Network


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

Hotspot


Dive into the research topics where Carmen Barnes is active.

Publication


Featured researches published by Carmen Barnes.


Nature Nanotechnology | 2014

In vivo endothelial siRNA delivery using polymeric nanoparticles with low molecular weight

James E. Dahlman; Carmen Barnes; Omar F. Khan; Aude Thiriot; Siddharth Jhunjunwala; Taylor E. Shaw; Yiping Xing; Hendrik B. Sager; Gaurav Sahay; Andrew Bader; Roman L. Bogorad; Hao Yin; Tim Racie; Yizhou Dong; Shan Jiang; Danielle Seedorf; Apeksha Dave; Kamaljeet Singh Sandhu; Matthew J. Webber; Tatiana Novobrantseva; Vera M. Ruda; Abigail K. R. Lytton-Jean; Christopher G. Levins; Brian T. Kalish; Dayna K. Mudge; Mario Perez; Ludmila Abezgauz; Partha Dutta; Lynelle Smith; Klaus Charisse

Dysfunctional endothelium contributes to more diseases than any other tissue in the body. Small interfering RNAs (siRNAs) can help in the study and treatment of endothelial cells in vivo by durably silencing multiple genes simultaneously, but efficient siRNA delivery has so far remained challenging. Here, we show that polymeric nanoparticles made of low-molecular-weight polyamines and lipids can deliver siRNA to endothelial cells with high efficiency, thereby facilitating the simultaneous silencing of multiple endothelial genes in vivo. Unlike lipid or lipid-like nanoparticles, this formulation does not significantly reduce gene expression in hepatocytes or immune cells even at the dosage necessary for endothelial gene silencing. These nanoparticles mediate the most durable non-liver silencing reported so far and facilitate the delivery of siRNAs that modify endothelial function in mouse models of vascular permeability, emphysema, primary tumour growth and metastasis.


Proceedings of the National Academy of Sciences of the United States of America | 2008

PPARα agonist fenofibrate suppresses tumor growth through direct and indirect angiogenesis inhibition

Dipak Panigrahy; Arja Kaipainen; Sui Huang; Catherine Butterfield; Carmen Barnes; Michael Fannon; Andrea Laforme; Deviney Chaponis; Judah Folkman; Mark W. Kieran

Angiogenesis and inflammation are central processes through which the tumor microenvironment influences tumor growth. We have demonstrated recently that peroxisome proliferator-activated receptor (PPAR)α deficiency in the host leads to overt inflammation that suppresses angiogenesis via excess production of thrombospondin (TSP)-1 and prevents tumor growth. Hence, we speculated that pharmacologic activation of PPARα would promote tumor growth. Surprisingly, the PPARα agonist fenofibrate potently suppressed primary tumor growth in mice. This effect was not mediated by cancer-cell-autonomous antiproliferative mechanisms but by the inhibition of angiogenesis and inflammation in the host tissue. Although PPARα-deficient tumors were still susceptible to fenofibrate, absence of PPARα in the host animal abrogated the potent antitumor effect of fenofibrate. In addition, fenofibrate suppressed endothelial cell proliferation and VEGF production, increased TSP-1 and endostatin, and inhibited corneal neovascularization. Thus, both genetic abrogation of PPARα as well as its activation by ligands cause tumor suppression via overlapping antiangiogenic pathways. These findings reveal the potential utility of the well tolerated PPARα agonists beyond their use as lipid-lowering drugs in anticancer therapy. Our results provide a mechanistic rationale for evaluating the clinical benefits of PPARα agonists in cancer treatment, alone and in combination with other therapies.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Lipopeptide nanoparticles for potent and selective siRNA delivery in rodents and nonhuman primates

Yizhou Dong; Kevin Love; J. Robert Dorkin; Sasilada Sirirungruang; Yunlong Zhang; Delai Chen; Roman L. Bogorad; Hao Yin; Yi Chen; Arturo Vegas; Christopher A. Alabi; Gaurav Sahay; Karsten Olejnik; Weiheng Wang; Avi Schroeder; Abigail K. R. Lytton-Jean; Daniel J. Siegwart; Akin Akinc; Carmen Barnes; Scott Barros; Mary Carioto; Kevin Fitzgerald; Julia Hettinger; Varun Kumar; Tatiana Novobrantseva; June Qin; William Querbes; Victor Koteliansky; Robert Langer; Daniel G. Anderson

Significance The safe, selective, and efficient delivery of siRNA is a key challenge to the broad application of siRNA therapeutics in humans. Motivated by the structure of lipoproteins, we developed lipopeptide nanomaterials for siRNA delivery. In vivo in mice, siRNA–lipopeptide particles provide the most potent delivery to hepatocytes (ED50 ∼ 0.002 mg/kg for FVII silencing), with the highest selectivity of delivery to hepatocytes over nontarget cell types (orders of magnitude), yet reported. These materials also show efficacy in nonhuman primates. siRNA therapeutics have promise for the treatment of a wide range of genetic disorders. Motivated by lipoproteins, we report lipopeptide nanoparticles as potent and selective siRNA carriers with a wide therapeutic index. Lead material cKK-E12 showed potent silencing effects in mice (ED50 ∼ 0.002 mg/kg), rats (ED50 < 0.01 mg/kg), and nonhuman primates (over 95% silencing at 0.3 mg/kg). Apolipoprotein E plays a significant role in the potency of cKK-E12 both in vitro and in vivo. cKK-E12 was highly selective toward liver parenchymal cell in vivo, with orders of magnitude lower doses needed to silence in hepatocytes compared with endothelial cells and immune cells in different organs. Toxicity studies showed that cKK-E12 was well tolerated in rats at a dose of 1 mg/kg (over 100-fold higher than the ED50). To our knowledge, this is the most efficacious and selective nonviral siRNA delivery system for gene silencing in hepatocytes reported to date.


Journal of Clinical Investigation | 2012

Epoxyeicosanoids stimulate multiorgan metastasis and tumor dormancy escape in mice

Dipak Panigrahy; Matthew L. Edin; Craig R. Lee; Sui Huang; Diane R. Bielenberg; Catherine Butterfield; Carmen Barnes; Akiko Mammoto; Ayala Luria; Ofra Benny; Deviney Chaponis; Andrew C. Dudley; Emily R. Greene; Jo-Anne Vergilio; Giorgio Pietramaggiori; Sandra S. Scherer-Pietramaggiori; Sarah Short; Meetu Seth; Fred B. Lih; Kenneth B. Tomer; Jun Yang; Reto A. Schwendener; Bruce D. Hammock; John R. Falck; Vijaya L. Manthati; Donald E. Ingber; Arja Kaipainen; Patricia A. D'Amore; Mark W. Kieran; Darryl C. Zeldin

Epoxyeicosatrienoic acids (EETs) are small molecules produced by cytochrome P450 epoxygenases. They are lipid mediators that act as autocrine or paracrine factors to regulate inflammation and vascular tone. As a result, drugs that raise EET levels are in clinical trials for the treatment of hypertension and many other diseases. However, despite their pleiotropic effects on cells, little is known about the role of these epoxyeicosanoids in cancer. Here, using genetic and pharmacological manipulation of endogenous EET levels, we demonstrate that EETs are critical for primary tumor growth and metastasis in a variety of mouse models of cancer. Remarkably, we found that EETs stimulated extensive multiorgan metastasis and escape from tumor dormancy in several tumor models. This systemic metastasis was not caused by excessive primary tumor growth but depended on endothelium-derived EETs at the site of metastasis. Administration of synthetic EETs recapitulated these results, while EET antagonists suppressed tumor growth and metastasis, demonstrating in vivo that pharmacological modulation of EETs can affect cancer growth. Furthermore, inhibitors of soluble epoxide hydrolase (sEH), the enzyme that metabolizes EETs, elevated endogenous EET levels and promoted primary tumor growth and metastasis. Thus, our data indicate a central role for EETs in tumorigenesis, offering a mechanistic link between lipid signaling and cancer and emphasizing the critical importance of considering possible effects of EET-modulating drugs on cancer.


The Journal of Pediatrics | 2010

Maternal Preeclampsia Predicts the Development of Bronchopulmonary Dysplasia

Anne Hansen; Carmen Barnes; Judah Folkman; Thomas F. McElrath

OBJECTIVE To test the hypothesis that exposure to preeclampsia is associated with an increased risk of bronchopulmonary dysplasia (BPD). STUDY DESIGN A prospective cohort study of 107 babies born between 23 and 32 weeks gestation, collecting maternal, neonatal, and placental data. RESULTS Of the 107 infants studied, 27 (25%) developed BPD. The bivariate odds ratio (OR) for the relationship between pre-eclampsia and BPD was 2.96 (95% confidence interval [CI] = 1.17 to 7.51; P = .01). When controlling for gestational age, birth weight z-score, chorioamnionitis, and other clinical confounders, the OR of developing BPD was 18.7 (95% CI = 2.44 to 144.76). Including the occurrence of preeclampsia, clinical chorioamnionitis, male sex, and maternal tobacco use in addition to gestational age and birth weight z-score accounted for 54% of the variability of the odds of developing BPD. CONCLUSIONS BPD is increased for infants exposed to preeclampsia. This has possible implications for the prevention of BPD with proangiogenic agents, such as vascular endothelial growth factor.


Journal of Biological Chemistry | 1999

Structural and Kinetic Studies of a Cisplatin-modified DNA Icosamer Binding to HMG1 Domain B

Elizabeth R. Jamieson; Matthew P. Jacobson; Carmen Barnes; Christine S. Chow; Stephen J. Lippard

The high mobility group (HMG) domain is a DNA-binding motif found in the non-histone chromosomal proteins, HMG1 and HMG2, and some transcription factors. Experimental evidence has demonstrated that HMG-domain proteins can play a role in sensitizing cells to the anticancer drug cisplatin. Fluorescence resonance energy transfer (FRET) experiments were performed in the present study to investigate structural changes that accompany complex formation between the HMG domain B of HMG1 and a cisplatin-modified, 20-base pair double-stranded DNA probe containing fluorescein and rhodamine tethered at its two ends. The binding affinity of HMG1 domain B for the cisplatin-modified DNA probe was investigated in fluorescence titration experiments, and a value of 60 ± 30 nm was determined for the dissociation constant. Single photon counting methods were employed to measure the fluorescence lifetime of the fluorescein donor in the presence and absence of HMG1 domain B. These FRET experiments revealed a distance change that was used to estimate a bend angle of 80–95° for the cisplatin-modified DNA upon protein binding. Stopped-flow fluorescence spectroscopic experiments afforded kinetic parameters for HMG1 domain B binding to the cisplatin-modified DNA probe, with k on = 1.1 ± 0.1 × 109 m −1 s−1 andk off = 30 ± 4 s−1.


Lymphatic Research and Biology | 2007

The Placenta Theory and the Origin of Infantile Hemangioma

Carmen Barnes; Emily A. Christison-Lagay; Judah Folkman

The pathogenesis of infantile hemangioma is unknown. In recent years, much of the focus has been placed at identifying the cell type(s) responsible for tumor initiation. New discoveries in infantile hemangioma suggest an involvement of progenitor cells in the pathogenesis of this vascular tumor. Both embryonic and extra-embryonic tissues have been postulated as potential sources for these progenitor cells. This review focuses on the placental theory, which proposes that a fetal placental progenitor is the cell type of origin for infantile hemangioma. Special emphasis will be placed on placental vasculogenesis and the presence and transit of placental progenitor cells during gestation.


Nano Letters | 2015

Dendrimer-Inspired Nanomaterials for the in Vivo Delivery of siRNA to Lung Vasculature

Omar F. Khan; Edmond Zaia; Siddharth Jhunjhunwala; Wen Xue; Wenxin Cai; Dong Soo Yun; Carmen Barnes; James E. Dahlman; Yizhou Dong; Jeisa M. Pelet; Matthew J. Webber; Jonathan K. Tsosie; Tyler Jacks; Robert Langer; Daniel G. Anderson

Targeted RNA delivery to lung endothelial cells has the potential to treat conditions that involve inflammation, such as chronic asthma and obstructive pulmonary disease. To this end, chemically modified dendrimer nanomaterials were synthesized and optimized for targeted small interfering RNA (siRNA) delivery to lung vasculature. Using a combinatorial approach, the free amines on multigenerational poly(amido amine) and poly(propylenimine) dendrimers were substituted with alkyl chains of increasing length. The top performing materials from in vivo screens were found to primarily target Tie2-expressing lung endothelial cells. At high doses, the dendrimer-lipid derivatives did not cause chronic increases in proinflammatory cytokines, and animals did not suffer weight loss due to toxicity. We believe these materials have potential as agents for the pulmonary delivery of RNA therapeutics.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Iron uptake in ferritin is blocked by binding of [Cr(TREN)(H2O)(OH)]2+, a slow dissociating model for [Fe(H2O)6]2+

Carmen Barnes; Elizabeth C. Theil; Kenneth N. Raymond

Ferritin concentrates iron as a hydrous ferric oxide in a protein cavity (8 nm in diameter) by using eight pores along the threefold symmetry axes of the octahedral supramolecular structure. The role of ligand exchange in the entry of Fe(II) hexahydrate into ferritin protein has been studied with [Cr(TREN)(H2O)(OH)]2+ [TREN = N(CH2CH2NH2)3], a model for Fe(H2O)62+ with only two exchangeable ligands. The results show that five different ferritin proteins, varying in pore structure, oxidation sites, and nucleation sites, bind Cr(TREN) at functional protein sites, based on inhibition of iron mineralization and oxidation. Properties of Cr(TREN)–ferritin adducts include an increased isoelectric point, a shift in the Cr(TREN) UV/vis spectrum consistent with exchange of water for protein carboxylate or thiolate ligands, binding affinities of 50–250 μM, and a slow rate of dissociation (k = 4 × 10−6 sec−1). The relationship of Cr(TREN) inhibition of iron oxidation and mineralization by Cr(TREN) to the known structures of the various ferritins tested suggests that Cr(TREN) plugs the ferritin pores, obstructing Fe(II) entry in folded and unfolded pores. Because only two exchangeable waters are sufficient for pore binding of Cr(TREN), the physiological Fe(II) donor must bind to the pore with few exchangeable ligands. These results show the advantage of using stable model complexes to explore properties of transient Fe–protein complexes during Fe mineralization in ferritin.


Pediatric Research | 2010

Correlation of 2-Methoxyestradiol Levels in Cord Blood and Complications of Prematurity

Carmen Barnes; Thomas F. McElrath; Judah Folkman; Anne Hansen

2-methoxyestradiol (2ME2) is a potent antiangiogenic molecule that inhibits the expression of hypoxia-inducible factor (HIF)-1α and, consequently, of VEGF and other HIF-1α target genes. Although 2ME2 is elevated during pregnancy in maternal serum, its presence in fetal fluids and its impact in neonatal health are unknown. In this study, we 1) described normal levels of 2ME2 in maternal blood, cord blood, breast milk, and amniotic fluid, and 2) compared a composite measure of perinatal outcome between infants born with high and low levels of 2ME2. We found that 2ME2 was significantly decreased in all fluids compared with prepartum maternal serum. After stratifying babies by 2ME2 exposure levels, we observed no differences in the vulnerability to impaired lung development or to complications involving aberrant angiogenesis or vascular leak, such as necrotizing enterocolitis (NEC), intraventricular hemorrhage (IVH), posthemorrhagic hydrocephalus (PHH), and retinopathy of prematurity (ROP). In summary, fetal 2ME2 concentrations do not appear to affect neonatal outcome.

Collaboration


Dive into the Carmen Barnes's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arja Kaipainen

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Daniel G. Anderson

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Dipak Panigrahy

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Stephen J. Lippard

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Anne Hansen

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hao Yin

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge