Network


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

Hotspot


Dive into the research topics where Victor Nussenzweig is active.

Publication


Featured researches published by Victor Nussenzweig.


Cell | 1997

TRAP Is Necessary for Gliding Motility and Infectivity of Plasmodium Sporozoites

Ali A. Sultan; Vandana Thathy; Ute Frevert; Kathryn J. H. Robson; Andrea Crisanti; Victor Nussenzweig; Ruth S. Nussenzweig; Robert Ménard

Many protozoans of the phylum Apicomplexa are invasive parasites that exhibit a substrate-dependent gliding motility. Plasmodium (malaria) sporozoites, the stage of the parasite that invades the salivary glands of the mosquito vector and the liver of the vertebrate host, express a surface protein called thrombospondin-related anonymous protein (TRAP) that has homologs in other Apicomplexa. By gene targeting in a rodent Plasmodium, we demonstrate that TRAP is critical for sporozoite infection of the mosquito salivary glands and the rat liver, and is essential for sporozoite gliding motility in vitro. This suggests that in Plasmodium sporozoites, and likely in other Apicomplexa, gliding locomotion and cell invasion have a common molecular basis.


Cell | 1992

The basolateral domain of the hepatocyte plasma membrane bears receptors for the circumsporozoite protein of Plasmodium falciparum sporozoites

Carla Cerami; Ute Frevert; Photini Sinnis; Béla Takács; Pedro Clavijo; Manuel J. Santos; Victor Nussenzweig

Minutes after injection into the circulation, malaria sporozoites enter hepatocytes. The speed and specificity of the invasion process suggest that it is receptor mediated. We show here that recombinant Plasmodium falciparum circumsporozoite protein (CS) binds specifically to regions of the plasma membrane of hepatocytes exposed to circulating blood in the Disse space. No binding has been detected in other organs, or even in other regions of the hepatocyte membrane. The interaction of CS with hepatocytes, as well as sporozoite invasion of HepG2 cells, is inhibited by synthetic peptides representing the evolutionarily conserved region II of CS. We conclude that region II is a sporozoite ligand for hepatocyte receptors localized to the basolateral domain of the plasma membrane. Our findings provide a rational explanation for the target cell specificity of malaria sporozoites.


Advances in Immunology | 1989

Rationale for the development of an engineered sporozoite malaria vaccine.

Victor Nussenzweig; Ruth S. Nussenzweig

Publisher Summary This chapter discusses the rationale for developing sporozoite vaccines. Sporozoites are the infective stages of the parasite found in the salivary glands of female Anopheles mosquitoes. Infection by Plasmodium vivax is also widespread and causes an acute or subacute disease, chronic anemia and splenomegaly but is not usually lethal or drug resistant. At the other end of the spectrum, Plasmodium malariue is well adapted to the human host and produces chronic illness that can persist for many years. The symptoms are similar to those of P. vivax infection, but relapses are less frequent. Attempts to control the spread of the infection by combating the Anopheles mosquito vectors have failed. Vaccine development has become an important research priority. Unfortunately, however, this task is complicated by several problems; some are associated with unique features of this parasite and others, of a more fundamental nature, are associated with the obstacles facing the design of rational, efficient, engineered subunit vaccines that contain multiple B and T cell epitopes. One of the problems in developing a malaria vaccine is that this parasite has a very complex life cycle and each stage bears different protective antigens. In spite of the formidable obstacles, considerable progress has been made in the past few years and a few candidate malaria vaccines have been tried in humans with partial success.


Nature | 2006

The circumsporozoite protein is an immunodominant protective antigen in irradiated sporozoites

Kota Arun Kumar; Gen Ichiro Sano; Silvia Beatriz Boscardin; Ruth S. Nussenzweig; Michel C. Nussenzweig; Fidel Zavala; Victor Nussenzweig

Malaria infection starts when mosquitoes inject sporozoites into the skin. The parasites enter the blood stream and make their way to the liver where they develop into the exo-erythrocytic forms (EEFs). Immunization with irradiated sporozoites (IrSp) leads to robust protection against malaria infection in rodents, monkeys and humans by eliciting antibodies to circumsporozoite protein (CS) that inhibit sporozoite infectivity, and T cells that destroy the EEFs. To study the role of non-CS antigens in protection, we produced CS transgenic mice that were tolerant to CS T-cell epitopes. Here we show that in the absence of T-cell-dependent immune responses to CS, protection induced by immunization with two doses of IrSp was greatly reduced. Thus, although hundreds of other Plasmodium genes are expressed in sporozoites and EEFs, CS is a dominant protective antigen. Nevertheless, sterile immunity could be obtained by immunization of CS transgenics with three doses of IrSp.


Experimental Parasitology | 1987

Stage-specific surface antigens expressed during the morphogenesis of vertebrate forms of Trypanosoma cruzi

Norma W. Andrews; Kyong-su Hong; Edith S. Robbins; Victor Nussenzweig

The origin of Trypanosoma cruzi slender and broad forms found in the circulation of the mammalian host has remained obscure and, unlike what has been proposed for African trypanosomes, no precise form-function relationship has been ascribed to them. We show here that parasites circulating in the blood of infected animals display a high degree of polymorphism. Around 10% of the forms found circulating in mice during the acute phase of infection were amastigotes, and the other 90% included slender and broad trypomastigotes and intermediate forms between amastigotes and trypomastigotes. Slender trypomastigotes, from blood or cell culture, undergo extracellularly morphological rearrangements in which the parasites become gradually broader and transform into amastigotes. By scanning electron microscopy a progressive internalization of the flagellum and reorganization of the cell shape in a helical fashion were observed in parasites undergoing transformation. After 48 hr of extracellular incubation the parasite population consisted exclusively of amastigotes with a short protruding flagellum. The morphological changes were associated with the expression of different surface antigens defined by monoclonal antibodies: the trypomastigote-specific antigens Ssp-1 (a 100-120-150-Mr glycoprotein), Ssp-2 (a 70-Mr glycoprotein), Ssp-3 (undefined), and Ssp-4, an amastigote-specific surface antigen. Ssp-4 was also detected on intracellular amastigotes (in vitro and in vivo). We conclude that trypomastigotes are programmed to develop into amastigotes whether or not they enter cells, and that the differentiation can occur in the blood of the vertebrate host. These findings raise some questions regarding conventional views on the life cycle of T. cruzi.


Cell | 2007

Plasmodium Circumsporozoite Protein Promotes the Development of the Liver Stages of the Parasite

Agam Prasad Singh; Carlos A. Buscaglia; Qian Wang; Agata Levay; Daniel R. Nussenzweig; John R. Walker; Elizabeth A. Winzeler; Hodaka Fujii; Beatriz M. A. Fontoura; Victor Nussenzweig

The liver stages of malaria are clinically silent but have a central role in the Plasmodium life cycle. Liver stages of the parasite containing thousands of merozoites grow inside hepatocytes for several days without triggering an inflammatory response. We show here that Plasmodium uses a PEXEL/VTS motif to introduce the circumsporozoite (CS) protein into the hepatocyte cytoplasm and a nuclear localization signal (NLS) to enter its nucleus. CS outcompetes NFkappaB nuclear import, thus downregulating the expression of many genes controlled by NFkappaB, including those involved in inflammation. CS also influences the expression of over one thousand host genes involved in diverse metabolic processes to create a favorable niche for the parasite growth. The presence of CS in the hepatocyte enhances parasite growth of the liver stages in vitro and in vivo. These findings have far reaching implications for drug and vaccine development against the liver stages of the malaria parasite.


Journal of Biological Chemistry | 2002

Infectivity-associated Changes in the Transcriptional Repertoire of the Malaria Parasite Sporozoite Stage

Kai Matuschewski; Jessica Ross; Stuart M. Brown; Karine Kaiser; Victor Nussenzweig; Stefan H. I. Kappe

Injection of Plasmodium salivary gland sporozoites into the vertebrate host byAnopheles mosquitoes initiates malaria infection. Sporozoites develop within oocysts in the mosquito midgut and then enter and mature in the salivary glands. Although morphologically similar, oocyst sporozoites and salivary gland sporozoites differ strikingly in their infectivity to the mammalian host, ability to elicit protective immune responses, and cell motility. Here, we show that differential gene expression coincides with these dramatic phenotypic differences. Using suppression subtractive cDNA hybridization we identified highly up-regulated mRNAs transcribed from 30 distinct genes in salivary gland sporozoites. Of those genes, 29 are not significantly expressed in the parasites blood stages. The most frequently recovered transcript encodes a protein kinase. Developmental up-regulation of specific mRNAs in the infectious transmission stage of Plasmodiumindicates that their translation products may have unique roles in hepatocyte infection and/or development of liver stages.


Journal of Cell Science | 2003

Myosin A tail domain interacting protein (MTIP) localizes to the inner membrane complex of Plasmodium sporozoites.

Lawrence W. Bergman; Karine Kaiser; Hisashi Fujioka; Isabelle Coppens; Thomas M. Daly; Sarah Fox; Kai Matuschewski; Victor Nussenzweig; Stefan H. I. Kappe

Apicomplexan host cell invasion and gliding motility depend on the parasites actomyosin system located beneath the plasma membrane of invasive stages. Myosin A (MyoA), a class XIV unconventional myosin, is the motor protein. A model has been proposed to explain how the actomyosin motor operates but little is known about the components, topology and connectivity of the motor complex. Using the MyoA neck and tail domain as bait in a yeast two-hybrid screen we identified MTIP, a novel 24 kDa protein that interacts with MyoA. Deletion analysis shows that the 15 amino-acid C-terminal tail domain of MyoA, rather than the neck domain, specifically interacts with MTIP. In Plasmodium sporozoites MTIP localizes to the inner membrane complex (IMC), where it is found clustered with MyoA. The data support a model for apicomplexan motility and invasion in which the MyoA motor protein is associated via its tail domain with MTIP, immobilizing it at the outer IMC membrane. The head domain of the immobilized MyoA moves actin filaments that, directly or via a bridging protein, connect to the cytoplasmic domain of a transmembrane protein of the TRAP family. The actin/TRAP complex is then redistributed by the stationary MyoA from the anterior to the posterior end of the zoite, leading to its forward movement on a substrate or to penetration of a host cell.


Human Vaccines | 2010

From the circumsporozoite protein to the RTS,S/AS candidate vaccine

Joe Cohen; Victor Nussenzweig; Johan Vekemans; Amanda J. Leach

The RTS,S/AS01E malaria vaccine candidate has recently entered phase 3 testing. Reaching this important milestone is the culmination of more than 20 years of research and development by GlaxoSmithKline and partners and collaborators. The vaccine has been developed to protect young children and infants living in sub-Saharan Africa against clinical and severe disease caused by Plasmodium falciparum infection. Over the past 9 years, RTS,S/AS candidate vaccine has been evaluated in multiple Phase 2 studies. The vaccine was shown to have a favourable safety profile and to be well tolerated in all age groups in which it was tested, including the intended target population of infants and young children in Sub-Saharan Africa. Data obtained so far suggest that RTS,S/AS can be co-administered with other vaccines included in the routine Expanded Program of Immunization (EPI). In Phase 2 testing, the vaccine candidate was shown to confer significant protection against P. falciparum infection and clinical disease, including severe malaria. Furthermore, a trend towards an indirect beneficial effect of the vaccine on non-malarial morbidities has been observed in several trials. In this paper, we will describe the genesis of the RTS,S/AS concept, including the rationale for selecting the circumsporozoite protein (CSP) as the target antigen. Early development history of the vaccine will be briefly described. We will present the most salient results from recent Phase 2 studies conducted in the target pediatric population, which have led to the decision to progress RTS,S/AS to Phase 3 testing. If the Phase 3 results confirm the observations made during Phase 2 testing, the RTS,S/AS vaccine, when broadly implemented and judiciously integrated with other malaria-prevention measures, would have a major public-health impact in Sub-Saharan Africa.


The EMBO Journal | 2002

Plasmodium sporozoite invasion into insect and mammalian cells is directed by the same dual binding system

Kai Matuschewski; Alvaro Nunes; Victor Nussenzweig; Robert Ménard

Plasmodium sporozoites, the transmission form of the malaria parasite, successively invade salivary glands in the mosquito vector and the liver in the mammalian host. Sporozoite capacity to invade host cells is mechanistically related to their ability to glide on solid substrates, both activities depending on the transmembrane protein TRAP. Here, we show that loss‐of‐ function mutations in two adhesive modules of the TRAP ectodomain, an integrin‐like A‐domain and a thrombospondin type I repeat, specifically decrease sporozoite invasion of host cells but do not affect sporozoite gliding and adhesion to cells. Irrespective of the target cell, i.e. in mosquitoes, rodents and cultured human or hamster cells, sporozoites bearing mutations in one module are less invasive, while those bearing mutations in both modules are non‐invasive. In Chinese hamster ovary cells, the TRAP modules interact with distinct cell receptors during sporozoite invasion, and thus act as independently active pass keys. As these modules are also present in other members of the TRAP family of proteins in Apicomplexa, they may account for the capacity of these parasites to enter many cell types of phylogenetically distant origins.

Collaboration


Dive into the Victor Nussenzweig's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fidel Zavala

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stephen Tomlinson

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Photini Sinnis

Johns Hopkins University

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