Searched for: person:cammem01
Inhibitory effect of capsule on natural transformation of Streptococcus pneumoniae
Ma, Sheya Xiao; Eichner, Hannes; Cammer, Michael; Weiser, Jeffrey N
The capsule of Streptococcus pneumoniae (Spn) is highly heterogeneous based on the expression of distinct polysaccharides. Spn transformation, controlled by the Com regulon, has been predominantly studied using unencapsulated laboratory strains. However, genomic studies revealed different rates of recombination events in clinical isolates of different serotypes. As these isolates were genetically distinct beyond capsule-encoding genes, the exact relationship between transformation and capsule remains unclear. Herein, we compared the transformability of a collection of isogenic capsule-switch strains. Strains with different capsule types and amounts significantly differed in their transformation frequency, with the unencapsulated strain having a higher frequency compared to encapsulated strains. A GFP-reporter of each strain monitoring the expression of a Com regulon-controlled gene showed similar kinetics, indicating differences in transformability were due to processes downstream of competence activation. The Com pilus, induced by competence, binds and takes in the donor DNA and is the central component of the transformation apparatus. The surface exposure of Com pilus significantly differed among serotypes with highly transformable strains having more cells binding ComGC antibody. Furthermore, electron microscopy demonstrated that transformability correlated with the proportion of cells bearing a Com pilus, which was affected by both the presence of capsule and serotype. Additionally, the unencapsulated strain displayed longer pili than encapsulated strains. Examination of capsule porosity revealed that serotypes with higher transformation frequencies had more porous capsules. Together, these results indicate that the capsule interferes with the assembly of Com pilus, thereby inhibiting the natural transformation of Spn.IMPORTANCEThe capsule is a major virulence factor of Streptococcus pneumoniae (Spn), providing a physical shield and exhibiting extensive diversity across at least 100 serotypes. Although natural transformation of Spn has predominantly been characterized in unencapsulated laboratory strains, clinical encapsulated isolates also exhibit transformability and demonstrate varied recombination rates during host carriage. We utilized otherwise genetically identical capsule-switch strains to isolate the effect of capsule on transformation. We demonstrate serotype- and quantity-dependent inhibition of transformation by the capsule, mediated through hindrance with the transformation pilus assembly and function. This study challenges the paradigm that unencapsulated laboratory strains fully recapitulate natural transformation dynamics. By redefining the capsule as a multifunctional modulator of Spn biology, balancing virulence and adaptability, our findings advance our understanding of pneumococcal evolution.
PMID: 40874614
ISSN: 2150-7511
CID: 5910472
Proximity between LAG-3 and the T cell receptor guides suppression of T cell activation and autoimmunity
Du, Jasper; Chen, Hui; You, Jia; Hu, Wei; Liu, Jia; Lu, Qiao; Zhang, Yong; Gao, Jie; Lin, Meng-Ju; Foster, Connor James Ryan; Rao, Eric; Cammer, Michael; Yin, Weiwei; Koide, Shohei; Lu, Catherine Pei-Ju; Chen, Wei; Lou, Jizhong; Wang, Jun
Therapeutically targeting pathogenic T cells in autoimmune diseases has been challenging. Although LAG-3, an inhibitory checkpoint receptor specifically expressed on activated T cells, is known to bind to major histocompatibility complex class II (MHC class II), we demonstrate that MHC class II interaction alone is insufficient for optimal LAG-3 function. Instead, LAG-3's spatial proximity to T cell receptor (TCR) but not CD4 co-receptor, facilitated by cognate peptide-MHC class II, is crucial in mediating CD4+ T cell suppression. Mechanistically, LAG-3 forms condensate with TCR signaling component CD3ε through its intracellular FSAL motif, disrupting CD3ε/lymphocyte-specific protein kinase (Lck) association. To exploit LAG-3's proximity to TCR and maximize LAG-3-dependent T cell suppression, we develop an Fc-attenuated LAG-3/TCR inhibitory bispecific antibody to bypass the requirement of cognate peptide-MHC class II. This approach allows for potent suppression of both CD4+ and CD8+ T cells and effectively alleviates autoimmune symptoms in mouse models. Our findings reveal an intricate and conditional checkpoint modulatory mechanism and highlight targeting of LAG-3/TCR cis-proximity for T cell-driven autoimmune diseases lacking effective and well-tolerated immunotherapies.
PMID: 40592325
ISSN: 1097-4172
CID: 5887772
De novo assembly of RNA m6A modification factors into viral genome-associated nuclear bodies drives HCMV RNA accumulation
Grande, Rebecca C; Lin, Chia-Ching; Cammer, Michael; Emesom, Ebube D; Khurram, Maaz Asher; Boutell, Chris; Denes, Lance T; Lionnet, Timothée; Wilson, Angus C; Mohr, Ian
The factors that install and recognize N6-methyladenosine (m6A) on RNA to regulate gene expression are well characterized, but how their spatial organization responds to physiological stress, including infection, is unclear. Here, we show that human cytomegalovirus (HCMV) infection induces accumulation of m6A methyltransferase subunits, including WTAP, together with nuclear m6A reader YTHDC1, into distinctive, membraneless nuclear bodies (NBs) overlapping with incoming virus genomes and immediate-early (IE) RNA transcripts. De novo assembly and integrity of these DNA-associated, IE, virus-activated NBs requires RNAPII transcription, METTL3 m6A methyltransferase activity, and m6A recognition by YTHDC1, but not new protein synthesis. Depleting YTHDC1 or WTAP limits the accumulation of critical HCMV RNAs required for virus DNA replication, interfering with virus reproduction. This reveals a surprising strategy whereby a discrete sub-nuclear RNA biogenesis compartment replete with RNAPII and m6A modification components is swiftly consolidated in proximity to infecting HCMV genomes to initialize and sustain virus gene expression.
PMID: 40560728
ISSN: 2211-1247
CID: 5889262
Paracrine regulations of IFN-γ secreting CD4+ T cells by lumican and biglycan are protective in allergic contact dermatitis
Maiti, George; Frikeche, Jihane; Loomis, Cynthia; Cammer, Michael; Eichman, Stephanie L; Chakravarti, Shukti
Allergic contact dermatitis (ACD) is a delayed-type IV hypersensitivity response driven by innate and adaptive immune cells. While specific immune regulations of these cell types are amply elucidated, their regulations by extracellular matrix (ECM) components and T cell mediated adaptive immunity in ACD remains unclear. Lumican and biglycan are ECM proteoglycans abundant in the dermis and lymph node, known to regulate innate immune myeloid cells, but have not been investigated in lymphoid cell regulations in ACD. By immunohistology we localized lumican and biglycan in skin biopsies of psoriatic patients. Using wild type (WT), lumican and biglycan knockout mice, we investigated CD4+T cell infiltration, activation and proliferation in the skin and draining lymph node (dLN) of CHS-challenged mice by immunohistochemistry and flow cytometry. We used the OT-II adoptive transfer model to test antigen specific CD4+T cell activation. We assessed interactions of the proteoglycans with LFA-1 on T cells by confocal microscopy. Compared to WTs, the knockouts showed severe ear inflammation, with increased CD4+T cells infiltration in the dermis. CHS-challenged knockout mice dLN showed increased T-bet, STAT1 and -STAT4 signaling, indicating enhanced Th1 commitment and proliferation. We found that WT lymph node fibroblastic reticular cells (FRCs) secrete lumican, biglycan and decorin, a related proteoglycan, while none are expressed by naive or activated T cells. Lumican and biglycan interact with LFA-1 on T cell surfaces, and in vitro all three proteoglycans suppress CD4+T cell activation. Secreted by dLN FRCs, lumican, biglycan, and possibly decorin interact with LFA-1 on CD4+T cells to restrict its activation and reduce dermatitis severity.
PMID: 40518026
ISSN: 1569-1802
CID: 5870662
Exercise-induced dysregulation of the adrenergic response in a mouse model of PKP2-arrhythmogenic cardiomyopathy
van Opbergen, Chantal Jm; Gutierrez, Lilian K; Bertoli, Giorgia; Zhang, Mingliang; Boyce, Sarah; Deng, Yan; Cammer, Michael; Liang, Feng-Xia; Delmar, Mario
BACKGROUND:Plakophilin-2 (PKP2) is a component of the desmosome. Pathogenic variants can lead to arrhythmogenic cardiomyopathy (PKP2-ACM). In PKP2-ACM patients, exercise and catecholamine surges negatively impact arrhythmia incidence and severity. OBJECTIVE:To characterize remodeling of the sympathetic input and adrenergic response in hearts of PKP2-deficient mice (PKP2cKO) subjected to endurance exercise. METHODS:transient dynamics. Separately, we evaluated distribution of sympathetic terminals in PKP2cKO trained hearts vs controls. RESULTS:Exercise led to increased abundance of sarcolemma β1-ARs in control, and decreased abundance in PKP2cKO-myocytes. OCT3 knockdown drastically reduced the response of trained PKP2cKO-myocytes to norepinephrine but not isoproterenol, indicating preserved response to native catecholamines by intracellular (dyad-associated) receptors in the setting of a reduced sarcolemma pool. In tissue, we observed reduced abundance of sympathetic terminals, and heterogeneous distribution across the myocardium. CONCLUSION/CONCLUSIONS:Endurance exercise in PKP2-deficient myocytes leads to reduced pool of functional β1-ARs in the sarcolemma and yet availability of intracellular receptors, which can activate selected (and heterogeneous) routes of intracellular signaling cascades. We speculate that remodeling of nerve terminals affects sympathetic input distribution and hence, regional modulation of excitability and conduction. These changes can facilitate cell-generated triggered activity and heterogeneity of the underlying substrate, setting the stage for life-threatening arrhythmias.
PMID: 40383179
ISSN: 1556-3871
CID: 5852682
Oxytocin induces embryonic diapause
Minder, Jessica L; Winokur, Sarah B; Stephens, Janaye; Tong, Jie; Cassel, Naomi L; Schuster, Luisa; Issa, Habon A; Cammer, Michael; Khatri, Latika; Moisan, Gaia; Alvarado-Torres, Maria; Aristizábal, Orlando; Wadghiri, Youssef Z; Kim, Sang Yong; Valtcheva, Silvana; Lu, Catherine Pei-Ju; Chao, Moses V; Froemke, Robert C
Embryonic development in many species, including case reports in humans, can be temporarily halted before implantation during a process called diapause. Facultative diapause occurs under conditions of maternal metabolic stress such as nursing. While molecular mechanisms of diapause have been studied, a natural inducing factor has yet to be identified. Here, we show that oxytocin induces embryonic diapause in mice. We show that gestational delays were triggered during nursing or optogenetic stimulation of oxytocin neurons simulating nursing patterns. Mouse blastocysts express oxytocin receptors, and oxytocin induced delayed implantation-like dispersion in cultured embryos. Last, oxytocin receptor-knockout embryos transferred into wild-type surrogates had low survival rates during diapause. Our results indicate that oxytocin coordinates timing of embryonic development with uterine progression through pregnancy, providing an evolutionarily conserved mechanism for ensuring successful reproduction.
PMCID:11881891
PMID: 40043121
ISSN: 2375-2548
CID: 5809752
Intra-serotype variation of Streptococcus pneumoniae capsule and its quantification
Eichner, Hannes; Wu, Cindy; Cammer, Michael; Tran, Elizabeth N H; Hirst, Timothy R; Paton, James C; Weiser, Jeffrey N
UNLABELLED: IMPORTANCE/OBJECTIVE:research should consider capsule size, not just its presence and type. The results imply that standardized vaccine efficacy tests may yield variable results depending on the capsule production of target strains.
PMID: 39950804
ISSN: 2165-0497
CID: 5793962
De novo assembly of RNA m6A modification factors into viral genome-associated nuclear bodies drives HCMV RNA accumulation
Grande, Rebecca C.; Lin, Chia-Ching; Cammer, Michael; Emesom, Ebube D.; Khurram, Maaz Asher; Boutell, Chris; Denes, Lance T.; Lionnet, Timothee; Wilson, Angus C.; Mohr, Ian
ISI:001537879400005
ISSN: 2211-1247
CID: 5905992
ABD-3, the confluence of powerful antibacterial modalities: ABDs delivering and expressing lss, the gene encoding lysostaphin
Ram, Geeta; Chiu, LiTing; Dey, Somrita; Ross, Hope F; Cammer, Michael; Novick, Richard P
In response to the antimicrobial resistance crisis, we have developed a powerful and versatile therapeutic platform, the Antibacterial Drone (ABD) system. The ABD consists of a highly mobile staphylococcal pathogenicity island re-purposed to deliver genes encoding antibacterial proteins. The chromosomally located island is induced by a co-resident helper phage, packaged in phage-like particles, and released in very high numbers upon phage-induced lysis. ABD particles specifically adsorb to bacteria causing an infection and deliver their DNA to these bacteria, where the bactericidal cargo genes are expressed, kill the bacteria, and cure the infection. Here, we report a major advance of the system, incorporation of the gene encoding a secreted, bactericidal, species-specific lytic enzyme, lysostsphin. This ABD not only kills the bacterium that has been attacked by the ABD, but also any surrounding bacteria that are sensitive to the lytic enzyme which is released by secretion and by lysis of the doomed cell. So while the killing field is thus expanded, there are no civilian casualties (bacteria that are insensitive to the ABD and its cargo protein(s) are not inadvertently killed). Without amplifying the number of ABD particles (which are not re-packaged), the expression and release of the cargo gene's product dramatically extend the effective reach of the ABD. A cargo gene that encodes a secreted bactericidal protein also enables the treatment of a mixed bacterial infection in which one of the infecting organisms is insensitive to the ABD delivery system but is sensitive to the ABD's secreted cargo protein.
PMCID:11373205
PMID: 39072634
ISSN: 1098-6596
CID: 5687332
Pulses of RhoA signaling stimulate actin polymerization and flow in protrusions to drive collective cell migration
Qian, Weiyi; Yamaguchi, Naoya; Lis, Patrycja; Cammer, Michael; Knaut, Holger
In animals, cells often move as collectives to shape organs, close wounds, or-in the case of disease-metastasize. To accomplish this, cells need to generate force to propel themselves forward. The motility of singly migrating cells is driven largely by an interplay between Rho GTPase signaling and the actin network. Whether cells migrating as collectives use the same machinery for motility is unclear. Using the zebrafish posterior lateral line primordium as a model for collective cell migration, we find that active RhoA and myosin II cluster on the basal sides of the primordium cells and are required for primordium motility. Positive and negative feedbacks cause RhoA and myosin II activities to pulse. These pulses of RhoA signaling stimulate actin polymerization at the tip of the protrusions and myosin-II-dependent actin flow and protrusion retraction at the base of the protrusions and deform the basement membrane underneath the migrating primordium. This suggests that RhoA-induced actin flow on the basal sides of the cells constitutes the motor that pulls the primordium forward, a scenario that likely underlies collective migration in other contexts.
PMID: 38096821
ISSN: 1879-0445
CID: 5588892