Monthly Archives

15 Articles

??7-Dehydrocholesterol Reductase

Data are representative of 3 indie experiments

Posted by Andre Olson on

Data are representative of 3 indie experiments. to target the IRE-1/XBP-1 pathway. Treatment of CLL cells with this inhibitor (B-I09) mimicked XBP-1 deficiency, including upregulation of IRE-1 expression and compromised BCR signaling. Moreover, B-I09 treatment did not affect the transport Quinestrol of secretory and integral membrane-bound proteins. Administration of B-I09 to CLL tumorCbearing mice suppressed leukemic progression by inducing apoptosis and did not cause systemic toxicity. Additionally, B-I09 and ibrutinib, an FDA-approved BTK inhibitor, synergized to induce apoptosis in B cell leukemia, lymphoma, and multiple myeloma. These data show that targeting XBP-1 has potential as a treatment strategy, not only for multiple myeloma, but also for mature B cell leukemia and lymphoma. Introduction The functional role of the ER stress response in mature B cell leukemia or lymphoma has been largely overlooked because leukemia and lymphoma cells do not expand their ER as do multiple myeloma (MM) cells. Recently, chronic lymphocytic leukemia (CLL), the most common adult leukemia, was shown to require activation of the ER stress response for survival (1). The IRE-1/XBP-1 pathway represents the most conserved ER stress-response pathway. IRE-1 contains a luminal stress-sensor domain name and a cytoplasmic kinase/RNase domain name (Supplemental Physique 1; supplemental material available online with this short article; doi:10.1172/JCI73448DS1). The RNase domain name is critical for the function of IRE-1 because it splices 26 nucleotides from your mRNA, causing a frame shift in translation (2C4). The spliced mRNA encodes a functional 54-kDa XBP-1s transcription factor. The role of XBP-1 in malignancy has not been validated by genetic deletion of the gene in mice. Thus, we deleted the gene from B cells of E-TCL1 transgenic mice (E-TCL1, herein referred to as XBP-1KO/E-TCL1), arguably the best CLL mouse model to date (5, 6). The E-TCL1 mouse model is usually clinically relevant because TCL1 expression is found Quinestrol in 90% of human CLL cases (1, 7). E-TCL1 mice develop leukemia with all clinical features of aggressive human CLL (6, 8) and have been used repeatedly for preclinical drug assessments (9C16). Using XBP-1KO/E-TCL1 mice, we examine the role of the IRE-1/XBP-1 pathway in tumor progression. Quinestrol While most transcription factors remain undruggable, the specific activation mechanism of XBP-1 renders IRE-1 an attractive target for therapeutic intervention. Although chemical screens have led to the identification of inhibitors of the IRE-1 RNase activity (17C20), there is a need to develop novel small molecules with improved cellular and in vivo efficacy. We synthesized and evaluated novel tricyclic chromenone inhibitors of IRE-1 RNase activity that potently suppress the expression of XBP-1 and induce apoptosis. We also decided the bioavailability and pharmacokinetics of our lead inhibitor, B-I09, and showed that B-I09, when administered as a single agent, effectively induces leukemic regression without causing systemic Rabbit polyclonal to ND2 toxicity in CLL-bearing E-TCL1 mice. Since the inhibition of the IRE-1/XBP-1 pathway compromises B cell receptor (BCR) signaling, we tested for any potential synergistic effect between B-I09 and the Brutons tyrosine kinase (BTK) inhibitor ibrutinib. Our results demonstrate the effectiveness of targeting both the IRE-1/XBP-1 and BCR signaling pathways to induce apoptosis in human B cell leukemia, lymphoma, and MM cells. Results XBP-1KO/E-TCL1 mice develop leukemia significantly more slowly than XBP-1WT/E-TCL1 mice. To investigate how the loss of XBP-1 can counter malignant progression of leukemia, we crossed B cellCspecific XBP-1KO mice (= 5 in each age group). (F) CD5+B220+ CLL cells purified from spleens of XBP-1WT/E-TCL1 and XBP-1KO/E-TCL1 mice were lysed to analyze for the expression of indicated proteins. Data shown in immunoblots are representative of 3 impartial experiments. (G) Spleens from 12-month-old age-matched XBP-1WT/E-TCL1 and XBP-1KO/E-TCL1 littermates and a WT mouse. (H) Kaplan-Meier analysis of overall survival of XBP-1KO/E-TCL1 mice (= 18). Four mice from your XBP-1KO/E-TCL1 group were censored (circled in reddish), as they were removed for other studies. XBP-1Cdeficient E-TCL1 CLL cells exhibit compromised BCR signaling. Constitutive BCR activation is usually a critical survival transmission for CLL cells (22, 23). To understand how the loss of XBP-1 may contribute to the slower progression of leukemia in E-TCL1 mice, we purified CLL cells from XBP-1WT/E-TCL1 and XBP-1KO/E-TCL1 littermates (Supplemental Physique 2, B and C), cultured them in LPS for 3 days, activated the BCR using F(ab)2 anti-mouse IgM, and lysed the cells. Cell lysates were immunoblotted for phospho-Syk and phospho-BTK because Syk and BTK are crucial BCR signaling molecules for CLL survival (22, 23). Compared with XBP-1WT/E-TCL1 CLL cells, XBP-1KO/E-TCL1 CLL cells are defective in Syk and BTK phosphorylation upon activation of the BCR (Physique ?(Figure2A).2A). Unlike naive normal B cells, XBP-1WT/E-TCL1 CLL cells synthesize significantly increased amounts of secretory forms of IgM and release them into culture medium in the absence of any activation (Physique ?(Physique2,2, B and C). The lack of XBP-1.

DGAT-1

J Immunol Methods

Posted by Andre Olson on

J Immunol Methods. produce inflammatory factors in response to pathogen recognition receptor (PRR) signaling, which might help to shape the biology of the TME. We determined that mouse ovarian tumors generate chemokines that are able to interact with receptors harbored by tumor-associated DCs. We also found that dsRNA triggers significant pro-inflammatory cytokine up-regulation in both human and mouse ovarian tumor cell lines, and that several PRR can simultaneously contribute to the stimulated inflammatory response displayed by these cells. Thus, dsRNA-activated PRRs may not only constitute potentially relevant drug targets for therapies aiming to prevent inflammation associated with leukocyte recruitment, or as co-adjuvants of therapeutic treatments, but also might have a role in development of nascent tumors, for example via activation of cancer cells by microbial molecules associated to pathogens, or with those appearing in circulation due to dysbiosis. cultured ID8-VegfA cancer cells (C) and normal tissues were subjected to RNA extraction followed by qPCR analysis. Data were analyzed with the Kruskal-Wallis Test (nonparametric ANOVA) followed by Dunn post-test comparisons. LN: Lymph nodes. (D). Analysis of Exodus-2 at the protein level was determined in solid mouse tumor by IHC. Staining of mouse ovarian tumors with CCL21 antibody (Left Panel) and isotype control (Right Panel) shows positive staining both in tumor islets and stroma. (100X magnification). Using qPCR analysis, we analyzed chemokine expression in samples collected from 20 independent solid tumors. We compared chemokine expression to that in immune organs, as well as in cultured ID8-VegfA cells recovered from different experiments. As shown in Figure ?Figure1C,1C, murine ovarian tumors express several Ruxolitinib sulfate chemokines at the RNA level such as ELC/CCL19 (interacts with CCR7); Exodus-2/CCL21 (interacts with CCR7); MIP-1/CCL3 (interacts with CCR1 and CCR5); MIP-1/CCL4 (interacts with CCR5); RANTES/CCL5 (interacts with CCR1, CCR3 and CCR5); and SDF-1/CXCL12 (interacts with CXCR4 Ruxolitinib sulfate and CXCR7). As expected, in most cases the overall levels of chemokines produced by tumors were lower than those of immunological organs, except in the case of MIP-1, or MIP-1, where the expression levels were not significantly different. In addition, with respect to MIP-1, tumor samples appear to express higher levels of the chemokine than those observed in Ruxolitinib sulfate tumor cells in culture. One possible explanation is that this chemokine is produced by tumor cells under the influence of the TME (e.g., different levels of oxygen, 3D environment, lactic acid accumulation, extracellular matrix interaction), or that other TME cells rather than cancer cells are responsible for the elevated expression of this chemokine. An immunohistochemistry analysis of solid tumors revealed the expression Alox5 of Exodus 2/CCL21 at the level of protein (Figure ?(Figure1D),1D), both in tumor islets and stroma, strongly suggesting that tumor cells can be a source of chemokines viability studies (Supplementary Figure 1E-1F). Additionally, we validated the protein array data with respect to IL-6 expression by means of ELISA experiments (Figure ?(Figure2G).2G). On the contrary, no differences in MCP-1/CCL2 expression were observed when using this technique. We also found that MIP-1/CCL4 is upregulated upon transfection with both poly (I:C) and poly (A:U). CXCL2, was present in the supernatants of mouse ovarian tumor cells (Figure ?(Figure2A),2A), but not upregulated upon dsRNA transfection as determined by array analysis (Figure ?(Figure2D),2D), and also showed no differences when analyzed by ELISA. Thus, both RANTES/CCL5 and IL-6 are molecules that were upregulated upon dsRNA transfection of cancer cells at the protein level as determined by two complementary methods. It has been reported that dsRNA can promote the upregulation of dsRNA-sensing PRRs in some cells [52]. In our studies we were able to determine, at the level of RNA, that PKR was the only dsRNA PRR affected by the transfection in these murine ovarian cancer Ruxolitinib sulfate cells, and only upon transfection with poly (A:U), indicating that PKR may participate in a positive feedback loop in response to dsRNA stimulation (Supplementary Figure 1G). PRR polymorphisms have been implicated in poor clinical outcomes in some cancers, an example of which is the overexpression of TLR3 in human ovarian tumors [29]. To further understand the mechanisms by.

mGlu6 Receptors

In keeping with the observed rhythmicity, BMAL1 bound to core clock genes, including and in GSCs, as measured by BMAL1 chromatin immunoprecipitation followed by deep sequencing (ChIP-seq; Supplementary Fig

Posted by Andre Olson on

In keeping with the observed rhythmicity, BMAL1 bound to core clock genes, including and in GSCs, as measured by BMAL1 chromatin immunoprecipitation followed by deep sequencing (ChIP-seq; Supplementary Fig. model systems, or serve as oncogenes (12,18,19), but, in others, their targeting is usually tumor suppressive (20C22). Recent systems analysis revealed that alteration of circadian genes is usually correlated with patient survival and clinical outcomes in several tumor types (23). Circadian networks in glioblastoma may be oncogenic with an association between gene variants and tumor incidence, and targeting circadian regulators may reduce tumor growth and improve efficacy of chemotherapy (24,25). Based on this background, we investigated the integrity of the core circadian circuitry within GSCs. RESULTS Genetic disruption of core circadian genes inhibits GSC growth To study the circadian rhythm and core circadian genes in glioblastoma, we monitored circadian clock activity utilizing a luciferase reporter driven by the promoter. Although MYC has been proposed to disrupt BET-IN-1 the normal circadian rhythm (26) and GSCs express high MYC levels (27), patient-derived GSCs and their differentiated progeny, displayed circadian rhythms with comparable properties to non-malignant brain cultures derived from epilepsy surgical resections (NMs), impartial of tumor genetics (Fig. 1ACD; Supplementary Fig. S1ACS1K). Consistent with the observed rhythmicity, BMAL1 bound to core clock genes, including and in GSCs, as measured by BMAL1 chromatin immunoprecipitation followed by deep sequencing (ChIP-seq; Supplementary Fig. S1LCS1N). Canonical rhythms observed in normal brain cells and GSCs suggest that cellular transformation maintains circadian rhythms, despite the activation of oncogenes. Open in a separate window Physique 1. Genetic disruption of core clock genes suppresses GSC growth despite strong circadian oscillation.(A-D) Bioluminescence of BMAL1::Luc in T387 (A) and T3565 (B) GSCs, non-malignant brain cultures (C), NSC (ENSA) (D), synchronized by 100 nM dexamethasone or 10 M forskolin. Data are representative of three experiments. (E and F) mRNA and protein expression of BMAL1 and CLOCK in T387 (E) and T3565 (F) GSCs transduced with shCONT, shBMAL1 or shCLOCK. Data are presented as mean SD. ***, P< 0.001. Statistical significance was determined by one-way ANOVA with Tukeys multiple comparison. N=3. (G and H) Relative cell numbers of T387 (G) and T3565 (H) GSCs transduced with shCONT, shBMAL1 or shCLOCK. Data are presented as mean SD. ***, P< 0.001. Statistical significance BET-IN-1 was determined by two-way ANOVA with Tukeys multiple comparison. N=4. (I and J) mRNA and protein expression of BMAL1 and CLOCK in non-malignant brain cultures (NM 263) (I) and NSC (ENSA) (J) transduced with shCONT, shBMAL1 or shCLOCK. Data are presented as mean SD. Ephb4 ***, P< 0.001. Statistical significance was determined by one-way ANOVA with Tukeys multiple comparison. N=3. (K and L) Relative cell numbers of nonmalignant brain cultures (K) and NSCs (L) transduced with shCONT, shBMAL1 or shCLOCK. Data are presented as mean SD. ***, P< 0.001. Statistical significance was determined by two-way BET-IN-1 ANOVA with Tukeys multiple comparison. N=4. (M-P) Protein expression of BMAL1 or CLOCK and relative cellular numbers in GSCs transduced with Cas9-sgCONT, Cas9-sgBMAL1 (M and N) or Cas9-sgCLOCK (O and P). Data are presented as mean SD. ***, P< 0.001. Statistical significance was determined by two-way ANOVA with Tukeys multiple comparison. N=3. To study the functional functions of core circadian genes, and were targeted by shRNA-mediated knockdown in patient-derived GSCs and NMs using two non-overlapping shRNAs compared to a control non-targeting shRNA sequence (shCONT). Targeting either or potently impaired proliferation in GSCs derived from multiple patients (Fig. 1ECH; Supplementary Fig. S2ACS2F). In contrast, targeting or minimally reduced cell proliferation in epilepsy-derived brain cultures or neural stem cells (NSCs) (Fig. 1ICL; Supplementary S2G and S2H), with modest anti-proliferative effects in DGCs (Supplementary Fig. S2ICS2L). Reduced GSC proliferation upon or knockdown BET-IN-1 was confirmed by CRISPR/Cas9-mediated knockout (Fig. 1MC1P). As partially compensates for the loss of CLOCK in some tissues (28), we measured mRNA expression in different cell types; while NSCs and NMs had the lowest and highest mRNA expression respectively, GSCs.

CASR

We cultured a number of different cell lines using the reduced static pressure-loadable two-chamber program, and examined cell development, cell routine, and cell morphology

Posted by Andre Olson on

We cultured a number of different cell lines using the reduced static pressure-loadable two-chamber program, and examined cell development, cell routine, and cell morphology. mesenchymal cells weren’t growth-suppressed, at 50 cm H2O actually. Phalloidin staining exposed that 50 cm H2O pressure fill vertically flattened and laterally widened columnar epithelial cells and produced actin dietary fiber distribution sparse, without influencing total phalloidin strength per cell. When the mucosal protectant irsogladine maleate (100 nM) was put into 50-cm-high culture moderate, MDCK cells had been reduced in quantity and their doubling period shortened. Cell morphology and proliferation are regarded as controlled from the Hippo signaling pathway. A pressure fill of 50 cm H2O improved serine-127 phosphorylation and cytoplasmic retention of YAP, the main constituent of the pathway, recommending that Hippo NVP-AAM077 Tetrasodium Hydrate (PEAQX) pathway was mixed up in pressure-induced cell development suppression. RNA sequencing of MDCK cells demonstrated a 50 cm H2O pressure fill upregulated procedure when erosive areas from the mucosa are becoming re-epithelialized by epithelial cell development beneath the condition of intraluminal pressure elevation. We’d a special fascination with cell shape modification induced by pressure fill, because mucosal epithelia contain columnar-shaped cells generally. We cultured numerous kinds of epithelial and mesenchymal cells utilizing a drinking water pressure-loadable two-chamber program, and examined adjustments in cell development cell and profiles morphology. Next, we examined protein expression from the Hippo pathway substances and tackled the Hippo signaling activity, and we comprehensively compared gene expression between non-loaded and pressure-loaded epithelial cells by RNA sequencing. Furthermore, we analyzed whether IM rescued the pressure-induced phenomena of epithelial cells. Pressure-induced phenotypes exposed a close hyperlink among morphology, cytoskeleton, and proliferation in columnar epithelial cells. Methods and Materials Cells, antibodies, and reagents MadinCDarby canine kidney (MDCK), NIH3T3, and TIG-1 cells had been bought and cultured as referred to in our earlier reviews (Ito et al., 2000, 2008; Hosokawa et al., 2011). Human being lung adenocarcinoma NCI-H441 cells (great deal no. 58294188) had been purchased through the American Type Tradition Collection (Manassas, VA, USA) and cultivated as previously referred to. Human digestive tract adenocarcinoma Caco-2, and human being gastric adenocarcinoma (signet-ring cell carcinoma) KATO-III and NUGC-4 cells had been purchased through the Riken BioResource Middle, Tsukuba, Japan. All tests using these cells had been performed within 4 weeks after NVP-AAM077 Tetrasodium Hydrate (PEAQX) resuscitation. MDCK, Caco-2, and NCI-H441 cell monolayer cultures on semipermeable membranes had been utilized as representative types of columnar epithelia (Volpe, 2011; Ren et al., 2016). KATO-III and NUGC-4 cells had been used as reps that are of epithelial source but possess a spherical morphology; this morphology well resembles that of signet-ring NVP-AAM077 Tetrasodium Hydrate (PEAQX) cell carcinoma cells (Sekiguchi et al., 1978; Nakashio et al., 1997). Major antibodies found in this research targeted MST2 (#3952; Cell Signaling, Beverly, MA, USA), LATS1 (C66B5; Cell Signaling), LATS2 (#A300-479A, Bethyl Laboratories, Montgomery, TX, USA), YAP (#4912; Cell Signaling), Phospho-YAP (Ser127; #4911, Cell signaling), TAZ (#HPA007415; Sigma-Aldrich, St. Louis, MO, USA), keratin 14 (LL002; Dako, Glostrup, Denmark), lamin B (M-20; Santa Cruz, Dallas, TX, USA), MCM7 (DCS-141; Medical & Biological Laboratories, Nagoya, Japan), -actin (Medical & Biological Laboratories), and GAPDH (Medical & Biological Laboratories). Peroxidase-conjugated supplementary antibodies useful for traditional western blot analysis had been bought from Amersham (Buckinghamshire, Britain). Phalloidin (rhodamine conjugated) and DAPI had been IGSF8 bought from Molecular Probes (Carlsbad, CA, USA) and Dojindo (Kumamoto, Japan), respectively. IM was supplied by Nippon Shinyaku Co kindly., Ltd. (Kyoto, Japan), and was dissolved in DMSO NVP-AAM077 Tetrasodium Hydrate (PEAQX) at a focus of just one 1 mM (share remedy). Blebbistatin and jasplakinolide had been bought from Wako Pure Chemical substance Sectors (Osaka, Japan) and BioVision, Inc. (SAN FRANCISCO BAY AREA, CA, USA), and was dissolved in DMSO at concentrations of 150 and 1.5 mM (share solution), respectively. Two-chamber tradition system for drinking water pressure loading Water pressure-loadable two-chamber tradition device once was described at length (Yoneshige et al., 2017). Quickly, the top chamber composite contains a long plastic material cylinder having a water-tight reference to a culture put in lined having a semipermeable membrane, and the machine was put into a 10-cm dish reduced chamber vertically. Between your two chambers, a porous (150 m, 200 cm2) silicon sheet was put to aid the semipermeable membrane against the moderate (drinking water pressure) put on the top chamber cylinder. Using this product, cells had been subjected to drinking water pressure amounts NVP-AAM077 Tetrasodium Hydrate (PEAQX) (cm H2O) dictated from the height from the moderate from the top towards the semipermeable membrane. Incomplete pressures.

Synthases/Synthetases

Fetuses in ZIKV-infected DENV-immune dams were normal sized, whereas fetal demise occurred in non-immune dams

Posted by Andre Olson on

Fetuses in ZIKV-infected DENV-immune dams were normal sized, whereas fetal demise occurred in non-immune dams. normal sized, whereas fetal demise occurred in non-immune dams. Moreover, reduced ZIKV Kv3 modulator 4 RNA is present in the placenta and fetuses of ZIKV-infected DENV-immune dams. DENV cross-reactive CD8+ T cells expand in the maternal spleen and decidua of ZIKV-infected dams, their depletion increases ZIKV infection in the placenta and fetus, and results in fetal demise. The inducement of cross-reactive CD8+ T cells via peptide immunization or adoptive transfer results in decreased ZIKV infection in the placenta. Prior DENV immunity can protect against ZIKV infection during pregnancy in mice, and CD8+ T cells are sufficient for this cross-protection. This has implications for understanding the natural history of ZIKV in DENV-endemic areas and the development of optimal ZIKV vaccines. Introduction Zika virus (ZIKV) is a positive-stranded, enveloped, RNA flavivirus in the family that is transmitted by species mosquitoes and sexual contact. ZIKV was first isolated in 1947 from a sentinel rhesus macaque in Uganda, and for decades, sporadic human case reports in Africa and Asia were associated with a self-limiting febrile illness. Outbreaks of ZIKV infection beyond its original range were reported in 2007 in Micronesia and from 2013 to 2014 in French Polynesia, where infection was associated with development of GuillainCBarr syndrome (GBS)1. Recently, there was a major epidemic of ZIKV in the Western Hemisphere, which also was associated with GBS. Additionally, infection of pregnant women was confirmed to cause congenital ZIKV syndrome, which includes microcephaly and other birth defects2,3. A successful pregnancy requires the maternal immune system to recognize and tolerate fetal tissues. Nonetheless, pregnant mammals must still mount robust immune response to pathogens4C6. Some pathogens including ZIKV ostensibly evade the immune system and breach the maternalCfetal interface. The primary barrier between the maternal and fetal compartments during pregnancy is the fetally derived placenta that is adjacent to and intercalated with the maternal decidua. Fetal macrophages (Hofbauer cells), placental fibroblasts, fetal endothelial cells and syncytiotrophoblasts, together with decidual stromal cells, macrophages, and lymphocytes of maternal origin, protect the fetus from pathogens present in maternal blood7C9. Several studies in animal models have demonstrated vertical transmission of ZIKV and its tropism for placental cells, including trophoblasts, endothelial cells, and macrophages10C15. Once ZIKV crosses the placental barrier, it can infect Kv3 modulator 4 neuronal progenitor cells in the fetal brain10,12,16C18. ZIKV and the closely related flavivirus DENV co-circulate in the same geographic ranges and are transmitted by the same mosquitoes. ZIKV and the four serotypes of dengue virus (DENV1C4) share 55.1C56.3% amino acid sequence identity. The adaptive immune response to DENV and its roles in protection versus pathogenesis is complex and remains incompletely understood19. Epidemiological data indicate that following primary infection by one DENV serotype, a second an infection using a different DENV serotype might trigger a far more serious type of dengue disease, revealing potential assignments for antibodies (Abs) and T cells in DENV pathogenesis. Two hypotheses have already been proposed to describe this sensation: Ab-dependent improvement (ADE) and T cell primary antigenic sin (TOAS). Many reports support the ADE model20C24 as the function for T cells continues to be less clear. Certainly, latest data indicate defensive assignments for serotype-specific and cross-reactive T cells against DENV infection in mice31C37 and individuals25C30. The role of T cells in ZIKV immunity continues to be explored in animal choices also. In nonhuman primates, the top of the Compact disc8+ T cell activation correlates with ZIKV RNA decrease, suggesting a defensive function for Compact disc8+ T cells in managing ZIKV replication38. In mice, Compact disc8+ T Kv3 modulator 4 cells broaden, display high cytolytic activity, and mediate viral clearance39. Predicated on amino acidity series and structural commonalities between ZIKV and DENV, many groups show cross-reactivity between DENV and ZIKV in both humoral40C45 and mobile replies46C49. One research in nonhuman primates demonstrated that preceding DENV exposure led to a decrease in the length of time of ZIKV viremia in DENV-immune pets, suggesting cross-protection50, although another combined group reported even more natural ramifications of DENV immunity on ZIKV infection and disease Ifng pathogenesis51. Research in mice show that DENV/ZIKV cross-reactive Abs can boost ZIKV pathogenesis41,42, whereas DENV/ZIKV cross-reactive Compact disc8+ T cells drive back ZIKV an infection46,49,52. Nevertheless, no study provides examined (i) how prior DENV publicity affects maternal and fetal final result of ZIKV an infection in being pregnant and (ii) the contribution of Compact disc8+ T cells to safeguard against or pathogenesis of ZIKV an infection during pregnancy. Appropriately, we investigated the final results of ZIKV infection during pregnancy in non-immune and DENV-immune mice using short-term sequential infection choices. Contact with DENV conferred security against maternal Prior.