Dectin-1 limits autoimmune neuroinflammation and promotes myeloid cell-astrocyte crosstalk via Card9-independent expression of Oncostatin M
Deerhake, ME;Danzaki, K;Inoue, M;Cardakli, ED;Nonaka, T;Aggarwal, N;Barclay, WE;Ji, RR;Shinohara, ML;
PMID: 33581044 | DOI: 10.1016/j.immuni.2021.01.004
Pathologic roles of innate immunity in neurologic disorders are well described, but their beneficial aspects are less understood. Dectin-1, a C-type lectin receptor (CLR), is largely known to induce inflammation. Here, we report that Dectin-1 limited experimental autoimmune encephalomyelitis (EAE), while its downstream signaling molecule, Card9, promoted the disease. Myeloid cells mediated the pro-resolution function of Dectin-1 in EAE with enhanced gene expression of the neuroprotective molecule, Oncostatin M (Osm), through a Card9-independent pathway, mediated by the transcription factor NFAT. Furthermore, we find that the Osm receptor (OsmR) functioned specifically in astrocytes to reduce EAE severity. Notably, Dectin-1 did not respond to heat-killed Mycobacteria, an adjuvant to induce EAE. Instead, endogenous Dectin-1 ligands, including galectin-9, in the central nervous system (CNS) were involved to limit EAE. Our study reveals a mechanism of beneficial myeloid cell-astrocyte crosstalk regulated by a Dectin-1 pathway and identifies potential therapeutic targets for autoimmune neuroinflammation.
Central and peripheral GLP-1 systems independently suppress eating
Brierley, DI;Holt, MK;Singh, A;de Araujo, A;McDougle, M;Vergara, M;Afaghani, MH;Lee, SJ;Scott, K;Maske, C;Langhans, W;Krause, E;de Kloet, A;Gribble, FM;Reimann, F;Rinaman, L;de Lartigue, G;Trapp, S;
PMID: 33589843 | DOI: 10.1038/s42255-021-00344-4
The anorexigenic peptide glucagon-like peptide-1 (GLP-1) is secreted from gut enteroendocrine cells and brain preproglucagon (PPG) neurons, which, respectively, define the peripheral and central GLP-1 systems. PPG neurons in the nucleus tractus solitarii (NTS) are widely assumed to link the peripheral and central GLP-1 systems in a unified gut-brain satiation circuit. However, direct evidence for this hypothesis is lacking, and the necessary circuitry remains to be demonstrated. Here we show that PPGNTS neurons encode satiation in mice, consistent with vagal signalling of gastrointestinal distension. However, PPGNTS neurons predominantly receive vagal input from oxytocin-receptor-expressing vagal neurons, rather than those expressing GLP-1 receptors. PPGNTS neurons are not necessary for eating suppression by GLP-1 receptor agonists, and concurrent PPGNTS neuron activation suppresses eating more potently than semaglutide alone. We conclude that central and peripheral GLP-1 systems suppress eating via independent gut-brain circuits, providing a rationale for pharmacological activation of PPGNTS neurons in combination with GLP-1 receptor agonists as an obesity treatment strategy.
Diet-dependent regulation of TGFβ impairs reparative innate immune responses after demyelination
Bosch-Queralt, M;Cantuti-Castelvetri, L;Damkou, A;Schifferer, M;Schlepckow, K;Alexopoulos, I;Lütjohann, D;Klose, C;Vaculčiaková, L;Masuda, T;Prinz, M;Monroe, KM;Di Paolo, G;Lewcock, JW;Haass, C;Simons, M;
PMID: 33619376 | DOI: 10.1038/s42255-021-00341-7
Proregenerative responses are required for the restoration of nervous-system functionality in demyelinating diseases such as multiple sclerosis (MS). Yet, the limiting factors responsible for poor CNS repair are only partially understood. Here, we test the impact of a Western diet (WD) on phagocyte function in a mouse model of demyelinating injury that requires microglial innate immune function for a regenerative response to occur. We find that WD feeding triggers an ageing-related, dysfunctional metabolic response that is associated with impaired myelin-debris clearance in microglia, thereby impairing lesion recovery after demyelination. Mechanistically, we detect enhanced transforming growth factor beta (TGFβ) signalling, which suppresses the activation of the liver X receptor (LXR)-regulated genes involved in cholesterol efflux, thereby inhibiting phagocytic clearance of myelin and cholesterol. Blocking TGFβ or promoting triggering receptor expressed on myeloid cells 2 (TREM2) activity restores microglia responsiveness and myelin-debris clearance after demyelinating injury. Thus, we have identified a druggable microglial immune checkpoint mechanism regulating the microglial response to injury that promotes remyelination.
Human neutralizing antibodies against SARS-CoV-2 require intact Fc effector functions for optimal therapeutic protection
Winkler, E;Gilchuk, P;Yu, J;Bailey, A;Chen, R;Chong, Z;Zost, S;Jang, H;Huang, Y;Allen, J;Case, J;Sutton, R;Carnahan, R;Darling, T;Boon, A;Mack, M;Head, R;Ross, T;Crowe, J;Diamond, M;
| DOI: 10.1016/j.cell.2021.02.026
SARS-CoV-2 has caused the global COVID-19 pandemic. Although passively delivered neutralizing antibodies against SARS-CoV-2 show promise in clinical trials, their mechanism of action in vivo is incompletely understood. Here we define correlates of protection of neutralizing human monoclonal antibodies (mAbs) in SARS-CoV-2-infected animals. Whereas Fc effector functions are dispensable when representative neutralizing mAbs are administered as prophylaxis, they are required for optimal protection as therapy. When given after infection, intact mAbs reduce SARS-CoV-2 burden and lung disease in mice and hamsters better than loss-of-function Fc variant mAbs. Fc engagement of neutralizing antibodies mitigates inflammation and improves respiratory mechanics, and transcriptional profiling suggests these phenotypes are associated with diminished innate immune signaling and preserved tissue repair. Immune cell depletions establish that neutralizing mAbs require monocytes and CD8+ T cells for optimal clinical and virological benefit. Thus, potently neutralizing mAbs utilize Fc effector functions during therapy to mitigate lung infection and disease.
Ventral arkypallidal neurons inhibit accumbal firing to promote reward consumption
Vachez, YM;Tooley, JR;Abiraman, K;Matikainen-Ankney, B;Casey, E;Earnest, T;Ramos, LM;Silberberg, H;Godynyuk, E;Uddin, O;Marconi, L;Le Pichon, CE;Creed, MC;
PMID: 33495635 | DOI: 10.1038/s41593-020-00772-7
The nucleus accumbens shell (NAcSh) and the ventral pallidum (VP) are critical for reward processing, although the question of how coordinated activity within these nuclei orchestrates reward valuation and consumption remains unclear. Inhibition of NAcSh firing is necessary for reward consumption, but the source of this inhibition remains unknown. Here, we report that a subpopulation of VP neurons, the ventral arkypallidal (vArky) neurons, project back to the NAcSh, where they inhibit NAcSh neurons in vivo in mice. Consistent with this pathway driving reward consumption via inhibition of the NAcSh, calcium activity of vArky neurons scaled with reward palatability (which was dissociable from reward seeking) and predicted the subsequent drinking behavior during a free-access paradigm. Activation of the VP-NAcSh pathway increased ongoing reward consumption while amplifying hedonic reactions to reward. These results establish a pivotal role for vArky neurons in the promotion of reward consumption through modulation of NAcSh firing in a value-dependent manner.
Specific Detection of Prostate Cancer Cells in Urine by RNA in Situ Hybridization
Eskra, JN;Rabizadeh, D;Zhang, J;Isaacs, WB;Luo, J;Pavlovich, CP;
PMID: 33617332 | DOI: 10.1097/JU.0000000000001691
Noninvasive tests that can accurately detect prostate cancer are urgently needed for prostate cancer diagnosis, surveillance, and prognosis. Exfoliated prostate cells captured in urine represent a promising resource for noninvasive detection of prostate cancer. We investigated performance of a novel cell-based urine test for detection of clinically significant prostate cancer. We previously developed a multiplex RNA in situ hybridization (RISH) assay targeting NKX3-1, PRAC1 and PCA3 that enables identification and quantification of malignant and benign prostate cells released into urine. We investigated application of the assay for prostate cancer detection in a cohort of 98 patients suspected of harboring prostate cancer. Urine was collected following digital rectal exam and the sediment was isolated and evaluated by RISH. Samples were scored based on cellular expression of RISH targets. Cells of prostate origin were defined by positivity for NKX3-1 and/or PRAC1, and prostate cancer cells by positivity for PCA3. Prostate cells (NKX3-1/PRAC1+ cells) were detected in 69 samples, among which 20 were positive for PCA3 (i.e., positive for prostate cancer cells). Comparison of RISH results with biopsy outcome and clinical variables revealed that positivity for cancer by RISH significantly correlated with intermediate/high risk cancer (p=0.003), PSA density (p=0.022), significant disease (p <0.0001), and Gleason score (p=0.003). The test was 95% specific and 51% sensitive for detection of clinically significant prostate cancer. Identification of exfoliated prostate cancer cells in urine by RISH provides a novel tool for highly specific and noninvasive detection of prostate cancer.
Plastic and reconstructive surgery
Stanton, E;Feng, J;Kondra, K;Sanchez, J;Jimenez, C;Brown, KS;Skiles, ML;Urata, MM;Chai, Y;Hammoudeh, JA;
PMID: 37224290 | DOI: 10.1097/PRS.0000000000010754
The standard graft material for alveolar cleft repair (ACR) is autogenous iliac crest. However, a promising alternative potential graft adjunct - newborn human umbilical cord mesenchymal stem cells (h-UCMSC) - has yet to be explored in vivo. Their capacity for self-renewal, multipotent differentiation, and proliferation allows h-UCMSC to be harnessed for regenerative medicine. Our study seeks to evaluate the efficacy of using tissue-derived h-UCMSC and their osteogenic capabilities in a murine model to improve ACR.Foxn1 mice were separated into three groups with the following calvarial defects: (1) no-treatment (empty defect; n=6), (2) poly (D,L-lactide-co-glycolide) (PLGA) scaffold (n=6), and (3) h-UCMSC with PLGA (n=4). Bilateral 2-mm diameter parietal bone critical-sized defects were created using a dental drill. Micro-CT imaging occurred at 1, 2, 3, and 4 weeks postoperatively. The mice were euthanized 4 weeks postoperatively for RNAscope analysis, immunohistochemistry, and histology.No mice experienced complications during the follow-up period. Micro-CT and histology demonstrated that the no-treatment (1) and PLGA-only (2) defects remained patent without significant defect size differences across groups. In contrast, the h-UCMSC with PLGA group (3) had significantly greater bone fill on micro-CT and histology.We demonstrate a successful calvarial defect model for the investigation of h-UCMSC-mediated osteogenesis and bone repair. Furthermore, evidence reveals that PLGA alone has neither short-term effects on bone formation nor any unwanted side effects, making it an attractive scaffold. Further investigation using h-UCMSC with PLGA in larger animals is warranted to advance future translation to patients requiring ACR.Our results demonstrate a successful murine calvarial defect model for the investigation of h-UCMSC-mediated osteogenesis and bone repair and provide preliminary evidence for the safe and efficacious use of this graft adjunct in alveolar cleft repair.
Pearce, L;He, D;Davidson, SM;Yellon, DM;
| DOI: 10.1161/circ.146.suppl_1.11926
Introduction: Myocardial infarction causes several types of injury to the myocardium including lethal cell injury and ‘no-reflow’ (NRF) /microvascular obstruction (MVO). Nonselective Rho Kinase (ROCK1/2) inhibitors such as Fasudil, ameliorate myocardial ischemia/reperfusion (I/R) injury but cause unwanted hypotension. Selective ROCK2 inhibitors (e.g.: KD025) are safe in clinical trials without causing haemodynamic compromise, however they have not been investigated in myocardial I/R. ROCK inhibitors prevent vascular smooth muscle cell (VSMC) contraction; such VSMC contraction/coronary spasm being features of ‘no-reflow’ (NRF) and microvascular obstruction (MVO). At present, there are limited therapies to improve ischemic MVO outcomes, and prognosis is poor. Hypothesis: Using a rat model, we hypothesised that, i) ROCK2 mRNA is expressed in myocardium and coronary vasculature and ii) The selective ROCK2 inhibitor KD025, would reduce infarct size (IS%) and NRF% (MVO) following I/R. Methods: RNA scope in-situ hybridisation was performed with a fluorescent, multiplex assay for ROCK1/2 & VSMC mRNA in myocardium and coronary vasculature. Male SD rats underwent in-vivo myocardial infarction with 30min ischemia, 180min reperfusion. 15min prior to reperfusion, the ROCK inhibitors Fasudil and KD025 or vehicle (DMSO) were administered i.p. For IS%, myocardium was stained with TTC, and regions not perfused with 1.5% Thioflavin S (NRF%), were visualised under UV light. Results: RNAscope confirmed the presence of ROCK2 mRNA within myocardium and VSMC of coronary arteries. Fasudil (10mg/kg) vs control significantly reduced regional IS% (30.3±4.4 vs52.9±3.8,p=0.02, n=15) and area of NRF% (12.4±2.8 vs28.6±2.2, p=0.001, n=15). However, there was significant hypotension;- Mean BP (mmHg) (72±3.9) vs control (84±2.3, p=0.007). KD025 (100mg/kg) did not reduce IS%, but significantly reduced the area of NRF% vs control (18.4±2.8 vs28.6±2.2,p=0.02, n=14) without hypotensive effect. Conclusions: Our results suggest that ROCK2 may be a prospective target in the management of coronary circulation reperfusion injury and ischemic MVO.
Journal of Oral and Maxillofacial Surgery
Stanton, E;Sanchez, J;Kondra, K;Jimenez, C;Urata, M;Hammoudeh, J;
| DOI: 10.1016/j.joms.2022.07.012
Background: The standard graft material for alveolar cleft repair (ACR) is autogenous iliac crest. However, a promising alternative potential graft adjunct - newborn human umbilical cord mesenchymal stem cells (h-UCMSC) - has yet to be explored in vivo. Their capacity for selfrenewal, multipotent differentiation, and proliferation allows h-UCMSC to be harnessed for regenerative medicine. Our study seeks to evaluate the efficacy of using tissue-derived hUCMSC and their osteogenic capabilities in a murine model to improve ACR. Methods: Foxn1 mice were separated into three groups with the following calvarial defects: (1) no-treatment (empty defect; n=6), (2) poly (D,L-lactide-co-glycolide) (PLGA) scaffold (n=6), and (3) h-UCMSC with PLGA (n=4). Bilateral 2-mm diameter parietal bone critical-sized defects were created using a dental drill. Micro-CT imaging occurred at 1, 2, 3, and 4 weeks postoperatively. The mice were euthanized 4 weeks postoperatively for RNAscope analysis, immunohistochemistry, and histology. Results: No mice experienced complications during the follow-up period. Micro-CT and histology demonstrated that the no-treatment (1) and PLGA-only (2) defects remained patent without significant defect size differences across groups. In contrast, the h-UCMSC with PLGA group (3) had significantly greater bone fill on micro-CT and histology. Conclusions: We demonstrate a successful calvarial defect model for the investigation of hUCMSC-mediated osteogenesis and bone repair. Furthermore, evidence reveals that PLGA alone has neither short-term effects on bone formation nor any unwanted side effects, making it an 3 attractive scaffold. Further investigation using h-UCMSC with PLGA in larger animals is warranted to advance future translation to patients requiring ACR. Clinical Relevance Statement: Our results demonstrate a successful murine calvarial defect model for the investigation of h-UCMSC-mediated osteogenesis and bone repair and provide preliminary evidence for the safe and efficacious use of this graft adjunct in alveolar cleft repair.
Barraza-Flores, P;Moghadaszadeh, B;Mitchell, B;Troiano, E;Mansur, A;Gupta, V;
| DOI: 10.1016/j.nmd.2022.07.331
SELENON-Related Myopathy (SELENON-RM) is a rare genetic disease caused by recessive mutations of the SELENON gene. It is characterized by the development of rigid spine, axial muscle weakness, and respiratory insufficiency. The most common histopathological feature in SELENON-RM patients is the presence of minicores in skeletal muscle biopsies, which are concentrated areas of mitochondrial depletion within fibers. Natural history data suggest that insulin-resistance as well as altered body mass index (BMI) are correlated with SELENON-RM prognosis. There is no cure or effective treatment for SELENON-RM. The SELENON gene encodes selenoprotein-N, an endoplasmic reticulum (ER) membrane glycoprotein with reducing catalytic activity. Selenoprotein-N has been shown to activate SERCA channels by reducing it at low Ca2+ concentrations in the ER. Additionally, it has also been shown to colocalize with Mitochondrial Associated Membranes (MAM), which are vital for mitochondrial function. However, the molecular mechanism(s) by which selenoprotein-N deficiency causes SELENON-RM is still unclear. A particular challenge has been the lack of cellular or animal models that exhibit readily assayable phenotypes. In this project, we aim to identify cellular and animal models suited for high throughput drug screening while elucidating the molecular mechanism of the disease. To do so, we tested selenoprotein-N deficient cells and zebrafish selenon-null models. Using different measures of metabolism, we found that Selenon-KO C2C12 cells and primary myoblasts isolated from Selenon-KO mice were metabolically impaired. We also assessed several phenotyping outcomes in zebrafish models, from embryonic development to adulthood. Our results showed muscle weakness during early development as well as reduced growth during the larval stage. Together, these data establish the potential for selenoprotein-N deficient cells and zebrafish as models for the discovery of therapeutic targets for SELENON-RM.
Mendell, J;Sahenk, Z;Lehman, K;Lowes, L;Reash, N;Iammarino, M;Alfano, L;Lewis, S;Church, K;Shell, R;Potter, R;Griffin, D;Pozsgai, E;Hogan, M;Hu, L;Mason, S;Darton, E;Rodino-Klapac, L;
| DOI: 10.1016/j.nmd.2022.07.251
Delandistrogene moxeparvovec (SRP-9001) is an investigational gene transfer therapy developed for targeted skeletal and cardiac muscle expression of micro-dystrophin (a shortened, functional dystrophin protein). The objective of this phase 1/2a, single-dose, open-label clinical trial (NCT03375164) is to evaluate the safety of systemic delivery of delandistrogene moxeparvovec in patients with Duchenne muscular dystrophy (DMD). Four ambulatory patients with DMD (≥4 to ≤8 years old) were enrolled. Patients were given an intravenous infusion of delandistrogene moxeparvovec at a dose of 2.0x1014 vg/kg (supercoiled qPCR, linear plasmid standard equivalent of 1.33x1014 vg/kg) and prednisone (1 mg/kg/day) 1 day pre- to 30 days post-gene delivery. The primary outcome measure is safety. The secondary outcome measures include micro-dystrophin expression in pre- and post-muscle biopsies (Week 12 post-infusion). Key efficacy outcome measures include North Star Ambulatory Assessment (NSAA) and timed function tests. Previously, data from 3 years post-treatment were presented. Treatment-related adverse events (TRAEs) were mild to moderate, occurred mostly in the first 90 days of treatment, and all resolved. No serious adverse events (AEs), study discontinuations, or AEs associated with clinically relevant complement activation were reported. All patients demonstrated a clinically meaningful improvement on NSAA. Patients treated with delandistrogene moxeparvovec generally maintained muscle strength (Time to Rise and 4-stair Climb) and showed improvement in ambulation ability (10-metre and 100-metre Walk/Run) from baseline to Year 3. The observed safety profile and the enduring response following treatment provide proof of concept for continuation of clinical trials assessing delandistrogene moxeparvovec using single-dose gene therapy in patients with DMD. We present the latest long-term (4-year) safety and functional data from this study.
Matrix biology : journal of the International Society for Matrix Biology
Rekad, Z;Izzi, V;Lamba, R;Ciais, D;Van Obberghen-Schilling, E;
PMID: 35537652 | DOI: 10.1016/j.matbio.2022.05.003
The extracellular matrix (ECM) is a fundamental component of the tissue of multicellular organisms that is comprised of an intricate network of multidomain proteins and associated factors, collectively known as the matrisome. The ECM creates a biophysical environment that regulates essential cellular processes such as adhesion, proliferation and migration and impacts cell fate decisions. The composition of the ECM varies across organs, developmental stages and diseases. Interestingly, most ECM genes generate transcripts that undergo extensive alternative splicing events, producing multiple protein variants from one gene thus enhancing ECM complexity and impacting matrix architecture. Extensive studies over the past several decades have linked ECM remodeling and expression of alternatively spliced ECM isoforms to cancer, and reprogramming of the alternative splicing patterns in cells has recently been proposed as a new hallmark of tumor progression. Indeed, tumor-associated alternative splicing occurs in both malignant and non-malignant cells of the tumor environment and growing evidence suggests that expression of specific ECM splicing variants could be a key step for stromal activation. In this review, we present a general overview of alternative splicing mechanisms, featuring examples of ECM components. The importance of ECM variant expression during essential physiological processes, such as tissue organization and embryonic development is discussed as well as the dysregulation of alternative splicing in cancer. The overall aim of this review is to address the complexity of the ECM by highlighting the importance of the yet-to-be-fully-characterized "alternative" matrisome in physiological and pathological states such as cancer.