNF in combination with the alkylating agent CCNU, which cured the majority of tumor-bearing mice, whereas monotherapies only had limited efficacy. In situ and ex vivo immunophenotypic and molecular profiling in the mouse models revealed that L19TNF and CCNU induced tumor DNA damage and treatment-associated tumor necrosis. In addition, this combination also up-regulated tumor endothelial cell adhesion molecules, promoted the infiltration of immune cells into the tumor, induced immunostimulatory pathways, and decreased immunosuppression pathways. MHC immunopeptidomics demonstrated that L19TNF and CCNU increased antigen presentation on MHC class I molecules. The antitumor activity was T cell dependent and completely abrogated in immunodeficient mouse models. On the basis of these encouraging results, we translated this treatment combination to patients with glioblastoma. The clinical translation is ongoing but already shows objective responses in three of five patients in the first recurrent glioblastoma patient cohort treated with L19TNF in combination with CCNU (NCT04573192).


Subject(s)
Glioblastoma , Animals , Mice , Glioblastoma/drug therapy , T-Lymphocytes , Neoplasm Recurrence, Local , Tumor Necrosis Factor-alpha , Disease Models, Animal , Lomustine
6.
EMBO Mol Med ; 15(1): e16789, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36382364

ABSTRACT

Many efforts targeting amyloid-ß (Aß) plaques for the treatment of Alzheimer's Disease thus far have resulted in failures during clinical trials. Regional and temporal heterogeneity of efficacy and dependence on plaque maturity may have contributed to these disappointing outcomes. In this study, we mapped the regional and temporal specificity of various anti-Aß treatments through high-resolution light-sheet imaging of electrophoretically cleared brains. We assessed the effect on amyloid plaque formation and growth in Thy1-APP/PS1 mice subjected to ß-secretase inhibitors, polythiophenes, or anti-Aß antibodies. Each treatment showed unique spatiotemporal Aß clearance, with polythiophenes emerging as a potent anti-Aß compound. Furthermore, aligning with a spatial-transcriptomic atlas revealed transcripts that correlate with the efficacy of each Aß therapy. As observed in this study, there is a striking dependence of specific treatments on the location and maturity of Aß plaques. This may also contribute to the clinical trial failures of Aß-therapies, suggesting that combinatorial regimens may be significantly more effective in clearing amyloid deposition.


Subject(s)
Alzheimer Disease , Microscopy , Mice , Animals , Mice, Transgenic , Amyloid beta-Peptides/metabolism , Alzheimer Disease/drug therapy , Brain/metabolism , Plaque, Amyloid/drug therapy , Disease Models, Animal , Amyloid beta-Protein Precursor , Presenilin-1/pharmacology
7.
Nat Biomed Eng ; 6(9): 1031-1044, 2022 09.
Article in English | MEDLINE | ID: mdl-35835994

ABSTRACT

Deposits of amyloid-ß (Aß) in the brains of rodents can be analysed by invasive intravital microscopy on a submillimetre scale, or via whole-brain images from modalities lacking the resolution or molecular specificity to accurately characterize Aß pathologies. Here we show that large-field multifocal illumination fluorescence microscopy and panoramic volumetric multispectral optoacoustic tomography can be combined to longitudinally assess Aß deposits in transgenic mouse models of Alzheimer's disease. We used fluorescent Aß-targeted probes (the luminescent conjugated oligothiophene HS-169 and the oxazine-derivative AOI987) to transcranially detect Aß deposits in the cortex of APP/PS1 and arcAß mice with single-plaque resolution (8 µm) and across the whole brain (including the hippocampus and the thalamus, which are inaccessible by conventional intravital microscopy) at sub-150 µm resolutions. Two-photon microscopy, light-sheet microscopy and immunohistochemistry of brain-tissue sections confirmed the specificity and regional distributions of the deposits. High-resolution multiscale optical and optoacoustic imaging of Aß deposits across the entire brain in rodents thus facilitates the in vivo study of Aß accumulation by brain region and by animal age and strain.


Subject(s)
Amyloid beta-Peptides , Plaque, Amyloid , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Oxazines , Plaque, Amyloid/pathology
8.
Neuron ; 110(7): 1097-1099, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35390289

ABSTRACT

Metastases are the main cause of death in cancer patients. In a recent issue of Cell, Gonzalez et al. (2022) analyze gene expression on the single-cell level in brain metastases from various primary tumors. By profiling metastatic tumor cells and their niche, they demonstrate distinctive and shared features across metastases.


Subject(s)
Brain Neoplasms , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Humans
9.
Cell ; 185(8): 1373-1388.e20, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35381199

ABSTRACT

Systemic sclerosis (scleroderma, SSc) is an incurable autoimmune disease with high morbidity and mortality rates. Here, we conducted a population-scale single-cell genomic analysis of skin and blood samples of 56 healthy controls and 97 SSc patients at different stages of the disease. We found immune compartment dysfunction only in a specific subtype of diffuse SSc patients but global dysregulation of the stromal compartment, particularly in a previously undefined subset of LGR5+-scleroderma-associated fibroblasts (ScAFs). ScAFs are perturbed morphologically and molecularly in SSc patients. Single-cell multiome profiling of stromal cells revealed ScAF-specific markers, pathways, regulatory elements, and transcription factors underlining disease development. Systematic analysis of these molecular features with clinical metadata associates specific ScAF targets with disease pathogenesis and SSc clinical traits. Our high-resolution atlas of the sclerodermatous skin spectrum will enable a paradigm shift in the understanding of SSc disease and facilitate the development of biomarkers and therapeutic strategies.


Subject(s)
Scleroderma, Systemic , Cells, Cultured , Fibroblasts/metabolism , Fibrosis , Humans , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/genetics , Skin/metabolism
10.
Stroke ; 52(5): 1856-1860, 2021 05.
Article in English | MEDLINE | ID: mdl-33722060

ABSTRACT

Background and Purpose: The classic presentation of chronic (stage III) hemorrhagic stroke lesions is a fluid-filled cavity. In one of the most commonly used animal models of intracerebral hemorrhage (ICH), we noticed additional solid material within the chronic lesion. We examined the composition of those chronic ICH lesions and compared them with human autopsy cases. Methods: ICH was induced in rats by the injection of collagenase in the striatum. Tissue sections after hematoma resolution corresponding to 3 different chronic time points­28, 42, and 73 to 85 days post-ICH­were selected. Human autopsy reports at the University Hospital of Zurich were searched between 1990 and 2019 for ICH, and 3 chronic cases were found. The rat and human sections were stained with a variety of histopathologic markers. Results: Extensive collagenous material was observed in the chronic lesion after hematoma resolution in both the rat model and human autopsy cases. Additional immunostaining revealed that the material consisted primarily of a loose network of collagen 3 intermingled with occasional GFAP (glial fibrillary acidic protein)-positive processes and collagen 4. Conclusions: A key feature of the chronic ICH lesion is a loose network of collagen 3. The collagenase rat model reproduces the morphology and composition of the chronic human ICH lesion. While identifying new features of ICH lesion pathology, these results are important for treatment and recovery strategies.


Subject(s)
Brain/metabolism , Cerebral Hemorrhage/metabolism , Collagen/metabolism , Gray Matter/metabolism , Animals , Brain/pathology , Cerebral Hemorrhage/pathology , Disease Models, Animal , Female , Glial Fibrillary Acidic Protein/metabolism , Gray Matter/pathology , Humans , Male , Rats , Rats, Sprague-Dawley
11.
Neuropathol Appl Neurobiol ; 47(3): 454-459, 2021 04.
Article in English | MEDLINE | ID: mdl-33249605

ABSTRACT

Coronavirus disease 19 (COVID-19) is a rapidly evolving pandemic caused by the coronavirus Sars-CoV-2. Clinically manifest central nervous system symptoms have been described in COVID-19 patients and could be the consequence of commonly associated vascular pathology, but the detailed neuropathological sequelae remain largely unknown. A total of six cases, all positive for Sars-CoV-2, showed evidence of cerebral petechial hemorrhages and microthrombi at autopsy. Two out of six patients showed an elevated risk for disseminated intravascular coagulopathy according to current criteria and were excluded from further analysis. In the remaining four patients, the hemorrhages were most prominent at the grey and white matter junction of the neocortex, but were also found in the brainstem, deep grey matter structures and cerebellum. Two patients showed vascular intramural inflammatory infiltrates, consistent with Sars-CoV-2-associated endotheliitis, which was associated by elevated levels of the Sars-CoV-2 receptor ACE2 in the brain vasculature. Distribution and morphology of patchy brain microbleeds was clearly distinct from hypertension-related hemorrhage, critical illness-associated microbleeds and cerebral amyloid angiopathy, which was ruled out by immunohistochemistry. Cerebral microhemorrhages in COVID-19 patients could be a consequence of Sars- CoV-2-induced endotheliitis and more general vasculopathic changes and may correlate with an increased risk of vascular encephalopathy.


Subject(s)
COVID-19/complications , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/virology , Vasculitis, Central Nervous System/pathology , Vasculitis, Central Nervous System/virology , Aged , Aged, 80 and over , Endothelial Cells/pathology , Female , Humans , Male , Retrospective Studies , SARS-CoV-2
12.
Nat Commun ; 11(1): 5729, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33184269

ABSTRACT

Vasocative-intestinal-peptide (VIP+) and somatostatin (SST+) interneurons are involved in modulating barrel cortex activity and perception during active whisking. Here we identify a developmental transition point of structural and functional rearrangements onto these interneurons around the start of active sensation at P14. Using in vivo two-photon Ca2+ imaging, we find that before P14, both interneuron types respond stronger to a multi-whisker stimulus, whereas after P14 their responses diverge, with VIP+ cells losing their multi-whisker preference and SST+ neurons enhancing theirs. Additionally, we find that Ca2+ signaling dynamics increase in precision as the cells and network mature. Rabies virus tracings followed by tissue clearing, as well as photostimulation-coupled electrophysiology reveal that SST+ cells receive higher cross-barrel inputs compared to VIP+ neurons at both time points. In addition, whereas prior to P14 both cell types receive direct input from the sensory thalamus, after P14 VIP+ cells show reduced inputs and SST+ cells largely shift to motor-related thalamic nuclei.


Subject(s)
Interneurons/metabolism , Somatostatin/metabolism , Vasoactive Intestinal Peptide/metabolism , Vibrissae/innervation , Vibrissae/metabolism , Animals , Calcium , Electrophysiology/methods , Female , Image Processing, Computer-Assisted , Male , Mice , Microscopy, Confocal , Models, Animal , Nervous System/growth & development , Neurons/metabolism , Rabbits , Thalamus/physiology , Vibrissae/diagnostic imaging , Vibrissae/growth & development
13.
Stroke ; 51(12): 3719-3722, 2020 12.
Article in English | MEDLINE | ID: mdl-33054673

ABSTRACT

BACKGROUND AND PURPOSE: Case series indicating cerebrovascular disorders in coronavirus disease 2019 (COVID-19) have been published. Comprehensive workups, including clinical characteristics, laboratory, electroencephalography, neuroimaging, and cerebrospinal fluid findings, are needed to understand the mechanisms. METHODS: We evaluated 32 consecutive critically ill patients with COVID-19 treated at a tertiary care center from March 9 to April 3, 2020, for concomitant severe central nervous system involvement. Patients identified underwent computed tomography, magnetic resonance imaging, electroencephalography, cerebrospinal fluid analysis, and autopsy in case of death. RESULTS: Of 32 critically ill patients with COVID-19, 8 (25%) had severe central nervous system involvement. Two presented with lacunar ischemic stroke in the early phase and 6 with prolonged impaired consciousness after termination of analgosedation. In all but one with delayed wake-up, neuroimaging or autopsy showed multiple cerebral microbleeds, in 3 with additional subarachnoid hemorrhage and in 2 with additional small ischemic lesions. In 3 patients, intracranial vessel wall sequence magnetic resonance imaging was performed for the first time to our knowledge. All showed contrast enhancement of vessel walls in large cerebral arteries, suggesting vascular wall pathologies with an inflammatory component. Reverse transcription-polymerase chain reactions for SARS-CoV-2 in cerebrospinal fluid were all negative. No intrathecal SARS-CoV-2-specific IgG synthesis was detectable. CONCLUSIONS: Different mechanisms of cerebrovascular disorders might be involved in COVID-19. Acute ischemic stroke might occur early. In a later phase, microinfarctions and vessel wall contrast enhancement occur, indicating small and large cerebral vessels involvement. Central nervous system disorders associated with COVID-19 may lead to long-term disabilities. Mechanisms should be urgently investigated to develop neuroprotective strategies.


Subject(s)
COVID-19/diagnostic imaging , Cerebral Arteries/diagnostic imaging , Cerebral Hemorrhage/diagnostic imaging , Cerebrovascular Disorders/diagnostic imaging , Ischemic Stroke/diagnostic imaging , Aged , Antibodies, Viral/cerebrospinal fluid , Brain Ischemia/diagnostic imaging , Brain Ischemia/etiology , COVID-19/cerebrospinal fluid , COVID-19/complications , COVID-19/physiopathology , COVID-19 Nucleic Acid Testing , COVID-19 Serological Testing , Cerebral Hemorrhage/etiology , Cerebrospinal Fluid/immunology , Cerebrospinal Fluid/virology , Cerebrovascular Disorders/cerebrospinal fluid , Cerebrovascular Disorders/etiology , Cerebrovascular Disorders/physiopathology , Consciousness Disorders/etiology , Consciousness Disorders/physiopathology , Contrast Media , Critical Illness , Electroencephalography , Female , Humans , Ischemic Stroke/etiology , Magnetic Resonance Imaging , Male , Middle Aged , SARS-CoV-2 , Severity of Illness Index , Switzerland , Tertiary Care Centers , Tomography, X-Ray Computed
15.
Nat Methods ; 16(11): 1105-1108, 2019 11.
Article in English | MEDLINE | ID: mdl-31527839

ABSTRACT

Light-sheet microscopy is an ideal technique for imaging large cleared samples; however, the community is still lacking instruments capable of producing volumetric images of centimeter-sized cleared samples with near-isotropic resolution within minutes. Here, we introduce the mesoscale selective plane-illumination microscopy initiative, an open-hardware project for building and operating a light-sheet microscope that addresses these challenges and is compatible with any type of cleared or expanded sample ( www.mesospim.org ).


Subject(s)
Microscopy, Fluorescence/instrumentation , Animals , Chick Embryo , Microscopy, Fluorescence/methods , Software
16.
J Cutan Pathol ; 46(8): 570-578, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30927294

ABSTRACT

BACKGROUND: Metastatic tumor spread is a complex multistep process. Due to the blood-brain barrier, metastasis to the central nervous system is restrictive with a distinct predilection for certain tumor types. In melanoma patients, brain metastasis is a common endpoint with the majority showing evidence of widespread disease at autopsy. In a previous murine melanoma model, we have shown that melanoma cells migrate along preexisting vessels into the brain, showing angiotropism/vascular co-option and pericytic mimicry. METHODS: Using conventional morphology and immunohistochemistry, we analyze brain metastases from eight autopsy cases. In addition, tissue clearing, which enables three-dimensional visualization over a distance of 100 µm is used. RESULTS: We show the angiotropic localization of melanoma deposits in the brains in all eight autopsy cases. Tissue clearing techniques have allowed visualization of melanoma cells in one case exclusively along the abluminal surface of brain blood vessels over a distance of 100 µm, thus showing pericytic mimicry. CONCLUSIONS: Our analyses show clear-cut evidence of angiotropism and pericytic mimicry of melanoma cells within the brain over some distance. In addition, these results support the hypothesis of metastasis along pathways other than hematogenous spread, or extravascular migratory metastasis (EVMM). During EVMM, melanoma cells may metastasize to the brain through pericytic mimicry, circumventing the blood-brain barrier.


Subject(s)
Blood-Brain Barrier , Brain Neoplasms , Cell Movement , Melanoma , Pericytes , Skin Neoplasms , Adult , Aged , Autopsy , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Female , Humans , Male , Melanoma/metabolism , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Pericytes/metabolism , Pericytes/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
17.
Brain ; 142(4): 885-902, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30805583

ABSTRACT

Brain calcifications are commonly detected in aged individuals and accompany numerous brain diseases, but their functional importance is not understood. In cases of primary familial brain calcification, an autosomally inherited neuropsychiatric disorder, the presence of bilateral brain calcifications in the absence of secondary causes of brain calcification is a diagnostic criterion. To date, mutations in five genes including solute carrier 20 member 2 (SLC20A2), xenotropic and polytropic retrovirus receptor 1 (XPR1), myogenesis regulating glycosidase (MYORG), platelet-derived growth factor B (PDGFB) and platelet-derived growth factor receptor ß (PDGFRB), are considered causal. Previously, we have reported that mutations in PDGFB in humans are associated with primary familial brain calcification, and mice hypomorphic for PDGFB (Pdgfbret/ret) present with brain vessel calcifications in the deep regions of the brain that increase with age, mimicking the pathology observed in human mutation carriers. In this study, we characterize the cellular environment surrounding calcifications in Pdgfbret/ret animals and show that cells around vessel-associated calcifications express markers for osteoblasts, osteoclasts and osteocytes, and that bone matrix proteins are present in vessel-associated calcifications. Additionally, we also demonstrate the osteogenic environment around brain calcifications in genetically confirmed primary familial brain calcification cases. We show that calcifications cause oxidative stress in astrocytes and evoke expression of neurotoxic astrocyte markers. Similar to previously reported human primary familial brain calcification cases, we describe high interindividual variation in calcification load in Pdgfbret/ret animals, as assessed by ex vivo and in vivo quantification of calcifications. We also report that serum of Pdgfbret/ret animals does not differ in calcification propensity from control animals and that vessel calcification occurs only in the brains of Pdgfbret/ret animals. Notably, ossification of vessels and astrocytic neurotoxic response is associated with specific behavioural and cognitive alterations, some of which are associated with primary familial brain calcification in a subset of patients.


Subject(s)
Astrocytes/metabolism , Ossification, Heterotopic/pathology , Proto-Oncogene Proteins c-sis/metabolism , Aged , Animals , Brain/pathology , Brain Diseases/genetics , Calcinosis/pathology , Female , Humans , Male , Mice , Mutation , Osteogenesis/physiology , Oxidative Stress , Pedigree , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/physiology , Receptor, Platelet-Derived Growth Factor beta/genetics , Sodium-Phosphate Cotransporter Proteins, Type III/genetics , Xenotropic and Polytropic Retrovirus Receptor
19.
PLoS Pathog ; 14(11): e1007424, 2018 11.
Article in English | MEDLINE | ID: mdl-30496289

ABSTRACT

Transmissible spongiform encephalopathies (TSEs) are caused by the prion, which consists essentially of PrPSc, an aggregated, conformationally modified form of the cellular prion protein (PrPC). Although TSEs can be experimentally transmitted by intracerebral inoculation, most instances of infection in the field occur through extracerebral routes. The epidemics of kuru and variant Creutzfeldt-Jakob disease were caused by dietary exposure to prions, and parenteral administration of prion-contaminated hormones has caused hundreds of iatrogenic TSEs. In all these instances, the development of postexposure prophylaxis relies on understanding of how prions propagate from the site of entry to the brain. While much evidence points to lymphoreticular invasion followed by retrograde transfer through peripheral nerves, prions are present in the blood and may conceivably cross the blood-brain barrier directly. Here we have addressed the role of the blood-brain barrier (BBB) in prion disease propagation using Pdgfbret/ret mice which possess a highly permeable BBB. We found that Pdgfbret/ret mice have a similar prion disease incubation time as their littermate controls regardless of the route of prion transmission. These surprising results indicate that BBB permeability is irrelevant to the initiation of prion disease, even when prions are administered parenterally.


Subject(s)
Blood-Brain Barrier/metabolism , Prion Diseases/metabolism , Prions/metabolism , Animals , Biological Transport , Brain/blood supply , Brain/pathology , Cattle , Creutzfeldt-Jakob Syndrome/pathology , Disease Models, Animal , Encephalopathy, Bovine Spongiform/pathology , Humans , Mice , Prion Diseases/transmission , Prion Proteins/metabolism , Prions/pathogenicity , Scrapie/pathology
20.
Child Obes ; 14(4): 227-236, 2018.
Article in English | MEDLINE | ID: mdl-29889565

ABSTRACT

Food addiction has become a popular notion in the media and scientific community, with many proposing that an addiction to food causes obesity. An article published in this journal in December 2017 by Tompkins et al. asserted that food addiction poses a barrier to the treatment of adolescent obesity. This review questions some of the methods, results, and perspectives offered by Tompkins et al. It also considers the extant evidence overall about this construct. The data about food addiction provide minimal support for the discriminant validity of this conceptualization relative to depressed binge eating. We believe that the evidence suggests that the potential harm outweighs the benefits of encouraging researchers, clinicians, and especially obese people to view food addiction as a cause of obesity or a barrier to effective weight management. Ultimately, this review concludes that the construct of food addiction does not serve the interests of those striving to lose weight permanently (i.e., weight controllers) nearly as well as an alternative perspective: weight controllers are athletes, not food addicts.


Subject(s)
Food Addiction , Obesity , Adolescent , Athletes , Behavior, Addictive , Binge-Eating Disorder , Body Weight , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...