Network


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

Hotspot


Dive into the research topics where Charlotte V. Hobbs is active.

Publication


Featured researches published by Charlotte V. Hobbs.


Cellular Microbiology | 2008

The Plasmodium TRAP/MIC2 family member, TRAP-Like Protein (TLP), is involved in tissue traversal by sporozoites

Cristina K. Moreira; Thomas J. Templeton; Catherine Lavazec; Rhian E. Hayward; Charlotte V. Hobbs; Hans Kroeze; Chris J. Janse; Andrew P. Waters; Photini Sinnis; Alida Coppi

In the apicomplexan protozoans motility and cell invasion are mediated by the TRAP/MIC2 family of transmembrane proteins, members of which link extracellular adhesion to the intracellular actomyosin motor complex. Here we characterize a new member of the TRAP/MIC2 family, named TRAP‐Like Protein (TLP), that is highly conserved within the Plasmodium genus. Similar to the Plasmodium sporozoite protein, TRAP, and the ookinete protein, CTRP, TLP possesses an extracellular domain architecture that is comprised of von Willebrand factor A (vWA) and thrombospondin type 1 (TSP1) domains, plus a short cytoplasmic domain. Comparison of the vWA domain of TLP genes from multiple Plasmodium falciparum isolates showed relative low sequence diversity, suggesting that the protein is not under selective pressures of the host immune system. Analysis of transcript levels by quantitative reverse transcription polymerase chain reaction (RT‐PCR) showed that TLP is predominantly expressed in salivary gland sporozoites of P. falciparum and P. berghei. Targeted disruption of P. berghei TLP resulted in a decreased capacity for cell traversal by sporozoites, and reduced infectivity of sporozoites in vivo, whereas in vitro sporozoite motility and hepatocyte invasion were unaffected. These results indicate a role of TLP in cell traversal by sporozoites.


The Journal of Infectious Diseases | 2009

HIV Protease Inhibitors Inhibit the Development of Preerythrocytic-Stage Plasmodium Parasites

Charlotte V. Hobbs; Tatiana Voza; Alida Coppi; Brian Kirmse; Kennan Marsh; William Borkowsky; Photini Sinnis

Recent studies have demonstrated that human immunodeficiency virus (HIV) protease inhibitors (PIs) exert inhibitory effects on erythrocytic stages of the human-malaria parasite Plasmodium falciparum in vitro and on erythrocytic stages of the rodent-malaria parasite Plasmodium chabaudi in vivo. Although it remains unclear how HIV PIs inhibit the parasite, the effect seen on parasite development in the erythrocytic stages is potent. The effect on preerythrocytic stages has not yet been investigated. Using the rodent parasite Plasmodium berghei, we screened a panel of HIV PIs in vitro for effects on the preerythrocytic stages. Our data indicated that the HIV PIs lopinavir and saquinavir affect preerythrocytic-stage parasite development in vitro. We then evaluated the effect of HIV PIs on preerythrocytic stages in vivo using the rodent parasite Plasmodium yoelii. We found that lopinavir/ritonavir had a dose-dependent effect on liver-stage parasite development. Given that sub-Saharan Africa is where the HIV/AIDS pandemic intersects with malaria, these results merit analysis in clinical settings.


The Journal of Infectious Diseases | 2013

HIV Treatments Have Malaria Gametocyte Killing and Transmission Blocking Activity

Charlotte V. Hobbs; Takeshi Tanaka; Olga Muratova; Jillian Van Vliet; William Borkowsky; Kim C. Williamson; Patrick E. Duffy

BACKGROUND  Millions of individuals being treated for human immunodeficiency virus (HIV) live in malaria-endemic areas, but the effects of these treatments on malaria transmission are unknown. While drugs like HIV protease inhibitors (PIs) and trimethoprim-sulfamethoxazole (TMP-SMX) have known activity against parasites during liver or asexual blood stages, their effects on transmission stages require further study. METHODS  The HIV PIs lopinavir and saquinavir, the nonnucleoside reverse-transcriptase inhibitor nevirapine, and the antibiotic TMP-SMX were assessed for activity against Plasmodium falciparum transmission stages. The alamarBlue assay was used to determine the effects of drugs on gametocyte viability, and exflagellation was assessed to determine the effects of drugs on gametocyte maturation. The effects of drug on transmission were assessed by calculating the mosquito oocyst count as a marker for infectivity, using standard membrane feeding assays. RESULTS  Lopinavir and saquinavir have gametocytocidal and transmission blocking activities at or approaching clinically relevant treatment levels, while nevirapine does not. TMP-SMX is not gametocytocidal, but at prophylactic levels it blocks transmission. CONCLUSIONS  Specific HIV treatments have gametocyte killing and transmission-blocking effects. Clinical studies are warranted to evaluate these findings and their potential impact on eradication efforts.


Journal of Acquired Immune Deficiency Syndromes | 2012

Unresolved Antiretroviral Treatment Management Issues in HIV-Infected Children

Shirin Heidari; Lynne M. Mofenson; Charlotte V. Hobbs; Mark F. Cotton; Richard Marlink; Elly Katabira

Abstract:Antiretroviral therapy in children has expanded dramatically in low-income and middle-income countries. The World Health Organization revised its pediatric HIV guidelines to recommend initiation of antiretroviral therapy in all HIV-infected children younger than 2 years, regardless of CD4 count or clinical stage. The number of children starting life-long antiretroviral therapy should therefore expand dramatically over time. The early initiation of antiretroviral therapy has indisputable benefits for children, but there is a paucity of definitive information on the potential adverse effects. In this review, a comprehensive literature search was conducted to provide an overview of our knowledge about the complications of treating pediatric HIV.Antiretroviral therapy in children, as in adults, is associated with enhanced survival, reduction in opportunistic infections, improved growth and neurocognitive function, and better quality of life. Despite antiretroviral therapy, HIV-infected children may continue to lag behind their uninfected peers in growth and development. In addition, epidemic concurrent conditions, such as tuberculosis, malaria, and malnutrition, can combine with HIV to yield more rapid disease progression and poor treatment outcomes.Additional studies are required to evaluate the long-term effects of antiretroviral therapy in HIV-infected infants, children, and adolescents, particularly in resource-limited countries where concomitant infections and conditions may enhance the risk of adverse effects. There is an urgent need to evaluate drug–drug interactions in children to determine optimal treatment regimens for both HIV and coinfections.


F1000 Medicine Reports | 2011

Drugs for malaria: something old, something new, something borrowed.

Charlotte V. Hobbs; Patrick E. Duffy

Malaria was estimated to cause 800,000 deaths and 225 million cases worldwide in 2010. Worryingly, the first-line treatment currently relies on a single drug class called artemisinins, and there are signs that the parasite is becoming resistant to these drugs. The good news is that new technology has given us new approaches to drug discovery. New drugs generated this way are probably 10-15 years away from the clinic. Other antimalarials that may offer hope include those rehabilitated after not being used for some time, those that act as inhibitors of resistance mechanisms, those that limit infection while allowing protective immunity to develop, and those which are drugs borrowed from other disease treatments. All of these offer new hope of turning the tables on malaria. In parallel with the effort to develop vaccines that interrupt malaria transmission, drugs that target the parasite during transmission to the mosquito or during its pre-erythrocytic development in the liver, may allow us to terminate the parasite’s spread.


Pediatric Infectious Disease Journal | 2013

Abnormal newborn screens and acylcarnitines in HIV-exposed and ARV-exposed infants

Brian Kirmse; Charlotte V. Hobbs; Inga Peter; Bryan LaPlante; Michele Caggana; Karen Kloke; Kimiyo Raymond; Marshall Summar; William Borkowsky

Background: Antiretroviral drugs (ARV), specifically nucleoside analogs, are toxic to mitochondrial oxidative phosphorylation. Other metabolic pathways, such as fatty acid oxidation, organic acid metabolism and amino acid metabolism, are dependent on normal oxidative phosphorylation but remain unexamined as potential points of ARV toxicity. Methods: We analyzed newborn screening data from New York and compared proportions of abnormal newborn metabolic screens in HIV antibody screen–positive and HIV screen–negative neonates. Subsequently, we compared acylcarnitine levels in ARV-exposed (n = 16) and ARV-unexposed (n = 14) HIV-exposed infants to assess for dysfunctional fatty and organic acid metabolism. Results: The rate of abnormal newborn metabolic screens in HIV screen–positive infants was higher than that in the general population (2.2% versus 1.2%; P = 0.00025), most of which were for disorders of mitochondria-related metabolism. Abnormal acylcarnitine levels occurred more frequently in ARV-exposed compared with ARV-unexposed infants (43% versus 0%; P = 0.02). Conclusions: A higher proportion of positive metabolic screens in HIV screen–positive neonates suggests that HIV or ARV exposure is associated with dysfunctional intermediary metabolism in newborns. Abnormal acylcarnitine levels were more frequent in ARV-exposed infants, suggesting that ARV may perturb normal fatty acid oxidation in some infants. Studies designed to validate and determine the clinical significance of these findings are warranted.


The Journal of Infectious Diseases | 2012

HIV Nonnucleoside Reverse Transcriptase Inhibitors and Trimethoprim-Sulfamethoxazole Inhibit Plasmodium Liver Stages

Charlotte V. Hobbs; Tatiana Voza; Patricia De La Vega; Jillian Vanvliet; Solomon Conteh; Scott R. Penzak; Michael P. Fay; Nicole M. Anders; Tiina Ilmet; Yonghua Li; William Borkowsky; Urszula Krzych; Patrick E. Duffy; Photini Sinnis

BACKGROUND Although nonnucleoside reverse transcriptase inhibitors (NNRTIs) are usually part of first-line treatment regimens for human immunodeficiency virus (HIV), their activity on Plasmodium liver stages remains unexplored. Additionally, trimethoprim-sulfamethoxazole (TMP-SMX), used for opportunistic infection prophylaxis in HIV-exposed infants and HIV-infected patients, reduces clinical episodes of malaria; however, TMP-SMX effect on Plasmodium liver stages requires further study. METHODS We characterized NNRTI and TMP-SMX effects on Plasmodium liver stages in vivo using Plasmodium yoelii. On the basis of these results, we conducted in vitro studies assessing TMP-SMX effects on the rodent parasites P. yoelii and Plasmodium berghei and on the human malaria parasite Plasmodium falciparum. RESULTS Our data showed NNRTI treatment modestly reduced P. yoelii liver stage parasite burden and minimally extended prepatent period. TMP-SMX administration significantly reduced liver stage parasite burden, preventing development of patent parasitemia in vivo. TMP-SMX inhibited development of rodent and P. falciparum liver stage parasites in vitro. CONCLUSIONS NNRTIs modestly affect liver stage Plasmodium parasites, whereas TMP-SMX prevents patent parasitemia. Because drugs that inhibit liver stages target parasites when they are present in lower numbers, these results may have implications for eradication efforts. Understanding HIV drug effects on Plasmodium liver stages will aid in optimizing treatment regimens for HIV-exposed and HIV-infected infected patients in malaria-endemic areas.


AIDS | 2013

The effect of antiretrovirals on Plasmodium falciparum liver stages.

Charlotte V. Hobbs; Patricia De La Vega; Scott R. Penzak; Jillian Van Vliet; Urszula Krzych; Photini Sinnis; William Borkowsky; Patrick E. Duffy

HIV and malaria overlap geographically, but the full impact of different antiretrovirals on malaria remains poorly understood. We examined the antimalarial activity of the HIV protease inhibitors lopinavir and saquinavir and the non-nucleoside reverse transcriptase inhibitor nevirapine on Plasmodium falciparum liver stages. Our results demonstrate that the HIV PI lopinavir inhibits liver stage parasites at clinically relevant concentrations, that is, at drug levels achieved in HIV-infected patients on standard dosing regimens. Because drugs that inhibit liver stages target parasites when they are present in lower numbers, these results might have implications for eradication efforts.


PLOS ONE | 2014

HIV Treatments Reduce Malaria Liver Stage Burden in a Non-Human Primate Model of Malaria Infection at Clinically Relevant Concentrations In Vivo

Charlotte V. Hobbs; Jillian Neal; Solomon Conteh; Liam Donnelly; Jingyang Chen; Kennan Marsh; Lynn Lambert; Sachy Orr-Gonzalez; Jessica Hinderer; Sara A. Healy; William Borkowsky; Scott R. Penzak; Sumana Chakravarty; Stephen L. Hoffman; Patrick E. Duffy

We have previously shown that the HIV protease inhibitor lopinavir-ritonavir (LPV-RTV) and the antibiotic trimethoprim sulfamethoxazole (TMP-SMX) inhibit Plasmodium liver stages in rodent malarias and in vitro in P. falciparum. Since clinically relevant levels are better achieved in the non-human-primate model, and since Plasmodium knowlesi is an accepted animal model for the study of liver stages of malaria as a surrogate for P. falciparum infection, we investigated the antimalarial activity of these drugs on Plasmodium knowlesi liver stages in rhesus macaques. We demonstrate that TMP-SMX and TMP-SMX+LPV-RTV (in combination), but not LPV-RTV alone, inhibit liver stage parasite development. Because drugs that inhibit the clinically silent liver stages target parasites when they are present in lower numbers, these results may have implications for eradication efforts.


AIDS | 2017

Buy one, get one free? Benefits of certain antiretrovirals against malaria.

Charlotte V. Hobbs; Sunil Parikh

Currently, the WHO recommends the use of combination ART for HIV management, with two nucleoside reverse transcriptase inhibitors (NRTIs) and as first line, a nonnucleoside reverse-transcriptase inhibitor (NNRTI), or, with second-line therapy, an HIV protease inhibitor, except for children under age 3 [2,3]. Interestingly, both in-vitro and in-vivo data demonstrate that HIV protease inhibitors kill various life cycle stages of malaria parasites [4–8]. Recent clinical studies have evaluated whether these data translate reduction in clinical malaria. To date, there have been a handful of studies in areas of varying malaria transmission intensity examining the impact of ARTon malaria in children, pregnant women, and adults. This is important because age, pregnancy, and transmission intensity impact acquisition of naturally acquired immunity to malaria.

Collaboration


Dive into the Charlotte V. Hobbs's collaboration.

Top Co-Authors

Avatar

Patrick E. Duffy

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jillian Neal

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Scott R. Penzak

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Brian Kirmse

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Chad Blackshear

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jingyang Chen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kengo Inagaki

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Photini Sinnis

Johns Hopkins University

View shared research outputs
Researchain Logo
Decentralizing Knowledge