119 results on '"Bennett FC"'
Search Results
2. FOXO-regulated transcription restricts overgrowth of Tsc mutant organs
- Author
-
Harvey, KF, Mattila, J, Sofer, A, Bennett, FC, Ramsey, MR, Ellisen, LW, Puig, O, Hariharan, IK, Harvey, KF, Mattila, J, Sofer, A, Bennett, FC, Ramsey, MR, Ellisen, LW, Puig, O, and Hariharan, IK
- Abstract
FOXO is thought to function as a repressor of growth that is, in turn, inhibited by insulin signaling. However, inactivating mutations in Drosophila melanogaster FOXO result in viable flies of normal size, which raises a question over the involvement of FOXO in growth regulation. Previously, a growth-suppressive role for FOXO under conditions of increased target of rapamycin (TOR) pathway activity was described. Here, we further characterize this phenomenon. We show that tuberous sclerosis complex 1 mutations cause increased FOXO levels, resulting in elevated expression of FOXO-regulated genes, some of which are known to antagonize growth-promoting pathways. Analogous transcriptional changes are observed in mammalian cells, which implies that FOXO attenuates TOR-driven growth in diverse species.
- Published
- 2008
3. Letters to the Editor
- Author
-
Mark L. Wolraich and Bennett Fc
- Subjects
Psychiatry and Mental health ,Behavioral pediatrics ,Pediatrics, Perinatology and Child Health ,Developmental and Educational Psychology ,Psychology ,Clinical psychology - Published
- 2003
- Full Text
- View/download PDF
4. LOW BIRTH WEIGHT
- Author
-
Jaction Jc, Bennett Fc, and La Pine Tr
- Subjects
Psychiatry and Mental health ,medicine.medical_specialty ,Low birth weight ,Obstetrics ,business.industry ,Birth weight ,Pediatrics, Perinatology and Child Health ,Developmental and Educational Psychology ,medicine ,medicine.symptom ,business - Published
- 1996
- Full Text
- View/download PDF
5. Cerebral palsy grown up.
- Author
-
Glew GM and Bennett FC
- Published
- 2011
- Full Text
- View/download PDF
6. Diet and female sex hormone concentrations: an intervention study for the type of fat consumed
- Author
-
Bennett, FC, primary and Ingram, DM, additional
- Published
- 1990
- Full Text
- View/download PDF
7. Results at age 8 years of early intervention for low-birth-weight premature infants. The Infant Health and Development Program.
- Author
-
McCarton CM, Brooks-Gunn J, Wallace IF, Bauer CR, Bennett FC, Bernbaum JC, Broyles RS, Casey PH, McCormick MC, Scott DT, Tyson J, Tonascia J, Meinert CL, Infant Health and Development Program Research Group, McCarton, C M, Brooks-Gunn, J, Wallace, I F, Bauer, C R, Bennett, F C, and Bernbaum, J C
- Abstract
Objective: To reevaluate at age 8 years children who had participated during the first 3 years of life in a randomized clinical trial of special services for low-birthweight (LBW) premature infants.Design: Follow-up of a randomized controlled trial of premature infants (< or = 37 weeks' gestation), stratified by 2 LBW groups (lighter [< or = 2000 g] and heavier [2001-2500 g]) and divided into intervention (n=377) and follow-up only (n=608) groups.Setting: Eight sites serving diverse populations.Participants: At age 8 years, 874 children were assessed: 336 in the intervention group and 538 in the follow-up only group.Intervention: The 3-year intervention consisted of home visits (birth to 3 years), child development center services (ages 1 to 3 years), and parent group meetings (ages 1 to 3 years).Primary Outcome Measures: Cognitive functioning (Weschler Intelligence Scale for Children-III; Peabody Picture Vocabulary Test-Revised); academic achievement (Woodcock-Johnson Tests of Achievement-Revised); and parental reports of school performance, behavior (Child Behavior Checklist), and health (Child General Health Survey).Results: At age 8 years, in the entire cohort and in the lighter LBW stratum, the intervention and follow-up only groups were similar on all primary outcome measures. Differences favoring the intervention group were found within the heavier LBW group: full-scale IQ score (4.4 points higher, P=.007), verbal IQ score (4.2 points higher, P=.01), performance IQ score (3.9 points higher, P=.02), mathematics achievement score (4.8 points higher, P=.04), and receptive vocabulary score (6.7 points higher, P=.001). On a physical functioning subscale, the whole intervention group received less favorable ratings, while the lighter LBW intervention group had lower maternal ratings assessing social limitations caused by behavior.Conclusion: Although at age 8 years there were modest intervention-related differences in the cognitive and academic skills of heavier LBW premature children, attenuation of the large favorable effects seen at 3 years was observed in both the heavier and lighter LBW groups. This indicates a need to develop additional intervention strategies for LBW premature children that can provide sustained benefits. [ABSTRACT FROM AUTHOR]- Published
- 1997
- Full Text
- View/download PDF
8. Parents' perceptions of helpful vs unhelpful types of support in managing the care of preadolescents with chronic conditions.
- Author
-
Garwick AW, Patterson JM, Bennett FC, and Blum RW
- Published
- 1998
- Full Text
- View/download PDF
9. Breaking the news: how families first learn about their child's chronic condition.
- Author
-
Garwick AW, Patterson J, Bennett FC, and Blum RW
- Published
- 1995
- Full Text
- View/download PDF
10. Outcomes of infants weighing less than 800 grams at birth: 15 years' experience.
- Author
-
La Pine TR, Jackson JC, and Bennett FC
- Published
- 1995
11. Diagnosing cerebral palsy -- the earlier the better.
- Author
-
Bennett FC
- Abstract
Recognizing cerebral palsy in the first year of life has substantial benefits for both the infant and the family. Here are the risk factors and motor signs to look for. [ABSTRACT FROM AUTHOR]
- Published
- 1999
12. Effects of electronic fetal-heart-rate monitoring, as compared with periodic auscultation, on the neurologic development of premature infants.
- Author
-
Shy KK, Luthy DA, Bennett FC, Whitfield M, Larson EB, van Belle G, Hughes JP, Wilson JA, and Stenchever MA
- Published
- 1990
13. Suppression of Huntington's Disease Somatic Instability by Transcriptional Repression and Direct CAG Repeat Binding.
- Author
-
Mathews EW, Coffey SR, Gärtner A, Belgrad J, Bragg RM, O'Reilly D, Cantle JP, McHugh C, Summers A, Fentz J, Schwagarus T, Cornelius A, Lingos I, Burch Z, Kovalenko M, Andrew MA, Bennett FC, Kordasiewicz HB, Marchionini DM, Wilkinson H, Vogt TF, Pinto RM, Khvorova A, Howland D, Wheeler VC, and Carroll JB
- Abstract
Huntington's disease (HD) arises from a CAG expansion in the huntingtin ( HTT ) gene beyond a critical threshold. A major thrust of current HD therapeutic development is lowering levels of mutant HTT mRNA (m HTT ) and protein (mHTT) with the aim of reducing the toxicity of these product(s). Human genetic data also support a key role for somatic instability (SI) in HTT 's CAG repeat - whereby it lengthens with age in specific somatic cell types - as a key driver of age of motor dysfunction onset. Thus, an attractive HD therapy would address both mHTT toxicity and SI, but to date the relationship between SI and HTT lowering remains unexplored. Here, we investigated multiple therapeutically-relevant HTT-lowering modalities to establish the relationship between HTT lowering and SI in HD knock-in mice. We find that repressing transcription of mutant Htt (m Htt ) provides robust protection from SI, using diverse genetic and pharmacological approaches (antisense oligonucleotides, CRISPR-Cas9 genome editing, the Lac repressor, and virally delivered zinc finger transcriptional repressor proteins, ZFPs). However, we find that small interfering RNA (siRNA), a potent HTT-lowering treatment, lowers HTT levels without influencing SI and that SI is also normal in mice lacking 50% of total HTT levels, suggesting HTT levels, per se , do not modulate SI in trans . Remarkably, modified ZFPs that bind the m Htt locus, but lack a repressive domain, robustly protect from SI, despite not reducing HTT mRNA or protein levels. These results have important therapeutic implications in HD, as they suggest that DNA-targeted HTT-lowering treatments may have significant advantages compared to other HTT-lowering approaches, and that interaction of a DNA-binding protein and HTT' s CAG repeats may provide protection from SI while sparing HTT expression.
- Published
- 2024
- Full Text
- View/download PDF
14. Microglia cannibalism during neurodevelopment results in necroptotic cell death.
- Author
-
Bennett FC and Bennett ML
- Subjects
- Animals, Humans, Cell Death physiology, Necroptosis physiology, Phagocytosis physiology, Cytophagocytosis physiology, Mice, Microglia physiology, Microglia metabolism, Neurons physiology
- Abstract
Clearance of dying neurons by microglia is critical to healthy neurodevelopment, but what else do microglia eat? A new study in PLOS Biology demonstrates that microglia not only eat neurons but each other, and that this "microglia cannibalism" causes necroptotic cell death., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2024 Bennett, Bennett. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
15. Role of Microglia in Central Nervous System Development and Plasticity.
- Author
-
Schafer DP, Stevens B, Bennett ML, and Bennett FC
- Abstract
The nervous system comprises a remarkably diverse and complex network of cell types, which must communicate with one another with speed, reliability, and precision. Thus, the developmental patterning and maintenance of these cell populations and their connections with one another pose a rather formidable task. Emerging data implicate microglia, the resident myeloid-derived cells of the central nervous system (CNS), in spatial patterning and synaptic wiring throughout the healthy, developing, and adult CNS. Importantly, new tools to specifically manipulate microglia function have revealed that these cellular functions translate, on a systems level, to effects on overall behavior. In this review, we give a historical perspective of work to identify microglia function in the healthy CNS, and highlight exciting new discoveries about their contributions to CNS development, maintenance, and plasticity., (Copyright © 2024 Cold Spring Harbor Laboratory Press; all rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
16. Microglia replacement by ER-Hoxb8 conditionally immortalized macrophages provides insight into Aicardi-Goutières Syndrome neuropathology.
- Author
-
Nemec KM, Uy G, Chaluvadi VS, Purnell FS, Elfayoumi B, O'Brien CA, Aisenberg WH, Lombroso SI, Guo X, Blank N, Oon CH, Yaqoob F, Temsamrit B, Rawat P, Thaiss CA, Wang Q, Bennett ML, and Bennett FC
- Abstract
Microglia, the brain's resident macrophages, can be reconstituted by surrogate cells - a process termed "microglia replacement." To expand the microglia replacement toolkit, we here introduce estrogen-regulated (ER) homeobox B8 (Hoxb8) conditionally immortalized macrophages, a cell model for generation of immune cells from murine bone marrow, as a versatile model for microglia replacement. We find that ER-Hoxb8 macrophages are highly comparable to primary bone marrow-derived (BMD) macrophages in vitro, and, when transplanted into a microglia-free brain, engraft the parenchyma and differentiate into microglia-like cells. Furthermore, ER-Hoxb8 progenitors are readily transducible by virus and easily stored as stable, genetically manipulated cell lines. As a demonstration of this system's power for studying the effects of disease mutations on microglia in vivo, we created stable, Adar1 -mutated ER-Hoxb8 lines using CRISPR-Cas9 to study the intrinsic contribution of macrophages to Aicardi-Goutières Syndrome (AGS), an inherited interferonopathy that primarily affects the brain and immune system. We find that Adar1 knockout elicited interferon secretion and impaired macrophage production in vitro, while preventing brain macrophage engraftment in vivo - phenotypes that can be rescued with concurrent mutation of Ifih1 (MDA5) in vitro, but not in vivo. Lastly, we extended these findings by generating ER-Hoxb8 progenitors from mice harboring a patient-specific Adar1 mutation (D1113H). We demonstrated the ability of microglia-specific D1113H mutation to drive interferon production in vivo, suggesting microglia drive AGS neuropathology. In sum, we introduce the ER-Hoxb8 approach to model microglia replacement and use it to clarify macrophage contributions to AGS.
- Published
- 2024
- Full Text
- View/download PDF
17. Microglia are not necessary for maintenance of blood-brain barrier properties in health, but PLX5622 alters brain endothelial cholesterol metabolism.
- Author
-
Profaci CP, Harvey SS, Bajc K, Zhang TZ, Jeffrey DA, Zhang AZ, Nemec KM, Davtyan H, O'Brien CA, McKinsey GL, Longworth A, McMullen TP, Capocchi JK, Gonzalez JG, Lawson DA, Arnold TD, Davalos D, Blurton-Jones M, Dabertrand F, Bennett FC, and Daneman R
- Subjects
- Animals, Mice, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor antagonists & inhibitors, Mice, Inbred C57BL, Male, Organic Chemicals, Microglia metabolism, Microglia drug effects, Blood-Brain Barrier metabolism, Blood-Brain Barrier drug effects, Cholesterol metabolism, Endothelial Cells metabolism, Endothelial Cells drug effects, Brain metabolism
- Abstract
Microglia, the resident immune cells of the central nervous system, are intimately involved in the brain's most basic processes, from pruning neural synapses during development to preventing excessive neuronal activity throughout life. Studies have reported both helpful and harmful roles for microglia at the blood-brain barrier (BBB) in the context of disease. However, less is known about microglia-endothelial cell interactions in the healthy brain. To investigate the role of microglia at a healthy BBB, we used the colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 to deplete microglia and analyzed the BBB ultrastructure, permeability, and transcriptome. Interestingly, we found that, despite their direct contact with endothelial cells, microglia are not necessary for the maintenance of BBB structure, function, or gene expression in the healthy brain. However, we found that PLX5622 treatment alters brain endothelial cholesterol metabolism. This effect was independent from microglial depletion, suggesting that PLX5622 has off-target effects on brain vasculature., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. Author Correction: Senescent glia link mitochondrial dysfunction and lipid accumulation.
- Author
-
Byrns CN, Perlegos AE, Miller KN, Jin Z, Carranza FR, Manchandra P, Beveridge CH, Randolph CE, Chaluvadi VS, Zhang SL, Srinivasan AR, Bennett FC, Sehgal A, Adams PD, Chopra G, and Bonini NM
- Published
- 2024
- Full Text
- View/download PDF
19. Senescent glia link mitochondrial dysfunction and lipid accumulation.
- Author
-
Byrns CN, Perlegos AE, Miller KN, Jin Z, Carranza FR, Manchandra P, Beveridge CH, Randolph CE, Chaluvadi VS, Zhang SL, Srinivasan AR, Bennett FC, Sehgal A, Adams PD, Chopra G, and Bonini NM
- Subjects
- Animals, Female, Humans, Male, Fibroblasts metabolism, Fibroblasts pathology, Longevity, Neurons metabolism, Neurons pathology, Oxidative Stress, Transcription Factor AP-1 metabolism, Lipids, Inflammation metabolism, Inflammation pathology, Aging metabolism, Aging pathology, Brain metabolism, Brain pathology, Brain cytology, Cellular Senescence, Drosophila melanogaster metabolism, Drosophila melanogaster cytology, Lipid Metabolism, Mitochondria metabolism, Mitochondria pathology, Neuroglia metabolism, Neuroglia pathology
- Abstract
Senescence is a cellular state linked to ageing and age-onset disease across many mammalian species
1,2 . Acutely, senescent cells promote wound healing3,4 and prevent tumour formation5 ; but they are also pro-inflammatory, thus chronically exacerbate tissue decline. Whereas senescent cells are active targets for anti-ageing therapy6-11 , why these cells form in vivo, how they affect tissue ageing and the effect of their elimination remain unclear12,13 . Here we identify naturally occurring senescent glia in ageing Drosophila brains and decipher their origin and influence. Using Activator protein 1 (AP1) activity to screen for senescence14,15 , we determine that senescent glia can appear in response to neuronal mitochondrial dysfunction. In turn, senescent glia promote lipid accumulation in non-senescent glia; similar effects are seen in senescent human fibroblasts in culture. Targeting AP1 activity in senescent glia mitigates senescence biomarkers, extends fly lifespan and health span, and prevents lipid accumulation. However, these benefits come at the cost of increased oxidative damage in the brain, and neuronal mitochondrial function remains poor. Altogether, our results map the trajectory of naturally occurring senescent glia in vivo and indicate that these cells link key ageing phenomena: mitochondrial dysfunction and lipid accumulation., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
20. Cross-talk between B cells, microglia and macrophages, and implications to central nervous system compartmentalized inflammation and progressive multiple sclerosis.
- Author
-
Touil H, Li R, Zuroff L, Moore CS, Healy L, Cignarella F, Piccio L, Ludwin S, Prat A, Gommerman J, Bennett FC, Jacobs D, Benjamins JA, Lisak RP, Antel JP, and Bar-Or A
- Abstract
Background: B cells can be enriched within meningeal immune-cell aggregates of multiple sclerosis (MS) patients, adjacent to subpial cortical demyelinating lesions now recognized as important contributors to progressive disease. This subpial demyelination is notable for a 'surface-in' gradient of neuronal loss and microglial activation, potentially reflecting the effects of soluble factors secreted into the CSF. We previously demonstrated that MS B-cell secreted products are toxic to oligodendrocytes and neurons. The potential for B-cell-myeloid cell interactions to propagate progressive MS is of considerable interest., Methods: Secreted products of MS-implicated pro-inflammatory effector B cells or IL-10-expressing B cells with regulatory potential were applied to human brain-derived microglia or monocyte-derived macrophages, with subsequent assessment of myeloid phenotype and function through measurement of their expression of pro-inflammatory, anti-inflammatory and homeostatic/quiescent molecules, and phagocytosis (using flow cytometry, ELISA and fluorescently-labeled myelin). Effects of secreted products of differentially activated microglia on B-cell survival and activation were further studied., Findings: Secreted products of MS-implicated pro-inflammatory B cells (but not IL-10 expressing B cells) substantially induce pro-inflammatory cytokine (IL-12, IL-6, TNFα) expression by both human microglia and macrophage (in a GM-CSF dependent manner), while down-regulating their expression of IL-10 and of quiescence-associated molecules, and suppressing their myelin phagocytosis. In contrast, secreted products of IL-10 expressing B cells upregulate both human microglia and macrophage expression of quiescence-associated molecules and enhance their myelin phagocytosis. Secreted factors from pro-inflammatory microglia enhance B-cell activation., Interpretation: Potential cross-talk between disease-relevant human B-cell subsets and both resident CNS microglia and infiltrating macrophages may propagate CNS-compartmentalized inflammation and injury associated with MS disease progression. These interaction represents an attractive therapeutic target for agents such as Bruton's tyrosine kinase inhibitors (BTKi) that modulate responses of both B cells and myeloid cells., Funding: Stated in Acknowledgments section of manuscript., Competing Interests: Declaration of interests H.T. and L.Z. received consulting fees from EMD Serono outside of the submitted work. D.J. has received consulting fees from Biogen, Genentech, Novartis, EMD Serono, Banner Life Sciences, Bristol Meyers Squibb, Horizon, Cycle and Sanofi Genzyme outside of the submitted work. A.BO. has received fees for advisory board participation and/or consulting from Abata, Accure, Atara Biotherapeutics, Biogen, BMS/Celgene/Receptos, GlaxoSmithKline, Gossamer, Immunic, Janssen/Actelion, Medimmune, Merck/EMD Serono, Novartis, Roche/Genentech, Sangamo, Sanofi-Genzyme, Viracta; and has received grant support to the University of Pennsylvania from Biogen Idec, Roche/Genentech, Merck/EMD Serono and Novartis. L.P. has recived grant support from Alector on projects not related to this work., (Copyright © 2023 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
21. CRISPR generation of CSF1R-G795A human microglia for robust microglia replacement in a chimeric mouse model.
- Author
-
Chadarevian JP, Davtyan H, Lombroso SI, Bennett FC, and Blurton-Jones M
- Subjects
- Adult, Animals, Mice, Infant, Newborn, Humans, Brain, Disease Models, Animal, Point Mutation, Microglia, Pluripotent Stem Cells
- Abstract
Chimeric mouse models have recently been developed to study human microglia in vivo. However, widespread engraftment of donor microglia within the adult brain has been challenging. Here, we present a protocol to introduce the G795A point mutation using CRISPR-Cas9 into the CSF1R locus of human pluripotent stem cells. We also describe an optimized microglial differentiation technique for transplantation into newborn or adult recipients. We then detail pharmacological paradigms to achieve widespread and near-complete engraftment of human microglia. For complete details on the use and execution of this protocol, please refer to Chadarevian et al. (2023).
1 ., Competing Interests: Declaration of interests M.B.-J., J.P.C., and H.D. are co-inventors on a pending patent filed by the University of California Regents (application 63/169,578) related to genetic modification of cells to confer resistance to CSF1R antagonists. M.B.-J. is a co-inventor of patent WO/2018/160496, related to the differentiation of human pluripotent stem cells into microglia. M.B.-J. is a co-founder of NovoGlia Inc. F.C.B. is a co-inventor on a pending patent filed by The Board of Trustees of The Leland Stanford Junior University (application 16/566,675) related to methods of microglia replacement., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
22. Human OPRM1 and murine Oprm1 promoter driven viral constructs for genetic access to μ-opioidergic cell types.
- Author
-
Salimando GJ, Tremblay S, Kimmey BA, Li J, Rogers SA, Wojick JA, McCall NM, Wooldridge LM, Rodrigues A, Borner T, Gardiner KL, Jayakar SS, Singeç I, Woolf CJ, Hayes MR, De Jonghe BC, Bennett FC, Bennett ML, Blendy JA, Platt ML, Creasy KT, Renthal WR, Ramakrishnan C, Deisseroth K, and Corder G
- Subjects
- Animals, Humans, Mice, Rats, Macaca, Receptors, Opioid, Receptors, Opioid, mu genetics, Transgenes, Analgesia, Chronic Pain, Induced Pluripotent Stem Cells
- Abstract
With concurrent global epidemics of chronic pain and opioid use disorders, there is a critical need to identify, target and manipulate specific cell populations expressing the mu-opioid receptor (MOR). However, available tools and transgenic models for gaining long-term genetic access to MOR+ neural cell types and circuits involved in modulating pain, analgesia and addiction across species are limited. To address this, we developed a catalog of MOR promoter (MORp) based constructs packaged into adeno-associated viral vectors that drive transgene expression in MOR+ cells. MORp constructs designed from promoter regions upstream of the mouse Oprm1 gene (mMORp) were validated for transduction efficiency and selectivity in endogenous MOR+ neurons in the brain, spinal cord, and periphery of mice, with additional studies revealing robust expression in rats, shrews, and human induced pluripotent stem cell (iPSC)-derived nociceptors. The use of mMORp for in vivo fiber photometry, behavioral chemogenetics, and intersectional genetic strategies is also demonstrated. Lastly, a human designed MORp (hMORp) efficiently transduced macaque cortical OPRM1+ cells. Together, our MORp toolkit provides researchers cell type specific genetic access to target and functionally manipulate mu-opioidergic neurons across a range of vertebrate species and translational models for pain, addiction, and neuropsychiatric disorders., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
23. Ionizable Lipid Nanoparticles for Therapeutic Base Editing of Congenital Brain Disease.
- Author
-
Palanki R, Bose SK, Dave A, White BM, Berkowitz C, Luks V, Yaqoob F, Han E, Swingle KL, Menon P, Hodgson E, Biswas A, Billingsley MM, Li L, Yiping F, Carpenter M, Trokhan A, Yeo J, Johana N, Wan TY, Alameh MG, Bennett FC, Storm PB, Jain R, Chan J, Weissman D, Mitchell MJ, and Peranteau WH
- Subjects
- Mice, Humans, Animals, Gene Editing, Lipids, Liposomes, RNA, Messenger genetics, RNA, Small Interfering genetics, Nanoparticles, Brain Diseases
- Abstract
Delivery of mRNA-based therapeutics to the perinatal brain holds great potential in treating congenital brain diseases. However, nonviral delivery platforms that facilitate nucleic acid delivery in this environment have yet to be rigorously studied. Here, we screen a diverse library of ionizable lipid nanoparticles (LNPs) via intracerebroventricular (ICV) injection in both fetal and neonatal mice and identify an LNP formulation with greater functional mRNA delivery in the perinatal brain than an FDA-approved industry standard LNP. Following in vitro optimization of the top-performing LNP (C3 LNP) for codelivery of an adenine base editing platform, we improve the biochemical phenotype of a lysosomal storage disease in the neonatal mouse brain, exhibit proof-of-principle mRNA brain transfection in vivo in a fetal nonhuman primate model, and demonstrate the translational potential of C3 LNPs ex vivo in human patient-derived brain tissues. These LNPs may provide a clinically translatable platform for in utero and postnatal mRNA therapies including gene editing in the brain.
- Published
- 2023
- Full Text
- View/download PDF
24. The enteric nervous system relays psychological stress to intestinal inflammation.
- Author
-
Schneider KM, Blank N, Alvarez Y, Thum K, Lundgren P, Litichevskiy L, Sleeman M, Bahnsen K, Kim J, Kardo S, Patel S, Dohnalová L, Uhr GT, Descamps HC, Kircher S, McSween AM, Ardabili AR, Nemec KM, Jimenez MT, Glotfelty LG, Eisenberg JD, Furth EE, Henao-Mejia J, Bennett FC, Pierik MJ, Romberg-Camps M, Mujagic Z, Prinz M, Schneider CV, Wherry EJ, Bewtra M, Heuckeroth RO, Levy M, and Thaiss CA
- Subjects
- Humans, Glucocorticoids pharmacology, Inflammation, Stress, Psychological, Enteric Nervous System physiology, Inflammatory Bowel Diseases
- Abstract
Mental health profoundly impacts inflammatory responses in the body. This is particularly apparent in inflammatory bowel disease (IBD), in which psychological stress is associated with exacerbated disease flares. Here, we discover a critical role for the enteric nervous system (ENS) in mediating the aggravating effect of chronic stress on intestinal inflammation. We find that chronically elevated levels of glucocorticoids drive the generation of an inflammatory subset of enteric glia that promotes monocyte- and TNF-mediated inflammation via CSF1. Additionally, glucocorticoids cause transcriptional immaturity in enteric neurons, acetylcholine deficiency, and dysmotility via TGF-β2. We verify the connection between the psychological state, intestinal inflammation, and dysmotility in three cohorts of IBD patients. Together, these findings offer a mechanistic explanation for the impact of the brain on peripheral inflammation, define the ENS as a relay between psychological stress and gut inflammation, and suggest that stress management could serve as a valuable component of IBD care., Competing Interests: Declaration of interests E.J.W. is an advisor for Danger Bio, Janssen, New Limit, Marengo, Pluto Immunotherapeutics Related Sciences, Rubius Therapeutics, Santa Ana Bio, Synthekine, and Surface Oncology. E.J.W. is a founder of and holds stock in Surface Oncology, Danger Bio, and Arsenal Biosciences., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
25. Molecular and metabolic heterogeneity of astrocytes and microglia.
- Author
-
Hasel P, Aisenberg WH, Bennett FC, and Liddelow SA
- Subjects
- Humans, Astrocytes metabolism, Brain metabolism, Microglia metabolism, Neurodegenerative Diseases metabolism
- Abstract
Astrocytes and microglia are central players in a myriad of processes in the healthy and diseased brain, ranging from metabolism to immunity. The crosstalk between these two cell types contributes to pathology in many if not all neuroinflammatory and neurodegenerative diseases. Recent advancements in integrative multimodal sequencing techniques have begun to highlight how heterogeneous both cell types are and the importance of metabolism to their regulation. We discuss here the transcriptomic, metabolic, and functional heterogeneity of astrocytes and microglia and highlight their interaction in health and disease., Competing Interests: Declaration of interests S.A.L. maintains a financial interest in AstronauTx Ltd. and sits on the Scientific Advisory Board of The BioAccess Fund. F.C.B is a consultant for Glia Biotherapeutics, Inc., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
26. Engineering an inhibitor-resistant human CSF1R variant for microglia replacement.
- Author
-
Chadarevian JP, Lombroso SI, Peet GC, Hasselmann J, Tu C, Marzan DE, Capocchi J, Purnell FS, Nemec KM, Lahian A, Escobar A, England W, Chaluvadi S, O'Brien CA, Yaqoob F, Aisenberg WH, Porras-Paniagua M, Bennett ML, Davtyan H, Spitale RC, Blurton-Jones M, and Bennett FC
- Subjects
- Animals, Humans, Mice, Aminopyridines pharmacology, Brain metabolism, Cell- and Tissue-Based Therapy methods, Microglia metabolism, Protein Engineering methods, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor genetics, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism
- Abstract
Hematopoietic stem cell transplantation (HSCT) can replace endogenous microglia with circulation-derived macrophages but has high mortality. To mitigate the risks of HSCT and expand the potential for microglia replacement, we engineered an inhibitor-resistant CSF1R that enables robust microglia replacement. A glycine to alanine substitution at position 795 of human CSF1R (G795A) confers resistance to multiple CSF1R inhibitors, including PLX3397 and PLX5622. Biochemical and cell-based assays show no discernable gain or loss of function. G795A- but not wildtype-CSF1R expressing macrophages efficiently engraft the brain of PLX3397-treated mice and persist after cessation of inhibitor treatment. To gauge translational potential, we CRISPR engineered human-induced pluripotent stem cell-derived microglia (iMG) to express G795A. Xenotransplantation studies demonstrate that G795A-iMG exhibit nearly identical gene expression to wildtype iMG, respond to inflammatory stimuli, and progressively expand in the presence of PLX3397, replacing endogenous microglia to fully occupy the brain. In sum, we engineered a human CSF1R variant that enables nontoxic, cell type, and tissue-specific replacement of microglia., (© 2022 Chadarevian et al.)
- Published
- 2023
- Full Text
- View/download PDF
27. Perivascular signals drive microglial appetite.
- Author
-
O'Brien CA and Bennett FC
- Subjects
- Microglia, Appetite
- Published
- 2023
- Full Text
- View/download PDF
28. Treatment of a genetic brain disease by CNS-wide microglia replacement.
- Author
-
Shibuya Y, Kumar KK, Mader MM, Yoo Y, Ayala LA, Zhou M, Mohr MA, Neumayer G, Kumar I, Yamamoto R, Marcoux P, Liou B, Bennett FC, Nakauchi H, Sun Y, Chen X, Heppner FL, Wyss-Coray T, Südhof TC, and Wernig M
- Subjects
- Animals, Bone Marrow Cells, Brain, Central Nervous System, Mice, Microglia, Brain Diseases, Hematopoietic Stem Cell Transplantation
- Abstract
Hematopoietic cell transplantation after myeloablative conditioning has been used to treat various genetic metabolic syndromes but is largely ineffective in diseases affecting the brain presumably due to poor and variable myeloid cell incorporation into the central nervous system. Here, we developed and characterized a near-complete and homogeneous replacement of microglia with bone marrow cells in mice without the need for genetic manipulation of donor or host. The high chimerism resulted from a competitive advantage of scarce donor cells during microglia repopulation rather than enhanced recruitment from the periphery. Hematopoietic stem cells, but not immediate myeloid or monocyte progenitor cells, contained full microglia replacement potency equivalent to whole bone marrow. To explore its therapeutic potential, we applied microglia replacement to a mouse model for Prosaposin deficiency, which is characterized by a progressive neurodegeneration phenotype. We found a reduction of cerebellar neurodegeneration and gliosis in treated brains, improvement of motor and balance impairment, and life span extension even with treatment started in young adulthood. This proof-of-concept study suggests that efficient microglia replacement may have therapeutic efficacy for a variety of neurological diseases.
- Published
- 2022
- Full Text
- View/download PDF
29. Microglia in antiviral immunity of the brain and spinal cord.
- Author
-
O'Brien CA, Bennett FC, and Bennett ML
- Subjects
- Humans, Brain, Spinal Cord, Central Nervous System, Antiviral Agents, Microglia
- Abstract
Viral infections of the central nervous system (CNS) are a significant cause of neurological impairment and mortality worldwide. As tissue resident macrophages, microglia are critical initial responders to CNS viral infection. Microglia seem to coordinate brain-wide antiviral responses of both brain resident cells and infiltrating immune cells. This review discusses how microglia may promote this antiviral response at a molecular level, from potential mechanisms of virus recognition to downstream cytokine responses and interaction with antiviral T cells. Recent advancements in genetic tools to specifically target microglia in vivo promise to further our understanding about the precise mechanistic role of microglia in CNS infection., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
30. Glia in neurodegeneration.
- Author
-
Bennett FC and Sloan SA
- Subjects
- Animals, Humans, Nerve Degeneration, Neurodegenerative Diseases, Neuroglia
- Published
- 2021
- Full Text
- View/download PDF
31. Please eat (only part) of me: synaptic phosphatidylserine cues microglia to feast: Two new studies identify how a common apoptotic cell flag is used to sculpt neural circuits.
- Author
-
Peet G, Bennett FC, and Bennett ML
- Subjects
- Animals, Apoptosis, Cues, Mice, Protein Isoforms, Receptors, G-Protein-Coupled, Synapses, Microglia, Phosphatidylserines
- Abstract
Microglia, the brain's tissue-resident macrophages, contribute to the developmental elimination of extranumerary synapses and to pathologic synapse loss in mouse models of neurodegeneration. Two papers published in The EMBO Journal reveal that phosphatidylserine (PS) is a neuronal cue for microglial synapse elimination., (© 2020 The Authors.)
- Published
- 2020
- Full Text
- View/download PDF
32. How Support of Early Career Researchers Can Reset Science in the Post-COVID19 World.
- Author
-
Gibson EM, Bennett FC, Gillespie SM, Güler AD, Gutmann DH, Halpern CH, Kucenas SC, Kushida CA, Lemieux M, Liddelow S, Macauley SL, Li Q, Quinn MA, Roberts LW, Saligrama N, Taylor KR, Venkatesh HS, Yalçın B, and Zuchero JB
- Subjects
- Achievement, Biomedical Research, Humans, Research Personnel education, Science education, Science trends, Universities, Career Mobility, Research trends, Research Personnel trends
- Abstract
The COVID19 crisis has magnified the issues plaguing academic science, but it has also provided the scientific establishment with an unprecedented opportunity to reset. Shoring up the foundation of academic science will require a concerted effort between funding agencies, universities, and the public to rethink how we support scientists, with a special emphasis on early career researchers., Competing Interests: Declaration of Interests Dr. Roberts serves as Editor-in-Chief of books for the American Psychiatric Association Publishing Division and as Editor-in-Chief of the journal Academic Medicine. Unrelated to this publication, Dr. Roberts serves as an advisor for the Bucksbaum Institute of the University of Chicago Pritzker School of Medicine and owns the small business Terra Nova Learning Systems., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
33. Neuroinflammation and EIF2 Signaling Persist despite Antiretroviral Treatment in an hiPSC Tri-culture Model of HIV Infection.
- Author
-
Ryan SK, Gonzalez MV, Garifallou JP, Bennett FC, Williams KS, Sotuyo NP, Mironets E, Cook K, Hakonarson H, Anderson SA, and Jordan-Sciutto KL
- Published
- 2020
- Full Text
- View/download PDF
34. Immune cell regulation of glia during CNS injury and disease.
- Author
-
Greenhalgh AD, David S, and Bennett FC
- Subjects
- Animals, Astrocytes immunology, Humans, Lymphocytes immunology, Macrophages immunology, Microglia immunology, Neutrophils immunology, Oligodendroglia immunology, Brain immunology, Central Nervous System Diseases immunology, Immune System, Neuroglia immunology
- Abstract
Glial cells are abundant in the CNS and are essential for brain development and homeostasis. These cells also regulate tissue recovery after injury and their dysfunction is a possible contributing factor to neurodegenerative and psychiatric disease. Recent evidence suggests that microglia, which are also the brain's major resident immune cells, provide disease-modifying regulation of the other major glial populations, namely astrocytes and oligodendrocytes. In addition, peripheral immune cells entering the CNS after injury and in disease may directly affect microglial, astrocyte and oligodendrocyte function, suggesting an integrated network of immune cell-glial cell communication.
- Published
- 2020
- Full Text
- View/download PDF
35. The influence of environment and origin on brain resident macrophages and implications for therapy.
- Author
-
Bennett ML and Bennett FC
- Subjects
- Animals, Cell Lineage physiology, Humans, Macrophages cytology, Brain cytology, Microglia cytology
- Abstract
Microglia are the tissue-resident macrophages of the brain and spinal cord. They are critical players in the development, normal function, and decline of the CNS. Unlike traditional monocyte-derived macrophages, microglia originate from primitive hematopoiesis in the embryonic yolk sac and self-renew throughout life. Microglia also have a unique genetic signature among tissue resident macrophages. Recent studies identify the contributions of both brain environment and developmental history to the transcriptomic identity of microglia. Here we review this emerging literature and discuss the potential implications of origin on microglial function, with particular focus on existing and future therapies using bone-marrow- or stem-cell-derived cells for the treatment of neurological diseases.
- Published
- 2020
- Full Text
- View/download PDF
36. Microglia Metabolic Breakdown Drives Alzheimer's Pathology.
- Author
-
Bennett FC and Liddelow SA
- Subjects
- Amyloid beta-Peptides, Brain, Humans, Microglia, Alzheimer Disease
- Abstract
Altered metabolic function is common in stressed immune cells, but alteration in brain microglia during neurodegeneration is not understood. In this issue, Baik et al. (2019) provide insight into microglial metabolism. They demonstrate a switch from oxidative phosphorylation to glycolysis following interaction with amyloid beta acutely, and breakdown in both pathways chronically., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
37. The immune system and psychiatric disease: a basic science perspective.
- Author
-
Bennett FC and Molofsky AV
- Subjects
- Animals, Behavior, Animal, Disease Models, Animal, Humans, Brain immunology, Brain pathology, Immune System pathology, Mental Disorders immunology, Mental Disorders pathology, Neurons immunology, Neurons pathology
- Abstract
Mental illness exerts a major burden on human health, yet evidence-based treatments are rudimentary due to a limited understanding of the underlying pathologies. Clinical studies point to roles for the immune system in psychiatric diseases, while basic science has revealed that the brain has an active and multi-cellular resident immune system that interacts with peripheral immunity and impacts behavior. In this perspective, we highlight evidence of immune involvement in human psychiatric disease and review data from animal models that link immune signaling to neuronal function and behavior. We propose a conceptual framework for linking advances in basic neuroimmunology to their potential relevance for psychiatric diseases, based on the subtypes of immune responses defined in peripheral tissues. Our goal is to identify novel areas of focus for future basic and translational studies that may reveal the potential of the immune system for diagnosing and treating mental illnesses., (© 2019 British Society for Immunology.)
- Published
- 2019
- Full Text
- View/download PDF
38. Spatiotemporal Tracking of Brain-Tumor-Associated Myeloid Cells in Vivo through Optical Coherence Tomography with Plasmonic Labeling and Speckle Modulation.
- Author
-
SoRelle ED, Yecies DW, Liba O, Bennett FC, Graef CM, Dutta R, Mitra S, Joubert LM, Cheshier S, Grant GA, and de la Zerda A
- Subjects
- Animals, Cell Line, Tumor, Contrast Media chemical synthesis, Disease Models, Animal, Female, Humans, Mice, Mice, Nude, Neoplasms, Experimental diagnostic imaging, Particle Size, Surface Properties, Brain Neoplasms diagnostic imaging, Cell Tracking, Contrast Media chemistry, Imaging, Three-Dimensional, Myeloid Cells pathology, Tomography, Optical Coherence
- Abstract
By their nature, tumors pose a set of profound challenges to the immune system with respect to cellular recognition and response coordination. Recent research indicates that leukocyte subpopulations, especially tumor-associated macrophages (TAMs), can exert substantial influence on the efficacy of various cancer immunotherapy treatment strategies. To better study and understand the roles of TAMs in determining immunotherapeutic outcomes, significant technical challenges associated with dynamically monitoring single cells of interest in relevant live animal models of solid tumors must be overcome. However, imaging techniques with the requisite combination of spatiotemporal resolution, cell-specific contrast, and sufficient signal-to-noise at increasing depths in tissue are exceedingly limited. Here we describe a method to enable high-resolution, wide-field, longitudinal imaging of TAMs based on speckle-modulating optical coherence tomography (SM-OCT) and spectral scattering from an optimized contrast agent. The approach's improvements to OCT detection sensitivity and noise reduction enabled high-resolution OCT-based observation of individual cells of a specific host lineage in live animals. We found that large gold nanorods (LGNRs) that exhibit a narrow-band, enhanced scattering cross-section can selectively label TAMs and activate microglia in an in vivo orthotopic murine model of glioblastoma multiforme. We demonstrated near real-time tracking of the migration of cells within these myeloid subpopulations. The intrinsic spatiotemporal resolution, imaging depth, and contrast sensitivity reported herein may facilitate detailed studies of the fundamental behaviors of TAMs and other leukocytes at the single-cell level in vivo , including intratumoral distribution heterogeneity and roles in modulating cancer proliferation.
- Published
- 2019
- Full Text
- View/download PDF
39. Isolation and Culture of Microglia.
- Author
-
Bohlen CJ, Bennett FC, and Bennett ML
- Subjects
- Animals, Mice, Rats, Cell Culture Techniques, Flow Cytometry, Microglia
- Abstract
Microglia represent 5-10% of cells in the central nervous system and contribute to the development, homeostasis, injury, and repair of neural tissues. As the tissue-resident macrophages of the central nervous system, microglia execute core innate immune functions such as detection of pathogens/damage, cytokine secretion, and phagocytosis. However, additional properties that are specific to microglia and their neural environment are beginning to be appreciated. This article describes approaches for purification of microglia by fluorescence-activated cell sorting using microglia-specific surface markers and for enrichment of microglia by magnetic sorting and immunopanning. Detailed information about culturing primary microglia at various developmental stages is also provided. Throughout, we focus on special considerations for handling microglia and compare the relative strengths or disadvantages of different protocols. © 2018 by John Wiley & Sons, Inc., (© 2018 John Wiley & Sons, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
40. A Combination of Ontogeny and CNS Environment Establishes Microglial Identity.
- Author
-
Bennett FC, Bennett ML, Yaqoob F, Mulinyawe SB, Grant GA, Hayden Gephart M, Plowey ED, and Barres BA
- Subjects
- Animals, Brain metabolism, Central Nervous System, Humans, Macrophages metabolism, Mice, Mice, Knockout, Microglia metabolism, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor, Brain cytology, Cell Lineage, Hematopoietic Stem Cells cytology, Macrophages cytology, Microglia cytology
- Abstract
Microglia, the brain's resident macrophages, are dynamic CNS custodians with surprising origins in the extra-embryonic yolk sac. The consequences of their distinct ontogeny are unknown but critical to understanding and treating brain diseases. We created a brain macrophage transplantation system to disentangle how environment and ontogeny specify microglial identity. We find that donor cells extensively engraft in the CNS of microglia-deficient mice, and even after exposure to a cell culture environment, microglia fully regain their identity when returned to the CNS. Though transplanted macrophages from multiple tissues can express microglial genes in the brain, only those of yolk-sac origin fully attain microglial identity. Transplanted macrophages of inappropriate origin, including primary human cells in a humanized host, express disease-associated genes and specific ontogeny markers. Through brain macrophage transplantation, we discover new principles of microglial identity that have broad applications to the study of disease and development of myeloid cell therapies., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
41. Diverse Requirements for Microglial Survival, Specification, and Function Revealed by Defined-Medium Cultures.
- Author
-
Bohlen CJ, Bennett FC, Tucker AF, Collins HY, Mulinyawe SB, and Barres BA
- Subjects
- Animals, Astrocytes metabolism, Cell Culture Techniques, Cholesterol metabolism, Culture Media, Conditioned metabolism, Humans, Interleukins metabolism, Macrophage Colony-Stimulating Factor metabolism, Mice, Microglia cytology, Microglia immunology, Microglia metabolism, Phagocytosis immunology, Rats, Serum, Transcriptome, Transforming Growth Factor beta metabolism, Transforming Growth Factor beta pharmacology, Transforming Growth Factor beta2 metabolism, Cell Survival drug effects, Cholesterol pharmacology, Interleukins pharmacology, Macrophage Colony-Stimulating Factor pharmacology, Microglia drug effects, Phagocytosis drug effects, Transforming Growth Factor beta2 pharmacology
- Abstract
Microglia, the resident macrophages of the CNS, engage in various CNS-specific functions that are critical for development and health. To better study microglia and the properties that distinguish them from other tissue macrophage populations, we have optimized serum-free culture conditions to permit robust survival of highly ramified adult microglia under defined-medium conditions. We find that astrocyte-derived factors prevent microglial death ex vivo and that this activity results from three primary components, CSF-1/IL-34, TGF-β2, and cholesterol. Using microglial cultures that have never been exposed to serum, we demonstrate a dramatic and lasting change in phagocytic capacity after serum exposure. Finally, we find that mature microglia rapidly lose signature gene expression after isolation, and that this loss can be reversed by engrafting cells back into an intact CNS environment. These data indicate that the specialized gene expression profile of mature microglia requires continuous instructive signaling from the intact CNS., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
42. Neurotoxic reactive astrocytes are induced by activated microglia.
- Author
-
Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, Bennett ML, Münch AE, Chung WS, Peterson TC, Wilton DK, Frouin A, Napier BA, Panicker N, Kumar M, Buckwalter MS, Rowitch DH, Dawson VL, Dawson TM, Stevens B, and Barres BA
- Subjects
- Animals, Astrocytes metabolism, Axotomy, Cell Culture Techniques, Cell Survival, Complement C1q metabolism, Disease Progression, Humans, Inflammation pathology, Interleukin-1alpha metabolism, Mice, Mice, Inbred C57BL, Microglia metabolism, Neurodegenerative Diseases pathology, Oligodendroglia pathology, Phagocytosis, Phenotype, Rats, Rats, Sprague-Dawley, Synapses pathology, Toxins, Biological metabolism, Tumor Necrosis Factor-alpha metabolism, Astrocytes classification, Astrocytes pathology, Cell Death, Central Nervous System pathology, Microglia pathology, Neurons pathology
- Abstract
Reactive astrocytes are strongly induced by central nervous system (CNS) injury and disease, but their role is poorly understood. Here we show that a subtype of reactive astrocytes, which we termed A1, is induced by classically activated neuroinflammatory microglia. We show that activated microglia induce A1 astrocytes by secreting Il-1α, TNF and C1q, and that these cytokines together are necessary and sufficient to induce A1 astrocytes. A1 astrocytes lose the ability to promote neuronal survival, outgrowth, synaptogenesis and phagocytosis, and induce the death of neurons and oligodendrocytes. Death of axotomized CNS neurons in vivo is prevented when the formation of A1 astrocytes is blocked. Finally, we show that A1 astrocytes are abundant in various human neurodegenerative diseases including Alzheimer's, Huntington's and Parkinson's disease, amyotrophic lateral sclerosis and multiple sclerosis. Taken together these findings help to explain why CNS neurons die after axotomy, strongly suggest that A1 astrocytes contribute to the death of neurons and oligodendrocytes in neurodegenerative disorders, and provide opportunities for the development of new treatments for these diseases.
- Published
- 2017
- Full Text
- View/download PDF
43. Pharmacokinetic and Pharmacodynamic Investigations of ION-353382, a Model Antisense Oligonucleotide: Using Alpha-2-Macroglobulin and Murinoglobulin Double-Knockout Mice.
- Author
-
Shemesh CS, Yu RZ, Gaus HJ, Seth PP, Swayze EE, Bennett FC, Geary RS, Henry SP, and Wang Y
- Subjects
- Animals, Enzyme-Linked Immunosorbent Assay, Gene Expression Regulation drug effects, Genetic Therapy, Hepatocytes drug effects, Hepatocytes metabolism, Humans, Liver drug effects, Liver metabolism, Mice, Mice, Knockout, Oligonucleotides, Antisense pharmacokinetics, Oligonucleotides, Antisense pharmacology, Pregnancy-Associated alpha 2-Macroglobulins antagonists & inhibitors, Scavenger Receptors, Class B antagonists & inhibitors, Serum Globulins antagonists & inhibitors, Oligonucleotides, Antisense genetics, Pregnancy-Associated alpha 2-Macroglobulins genetics, Scavenger Receptors, Class B genetics, Serum Globulins genetics
- Abstract
To investigate the pharmacokinetics (PKs) and pharmacodynamics (PDs) for ION-353382, an antisense oligonucleotide (ASO) targeting scavenger receptor class B type I (SRB1) mRNA, using alpha-2-macroglobulin (A2M), murinoglobulin double-knockout (DKO), and wild-type mice. Wild-type and DKO homozygous mice were administered a single subcutaneous injection of ION-353382 at 0, 5, 15, 30, and 60 mg/kg. Mice were sacrificed at 72 h with plasma and organs harvested. Both liquid chromatography-mass spectrometry (LC-MS) and enzyme-linked immunosorbent assay (ELISA) were used to determine ASO exposure with real-time PCR for SRB1 expression. Immunohistochemistry was evaluated to explore hepatic uptake of ASOs. The total plasma protein binding and profiling was assessed. Finally, two-dimensional gel electrophoresis identified protein expression differences. PK exposures were comparable between wild-type and DKO mice in plasma, liver, and kidney, yet a near twofold reduction in EC50 was revealed for DKO mice based on an inhibitory effect liver exposure response model. Total plasma protein binding and profiling revealed no major dissimilarities between both groups. Plasma proteome fingerprinting confirmed protein expression variations related to A2M. Histological examination revealed enhanced ASO distribution into hepatocytes and less nonparenchymal uptake for DKO mice compared to wild-type mice. Knocking out A2M showed improved PD activities without an effect on total plasma and tissue exposure kinetics. Binding to A2M could mediate ASOs to nonproductive compartments, and thus, decreased binding of ASOs to A2M could potentially improve ASO pharmacology.
- Published
- 2016
- Full Text
- View/download PDF
44. Neurodevelopmental and Cognitive Outcomes in Children With Intestinal Failure.
- Author
-
Chesley PM, Sanchez SE, Melzer L, Oron AP, Horslen SP, Bennett FC, and Javid PJ
- Subjects
- Child, Child, Preschool, Female, Humans, Infant, Infant, Extremely Low Birth Weight, Infant, Newborn, Intellectual Disability complications, Intellectual Disability psychology, Male, Prospective Studies, Psychometrics, Short Bowel Syndrome complications, Intellectual Disability physiopathology, Short Bowel Syndrome physiopathology, Survivors
- Abstract
Objectives: Recent advances in medical and surgical management have led to improved long-term survival in children with intestinal failure. Yet, limited data exist on their neurodevelopmental and cognitive outcomes. The aim of the present study was to measure neurodevelopmental outcomes in children with intestinal failure., Methods: Children enrolled in a regional intestinal failure program underwent prospective neurodevelopmental and psychometric evaluation using a validated scoring tool. Cognitive impairment was defined as a mental developmental index <70. Neurodevelopmental impairment was defined as cerebral palsy, visual or hearing impairment, or cognitive impairment. Univariate analyses were performed using the Wilcoxon rank-sum test. Data are presented as median (range)., Results: Fifteen children with a remnant bowel length of 18 (5-85) cm were studied at age 17 (12-67) months. Thirteen patients remained dependent on parenteral nutrition. Twelve (80%) subjects scored within the normal range on cognitive testing. Each child with cognitive impairment was noted to have additional risk factors independent of intestinal failure including cardiac arrest and extreme prematurity. On univariate analysis, cognitive impairment was associated with longer inpatient hospital stays, increased number of surgical procedures, and prematurity (P < 0.02). In total, 4 (27%) children demonstrated findings consistent with neurodevelopmental impairment., Conclusions: A majority of children with intestinal failure demonstrated normal neurodevelopmental and cognitive outcomes on psychometric testing. These data suggest that children with intestinal failure without significant comorbidity may be at low risk for long-term neurodevelopmental impairment.
- Published
- 2016
- Full Text
- View/download PDF
45. New tools for studying microglia in the mouse and human CNS.
- Author
-
Bennett ML, Bennett FC, Liddelow SA, Ajami B, Zamanian JL, Fernhoff NB, Mulinyawe SB, Bohlen CJ, Adil A, Tucker A, Weissman IL, Chang EF, Li G, Grant GA, Hayden Gephart MG, and Barres BA
- Subjects
- Aged, Animals, Antibodies, Monoclonal immunology, Biomarkers, Brain embryology, Brain growth & development, Cell Division, Cell Lineage, Child, Endotoxemia pathology, Gene Expression Profiling, Gene Expression Regulation, Developmental, Humans, Lipopolysaccharides toxicity, Macrophages chemistry, Membrane Proteins deficiency, Membrane Proteins genetics, Membrane Proteins immunology, Mice, Mice, Knockout, Microglia physiology, Middle Aged, Nerve Crush, Nerve Tissue Proteins genetics, Nerve Tissue Proteins immunology, Optic Nerve Injuries pathology, Organ Specificity, Rabbits, Sciatic Nerve injuries, Sciatic Nerve pathology, Sequence Analysis, RNA, Temporal Lobe metabolism, Transcriptome, Brain cytology, Membrane Proteins analysis, Microglia chemistry, Nerve Tissue Proteins analysis
- Abstract
The specific function of microglia, the tissue resident macrophages of the brain and spinal cord, has been difficult to ascertain because of a lack of tools to distinguish microglia from other immune cells, thereby limiting specific immunostaining, purification, and manipulation. Because of their unique developmental origins and predicted functions, the distinction of microglia from other myeloid cells is critically important for understanding brain development and disease; better tools would greatly facilitate studies of microglia function in the developing, adult, and injured CNS. Here, we identify transmembrane protein 119 (Tmem119), a cell-surface protein of unknown function, as a highly expressed microglia-specific marker in both mouse and human. We developed monoclonal antibodies to its intracellular and extracellular domains that enable the immunostaining of microglia in histological sections in healthy and diseased brains, as well as isolation of pure nonactivated microglia by FACS. Using our antibodies, we provide, to our knowledge, the first RNAseq profiles of highly pure mouse microglia during development and after an immune challenge. We used these to demonstrate that mouse microglia mature by the second postnatal week and to predict novel microglial functions. Together, we anticipate these resources will be valuable for the future study and understanding of microglia in health and disease.
- Published
- 2016
- Full Text
- View/download PDF
46. Regional brain activation during verbal declarative memory in metastatic breast cancer.
- Author
-
Kesler SR, Bennett FC, Mahaffey ML, and Spiegel D
- Subjects
- Brain physiopathology, Breast Neoplasms pathology, Female, Humans, Hydrocortisone metabolism, Magnetic Resonance Imaging, Memory, Middle Aged, Neoplasm Metastasis, Neuropsychological Tests, Verbal Behavior, Breast Neoplasms physiopathology, Breast Neoplasms psychology, Memory Disorders physiopathology
- Abstract
Purpose: To determine the neurofunctional basis of verbal memory dysfunction in women with metastatic breast cancer. This objective was based on previous research suggesting memory and other cognitive deficits in this population. We attempted to determine if verbal memory impairments were related to the most commonly studied disease parameters including adjuvant chemotherapy and chronic stress-related disruption of limbic system structures., Experimental Design: We used functional magnetic resonance imaging to test our hypothesis that women with breast cancer would show significantly lower brain activation during verbal declarative memory tasks compared with age and education-matched healthy female controls. We also assessed several stress-related variables including diurnal cortisol levels to test our hypothesis that women with breast cancer would show higher stress and this would contribute to brain activation deficits during memory tasks., Results: Women with breast cancer had significantly lower prefrontal cortex activation during the memory encoding condition compared with controls. However, the breast cancer group showed significantly greater activation than controls during the recall condition in multiple, diffuse brain regions. There were no significant differences between the groups in stress-related variables. Women who were treated with cyclophosphamide, methotrexate, and 5-fluorouracil chemotherapy showed lower prefrontal cortex activation during memory encoding., Conclusions: These results suggest that women with metastatic breast cancer may be at risk for verbal memory impairments as a result of altered functional brain activation profiles. These findings may be associated with chemotherapy type and/or other aspects of the breast cancer disease process.
- Published
- 2009
- Full Text
- View/download PDF
47. Myosin-IIA and ICAM-1 regulate the interchange between two distinct modes of T cell migration.
- Author
-
Jacobelli J, Bennett FC, Pandurangi P, Tooley AJ, and Krummel MF
- Subjects
- Animals, Cell Adhesion immunology, Cell Line, Immunoblotting, Intercellular Adhesion Molecule-1 metabolism, Mice, Microscopy, Confocal, Nonmuscle Myosin Type IIA metabolism, T-Lymphocytes cytology, T-Lymphocytes metabolism, Transfection, Cell Movement immunology, Intercellular Adhesion Molecule-1 immunology, Nonmuscle Myosin Type IIA immunology, T-Lymphocytes immunology
- Abstract
How T cells achieve rapid chemotactic motility under certain circumstances and efficient cell surface surveillance in others is not fully understood. We show that T lymphocytes are motile in two distinct modes: a fast "amoeboid-like" mode, which uses sequential discontinuous contacts to the substrate; and a slower mode using a single continuously translating adhesion, similar to mesenchymal motility. Myosin-IIA is necessary for fast amoeboid motility, and our data suggests that this occurs via cyclical rear-mediated compressions that eliminate existing adhesions while licensing subsequent ones at the front of the cell. Regulation of Myosin-IIA function in T cells is thus a key mechanism to regulate surface contact area and crawling velocity within different environments. This can provide T lymphocytes with motile and adhesive properties that are uniquely suited toward alternative requirements for immune surveillance and response.
- Published
- 2009
- Full Text
- View/download PDF
48. FOXO-regulated transcription restricts overgrowth of Tsc mutant organs.
- Author
-
Harvey KF, Mattila J, Sofer A, Bennett FC, Ramsey MR, Ellisen LW, Puig O, and Hariharan IK
- Subjects
- Animals, Cell Proliferation, Congenital Abnormalities genetics, Drosophila melanogaster cytology, Female, Gene Expression Regulation, Developmental genetics, Growth Inhibitors genetics, Growth Inhibitors metabolism, Male, Phosphatidylinositol 3-Kinases genetics, Protein Kinases, Regulatory Elements, Transcriptional genetics, Species Specificity, TOR Serine-Threonine Kinases, Transcription, Genetic genetics, Up-Regulation genetics, Drosophila Proteins genetics, Drosophila melanogaster embryology, Drosophila melanogaster genetics, Forkhead Transcription Factors genetics, Mutation genetics, Organogenesis genetics
- Abstract
FOXO is thought to function as a repressor of growth that is, in turn, inhibited by insulin signaling. However, inactivating mutations in Drosophila melanogaster FOXO result in viable flies of normal size, which raises a question over the involvement of FOXO in growth regulation. Previously, a growth-suppressive role for FOXO under conditions of increased target of rapamycin (TOR) pathway activity was described. Here, we further characterize this phenomenon. We show that tuberous sclerosis complex 1 mutations cause increased FOXO levels, resulting in elevated expression of FOXO-regulated genes, some of which are known to antagonize growth-promoting pathways. Analogous transcriptional changes are observed in mammalian cells, which implies that FOXO attenuates TOR-driven growth in diverse species.
- Published
- 2008
- Full Text
- View/download PDF
49. Fat cadherin modulates organ size in Drosophila via the Salvador/Warts/Hippo signaling pathway.
- Author
-
Bennett FC and Harvey KF
- Subjects
- Animals, Apoptosis, Cadherins genetics, Cadherins physiology, Cell Adhesion Molecules genetics, Cell Cycle, Cell Cycle Proteins metabolism, Cell Proliferation, Cyclin E, Drosophila embryology, Drosophila genetics, Drosophila Proteins genetics, Drosophila Proteins metabolism, Eye anatomy & histology, Eye embryology, Inhibitor of Apoptosis Proteins, Intracellular Signaling Peptides and Proteins, Membrane Proteins metabolism, Organ Size, Phenotype, Protein Kinases metabolism, Protein Serine-Threonine Kinases metabolism, Retina cytology, Wings, Animal anatomy & histology, Cell Adhesion Molecules physiology, Drosophila physiology, Drosophila Proteins physiology, Signal Transduction
- Abstract
Background: The atypical Fat cadherin has long been known to control cell proliferation and organ size in Drosophila, but the mechanism by which Fat controls these processes has remained elusive. A newly emerging signaling pathway that controls organ size during development is the Salvador/Warts/Hippo pathway., Results: Here we demonstrate that Fat limits organ size by modulating activity of the Salvador/Warts/Hippo pathway. ft interacts genetically with positive and negative regulators of this pathway, and tissue lacking fat closely phenocopies tissue deficient for genes that normally promote Salvador/Warts/Hippo pathway activity. Cells lacking fat grow and proliferate more quickly than their wild-type counterparts and exhibit delayed cell-cycle exit as a result of elevated expression of Cyclin E. fat mutant cells display partial insensitivity to normal developmental apoptosis cues and express increased levels of the anti-apoptotic DIAP1 protein. Collectively, these defects lead to increased organ size and organism lethality in fat mutant animals. Fat modulates Salvador/Warts/Hippo pathway activity by promoting abundance and localization of Expanded protein at the apical membrane of epithelial tissues., Conclusions: Fat restricts organ size during Drosophila development via the Salvador/Warts/Hippo pathway. These studies aid our understanding of developmental organ size control and have implications for human hyperproliferative disorders, such as cancers.
- Published
- 2006
- Full Text
- View/download PDF
50. Early intervention in low birth weight premature infants: results at 18 years of age for the Infant Health and Development Program.
- Author
-
McCormick MC, Brooks-Gunn J, Buka SL, Goldman J, Yu J, Salganik M, Scott DT, Bennett FC, Kay LL, Bernbaum JC, Bauer CR, Martin C, Woods ER, Martin A, and Casey PH
- Subjects
- Adolescent, Child, Child Behavior, Child Development, Cognition, Educational Status, Female, Follow-Up Studies, Health Status, Humans, Infant, Newborn, Intelligence, Male, Randomized Controlled Trials as Topic, Risk-Taking, Early Intervention, Educational, Infant, Low Birth Weight, Infant, Premature
- Abstract
Objective: To assess whether improvements in cognitive and behavioral development seen in preschool educational programs persist, we compared those in a multisite randomized trial of such a program over the first 3 years of life (INT) to those with follow-up only (FUO) at 18 months of age., Methods: This was a prospective follow-up of the Infant Health and Development Program at 8 sites heterogeneous for sociodemographic characteristics. Originally 985 children were randomized to the INT (n = 377) or FUO (n = 608) groups within 2 birth weight strata: heavier low birth weight (HLBW; 2001-2499 g) and lighter low birth weight (LLBW; < or = 2000 g). Primary outcome measures were the Peabody Picture Vocabulary Test (PPVT-III), reading and mathematics subscales of the Woodcock-Johnson Tests of Achievement, youth self-report on the Total Behavior Problem Index, and high-risk behaviors on the Youth Risk Behavior Surveillance System (YRBSS). Secondary outcomes included Weschler full-scale IQ, caregiver report on the Total Behavior Problem Index, and caregiver and youth self-reported physical health using the Medical Outcome Study measure. Assessors were masked as to study status., Results: We assessed 636 youths at 18 years (64.6% of the 985, 72% of whom had not died or refused at prior assessments). After adjusting for cohort attrition, differences favoring the INT group were seen on the Woodcock-Johnson Tests of Achievement in math (5.1 points), YRBSS (-0.7 points), and the PPVT-III (3.8 points) in the HLBW youth. In the LLBW youth, the Woodcock-Johnson Tests of Achievement in reading was higher in the FUO than INT group (4.2)., Conclusions: The findings in the HLBW INT group provide support for preschool education to make long-term changes in a diverse group of children who are at developmental risk. The lack of observable benefit in the LLBW group raises questions about the biological and educational factors that foster or inhibit sustained effects of early educational intervention.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.