112 results on '"Roberta Brambilla"'
Search Results
2. Chorioamnionitis accelerates granule cell and oligodendrocyte maturation in the cerebellum of preterm nonhuman primates
- Author
-
Josef Newman, Xiaoying Tong, April Tan, Toni Yeasky, Vanessa Nunes De Paiva, Pietro Presicce, Paranthaman S. Kannan, Kevin Williams, Andreas Damianos, Marione Tamase Newsam, Merline K. Benny, Shu Wu, Karen C. Young, Lisa A. Miller, Suhas G. Kallapur, Claire A. Chougnet, Alan H. Jobe, Roberta Brambilla, and Augusto F. Schmidt
- Subjects
Chorioamnionitis ,Cerebellum ,Granule cell ,Purkinje cell ,Oligodendrocyte ,Maturation ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Preterm birth is often associated with chorioamnionitis and leads to increased risk of neurodevelopmental disorders, such as autism. Preterm birth can lead to cerebellar underdevelopment, but the mechanisms of disrupted cerebellar development in preterm infants are not well understood. The cerebellum is consistently affected in people with autism spectrum disorders, showing reduction of Purkinje cells, decreased cerebellar grey matter, and altered connectivity. Methods Preterm rhesus macaque fetuses were exposed to intra-amniotic LPS (1 mg, E. coli O55:B5) at 127 days (80%) gestation and delivered by c-section 5 days after injections. Maternal and fetal plasma were sampled for cytokine measurements. Chorio-decidua was analyzed for immune cell populations by flow cytometry. Fetal cerebellum was sampled for histology and molecular analysis by single-nuclei RNA-sequencing (snRNA-seq) on a 10× chromium platform. snRNA-seq data were analyzed for differences in cell populations, cell-type specific gene expression, and inferred cellular communications. Results We leveraged snRNA-seq of the cerebellum in a clinically relevant rhesus macaque model of chorioamnionitis and preterm birth, to show that chorioamnionitis leads to Purkinje cell loss and disrupted maturation of granule cells and oligodendrocytes in the fetal cerebellum at late gestation. Purkinje cell loss is accompanied by decreased sonic hedgehog signaling from Purkinje cells to granule cells, which show an accelerated maturation, and to oligodendrocytes, which show accelerated maturation from pre-oligodendrocytes into myelinating oligodendrocytes. Conclusion These findings suggest a role of chorioamnionitis on disrupted cerebellar maturation associated with preterm birth and on the pathogenesis of neurodevelopmental disorders among preterm infants.
- Published
- 2024
- Full Text
- View/download PDF
3. Peripherally derived myeloid cells induce disease-dependent phenotypic changes in microglia
- Author
-
Estrid Thougaard, Brianna Carney, Agnieszka Wlodarczyk, Roberta Brambilla, and Kate Lykke Lambertsen
- Subjects
myeloid cells ,microglia ,multiple sclerosis ,ischemic stroke ,neuroinflammation ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
In central nervous system (CNS) injury and disease, peripherally derived myeloid cells infiltrate the CNS parenchyma and interact with resident cells, propagating the neuroinflammatory response. Because peripheral myeloid populations differ profoundly depending on the type and phase of injury, their crosstalk with CNS resident cells, particularly microglia, will lead to different functional outcomes. Thus, understanding how peripheral myeloid cells affect the phenotype and function of microglia in different disease conditions and phases may lead to a better understanding of disease-specific targetable pathways for neuroprotection and neurorepair. To this end, we set out to develop an in vitro system to investigate the communication between peripheral myeloid cells and microglia, with the goal of uncovering potential differences due to disease type and timing. We isolated peripheral myeloid cells from mice undergoing experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis, or acute cerebral ischemia by permanent middle cerebral artery occlusion (pMCAO) at different times after disease and probed their ability to change the phenotype of primary microglia isolated from the brain of adult mice. We identified changes not only dependent on the disease model, but also on the timepoint after disease onset from which the myeloid cells were isolated. Peripheral myeloid cells from acute EAE induced morphological changes in microglia, followed by increases in expression of genes involved in inflammatory signaling. Conversely, it was the peripheral myeloid cells from the chronic phase of pMCAO that induced gene expression changes in genes involved in inflammatory signaling and phagocytosis, which was not followed by a change in morphology. This underscores the importance of understanding the role of infiltrating myeloid cells in different disease contexts and phases. Furthermore, we showed that our assay is a valuable tool for investigating myeloid cell interactions in a range of CNS neuroinflammatory conditions.
- Published
- 2023
- Full Text
- View/download PDF
4. Distinct myeloid population phenotypes dependent on TREM2 expression levels shape the pathology of traumatic versus demyelinating CNS disorders
- Author
-
Han Gao, Jiawei Di, Bettina Hjelm Clausen, Nanxiang Wang, Xizhong Zhu, Tianlun Zhao, Yanyu Chang, Mao Pang, Yang Yang, Ronghan He, Yuge Wang, Liangming Zhang, Bin Liu, Wei Qiu, Kate Lykke Lambertsen, Roberta Brambilla, and Limin Rong
- Subjects
Biology (General) ,QH301-705.5 - Published
- 2023
- Full Text
- View/download PDF
5. Circulating extracellular vesicles activate the pyroptosis pathway in the brain following ventilation-induced lung injury
- Author
-
Laura Chavez, Julia Meguro, Shaoyi Chen, Vanessa Nunes de Paiva, Ronald Zambrano, Julia M. Eterno, Rahul Kumar, Matthew R. Duncan, Merline Benny, Karen C. Young, W. Dalton Dietrich, Roberta Brambilla, Shu Wu, and Augusto F. Schmidt
- Subjects
Prematurity ,Mechanical ventilation ,Lung injury ,Brain injury ,Gasdermin D ,Pyroptosis ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Mechanical ventilation of preterm newborns causes lung injury and is associated with poor neurodevelopmental outcomes. However, the mechanistic links between ventilation-induced lung injury (VILI) and brain injury is not well defined. Since circulating extracellular vesicles (EVs) are known to link distant organs by transferring their cargos, we hypothesized that EVs mediate inflammatory brain injury associated with VILI. Methods Neonatal rats were mechanically ventilated with low (10 mL/kg) or high (25 mL/kg) tidal volume for 1 h on post-natal day 7 followed by recovery for 2 weeks. Exosomes were isolated from the plasma of these rats and adoptively transferred into normal newborn rats. We assessed the effect of mechanical ventilation or exosome transfer on brain inflammation and activation of the pyroptosis pathway by western blot and histology. Results Injurious mechanical ventilation induced similar markers of inflammation and pyroptosis, such as increased IL-1β and activated caspase-1/gasdermin D (GSDMD) in both lung and brain, in addition to inducing microglial activation and cell death in the brain. Isolated EVs were enriched for the exosomal markers CD9 and CD81, suggesting enrichment for exosomes. EVs isolated from neonatal rats with VILI had increased caspase-1 but not GSDMD. Adoptive transfer of these EVs led to neuroinflammation with microglial activation and activation of caspase-1 and GSDMD in the brain similar to that observed in neonatal rats that were mechanically ventilated. Conclusions These findings suggest that circulating EVs can contribute to the brain injury and poor neurodevelopmental outcomes in preterm infants with VILI through activation of GSDMD.
- Published
- 2021
- Full Text
- View/download PDF
6. Transcriptional abnormalities in induced pluripotent stem cell-derived oligodendrocytes of individuals with primary progressive multiple sclerosis
- Author
-
Melanie J. Plastini, Haritha L. Desu, Maureen C. Ascona, Anna L. Lang, Mario A. Saporta, and Roberta Brambilla
- Subjects
oligodendrocytes ,human induced pluripotent stem cells ,primary progressive multiple sclerosis ,RNA-sequencing ,inflammasome ,NLRP2 ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Multiple sclerosis (MS) is the most common neurological disorder in young adults and is classically defined as a chronic inflammatory demyelinating disease of the central nervous system (CNS). Although MS affects millions of people worldwide, its underlying cause remains unknown making discovery of effective treatments challenging. Whether intrinsic or extrinsic factors contribute to MS initiation and progression is still unclear. This is especially true for primary progressive MS (PPMS), the rarest form of the disease, in which progressive and irreversible loss of neurological function is often observed in the absence of an overt immune-inflammatory response. To test the hypothesis that intrinsic dysfunction in oligodendrocytes (OLs), the primary targets of damage in MS, may contribute to PPMS etiopathology, we differentiated human induced pluripotent stem cell (hiPSC) lines derived from PPMS and healthy individuals into mature OLs to compare their transcriptional profile. PPMS derived OLs displayed hundreds of differentially expressed genes compared to control OLs, many associated with cell adhesion, apoptosis and inflammation, including the inflammasome component Nlrp2, which was highly upregulated. NLRP2 immunoreactivity in OLs was confirmed in post-mortem PPMS brain tissues, with higher expression than in control tissues. Altogether, our findings suggest that mature OLs in PPMS affected individuals carry intrinsic abnormalities that could contribute, at least in part, to the pathophysiology of this form of the disease.
- Published
- 2022
- Full Text
- View/download PDF
7. Selective Inhibition of Soluble Tumor Necrosis Factor Alters the Neuroinflammatory Response following Moderate Spinal Cord Injury in Mice
- Author
-
Minna Christiansen Lund, Ditte Gry Ellman, Pernille Vinther Nielsen, Stefano Raffaele, Marta Fumagalli, Raphael Guzman, Matilda Degn, Roberta Brambilla, Morten Meyer, Bettina Hjelm Clausen, and Kate Lykke Lambertsen
- Subjects
XPro1595 ,neuroinflammation ,microglia ,CNS trauma ,leukocyte infiltration ,Biology (General) ,QH301-705.5 - Abstract
Clinical and animal model studies have implicated inflammation and glial and peripheral immune cell responses in the pathophysiology of spinal cord injury (SCI). A key player in the inflammatory response after SCI is the pleiotropic cytokine tumor necrosis factor (TNF), which exists both in both a transmembrane (tmTNF) and a soluble (solTNF) form. In the present study, we extend our previous findings of a therapeutic effect of topically blocking solTNF signaling after SCI for three consecutive days on lesion size and functional outcome to study the effect on spatio-temporal changes in the inflammatory response after SCI in mice treated with the selective solTNF inhibitor XPro1595 and compared to saline-treated mice. We found that despite comparable TNF and TNF receptor levels between XPro1595- and saline-treated mice, XPro1595 transiently decreased pro-inflammatory interleukin (IL)-1β and IL-6 levels and increased pro-regenerative IL-10 levels in the acute phase after SCI. This was complemented by a decrease in the number of infiltrated leukocytes (macrophages and neutrophils) in the lesioned area of the spinal cord and an increase in the number of microglia in the peri-lesion area 14 days after SCI, followed by a decrease in microglial activation in the peri-lesion area 21 days after SCI. This translated into increased myelin preservation and improved functional outcomes in XPro1595-treated mice 35 days after SCI. Collectively, our data suggest that selective targeting of solTNF time-dependently modulates the neuroinflammatory response by favoring a pro-regenerative environment in the lesioned spinal cord, leading to improved functional outcomes.
- Published
- 2023
- Full Text
- View/download PDF
8. IC100: a novel anti-ASC monoclonal antibody improves functional outcomes in an animal model of multiple sclerosis
- Author
-
Haritha L. Desu, Melanie Plastini, Placido Illiano, Helen M. Bramlett, W. Dalton Dietrich, Juan Pablo de Rivero Vaccari, Roberta Brambilla, and Robert W. Keane
- Subjects
IC100 ,Inflammasome ,Neuroinflammation ,Multiple sclerosis ,Experimental autoimmune encephalomyelitis ,Caspase-1 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background The inflammasome adaptor apoptosis-associated speck-like protein containing a CARD (ASC) is involved in immune signaling by bridging the interactions between inflammasome sensors and caspase-1. Strong experimental evidence has shown that ASC−/− mice are protected from disease progression in animal models of multiple sclerosis (MS), suggesting that targeting inflammasome activation via ASC inhibition may be a promising therapeutic strategy in MS. Thus, the goal of our study is to test the efficacy of IC100, a novel humanized antibody targeting ASC, in preventing and/or suppressing disease in the experimental autoimmune encephalomyelitis (EAE) model of MS. Methods We employed the EAE model of MS where disease was induced by immunization of C57BL/6 mice with myelin oligodendrocyte glycoprotein peptide 35–55 (MOG35–55). Mice were treated with vehicle or increasing doses of IC100 (10, 30, and 45 mg/kg) and clinical disease course was evaluated up to 35 days post EAE induction. Immune cell infiltration into the spinal cord and microglia responses were assessed. Results We show that IC100 treatment reduced the severity of EAE when compared to vehicle-treated controls. At a dose of 30 mg/kg, IC100 significantly reduced the number of CD4+ and CD8+ T cells and CD11b+MHCII+ activated myeloid cells entering the spinal cord from the periphery, and reduced the number of total and activated microglia. Conclusions These data indicate that IC100 suppresses the immune-inflammatory response that drives EAE development and progression, thereby identifying ASC as a promising target for the treatment of MS as well as other neurological diseases with a neuroinflammatory component.
- Published
- 2020
- Full Text
- View/download PDF
9. The Inflammatory Response after Moderate Contusion Spinal Cord Injury: A Time Study
- Author
-
Minna Christiansen Lund, Ditte Gry Ellman, Maiken Nissen, Pernille Sveistrup Nielsen, Pernille Vinther Nielsen, Carina Jørgensen, Ditte Caroline Andersen, Han Gao, Roberta Brambilla, Matilda Degn, Bettina Hjelm Clausen, and Kate Lykke Lambertsen
- Subjects
neuroinflammation ,cytokines ,tumor necrosis factor ,immune cells ,microglia ,Biology (General) ,QH301-705.5 - Abstract
Spinal cord injury (SCI) initiates detrimental cellular and molecular events that lead to acute and delayed neuroinflammation. Understanding the role of the inflammatory response in SCI requires insight into the temporal and cellular synthesis of inflammatory mediators. We subjected C57BL/6J mice to SCI and investigated inflammatory reactions. We examined activation, recruitment, and polarization of microglia and infiltrating immune cells, focusing specifically on tumor necrosis factor (TNF) and its receptors TNFR1 and TNFR2. In the acute phase, TNF expression increased in glial cells and neuron-like cells, followed by infiltrating immune cells. TNFR1 and TNFR2 levels increased in the delayed phase and were found preferentially on neurons and glial cells, respectively. The acute phase was dominated by the infiltration of granulocytes and macrophages. Microglial/macrophage expression of Arg1 increased from 1–7 days after SCI, followed by an increase in Itgam, Cx3cr1, and P2ry12, which remained elevated throughout the study. By 21 and 28 days after SCI, the lesion core was populated by galectin-3+, CD68+, and CD11b+ microglia/macrophages, surrounded by a glial scar consisting of GFAP+ astrocytes. Findings were verified in postmortem tissue from individuals with SCI. Our findings support the consensus that future neuroprotective immunotherapies should aim to selectively neutralize detrimental immune signaling while sustaining pro-regenerative processes.
- Published
- 2022
- Full Text
- View/download PDF
10. Dynamic Responses of Microglia in Animal Models of Multiple Sclerosis
- Author
-
Melanie J. Plastini, Haritha L. Desu, and Roberta Brambilla
- Subjects
microglia ,neuroinflammation ,neurorepair ,multiple sclerosis ,neurological disease ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Microglia play an essential role in maintaining central nervous system (CNS) homeostasis, as well as responding to injury and disease. Most neurological disorders feature microglial activation, a process whereby microglia undergo profound morphological and transcriptional changes aimed at containing CNS damage and promoting repair, but often resulting in overt inflammation that sustains and propagates the neurodegenerative process. This is especially evident in multiple sclerosis (MS), were microglial activation and microglia-driven neuroinflammation are considered key events in the onset, progression, and resolution of the disease. Our understanding of microglial functions in MS has widened exponentially in the last decade by way of new tools and markers to discriminate microglia from other myeloid populations. Consequently, the complex functional and phenotypical diversity of microglia can now be appreciated. This, in combination with a variety of animal models that mimic specific features and processes of MS, has contributed to filling the gap of knowledge in the cascade of events underlying MS pathophysiology. The purpose of this review is to present the most up to date knowledge of the dynamic responses of microglia in the commonly used animal models of MS, specifically the immune-mediated experimental autoimmune encephalomyelitis (EAE) model, and the chemically-induced cuprizone and lysolecithin models. Elucidating the spectrum of microglial functions in these models, from detrimental to protective, is essential to identify emerging targets for therapy and guide drug discovery efforts.
- Published
- 2020
- Full Text
- View/download PDF
11. Fibrotic scar after experimental autoimmune encephalomyelitis inhibits oligodendrocyte differentiation
- Author
-
Stephanie L. Yahn, Jiajun Li, Irene Goo, Han Gao, Roberta Brambilla, and Jae K. Lee
- Subjects
Fibrosis ,EAE ,MS ,Myelination ,Fibrotic scar ,Perivascular fibroblasts ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Remyelination failure is a crucial component of disease progression in the autoimmune demyelinating disease Multiple Sclerosis (MS). The regenerative capacity of oligodendrocyte progenitor cells (OPCs) to replace myelinating oligodendrocytes is likely influenced by many aspects of the lesion environment including inflammatory signaling and extracellular matrix (ECM) deposition. These features of MS lesions are typically attributed to infiltrating leukocytes and reactive astrocytes. Here we demonstrate that fibroblasts also contribute to the inhibitory environment in the animal model of MS, experimental autoimmune encephalomyelitis (EAE). Using Col1α1GFP transgenic mice, we show that perivascular fibroblasts are activated in the spinal cord at EAE onset, and infiltrate the parenchyma by the peak of behavioral deficits where they are closely associated with areas of demyelination, myeloid cell accumulation, and ECM deposition. We further show that both fibroblast conditioned media and fibroblast ECM inhibit the differentiation of OPCs into mature oligodendrocytes. Taken together, our results indicate that the fibrotic scar is a major component of EAE pathology that leads to an inhibitory environment for remyelination, thus raising the possibility that anti-fibrotic mechanisms may serve as novel therapeutic targets for MS.
- Published
- 2020
- Full Text
- View/download PDF
12. TNFR2 Signaling Regulates the Immunomodulatory Function of Oligodendrocyte Precursor Cells
- Author
-
Haritha L. Desu, Placido Illiano, James S. Choi, Maureen C. Ascona, Han Gao, Jae K. Lee, and Roberta Brambilla
- Subjects
oligodendrocytes ,neuroinflammation ,remyelination ,cytokines ,TNF ,multiple sclerosis ,Cytology ,QH573-671 - Abstract
Multiple sclerosis (MS) is a neuroimmune disorder characterized by inflammation, CNS demyelination, and progressive neurodegeneration. Chronic MS patients exhibit impaired remyelination capacity, partly due to the changes that oligodendrocyte precursor cells (OPCs) undergo in response to the MS lesion environment. The cytokine tumor necrosis factor (TNF) is present in the MS-affected CNS and has been implicated in disease pathophysiology. Of the two active forms of TNF, transmembrane (tmTNF) and soluble (solTNF), tmTNF signals via TNFR2 mediating protective and reparative effects, including remyelination, whereas solTNF signals predominantly via TNFR1 promoting neurotoxicity. To better understand the mechanisms underlying repair failure in MS, we investigated the cellular responses of OPCs to inflammatory exposure and the specific role of TNFR2 signaling in their modulation. Following treatment of cultured OPCs with IFNγ, IL1β, and TNF, we observed, by RNA sequencing, marked inflammatory and immune activation of OPCs, accompanied by metabolic changes and dysregulation of their proliferation and differentiation programming. We also established the high likelihood of cell–cell interaction between OPCs and microglia in neuroinflammatory conditions, with OPCs able to produce chemokines that can recruit and activate microglia. Importantly, we showed that these functions are exacerbated when TNFR2 is ablated. Together, our data indicate that neuroinflammation leads OPCs to shift towards an immunomodulatory phenotype while diminishing their capacity to proliferate and differentiate, thus impairing their repair function. Furthermore, we demonstrated that TNFR2 plays a key role in this process, suggesting that boosting TNFR2 activation or its downstream signals could be an effective strategy to restore OPC reparative capacity in demyelinating disease.
- Published
- 2021
- Full Text
- View/download PDF
13. Topical Administration of a Soluble TNF Inhibitor Reduces Infarct Volume After Focal Cerebral Ischemia in Mice
- Author
-
Minna Yli-Karjanmaa, Bettina Hjelm Clausen, Matilda Degn, Hans Gram Novrup, Ditte Gry Ellman, Peter Toft-Jensen, David E. Szymkowski, Allan Stensballe, Morten Meyer, Roberta Brambilla, and Kate Lykke Lambertsen
- Subjects
ischemic stroke ,behavior ,cytokines ,microglial activation ,neuroprotection ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
BackgroundTumor necrosis factor, which exists both as a soluble (solTNF) and a transmembrane (tmTNF) protein, plays an important role in post-stroke inflammation. The objective of the present study was to test the effect of topical versus intracerebroventricular administration of XPro1595 (a solTNF inhibitor) and etanercept (a solTNF and tmTNF inhibitor) compared to saline on output measures such as infarct volume and post-stroke inflammation in mice.MethodsAdult male C57BL/6 mice were treated topically (2.5 mg/ml/1μl/h for 3 consecutive days) or intracerebroventricularly (1.25 mg/kg/0.5 ml, once) with saline, XPro1595, or etanercept immediately after permanent middle cerebral artery occlusion (pMCAO). Mice were allowed to survive 1 or 3 days. Infarct volume, microglial and leukocyte profiles, and inflammatory markers were evaluated.ResultsWe found that topical, and not intracerebroventricular, administration of XPro1595 reduced infarct volume at both 1 and 3 days after pMCAO. Etanercept showed no effect. We observed no changes in microglial or leukocyte populations. XPro1595 increased gene expression of P2ry12 at 1 day and Trem2 at 1 and 3 days, while decreasing Cx3cr1 expression at 1 and 3 days after pMCAO, suggesting a change in microglial activation toward a phagocytic phenotype.ConclusionOur data demonstrate that topical administration of XPro1595 for 3 consecutive days decreases infarct volumes after ischemic stroke, while modifying microglial activation and the inflammatory response post-stroke. This suggests that inhibitors of solTNF hold great promise for future neuroprotective treatment in ischemic stroke.
- Published
- 2019
- Full Text
- View/download PDF
14. Conditional Ablation of Myeloid TNF Improves Functional Outcome and Decreases Lesion Size after Spinal Cord Injury in Mice
- Author
-
Ditte Gry Ellman, Minna Christiansen Lund, Maiken Nissen, Pernille Sveistrup Nielsen, Charlotte Sørensen, Emilie Boye Lester, Estrid Thougaard, Louise Helskov Jørgensen, Sergei A. Nedospasov, Ditte Caroline Andersen, Jane Stubbe, Roberta Brambilla, Matilda Degn, and Kate Lykke Lambertsen
- Subjects
tumor necrosis factor ,spinal cord injury ,myeloid cells ,functional outcome ,Cytology ,QH573-671 - Abstract
Spinal cord injury (SCI) is a devastating condition consisting of an instant primary mechanical injury followed by a secondary injury that progresses for weeks to months. The cytokine tumor necrosis factor (TNF) plays an important role in the pathophysiology of SCI. We investigated the effect of myeloid TNF ablation (peripheral myeloid cells (macrophages and neutrophils) and microglia) versus central myeloid TNF ablation (microglia) in a SCI contusion model. We show that TNF ablation in macrophages and neutrophils leads to reduced lesion volume and improved functional outcome after SCI. In contrast, TNF ablation in microglia only or TNF deficiency in all cells had no effect. TNF levels tended to be decreased 3 h post-SCI in mice with peripheral myeloid TNF ablation and was significantly decreased 3 days after SCI. Leukocyte and microglia populations and all other cytokines (IL-1β, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12, and IFNγ) and chemokines (CCL2, CCL5, and CXCL1) investigated, in addition to TNFR1 and TNFR2, were comparable between genotypes. Analysis of post-SCI signaling cascades demonstrated that the MAPK kinase SAPK/JNK decreased and neuronal Bcl-XL levels increased post-SCI in mice with ablation of TNF in peripheral myeloid cells. These findings demonstrate that peripheral myeloid cell-derived TNF is pathogenic in SCI.
- Published
- 2020
- Full Text
- View/download PDF
15. Genetic Ablation of Soluble TNF Does Not Affect Lesion Size and Functional Recovery after Moderate Spinal Cord Injury in Mice
- Author
-
Ditte Gry Ellman, Matilda Degn, Minna Christiansen Lund, Bettina Hjelm Clausen, Hans Gram Novrup, Simon Bertram Flæng, Louise Helskov Jørgensen, Lujitha Suntharalingam, Åsa Fex Svenningsen, Roberta Brambilla, and Kate Lykke Lambertsen
- Subjects
Pathology ,RB1-214 - Abstract
Traumatic spinal cord injury (SCI) is followed by an instant increase in expression of the microglial-derived proinflammatory cytokine tumor necrosis factor (TNF) within the lesioned cord. TNF exists both as membrane-anchored TNF (mTNF) and as cleaved soluble TNF (solTNF). We previously demonstrated that epidural administration of a dominant-negative inhibitor of solTNF, XPro1595, to the contused spinal cord resulted in changes in Iba1 protein expression in microglia/macrophages, decreased lesion volume, and improved locomotor function. Here, we extend our studies using mice expressing mTNF, but no solTNF (mTNFΔ/Δ), to study the effect of genetic ablation of solTNF on SCI. We demonstrate that TNF levels were significantly decreased within the lesioned spinal cord 3 days after SCI in mTNFΔ/Δ mice compared to littermates. This decrease did, however, not translate into significant changes in other pro- and anti-inflammatory cytokines (IL-10, IL-1β, IL-6, IL-5, IL-2, CXCL1, CCL2, or CCL5), despite a tendency towards increased IL-10 and decreased IL-1β, TNFR1, and TNFR2 levels in mTNFΔ/Δ mice. In addition, microglial and leukocyte infiltration, activation state (Iba1, CD11b, CD11c, CD45, and MHCII), lesion size, and functional outcome after moderate SCI were comparable between genotypes. Collectively, our data demonstrate that genetic ablation of solTNF does not significantly modulate postlesion outcome after SCI.
- Published
- 2016
- Full Text
- View/download PDF
16. Cell-Autonomous Cxcl1 Sustains Tolerogenic Circuitries and Stromal Inflammation via Neutrophil-Derived TNF in Pancreatic Cancer
- Author
-
Anna Bianchi, Iago De Castro Silva, Nilesh U. Deshpande, Samara Singh, Siddharth Mehra, Vanessa T. Garrido, Xinyu Guo, Luis A. Nivelo, Despina S. Kolonias, Shannon J. Saigh, Eric Wieder, Christine I. Rafie, Austin R. Dosch, Zhiqun Zhou, Oliver Umland, Haleh Amirian, Ifeanyichukwu C. Ogobuiro, Jian Zhang, Yuguang Ban, Carina Shiau, Nagaraj S. Nagathihalli, Elizabeth A. Montgomery, William L. Hwang, Roberta Brambilla, Krishna Komanduri, Alejandro V. Villarino, Eneda Toska, Ben Z. Stanger, Dmitry I. Gabrilovich, Nipun B. Merchant, and Jashodeep Datta
- Subjects
Oncology - Abstract
We have shown that KRAS–TP53 genomic coalteration is associated with immune-excluded microenvironments, chemoresistance, and poor survival in pancreatic ductal adenocarcinoma (PDAC) patients. By treating KRAS–TP53 cooperativity as a model for high-risk biology, we now identify cell-autonomous Cxcl1 as a key mediator of spatial T-cell restriction via interactions with CXCR2+ neutrophilic myeloid-derived suppressor cells in human PDAC using imaging mass cytometry. Silencing of cell-intrinsic Cxcl1 in LSL-KrasG12D/+;Trp53R172H/+;Pdx-1Cre/+(KPC) cells reprograms the trafficking and functional dynamics of neutrophils to overcome T-cell exclusion and controls tumor growth in a T cell–dependent manner. Mechanistically, neutrophil-derived TNF is a central regulator of this immunologic rewiring, instigating feed-forward Cxcl1 overproduction from tumor cells and cancer-associated fibroblasts (CAF), T-cell dysfunction, and inflammatory CAF polarization via transmembrane TNF–TNFR2 interactions. TNFR2 inhibition disrupts this circuitry and improves sensitivity to chemotherapy in vivo. Our results uncover cancer cell–neutrophil cross-talk in which context-dependent TNF signaling amplifies stromal inflammation and immune tolerance to promote therapeutic resistance in PDAC. Significance: By decoding connections between high-risk tumor genotypes, cell-autonomous inflammatory programs, and myeloid-enriched/T cell–excluded contexts, we identify a novel role for neutrophil-derived TNF in sustaining immunosuppression and stromal inflammation in pancreatic tumor microenvironments. This work offers a conceptual framework by which targeting context-dependent TNF signaling may overcome hallmarks of chemoresistance in pancreatic cancer.
- Published
- 2023
- Full Text
- View/download PDF
17. Selective Inhibition of Soluble Tumor Necrosis Factor Alters the Neuroinflammatory Response following Moderate Spinal Cord Injury in Mice
- Author
-
Lambertsen, Minna Christiansen Lund, Ditte Gry Ellman, Pernille Vinther Nielsen, Stefano Raffaele, Marta Fumagalli, Raphael Guzman, Matilda Degn, Roberta Brambilla, Morten Meyer, Bettina Hjelm Clausen, and Kate Lykke
- Subjects
XPro1595 ,neuroinflammation ,microglia ,CNS trauma ,leukocyte infiltration - Abstract
Clinical and animal model studies have implicated inflammation and glial and peripheral immune cell responses in the pathophysiology of spinal cord injury (SCI). A key player in the inflammatory response after SCI is the pleiotropic cytokine tumor necrosis factor (TNF), which exists both in both a transmembrane (tmTNF) and a soluble (solTNF) form. In the present study, we extend our previous findings of a therapeutic effect of topically blocking solTNF signaling after SCI for three consecutive days on lesion size and functional outcome to study the effect on spatio-temporal changes in the inflammatory response after SCI in mice treated with the selective solTNF inhibitor XPro1595 and compared to saline-treated mice. We found that despite comparable TNF and TNF receptor levels between XPro1595- and saline-treated mice, XPro1595 transiently decreased pro-inflammatory interleukin (IL)-1β and IL-6 levels and increased pro-regenerative IL-10 levels in the acute phase after SCI. This was complemented by a decrease in the number of infiltrated leukocytes (macrophages and neutrophils) in the lesioned area of the spinal cord and an increase in the number of microglia in the peri-lesion area 14 days after SCI, followed by a decrease in microglial activation in the peri-lesion area 21 days after SCI. This translated into increased myelin preservation and improved functional outcomes in XPro1595-treated mice 35 days after SCI. Collectively, our data suggest that selective targeting of solTNF time-dependently modulates the neuroinflammatory response by favoring a pro-regenerative environment in the lesioned spinal cord, leading to improved functional outcomes.
- Published
- 2023
- Full Text
- View/download PDF
18. Data from Cell-Autonomous Cxcl1 Sustains Tolerogenic Circuitries and Stromal Inflammation via Neutrophil-Derived TNF in Pancreatic Cancer
- Author
-
Jashodeep Datta, Nipun B. Merchant, Dmitry I. Gabrilovich, Ben Z. Stanger, Eneda Toska, Alejandro V. Villarino, Krishna Komanduri, Roberta Brambilla, William L. Hwang, Elizabeth A. Montgomery, Nagaraj S. Nagathihalli, Carina Shiau, Yuguang Ban, Jian Zhang, Ifeanyichukwu C. Ogobuiro, Haleh Amirian, Oliver Umland, Zhiqun Zhou, Austin R. Dosch, Christine I. Rafie, Eric Wieder, Shannon J. Saigh, Despina S. Kolonias, Luis A. Nivelo, Xinyu Guo, Vanessa T. Garrido, Siddharth Mehra, Samara Singh, Nilesh U. Deshpande, Iago De Castro Silva, and Anna Bianchi
- Abstract
We have shown that KRAS–TP53 genomic coalteration is associated with immune-excluded microenvironments, chemoresistance, and poor survival in pancreatic ductal adenocarcinoma (PDAC) patients. By treating KRAS–TP53 cooperativity as a model for high-risk biology, we now identify cell-autonomous Cxcl1 as a key mediator of spatial T-cell restriction via interactions with CXCR2+ neutrophilic myeloid-derived suppressor cells in human PDAC using imaging mass cytometry. Silencing of cell-intrinsic Cxcl1 in LSL-KrasG12D/+;Trp53R172H/+;Pdx-1Cre/+(KPC) cells reprograms the trafficking and functional dynamics of neutrophils to overcome T-cell exclusion and controls tumor growth in a T cell–dependent manner. Mechanistically, neutrophil-derived TNF is a central regulator of this immunologic rewiring, instigating feed-forward Cxcl1 overproduction from tumor cells and cancer-associated fibroblasts (CAF), T-cell dysfunction, and inflammatory CAF polarization via transmembrane TNF–TNFR2 interactions. TNFR2 inhibition disrupts this circuitry and improves sensitivity to chemotherapy in vivo. Our results uncover cancer cell–neutrophil cross-talk in which context-dependent TNF signaling amplifies stromal inflammation and immune tolerance to promote therapeutic resistance in PDAC.Significance:By decoding connections between high-risk tumor genotypes, cell-autonomous inflammatory programs, and myeloid-enriched/T cell–excluded contexts, we identify a novel role for neutrophil-derived TNF in sustaining immunosuppression and stromal inflammation in pancreatic tumor microenvironments. This work offers a conceptual framework by which targeting context-dependent TNF signaling may overcome hallmarks of chemoresistance in pancreatic cancer.
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Table S1 from Cell-Autonomous Cxcl1 Sustains Tolerogenic Circuitries and Stromal Inflammation via Neutrophil-Derived TNF in Pancreatic Cancer
- Author
-
Jashodeep Datta, Nipun B. Merchant, Dmitry I. Gabrilovich, Ben Z. Stanger, Eneda Toska, Alejandro V. Villarino, Krishna Komanduri, Roberta Brambilla, William L. Hwang, Elizabeth A. Montgomery, Nagaraj S. Nagathihalli, Carina Shiau, Yuguang Ban, Jian Zhang, Ifeanyichukwu C. Ogobuiro, Haleh Amirian, Oliver Umland, Zhiqun Zhou, Austin R. Dosch, Christine I. Rafie, Eric Wieder, Shannon J. Saigh, Despina S. Kolonias, Luis A. Nivelo, Xinyu Guo, Vanessa T. Garrido, Siddharth Mehra, Samara Singh, Nilesh U. Deshpande, Iago De Castro Silva, and Anna Bianchi
- Abstract
Differentially expressed pathways comparing transcriptomes in KRAS-TP53 co-altered (n=23) and KRAS-altered/TP53WT (n=5) derived from Cancer Cell Line Encyclopedia
- Published
- 2023
- Full Text
- View/download PDF
20. Increased activity of IRE1 improves the clinical presentation of EAE
- Author
-
Valerie Bracchi-Ricard, Kayla Nguyen, Daniela Ricci, Brian Gaudette, Jorge Henao-Meija, Roberta Brambilla, Tetyana Martynyuk, Tali Gidalevitz, David Allman, John R. Bethea, and Yair Argon
- Subjects
Article - Abstract
Activation of the ER stress sensor IRE1α contributes to neuronal development and is known to induce neuronal remodelingin vitroandin vivo. On the other hand, excessive IRE1 activity is often detrimental and may contribute to neurodegeneration. To determine the consequences of increased activation of IRE1α, we used a mouse model expressing a C148S variant of IRE1α with increased and sustained activation. Surprisingly, the mutation did not affect the differentiation of highly secretory antibody-producing cells, but exhibited a strong protective effect in a mouse model of experimental autoimmune encephalomyelitis (EAE). Significant improvement in motor function was found in IRE1C148S mice with EAE relative to WT mice. Coincident with this improvement, there was reduced microgliosis in the spinal cord of IRE1C148S mice, with reduced expression of pro-inflammatory cytokine genes. This was accompanied by reduced axonal degeneration and enhanced CNPase levels, suggestiing improved myelin integrity. Interestingly, while the IRE1C148S mutation is expressed in all cells, the reduction in proinflammatory cytokines and in the activation of microglial activation marker IBA1, along with preservation of phagocytic gene expression, all point to microglia as the cell type contributing to the clinical improvement in IRE1C148S animals. Our data suggest that sustained increase in IRE1α activity can be protectivein vivo, and that this protection is cell type and context dependent. Considering the overwhelming but conflicting evidence for the role of the ER stress in neurological diseases, a better understanding of the function of ER stress sensors in physiological contexts is clearly needed.
- Published
- 2023
21. The Role of Tumor Necrosis Factor Following Spinal Cord Injury: A Systematic Review
- Author
-
Minna Christiansen Lund, Bettina Hjelm Clausen, Roberta Brambilla, and Kate Lykke Lambertsen
- Subjects
Cellular and Molecular Neuroscience ,Cell Biology ,General Medicine - Abstract
Pre-clinical studies place tumor necrosis factor (TNF) as a central player in the inflammatory response after spinal cord injury (SCI), and blocking its production and/or activity has been proposed as a possible treatment option after SCI. This systematic review provides an overview of the literature on the temporal and cellular expression of TNF after SCI and clarifies the potential for its therapeutic manipulation in SCI. A systematic search was performed in EMBASE (Ovid), MEDLINE (Ovid), and Web of Science (Core Collection). The search terms were the MeSH forms of tumor necrosis factor and spinal cord injury in the different databases, and the last search was performed on February 3, 2021. We found twenty-four articles examining the expression of TNF, with most using a thoracic contusive SCI model in rodents. Two articles described the expression of TNF receptors in the acute phase after SCI. Twenty-one articles described the manipulation of TNF signaling using genetic knock-out, pharmaceutical inhibition, or gain-of-function approaches. Overall, TNF expression increased rapidly after SCI, within the first hours, in resident cells (neurons, astrocytes, oligodendrocytes, and microglia) and again in macrophages in the chronic phase after injury. The review underscores the complexity of TNF's role after SCI and indicates that TNF inhibition is a promising therapeutic option. This review concludes that TNF plays a significant role in the inflammatory response after SCI and suggests that targeting TNF signaling is a feasible therapeutic approach.
- Published
- 2022
- Full Text
- View/download PDF
22. Single-nucleus RNA-seq reveals disrupted cell maturation by chorioamnionitis in the preterm cerebellum of nonhuman primates
- Author
-
Josef Newman, Xiaoying Tong, April Tan, Vanessa Nunes De Paiva, Pietro Presicce, Paranthaman S Kannan, Kevin Williams, Andreas Damianos, Marione Tamase Newsam, Merline Benny, Shu Wu, Karen Young, Lisa A. Miller, Suhas G Kallapur, Claire A Chougnet, Alan H Jobe, Roberta Brambilla, and Augusto F. Schmidt
- Abstract
Preterm birth is often associated with chorioamnionitis and increased risk of neurodevelopmental disorders. Preterm birth also leads to cerebellar underdevelopment but the mechanisms of disrupted cerebellar development in preterm infants are little understood. Here, we leveraged single-nuclei RNA-sequencing of the cerebellum in a rhesus macaque model of chorioamnionitis and preterm birth, to show that chorioamnionitis leads to Purkinje cell loss and disrupted maturation of granule cells and oligodendrocytes in the fetal cerebellum at late gestation. Purkinje cell loss is accompanied by decreased sonic hedgehog signaling from Purkinje cells to granule cells, which show an accelerated maturation. Chorioamnionitis also accelerated pre-oligodendrocyte maturation into myelinating oligodendrocytes, confirmed by increased expression of myelin basic protein in the cerebellum of chorioamnionitis-exposed fetuses. These findings are consistent with reported histopathological findings in individuals with autism and suggest a novel mechanism through which perinatal inflammation contributes to neurodevelopmental disorders.
- Published
- 2022
- Full Text
- View/download PDF
23. Adult-Onset Deficiency of Mitochondrial Complex III in a Mouse Model of Alzheimer's Disease Decreases Amyloid Beta Plaque Formation
- Author
-
Milena Pinto, Francisca Diaz, Nadee Nissanka, Chelsey S. Guastucci, Placido Illiano, Roberta Brambilla, and Carlos T. Moraes
- Subjects
Mice, Knockout ,Amyloid ,Amyloid beta-Peptides ,Neuroscience (miscellaneous) ,Plaque, Amyloid ,Cellular and Molecular Neuroscience ,Amyloid beta-Protein Precursor ,Disease Models, Animal ,Electron Transport Complex III ,Mice ,Oxidative Stress ,Neurology ,Alzheimer Disease ,Animals - Abstract
For decades, mitochondrial dysfunctions and the generation of reactive oxygen species have been proposed to promote the development and progression of the amyloid pathology in Alzheimer's disease, but this association is still debated. It is unclear whether different mitochondrial dysfunctions, such as oxidative phosphorylation deficiency and oxidative stress, are triggers or rather consequences of the formation of amyloid aggregates. Likewise, the role of the different mitochondrial oxidative phosphorylation complexes in Alzheimer's patients' brain remains poorly understood. Previous studies showed that genetic ablation of oxidative phosphorylation enzymes from early age decreased amyloid pathology, which were unexpected results. To better model oxidative phosphorylation defects in aging, we induced the ablation of mitochondrial Complex III (CIII
- Published
- 2022
24. IC100: a novel anti-ASC monoclonal antibody improves functional outcomes in an animal model of multiple sclerosis
- Author
-
Roberta Brambilla, Melanie J. Plastini, Robert W. Keane, Placido Illiano, W. Dalton Dietrich, Haritha L. Desu, Juan Pablo de Rivero Vaccari, and Helen M. Bramlett
- Subjects
Encephalomyelitis, Autoimmune, Experimental ,Immunology ,Caspase 1 ,Antibodies, Monoclonal, Humanized ,ASC ,lcsh:RC346-429 ,Myelin oligodendrocyte glycoprotein ,Inflammasome ,Multiple sclerosis ,Cellular and Molecular Neuroscience ,Mice ,Neuroinflammation ,medicine ,Animals ,Humans ,lcsh:Neurology. Diseases of the nervous system ,Experimental autoimmune encephalomyelitis ,biology ,Microglia ,business.industry ,IL-1 ,General Neuroscience ,Research ,Antibodies, Monoclonal ,PYCARD ,Recovery of Function ,medicine.disease ,CARD Signaling Adaptor Proteins ,Mice, Inbred C57BL ,Pycard ,medicine.anatomical_structure ,Neurology ,Spinal Cord ,Caspase-1 ,biology.protein ,Female ,business ,IC100 ,medicine.drug - Abstract
BackgroundThe inflammasome adaptor apoptosis-associated speck-like protein containing a CARD (ASC) is involved in immune signaling by bridging the interactions between inflammasome sensors and caspase-1. Strong experimental evidence has shown that ASC−/−mice are protected from disease progression in animal models of multiple sclerosis (MS), suggesting that targeting inflammasome activation via ASC inhibition may be a promising therapeutic strategy in MS. Thus, the goal of our study is to test the efficacy of IC100, a novel humanized antibody targeting ASC, in preventing and/or suppressing disease in the experimental autoimmune encephalomyelitis (EAE) model of MS.MethodsWe employed the EAE model of MS where disease was induced by immunization of C57BL/6 mice with myelin oligodendrocyte glycoprotein peptide 35–55 (MOG35–55). Mice were treated with vehicle or increasing doses of IC100 (10, 30, and 45 mg/kg) and clinical disease course was evaluated up to 35 days post EAE induction. Immune cell infiltration into the spinal cord and microglia responses were assessed.ResultsWe show that IC100 treatment reduced the severity of EAE when compared to vehicle-treated controls. At a dose of 30 mg/kg, IC100 significantly reduced the number of CD4+and CD8+T cells and CD11b+MHCII+activated myeloid cells entering the spinal cord from the periphery, and reduced the number of total and activated microglia.ConclusionsThese data indicate that IC100 suppresses the immune-inflammatory response that drives EAE development and progression, thereby identifying ASC as a promising target for the treatment of MS as well as other neurological diseases with a neuroinflammatory component.
- Published
- 2020
- Full Text
- View/download PDF
25. Increased Neuroprotective Microglia and Photoreceptor Survival in the Retina from a Peptide Inhibitor of Myeloid Differentiation Factor 88 (MyD88)
- Author
-
Abigail S. Hackam, Roberta Brambilla, Tal Carmy, Placido Illiano, and Kimberly Garces
- Subjects
Male ,0301 basic medicine ,Retinal degeneration ,Interleukin-27 ,Anti-Inflammatory Agents ,Apoptosis ,Biology ,Interleukin-1 receptor ,Neuroprotection ,Retina ,Toll-like receptor (TLR) ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Animals ,Photoreceptor Cells ,Receptor ,Cells, Cultured ,Chemokine CCL2 ,Arginase ,Microglia ,Retinal Degeneration ,Retinal ,General Medicine ,MyD88 ,medicine.disease ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Myeloid Differentiation Factor 88 ,Female ,sense organs ,Oligopeptides ,030217 neurology & neurosurgery - Abstract
Myeloid differentiation factor 88 (MyD88) is an adaptor protein for the Toll-like receptor (TLR) and interleukin 1 receptor (IL-1R) families of innate immunity receptors that mediate inflammatory responses to cellular injury. TLR/IL1R/MyD88 signaling is known to contribute to retinal degeneration, although how MyD88 regulates neuronal survival, and the effect of MyD88 on the inflammatory environment in the retina, is mostly unknown. In this study, we tested the hypothesis that blocking MyD88-mediated signaling early in retinal degeneration promotes transition of microglia towards a neuroprotective anti-inflammatory phenotype, resulting in enhanced photoreceptor survival. We also tested whether systemic delivery of a pharmacologic MyD88 inhibitor has therapeutic potential. The rd10 mouse model of retinal degeneration was injected intraperitoneally with increasing doses of a MyD88 blocking peptide or control peptide early in degeneration, and inflammatory responses and photoreceptor survival were measured at specific time points using flow cytometry, cytokine profiling, and electroretinograms. Our results demonstrated that rd10 mice injected with a low dose of MyD88 inhibitor peptide showed increased rod photoreceptor function and reduced apoptosis compared with control peptide and uninjected mice. MyD88 inhibition also resulted in fewer microglia/macrophage cells in the photoreceptor layer whereas total peripheral and retinal macrophage were not changed. Furthermore, increased number of cells expressing the Arg1 marker of neuroprotective microglia in the photoreceptor layer and higher MCP-1 and anti-inflammatory cytokine IL-27 were associated with photoreceptor survival. Therefore, these data suggest that the MyD88 inhibitor modified the retina environment to become less inflammatory, leading to improved photoreceptor function and survival.
- Published
- 2020
- Full Text
- View/download PDF
26. The mutual interplay of gut microbiota, diet and human disease
- Author
-
Cinzia Parolini, Placido Illiano, and Roberta Brambilla
- Subjects
Central Nervous System ,0301 basic medicine ,medicine.drug_class ,brain ,Antibiotics ,Disease ,Gut flora ,digestive system ,Biochemistry ,functional food ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,cardiovascular disease ,Neoplasms ,medicine ,Humans ,cancer ,Microbiome ,Molecular Biology ,gut microbiota ,biology ,Probiotics ,Brain ,Cell Biology ,central nervous system ,biology.organism_classification ,medicine.disease ,Gastrointestinal Microbiome ,Prebiotics ,030104 developmental biology ,probiotics ,Cardiovascular Diseases ,030220 oncology & carcinogenesis ,Immunology ,prebiotics ,Dysbiosis ,Bacteria - Abstract
The intestinal milieu harbours the gut microbiota, consisting of a complex community of bacteria, archaea, fungi, viruses and protozoans that bring to the host organism an endowment of cells and genes more numerous than its own. In the last 10 years, mounting evidence has highlighted the prominent influence of the gut mutualistic bacterial communities on human health. Microbial colonization occurs alongside with immune system development and plays a role in intestinal physiology. The community of the gut microbiota does not undergo significant fluctuations throughout adult life. However, bacterial infections, antibiotic treatment, lifestyle, surgery and diet might profoundly affect it. Gut microbiota dysbiosis, defined as marked alterations in the amount and function of the intestinal microorganisms, is correlated with the aetiology of chronic noncommunicable diseases, ranging from cardiovascular, neurologic, respiratory and metabolic illnesses to cancer. In this review, we focus on the interplay among gut microbiota, diet and host to provide a perspective on the role of microbiota and their unique metabolites in the pathogenesis and/or progression of various human disorders. We discuss interventions based on microbiome studies, that is faecal microbiota transplantation, probiotics and prebiotics, to introduce the concept that correcting gut dysbiosis can ameliorate disease symptoms, thus offering a new approach towards disease treatment.
- Published
- 2020
- Full Text
- View/download PDF
27. Mild deficiency of mitochondrial Complex III in a mouse model of Alzheimer’s disease decreases amyloid beta plaque formation
- Author
-
Roberta Brambilla, Chelsey S Guastucci, Milena F. Pinto, Francisca P. Díaz, Placido Illiano, Carlos T. Moraes, and Nadee Nissanka
- Subjects
Text mining ,biology ,Amyloid beta ,Chemistry ,business.industry ,mental disorders ,biology.protein ,Mitochondrial complex III ,Disease ,business ,Molecular biology - Abstract
Background: For decades, mitochondrial dysfunctions and the generation of reactive oxygen species have been proposed to promote the development and progression of the amyloid pathology in Alzheimer’s disease, but this association is still debated. In particular, it is still unclear if mitochondrial dysfunctions are a trigger or rather a consequence of the formation of amyloid aggregates, and in particular, the role of the different mitochondrial oxidative phosphorylation complexes in Alzheimer’s patients’ brain remains poorly understood. Methods: To study how mitochondrial Complex III defects affect amyloid beta pathology in vivo , we partially knocked out mitochondrial Complex III (CIII KO ) in mature forebrain neurons of an Alzheimer’s mouse model that develops plaque pathology (APP/PS1). Results: We found that Complex III dysfunction in adult neurons induced mild oxidative stress which did not correlate with increased amyloid beta accumulation. In fact, CIII KO -AD mice showed decreased plaque number, decreased Aβ42 toxic fragment and altered amyloid precursor protein cleavage pathway. Conclusions: Our results support a model in which mitochondrial dysfunction is not the cause of amyloid oligomer accumulation but rather a consequence of amyloid beta toxicity.
- Published
- 2021
- Full Text
- View/download PDF
28. Deconstructing Noncovalent Kelch-like ECH-Associated Protein 1 (Keap1) Inhibitors into Fragments to Reconstruct New Potent Compounds
- Author
-
Anthony D. Garcia, Kim T. Tran, Lars Jakobsen Høj, Jakob S. Pallesen, Tommy N. Johansen, Giuseppe Marseglia, Michael Gajhede, Federico Munafò, Michael Sattler, Grzegorz M Popowicz, Haritha L. Desu, Dilip Narayanan, Sara Marie Øie Solbak, Anders Bach, Louis M.E. Sørensen, Rosa M. C. Carmona, Roberta Brambilla, University of Copenhagen = Københavns Universitet (KU), University of Parma = Università degli studi di Parma [Parme, Italie], Institut des Sciences Chimiques de Rennes (ISCR), Centre National de la Recherche Scientifique (CNRS)-Institut de Chimie du CNRS (INC)-Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES)-Ecole Nationale Supérieure de Chimie de Rennes (ENSCR)-Institut National des Sciences Appliquées - Rennes (INSA Rennes), Institut National des Sciences Appliquées (INSA)-Université de Rennes (UNIV-RENNES)-Institut National des Sciences Appliquées (INSA), University of Miami Leonard M. Miller School of Medicine (UMMSM), University of Southern Denmark (SDU), Helmholtz-Zentrum München (HZM), This research was supported by the Lundbeck Foundation (grant R190-2014-3710 for A.B.), the A. P. MøllerFoundation for the Advancement of Medical Science (grant 14-28 for A.B.), the Hørslev Foundation (grant203866-MIA for A.B.), the Augustinus Foundation (grant 14-1571 for A.B.), and the Drug ResearchAcademy/Lundbeck Foundation (scholarship for K.T.T.). We also acknowledge funding from the EuropeanUnion’s Framework Programme for Researc and Innovation Horizon 2020 (2014-2020) under the MarieSkłodowska-Curie Grant Agreement No. 675555, Accelerated Early staGe drug discovery (AEGIS) and theHelmholtz Center Munich to M.S. and G.P., and access to NMR measurements at the Bavarian NMR Center andat University of Copenhagen (the latter supported by grant #10-085264 from The Danish Research Council forIndependent Research | Nature and Universe and grant R77-A6742 from the Lundbeck Foundation). We thankall the staff at the European beamlines (BioMAX at MAX IV, Sweden, ID29 and ID30a at ESRF, France, and P13and P14 at DESY, Germany) for beamtime and their support and help., University of Copenhagen = Københavns Universitet (UCPH), Università degli studi di Parma = University of Parma (UNIPR), Université de Rennes (UR)-Institut National des Sciences Appliquées - Rennes (INSA Rennes), Institut National des Sciences Appliquées (INSA)-Institut National des Sciences Appliquées (INSA)-Ecole Nationale Supérieure de Chimie de Rennes (ENSCR)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS), and Helmholtz Zentrum München = German Research Center for Environmental Health
- Subjects
Magnetic Resonance Spectroscopy ,Stereochemistry ,NF-E2-Related Factor 2 ,Binding pocket ,Molecular Dynamics Simulation ,Crystallography, X-Ray ,Ligands ,01 natural sciences ,Small Molecule Libraries ,03 medical and health sciences ,Structure-Activity Relationship ,Drug Stability ,Microsomes ,Drug Discovery ,[CHIM]Chemical Sciences ,Humans ,Protein Interaction Maps ,030304 developmental biology ,Therapeutic strategy ,0303 health sciences ,Control diseases ,Binding Sites ,Kelch-Like ECH-Associated Protein 1 ,Chemistry ,Surface Plasmon Resonance ,KEAP1 ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Microsome ,Molecular Medicine ,Protein Binding - Abstract
Targeting the protein–protein interaction (PPI) between nuclear factor erythroid 2-related factor 2 (Nrf2) and Kelch-like ECH-associated protein 1 (Keap1) is a potential therapeutic strategy to control diseases involving oxidative stress. Here, six classes of known small-molecule Keap1–Nrf2 PPI inhibitors were dissected into 77 fragments in a fragment-based deconstruction reconstruction (FBDR) study and tested in four orthogonal assays. This gave 17 fragment hits of which six were shown by X-ray crystallography to bind in the Keap1 Kelch binding pocket. Two hits were merged into compound 8 with a 220–380-fold stronger affinity (Ki = 16 μM) relative to the parent fragments. Systematic optimization resulted in several novel analogues with Ki values of 0.04–0.5 μM, binding modes determined by X-ray crystallography, and enhanced microsomal stability. This demonstrates how FBDR can be used to find new fragment hits, elucidate important ligand–protein interactions, and identify new potent inhibitors of the Keap1–Nrf2 PPI. Targeting the protein-protein interaction (PPI) between nuclear factor erythroid 2-related factor 2 (Nrf2) and Kelch-like ECH-associated protein 1 (Keap1) is a potential therapeutic strategy to control diseases involving oxidative stress. Here, six classes of known small-molecule Keap1-Nrf2 PPI inhibitors were dissected into 77 fragments in a fragment-based deconstruction reconstruction (FBDR) study and tested in four orthogonal assays. This gave 17 fragment hits of which six were shown by X-ray crystallography to bind in the Keap1 Kelch binding pocket. Two hits were merged into compound 8 with a 220-380-fold stronger affinity (Ki = 16 μM) relative to the parent fragments. Systematic optimization resulted in several novel analogues with Ki values of 0.04-0.5 μM, binding modes determined by X-ray crystallography, and enhanced microsomal stability. This demonstrates how FBDR can be used to find new fragment hits, elucidate important ligand-protein interactions, and identify new potent inhibitors of the Keap1-Nrf2 PPI.
- Published
- 2021
- Full Text
- View/download PDF
29. Conditional Ablation of Myeloid TNF Improves Functional Outcome and Decreases Lesion Size after Spinal Cord Injury in Mice
- Author
-
Emilie Boye Lester, Pernille Sveistrup Nielsen, Roberta Brambilla, Charlotte Sørensen, Minna Christiansen Lund, Matilda Degn, Sergei A. Nedospasov, Estrid Thougaard, Louise Helskov Jørgensen, Ditte Caroline Andersen, Ditte Gry Ellman, Maiken Nissen, Jane Stubbe, and Kate Lykke Lambertsen
- Subjects
Chemokine ,Myeloid ,MAP Kinase Signaling System ,Neutrophils ,medicine.medical_treatment ,tumor necrosis factor ,CX3C Chemokine Receptor 1 ,bcl-X Protein ,CCL2 ,Motor Activity ,Article ,functional outcome ,Lesion ,Mice ,medicine ,Animals ,lcsh:QH301-705.5 ,Spinal Cord Injuries ,Inflammation ,Microglia ,biology ,business.industry ,Tumor Necrosis Factor-alpha ,Macrophages ,General Medicine ,Recovery of Function ,spinal cord injury ,CXCL1 ,STAT Transcription Factors ,medicine.anatomical_structure ,Cytokine ,lcsh:Biology (General) ,Spinal Cord ,myeloid cells ,biology.protein ,Cancer research ,Tumor necrosis factor alpha ,medicine.symptom ,business ,Proto-Oncogene Proteins c-akt ,Gene Deletion - Abstract
Spinal cord injury (SCI) is a devastating condition consisting of an instant primary mechanical injury followed by a secondary injury that progresses for weeks to months. The cytokine tumor necrosis factor (TNF) plays an important role in the pathophysiology of SCI. We investigated the effect of myeloid TNF ablation (peripheral myeloid cells (macrophages and neutrophils) and microglia) versus central myeloid TNF ablation (microglia) in a SCI contusion model. We show that TNF ablation in macrophages and neutrophils leads to reduced lesion volume and improved functional outcome after SCI. In contrast, TNF ablation in microglia only or TNF deficiency in all cells had no effect. TNF levels tended to be decreased 3 h post-SCI in mice with peripheral myeloid TNF ablation and was significantly decreased 3 days after SCI. Leukocyte and microglia populations and all other cytokines (IL-1&beta, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12, and IFN&gamma, ) and chemokines (CCL2, CCL5, and CXCL1) investigated, in addition to TNFR1 and TNFR2, were comparable between genotypes. Analysis of post-SCI signaling cascades demonstrated that the MAPK kinase SAPK/JNK decreased and neuronal Bcl-XL levels increased post-SCI in mice with ablation of TNF in peripheral myeloid cells. These findings demonstrate that peripheral myeloid cell-derived TNF is pathogenic in SCI.
- Published
- 2020
- Full Text
- View/download PDF
30. Tumor Necrosis Factor Inhibition in the Acute Management of Traumatic Optic Neuropathy
- Author
-
Daniel Pelaez, Ryan A. Gallo, David T. Tse, Wensi Tao, Brian C. Tse, Galina Dvoriantchikova, Steven Pappas, John Y. Lee, Roberta Brambilla, and Dmitry Ivanov
- Subjects
Retinal Ganglion Cells ,Pathology ,medicine.medical_specialty ,genetic structures ,Cell Survival ,Nerve Crush ,Injections, Subcutaneous ,traumatic optic neuropathy ,tumor necrosis factor ,Real-Time Polymerase Chain Reaction ,Neuroprotection ,Retinal ganglion ,Etanercept ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Electroretinography ,medicine ,Animals ,Retina ,Eye Movements, Strabismus, Amblyopia and Neuro-Ophthalmology ,Tumor Necrosis Factor-alpha ,business.industry ,Immunohistochemistry ,eye diseases ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,Gene Expression Regulation ,Optic Nerve Injuries ,Acute Disease ,030221 ophthalmology & optometry ,Optic nerve ,Tumor necrosis factor alpha ,sense organs ,business ,etanercept ,Immunosuppressive Agents ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Purpose To determine the effectiveness of etanercept, a tumor necrosis factor (TNF) inhibitor, in conferring neuroprotection to retinal ganglion cells (RGCs) and improving visual outcomes after optic nerve trauma with either optic nerve crush (ONC) or sonication-induced traumatic optic neuropathy (SI-TON) in mice. Methods Mouse optic nerves were unilaterally subjected to ONC (n = 20) or SI-TON (n = 20). TNF expression was evaluated by using immunohistochemistry and quantitative RT-PCR (qRT-PCR) in optic nerves harvested 6 and 24 hours post ONC (n = 10) and SI-TON (n = 10). Mice in each injury group received daily subcutaneous injections of either etanercept (10 mg/kg of body weight; five mice) or vehicle (five mice) for 7 days. Pattern electroretinograms were performed on all mice at 1 and 2 weeks after injury. ONC mice were killed at 2 weeks after injury, while SI-TON mice were euthanized at 4 weeks after injury. Whole retina flat-mounts were used for RGC quantification. Results Immunohistochemistry and qRT-PCR showed upregulation of TNF protein and gene expression within 24 hours after injury. In both models, etanercept use immediately following optic nerve injury led to higher RGC survival when compared to controls, which was comparable between the two models (24.23% in ONC versus 20.42% in SI-TON). In both models, 1 and 2 weeks post injury, mice treated with etanercept had significantly higher a-wave amplitudes than untreated injured controls. Conclusions Treatment with etanercept significantly reduced retinal damage and improved visual function in both animal models of TON. These findings suggest that reducing TNF activity in injured optic nerves constitutes an effective therapeutic approach in an acute setting.
- Published
- 2018
31. Corrigendum to: Oligodendrocytes modulate the immune-inflammatory response in EAE via TNFR2 signaling
- Author
-
Kate Lykke Lambertsen, Pernille M. Madsen, Haritha L. Desu, Yoleinny Florimon, Roberta Brambilla, Juan Pablo de Rivero Vaccari, Bettina Hjelm Clausen, Ditte Gry Ellman, and Robert W. Keane
- Subjects
Behavioral Neuroscience ,Text mining ,Immune system ,Endocrine and Autonomic Systems ,business.industry ,Inflammatory response ,Immunology ,Medicine ,business - Published
- 2021
- Full Text
- View/download PDF
32. Oligodendrocytes modulate the immune-inflammatory response in EAE via TNFR2 signaling
- Author
-
Bettina Hjelm Clausen, Juan Pablo de Rivero Vaccari, Yoleinny Florimon, Roberta Brambilla, Robert W. Keane, Pernille M. Madsen, Kate Lykke Lambertsen, Ditte Gry Ellman, and Haritha L. Desu
- Subjects
0301 basic medicine ,Encephalomyelitis, Autoimmune, Experimental ,Multiple Sclerosis ,medicine.medical_treatment ,Immunology ,Inflammation ,Biology ,Article ,Proinflammatory cytokine ,03 medical and health sciences ,Behavioral Neuroscience ,Mice ,0302 clinical medicine ,medicine ,Animals ,Receptors, Tumor Necrosis Factor, Type II ,Pathophysiology of multiple sclerosis ,Neuroinflammation ,Endocrine and Autonomic Systems ,Experimental autoimmune encephalomyelitis ,Inflammasome ,medicine.disease ,Mice, Inbred C57BL ,Oligodendroglia ,030104 developmental biology ,Cytokine ,Tumor necrosis factor alpha ,Female ,medicine.symptom ,030217 neurology & neurosurgery ,medicine.drug ,Signal Transduction - Abstract
The pleotropic cytokine tumor necrosis factor (TNF) is involved in the pathophysiology of multiple sclerosis (MS). In various models of MS, including experimental autoimmune encephalomyelitis (EAE), the membrane-bound form of TNF (tmTNF), which signals primarily via TNFR2, mediates protective and reparative effects, whereas the soluble form (solTNF), which signals primarily via TNFR1, promotes pro-inflammatory and detrimental functions. In this study, we investigated the role of TNFR2 expressed in oligodendrocytes in the early phase of EAE pathogenesis. We demonstrated that mice with specific ablation of oligodendroglial TNFR2 displayed early onset and higher peak of motor dysfunction when subjected to EAE, in advance of which accelerated infiltration of immune cells was observed as early as 10 days post EAE induction. The immune cell influx was preceded by microglial activation and increased blood brain barrier permeability. Lack of oligodendroglial TNFR2 accelerated the expression of inflammatory cytokines as well as expression and activation of the inflammasome. Gene expression profiling of oligodendrocytes sorted from the spinal cord 14 days post EAE induction showed robust upregulation of inflammatory genes, some of which were elevated in cells lacking TNFR2 compared to controls. Together, our data demonstrate that oligodendrocytes are directly involved in inflammation and immune modulation in CNS disease and this function is regulated, at least in part, by TNFR2.
- Published
- 2019
33. Topical administration of a soluble TNF inhibitor reduces infarct volume after focal cerebral ischemia in mice
- Author
-
Yli-Karjanmaa, M., Bettina Hjelm Clausen, Degn, M., Novrup, H. G., Ellman, D. G., Szymkowski, D. E., Morten Meyer, Roberta Brambilla, and Kate L. Lambertsen
- Published
- 2019
34. TNF deficiency causes alterations in the spatial organization of neurogenic zones and alters the number of microglia and neurons in the cerebral cortex
- Author
-
Nellie Anne Martin, Alexandre Breton, Daniel C. Anthony, Jan Bert Gramsbergen, Roberta Brambilla, Lars Henrik Frich, Kathrine Solevad Larsen, Morten Meyer, Minna Liisa Kyllikki Yli-Karjanmaa, Peter Toft jensen, Kate Lykke Lambertsen, Lubov Nathanson, Pernille Vinther Nielsen, Stephanie Lindeman Carlsen, Jane Stubbe, Minna Christiansen Lund, Helen B. Stolp, Lotte K. Kristensen, Christina Fenger, and Bente Finsen
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,Neurogenesis ,Immunology ,Hippocampus ,Etanercept ,TNF inhibitors ,03 medical and health sciences ,Behavioral Neuroscience ,Mice ,0302 clinical medicine ,Cognition ,Memory ,Internal medicine ,Medicine ,Animals ,Wnt Signaling Pathway ,Cerebral Cortex ,Mice, Knockout ,Neurons ,Microglia ,Endocrine and Autonomic Systems ,business.industry ,Tumor Necrosis Factor-alpha ,Dentate gyrus ,Granule cell ,TNF inhibitor ,Barnes maze ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Cytokines ,Tumor necrosis factor alpha ,Tumor Necrosis Factor Inhibitors ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Background: Although tumor necrosis factor (TNF) inhibitors are used to treat chronic inflammatory diseases, there is little information about how long-term inhibition of TNF affects the homeostatic functions that TNF maintains in the intact CNS. Materials and methods: To assess whether developmental TNF deficiency causes alterations in the naïve CNS, we estimated the number of proliferating cells, microglia, and neurons in the developing neocortex of E13.5, P7 and adult TNF knock out (TNF −/−) mice and wildtype (WT) littermates. We also measured changes in gene and protein expression and monoamine levels in adult WT and TNF −/− mice. To evaluate long-term effects of TNF inhibitors, we treated healthy adult C57BL/6 mice with either saline, the selective soluble TNF inhibitor XPro1595, or the nonselective TNF inhibitor etanercept. We estimated changes in cell number and protein expression after two months of treatment. We assessed the effects of TNF deficiency on cognition by testing adult WT and TNF −/− mice and mice treated with saline, XPro1595, or etanercept with specific behavioral tasks. Results: TNF deficiency decreased the number of proliferating cells and microglia and increased the number of neurons. At the same time, TNF deficiency decreased the expression of WNT signaling-related proteins, specifically Collagen Triple Helix Repeat Containing 1 (CTHRC1) and Frizzled receptor 6 (FZD6). In contrast to XPro1595, long-term inhibition of TNF with etanercept in adult C57BL/6 mice decreased the number of BrdU + cells in the granule cell layer of the dentate gyrus. Etanercept, but not XPro1595, also impaired spatial learning and memory in the Barnes maze memory test. Conclusion: TNF deficiency impacts the organization of neurogenic zones and alters the cell composition in brain. Long-term inhibition of TNF with the nonselective TNF inhibitor etanercept, but not the soluble TNF inhibitor XPro1595, decreases neurogenesis in the adult mouse hippocampus and impairs learning and memory after two months of treatment.
- Published
- 2019
- Full Text
- View/download PDF
35. Neuroinflammation, the thread connecting neurological disease : Cluster: 'Neuroinflammatory mechanisms in neurodegenerative disorders'
- Author
-
Roberta Brambilla
- Subjects
Inflammation ,Cellular and Molecular Neuroscience ,Review Literature as Topic ,business.industry ,Medicine ,Animals ,Humans ,Neurodegenerative Diseases ,Neurology (clinical) ,business ,Neuroscience ,Neuroinflammation ,Pathology and Forensic Medicine - Published
- 2019
- Full Text
- View/download PDF
36. The contribution of astrocytes to the neuroinflammatory response in multiple sclerosis and experimental autoimmune encephalomyelitis
- Author
-
Roberta Brambilla
- Subjects
0301 basic medicine ,Encephalomyelitis, Autoimmune, Experimental ,Multiple Sclerosis ,Population ,Central nervous system ,Disease ,medicine.disease_cause ,Article ,Pathology and Forensic Medicine ,Autoimmunity ,Multiple sclerosis ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Astroglia ,0302 clinical medicine ,Neuroinflammation ,medicine ,Animals ,Humans ,education ,Demyelinating Disorder ,Demyelinating disorder ,Inflammation ,education.field_of_study ,Neuroimmune disease ,Experimental autoimmune encephalomyelitis ,business.industry ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Astrocytes ,Neurology (clinical) ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Neuroinflammation is the coordinated response of the central nervous system (CNS) to threats to its integrity posed by a variety of conditions, including autoimmunity, pathogens and trauma. Activated astrocytes, in concert with other cellular elements of the CNS and immune system, are important players in the modulation of the neuroinflammatory response. During neurological disease, they produce and respond to cellular signals that often lead to dichotomous processes, which can promote further damage or contribute to repair. This occurs also in multiple sclerosis (MS), where astrocytes are now recognized as key components of its immunopathology. Evidence supporting this role has emerged not only from studies in MS patients, but also from animal models, among which the experimental autoimmune encephalomyelitis (EAE) model has proved especially instrumental. Based on this premise, the purpose of the present review is to summarize the current knowledge of astrocyte behavior in MS and EAE. Following a brief description of the pathological characteristics of the two diseases and the main functional roles of astrocytes in CNS physiology, we will delve into the specific responses of this cell population, analyzing MS and EAE in parallel. We will define the temporal and anatomical profile of astroglial activation, then focus on key processes they participate in. These include: (1) production and response to soluble mediators (e.g., cytokines and chemokines), (2) regulation of oxidative stress, and (3) maintenance of BBB integrity and function. Finally, we will review the state of the art on the available methods to measure astroglial activation in vivo in MS patients, and how this could be exploited to optimize diagnosis, prognosis and treatment decisions. Ultimately, we believe that integrating the knowledge obtained from studies in MS and EAE may help not only better understand the pathophysiology of MS, but also uncover new signals to be targeted for therapeutic intervention.
- Published
- 2019
- Full Text
- View/download PDF
37. Response
- Author
-
Cristina Giannattasio, Antonio Vincenti, Monica Failla, Anna Capra, Antonio Cirò, Sergio De Ceglia, Gaetano Gentile, Roberta Brambilla, and Giuseppe Mancia
- Subjects
Internal Medicine - Published
- 2019
- Full Text
- View/download PDF
38. Oligodendroglial TNFR2 Mediates Membrane TNF-Dependent Repair in Experimental Autoimmune Encephalomyelitis by Promoting Oligodendrocyte Differentiation and Remyelination
- Author
-
Dario Motti, Roberta Brambilla, Pernille M. Madsen, John R. Bethea, Shaffiat Karmally, Kate Lykke Lambertsen, and David E. Szymkowski
- Subjects
Male ,0301 basic medicine ,Neuroglia/metabolism ,Cell Survival/genetics ,Mice ,Myelin ,0302 clinical medicine ,Neuroinflammation ,Neural Stem Cells ,Myelin Sheath ,Mice, Knockout ,Tumor Necrosis Factor-alpha/metabolism ,Behavior, Animal ,General Neuroscience ,Experimental autoimmune encephalomyelitis ,Neurodegeneration ,Cell Differentiation ,Articles ,Receptors, Tumor Necrosis Factor, Type II/genetics ,medicine.anatomical_structure ,Female ,Demyelination ,medicine.symptom ,Neuroglia ,Nerve Regeneration/genetics ,Encephalomyelitis, Autoimmune, Experimental ,Cell Survival ,Encephalomyelitis, Autoimmune, Experimental/genetics ,Cell Differentiation/genetics ,Inflammation ,Biology ,Gene Expression Regulation/genetics ,Multiple sclerosis ,03 medical and health sciences ,medicine ,Receptors, Tumor Necrosis Factor, Type II ,Animals ,Remyelination ,Cytokine ,Tumor Necrosis Factor-alpha ,Oligodendrocyte differentiation ,medicine.disease ,Axons ,Nerve Regeneration ,Axons/pathology ,030104 developmental biology ,Gene Expression Regulation ,Cancer research ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Tumor necrosis factor (TNF) is associated with the pathophysiology of various neurological disorders, including multiple sclerosis. It exists as a transmembrane form tmTNF, signaling via TNF receptor 2 (TNFR2) and TNFR1, and a soluble form, solTNF, signaling via TNFR1. Multiple sclerosis is associated with the detrimental effects of solTNF acting through TNFR1, while tmTNF promotes repair and remyelination. Here we demonstrate that oligodendroglial TNFR2 is a key mediator of tmTNF-dependent protection in experimental autoimmune encephalomyelitis (EAE). CNP-cre:TNFR2fl/flmice with TNFR2 ablation in oligodendrocytes show exacerbation of the disease with increased axon and myelin pathology, reduced remyelination, and increased loss of oligodendrocyte precursor cells and mature oligodendrocytes. The clinical course of EAE is not improved by the solTNF inhibitor XPro1595 in CNP-cre:TNFR2fl/flmice, indicating that for tmTNF to promote recovery TNFR2 in oligodendrocytes is required. We show that TNFR2 drives differentiation of oligodendrocyte precursor cells, but not proliferation or survival. TNFR2 ablation leads to dysregulated expression of microRNAs, among which are regulators of oligodendrocyte differentiation and inflammation, including miR-7a. Our data provide the first directin vivoevidence that TNFR2 in oligodendrocytes is important for oligodendrocyte differentiation, thereby sustaining tmTNF-dependent repair in neuroimmune disease. Our studies identify TNFR2 in the CNS as a molecular target for the development of remyelinating agents, addressing the most pressing need in multiple sclerosis therapy nowadays.SIGNIFICANCE STATEMENTOur study, using novel TNF receptor 2 (TNFR2) conditional KO mice with selective TNFR2 ablation in oligodendrocytes, provides the first direct evidence that TNFR2 is an important signal for oligodendrocyte differentiation. Following activation by transmembrane TNF, TNFR2 initiates pathways that drive oligodendrocytes into a reparative mode contributing to remyelination following disease. This identifies TNFR2 as a new molecular target for the development of therapeutic agents in multiple sclerosis.
- Published
- 2016
- Full Text
- View/download PDF
39. Anti-TNF therapy alters neurogenesis and affects learning and memory
- Author
-
Minna Yli-Karjanmaa, Kathrine Solevad Larsen, Stephanie Lindeman Carlsen, Szymkowski, David E., Jane Stubbe, Lars Henrik Frich, Roberta Brambilla, and Kate L. Lambertsen
- Abstract
Background: Non-selective inhibition of TNF can cause suppression of the immune system and due to this inhibitors are used for long-term treatment of peripheral autoimmune diseases such as rheumatoid arthritis and Crohn’s disease. Inhibition of TNF is known to cause demyelination but otherwise little is known about the effects of long-term treatment in central nervous system (CNS) when CNS itself is not affected by an autoimmune disease or an insult. Since TNF signaling is associated with synaptic function and plasticity and there is evidence that it is involved in learning and memory, it is possible that long-term treatment with TNF inhibitors could alter hippocampal functions in otherwise healthy brain. Aim of the study: To test the effect of a long-term inhibition of TNF in learning and memory and cell proliferation in hippocampus. Methods: Adult male mice (C57BL6/J) were divided in three groups; treated with non-selective TNF inhibitor etanercept, selective soluble TNF (solTNF) inhibitor XPro1595 or saline. Drugs were administered 10 mg/kg subcutaneously every third day for two months. Spatial learning and memory were tested on Barnes maze after the treatment period. Proliferation marker BrdU was administered i.p. in the beginning of the treatment period and EdU before the Barnes maze test. The number of BrdU+ and EdU+ cells were counted in hippocampal dentate gyrus. Results: We found that non-selective inhibition of TNF with etanercept impairs learning and memory while animals treated with solTNF inhibitor XPro1595 express normal behaviour. Also number of BrdU+ cells was decreased after treatment with etanercept suggesting non-selective inhibition of TNF to alter neurogenesis in hippocampus. No differences in the number of EdU+ positive cells were seen between the treatment groups.Conclusion: Non-selective inhibition of TNF can impair learning and memory and decrease neurogenesis, while inhibiting only solTNF does not cause a decline in cognitive functions.
- Published
- 2018
40. High content analysis of phagocytic activity and cell morphology with PuntoMorph
- Author
-
Vanessa Ann Peters, Roberta Brambilla, Camilla Dalby-Hansen, Hassan Al-Ali, Han Gao, and Yan Shi
- Subjects
0301 basic medicine ,Phenotypic screening ,Phagocytosis ,Healthy tissue ,Biology ,Cell morphology ,Article ,Pattern Recognition, Automated ,03 medical and health sciences ,0302 clinical medicine ,Image processing ,Drug Discovery ,medicine ,Image Processing, Computer-Assisted ,Animals ,Cells, Cultured ,Fluorescent Dyes ,Cerebral Cortex ,Microglia ,Drug discovery ,General Neuroscience ,Morphometry ,High content analysis ,Phenotype ,Microspheres ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Microscopy, Fluorescence ,High-content screening ,Cell Body ,Immunology ,030217 neurology & neurosurgery ,Homeostasis ,Algorithms - Abstract
Background Phagocytosis is essential for maintenance of normal homeostasis and healthy tissue. As such, it is a therapeutic target for a wide range of clinical applications. The development of phenotypic screens targeting phagocytosis has lagged behind, however, due to the difficulties associated with image-based quantification of phagocytic activity. New method We present a robust algorithm and cell-based assay system for high content analysis of phagocytic activity. The method utilizes fluorescently labeled beads as a phagocytic substrate with defined physical properties. The algorithm employs statistical modeling to determine the mean fluorescence of individual beads within each image, and uses the information to conduct an accurate count of phagocytosed beads. In addition, the algorithm conducts detailed and sophisticated analysis of cellular morphology, making it a standalone tool for high content screening. Results We tested our assay system using microglial cultures. Our results recapitulated previous findings on the effects of microglial stimulation on cell morphology and phagocytic activity. Moreover, our cell-level analysis revealed that the two phenotypes associated with microglial activation, specifically cell body hypertrophy and increased phagocytic activity, are not highly correlated. This novel finding suggests the two phenotypes may be under the control of distinct signaling pathways. Comparison with existing methods We demonstrate that our assay system outperforms preexisting methods for quantifying phagocytic activity in multiple dimensions including speed, accuracy, and resolution. Conclusions We provide a framework to facilitate the development of high content assays suitable for drug screening. For convenience, we implemented our algorithm in a standalone software package, PuntoMorph.
- Published
- 2017
- Full Text
- View/download PDF
41. Predictors of Zero X-Ray Ablation for Supraventricular Tachycardias in a Nationwide Multicenter Experience
- Author
-
Antonio Pani, Belotti Giuseppina, Carlo Bonanno, Maria Grazia Bongiorni, Nicola Bottoni, Roberta Brambilla, Sergio de Ceglia, Paolo Della Bella, Giovanni de Vito, Daniele Malaspina, Endrj Menardi, Velia Napoli, Maria Silvia Negroni, Salvatore Ocello, Daniela Orsida, Claudio Pandozi, Stefano Pedretti, Diego Penela, Patrizia Pepi, Luca Rossi, Giovanni Rovaris, Alice Scopinaro, Antonello Vincenti, Graziana Viola, Valerio Zacà, Franco Zoppo, Pasquale Vergara, Sandra Badolati, Claudia Baiocchi, Sebastiano Belletti, Andrea Di Cori, Marco Galeazzi, Edoardo Gandolfi, Matteo Iori, Francesco Isola, Riccardo Massa, Giovanni Motta, Massimo Pala, Elena Piazzi, Fabio Quartieri, Luca Segreti, Antonello Vado, Gabriele Vignati, Pani, A, Belotti, G, Bonanno, C, Bongiorni, M, Bottoni, N, Brambilla, R, de Ceglia, S, Della Bella, P, de Vito, G, Malaspina, D, Menardi, E, Napoli, V, Negroni, M, Ocello, S, Orsida, D, Pandozi, C, Pedretti, S, Penela, D, Pepi, P, Rossi, L, Rovaris, G, Scopinaro, A, Vincenti, A, Viola, G, Zacà, V, Zoppo, F, and Vergara, P
- Subjects
Tachycardia ,Male ,medicine.medical_specialty ,workflow ,medicine.medical_treatment ,tachycardia, atrioventricular nodal reentry ,Catheter ablation ,radiation exposure ,030204 cardiovascular system & hematology ,tachycardia ,Radiation Dosage ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,0302 clinical medicine ,Imaging, Three-Dimensional ,Physiology (medical) ,Internal medicine ,X-rays ,medicine ,Tachycardia, Supraventricular ,Fluoroscopy ,Humans ,Prospective Studies ,Prospective cohort study ,atrioventricular nodal reentry ,catheter ablation ,fluoroscopy ,supraventricular ,medicine.diagnostic_test ,business.industry ,Hazard ratio ,Body Surface Potential Mapping ,Middle Aged ,Ablation ,Confidence interval ,Treatment Outcome ,Italy ,Surgery, Computer-Assisted ,Mapping system ,Cardiology ,Catheter Ablation ,Female ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Follow-Up Studies - Abstract
Background: This multicenter, prospective study evaluated the determinants of zero-fluoroscopy (ZFL) ablation of supraventricular tachycardias. Methods and Results: Four hundred thirty patients (215 male, 55.4±22.1 years) with indication to electrophysiological study or ablation of supraventricular tachycardias were enrolled. All participating physicians agreed to follow the as low as reasonably achievable policy. A procedure was defined as ZFL when no fluoroscopy was used. The total fluoroscopy time inversely correlated to the number of procedures previously performed by each operator since study start ( r =−0.112; P =0.02). Two hundred eighty-nine procedures (67.2%) were ZFL; multivariable analysis identified as predictors of ZFL: procedure after the 30th for each operator, compared with procedures up to the ninth ( P =0.011; hazard ratio, 3.49; 95% confidence interval [CI], 1.79–6.80); the type of arrhythmia ( P =0.031; electrophysiological study and atrioventricular nodal reentry tachycardia ablation having the highest probability of ZFL; hazard ratio, 6.87; 95% CI, 2.08–22.7 and hazard ratio, 2.02; 95% CI, 1.04–3.91, respectively); the operator’s ( P =0.002) and patient’s age ( P =0.009). Among operators, achievement of ZFL varied from 0% to 100%; 8 (22.8%) operators achieved ZFL in 75% of their procedures. The probability of ZFL increased by 2.8% (hazard ratio, 0.98; 95% CI, 0.97–0.99) as patient’s age decreased by 1 year. Acute procedural success was obtained in all cases. Conclusions: The use of 3-dimensional mapping system completely avoided the use of fluoroscopy in most cases, with very low fluoroscopy time in the remaining and high safety and effectiveness profiles. Achievement of ZFL was predicted by the type of arrhythmia, operator’s experience, and patient’s age.
- Published
- 2017
42. Mitochondrial DNA Double-Strand Breaks in Oligodendrocytes Cause Demyelination, Axonal Injury, and CNS Inflammation
- Author
-
Roberta Brambilla, Mehran Taherian, Dmitry Ivanov, Han Gao, Galina Dvoriantchikova, Milena F. Pinto, Carlos T. Moraes, Stephanie McCarthy, Shreyans Patel, Kenji F. Tanaka, Claudia V. Pereira, Shaffiat Karmally, and Pernille M. Madsen
- Subjects
0301 basic medicine ,Central Nervous System ,Male ,Axonal loss ,Mitochondrion ,DNA, Mitochondrial/genetics ,Nerve Degeneration/genetics ,Mice ,0302 clinical medicine ,Inflammation/genetics ,DNA Breaks, Double-Stranded ,Research Articles ,General Neuroscience ,Experimental autoimmune encephalomyelitis ,Mitochondria ,Oligodendroglia/pathology ,Oligodendroglia ,medicine.anatomical_structure ,Locomotion/physiology ,Central Nervous System/pathology ,Female ,Demyelination ,Locomotion ,Mitochondrial DNA ,Encephalomyelitis, Autoimmune, Experimental ,Encephalomyelitis, Autoimmune, Experimental/genetics ,Demyelinating Diseases/genetics ,Mice, Transgenic ,Biology ,Neuroprotection ,DNA, Mitochondrial ,Multiple sclerosis ,03 medical and health sciences ,medicine ,Animals ,Animal model ,Oxidative phosphorylation ,Remyelination ,Inflammation ,medicine.disease ,Oligodendrocyte ,Axons ,Mice, Inbred C57BL ,030104 developmental biology ,Axons/pathology ,Nerve Degeneration ,Neuroscience ,030217 neurology & neurosurgery ,Demyelinating Diseases - Abstract
Mitochondrial dysfunction has been implicated in the pathophysiology of neurodegenerative disorders, including multiple sclerosis (MS). To date, the investigation of mitochondrial dysfunction in MS has focused exclusively on neurons, with no studies exploring whether dysregulation of mitochondrial bioenergetics and/or genetics in oligodendrocytes might be associated with the etiopathogenesis of MS and other demyelinating syndromes. To address this question, we established a mouse model where mitochondrial DNA (mtDNA) double-strand breaks (DSBs) were specifically induced in myelinating oligodendrocytes (PLP:mtPstI mice) by expressing a mitochondrial-targeted endonuclease, mtPstI, starting at 3 weeks of age. In both female and male mice, DSBs of oligodendroglial mtDNA caused impairment of locomotor function, chronic demyelination, glial activation, and axonal degeneration, which became more severe with time of induction. In addition, after short transient induction of mtDNA DSBs, PLP:mtPstI mice showed an exacerbated response to experimental autoimmune encephalomyelitis. Together, our data demonstrate that mtDNA damage can cause primary oligodendropathy, which in turn triggers demyelination, proving PLP:mtPstI mice to be a useful tool to study the pathological consequences of mitochondrial dysfunction in oligodendrocytes. In addition, the demyelination and axonal loss displayed by PLP:mtPstI mice recapitulate some of the key features of chronic demyelinating syndromes, including progressive MS forms, which are not accurately reproduced in the models currently available. For this reason, the PLP:mtPstI mouse represents a unique and much needed platform for testing remyelinating therapies.SIGNIFICANCE STATEMENTIn this study, we show that oligodendrocyte-specific mitochondrial DNA double-strand breaks in PLP:mtPstI mice cause oligodendrocyte death and demyelination associated with axonal damage and glial activation. Hence, PLP:mtPstI mice represent a unique tool to study the pathological consequences of mitochondrial dysfunction in oligodendrocytes, as well as an ideal platform to test remyelinating and neuroprotective agents.
- Published
- 2017
- Full Text
- View/download PDF
43. Topical administration of TNF-inhibitor XPro1595 decreases infarct volume after experimental stroke
- Author
-
Minna Liisa Kyllikki Yli-Karjanmaa, Bettina Hjelm Clausen, Hans Gram Novrup, Szymkowski, David E., Morten Meyer, Roberta Brambilla, and Lambertsen, Kate L.
- Published
- 2017
44. Neuronal Ablation of IKK2 Decreases Lesion Size and Improves Functional Outcome after Spinal Cord Injury in Mice
- Author
-
Ditte Gry Ellman, Hans Gram Novrup, Louise Helskov Jørgensen, Minna Christiansen Lund, Minna Liisa Kyllikki Yli-Karjanmaa, Pernille Marie Madsen, Jonas Heinrich Vienhues, Safinaz Dursun, Bethea, John R., Karin Lykke-Hartmann, Roberta Brambilla, and Kate L. Lambertsen
- Subjects
Article - Abstract
Nuclear factor-kappa B (NF-κB) is a key modulator of inflammation and secondary injury responses in neurodegenerative disease, including spinal cord injury (SCI). Inhibition of astroglial NF-κB reduces inflammation, enhances oligodendrogenesis and improves functional recovery after SCI, however the contribution of neuronal NF-κB to secondary inflammatory responses following SCI has yet to be investigated. We demonstrate that conditional ablation of IKK2 in Synapsin 1-expressing neurons in mice (Syn1creIKK2fl/fl) reduces activation of the classical NF-κB signaling pathway, resulting in impaired motor function and altered memory retention under naïve conditions. Following induction of a moderate SCI phosphorylated NF-κB levels decreased in the spinal cord of Syn1creIKK2fl/fl mice compared to controls, resulting in improvement in functional recovery. Histologically, Syn1creIKK2fl/fl mice exhibited reduced lesion volume but comparable microglial/leukocyte responses after SCI. In parallel, interleukin (IL)-1β expression was significantly decreased within the lesioned spinal cord, whereas IL-5, IL-6, IL-10, tumor necrosis factor (TNF) and chemokine (C-X-C motif) ligand 1 were unchanged compared to control mice. We conclude that conditional ablation of IKK2 in neurons, resulting in reduced neuronal NF-B signaling, and lead to protective effects after SCI and propose the neuronal classical NF-κB pathway as a potential target for the development of new therapeutic, neuroprotective strategies for SCI.
- Published
- 2017
45. Neuropathic pain-induced depressive-like behavior and hippocampal neurogenesis and plasticity are dependent on TNFR1 signaling
- Author
-
Spencer Summers, Winston M. Walters, Roberta Brambilla, Mariagrazia Grilli, Anna Dellarole, John R. Bethea, Danielle Bernardes, and Paul D. Morton
- Subjects
medicine.medical_specialty ,Hot Temperature ,Anhedonia ,Neurogenesis ,Immunology ,Drinking Behavior ,Hippocampal formation ,Hippocampus ,Receptors, Tumor Necrosis Factor ,Article ,Food Preferences ,Mice ,Sciatica ,Behavioral Neuroscience ,Myelin ,Internal medicine ,Neuroplasticity ,Pressure ,medicine ,Animals ,Single-Blind Method ,Mice, Knockout ,Neuronal Plasticity ,Depression ,Tumor Necrosis Factor-alpha ,Endocrine and Autonomic Systems ,Sciatic Nerve ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Endocrinology ,Hyperalgesia ,Receptors, Tumor Necrosis Factor, Type I ,Neuropathic pain ,Exploratory Behavior ,Neuralgia ,Tumor necrosis factor alpha ,Sciatic nerve ,medicine.symptom ,Corticosterone ,Psychology ,Neuroscience ,Signal Transduction - Abstract
Patients suffering from neuropathic pain have a higher incidence of mood disorders such as depression. Increased expression of tumor necrosis factor (TNF) has been reported in neuropathic pain and depressive-like conditions and most of the pro-inflammatory effects of TNF are mediated by the TNF receptor 1 (TNFR1). Here we sought to investigate: (1) the occurrence of depressive-like behavior in chronic neuropathic pain and the associated forms of hippocampal plasticity, and (2) the involvement of TNFR1-mediated TNF signaling as a possible regulator of such events. Neuropathic pain was induced by chronic constriction injury of the sciatic nerve in wild-type and TNFR1(-/-) mice. Anhedonia, weight loss and physical state were measured as symptoms of depression. Hippocampal neurogenesis, neuroplasticity, myelin remodeling and TNF/TNFRs expression were analyzed by immunohistochemical analysis and western blot assay. We found that neuropathic pain resulted in the development of depressive symptoms in a time dependent manner and was associated with profound hippocampal alterations such as impaired neurogenesis, reduced expression of neuroplasticity markers and myelin proteins. The onset of depressive-like behavior also coincided with increased hippocampal levels of TNF, and decreased expression of TNF receptor 2 (TNFR2), which were all fully restored after mice spontaneously recovered from pain. Notably, TNFR1(-/-) mice did not develop depressive-like symptoms after injury, nor were there changes in hippocampal neurogenesis and plasticity. Our data show that neuropathic pain induces a cluster of depressive-like symptoms and profound hippocampal plasticity that are dependent on TNF signaling through TNFR1.
- Published
- 2014
- Full Text
- View/download PDF
46. Fibrotic scar after experimental autoimmune encephalomyelitis inhibits oligodendrocyte differentiation
- Author
-
Jae K. Lee, Roberta Brambilla, Jiajun Li, Han Gao, Stephanie L. Yahn, and Irene Goo
- Subjects
0301 basic medicine ,Male ,Myeloid ,Encephalomyelitis, Autoimmune, Experimental ,Mice, Transgenic ,Biology ,OPCs ,Article ,lcsh:RC321-571 ,Extracellular matrix ,03 medical and health sciences ,Myelination ,0302 clinical medicine ,medicine ,Demyelinating disease ,Animals ,Myeloid Cells ,Perivascular fibroblasts ,Remyelination ,Fibroblast ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,030304 developmental biology ,0303 health sciences ,EAE ,Multiple sclerosis ,Experimental autoimmune encephalomyelitis ,Oligodendrocyte differentiation ,Cell Differentiation ,MS ,Fibroblasts ,medicine.disease ,Fibrosis ,White Matter ,Mice, Inbred C57BL ,Oligodendroglia ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Spinal Cord ,Cancer research ,Fibrotic scar ,030217 neurology & neurosurgery - Abstract
Remyelination failure is a crucial component of disease progression in the autoimmune demyelinating disease Multiple Sclerosis (MS). The regenerative capacity of oligodendrocyte progenitor cells (OPCs) to replace myelinating oligodendrocytes is likely influenced by many aspects of the lesion environment including inflammatory signaling and extracellular matrix (ECM) deposition. These features of MS lesions are typically attributed to infiltrating leukocytes and reactive astrocytes. Here we demonstrate that fibroblasts also contribute to the inhibitory environment in the animal model of MS, experimental autoimmune encephalomyelitis (EAE). Using Col1α1GFPtransgenic mice, we show that perivascular fibroblasts are activated in the spinal cord at EAE onset, and infiltrate the parenchyma by the peak of behavioral deficits where they are closely associated with areas of demyelination, myeloid cell accumulation, and ECM deposition. We further show that both fibroblast conditioned media and fibroblast ECM inhibit the differentiation of OPCs into mature oligodendrocytes. Taken together, our results indicate that the fibrotic scar is a major component of EAE pathology that leads to an inhibitory environment for remyelination, thus raising the possibility that anti-fibrotic mechanisms may serve as novel therapeutic targets for MS.
- Published
- 2019
47. Astrocytes play a key role in EAE pathophysiology by orchestrating in the CNS the inflammatory response of resident and peripheral immune cells and by suppressing remyelination
- Author
-
Jessica Jopek Ashbaugh, Paul D. Morton, Roberta Brambilla, John R. Bethea, Kate Lykke Lambertsen, and Shaffiat Karmally
- Subjects
biology ,Experimental autoimmune encephalomyelitis ,Inflammation ,medicine.disease ,Myelin oligodendrocyte glycoprotein ,Astrogliosis ,Cellular and Molecular Neuroscience ,Myelin ,Immune system ,medicine.anatomical_structure ,Neurology ,Immunology ,medicine ,biology.protein ,medicine.symptom ,Remyelination ,Neuroinflammation - Abstract
Astrocytes respond to insult with a process of cellular activation known as reactive astrogliosis. One of the key signals regulating this phenomenon is the transcription factor nuclear factor-kappa B (NF-κB), which is responsible for modulating inflammation, cell survival, and cell death. In astrocytes, following trauma or disease, the expression of NF-κB-dependent genes is highly activated. We previously demonstrated that inactivation of astroglial NF-κB in vivo (GFAP-IκBα-dn mice) leads to improved functional outcome in experimental autoimmune encephalomyelitis (EAE), and this is accompanied by reduction of pro-inflammatory gene expression in the CNS. Here we extend our studies to show that recovery from EAE in GFAP-IκBα-dn mice is associated with reduction of peripheral immune cell infiltration into the CNS at the chronic phase of EAE. This is not dependent on a less permeable blood-brain barrier, but rather on a reduced immune cell mobilization from the periphery. Furthermore, once inside the CNS, the ability of T cells to produce pro-inflammatory cytokines is diminished during acute disease. In parallel, we found that the number of total and activated microglial cells is reduced, suggesting that functional improvement in GFAP-IκBα-dn mice is dependent upon reduction of the overall inflammatory response within the CNS sustained by both resident and infiltrating cells. This results in preservation of myelin compaction and enhanced remyelination, as shown by electron microscopy analysis of the spinal cord. Collectively our data indicate that astrocytes are key players in driving CNS inflammation and are directly implicated in the pathophysiology of EAE, since blocking their pro-inflammatory capability results in protection from the disease.
- Published
- 2013
- Full Text
- View/download PDF
48. Prolonged stimulation of a brainstem raphe region attenuates experimental autoimmune encephalomyelitis
- Author
-
Melissa M. Carballosa-Gautam, Stephanie S. Sloley, Pernille M. Madsen, Ian D. Hentall, Alberto Vitores, and Roberta Brambilla
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Encephalomyelitis, Autoimmune, Experimental ,Multiple Sclerosis ,Gene Expression ,Stimulation ,Electric Stimulation Therapy ,multiple sclerosis ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Internal medicine ,nucleus raphe magnus ,medicine ,Microstimulation ,Animals ,Myelin Sheath ,Nucleus raphe magnus ,Inflammation ,biology ,Raphe ,business.industry ,General Neuroscience ,Experimental autoimmune encephalomyelitis ,myelination ,medicine.disease ,Oligodendrocyte ,cytokines ,Electric Stimulation ,Myelin basic protein ,deep brain stimulation ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Spinal Cord ,Immunology ,biology.protein ,Cytokines ,Raphe Nuclei ,Female ,Raphe nuclei ,business ,030217 neurology & neurosurgery - Abstract
Multiple sclerosis (MS), a neuroinflammatory disease, has few treatment options, none entirely adequate. We studied whether prolonged electrical microstimulation of a hindbrain region (the nucleus raphe magnus) can attenuate experimental autoimmune encephalomyelitis, a murine model of MS induced by MOG35-55 injection. Eight days after symptoms emerged, a wireless electrical stimulator with an attached microelectrode was implanted cranially, and daily intermittent stimulation was begun in awake, unrestrained mice. The thoracic spinal cord was analyzed for changes in histology (on day 29) and gene expression (on day 37), with a focus on myelination and cytokine production. Controls, with inactive implants, showed a phase of disease exacerbation on days 19–25 that stimulation for >16 days eliminated. Prolonged stimulation also reduced numbers of infiltrating immune cells and increased numbers of myelinated axons. It additionally lowered genetic expression of some pro-inflammatory cytokines (interferon gamma and tumor necrosis factor) and platelet-derived growth factor receptor alpha, a marker of oligodendrocyte precursors, while raising expression of myelin basic protein. Studies of restorative treatments for MS might profitably consider ways to stimulate the raphe magnus, directly or via its inputs, or to emulate its serotonergic and peptidergic output.
- Published
- 2016
- Full Text
- View/download PDF
49. Conditional ablation of myeloid TNF increases lesion volume after experimental stroke in mice, possibly via altered ERK1/2 signaling
- Author
-
Kate Lykke Lambertsen, Sergei A. Nedospasov, Mithula Sivasaravanaparan, Torben Fogtmann, Maria Gammelstrup Andersen, Han Gao, Jae K. Lee, Bjarne Winther Kristensen, Tomas Deierborg, Svend Hvidsten, Roberta Brambilla, Michelle Trojanowsky, Bente Finsen, Matilda Degn, Bettina Hjelm Clausen, Christina Baun, Morten Meyer, and Sara Thornby Bak
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Male ,medicine.medical_treatment ,Interleukin-1beta ,Interleukin-6/metabolism ,MAP Kinase Signaling System/physiology ,Brain Ischemia ,Neuroprotection/physiology ,Mice ,0302 clinical medicine ,Leukocytes ,Myeloid Cells ,Mice, Knockout ,Tumor Necrosis Factor-alpha/metabolism ,Multidisciplinary ,Microglia ,Inflammation/metabolism ,Cytokines/metabolism ,Myeloid Cells/metabolism ,Neuroprotection ,Stroke ,CXCL1 ,medicine.anatomical_structure ,Cytokine ,Knockout mouse ,Leukocytes/metabolism ,Cytokines ,Tumor necrosis factor alpha ,Signal Transduction ,medicine.medical_specialty ,MAP Kinase Signaling System ,Ischemia ,Article ,Microglia/metabolism ,03 medical and health sciences ,Internal medicine ,medicine ,Animals ,Neuroinflammation ,Inflammation ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,business.industry ,Signal Transduction/physiology ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Stroke/metabolism ,Interleukin-5/metabolism ,Immunology ,Interleukin-1beta/metabolism ,Brain Ischemia/metabolism ,Interleukin-5 ,business ,030217 neurology & neurosurgery - Abstract
Microglia are activated following cerebral ischemia and increase their production of the neuro- and immunomodulatory cytokine tumor necrosis factor (TNF). To address the function of TNF from this cellular source in focal cerebral ischemia we used TNF conditional knock out mice (LysMcreTNFfl/fl) in which the TNF gene was deleted in cells of the myeloid lineage, including microglia. The deletion reduced secreted TNF levels in lipopolysaccharide-stimulated cultured primary microglia by ~93%. Furthermore, phosphorylated-ERK/ERK ratios were significantly decreased in naïve LysMcreTNFfl/fl mice demonstrating altered ERK signal transduction. Micro-PET using 18[F]-fluorodeoxyglucose immediately after focal cerebral ischemia showed increased glucose uptake in LysMcreTNFfl/fl mice, representing significant metabolic changes, that translated into increased infarct volumes at 24 hours and 5 days compared to littermates (TNFfl/fl). In naïve LysMcreTNFfl/fl mice cytokine levels were low and comparable to littermates. At 6 hours, TNF producing microglia were reduced by 56% in the ischemic cortex in LysMcreTNFfl/fl mice compared to littermate mice, whereas no TNF+ leukocytes were detected. At 24 hours, pro-inflammatory cytokine (TNF, IL-1β, IL-6, IL-5 and CXCL1) levels were significantly lower in LysMcreTNFfl/fl mice, despite comparable infiltrating leukocyte populations. Our results identify microglial TNF as beneficial and neuroprotective in the acute phase and as a modulator of neuroinflammation at later time points after experimental ischemia, which may contribute to regenerative recovery.
- Published
- 2016
- Full Text
- View/download PDF
50. Pioglitazone ameliorates the phenotype of a novel Parkinson’s disease mouse model by reducing neuroinflammation
- Author
-
Francisca Diaz, Nadee Nissanka, Carlos T. Moraes, Roberta Brambilla, Milena F. Pinto, and Susana Peralta
- Subjects
0301 basic medicine ,Agonist ,medicine.medical_specialty ,Parkinson's disease ,medicine.drug_class ,Dopamine ,Clinical Neurology ,Mice, Transgenic ,Substantia nigra ,Striatum ,Midbrain ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Neuroinflammation ,Mesencephalon ,Internal medicine ,medicine ,Animals ,Molecular Biology ,Inflammation ,Mice, Knockout ,Pioglitazone ,business.industry ,Dopaminergic Neurons ,Dopaminergic ,Parkinson Disease ,medicine.disease ,Corpus Striatum ,Mitochondria ,3. Good health ,Substantia Nigra ,Disease Models, Animal ,Phenotype ,030104 developmental biology ,Endocrinology ,nervous system ,Thiazolidinediones ,Neurology (clinical) ,business ,Neuroscience ,030217 neurology & neurosurgery ,Research Article ,medicine.drug - Abstract
Background Parkinson’s disease (PD) is a progressive neurodegenerative disorder characterized by motor and non-motor symptoms. The cause of the motor symptoms is the loss of dopaminergic neurons in the substantia nigra with consequent depletion of dopamine in the striatum. Although the etiology of PD is unknown, mitochondrial dysfunctions, including cytochrome c oxidase (Complex IV) impairment in dopaminergic neurons, have been associated with the disease’s pathophysiology. In order to analyze the role of Complex IV in PD, we knocked out Cox10 (essential for the maturation of COXI, a catalytic subunit of Complex IV) in dopaminergic neurons. We also tested whether the resulting phenotype was improved by stimulating the PPAR-γ pathway. Results Cox10/DAT-cre mice showed decreased numbers of TH+ and DAT+ cells in the substantia nigra, early striatal dopamine depletion, motor defects reversible with L-DOPA treatment and hypersensitivity to L-DOPA with hyperkinetic behavior. We found that chronic pioglitazone (PPAR-γ agonist) treatment ameliorated the motor phenotype in Cox10/DAT-cre mice. Although neither mitochondrial function nor the number of dopaminergic neurons was improved, neuroinflammation in the midbrain and the striatum was decreased. Conclusions By triggering a mitochondrial Complex IV defect in dopaminergic neurons, we created a new mouse model resembling the late stages of PD with massive degeneration of dopaminergic neurons and striatal dopamine depletion. The motor phenotypes were improved by Pioglitazone treatment, suggesting that targetable secondary pathways can influence the development of certain forms of PD. Electronic supplementary material The online version of this article (doi:10.1186/s13024-016-0090-7) contains supplementary material, which is available to authorized users.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.