Network


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

Hotspot


Dive into the research topics where Larry S. McDaniel is active.

Publication


Featured researches published by Larry S. McDaniel.


Infection and Immunity | 2001

PspC, a Pneumococcal Surface Protein, Binds Human Factor H

Sandhya Dave; Alexis Brooks-Walter; Michael K. Pangburn; Larry S. McDaniel

ABSTRACT PspC was found to bind human complement factor H (FH) by Western blot analysis of D39 (pspC+) and an isogenic mutant TRE108 (pspC). We confirmed that PspA does not bind FH, while purified PspC binds FH very strongly. The binding of FH to exponentially growing pneumococci varied among different isolates when analyzed by fluorescence activated cell sorting analysis.


Journal of Immunology | 2004

Dual Roles of PspC, a Surface Protein of Streptococcus pneumoniae, in Binding Human Secretory IgA and Factor H

Sandhya Dave; Stephanie Carmicle; Sven Hammerschmidt; Michael K. Pangburn; Larry S. McDaniel

Streptococcus pneumoniae, also known as the pneumococcus, contains several surface proteins that along with the polysaccharide capsule function in antiphagocytic activities and evasion of the host immune system. These pneumococcal proteins interact with the host immune system in various ways and possess a wide range of biological activities that suggests that they may be involved at different stages of pneumococcal infection. PspC, also known as CbpA and SpsA, is one of several pneumococcal surface proteins that binds host proteins, including factor H (FH) and secretory IgA (sIgA) via the secretory component. Previous work by our laboratory has demonstrated that PspC on the surface of live pneumococcal cells binds FH. This paper provides evidence that FH activity is maintained in the presence of PspC and that the PspC binding site is located in the short consensus repeat 6–10 region of FH. We also report for the first time that although both FH and sIgA binding has been localized to the α-helical domain of PspC, the binding of FH to PspC is not inhibited by sIgA. ELISA, surface plasmon resonance, and flow cytometry indicate that the two host proteins do not compete for binding with PspC and likely do not share the same binding sites. We confirmed by Western analysis that the binding sites are separate using recombinant PspC proteins. These PspC variants bind FH yet fail to bind sIgA. Thus, we conclude that FH and sIgA can bind concurrently to the α-helical region of PspC.


Infection and Immunity | 2001

Characterization of binding of human lactoferrin to pneumococcal surface protein A

Anders P. Hakansson; Hazeline Roche; Shaper Mirza; Larry S. McDaniel; Alexis Brooks-Walter; David E. Briles

ABSTRACT Human lactoferrin is an iron-binding glycoprotein that is particularly prominent in exocrine secretions and leukocytes and is also found in serum, especially during inflammation. It is able to sequester iron from microbes and has immunomodulatory functions, including inhibition of both complement activation and cytokine production. This study used mutants lacking pneumococcal surface protein A (PspA) and PspC to demonstrate that the binding of human lactoferrin to the surface of Streptococcus pneumoniae was entirely dependent on PspA. Lactoferrin bound both family 1 and family 2 PspAs. Binding of lactoferrin to PspA was shown by surface colocalization with PspA and was verified by the lack of binding to PspA-negative mutants. Lactoferrin was expressed on the body of the cells but was largely absent from the poles. PspC showed exactly the same distribution on the pneumococcal surface as PspA but did not bind lactoferrin. PspAs binding site for lactoferrin was mapped using recombinant fragments of PspA of families 1 and 2. Binding of human lactoferrin was detected primarily in the C-terminal half of the α-helical domain of PspA (amino acids 167 to 288 of PspA/Rx1), with no binding to the N-terminal 115 amino acids in either strain. The interaction was highly specific. As observed previously, bovine lactoferrin bound poorly to PspA. Human transferrin did not bind PspA at all. The binding of lactoferrin to S. pneumoniae might provide a way for the bacteria to interfere with host immune functions or to aid in the acquisition of iron at the site of infection.


Infection and Immunity | 2003

Pneumolysin-Dependent and -Independent Gene Expression Identified by cDNA Microarray Analysis of THP-1 Human Mononuclear Cells Stimulated by Streptococcus pneumoniae

P. David Rogers; Justin Thornton; Katherine S. Barker; D. Olga McDaniel; Gordon S. Sacks; Edwin Swiatlo; Larry S. McDaniel

ABSTRACT Pneumolysin is an important virulence factor of Streptococcus pneumoniae, interacting with the membranes of host cells to elicit a multitude of inflammatory responses. We used cDNA microarrays to identify genes which are responsive to S. pneumoniae in a pneumolysin-dependent and -independent fashion. The THP-1 human monocytic cell line was coincubated for 3 h with medium alone, with the virulent type 2 S. pneumoniae strain D39, or with the isogenic strain PLN, which does not express pneumolysin. RNA was isolated from the monocytes and hybridized on cDNA microarrays. Of 4,133 genes evaluated, 142 were found to be responsive in a pneumolysin-dependent fashion, whereas 40 were found to be responsive independent of pneumolysin. Genes that were up-regulated in cells exposed to D39 relative to those exposed to PLN included genes encoding proteins such as mannose binding lectin 1, lysozyme, α-1 catenin, cadherin 17, caspases 4 and 6, macrophage inflammatory protein 1β (MIP-1β), interleukin 8 (IL-8), monocyte chemotactic protein 3 (MCP-3), IL-2 receptor β (IL-2Rβ), IL-15 receptor α (IL-15Rα), interferon receptor 2, and prostaglandin E synthase. Down-regulated genes included those encoding complement component receptor 2/CD21, platelet-activating factor acetylhydrolase, and oxidized low-density lipoprotein receptor 1 (OLR1). Pneumolysin-independent responses included down-regulation of the genes encoding CD68, CD53, CD24, transforming growth factor β2, and signal transducers and activators of transcription 1. These results demonstrate the striking effects of pneumolysin on the host cell upon exposure to S. pneumoniae.


Mbio | 2012

Nontypeable Pneumococci Can Be Divided into Multiple cps Types, Including One Type Expressing the Novel Gene pspK

In Ho Park; Kyung Hyo Kim; Ana Lucia Andrade; David E. Briles; Larry S. McDaniel; Moon H. Nahm

ABSTRACT Although virulence of Streptococcus pneumoniae is associated with its capsule, some pathogenic S. pneumoniae isolates lack capsules and are serologically nontypeable (NT). We obtained 64 isolates that were identified as NT “pneumococci” (i.e., bacteria satisfying the conventional definition but without the multilocus sequence typing [MLST]-based definition of S. pneumoniae) by the traditional criteria. All 64 were optochin sensitive and had lytA, and 63 had ply. Twelve isolates had cpsA, suggesting the presence of a conventional but defective capsular polysaccharide synthesis (cps) locus. The 52 cpsA-negative isolates could be divided into three null capsule clades (NCC) based on aliC (aliB-like ORF1), aliD (aliB-like ORF2), and our newly discovered gene, pspK, in their cps loci. pspK encodes a protein with a long alpha-helical region containing an LPxTG motif and a YPT motif known to bind human pIgR. There were nine isolates in NCC1 (pspK+ but negative for aliC and aliD), 32 isolates in NCC2 (aliC+ aliD+ but negative for pspK), and 11 in NCC3 (aliD+ but negative for aliC and pspK). Among 52 cpsA-negative isolates, 41 were identified as S. pneumoniae by MLST analysis. All NCC1 and most NCC2 isolates were S. pneumoniae, whereas all nine NCC3 and two NCC2 isolates were not S. pneumoniae. Several NCC1 and NCC2 isolates from multiple individuals had identical MLST and cps regions, showing that unencapsulated S. pneumoniae can be infectious among humans. Furthermore, NCC1 and NCC2 S. pneumoniae isolates could colonize mice as well as encapsulated S. pneumoniae, although S. pneumoniae with an artificially disrupted cps locus did not. Moreover, an NCC1 isolate with pspK deletion did not colonize mice, suggesting that pspK is critical for colonization. Thus, PspK may provide pneumococci a means of surviving in the nasopharynx without capsule. IMPORTANCE The presence of a capsule is critical for many pathogenic bacteria, including pneumococci. Reflecting the pathogenic importance of the pneumococcal capsule, pneumococcal vaccines are designed to elicit anticapsule antibodies. Additional evidence for the pathogenic importance of the pneumococcal capsule is the fact that in pneumococci all the genes necessary for capsule production are together in one genetic locus, which is called the cps locus. However, there are occasional pathogenic pneumococci without capsules, and how they survive in the host without the capsule is unknown. Here, we show that in these acapsular pneumococci, the cps loci have been replaced with various novel genes and they can colonize mouse nasopharynges as well as capsulated pneumococci. Since the genes that replace the cps loci are likely to be important in host survival, they may show new and/or alternative capsule-independent survival mechanisms used by pneumococci. The presence of a capsule is critical for many pathogenic bacteria, including pneumococci. Reflecting the pathogenic importance of the pneumococcal capsule, pneumococcal vaccines are designed to elicit anticapsule antibodies. Additional evidence for the pathogenic importance of the pneumococcal capsule is the fact that in pneumococci all the genes necessary for capsule production are together in one genetic locus, which is called the cps locus. However, there are occasional pathogenic pneumococci without capsules, and how they survive in the host without the capsule is unknown. Here, we show that in these acapsular pneumococci, the cps loci have been replaced with various novel genes and they can colonize mouse nasopharynges as well as capsulated pneumococci. Since the genes that replace the cps loci are likely to be important in host survival, they may show new and/or alternative capsule-independent survival mechanisms used by pneumococci.


Fems Immunology and Medical Microbiology | 2009

Serum resistance and biofilm formation in clinical isolates of Acinetobacter baumannii

Lauren B. King; Edwin Swiatlo; Andrea Swiatlo; Larry S. McDaniel

Acinetobacter baumannii has few known virulence factors and yet causes a variety of opportunistic infections. Many gram-negative bacteria are directly killed by complement, but we hypothesized that A. baumannii would be resistant to serum killing. A serum bactericidal assay assessed the resistance of seven A. baumannii isolates to serum killing, and C2-deficient serum was used to examine its activation of the alternative pathway. Flow cytometry was utilized to determine whether complement regulator factor H (FH) was bound by A. baumannii, and to assay C3 deposition on cells. A microtiter biofilm assay compared biofilm production among isolates. Of seven isolates, four were serum sensitive and three were serum resistant. The C2-deficient serum demonstrated that A. baumannii can activate the alternative pathway. None of the isolates bound FH. Serum-resistant strains accumulated little C3 when exposed to human serum, while sensitive strains had a high amount of surface C3 deposition. Biofilm production varied extensively among strains. Most serum-resistant isolates formed a substantial amount of biofilm, while sensitive isolates produced negligible amounts of biofilm. Our data indicate that some strains of A. baumannii are resistant to serum killing and produce biofilms and by understanding the resistance mechanisms used by this bacterium, we can further elucidate its complex pathogenicity.


Infection and Immunity | 2007

Pneumolysin, PspA, and PspC Contribute to Pneumococcal Evasion of Early Innate Immune Responses during Bacteremia in Mice

Lisa R. Quin; Quincy C. Moore; Larry S. McDaniel

ABSTRACT The pneumococcal virulence factors include capsule, PspA, PspC, and Ply. Cytometric analysis demonstrated that the greatest levels of C3 deposition were on a Δply PspA− PspC− mutant. Also, Ply, PspA, and PspC expression resulted in C3 degradation in vitro and in vivo. Finally, blood clearance assays demonstrated that there was enhanced clearance of Δply PspA− PspC− pneumococci compared to the clearance of nonencapsulated pneumococci.


Infection and Immunity | 2007

Factor H Binding to PspC of Streptococcus pneumoniae Increases Adherence to Human Cell Lines In Vitro and Enhances Invasion of Mouse Lungs In Vivo

Lisa R. Quin; Chinwendu Onwubiko; Quincy C. Moore; Megumi Fujioka Mills; Larry S. McDaniel; Stephanie Carmicle

ABSTRACT Pneumococcal surface protein C (PspC) binds to both human secretory immunoglobulin A (sIgA) and complement factor H (FH). FH, a regulator of the alternative pathway of complement, can also mediate adherence of different host cells. Since PspC contributes to adherence and invasion of host cells, we hypothesized that the interaction of PspC with FH may also mediate adherence of pneumococci to human cells. In this study, we investigated FH- and sIgA-mediated pneumococcal adherence to human cell lines in vitro. Adherence assays demonstrated that preincubation of Streptococcus pneumoniae D39 with FH increased adherence to human umbilical vein endothelial cells (HUVEC) 5-fold and to lung epithelial cells (SK-MES-1) 18-fold, relative to that of D39 without FH on the surface. The presence of sIgA enhanced adherence to SK-MES-1 6-fold and to pharyngeal epithelial cells (Detroit 562) 14-fold. Furthermore, sIgA had an additive effect on adherence to HUVEC; specifically, preincubation of D39 with both FH and sIgA led to a 21-fold increase in adherence. Finally, using a mouse model, we examined the significance of the FH-PspC interaction in pneumococcal nasal colonization and lung invasion. Mice intranasally infected with D39 preincubated with FH had increased bacteremia and lung invasion, but they had similar levels of nasopharyngeal colonization compared to that of mice challenged with D39 without FH.


The Journal of Infectious Diseases | 2005

In Vivo Binding of Complement Regulator Factor H by Streptococcus pneumoniae

Lisa R. Quin; Stephanie Carmicle; Sandhya Dave; Michael K. Pangburn; Jason P. Evenhuis; Larry S. McDaniel

Pneumococcal surface protein C (PspC) binds to the complement regulatory protein factor H (FH), which inhibits alternative pathway activation. In the present study, using a mouse model of systemic infection and flow-cytometric analyses, we demonstrated an in vivo interaction between FH and pneumococci and showed differential FH binding during bacteremia. Flow-cytometric analyses of pneumococci harvested after intraperitoneal (ip) challenge demonstrated increased binding of FH, compared with that after intravenous (iv) challenge. Real-time polymerase chain reaction analyses of PspC mRNA showed that, relative to pneumococci grown in vitro, those recovered from the blood of mice 24 h after iv challenge exhibited 23-fold higher mRNA levels; however, after ip challenge, PspC mRNA induction was increased 870-fold. A subsequent increase in PspC expression was detected by flow cytometry using a monoclonal antibody against PspC. Furthermore, pneumococci with FH bound to complement before exposure had increased proliferation, compared with pneumococci not pretreated with FH. These results suggest that the interaction between PspC and FH contributes to pneumococcal virulence.


Infection and Immunity | 2003

Relative Roles of Genetic Background and Variation in PspA in the Ability of Antibodies to PspA To Protect against Capsular Type 3 and 4 Strains of Streptococcus pneumoniae

Hazeline Roche; Bing Ren; Larry S. McDaniel; Anders P. Hakansson; David E. Briles

ABSTRACT Pneumococcal surface protein A (PspA) is able to elicit antibodies in mice and humans that can protect mice against fatal infection with Streptococcus pneumoniae. It has been observed that immunization with a single family 1 PspA can protect mice against infections with capsular type 3 or 6B strains expressing PspA family 1 or 2. However, several studies have shown that immunity to PspA is less efficacious against several capsular type 4 strains than against strains of capsular types 3, 6A, and 6B. To determine whether the greater difficulty in protecting against capsular type 4 strains resulted from differences in their PspAs or from differences in their genetic backgrounds, we performed protection experiments using four different challenge strains: a capsular type 3 strain expressing a family 1 PspA (WU2), a capsular type 4 strain expressing a family 2 PspA (TIGR4), and genetically engineered variants of WU2 and TIGR4 expressing each others PspAs. Prior to infection, the mice were immunized with recombinant family 1 or family 2 PspA. The results revealed that much of the difficulty in protecting against capsular type 4 strains was eliminated when mice were immunized with a homologous PspA of the same PspA family. However, regardless of which PspA the strains expressed, those on the TIGR4 background were about twice as hard to protect against as WU2 strains expressing the same PspA based on the efficacy rates seen in our experiments. These results point out the importance of including more than one PspA in any PspA vaccines developed for human use.

Collaboration


Dive into the Larry S. McDaniel's collaboration.

Top Co-Authors

Avatar

David E. Briles

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Edwin Swiatlo

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lance E. Keller

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Quincy C. Moore

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Mary E. Marquart

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lisa R. Quin

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Melissa E. Sanders

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Chinwendu Onwubiko

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jessica L. Bradshaw

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Haley R. Pipkins

University of Mississippi Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge