Network


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

Hotspot


Dive into the research topics where Brian M. Friedrich is active.

Publication


Featured researches published by Brian M. Friedrich.


The FASEB Journal | 2002

Aldose reductase mediates cytotoxic signals of hyperglycemia and TNF-alpha in human lens epithelial cells.

Kota V. Ramana; Brian M. Friedrich; Aruni Bhatnagar; Satish K. Srivastava

Chronic hyperglycemia and cytokines such as tumor necrosis factor α (TNF‐α) cause oxidative stress leading to dysregulated cell growth or apoptosis that contributes to the development of inflammation and secondary complications of diabetes. However, the mechanisms regulating hyperglycemic or cytokine injury are not well understood. Herein we report that inhibition of the polyol pathway enzyme aldose reductase (AR) by two structurally unrelated inhibitors—sorbinil and tolrestat—prevents, in the human lens epithelial cell line B‐3, the apoptosis and activation of caspase‐3 caused by exposure to high glucose levels or TNF‐α. Inhibition of AR attenuated TNF‐α and hyperglycemia‐induced activation of protein kinase C (PKC), phosphorylation of the inhibitory subunit of nuclear factor‐κB (NF‐κB), and stimulation of NF‐κB, but it did not prevent the activation of NF‐κB and PKC by phorbol ester. Inhibition of AR also attenuated the increase in p38 mitogen‐activated protein kinase and c‐Jun N‐terminal kinase phosphorylation. These signaling pathways were also inhibited in cells in which the expression of AR was reduced by antisense ablation. Collectively, these results identify a new participant in apoptotic signaling and suggest that AR is an obligatory mediator of the apoptotic events upstream of PKC. These observations could provide new insights into the pathophysiology of diabetes and the role of aberrant glucose metabolism in apoptotic cell death.


Virus Research | 2011

Host factors mediating HIV-1 replication.

Brian M. Friedrich; Natallia Dziuba; Guangyu Li; Mark A. Endsley; James L. Murray; Monique R. Ferguson

Human immunodeficiency virus type 1(HIV-1) infection is the leading cause of death worldwide in adults attributable to infectious diseases. Although the majority of infections are in sub-Saharan Africa and Southeast Asia, HIV-1 is also a major health concern in most countries throughout the globe. While current antiretroviral treatments are generally effective, particularly in combination therapy, limitations exist due to drug resistance occurring among the drug classes. Traditionally, HIV-1 drugs have targeted viral proteins, which are mutable targets. As cellular genes mutate relatively infrequently, host proteins may prove to be more durable targets than viral proteins. HIV-1 replication is dependent upon cellular proteins that perform essential roles during the viral life cycle. Maraviroc is the first FDA-approved antiretroviral drug to target a cellular factor, HIV-1 coreceptor CCR5, and serves to intercept viral-host protein-protein interactions mediating entry. Recent large-scale siRNA and shRNA screens have revealed over 1000 candidate host factors that potentially support HIV-1 replication, and have implicated new pathways in the viral life cycle. These host proteins and cellular pathways may represent important targets for future therapeutic discoveries. This review discusses critical cellular factors that facilitate the successive steps in HIV-1 replication.


Virology | 2008

A critical role for CD63 in HIV replication and infection of macrophages and cell lines

Hui Chen; Natallia Dziuba; Brian M. Friedrich; Jana J. von Lindern; James L. Murray; Daniel R. Rojo; Thomas W. Hodge; William A. O'Brien; Monique R. Ferguson

HIV infection typically involves interaction of Env with CD4 and a chemokine coreceptor, either CCR5 or CXCR4. Other cellular factors supporting HIV replication have also been characterized. We previously demonstrated a role for CD63 in early HIV infection events in macrophages via inhibition by anti-CD63 antibody pretreatment. To confirm the requirement for CD63 in HIV replication, we decreased CD63 expression using CD63-specific short interfering RNAs (siRNA), and showed inhibition of HIV replication in macrophages. Surprisingly, pretreatment with CD63 siRNA not only silenced CD63 expression by 90%, but also inhibited HIV-1 replication in a cultured cell line (U373-MAGI) which had been previously shown to be insensitive to CD63 monoclonal antibody inhibition. Although the anti-CD63 antibody was previously shown to inhibit early HIV infection events only in macrophages, we now show a potential role for CD63 in later HIV replication events in macrophages and cell lines. Further delineation of the role of CD63 in HIV replication may lead to development of novel therapeutic compounds.


Journal of Clinical Microbiology | 2009

Dried-Plasma Transport Using a Novel Matrix and Collection System for Human Immunodeficiency Virus and Hepatitis C Virus Virologic Testing

Robert M. Lloyd; D. A. Burns; Joe Huong; R. L. Mathis; Mark A. Winters; M. Tanner; A. De La Rosa; Belinda Yen-Lieberman; W. Armstrong; A. Taege; Daniel McClernon; J. L. Wetshtein; Brian M. Friedrich; Monique R. Ferguson; William A. O'Brien; P. M. Feorino; Mark Holodniy

ABSTRACT A novel method for the collection and transportation of dried-blood-plasma samples, SampleTanker (ST), was developed and compared to standard shipping protocols for frozen-plasma specimens containing human immunodeficiency virus type 1 (HIV-1) and/or hepatitis C virus (HCV). Matched frozen and dried 1-ml EDTA-containing plasma samples were collected and analyzed by several molecular-based virologic assays. After addition of 1.175 ml of reconstitution buffer, 1.035 ml of dried plasma was recovered. Mean intra-assay variances were 0.05, 0.05, and 0.06 log10 copies/ml for the Versant, Amplicor, and NucliSens QT HIV-1 load assays, respectively (P, not significant). However, mean HIV-1 viral load was consistently reduced in dried samples by 0.32 to 0.51 log10 copies/ml, depending on assay type (P < 0.05). Infectious HIV-1 was not recovered from dried ST plasma. There was no significant difference in HIV-1 viral load results obtained using ST after 8 weeks of storage at ambient temperature. Compared to frozen plasma, HIV-1 genotypic results were >99% concordant at the nucleotide and amino acid levels, as well as for resistance-associated mutations. We further demonstrated successful detection of multiple analytes, including HIV-1 viral load, HIV-1 antiretroviral resistance genotype, and HCV genotype, from a single ST unit. Dried plasma collected with ST yielded comparable results to frozen samples for multiple-analyte clinical testing. As such, ST could be a useful alternative for virologic tests and clinical trials worldwide by significantly diminishing transportation cost and the sample volume restrictions associated with dried-blood-spot technology.


Retrovirology | 2011

A Functional Role for ADAM10 in Human Immunodeficiency Virus Type-1 Replication

Brian M. Friedrich; James L. Murray; Guangyu Li; Jinsong Sheng; Thomas W. Hodge; Donald H. Rubin; William A. O'Brien; Monique R. Ferguson

BackgroundGene trap insertional mutagenesis was used as a high-throughput approach to discover cellular genes participating in viral infection by screening libraries of cells selected for survival from lytic infection with a variety of viruses. Cells harboring a disrupted ADAM10 (A Disintegrin and Metalloprotease 10) allele survived reovirus infection, and subsequently ADAM10 was shown by RNA interference to be important for replication of HIV-1.ResultsSilencing ADAM10 expression with small interfering RNA (siRNA) 48 hours before infection significantly inhibited HIV-1 replication in primary human monocyte-derived macrophages and in CD4+ cell lines. In agreement, ADAM10 over-expression significantly increased HIV-1 replication. ADAM10 down-regulation did not inhibit viral reverse transcription, indicating that viral entry and uncoating are also independent of ADAM10 expression. Integration of HIV-1 cDNA was reduced in ADAM10 down-regulated cells; however, concomitant 2-LTR circle formation was not detected, suggesting that HIV-1 does not enter the nucleus. Further, ADAM10 silencing inhibited downstream reporter gene expression and viral protein translation. Interestingly, we found that while the metalloprotease domain of ADAM10 is not required for HIV-1 replication, ADAM15 and γ-secretase (which proteolytically release the extracellular and intracellular domains of ADAM10 from the plasma membrane, respectively) do support productive infection.ConclusionsWe propose that ADAM10 facilitates replication at the level of nuclear trafficking. Collectively, our data support a model whereby ADAM10 is cleaved by ADAM15 and γ-secretase and that the ADAM10 intracellular domain directly facilitates HIV-1 nuclear trafficking. Thus, ADAM10 represents a novel cellular target class for development of antiretroviral drugs.


Virology | 2011

A Post-Entry Role for CD63 in Early HIV-1 Replication

Guangyu Li; Natallia Dziuba; Brian M. Friedrich; James L. Murray; Monique R. Ferguson

Macrophages and CD4(+) lymphocytes are the major reservoirs for HIV-1 infection. CD63 is a tetraspanin transmembrane protein, which has been shown to play an essential role during HIV-1 replication in macrophages. In this study, we further confirm the requirement of CD63 in early HIV-1 replication events in both macrophages and a CD4(+) cell line. Further analysis revealed that viral attachment and cell-cell fusion were unaffected by CD63 silencing. However, CD63-depleted macrophages showed a significant decrease in the initiation and completion of HIV-1 reverse transcription, affecting subsequent events of the HIV-1 life cycle. Integration of HIV-1 cDNA as well as the formation of 2-LTR circles was notably reduced. Reporter assays showed that CD63 down regulation reduced production of the early HIV protein Tat. In agreement, CD63 silencing also inhibited production of the late protein p24. These findings suggest that CD63 plays an early post-entry role prior to or at the reverse transcription step.


Virology Journal | 2010

Quantitative PCR used to assess HIV-1 integration and 2-LTR circle formation in human macrophages, peripheral blood lymphocytes and a CD4+ cell line.

Brian M. Friedrich; Guangyu Li; Natallia Dziuba; Monique R. Ferguson

BackgroundIntegration is an intermediate step in the HIV life cycle and is defined as the insertion of HIV-1 proviral DNA into the host chromosome. If integration does not occur when HIV-1 cDNA enters the nucleus, it circularizes upon itself and forms a 2-LTR circle. Monitoring the level of integrated HIV-1 cDNA in different primary cell subsets is very important, particularly regarding the effect of HAART in HIV-1 infected individuals. Because of limitations of prior HIV-1 integration assays, there is limited data on the level of integration and 2-LTR circle formation in primary cell subsets, particularly in human monocyte-derived macrophages and peripheral blood lymphocytes (PBL).ResultsIn this study, we utilized a well-defined, sensitive two-step quantitative real-time PCR method to detect HIV-1 integration as well as conventional real-time PCR to detect 2-LTR circle formation in human macrophages and PBL isolated from six different healthy donors, as well as U373 CD4+ cells by infecting with HIV-1SX (R5) or dual-tropic isolate HIV-189.6 (R5/X4) virus strains. We used the FDA-approved integrase inhibitor, raltegravir, to determine quantitative differences of integrated HIV viral cDNA in HIV-1 infected cells with and without raltegravir treatment. Our results show that integration and 2-LTR circle formation can be assessed in primary macrophages, PBL, and a CD4+ cell line by this method. Specifically, our results demonstrate that this two-step real-time PCR method can distinguish between HIV-1 integrated viral cDNA and non-integrated nuclear HIV-1 2-LTR circles caused by impaired integration with raltegravir-treatment. This further confirms that only integrated HIV-1 cDNA can be specifically amplified and quantified by two-step PCR without non-specifically detecting non-integrated viral cDNA.ConclusionThese results consistently demonstrate that the well-established real-time PCR assays used are robust, sensitive and quantitative for the detection of HIV-1 integration and 2-LTR circle formation in physiologically relevant human macrophages and PBL using lab-adapted virus strains, instead of pseudovirus. With two-step real-time PCR, we show that unintegrated, nuclear HIV-1 cDNA is not detected in raltegravir-treated cells, while specific for only integrated HIV-1 cDNA in non-treated cells. These methods could be applied as a useful tool in further monitoring specific therapy in HIV-1 infected individuals.


Chemico-Biological Interactions | 2003

Role of aldose reductase in TNF-α-induced apoptosis of vascular endothelial cells

Deepak Chandra; Kota V. Ramana; Brian M. Friedrich; Sanjay Srivastava; Aruni Bhatnagar; Satish K. Srivastava

Abstract Apoptosis of vascular endothelial cells (VECs) and concomitant proliferation of the underlying vascular smooth muscle cells (VSMCs) in large arteries are the key features of atherosclerosis and restenosis. However, the mechanisms underlying endothelial cell death and abnormal smooth muscle cell proliferation during the development of vascular lesions remain unclear. We have previously demonstrated that treatment with inhibitors of the aldehyde-metabolizing enzyme and aldose reductase (AR) attenuates restenosis of balloon-injured rat carotid arteries. The inhibition of AR also prevents the apoptosis of VECs induced by the tumor necrosis factor-alpha (TNF-α). Apoptosis of the VECs was determined by the incorporation of [3H]-thymidine and the activation of caspase-3. Stimulation of the VECs with TNF-α led to an increase in the DNA-binding activity of the transcription factor, nuclear factor-kappa binding protein (NF-κB) and the induction of the adhesion molecule (ICAM)-1. Treatment of VECs with the AR inhibitor, tolrestat, prevented the activation of NF-κB and diminished ICAM-1 induction stimulated by TNF-α. These results indicate an obligatory requirement of AR activity in the transduction of intracellular signaling initiated by the ligation of the TNF-α receptors leading to the activation of NF-κB. Although the specific signaling events interrupted by AR inhibition remain unknown, our results suggest that product(s) of AR catalysis may be essential for NF-κB activation. These observations could form the basis of future investigations into the therapeutic utility of AR inhibitors in preserving endothelial function and integrity during atherosclerosis and diabetes.


Journal of Reproductive Immunology | 2011

Rapid clearance of herpes simplex virus type 2 by CD8+ T cells requires high level expression of effector T cell functions

Michelle H. Nelson; Melanie D. Bird; Chin Fun Chu; Alison J. Johnson; Brian M. Friedrich; Windy Allman; Gregg N. Milligan

CD8(+) T cells are important for resolution of HSV-2 lesions from the female genital epithelium. It is uncertain whether optimal clearance of viruses such as HSV-2 that cause a limited, non-systemic infection solely requires expression of effector functions by infiltrating CD8(+) T lymphocytes, or if the clearance rate is reflective of the expression level of critical effector functions. To address this, CD8(+) T cells from normal OT-I mice or OT-I mice deficient in IFNγ (IFNγ(-/-)) or the IFNγ receptor (IFNγR(-/-)) were activated in vitro in the presence of IFNγ or IL-4 to generate a series of effector populations (Tc1 and Tc2-like respectively) that secreted different levels of IFNγ and expressed different levels of HSV-specific cytolytic function. Compared with Tc1 cells, Tc2-like cells produced the type 2 cytokines IL-4 and IL-5, exhibited decreased IFNγ secretion, diminished proliferation in vitro, and decreased antigen-specific cytolysis in vivo. Clearance of an ovalbumin-expressing HSV-2 strain (HSV-2 tk(-) OVA) by adoptively transferred Tc2-like cells was delayed relative to Tc1 cell recipients. Because donor Tc2-like cells proliferated in vivo and infiltrated the infected genital epithelium similar to Tc1 cells, the diminished virus clearance by Tc2-like effector cells correlated with reduced expression of critical effector functions. Together, these results suggest that high level expression of protective T cell functions by effector T cells is necessary for optimal clearance of HSV-2 from the genital epithelium. These results have important implications for vaccines designed to elicit CD8(+) T cells against viruses such as HSV-2 that infect the genital tract.


Vaccine | 2016

Supramolecular peptide hydrogel adjuvanted subunit vaccine elicits protective antibody responses against West Nile virus.

Brian M. Friedrich; David W. C. Beasley; Jai S. Rudra

A crucial issue in vaccine development is to balance safety with immunogenicity. The low immunogenicity of most subunit antigens warrants a search for adjuvants able to stimulate both cell-mediated and humoral immunity. In recent years, successful applications of nanotechnology and bioengineering in the field of vaccine development have enabled the production of novel adjuvant technologies. In this work, we investigated totally synthetic and supramolecular peptide hydrogels as novel vaccine adjuvants in conjunction with the immunoprotective envelope protein domain III (EIII) of West Nile virus as an immunogen in a mouse model. Our results indicate that, compared to the clinically approved adjuvant alum, peptide hydrogel adjuvanted antigen elicited stronger antibody responses and conferred significant protection against mortality after virus challenge. The high chemical definition and biocompatibility of self-assembling peptide hydrogels makes them attractive as immune adjuvants for the production of subunit vaccines against viral and bacterial infections where antibody-mediated protection is desirable.

Collaboration


Dive into the Brian M. Friedrich's collaboration.

Top Co-Authors

Avatar

Monique R. Ferguson

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Guangyu Li

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

James L. Murray

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Kota V. Ramana

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Natallia Dziuba

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Satish K. Srivastava

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

William A. O'Brien

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Thomas W. Hodge

Centers for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Alison J. Johnson

University of Texas Medical Branch

View shared research outputs
Researchain Logo
Decentralizing Knowledge