Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Carolyn D. Kramer is active.

Publication


Featured researches published by Carolyn D. Kramer.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2014

Interleukin 1 Receptor 1 and Interleukin 1β Regulate Megakaryocyte Maturation, Platelet Activation, and Transcript Profile During Inflammation in Mice and Humans

Lea M. Beaulieu; Elaine Lin; Eric Mick; Milka Koupenova; Ellen O. Weinberg; Carolyn D. Kramer; Caroline Attardo Genco; Martin G. Larson; Emelia J. Benjamin; Jane E. Freedman

Objective— Interleukin 1 Receptor 1 (IL1R1) and its ligand, IL1&bgr;, are upregulated in cardiovascular disease, obesity, and infection. Previously, we reported a higher level of IL1R1 transcripts in platelets from obese individuals of the Framingham Heart Study (FHS), but its functional effect in platelets has never been described. Additionally, IL1&bgr; levels are increased in atherosclerotic plaques and in bacterial infections. The aim of this work is to determine whether IL1&bgr;, through IL1R1, can activate platelets and megakaryocytes to promote atherothrombosis. Approach and Results— We found that IL1&bgr;-related genes from platelets, as measured in 1819 FHS participants, were associated with increased body mass index, and a direct relationship was shown in wild-type mice fed a high-fat diet. Mechanistically, IL1&bgr; activated nuclear factor-&kgr;B and mitogen-activated protein kinase signaling pathways in megakaryocytes. IL1&bgr;, through IL1R1, increased ploidy of megakaryocytes to 64+ N by 2-fold over control. IL1&bgr; increased agonist-induced platelet aggregation by 1.2-fold with thrombin and 4.2-fold with collagen. IL1&bgr; increased adhesion to both collagen and fibrinogen, and heterotypic aggregation by 1.9-fold over resting. High fat diet-enhanced platelet adhesion was absent in IL1R1−/− mice. Wild-type mice infected with Porphyromonas gingivalis had circulating heterotypic aggregates (1.5-fold more than control at 24 hours and 6.2-fold more at 6 weeks) that were absent in infected IL1R1−/− and IL1&bgr;−/− mice. Conclusions— In summary, IL1R1- and IL1&bgr;-related transcripts are elevated in the setting of obesity. IL1R1/IL1&bgr; augment both megakaryocyte and platelet functions, thereby promoting a prothrombotic environment during infection and obesity; potentially contributing to the development of atherothrombotic disease.


PLOS Pathogens | 2014

Distinct Lipid A Moieties Contribute to Pathogen-Induced Site-Specific Vascular Inflammation

Connie Slocum; Stephen R. Coats; Ning Hua; Carolyn D. Kramer; George K. Papadopoulos; Ellen O. Weinberg; Cynthia V. Gudino; James A. Hamilton; Richard P. Darveau; Caroline Attardo Genco

Several successful pathogens have evolved mechanisms to evade host defense, resulting in the establishment of persistent and chronic infections. One such pathogen, Porphyromonas gingivalis, induces chronic low-grade inflammation associated with local inflammatory bone loss and systemic inflammation manifested as atherosclerosis. P. gingivalis expresses an atypical lipopolysaccharide (LPS) structure containing heterogeneous lipid A species, that exhibit Toll-like receptor-4 (TLR4) agonist or antagonist activity, or are non-activating at TLR4. In this study, we utilized a series of P. gingivalis lipid A mutants to demonstrate that antagonistic lipid A structures enable the pathogen to evade TLR4-mediated bactericidal activity in macrophages resulting in systemic inflammation. Production of antagonistic lipid A was associated with the induction of low levels of TLR4-dependent proinflammatory mediators, failed activation of the inflammasome and increased bacterial survival in macrophages. Oral infection of ApoE−/− mice with the P. gingivalis strain expressing antagonistic lipid A resulted in vascular inflammation, macrophage accumulation and atherosclerosis progression. In contrast, a P. gingivalis strain producing exclusively agonistic lipid A augmented levels of proinflammatory mediators and activated the inflammasome in a caspase-11-dependent manner, resulting in host cell lysis and decreased bacterial survival. ApoE−/− mice infected with this strain exhibited diminished vascular inflammation, macrophage accumulation, and atherosclerosis progression. Notably, the ability of P. gingivalis to induce local inflammatory bone loss was independent of lipid A expression, indicative of distinct mechanisms for induction of local versus systemic inflammation by this pathogen. Collectively, our results point to a pivotal role for activation of the non-canonical inflammasome in P. gingivalis infection and demonstrate that P. gingivalis evades immune detection at TLR4 facilitating chronic inflammation in the vasculature. These studies support the emerging concept that pathogen-mediated chronic inflammatory disorders result from specific pathogen-mediated evasion strategies resulting in low-grade chronic inflammation.


Experimental Parasitology | 2008

Dermacentor variabilis: Regulation of fibroblast migration by tick salivary gland extract and saliva

Carolyn D. Kramer; Zachary Nahmias; Derek D. Norman; Tara A. Mulvihill; Lewis B. Coons; Judith A. Cole

We examined the effects of tick SGx and saliva on basal- and platelet-derived growth factor (PDGF)-stimulated cell migration and extracellular signal-regulated kinase (ERK) signaling in fibroblasts. Repair of injured monolayers was delayed by SGx pretreatment and was not associated with reductions in cell number. In migration assays, SGx suppressed both basal- and PDGF-stimulated fibroblast movement. Furthermore, SGx and saliva reduced PDGF-stimulated ERK activity. Thus, the delayed repair of monolayer injuries resulted from SGx inhibiting fibroblast migratory responses to chemotactic signals. SGx also suppressed injury- and growth factor-induced ERK activation in renal epithelial OK cells. Our data suggest that maintenance of the tick feeding lesion results, in part, from suppressing ERK signaling and fibroblast migration, events playing integral roles in the wound healing response. The effects of SGx on cells not involved in wound healing suggest that a constituent(s) in tick saliva has global effects on the ERK signaling pathway.


Experimental Parasitology | 2011

Tick saliva regulates migration, phagocytosis, and gene expression in the macrophage-like cell line, IC-21

Carolyn D. Kramer; Nina M. Poole; Lewis B. Coons; Judith A. Cole

We studied the effects of tick saliva on cell migration, cell signaling, phagocytosis, and gene expression in the murine macrophage cell line, IC-21. Saliva increased both basal- and platelet-derived growth factor (PDGF)-stimulated migration in IC-21 cells. However, saliva did not affect PDGF-stimulated extracellular signal-regulated kinase (ERK) activity. Zymosan-mediated interleukin-1 receptor associated kinase (IRAK) activity increased when cells were pretreated with saliva. Saliva suppressed phagocytosis of zymosan particles by IC-21 cells. An RT(2) Profiler™ PCR Array revealed that saliva regulates gene expression in a manner consistent with an immune response skewed toward a Th2 reaction, which is characterized by production of anti-inflammatory cytokines IL-4 and IL-10. Our results using IC-21 cells suggest that Dermacentor variabilis has evolved a mechanism for regulating macrophage function, which may contribute to the ticks ability to modulate immune function.


Ticks and Tick-borne Diseases | 2013

Effects of tick saliva on the migratory and invasive activity of Saos-2 osteosarcoma and MDA-MB-231 breast cancer cells.

Nina M. Poole; Lilian Nyindodo-Ogari; Carolyn D. Kramer; Lewis B. Coons; Judith A. Cole

In previous studies we showed that tick saliva modulates the migratory activity of cells involved in the wound healing response. Since cell migration is a prerequisite for tumor invasion and metastasis, we examined the effects of tick saliva on the migratory and invasive activity of Saos-2 osteosarcoma and MDA-MB-231 (MB-231) breast cancer cells and the potential signaling pathways that may be affected. Saliva inhibited basal and agonist-induced Saos-2 and MB-231 migration and invasion through a matrigel-coated filter. In the Saos-2 cells, saliva suppressed epidermal growth factor (EGF)-activation of Akt/Protein Kinase B, however, only basal extracellular signal-regulated kinase (ERK) activity was affected in MB-231 cells. EGF receptor (EGFR) overexpression masked the effect of saliva on MB-231 cells, but its ability to inhibit MB-231 migration was enhanced by the EGFR inhibitor PD 168393 and MEK inhibitor U0126. Our data indicate that the mechanisms ticks have evolved to regulate the wound healing response have generalized effects on the migratory and invasive activities of metastatic cancer cells.


BMC Genomics | 2014

Distinct gene signatures in aortic tissue from ApoE-/- mice exposed to pathogens or Western diet

Carolyn D. Kramer; Ellen O. Weinberg; Adam C. Gower; Xianbao He; Samrawit Mekasha; Connie Slocum; Lea M. Beaulieu; Lee M. Wetzler; Yuriy O. Alekseyev; Frank C. Gibson; Jane E. Freedman; Robin R. Ingalls; Caroline Attardo Genco

BackgroundAtherosclerosis is a progressive disease characterized by inflammation and accumulation of lipids in vascular tissue. Porphyromonas gingivalis (Pg) and Chlamydia pneumoniae (Cp) are associated with inflammatory atherosclerosis in humans. Similar to endogenous mediators arising from excessive dietary lipids, these Gram-negative pathogens are pro-atherogenic in animal models, although the specific inflammatory/atherogenic pathways induced by these stimuli are not well defined. In this study, we identified gene expression profiles that characterize P. gingivalis, C. pneumoniae, and Western diet (WD) at acute and chronic time points in aortas of Apolipoprotein E (ApoE-/-) mice.ResultsAt the chronic time point, we observed that P. gingivalis was associated with a high number of unique differentially expressed genes compared to C. pneumoniae or WD. For the top 500 differentially expressed genes unique to each group, we observed a high percentage (76%) that exhibited decreased expression in P. gingivalis-treated mice in contrast to a high percentage (96%) that exhibited increased expression in WD mice. C. pneumoniae treatment resulted in approximately equal numbers of genes that exhibited increased and decreased expression. Gene Set Enrichment Analysis (GSEA) revealed distinct stimuli-associated phenotypes, including decreased expression of mitochondrion, glucose metabolism, and PPAR pathways in response to P. gingivalis but increased expression of mitochondrion, lipid metabolism, carbohydrate and amino acid metabolism, and PPAR pathways in response to C. pneumoniae; WD was associated with increased expression of immune and inflammatory pathways. DAVID analysis of gene clusters identified by two-way ANOVA at acute and chronic time points revealed a set of core genes that exhibited altered expression during the natural progression of atherosclerosis in ApoE-/- mice; these changes were enhanced in P. gingivalis-treated mice but attenuated in C. pneumoniae-treated mice. Notable differences in the expression of genes associated with unstable plaques were also observed among the three pro-atherogenic stimuli.ConclusionsDespite the common outcome of P. gingivalis, C. pneumoniae, and WD on the induction of vascular inflammation and atherosclerosis, distinct gene signatures and pathways unique to each pro-atherogenic stimulus were identified. Our results suggest that pathogen exposure results in dysregulated cellular responses that may impact plaque progression and regression pathways.


Journal of Visualized Experiments | 2014

A Mouse Model for Pathogen-induced Chronic Inflammation at Local and Systemic Sites

George K. Papadopoulos; Carolyn D. Kramer; Connie S. Slocum; Ellen O. Weinberg; Ning Hua; Cynthia V. Gudino; James A. Hamilton; Caroline Attardo Genco

Chronic inflammation is a major driver of pathological tissue damage and a unifying characteristic of many chronic diseases in humans including neoplastic, autoimmune, and chronic inflammatory diseases. Emerging evidence implicates pathogen-induced chronic inflammation in the development and progression of chronic diseases with a wide variety of clinical manifestations. Due to the complex and multifactorial etiology of chronic disease, designing experiments for proof of causality and the establishment of mechanistic links is nearly impossible in humans. An advantage of using animal models is that both genetic and environmental factors that may influence the course of a particular disease can be controlled. Thus, designing relevant animal models of infection represents a key step in identifying host and pathogen specific mechanisms that contribute to chronic inflammation. Here we describe a mouse model of pathogen-induced chronic inflammation at local and systemic sites following infection with the oral pathogen Porphyromonas gingivalis, a bacterium closely associated with human periodontal disease. Oral infection of specific-pathogen free mice induces a local inflammatory response resulting in destruction of tooth supporting alveolar bone, a hallmark of periodontal disease. In an established mouse model of atherosclerosis, infection with P. gingivalis accelerates inflammatory plaque deposition within the aortic sinus and innominate artery, accompanied by activation of the vascular endothelium, an increased immune cell infiltrate, and elevated expression of inflammatory mediators within lesions. We detail methodologies for the assessment of inflammation at local and systemic sites. The use of transgenic mice and defined bacterial mutants makes this model particularly suitable for identifying both host and microbial factors involved in the initiation, progression, and outcome of disease. Additionally, the model can be used to screen for novel therapeutic strategies, including vaccination and pharmacological intervention.


PLOS ONE | 2015

Specific Inflammatory Stimuli Lead to Distinct Platelet Responses in Mice and Humans.

Lea M. Beaulieu; Lauren Clancy; Emelia J. Benjamin; Carolyn D. Kramer; Ellen O. Weinberg; Xianbao He; Samrawit Mekasha; Eric Mick; Robin R. Ingalls; Caroline Attardo Genco; Jane E. Freedman

Introduction Diverse and multi-factorial processes contribute to the progression of cardiovascular disease. These processes affect cells involved in the development of this disease in varying ways, ultimately leading to atherothrombosis. The goal of our study was to compare the differential effects of specific stimuli – two bacterial infections and a Western diet – on platelet responses in ApoE-/- mice, specifically examining inflammatory function and gene expression. Results from murine studies were verified using platelets from participants of the Framingham Heart Study (FHS; n = 1819 participants). Methods Blood and spleen samples were collected at weeks 1 and 9 from ApoE-/- mice infected with Porphyromonas gingivalis or Chlamydia pneumoniae and from mice fed a Western diet for 9 weeks. Transcripts based on data from a Western diet in ApoE-/- mice were measured in platelet samples from FHS using high throughput qRT-PCR. Results At week 1, both bacterial infections increased circulating platelet-neutrophil aggregates. At week 9, these cells individually localized to the spleen, while Western diet resulted in increased platelet-neutrophil aggregates in the spleen only. Microarray analysis of platelet RNA from infected or Western diet-fed mice at week 1 and 9 showed differential profiles. Genes, such as Serpina1a, Ttr, Fgg, Rpl21, and Alb, were uniquely affected by infection and diet. Results were reinforced in platelets obtained from participants of the FHS. Conclusion Using both human studies and animal models, results demonstrate that variable sources of inflammatory stimuli have the ability to influence the platelet phenotype in distinct ways, indicative of the diverse function of platelets in thrombosis, hemostasis, and immunity.


Anaerobe | 2017

Distinct roles for dietary lipids and Porphyromonas gingivalis infection on atherosclerosis progression and the gut microbiota

Carolyn D. Kramer; Alexandra M. Simas; Xianbao He; Robin R. Ingalls; Ellen O. Weinberg; Caroline Attardo Genco


Arteriosclerosis, Thrombosis, and Vascular Biology | 2014

Abstract 272: Targeted Activatable Cell-Penetrating Peptides Identify Atherosclerotic Plaques in Mice with Noninvasive Magnetic Resonance Imaging

Heidi A. Schwanz; Ning Hua; Connie Slocum; Carolyn D. Kramer; Michael Whitney; Tao Jiang; Caroline Attardo Genco; Roger Y. Tsien; James A. Hamilton

Collaboration


Dive into the Carolyn D. Kramer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jane E. Freedman

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lea M. Beaulieu

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge