Network


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

Hotspot


Dive into the research topics where Paula S. Hochman is active.

Publication


Featured researches published by Paula S. Hochman.


Immunity | 1998

Lymph Node Genesis Is Induced by Signaling through the Lymphotoxin β Receptor

Paul D. Rennert; Danelle James; Fabienne Mackay; Jeffrey L. Browning; Paula S. Hochman

Abstract We investigated lymphotoxin (LT) and TNF function in lymph node genesis and cellular organization by manipulating LTβ-R and TNF-R signaling. Lymph nodes developed in LTα −/− mice treated in utero with agonist anti-LTβ-R monoclonal antibody. Thus, LTβ-R signaling mediates lymph node genesis. Surprisingly, mucosal lymph nodes that can develop independently of LTαβ/LTβ-R interaction were generated. Normal mice treated in utero with LTβ-R-Ig and TNF-R55-Ig or anti-TNF lacked all lymph nodes, indicating that TNF signaling contributes to lymph node genesis. Lymph nodes generated in LTα −/− mice had disrupted cellular organization. Therefore, LTβ-R signaling during gestation is not sufficient to establish normal cellular microarchitecture. We conclude that LT and TNF play critical roles in the genesis and cellular organization of lymph nodes.


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 Immunology | 2003

Cutting Edge: Germinal Centers Formed in the Absence of B Cell-Activating Factor Belonging to the TNF Family Exhibit Impaired Maturation and Function

Kalpit A. Vora; Li Chun Wang; Sambasiva Rao; Zhong-Ying Liu; Gerard R. Majeau; Anne H. Cutler; Paula S. Hochman; Martin L. Scott; Susan L. Kalled

Germinal centers (GCs) form in B cell follicles and require specific signals for development and maintenance. B cell-activating factor belonging to the TNF family (BAFF) is a fundamental B cell survival factor and therefore may influence GC reactions and subsequent Ab responses. To test this possibility, the effect of BAFF neutralization in immunized mice was assessed. Using B cell maturation Ag-Fc, we demonstrate that BAFF blockade does not inhibit GC formation or somatic hypermutation. However, GCs in B cell maturation Ag-Fc-treated mice dissipated more rapidly than those of control mice and did not form a mature follicular dendritic cell reticulum. Examination of immunized BAFF-null mice validated the BAFF-independent nature of GC formation. Furthermore, Ab responses, including high-affinity responses, were attenuated. This is the first evidence that BAFF is required for maintenance, but not initiation, of the GC reaction, and it further hints that somatic hypermutation within the GC and selection of Ag-specific high-affinity Ab could be uncoupled.


Journal of Immunology | 2002

Alefacept, an Immunomodulatory Recombinant LFA-3/IgG1 Fusion Protein, Induces CD16 Signaling and CD2/CD16-Dependent Apoptosis of CD2+ Cells

Antonio J. da Silva; Margot Brickelmaier; Gerard R. Majeau; Zhifang Li; Lihe Su; Yen-Ming Hsu; Paula S. Hochman

Alefacept, an immunomodulatory recombinant fusion protein composed of the first extracellular domain of LFA-3 fused to the human IgG1 hinge, CH2, and CH3 domains, has recently been shown in phase II and III clinical trials to safely reduce disease expression in patients with chronic plaque psoriasis. Alefacept modulates the function of and selectively induces apoptosis of CD2+ human memory-effector T cells in vivo. We have sought to gain further understanding of the mechanisms of action that influence the biological activity of alefacept and may contribute to its efficacy and patient responsiveness. Specifically evaluated is the ability of alefacept to activate intracellular signals mediated via CD2 and/or FcγRIII (CD16). Experimentation using isoforms of alefacept engineered to have amino acid substitutions in the IgG1 CH2 domain that impact FcγR binding indicate that alefacept mediates cognate interactions between cells expressing human CD2 and CD16 to activate cells, e.g., increase extracellular signal-regulated kinase phosphorylation, up-regulate cell surface expression of the activation marker CD25, and induce release of granzyme B. In the systems used, this signaling is shown to require binding to CD2 and CD16 and be mediated through CD16, but not CD2. Experimentation using human CD2-transgenic mice and isoforms of alefacept confirmed the requirement for FcγR binding for detection of the pharmacological effects of alefacept in vivo. Thus alefacept acts as an effector molecule, mediating cognate interactions to activate FcγR+ cells (e.g., NK cells) to induce apoptosis of sensitive CD2+ target cells.


Cellular and Molecular Life Sciences | 1998

THE STRUCTURE OF HUMAN INTERFERON-BETA : IMPLICATIONS FOR ACTIVITY

Michael Karpusas; Adrian Whitty; Laura Runkel; Paula S. Hochman

Abstract. Interferons (IFNs) are potent extracellular protein mediators of host defence and homoeostasis. This article reviews the structure of human IFN-β (HuIFN-β), in particular in relation to its activity. The recently determined crystal structure of HuIFN-β provides a framework for understanding of the mechanism of differentiation of type I IFNs by their common receptor. Insights are generated by comparison with the structures of other type I IFNs and from the interpretation of existing mutagenesis data. The details of the observed carbohydrate structure, together with biochemical data, implicate the glycosylation of HuIFN-β, which is uncommon among type I IFNs, as an important factor in the solubility, stability and, consequently, activity of the protein. Finally, these structural implications are discussed in the context of the clinical use of HuIFN-β.


Journal of Biological Chemistry | 1996

Lymphotoxin β Receptor Triggering Induces Activation of the Nuclear Factor κB Transcription Factor in Some Cell Types

Fabienne Mackay; Gerard R. Majeau; Paula S. Hochman; Jeffrey L. Browning

NFκB is a pleiotropic transcription factor capable of activating the expression of a great variety of genes critical for the immunoinflammatory response. Tumor necrosis factor α (TNFα) and lymphotoxin α (LTα, originally TNFβ) are potent nuclear factor κB (NFκB) activators in various cell types. The LTα molecule, in addition to being secreted as a soluble trimer, can also form membrane-anchored heterotrimers with the LTβ chain, another member of the TNF family. The LTα1β2 heterotrimer binds a specific receptor, called the LTβ receptor (LTβ-R), which is also a member of the TNF receptor family. Here, we show that engagement of LTβ-R with a soluble form of LTα1β2 or with a specific anti-LTβ-R agonistic monoclonal antibody CBE11 quickly induces activation of NFκB in HT-29 and WiDr human adenocarcinomas. LTβ-R triggering activates NFκB and induces proliferation in WI-38 human lung fibroblasts. No NFκB activation is observed in human umbilical vein endothelial cells, correlating with the inability of LTβ-R activation to induce expression of NFκB-dependent cell surface adhesion molecules. Thus, like several other members of the TNF receptor family, the LTβ-R can activate NFκB following receptor ligation in some but not all LTβ-R-positive cells.


Journal of Interferon and Cytokine Research | 2002

Comparison of Gene Expression Patterns Induced by Treatment of Human Umbilical Vein Endothelial Cells with IFN-α2b vs. IFN-β1a: Understanding the Functional Relationship Between Distinct Type I Interferons That Act Through a Common Receptor

Antonio J. da Silva; Margot Brickelmaier; Gerard R. Majeau; Alexander V. Lukashin; John Peyman; Adrian Whitty; Paula S. Hochman

We analyzed whether interferon-alpha 2b (IFN-alpha 2b) and IFN-beta 1a engage their common receptor to generate activated receptor complexes possessing distinct signaling properties. Human vascular endothelial cells (HUVEC) are 100-1000-fold more sensitive to IFN-beta 1a than to IFN-alpha 2b in in vitro assays. An nonarray-based expression profiling (GeneCalling) technology was employed to compare the patterns and levels of gene expression induced by these IFN as the broadest means by which signaling events could be measured. To distinguish subtype-related differences from dose-related effects, RNA was prepared from HUVEC treated with 50-5000 pg/ml of each IFN. The results showed that at 50 pg/ml IFN, only a subset of the genes induced by IFN-beta 1a were also induced by IFN-alpha 2b and that individual genes were induced to higher levels by IFN-beta 1a. In contrast, at 5000 pg/ml, both subtypes induced essentially identical sets of genes to similar levels of expression. No genes were seen to be induced uniquely by IFN-alpha 2b but not by IFN-beta 1a. The results show that the two IFN are intrinsically capable of inducing similar gene induction responses and do not provide evidence that they generate activated receptor complexes possessing distinct signaling properties. In contrast, the two IFN generate gene induction patterns that are both qualitatively and quantitatively distinct at subsaturating and potentially physiologically more relevant concentrations.


Transplantation | 1996

Short course single agent therapy with an LFA-3-IgG1 fusion protein prolongs primate cardiac allograft survival.

Richard J. Kaplon; Paula S. Hochman; Robert E. Michler; P. Kwiatkowski; Niloo M. Edwards; Carole L. Berger; He Xu; Werner Meier; Barbara P. Wallner; Patricia L. Chisholm; Charles C. Marboe

The interaction of T cell costimulatory molecules with their ligands is required for optimal T cell activation. Interference with such interactions can induce antigen unresponsiveness and delay xeno- and allograft rejection. We have previously shown that LFA3TIP, a soluble human lymphocyte function-associated antigen (LFA)-3 construct, binds CD2 and inhibits responses of human T cells in vitro. This study reports the first use of a human fusion protein, LFA3TIP, to significantly prolong primate cardiac allograft survival. Based on our observations that LFA3TIP inhibits baboon allogeneic mixed lymphocyte reactions, we gave baboon recipients of heterotopic cardiac allografts injections of LFA3TIP, 3 mg/kg i.v., for 12 consecutive days, starting 2 days before transplantation. This regimen delayed graft rejection from an average of 10.6 +/- 2.3 days for human IgG-treated controls (n = 5) to an average of 18.0 +/- 5.3 days for LFA3TIP-injected animals (n = 7; P < or = 0.01). Grafts from LFA3TIP-treated animals showed markedly diminished coronary endothelialitis as compared with control animals. LFA3TIP reached peak serum levels of approximately 100 micrograms/ml after 7-9 injections and persisted in the 10-micrograms/ml range for 1 to 2 weeks after the final injection. Despite these blood levels, circulating antibodies to LFA3TIP were not detected in the serum. No renal or hepatic toxicity was noted. The possible mechanism by which LFA3TIP acts to inhibit graft rejection is discussed; success in prolonging graft survival when LFA3TIP is used as a single-agent therapy suggests great potential for this novel therapeutic agent.


Journal of Immunology | 2004

Protection against Progressive Leishmaniasis by IFN-β

Jochen Mattner; Alexandra Wandersee-Steinhäuser; Andreas Pahl; Martin Röllinghoff; Gerard R. Majeau; Paula S. Hochman; Christian Bogdan

Type I IFNs (IFN-αβ) exert potent antiviral and immunoregulatory activities during viral infections, but their role in bacterial or protozoan infections is poorly understood. In this study, we demonstrate that the application of low, but not of high doses of IFN-β protects 60 or 100% of BALB/c mice from progressive cutaneous and fatal visceral disease after infection with a high (106) or low (104) number of Leishmania major parasites, respectively. IFN-β treatment of BALB/c mice restored the NK cell cytotoxic activity, increased the lymphocyte proliferation, and augmented the production of IFN-γ and IL-12 in the draining lymph node. Low, but not high doses of IFN-β caused enhanced tyrosine phosphorylation of STAT1 and STAT4, suppressed the levels of suppressor of cytokine signaling-1, and up-regulated the expression of inducible NO synthase in vivo. The IFN-β-induced increase of IFN-γ production was dependent on STAT4. Protection by IFN-β strictly required the presence of inducible NO synthase. In the absence of STAT4 or IL-12, IFN-β led to an amelioration of the cutaneous and visceral disease, but was unable to prevent its progression. These results identify IFN-β as a novel cytokine with a strong, dose-dependent protective effect against progressive cutaneous leishmaniasis that results from IL-12- and STAT4-dependent as well as -independent events.


Cell Adhesion and Communication | 1999

Low affinity binding of an LFA-3/IgG1 fusion protein to CD2+ T cells is independent of cell activation

Gerard R. Majeau; Adrian Whitty; Kalvin Yim; Werner Meier; Paula S. Hochman

Quantitative analysis of binding of the bivalent recombinant soluble fusion protein, LFA-3/IgG1, shows that the fusion protein binds to human CD2+ PBLs primarily through low affinity (KD approximately 140 microM) but also through high avidity (90 nM) interactions. The concentration dependence for LFA-3/IgG1 PBL binding took the form of two overlapping bell-shaped curves separated by a clear and reproducible minimum. This was accounted for in part by minor heterogeneity in the LFA-3/IgG1 preparations, and potentially by the ability of the ligand to bind to both CD2 and Fc receptors (FcR), best evidenced by the distinct binding properties of the fusion protein to NK and T cells. The low affinity LFA-3/ IgG1 binding to T cells is consistent with binding to CD2 only, and is in agreement with the low affinity reported for interactions between soluble forms of LFA-3 and CD2 by surface plasmon resonance technology. Moreover, as the low affinity determinations are similar for CD2 on resting and activated T cells, although the CD2 molecule has been reported to be altered to reveal new epitopes upon T cell activation, the binding data argue against multiple cell activation-dependent affinity states of CD2 for LFA-3 binding. This is distinct from that observed with other adhesion partners, and suggests that the different adhesion pathways utilize distinct mechanisms to mediate cell adhesion.

Collaboration


Dive into the Paula S. Hochman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge