Network


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

Hotspot


Dive into the research topics where Tatiana Novobrantseva is active.

Publication


Featured researches published by Tatiana Novobrantseva.


Nature Biotechnology | 2011

Therapeutic siRNA silencing in inflammatory monocytes in mice

Florian Leuschner; Partha Dutta; Rostic Gorbatov; Tatiana Novobrantseva; Jessica S. Donahoe; Gabriel Courties; Kang Mi Lee; James I. Kim; James F. Markmann; Brett Marinelli; Peter Panizzi; Won Woo Lee; Yoshiko Iwamoto; Hila Epstein-Barash; William Cantley; Jamie Wong; Virna Cortez-Retamozo; Andita Newton; Kevin Love; Peter Libby; Mikael J. Pittet; Filip K. Swirski; Victor Koteliansky; Robert Langer; Ralph Weissleder; Daniel G. Anderson; Matthias Nahrendorf

Excessive and prolonged activity of inflammatory monocytes is a hallmark of many diseases with an inflammatory component. In such conditions, precise targeting of these cells could be therapeutically beneficial while sparing many essential functions of the innate immune system, thus limiting unwanted effects. Inflammatory monocytes—but not the noninflammatory subset—depend on the chemokine receptor CCR2 for localization to injured tissue. Here we present an optimized lipid nanoparticle and a CCR2-silencing short interfering RNA that, when administered systemically in mice, show rapid blood clearance, accumulate in spleen and bone marrow, and localize to monocytes. Efficient degradation of CCR2 mRNA in monocytes prevents their accumulation in sites of inflammation. Specifically, the treatment attenuates their number in atherosclerotic plaques, reduces infarct size after coronary artery occlusion, prolongs normoglycemia in diabetic mice after pancreatic islet transplantation, and results in reduced tumor volumes and lower numbers of tumor-associated macrophages.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Origins of tumor-associated macrophages and neutrophils

Virna Cortez-Retamozo; Martin Etzrodt; Andita Newton; Philipp J. Rauch; Aleksey Chudnovskiy; Cedric R. Berger; Russell J.H. Ryan; Yoshiko Iwamoto; Brett Marinelli; Rostic Gorbatov; Reza Forghani; Tatiana Novobrantseva; Victor Koteliansky; Jose-Luiz Figueiredo; John W. Chen; Daniel G. Anderson; Matthias Nahrendorf; Filip K. Swirski; Ralph Weissleder; Mikael J. Pittet

Tumor-associated macrophages (TAMs) and tumor-associated neutrophils (TANs) can control cancer growth and exist in almost all solid neoplasms. The cells are known to descend from immature monocytic and granulocytic cells, respectively, which are produced in the bone marrow. However, the spleen is also a recently identified reservoir of monocytes, which can play a significant role in the inflammatory response that follows acute injury. Here, we evaluated the role of the splenic reservoir in a genetic mouse model of lung adenocarcinoma driven by activation of oncogenic Kras and inactivation of p53. We found that high numbers of TAM and TAN precursors physically relocated from the spleen to the tumor stroma, and that recruitment of tumor-promoting spleen-derived TAMs required signaling of the chemokine receptor CCR2. Also, removal of the spleen, either before or after tumor initiation, reduced TAM and TAN responses significantly and delayed tumor growth. The mechanism by which the spleen was able to maintain its reservoir capacity throughout tumor progression involved, in part, local accumulation in the splenic red pulp of typically rare extramedullary hematopoietic stem and progenitor cells, notably granulocyte and macrophage progenitors, which produced CD11b+ Ly-6Chi monocytic and CD11b+ Ly-6Ghi granulocytic cells locally. Splenic granulocyte and macrophage progenitors and their descendants were likewise identified in clinical specimens. The present study sheds light on the origins of TAMs and TANs, and positions the spleen as an important extramedullary site, which can continuously supply growing tumors with these cells.


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.


Journal of Clinical Investigation | 2005

Attenuated liver fibrosis in the absence of B cells

Tatiana Novobrantseva; Gerard R. Majeau; Aldo Amatucci; Sophia Kogan; Ian Brenner; Stefano Casola; Mark J. Shlomchik; Victor Koteliansky; Paula S. Hochman

Analysis of mononuclear cells in the adult mouse liver revealed that B cells represent as much as half of the intrahepatic lymphocyte population. Intrahepatic B cells (IHB cells) are phenotypically similar to splenic B2 cells but express lower levels of CD23 and CD21 and higher levels of CD5. IHB cells proliferate as well as splenic B cells in response to anti-IgM and LPS stimulation in vitro. VDJ gene rearrangements in IHB cells contain insertions of N,P region nucleotides characteristic of B cells maturing in the adult bone marrow rather than in the fetal liver. To evaluate whether B cells can have an impact on liver pathology, we compared CCl4-induced fibrosis development in B cell-deficient and wild-type mice. CCl4 caused similar acute liver injury in mutant and wild-type mice. However, following 6 weeks of CCl4 treatment, histochemical analyses showed markedly reduced collagen deposition in B cell-deficient as compared with wild-type mice. By analyzing mice that have normal numbers of B cells but lack either T cells or immunoglobulin in the serum, we established that B cells have an impact on fibrosis in an antibody- and T cell-independent manner.


Journal of Biological Chemistry | 2012

Kuppfer Cells Trigger Nonalcoholic Steatohepatitis Development in Diet-induced Mouse Model through Tumor Necrosis Factor-α Production

Annie‐Carole Tosello‐Trampont; Susan G. Landes; Virginia Nguyen; Tatiana Novobrantseva; Young S. Hahn

Background: The mechanisms triggering nonalcoholic steatohepatitis (NASH) remain poorly defined. Results: Kupffer cells are the first responding cells to hepatocyte injuries, leading to TNFα production, chemokine induction, and monocyte recruitment. The silencing of TNFα in myeloid cells reduces NASH progression. Conclusion: Increase of TNFα-producing Kupffer cells is crucial for triggering NASH via monocyte recruitment. Significance: Myeloid cells-targeted silencing of TNFα might be a tenable therapeutic approach. Nonalcoholic steatohepatitis (NASH), characterized by lipid deposits within hepatocytes (steatosis), is associated with hepatic injury and inflammation and leads to the development of fibrosis, cirrhosis, and hepatocarcinoma. However, the pathogenic mechanism of NASH is not well understood. To determine the role of distinct innate myeloid subsets in the development of NASH, we examined the contribution of liver resident macrophages (i.e. Kupffer cells) and blood-derived monocytes in triggering liver inflammation and hepatic damage. Employing a murine model of NASH, we discovered a previously unappreciated role for TNFα and Kupffer cells in the initiation and progression of NASH. Sequential depletion of Kupffer cells reduced the incidence of liver injury, steatosis, and proinflammatory monocyte infiltration. Furthermore, our data show a differential contribution of Kupffer cells and blood monocytes during the development of NASH; Kupffer cells increased their production of TNFα, followed by infiltration of CD11bintLy6Chi monocytes, 2 and 10 days, respectively, after starting the methionine/choline-deficient (MCD) diet. Importantly, targeted knockdown of TNFα expression in myeloid cells decreased the incidence of NASH development by decreasing steatosis, liver damage, monocyte infiltration, and the production of inflammatory chemokines. Our findings suggest that the increase of TNFα-producing Kupffer cells in the liver is crucial for the early phase of NASH development by promoting blood monocyte infiltration through the production of IP-10 and MCP-1.


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.


Nature Communications | 2014

Degradable lipid nanoparticles with predictable in vivo siRNA delivery activity

Kathryn A. Whitehead; J. Robert Dorkin; Arturo Vegas; Philip H. Chang; Omid Veiseh; Jonathan C. F. Matthews; Owen S. Fenton; Yunlong Zhang; Karsten Olejnik; Volkan Yesilyurt; Delai Chen; Scott Barros; Boris Klebanov; Tatiana Novobrantseva; Robert Langer; Daniel G. Anderson

One of the most significant challenges in the development of clinically-viable delivery systems for RNA interference therapeutics is to understand how molecular structures influence delivery efficacy. To this end, we synthesized 1400 degradable lipidoids and evaluated their transfection ability and structure function activity. Here we show that lipidoid nanoparticles mediate potent gene knockdown in hepatocytes and immune cell populations upon IV administration to mice (siRNA EC50 values as low as 0.01 mg/kg). Surprisingly, we identify four necessary and sufficient structural and pKa criteria that robustly predict the ability of nanoparticles to mediate greater than 95% protein silencing in vivo. Because these efficacy criteria can be dictated through chemical design, this discovery could eliminate our dependence on time-consuming and expensive cell culture assays and animal testing. Herein, we identify promising degradable lipidoids and describe new design criteria that reliably predict in vivo siRNA delivery efficacy without any prior biological testing.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Combinatorial synthesis of chemically diverse core-shell nanoparticles for intracellular delivery

Daniel J. Siegwart; Kathryn A. Whitehead; Lutz Nuhn; Gaurav Sahay; Hao Cheng; Shan Jiang; Minglin Ma; Abigail K. R. Lytton-Jean; Arturo Vegas; Patrick Fenton; Christopher G. Levins; Kevin Love; Haeshin Lee; Christina Cortez; Sean P. Collins; Ying Fei Li; Janice Jang; William Querbes; Christopher Zurenko; Tatiana Novobrantseva; Robert Langer; Daniel G. Anderson

Analogous to an assembly line, we employed a modular design for the high-throughput study of 1,536 structurally distinct nanoparticles with cationic cores and variable shells. This enabled elucidation of complexation, internalization, and delivery trends that could only be learned through evaluation of a large library. Using robotic automation, epoxide-functionalized block polymers were combinatorially cross-linked with a diverse library of amines, followed by measurement of molecular weight, diameter, RNA complexation, cellular internalization, and in vitro siRNA and pDNA delivery. Analysis revealed structure-function relationships and beneficial design guidelines, including a higher reactive block weight fraction, stoichiometric equivalence between epoxides and amines, and thin hydrophilic shells. Cross-linkers optimally possessed tertiary dimethylamine or piperazine groups and potential buffering capacity. Covalent cholesterol attachment allowed for transfection in vivo to liver hepatocytes in mice. The ability to tune the chemical nature of the core and shell may afford utility of these materials in additional applications.


Molecular Therapy | 2011

Influence of cationic lipid composition on gene silencing properties of lipid nanoparticle formulations of siRNA in antigen-presenting cells.

Genc Basha; Tatiana Novobrantseva; Nicole Rosin; Yuen Yi C. Tam; Ismail Hafez; Matthew Wong; Tsukasa Sugo; Vera M. Ruda; June Qin; Boris Klebanov; Marco A. Ciufolini; Akin Akinc; Ying K. Tam; Michael J. Hope; Pieter R. Cullis

Lipid nanoparticles (LNPs) are currently the most effective in vivo delivery systems for silencing target genes in hepatocytes employing small interfering RNA. Antigen-presenting cells (APCs) are also potential targets for LNP siRNA. We examined the uptake, intracellular trafficking, and gene silencing potency in primary bone marrow macrophages (bmMΦ) and dendritic cells of siRNA formulated in LNPs containing four different ionizable cationic lipids namely DLinDAP, DLinDMA, DLinK-DMA, and DLinKC2-DMA. LNPs containing DLinKC2-DMA were the most potent formulations as determined by their ability to inhibit the production of GAPDH target protein. Also, LNPs containing DLinKC2-DMA were the most potent intracellular delivery agents as indicated by confocal studies of endosomal versus cytoplamic siRNA location using fluorescently labeled siRNA. DLinK-DMA and DLinKC2-DMA formulations exhibited improved gene silencing potencies relative to DLinDMA but were less toxic. In vivo results showed that LNP siRNA systems containing DLinKC2-DMA are effective agents for silencing GAPDH in APCs in the spleen and peritoneal cavity following systemic administration. Gene silencing in APCs was RNAi mediated and the use of larger LNPs resulted in substantially reduced hepatocyte silencing, while similar efficacy was maintained in APCs. These results are discussed with regard to the potential of LNP siRNA formulations to treat immunologically mediated diseases.


Journal of the American College of Cardiology | 2014

In vivo silencing of the transcription factor IRF5 reprograms the macrophage phenotype and improves infarct healing

Gabriel Courties; Timo Heidt; Matthew Sebas; Yoshiko Iwamoto; Derrick Jeon; Jessica Truelove; Benoit Tricot; Greg Wojtkiewicz; Partha Dutta; Hendrik B. Sager; Anna Borodovsky; Tatiana Novobrantseva; Boris Klebanov; Kevin Fitzgerald; Daniel G. Anderson; Peter Libby; Filip K. Swirski; Ralph Weissleder; Matthias Nahrendorf

OBJECTIVES The aim of this study was to test whether silencing of the transcription factor interferon regulatory factor 5 (IRF5) in cardiac macrophages improves infarct healing and attenuates post-myocardial infarction (MI) remodeling. BACKGROUND In healing wounds, the M1 toward M2 macrophage phenotype transition supports resolution of inflammation and tissue repair. Persistence of inflammatory M1 macrophages may derail healing and compromise organ functions. The transcription factor IRF5 up-regulates genes associated with M1 macrophages. METHODS Here we used nanoparticle-delivered small interfering ribonucleic acid (siRNA) to silence IRF5 in macrophages residing in MIs and in surgically-induced skin wounds in mice. RESULTS Infarct macrophages expressed high levels of IRF5 during the early inflammatory wound-healing stages (day 4 after coronary ligation), whereas expression of the transcription factor decreased during the resolution of inflammation (day 8). Following in vitro screening, we identified an siRNA sequence that, when delivered by nanoparticles to wound macrophages, efficiently suppressed expression of IRF5 in vivo. Reduction of IRF5 expression, a factor that regulates macrophage polarization, reduced expression of inflammatory M1 macrophage markers, supported resolution of inflammation, accelerated cutaneous and infarct healing, and attenuated development of post-MI heart failure after coronary ligation as measured by protease targeted fluorescence molecular tomography-computed tomography imaging and cardiac magnetic resonance imaging (p < 0.05). CONCLUSIONS This work identified a new therapeutic avenue to augment resolution of inflammation in healing infarcts by macrophage phenotype manipulation. This therapeutic concept may be used to attenuate post-MI remodeling and heart failure.

Collaboration


Dive into the Tatiana Novobrantseva's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fougerolles Antonin De

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Akin Akinc

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Daniel G. Anderson

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Pamela Tan

Alnylam Pharmaceuticals

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