Network


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

Hotspot


Dive into the research topics where Brian J. Balin is active.

Publication


Featured researches published by Brian J. Balin.


Neurobiology of Aging | 2004

Chlamydia pneumoniae induces Alzheimer-like amyloid plaques in brains of BALB/c mice

C. Scott Little; Christine J. Hammond; Angela MacIntyre; Brian J. Balin; Denah M. Appelt

Amyloid deposits resembling plaques found in Alzheimers disease (AD) brains were formed in the brains of non-transgenic BALB/c mice following intranasal infection with Chlamydia pneumoniae. The mice were infected at 3 months of age with C. pneumoniae isolated from an AD brain. Infection was confirmed by light and electron microscopy in olfactory tissues of the mice. C. pneumoniae was still evident in these tissues 3 months after the initial infection indicating that a persistent infection had been established. Amyloid beta (Abeta) 1-42 immunoreactive deposits were identified in the brains of infected BALB/c mice up to 3 months post-infection with the density, size, and number of deposits increasing as the infection progressed. A subset of deposits exhibited thioflavin-s labeling. Intracellular Abeta1-42 labeling was observed in neuronal cells. Experimental induction of amyloid deposition in brains of non-transgenic BALB/c mice following infection with C. pneumoniae may be a useful model for furthering our understanding of mechanisms, linked to infection, involved in the initiation of the pathogenesis of sporadic AD.


Journal of Alzheimer's Disease | 2016

Microbes and Alzheimer's Disease

Ruth F. Itzhaki; Richard Lathe; Brian J. Balin; Melvyn J. Ball; Elaine L. Bearer; Heiko Braak; María J. Bullido; Chris Carter; Mario Clerici; S. Louise Cosby; Kelly Del Tredici; Hugh J. Field; Tamas Fulop; Claudio Grassi; W. Sue T. Griffin; Jürgen Haas; Alan P. Hudson; Angela R. Kamer; Douglas B. Kell; Federico Licastro; Luc Letenneur; Hugo Lövheim; Roberta Mancuso; Judith Miklossy; Carola Otth; Anna Teresa Palamara; George Perry; Chris M. Preston; Etheresia Pretorius; Timo E. Strandberg

We are researchers and clinicians working on Alzheimer’s disease (AD) or related topics, and we write to express our concern that one particular aspect of the disease has been neglected, even thoug ...


Journal of Histochemistry and Cytochemistry | 1996

Localization of transglutaminase in hippocampal neurons: implications for Alzheimer's disease.

Denah M. Appelt; G.C. Kopen; L.J. Boyne; Brian J. Balin

The purpose of this investigation was to identify and localize tissue transglutaminase (TGase) within neurons from the hippocampi of normal aged individuals and of those with confirmed Alzheimers disease (AD). This enzyme may be a factor in the molecular mechanisms of neurodegeneration and formation of insoluble macromolecular complexes found in the neurons of normal aged and AD brain tissue. An antibody made to the extracellular TGase, coagulation factor XIIIa, was found to be specific for purified intracellular guinea pig liver tissue TGase. The specificity for liver tissue TGase has enabled us to identify tissue TGase(s) within rat hippocampal neurons and within neurons from normal aged and AD hippocampal tissues. Degenerating neurons from the AD hippocampus, compared to neurons from the normal aged hippocampus, exhibited increased immunoreactivity for TGase and demonstrated co-labeling for PHF1 and anti-TGase. Our results suggest that TGase may be associated with the neurofibrillary degeneration observed in AD, thereby implicating TGase as a potential factor in the pathogenesis of Alzheimers disease.


Journal of Neuroscience Research | 2003

Chlamydia pneumoniae infection promotes the transmigration of monocytes through human brain endothelial cells

Angela MacIntyre; R. Abramov; Christine J. Hammond; Alan P. Hudson; E.J. Arking; Christopher S. Little; Denah M. Appelt; Brian J. Balin

We have investigated the effects of Chlamydia pneumoniae on human brain endothelial cells (HBMECs) and human monocytes as a mechanism for breaching the blood‐brain barrier (BBB) in Alzheimers disease (AD). HBMECs and peripheral blood monocytes may be key components in controlling the entry of C. pneumoniae into the human brain. Our results indicate that C. pneumoniae infects blood vessels and monocytes in AD brain tissues compared with normal brain tissue. C. pneumoniae infection stimulates transendothelial entry of monocytes through HBMECs. This entry is facilitated by the up‐regulation of VCAM‐1 and ICAM‐1 on HBMECs and a corresponding increase of LFA‐1, VLA‐4, and MAC‐1 on monocytes. C. pneumoniae infection in HBMECs and THP‐1 monocytes up‐regulates monocyte transmigration threefold in an in vitro brain endothelial monolayer. In this way, C. pneumoniae infection in these cell types may contribute to increased monocyte migration and promote inflammation within the CNS resulting from infection at the level of the vasculature. Thus, infection at the level of the vasculature may be a key initiating factor in the pathogenesis of neurodegenerative diseases such as sporadic AD.


BMC Neuroscience | 2010

Immunohistological detection of Chlamydia pneumoniae in the Alzheimer's disease brain

Christine J. Hammond; Loretta R Hallock; Raymond J Howanski; Denah M. Appelt; C. Scott Little; Brian J. Balin

BackgroundSporadic late-onset Alzheimers disease (AD) appears to evolve from an interplay between genetic and environmental factors. One environmental factor that continues to be of great interest is that of Chlamydia pneumoniae infection and its association with late-onset disease. Detection of this organism in clinical and autopsy samples has proved challenging using a variety of molecular and histological techniques. Our current investigation utilized immunohistochemistry with a battery of commercially available anti-C. pneumoniae antibodies to determine whether C. pneumoniae was present in areas typically associated with AD neuropathology from 5 AD and 5 non-AD control brains.ResultsImmunoreactivity for C. pneumoniae antigens was observed both intracellularly in neurons, neuroglia, endothelial cells, and peri-endothelial cells, and extracellularly in the frontal and temporal cortices of the AD brain with multiple C. pneumoniae-specific antibodies. This immunoreactivity was seen in regions of amyloid deposition as revealed by immunolabeling with two different anti-beta amyloid antibodies. Thioflavin S staining, overlaid with C. pneumoniae immunolabeling, demonstrated no direct co-localization of the organism and amyloid plaques. Further, the specificity of C. pneumoniae labeling of AD brain sections was demonstrated using C. pneumoniae antibodies pre-absorbed against amyloid β 1-40 and 1-42 peptides.ConclusionsAnti-C. pneumoniae antibodies, obtained commercially, identified both typical intracellular and atypical extracellular C. pneumoniae antigens in frontal and temporal cortices of the AD brain. C. pneumoniae, amyloid deposits, and neurofibrillary tangles were present in the same regions of the brain in apposition to one another. Although additional studies are required to conclusively characterize the nature of Chlamydial immunoreactivity in the AD brain, these results further implicate C. pneumoniae infection with the pathogenesis of Alzheimers disease.


Annals of the New York Academy of Sciences | 2006

Association between Sézary T Cell‐activating Factor, Chlamydia pneumoniae, and Cutaneous T Cell Lymphoma

J. Todd Abrams; Brian J. Balin; Eric C. Vonderheid

Abstract: Séezary T cell‐activating factor (SAF) was originally defined as an inducer of functional interleukin‐2 (IL‐2) receptors on normal and malignant T cells in patients suffering from Sézary syndrome. In fact, a combination of SAF and IL‐2 stimulated the propagation of T cell lines from the peripheral blood mononuclear cells (PBMC) of those patients, with approximately one third of those cell lines containing the predominant malignant clone as determined via cytogenetic and/or T cell receptor gene rearrangement analysis. Although the primary source of SAF was mitogen‐stimulated PBMC of a patient with Sézary syndrome, we were unable to isolate the gene encoding SAF from eukaryotic libraries. However, we observed SAF activity in the cytoplasm of one of the malignant cell lines in a complex containing RNA and DNA. This observation led us to consider the possibility that SAF is not of eukaryotic origin. Intracellular pathogens replicate in the cytoplasm of host cells and contain proteins, DNA, and RNA. Using a panel of antichlamydial antibodies with confirmation from polymerase chain reaction primers, we found that most patients with mycosis fungoides were positive for these determinants. Immunoelectron microscopy and protein blotting further confirmed antibody reactivity. We showed that Chlamydia pneumoniae were capable of infecting normal human keratinocytes in culture. We also demonstrated that C. pneumoniae antigen expression was associated with active disease because these determinants were not expressed after psoralen and ultraviolet A therapy. We hypothesize that chronic infection by C. pneumoniae leads to expansion of C. pneumoniae‐specific T cells, thereby potentiating the development of cutaneous T cell lymphoma.


BMC Neuroscience | 2008

Inhibition of apoptosis in neuronal cells infected with Chlamydophila (Chlamydia) pneumoniae

Denah M. Appelt; Maria Roupas; Deana S. Way; Marcus G. Bell; Elizabeth V. Albert; Christine J. Hammond; Brian J. Balin

BackgroundChlamydophila (Chlamydia) pneumoniae is an intracellular bacterium that has been identified within cells in areas of neuropathology found in Alzheimer disease (AD), including endothelia, glia, and neurons. Depending on the cell type of the host, infection by C. pneumoniae has been shown to influence apoptotic pathways in both pro- and anti-apoptotic fashions. We have hypothesized that persistent chlamydial infection of neurons may be an important mediator of the characteristic neuropathology observed in AD brains. Chronic and/or persistent infection of neuronal cells with C. pneumoniae in the AD brain may affect apoptosis in cells containing chlamydial inclusions.ResultsSK-N-MC neuroblastoma cells were infected with the respiratory strain of C. pneumoniae, AR39 at an MOI of 1. Following infection, the cells were either untreated or treated with staurosporine and then examined for apoptosis by labeling for nuclear fragmentation, caspase activity, and membrane inversion as indicated by annexin V staining. C. pneumoniae infection was maintained through 10 days post-infection. At 3 and 10 days post-infection, the infected cell cultures appeared to inhibit or were resistant to the apoptotic process when induced by staurosporine. This inhibition was demonstrated quantitatively by nuclear profile counts and caspase 3/7 activity measurements.ConclusionThese data suggest that C. pneumoniae can sustain a chronic infection in neuronal cells by interfering with apoptosis, which may contribute to chronic inflammation in the AD brain.


Infection and Immunity | 2005

Age Alterations in Extent and Severity of Experimental Intranasal Infection with Chlamydophila pneumoniae in BALB/c Mice

Christopher S. Little; Andrew Bowe; Richard Lin; Jason Litsky; Robert M Fogel; Brian J. Balin; Kerin L. Fresa-Dillon

ABSTRACT The intracellular bacterium Chlamydophila (“Chlamydia”) pneumoniae is a pathogen for several respiratory diseases and may be a factor in the pathogenesis of chronic diseases of aging including atherosclerosis and Alzheimers disease. We assessed whether aging is coupled with increased burden of infection in BALB/c mice after intranasal infection by C. pneumoniae. Six- and twenty-month-old BALB/c mice were infected intranasally with 5 × 104 inclusion forming units (IFU) or 5 × 105 IFU of C. pneumoniae. Lung, brain, and heart tissue were analyzed for infectious C. pneumoniae and for Chlamydophila antigen by immunohistochemistry. At both doses, aging was associated with a decreased proportion of animals that cleared infection from the lung and greater burden of infectious organism within the lung. We observed dose-dependent spread to the heart/ascending aorta in animals infected with C. pneumoniae. In mice given 5 × 104 IFU, spread to the heart by day 14 was only observed in old mice. By day 28, all animals inoculated with 5 × 104 IFU showed evidence of spread to the heart, although higher C. pneumoniae titers were observed in the hearts from old mice. In mice inoculated with 5 × 105 IFU, spread of C. pneumoniae to the heart was evident by day 14, with no discernible age effect. C. pneumoniae was also recovered from the central nervous system (brain and olfactory bulb) of all mice by day 28 postinfection, with higher C. pneumoniae titers in old animals than in young animals. Our results suggest that infection with C. pneumoniae may be more severe in old animals.


Frontiers in Aging Neuroscience | 2014

Detection of bacterial antigens and Alzheimer's disease-like pathology in the central nervous system of BALB/c mice following intranasal infection with a laboratory isolate of Chlamydia pneumoniae.

Christopher S. Little; Timothy Joyce; Christine J. Hammond; Hazem Matta; David Cahn; Denah M. Appelt; Brian J. Balin

Pathology consistent with that observed in Alzheimer’s disease (AD) has previously been documented following intranasal infection of normal wild-type mice with Chlamydia pneumoniae (Cpn) isolated from an AD brain (96-41). In the current study, BALB/c mice were intranasally infected with a laboratory strain of Cpn, AR-39, and brain and olfactory bulbs were obtained at 1–4 months post-infection (pi). Immunohistochemistry for amyloid beta or Cpn antigens was performed on sections from brains of infected or mock-infected mice. Chlamydia-specific immunolabeling was identified in olfactory bulb tissues and in cerebrum of AR-39 infected mice. The Cpn specific labeling was most prominent at 1 month pi and the greatest burden of amyloid deposition was noted at 2 months pi, whereas both decreased at 3 and 4 months. Viable Cpn was recovered from olfactory bulbs of 3 of 3 experimentally infected mice at 1 and 3 months pi, and in 2 of 3 mice at 4 months pi. In contrast, in cortical tissues of infected mice at 1 and 4 months pi no viable organism was obtained. At 3 months pi, only 1 of 3 mice had a measurable burden of viable Cpn from the cortical tissues. Mock-infected mice (0 of 3) had no detectable Cpn in either olfactory bulbs or cortical tissues. These data indicate that the AR-39 isolate of Cpn establishes a limited infection predominantly in the olfactory bulbs of BALB/c mice. Although infection with the laboratory strain of Cpn promotes deposition of amyloid beta, this appears to resolve following reduction of the Cpn antigen burden over time. Our data suggest that infection with the AR-39 laboratory isolate of Cpn results in a different course of amyloid beta deposition and ultimate resolution than that observed following infection with the human AD-brain Cpn isolate, 96-41. These data further support that there may be differences, possibly in virulence factors, between Cpn isolates in the generation of sustainable AD pathology.


Antimicrobial Agents and Chemotherapy | 2012

NLRP3 Inflammasome Is a Target for Development of Broad-Spectrum Anti-Infective Drugs

James D. Thacker; Brian J. Balin; Denah M. Appelt; Sihem Sassi-Gaha; Mitali Purohit; Richard F. Rest; Carol M. Artlett

ABSTRACT We describe the molecular mode of action and pharmacodynamics of a new molecular entity (NME) that induces the NLRP3 inflammasome-mediated innate immune response. This innate response reduces the pathogen load in an experimentally induced methicillin-resistant Staphylococcos aureus infection, enhances survival in an experimentally induced Gram-negative bacteremia, and overrides the escape mechanism of an obligate intracellular pathogen, viz. Chlamydia pneumoniae. Furthermore, the NME is more effective than standard-of-care antibiotic therapy in a clinically established multifactorial bacterial infection. Analysis of transcriptional regulation of inflammasome signaling genes and innate/adaptive immune genes revealed consistent and significant host changes responsible for the improved outcomes in these infections. These studies pave the way for the development of first-in-class drugs that enhance inflammasome-mediated pathogen clearance and identify the NLRP3 inflammasome as a drug target to address the global problem of emerging new infectious diseases and the reemergence of old diseases in an antibiotic-resistant form.

Collaboration


Dive into the Brian J. Balin's collaboration.

Top Co-Authors

Avatar

Denah M. Appelt

Philadelphia College of Osteopathic Medicine

View shared research outputs
Top Co-Authors

Avatar

Christine J. Hammond

Philadelphia College of Osteopathic Medicine

View shared research outputs
Top Co-Authors

Avatar

Susan T. Hingley

Philadelphia College of Osteopathic Medicine

View shared research outputs
Top Co-Authors

Avatar

Christopher S. Little

Philadelphia College of Osteopathic Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Juliana Zoga

Philadelphia College of Osteopathic Medicine

View shared research outputs
Top Co-Authors

Avatar

C. Scott Little

Philadelphia College of Osteopathic Medicine

View shared research outputs
Top Co-Authors

Avatar

Marcus G. Bell

Philadelphia College of Osteopathic Medicine

View shared research outputs
Top Co-Authors

Avatar

Morgan M Devins

Philadelphia College of Osteopathic Medicine

View shared research outputs
Top Co-Authors

Avatar

Ahmad B. Cader

Philadelphia College of Osteopathic Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge