Network


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

Hotspot


Dive into the research topics where James C. Brooks is active.

Publication


Featured researches published by James C. Brooks.


Journal of Immunology | 2007

A New Population of Cells Lacking Expression of CD27 Represents a Notable Component of the B Cell Memory Compartment in Systemic Lupus Erythematosus

Chungwen Wei; Jennifer H. Anolik; Amedeo J. Cappione; Bo Zheng; Aimee E. Pugh-Bernard; James C. Brooks; Eun-Hyung Lee; Eric C. B. Milner; Iñaki Sanz

Human memory B cells comprise isotype-switched and nonswitched cells with both subsets displaying somatic hypermutation. In addition to somatic hypermutation, CD27 expression has also been considered a universal memory B cell marker. We describe a new population of memory B cells containing isotype-switched (IgG and IgA) and IgM-only cells and lacking expression of CD27 and IgD. These cells are present in peripheral blood and tonsils of healthy subjects and display a degree of hypermutation comparable to CD27+ nonswitched memory cells. As conventional memory cells, they proliferate in response to CpG DNA and fail to extrude rhodamine. In contrast to other recently described CD27-negative (CD27neg) memory B cells, they lack expression of FcRH4 and recirculate in the peripheral blood. Although CD27neg memory cells are relatively scarce in healthy subjects, they are substantially increased in systemic lupus erythematosus (SLE) patients in whom they frequently represent a large fraction of all memory B cells. Yet, their frequency is normal in patients with rheumatoid arthritis or chronic hepatitis C. In SLE, an increased frequency of CD27neg memory cells is significantly associated with higher disease activity index, a history of nephritis, and disease-specific autoantibodies (anti-dsDNA, anti-Smith (Sm), anti-ribonucleoprotein (RNP), and 9G4). These findings enhance our understanding of the B cell diversification pathways and provide mechanistic insight into the immunopathogenesis of SLE.


Journal of Biological Chemistry | 2006

Cytoplasmic Targeting Motifs Control Localization of Toll-like Receptor 9

Cynthia A. Leifer; James C. Brooks; Karin Hoelzer; Jody L. Lopez; Margaret N. Kennedy; Alessandra Mazzoni; David M. Segal

Toll-like receptors (TLRs) are essential for host defense. Although several TLRs reside on the cell surface, nucleic acid recognition of TLRs occurs intracellularly. For example, the receptor for CpG containing bacterial and viral DNA, TLR9, is retained in the endoplasmic reticulum. Recent evidence suggests that the localization of TLR9 is critical for appropriate ligand recognition. Here we have defined which structural features of the TLR9 molecule control its intracellular localization. Both the cytoplasmic and ectodomains of TLR9 contain sufficient information, whereas the transmembrane domain plays no role in intracellular localization. We identify a 14-amino acid stretch that directs TLR9 intracellularly and confers intracellular localization to the normally cell surface-expressed TLR4. Truncation or mutation of the cytoplasmic tail of TLR9 reveals a vesicle localization motif that targets early endosomes. We propose a model whereby modification of the cytoplasmic tail of TLR9 results in trafficking to early endosomes where it encounters CpG DNA.


Immunology and Cell Biology | 2009

TLR9 traffics through the Golgi complex to localize to endolysosomes and respond to CpG DNA

Annapoorani Chockalingam; James C. Brooks; Jody Cameron; Lisa K Blum; Cynthia A. Leifer

Toll‐like receptor 9 (TLR9) promiscuously binds self‐ and microbial DNA, but only microbial DNA elicits an inflammatory response. How TLR9 discriminates between self‐ and foreign DNA is unclear, but inappropriate localization of TLR9 permits response to self‐DNA, suggesting that TLR9 localization and trafficking are critical components. The molecular mechanisms controlling the movement of TLR9 may provide new insight into the recognition of DNA in normal and in pathological conditions such as autoimmune systemic lupus erythematosus. We have shown earlier that TLR9 is retained in the endoplasmic reticulum (ER) and it moves to endolysosomes to recognize CpG DNA. Other studies have suggested that TLR9 bypasses the Golgi complex to access endolysosomes. Here, we show that TLR9 translocates from ER to endolysosomes through the Golgi complex and that Golgi export is required for optimal TLR9 signaling. In all, 6–13% of TLR9 constitutively exits the ER, moves through the Golgi complex and resides in lysosomal‐associated membrane protein‐1‐positive vesicles. TLR9 bound to CpG DNA had glycan modifications indicative of Golgi processing confirming that TLR9 travels through the Golgi complex to access CpG DNA in endolysosomes. Together, these data support a model where TLR9 uses traditional secretory pathways and does not bypass the Golgi complex.


European Journal of Immunology | 2011

Negative regulation of signaling by a soluble form of toll‐like receptor 9

Annapoorani Chockalingam; Jody Cameron; James C. Brooks; Cynthia A. Leifer

Nucleic acid structures are highly conserved through evolution and when self nucleic acids are aberrantly detected by toll‐like receptors (TLRs) they contribute to autoimmune disease. For this reason, multiple regulatory mechanisms exist to prevent immune responses to self nucleic acids. TLR9 is a nucleic acid‐sensing TLR that is regulated at multiple levels including association with accessory proteins, intracellular localization and proteolytic processing. In the endolysosomal compartment TLR9 is proteolytically processed to an 80 kDa form (p80) and this processing is a prerequisite for activation. Here, we identified a soluble form of TLR9 (sTLR9) generated by a novel proteolytic event that cleaved TLR9 between amino acids 724–735. Similar to p80, sTLR9 was generated in endosomes. However, generation of sTLR9 was independent of the cysteine protease cathepsin B, active at acidic pH, but partially dependent on cathepsin S, a protease active at neutral pH. Most importantly, sTLR9 inhibited TLR9‐dependent signaling. Altogether, these data support a model where an intrinsic proteolytic processing mechanism negatively regulates TLR9 signaling. A proper balance between the independent proteolytic events probabably contributes to regulation of TLR9‐mediated innate immunity and prevention of autoimmune disease.


Journal of Medicinal Chemistry | 2015

Structure–Activity Relationship in a Purine-Scaffold Compound Series with Selectivity for the Endoplasmic Reticulum Hsp90 Paralog Grp94

Hardik J. Patel; Pallav D. Patel; Stefan O. Ochiana; Pengrong Yan; Weilin Sun; Maulik R. Patel; Smit K. Shah; Elisa Tramentozzi; James C. Brooks; Alexander Bolaender; Liza Shrestha; Ralph Stephani; Paola Finotti; Cynthia A. Leifer; Zihai Li; Daniel T. Gewirth; Tony Taldone; Gabriela Chiosis

Grp94 is involved in the regulation of a restricted number of proteins and represents a potential target in a host of diseases, including cancer, septic shock, autoimmune diseases, chronic inflammatory conditions, diabetes, coronary thrombosis, and stroke. We have recently identified a novel allosteric pocket located in the Grp94 N-terminal binding site that can be used to design ligands with a 2-log selectivity over the other Hsp90 paralogs. Here we perform extensive SAR investigations in this ligand series and rationalize the affinity and paralog selectivity of choice derivatives by molecular modeling. We then use this to design 18c, a derivative with good potency for Grp94 (IC50 = 0.22 μM) and selectivity over other paralogs (>100- and 33-fold for Hsp90α/β and Trap-1, respectively). The paralog selectivity and target-mediated activity of 18c was confirmed in cells through several functional readouts. Compound 18c was also inert when tested against a large panel of kinases. We show that 18c has biological activity in several cellular models of inflammation and cancer and also present here for the first time the in vivo profile of a Grp94 inhibitor.


Biochemical and Biophysical Research Communications | 2012

Heat Shock Protein gp96 regulates Toll-Like Receptor 9 proteolytic processing and conformational stability

James C. Brooks; Weilin Sun; Gabriela Chiosis; Cynthia A. Leifer

Nucleic acid-sensing Toll-like receptors (TLRs) initiate innate immune responses to foreign RNA and DNA, yet can detect and respond to host DNA. To avoid autoimmune pathologies, nucleic acid sensing TLRs are tightly regulated. TLR9 primarily resides in the endoplasmic reticulum, traffics to endosomes, is proteolytically processed and responds to DNA. The heat shock protein gp96 is one of several accessory proteins that regulate intracellular trafficking of TLR9. In the absence of gp96, TLR9 fails to exit the endoplasmic reticulum, and therefore gp96-deficient macrophages fail to respond to CpG DNA. However, absence of gp96 precludes studies on potential chaperoning functions of gp96 for TLR9. Here we demonstrate that pharmacologic interference with gp96 function inhibits TLR9 signaling. TLR9 remains associated with gp96 during intracellular trafficking, and gp96-specific inhibitors increase TLR9 sensitivity to proteolytic degradation. We propose that gp96 is critical for both TLR9 egress from the ER, and for protein conformational stability in the endosomal compartment. These studies highlight the importance of examining gp96-specific inhibitors for modulating TLR9 activation, and the treatment autoimmune diseases.


Journal of Immunology | 2016

Complex Negative Regulation of TLR9 by Multiple Proteolytic Cleavage Events

Siddhartha Sinha; Jody Cameron; James C. Brooks; Cynthia A. Leifer

TLR9 is an innate immune receptor important for recognizing DNA of host and foreign origin. A mechanism proposed to prevent excessive response to host DNA is the requirement for proteolytic cleavage of TLR9 in endosomes to generate a mature form of the receptor (TLR9471–1032). We previously described another cleavage event in the juxtamembrane region of the ectodomain that generated a dominant-negative form of TLR9. Thus, there are at least two independent cleavage events that regulate TLR9. In this study, we investigated whether an N-terminal fragment of TLR9 could be responsible for regulation of the mature or negative-regulatory form. We show that TLR9471–1032, corresponding to the proteolytically cleaved form, does not function on its own. Furthermore, activity is not rescued by coexpression of the N-terminal fragment (TLR91–440), inclusion of the hinge region (TLR9441–1032), or overexpression of UNC93B1, the last of which is critical for trafficking and cleavage of TLR9. TLR91–440 coimmunoprecipitates with full-length TLR9 and TLR9471–1032 but does not rescue the native glycosylation pattern; thus, inappropriate trafficking likely explains why TLR9471–1032 is nonfunctional. Lastly, we show that TLR9471–1032 is also a dominant-negative regulator of TLR9 signaling. Together, these data provide a new perspective on the complexity of TLR9 regulation by proteolytic cleavage and offer potential ways to inhibit activity through this receptor, which may dampen autoimmune inflammation.


Archive | 2012

Regulation of Nucleic Acid Sensing Toll-Like Receptors in Systemic Lupus Erythematosus

Cynthia A. Leifer; James C. Brooks

Autoimmune disease is an aberrant response of the immune system to self. Unlike the adaptive immune system, the innate immune system is not selected during development and was originally thought to inherently discriminate between host and foreign molecular structures (Janeway, 1989). This is true for many innate immune receptor ligands including lipopolysaccharide, which is only synthesized by Gram-negative bacteria. However, some innate immune receptors detect nucleic acids that are shared between microbes and the host. Immune complexes containing nucleic acids are a hallmark of Systemic Lupus Erythematosus (SLE), and the nucleic acid sensing Toll-like receptors (TLRs) respond to the DNA and RNA within these complexes thereby contributing to disease. Defects in regulation of this class of innate immune receptors likely play a key role in precipitation of disease. Here we review nucleic acid sensing TLRs in SLE and recent advances in our understanding of the regulatory mechanisms governing TLR activity. Since breakdown of regulatory mechanisms controlling response of nucleic acid-sensing TLRs likely contributes to development of SLE, targeting specific proteins in these regulatory pathways has the potential to block nucleic acid-driven autoimmune inflammation.


Journal of Immunology | 2010

The Heat Shock Protein gp96 play a multifaceted role in regulating Toll-like receptor 9

Cynthia A. Leifer; James C. Brooks; Jody Cameron; Gabriela Chiosis


Journal of Immunology | 2010

Toll-like receptor 9 is alternatively cleaved to generate a soluble form of the receptor that downregulates response to CpG-DNA

Annapoorani Chockalingam; James C. Brooks; Jody Cameron; Cynthia A. Leifer

Collaboration


Dive into the James C. Brooks's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gabriela Chiosis

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Weilin Sun

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Aimee E. Pugh-Bernard

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Alessandra Mazzoni

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Alexander Bolaender

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge