M. T. Pham et al. ( 2018)
NeuroReport 29 7 588--593
Generation of human vascularized brain organoids
The aim of this study was to vascularize brain organoids with a patient's own endothelial cells (ECs). Induced pluripotent stem cells (iPSCs) of one UC Davis patient were grown into whole-brain organoids. Simultaneously,iPSCs from the same patient were differentiated into ECs. On day 34,the organoid was re-embedded in Matrigel with 250 000 ECs. Vascularized organoids were grown in vitro for 3-5 weeks or transplanted into immunodeficient mice on day 54,and animals were perfused on day 68. Coating of brain organoids on day 34 with ECs led to robust vascularization of the organoid after 3-5 weeks in vitro and 2 weeks in vivo. Human CD31-positive blood vessels were found inside and in-between rosettes within the center of the organoid after transplantation. Vascularization of brain organoids with a patient's own iPSC-derived ECs is technically feasible.
View Publication
I. O. Peshkova et al. (nov 2019)
Nature communications 10 1 5046
Abdominal aortic aneurysm (AAA) is a prevalent life-threatening disease,where aortic wall degradation is mediated by accumulated immune cells. Although cytokines regulate inflammation within the aorta,their contribution to AAA via distant alterations,particularly in the control of hematopoietic stem cell (HSC) differentiation,remains poorly defined. Here we report a pathogenic role for the interleukin-27 receptor (IL-27R) in AAA,as genetic ablation of IL-27R protects mice from the disease development. Mitigation of AAA is associated with a blunted accumulation of myeloid cells in the aorta due to the attenuation of Angiotensin II (Ang II)-induced HSC expansion. IL-27R signaling is required to induce transcriptional programming to overcome HSC quiescence and increase differentiation and output of mature myeloid cells in response to stress stimuli to promote their accumulation in the diseased aorta. Overall,our studies illuminate how a prominent vascular disease can be distantly driven by a cytokine-dependent regulation of bone marrow precursors.
View Publication
M. D. Perry et al. (sep 2019)
Cardiovascular research
Pharmacological activation of IKr in models of long QT Type 2 risks overcorrection of repolarization.
AIMS Current treatment for congenital long QT syndrome Type 2 (cLQTS2),an electrical disorder that increases the risk of life-threatening cardiac arrhythmias,is aimed at reducing the incidence of arrhythmia triggers (beta-blockers) or terminating the arrhythmia after onset (implantable cardioverter-defibrillator). An alternative strategy is to target the underlying disease mechanism,which is reduced rapid delayed rectifier current (IKr) passed by Kv11.1 channels. Small molecule activators of Kv11.1 have been identified but the extent to which these can restore normal cardiac signalling in cLQTS2 backgrounds remains unclear. Here,we examined the ability of ICA-105574,an activator of Kv11.1 that impairs transition to the inactivated state,to restore function to heterozygous Kv11.1 channels containing either inactivation enhanced (T618S,N633S) or expression deficient (A422T) mutations. METHODS AND RESULTS ICA-105574 effectively restored Kv11.1 current from heterozygous inactivation enhanced or expression defective mutant channels in heterologous expression systems. In a human-induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) model of cLQTS2 containing the expression defective Kv11.1 mutant A422T,cardiac repolarization,estimated from the duration of calcium transients in isolated cells and the rate corrected field potential duration (FPDc) in culture monolayers of cells,was significantly prolonged. The Kv11.1 activator ICA-105574 was able to reverse the prolonged repolarization in a concentration-dependent manner. However,at higher doses,ICA-105574 produced a shortening of the FPDc compared to controls. In vitro and in silico analysis suggests that this overcorrection occurs as a result of a temporal redistribution of the peak IKr to much earlier in the plateau phase of the action potential,which results in early repolarization. CONCLUSION Kv11.1 activators,which target the primary disease mechanism,provide a possible treatment option for cLQTS2,with the caveat that there may be a risk of overcorrection that could itself be pro-arrhythmic.
View Publication
J. A. Pearson et al. ( 2019)
Frontiers in immunology 10 2654
Norovirus Changes Susceptibility to Type 1 Diabetes by Altering Intestinal Microbiota and Immune Cell Functions.
Environmental factors contribute to Type 1 diabetes (T1D) susceptibility. The gut microbiome,which includes bacteria,viruses,and fungi,contributes to this environmental influence,and can induce immunological changes. The gut viral component of the microbiome,related to T1D has mostly focused on coxsackieviruses and rotavirus. The role of norovirus,another common enteric virus,in susceptibility to T1D was hitherto unknown. Norovirus is highly infectious and encountered by many children. We studied the mouse norovirus 4 (MNV4),related to human noroviruses,in the Non-obese diabetic (NOD) mouse model,to determine its role in influencing susceptibility to T1D. We infected MNV-free NOD mice with MNV4 by exposing the mice to MNV4-positive bedding from an endemically-infected mouse colony to mimic a natural infection. Control MNV-free NOD mice were exposed to MNV-free bedding from the same colony. Interestingly,MNV4 infection protected NOD mice from the development of T1D and was associated with an expansion of Tregs and reduced proinflammatory T cells. We also found MNV4 significantly modified the gut commensal bacteria composition,promoting increased $\alpha$-diversity and Firmicutes/Bacteroidetes ratio. To elucidate whether T1D protection was directly related to MNV4,or indirectly through modulating gut microbiota,we colonized germ-free (GF) NOD mice with the MNV4-containing or non-MNV4-containing viral filtrate,isolated from filtered fecal material. We found that MNV4 induced significant changes in mucosal immunity,including altered Tuft cell markers,cytokine secretion,antiviral immune signaling markers,and the concentration of mucosal antibodies. Systemically,MNV4-infection altered the immune cells including B cell subsets,macrophages and T cells,and especially induced an increase in Treg number and function. Furthermore,in vitro primary exposure of the norovirus filtrate to na{\{i}}ve splenocytes identified significant increases in the proportion of activated and CTLA4-expressing Tregs. Our data provide novel knowledge that norovirus can protect NOD mice from T1D development by inducing the expansion of Tregs and reducing inflammatory T cells. Our study also highlights the importance of distinguishing the mucosal immunity mediated by bacteria from that by enteric viruses."
View Publication
M. Pardons et al. (nov 2019)
Cell reports 29 9 2783--2795.e5
Latency-Reversing Agents Induce Differential Responses in Distinct Memory CD4 T Cell Subsets in Individuals on Antiretroviral Therapy.
Latent proviruses persist in central (TCM),transitional (TTM),and effector (TEM) memory cells. We measured the levels of cellular factors involved in HIV gene expression in these subsets. The highest levels of acetylated H4,active nuclear factor $\kappa$B (NF-$\kappa$B),and active positive transcription elongation factor b (P-TEFb) were measured in TEM,TCM,and TTM cells,respectively. Vorinostat and romidepsin display opposite abilities to induce H4 acetylation across subsets. Protein kinase C (PKC) agonists are more efficient at inducing NF-$\kappa$B phosphorylation in TCM cells but more potent at activating PTEF-b in the TEM subset. We selected the most efficient latency-reversing agents (LRAs) and measured their ability to reverse latency in each subset. While ingenol alone has modest activities in the three subsets,its combination with a histone deacetylase inhibitor (HDACi) dramatically increases latency reversal in TCM cells. Altogether,these results indicate that cellular HIV reservoirs are differentially responsive to common LRAs and suggest that combination of compounds will be required to achieve latency reversal in all subsets.
View Publication
Q. Pan et al. (dec 2019)
Cell and tissue research
Characterizing the effects of hypoxia on the metabolic profiles of mesenchymal stromal cells derived from three tissue sources using chemical isotope labeling liquid chromatography-mass spectrometry.
Microenvironmental factors such as oxygen concentration mediate key effects on the biology of mesenchymal stromal cells (MSCs). Herein,we performed an in-depth characterization of the metabolic behavior of MSCs derived from the placenta,umbilical cord,and adipose tissue (termed hPMSCs,UC-MSCs,and AD-MSCs,respectively) at physiological (hypoxic; 5{\%} oxygen [O2]) and standardized (normoxic; 21{\%} O2) O2 concentrations using chemical isotope labeling liquid chromatography-mass spectrometry. 12C- and 13C-isotope dansylation (Dns) labeling was used to analyze the amine/phenol submetabolome,and 2574 peak pairs or metabolites were detected and quantified,from which 52 metabolites were positively identified using a library of 275 Dns-metabolite standards; 2189 metabolites were putatively identified. Next,we identified six metabolites using the Dns library,as well as 14 hypoxic biomarkers from the human metabolome database out of 96 altered metabolites. Ultimately,metabolic pathway analyses were performed to evaluate the associated pathways. Based on pathways identified using the Kyoto Encyclopedia of Genes and Genomes,we identified significant changes in the metabolic profiles of MSCs in response to different O2 concentrations. These results collectively suggest that O2 concentration has the strongest influence on hPMSCs metabolic characteristics,and that 5{\%} O2 promotes arginine and proline metabolism in hPMSCs and UC-MSCs but decreases gluconeogenesis (alanine-glucose) rates in hPMSCs and AD-MSCs. These changes indicate that MSCs derived from different sources exhibit distinct metabolic profiles.
View Publication
B. Novotn\'a et al. (dec 2019)
Journal of medicinal chemistry 62 23 10676--10690
Enzymatic Preparation of 2'-5',3'-5'-Cyclic Dinucleotides, Their Binding Properties to Stimulator of Interferon Genes Adaptor Protein, and Structure/Activity Correlations.
Cyclic dinucleotides are second messengers in the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway,which plays an important role in recognizing tumor cells and viral or bacterial infections. They bind to the STING adaptor protein and trigger expression of cytokines via TANK binding kinase 1 (TBK1)/interferon regulatory factor 3 (IRF3) and inhibitor of nuclear factor-$\kappa$B (I$\kappa$B) kinase (IKK)/nuclear factor-$\kappa$B (NF$\kappa$B) signaling cascades. In this work,we describe an enzymatic preparation of 2'-5',3'-5'-cyclic dinucleotides (2'3'CDNs) with use of cyclic GMP-AMP synthases (cGAS) from human,mouse,and chicken. We profile substrate specificity of these enzymes by employing a small library of nucleotide-5'-triphosphate (NTP) analogues and use them to prepare 33 2'3'CDNs. We also determine affinity of these CDNs to five different STING haplotypes in cell-based and biochemical assays and describe properties needed for their optimal activity toward all STING haplotypes. Next,we study their effect on cytokine and chemokine induction by human peripheral blood mononuclear cells (PBMCs) and evaluate their cytotoxic effect on monocytes. Additionally,we report X-ray crystal structures of two new CDNs bound to STING protein and discuss structure-activity relationship by using quantum and molecular mechanical (QM/MM) computational modeling.
View Publication
L. R. Neitzel et al. ( 2019)
Mechanisms of development 156 20--31
Developmental regulation of Wnt signaling by Nagk and the UDP-GlcNAc salvage pathway.
In a screen for human kinases that regulate Xenopus laevis embryogenesis,we identified Nagk and other components of the UDP-GlcNAc glycosylation salvage pathway as regulators of anteroposterior patterning and Wnt signaling. We find that the salvage pathway does not affect other major embryonic signaling pathways (Fgf,TGF$\beta$,Notch,or Shh),thereby demonstrating specificity for Wnt signaling. We show that the role of the salvage pathway in Wnt signaling is evolutionarily conserved in zebrafish and Drosophila. Finally,we show that GlcNAc is essential for the growth of intestinal enteroids,which are highly dependent on Wnt signaling for growth and maintenance. We propose that the Wnt pathway is sensitive to alterations in the glycosylation state of a cell and acts as a nutritional sensor in order to couple growth/proliferation with its metabolic status. We also propose that the clinical manifestations observed in congenital disorders of glycosylation (CDG) in humans may be due,in part,to their effects on Wnt signaling during development.
View Publication
J. A. Neil et al. ( 2019)
Nature microbiology 4 10 1737--1749
IFN-I and IL-22 mediate protective effects of intestinal viral infection.
Products derived from bacterial members of the gut microbiota evoke immune signalling pathways of the host that promote immunity and barrier function in the intestine. How immune reactions to enteric viruses support intestinal homeostasis is unknown. We recently demonstrated that infection by murine norovirus (MNV) reverses intestinal abnormalities following depletion of bacteria,indicating that an intestinal animal virus can provide cues to the host that are typically attributed to the microbiota. Here,we elucidate mechanisms by which MNV evokes protective responses from the host. We identify an important role for the viral protein NS1/2 in establishing local replication and a type I interferon (IFN-I) response in the colon. We further show that IFN-I acts on intestinal epithelial cells to increase the proportion of CCR2-dependent macrophages and interleukin (IL)-22-producing innate lymphoid cells,which in turn promote pSTAT3 signalling in intestinal epithelial cells and protection from intestinal injury. In addition,we demonstrate that MNV provides a striking IL-22-dependent protection against early-life lethal infection by Citrobacter rodentium. These findings demonstrate novel ways in which a viral member of the microbiota fortifies the intestinal barrier during chemical injury and infectious challenges.
View Publication
B. Ndreshkjana et al. ( 2019)
Cell death {\&} disease 10 6 379
Combination of 5-fluorouracil and thymoquinone targets stem cell gene signature in colorectal cancer cells.
Cancer stem cells (CSCs) residing in colorectal cancer tissues have tumorigenic capacity and contribute to chemotherapeutic resistance and disease relapse. It is well known that the survival of colorectal CSCs after 5-fluorouracil (5-FU)-based therapy leads to cancer recurrence. Thus CSCs represent a promising drug target. Here,we designed and synthesized novel hybrid molecules linking 5-FU with the plant-derived compound thymoquinone (TQ) and tested the potential of individual compounds and their combination to eliminate colorectal CSCs. Both,Combi and SARB hybrid showed augmented cytotoxicity against colorectal cancer cells,but were non-toxic to organoids prepared from healthy murine small intestine. NanoString analysis revealed a unique signature of deregulated gene expression in response to the combination of TQ and 5-FU (Combi) and SARB treatment. Importantly,two principle stem cell regulatory pathways WNT/{\ss}-Catenin and PI3K/AKT were found to be downregulated after Combi and hybrid treatment. Furthermore,both treatments strikingly eliminated CD133+ CSC population,accompanying the depleted self-renewal capacity by eradicating long-term propagated 3D tumor cell spheres at sub-toxic doses. In vivo xenografts on chicken eggs of SARB-treated HCT116 cells showed a prominent nuclear {\ss}-Catenin and E-cadherin staining. This was in line with the reduced transcriptional activity of {\ss}-Catenin and diminished cell adhesion under SARB exposure. In contrast to 5-FU,both,Combi and SARB treatment effectively reduced the angiogenic capacity of the remaining resistant tumor cells. Taken together,combination or hybridization of single compounds target simultaneously a broader spectrum of oncogenic pathways leading to an effective eradication of colorectal cancer cells.
View Publication
D. Nag et al. (aug 2019)
Clinical cancer research : an official journal of the American Association for Cancer Research 25 15 4791--4807
Auranofin Protects Intestine against Radiation Injury by Modulating p53/p21 Pathway and Radiosensitizes Human Colon Tumor.
PURPOSE The radiosensitivity of the normal intestinal epithelium is the major limiting factor for definitive radiotherapy against abdominal malignancies. Radiosensitizers,which can be used without augmenting radiation toxicity to normal tissue,are still an unmet need. Inhibition of proteosomal degradation is being developed as a major therapeutic strategy for anticancer therapy as cancer cells are more susceptible to proteasomal inhibition-induced cytotoxicity compared with normal cells. Auranofin,a gold-containing antirheumatoid drug,blocks proteosomal degradation by inhibiting deubiquitinase inhibitors. In this study,we have examined whether auranofin selectively radiosensitizes colon tumors without promoting radiation toxicity in normal intestine. EXPERIMENTAL DESIGN The effect of auranofin (10 mg/kg i.p.) on the radiation response of subcutaneous CT26 colon tumors and the normal gastrointestinal epithelium was determined using a mouse model of abdominal radiation. The effect of auranofin was also examined in a paired human colonic organoid system using malignant and nonmalignant tissues from the same patient. RESULTS Both in the mouse model of intestinal injury and in the human nonmalignant colon organoid culture,auranofin pretreatment prevented radiation toxicity and improved survival with the activation of p53/p21-mediated reversible cell-cycle arrest. However,in a mouse model of abdominal tumor and in human malignant colonic organoids,auranofin inhibited malignant tissue growth with inhibition of proteosomal degradation,induction of endoplasmic reticulum stress/unfolded protein response,and apoptosis. CONCLUSIONS Our data suggest that auranofin is a potential candidate to be considered as a combination therapy with radiation to improve therapeutic efficacy against abdominal malignancies.
View Publication
F. Muhammad et al. (nov 2019)
Scientific reports 9 1 16941
Experimental autoimmune uveoretinitis (EAU) is a mouse model of human autoimmune uveitis marked by ocular autoantigen-specific regulatory immunity in the spleen. The melanocortin 5 receptor (MC5r) and adenosine 2 A receptor (A2Ar) are required for induction of post-EAU regulatory T cells (Tregs) which provide resistance to EAU. We show that blocking the PD-1/PD-L1 pathway prevented suppression of EAU by post-EAU Tregs. A2Ar induction of PD-1+FoxP3+ Tregs in uveitis patients was similar compared to healthy controls,but was significantly reduced with melanocortin stimulation. Further,lower body mass index correlated with responsiveness to stimulation of this pathway. These observations indicate an importance of the PD-1/PD-L1 pathway to provide resistance to relapsing uveitis and shows a reduced capacity of uveitis patients to induce Tregs when stimulated through melanocortin receptors,but that it is possible to bypass this part of the pathway through direct stimulation of A2Ar.
View Publication