Network


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

Hotspot


Dive into the research topics where Brian D. Brown is active.

Publication


Featured researches published by Brian D. Brown.


Nature Immunology | 2012

Deciphering the transcriptional network of the dendritic cell lineage

Jennifer Miller; Brian D. Brown; Tal Shay; Emmanuel L. Gautier; Vladimir Jojic; Ariella Cohain; Gaurav Pandey; Marylene Leboeuf; Kutlu G. Elpek; Julie Helft; Daigo Hashimoto; Andrew Chow; Jeremy Price; Melanie Greter; Milena Bogunovic; Angelique Bellemare-Pelletier; Paul S. Frenette; Gwendalyn J. Randolph; Shannon J. Turley; Miriam Merad

Although much progress has been made in the understanding of the ontogeny and function of dendritic cells (DCs), the transcriptional regulation of the lineage commitment and functional specialization of DCs in vivo remains poorly understood. We made a comprehensive comparative analysis of CD8+, CD103+, CD11b+ and plasmacytoid DC subsets, as well as macrophage DC precursors and common DC precursors, across the entire immune system. Here we characterized candidate transcriptional activators involved in the commitment of myeloid progenitor cells to the DC lineage and predicted regulators of DC functional diversity in tissues. We identified a molecular signature that distinguished tissue DCs from macrophages. We also identified a transcriptional program expressed specifically during the steady-state migration of tissue DCs to the draining lymph nodes that may control tolerance to self tissue antigens.


Nature Medicine | 2006

Endogenous microRNA regulation suppresses transgene expression in hematopoietic lineages and enables stable gene transfer.

Brian D. Brown; Mary Anna Venneri; Anna Zingale; Lucia Sergi Sergi; Luigi Naldini

MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene expression by repressing translation of target cellular transcripts. Increasing evidence indicates that miRNAs have distinct expression profiles and play crucial roles in numerous cellular processes, although the extent of miRNA regulation is not well known. By challenging mice with lentiviral vectors encoding target sequences of endogenous miRNAs, we show the efficiency of miRNAs in sharply segregating gene expression among different tissues. Transgene expression from vectors incorporating target sequences for mir-142-3p was effectively suppressed in intravascular and extravascular hematopoietic lineages, whereas expression was maintained in nonhematopoietic cells. This expression profile, which could not be attained until now, enabled stable gene transfer in immunocompetent mice, thus overcoming a major hurdle to successful gene therapy. Our results provide novel in situ evidence of miRNA regulation and demonstrate a new paradigm in vector design with applications for genetic engineering and therapeutic gene transfer.


Nature Reviews Genetics | 2009

Exploiting and antagonizing microRNA regulation for therapeutic and experimental applications

Brian D. Brown; Luigi Naldini

New technologies are emerging that utilize artificial microRNA (miRNA) target sites to exploit or inhibit endogenous miRNA regulation. This approach has been used to improve cell-specific targeting for gene and stem cell therapy studies and for animal transgenics, and also to reduce the toxicity of oncolytic viruses and to attenuate viral vaccines. Artificial targets have also been used to sponge or decoy miRNAs as a way to study their functions. This article considers the benefits of this approach and design considerations for future studies.


Nature Methods | 2009

Stable knockdown of microRNA in vivo by lentiviral vectors

Bernhard Gentner; Giulia Schira; Alice Giustacchini; Mario Amendola; Brian D. Brown; Maurilio Ponzoni; Luigi Naldini

Studying microRNA function in vivo requires genetic strategies to generate loss-of-function phenotypes. We used lentiviral vectors to stably and specifically knock down microRNA by overexpressing microRNA target sequences from polymerase II promoters. These vectors effectively inhibited regulation of reporter constructs and natural microRNA targets. We used bone marrow reconstitution with hematopoietic stem cells stably overexpressing miR-223 target sequence to phenocopy the genetic miR-223 knockout mouse, indicating robust interference of microRNA function in vivo.


Nature Reviews Immunology | 2011

Studying the mononuclear phagocyte system in the molecular age

Andrew Chow; Brian D. Brown; Miriam Merad

The mononuclear phagocyte system (MPS) comprises monocytes, macrophages and dendritic cells. Tissue phagocytes share several cell surface markers, phagocytic capability and myeloid classification; however, the factors that regulate the differentiation, homeostasis and function of macrophages and dendritic cells remain largely unknown. The purpose of this manuscript is to review the tools that are currently available and those that are under development to study the origin and function of mononuclear phagocytes.


Nucleic Acids Research | 2011

Identification and remediation of biases in the activity of RNA ligases in small-RNA deep sequencing

Anitha Jayaprakash; Omar J. Jabado; Brian D. Brown; Ravi Sachidanandam

Deep sequencing of small RNAs (sRNA-seq) is now the gold standard for small RNA profiling and discovery. Biases in sRNA-seq have been reported, but their etiology remains unidentified. Through a comprehensive series of sRNA-seq experiments, we establish that the predominant cause of the bias is the RNA ligases. We further demonstrate that RNA ligases have strong sequence-specific biases which distort the small RNA profiles considerably. We have devised a pooled adapter strategy to overcome this bias, and validated the method through data derived from microarray and qPCR. In light of our findings, published small RNA profiles, as well as barcoding strategies using adapter-end modifications, may need to be revisited. Importantly, by providing a wide spectrum of substrate for the ligase, the pooled-adapter strategy developed here provides a means to overcome issues of bias, and generate more accurate small RNA profiles.


Journal of Clinical Investigation | 2012

Cross-presenting CD103+ dendritic cells are protected from influenza virus infection.

Julie Helft; Balaji Manicassamy; Pierre Guermonprez; Daigo Hashimoto; Aymeric Silvin; Judith Agudo; Brian D. Brown; Mirco Schmolke; Jennifer Miller; Marylene Leboeuf; Kenneth M. Murphy; Adolfo García-Sastre; Miriam Merad

CD8+ cytotoxic T cells are critical for viral clearance from the lungs upon influenza virus infection. The contribution of antigen cross-presentation by DCs to the induction of anti-viral cytotoxic T cells remains controversial. Here, we used a recombinant influenza virus expressing a nonstructural 1-GFP (NS1-GFP) reporter gene to visualize the route of antigen presentation by lung DCs upon viral infection in mice. We found that lung CD103+ DCs were the only subset of cells that carried intact GFP protein to the draining LNs. Strikingly, lung migratory CD103+ DCs were not productively infected by influenza virus and thus were able to induce virus-specific CD8+ T cells through the cross-presentation of antigens from virally infected cells. We also observed that CD103+ DC resistance to infection correlates with an increased anti-viral state in these cells that is dependent on the expression of type I IFN receptor. These results show that efficient cross-priming by migratory lung DCs is coupled to the acquisition of an anti-viral status, which is dependent on the type I IFN signaling pathway.


Nature Communications | 2014

microRNA-181a has a critical role in ovarian cancer progression through the regulation of the epithelial-mesenchymal transition.

Aditya Parikh; Christine Elaine Lee; Peronne Joseph; Sergio Marchini; Alessia Baccarini; V. Kolev; Chiara Romualdi; Robert Fruscio; Hardik Shah; Feng Wang; Gavriel Mullokandov; David A. Fishman; Maurizio D’Incalci; Jamal Rahaman; Tamara Kalir; Raymond W. Redline; Brian D. Brown; Goutham Narla; Analisa DiFeo

Ovarian cancer is a leading cause of cancer deaths among women. Effective targets to treat advanced epithelial ovarian cancer (EOC) and biomarkers to predict treatment response are still lacking because of the complexity of pathways involved in ovarian cancer progression. Here we show that miR-181a promotes TGF-β-mediated epithelial-to-mesenchymal transition via repression of its functional target, Smad7. miR-181a and phosphorylated Smad2 are enriched in recurrent compared with matched-primary ovarian tumours and their expression is associated with shorter time to recurrence and poor outcome in patients with EOC. Furthermore, ectopic expression of miR-181a results in increased cellular survival, migration, invasion, drug resistance and in vivo tumour burden and dissemination. In contrast, miR-181a inhibition via decoy vector suppression and Smad7 re-expression results in significant reversion of these phenotypes. Combined, our findings highlight an unappreciated role for miR-181a, Smad7, and the TGF-β signalling pathway in high-grade serous ovarian cancer.


Immunity | 2016

Expansion and Activation of CD103+ Dendritic Cell Progenitors at the Tumor Site Enhances Tumor Responses to Therapeutic PD-L1 and BRAF Inhibition

Hélène Salmon; Juliana Idoyaga; Adeeb Rahman; Marylene Leboeuf; Romain Remark; Stefan Jordan; Maria Casanova-Acebes; Makhzuna Khudoynazarova; Judith Agudo; Navpreet Tung; Svetoslav Chakarov; Christina Rivera; Brandon Hogstad; Marcus Bosenberg; Daigo Hashimoto; Sacha Gnjatic; Nina Bhardwaj; Anna Karolina Palucka; Brian D. Brown; Joshua Brody; Florent Ginhoux; Miriam Merad

Large numbers of melanoma lesions develop resistance to targeted inhibition of mutant BRAF or fail to respond to checkpoint blockade. We explored whether modulation of intratumoral antigen-presenting cells (APCs) could increase responses to these therapies. Using mouse melanoma models, we found that CD103(+) dendritic cells (DCs) were the only APCs transporting intact antigens to the lymph nodes and priming tumor-specific CD8(+) T cells. CD103(+) DCs were required to promote anti-tumoral effects upon blockade of the checkpoint ligand PD-L1; however, PD-L1 inhibition only led to partial responses. Systemic administration of the growth factor FLT3L followed by intratumoral poly I:C injections expanded and activated CD103(+) DC progenitors in the tumor, enhancing responses to BRAF and PD-L1 blockade and protecting mice from tumor rechallenge. Thus, the paucity of activated CD103(+) DCs in tumors limits checkpoint-blockade efficacy and combined FLT3L and poly I:C therapy can enhance tumor responses to checkpoint and BRAF blockade.


Cell | 2014

TLR Signals Induce Phagosomal MHC-I Delivery from the Endosomal Recycling Compartment to Allow Cross-Presentation

Priyanka Nair-Gupta; Alessia Baccarini; Navpreet Tung; Fabian Seyffer; Oliver Florey; Yunjie Huang; Meenakshi Banerjee; Michael Overholtzer; Paul A. Roche; Robert Tampé; Brian D. Brown; Derk Amsen; Sidney W. Whiteheart; J. Magarian Blander

Adaptation of the endoplasmic reticulum (ER) pathway for MHC class I (MHC-I) presentation in dendritic cells enables cross-presentation of peptides derived from phagocytosed microbes, infected cells, or tumor cells to CD8 T cells. How these peptides intersect with MHC-I molecules remains poorly understood. Here, we show that MHC-I selectively accumulate within phagosomes carrying microbial components, which engage Toll-like receptor (TLR) signaling. Although cross-presentation requires Sec22b-mediated phagosomal recruitment of the peptide loading complex from the ER-Golgi intermediate compartment (ERGIC), this step is independent of TLR signaling and does not deliver MHC-I. Instead, MHC-I are recruited from an endosomal recycling compartment (ERC), which is marked by Rab11a, VAMP3/cellubrevin, and VAMP8/endobrevin and holds large reserves of MHC-I. While Rab11a activity stocks ERC stores with MHC-I, MyD88-dependent TLR signals drive IκB-kinase (IKK)2-mediated phosphorylation of phagosome-associated SNAP23. Phospho-SNAP23 stabilizes SNARE complexes orchestrating ERC-phagosome fusion, enrichment of phagosomes with ERC-derived MHC-I, and subsequent cross-presentation during infection.

Collaboration


Dive into the Brian D. Brown's collaboration.

Top Co-Authors

Avatar

Alessia Baccarini

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Judith Agudo

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Luigi Naldini

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Miriam Merad

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lucia Sergi Sergi

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Gavriel Mullokandov

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Joshua Brody

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Ravi Sachidanandam

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge