Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Am J Pathol ; 191(12): 2133-2146, 2021 12.
Article in English | MEDLINE | ID: mdl-34428423

ABSTRACT

Murine tumors are indispensable model systems in preclinical immuno-oncology research. While immunologic heterogeneity is well-known to be an important factor that can influence treatment outcome, there is a severe paucity of data concerning the nature of this heterogeneity in murine tumor models. Using serial sectioning methodology combined with IHC analysis and whole-slide image analysis, the depth-dependent variation in immune-cell abundance in tumor specimens was investigated at single-cell resolution. Specifically, intra- and intertumor variability in cell density of nine immune-cell biomarkers was quantified in multiple murine tumor models. The analysis showed that intertumor variability was typically the dominant source of variation in measurements of immune-cell densities. Statistical power analysis revealed the effect of group size and variance in immune-cell density on the predictive power of detecting a statistically meaningful fold-change in immune-cell density. Intertumor variability in the ratio of immune-cell densities showed distinct patterns in select tumor models and revealed the existence of strong correlations between select biomarker pairs. Furthermore, the relative proportion of immune cells at different depths across tumor samples was preserved in some but not all tumor models, thereby revealing the existence of compositional heterogeneity. Taken together, these results reveal novel insights into the nature of immunologic heterogeneity, which is not accessible through typical omics approaches.


Subject(s)
Lymphocytes, Tumor-Infiltrating/pathology , Neoplasms/immunology , Animals , Biological Variation, Individual , Cell Count , Chemotaxis, Leukocyte/physiology , Female , Gene Expression Regulation, Neoplastic , Immunophenotyping , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Neoplasms/genetics , Neoplasms/pathology , Transplantation, Isogeneic , Tumor Cells, Cultured , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
2.
Regul Toxicol Pharmacol ; 123: 104934, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33872740

ABSTRACT

Systemic toxicity assessments for oral or parenteral drugs often utilize the concentration of drug in plasma to enable safety margin calculations for human risk assessment. For topical drugs, there is no standard method for measuring drug concentrations in the stratum basale of the viable epidermis. This is particularly important since the superficial part of the epidermis, the stratum corneum (SC), is nonviable and where most of a topically applied drug remains, never penetrating deeper into the skin. We investigated the relative concentrations of a prototype kinase inhibitor using punch biopsy, laser capture microdissection, and imaging mass spectrometry methods in the SC, stratum basale, and dermis of minipig skin following topical application as a cream formulation. The results highlight the value of laser capture microdissection and mass spectrometry imaging in quantifying the large difference in drug concentration across the skin and even within the epidermis, and supports use of these methods for threshold-based toxicity risk assessments in specific anatomic locations of the skin, like of the stratum basale.


Subject(s)
Pharmaceutical Preparations/metabolism , Skin Absorption/physiology , Skin/metabolism , Animals , Epidermis , Humans , Mass Spectrometry , Risk Assessment , Swine , Swine, Miniature/physiology
3.
Regul Toxicol Pharmacol ; 112: 104587, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32006671

ABSTRACT

Adalimumab, a recombinant fully human monoclonal antibody targeting tumor necrosis factor (TNF), is approved in the United States and Europe to treat various inflammatory and autoimmune indications. Biosimilars are approved biologics highly similar, but not identical, to approved biotherapeutics. To support clinical development of PF-06410293, an adalimumab biosimilar, nonclinical studies evaluated the structural, functional, toxicologic, and toxicokinetic similarity to originator adalimumab sourced from the United States (adalimumab-US) and European Union (adalimumab-EU). Structural similarity was assessed by peptide mapping. Biologic activity was measured via inhibition of TNF-induced apoptosis and Fc-based functionality assessments. In vivo nonclinical similarity was evaluated in a toxicity study in cynomolgus monkeys administered subcutaneous PF-06410293 or adalimumab-EU (0 or 157 mg/kg/week). Peptide mapping demonstrated PF-06410293, adalimumab-US, and adalimumab-EU had identical amino acid sequences. Comparative functional and binding assessments were similar. Effects of PF-06410293 and adalimumab-EU were similar and limited to pharmacologically mediated decreased cellularity of lymphoid follicles and germinal centers in spleen. Toxicokinetics were similar; maximum plasma concentration and area-under-the-concentration-time curve ratio of PF-06410293:adalimumab-EU ranged from 1.0 to 1.2. These studies supported PF-06410293 entry into clinical development. Many regulatory agencies now only request nonclinical in vivo testing if there is residual uncertainty regarding biosimilarity after in vitro analytical studies.


Subject(s)
Adalimumab/pharmacokinetics , Biosimilar Pharmaceuticals/pharmacokinetics , Adalimumab/blood , Adalimumab/chemistry , Animals , Biosimilar Pharmaceuticals/blood , Biosimilar Pharmaceuticals/chemistry , European Union , Female , Humans , Macaca fascicularis , Male , Tissue Distribution , U937 Cells , United States
4.
J Allergy Clin Immunol ; 137(4): 1079-1090, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27059729

ABSTRACT

BACKGROUND: Tofacitinib is an oral Janus kinase inhibitor being investigated for psoriasis. OBJECTIVE: We sought to elucidate the molecular mechanisms underlying the clinical efficacy of tofacitinib in patients with psoriasis. METHODS: Twelve patients with plaque psoriasis were randomized (3:1) to receive 10 mg of tofacitinib or placebo twice daily for 12 weeks. Biopsy specimens were taken from nonlesional (baseline) and lesional (baseline, days 1 and 3, and weeks 1, 2, 4, and 12) skin. Biopsy specimens were examined for psoriatic epidermal features (thickness, Ki67(+) keratinocytes and keratin 16 [KRT16] mRNA expression, and phosphorylated signal transducer and activator of transcription [pSTAT](+) nuclei) and T-cell and dendritic cell (DC) subsets by using immunohistochemistry, and mRNA transcripts were quantified by using a microarray. RESULTS: In lesional skin keratinocyte pSTAT1 and pSTAT3 staining was increased at baseline but reduced after 1 day of tofacitinib (baseline, median of 1290 pSTAT1(+) cells/µm(2); day 1, median of 332 pSTAT1(+) cells/µm(2); and nonlesional, median of 155 pSTAT1(+) cells/µm(2)). Epidermal thickness and KRT16 mRNA expression were significantly and progressively reduced after days 1 and 3 of tofacitinib administration, respectively (eg, KRT16 decreased 2.74-fold, day 3 vs baseline, P = .016). Decreases in DC and T-cell numbers were observed after weeks 1 and 2, respectively. At week 4, significant decreases in IL-23/TH17 pathways were observed that persisted through week 12. Improvements in clinical and histologic features were strongly associated with changes in expression of psoriasis-related genes and reduction in IL-17 gene expression. CONCLUSIONS: Tofacitinib has a multitiered response in patients with psoriasis: (1) rapid attenuation of keratinocyte Janus kinase/STAT signaling; (2) removal of keratinocyte-induced cytokine signaling, leading to reductions in pathologic DC and T-cell numbers to nonlesional levels; and (3) inhibition of the IL-23/TH17 pathway.


Subject(s)
Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Psoriasis/drug therapy , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Biopsy , Double-Blind Method , Drug Administration Schedule , Female , Humans , Keratinocytes/immunology , Keratinocytes/metabolism , Male , Middle Aged , Psoriasis/immunology , Psoriasis/metabolism , Psoriasis/pathology , Signal Transduction , Skin/immunology , Skin/metabolism , Skin/pathology , Treatment Outcome , Young Adult
5.
BMC Genomics ; 15: 485, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24942259

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are small, non-coding RNAs that regulate protein levels post-transcriptionally. miRNAs play important regulatory roles in many cellular processes and have been implicated in several diseases. Recent studies have reported significant levels of miRNAs in a variety of body fluids, raising the possibility that miRNAs could serve as useful biomarkers. Next-generation sequencing (NGS) is increasingly employed in biomedical investigations. Although concordance between this platform and qRT-PCR based assays has been reported in high quality specimens, information is lacking on comparisons in biofluids especially urine. Here we describe the changes in miRNA expression patterns in a rodent model of renal tubular injury (gentamicin). Our aim is to compare RNA sequencing and qPCR based miRNA profiling in urine specimen from control and rats with confirmed tubular injury. RESULTS: Our preliminary examination of the concordance between miRNA-seq and qRT-PCR in urine specimen suggests minimal agreement between platforms probably due to the differences in sensitivity. Our results suggest that although miRNA-seq has superior specificity, it may not detect low abundant miRNAs in urine samples. Specifically, miRNA-seq did not detect some sequences which were identified by qRT-PCR. On the other hand, the qRT-PCR analysis was not able to detect the miRNA isoforms, which made up the majority of miRNA changes detected by NGS. CONCLUSIONS: To our knowledge, this is the first time that miRNA profiling platforms including NGS have been compared in urine specimen. miRNAs identified by both platforms, let-7d, miR-203, and miR-320, may potentially serve as promising novel urinary biomarkers for drug induced renal tubular epithelial injury.


Subject(s)
Kidney Tubules/metabolism , MicroRNAs/genetics , MicroRNAs/urine , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Acute Kidney Injury/urine , Animals , Biomarkers , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Regulatory Networks , Gentamicins/administration & dosage , Gentamicins/adverse effects , Gentamicins/toxicity , High-Throughput Nucleotide Sequencing , Kidney Tubules/drug effects , Kidney Tubules/pathology , Male , RNA Interference , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction
6.
Toxicol Pathol ; 42(1): 229-42, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24226507

ABSTRACT

Pancreatic toxicity commonly affects the endocrine or exocrine pancreas. However, it can also occur at the endocrine-exocrine interface (EEI), where the capillary network of the islet merges with the capillaries of the surrounding acinar tissue, that is, the insulo-acinar portal system. The goal of this article is to describe a novel, test article-induced pancreatic toxicity that originated at the EEI and to summarize investigations into the mechanistic basis of the injury. This injury was initially characterized by light microscopy in 7/14 day-toxicity studies in Sprague-Dawley (Crl: CD®[SD]) rats with undisclosed test articles. Microvascular injury at the interface resulted in peri-islet serum exudation, fibrin deposition, hemorrhage, inflammation, and secondary degeneration/necrosis of surrounding exocrine tissue. More chronic injury presented as islet fibrosis and lobular atrophy. Direct cytotoxicity affecting the capillary endothelium at the EEI was confirmed ultrastructurally on day 4. Endothelial microparticle and blood flow studies further confirmed endothelial involvement. Similar lesions occurred less frequently in 2 other rat strains and not in the mouse, dog, or cynomolgus macaque. In summary, in vivo and investigative study data confirmed primary endothelial cytotoxicity in the pathogenesis of this lesion and suggested that the lesion may be rat/rat strain-specific and of uncertain relevance for human safety risk assessment.


Subject(s)
Islets of Langerhans/drug effects , Lead/toxicity , Pancreas, Exocrine/drug effects , Pancreas/drug effects , Pancreatitis/pathology , Animals , Capillaries/drug effects , Capillaries/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Hemodynamics , Hemorrhage/chemically induced , Hemorrhage/pathology , Islets of Langerhans/pathology , Male , Pancreas/pathology , Pancreas, Exocrine/pathology , Pancreatitis/chemically induced , Portal System/drug effects , Portal System/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Risk Assessment , Toxicity Tests, Acute
7.
Sci Rep ; 14(1): 8496, 2024 04 11.
Article in English | MEDLINE | ID: mdl-38605049

ABSTRACT

We present a rigorous validation strategy to evaluate the performance of Ultivue multiplex immunofluorescence panels. We have quantified the accuracy and precision of four different multiplex panels (three human and one mouse) in tumor specimens with varying levels of T cell density. Our results show that Ultivue panels are typically accurate wherein the relative difference in cell proportion between a multiplex image and a 1-plex image is less than 20% for a given biomarker. Ultivue panels exhibited relatively high intra-run precision (CV ≤ 25%) and relatively low inter-run precision (CV >> 25%) which can be remedied by using local intensity thresholding to gate biomarker positivity. We also evaluated the reproducibility of cell-cell distance estimates measured from multiplex images which show high intra- and inter-run precision. We introduce a new metric, multiplex labeling efficiency, which can be used to benchmark the overall fidelity of the multiplex data across multiple batch runs. Taken together our results provide a comprehensive characterization of Ultivue panels and offer practical guidelines for analyzing multiplex images.


Subject(s)
Neoplasms , Animals , Humans , Mice , Biomarkers , Formaldehyde , Neoplasms/pathology , Paraffin Embedding/methods , Reproducibility of Results
8.
NMR Biomed ; 26(12): 1742-50, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23939596

ABSTRACT

Fingolimod (FTY720) is an orally available sphingosine-1-phosphate (S1P) receptor modulator reducing relapse frequency in patients with relapsing-remitting multiple sclerosis (RRMS). In addition to immunosuppression, neuronal protection by FTY720 has also been suggested, but remains controversial. Axial and radial diffusivities derived from in vivo diffusion tensor imaging (DTI) were employed as noninvasive biomarkers of axonal injury and demyelination to assess axonal protection by FTY720 in experimental autoimmune encephalomyelitis (EAE) mice. EAE was induced through active immunization of C57BL/6 mice using myelin oligodendrocyte glycoprotein peptide 35-55 (MOG(35-55)). We evaluated both the prophylactic and therapeutic treatment effect of FTY720 at doses of 3 and 10 mg/kg on EAE mice by daily clinical scoring and end-point in vivo DTI. Prophylactic administration of FTY720 suppressed the disease onset and prevented axon and myelin damage when compared with EAE mice without treatment. Therapeutic treatment by FTY720 did not prevent EAE onset, but reduced disease severity, improving axial and radial diffusivity towards the control values without statistical significance. Consistent with previous findings, in vivo DTI-derived axial and radial diffusivity correlated with clinical scores in EAE mice. The results support the use of in vivo DTI as an effective outcome measure for preclinical drug development.


Subject(s)
Diffusion Tensor Imaging , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Propylene Glycols/therapeutic use , Sphingosine/analogs & derivatives , Animals , Anisotropy , Disease Progression , Female , Fingolimod Hydrochloride , Mice , Mice, Inbred C57BL , Myelin Basic Protein/metabolism , Sphingosine/therapeutic use , Spine/pathology
9.
J Immunol ; 186(9): 5151-61, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21441446

ABSTRACT

Although the cellular immune response is essential for controlling SIV replication in Asian macaques, its role in maintaining nonpathogenic SIV infection in natural hosts such as sooty mangabeys (SM) remains to be defined. We have previously shown that similar to rhesus macaques (RM), SM are able to mount a T lymphocyte response against SIV infection. To investigate early control of SIV replication in natural hosts, we performed a detailed characterization of SIV-specific cellular immunity and viral control in the first 6 mo following SIV infection in SM. Detection of the initial SIV-specific IFN-γ ELISPOT response in SIVsmE041-infected SM coincided temporally with a decline in peak plasma viremia and was similar in magnitude, specificity, and breadth to SIVsmE041-infected and SIVmac239-infected RM. Despite these similarities, SM showed a greater reduction in postpeak plasma viremia and a more rapid disappearance of productively SIV-infected cells from the lymph node compared with SIVmac239-infected RM. The early Gag-specific CD8(+) T lymphocyte response was significantly more polyfunctional in SM compared with RM, and granzyme B-positive CD8(+) T lymphocytes were present at significantly higher frequencies in SM even prior to SIV infection. These findings suggest that the early SIV-specific T cell response may be an important determinant of lymphoid tissue viral clearance and absence of lymph node immunopathology in natural hosts of SIV infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymph Nodes/immunology , Lymph Nodes/virology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Animals , Cell Separation , Cercocebus atys , Flow Cytometry , In Situ Hybridization , RNA, Viral/blood , Simian Acquired Immunodeficiency Syndrome/blood , Simian Acquired Immunodeficiency Syndrome/virology , Viremia/blood
10.
Am J Pathol ; 177(2): 777-91, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20595631

ABSTRACT

Antiretroviral drugs suppress virus burden in the cerebrospinal fluid of HIV-infected individuals; however, the direct effect of antiretrovirals on virus replication in brain parenchyma is poorly understood. We investigated the effect of short-term combined antiretroviral therapy (CART) on brain virus burden in rhesus monkeys using the CD8-depletion model of accelerated simian immunodeficiency virus (SIV) encephalitis. Four monkeys received CART (consisting of the nonpenetrating agents PMPA and RCV) for four weeks, beginning 28 days after SIV inoculation. Lower virus burdens were measured by real-time RT-PCR in four of four regions of brain from monkeys that received CART as compared with four SIV-infected, untreated controls; however, the difference was only significant for the frontal cortex (P < 0.05). In contrast, significantly lower virus burdens were measured in plasma and four of five lymphoid compartments from animals that received CART. Surprisingly, despite normalization of neuronal function in treated animals, the numbers of activated macrophages/microglia and the magnitude of TNF-alpha mRNA expression in brain were similar between treated animals and controls. These results suggest that short-term therapy with antiretrovirals that fail to penetrate the blood-cerebrospinal fluid barrier can reduce brain virus burden provided systemic virus burden is suppressed; however, longer treatment may be required to completely resolve encephalitic lesions and microglial activation, which may reflect the longer half-life of the principal target cells of HIV/SIV in the brain (macrophages) versus lymphoid tissues (T lymphocytes).


Subject(s)
Anti-Retroviral Agents/pharmacology , Brain , CD8-Positive T-Lymphocytes/virology , Macaca mulatta/virology , Simian Immunodeficiency Virus/drug effects , Viral Load , Animals , Anti-Retroviral Agents/therapeutic use , Brain/drug effects , Brain/metabolism , Brain/virology , CD8-Positive T-Lymphocytes/immunology , Gene Products, gag/genetics , Gene Products, gag/metabolism , Humans , Lymphoid Tissue/metabolism , Lymphoid Tissue/pathology , Lymphoid Tissue/virology , Macrophages/metabolism , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Virus Replication/drug effects
11.
J Infect Dis ; 201(8): 1155-63, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20214475

ABSTRACT

BACKGROUND: Worldwide, approximately 90% of all human immunodeficiency virus (HIV) transmissions occur mucosally; almost all involve R5 strains. Risks of sexual HIV acquisition are highest for rectal, then vaginal, and finally oral exposures. METHODS: Mucosal lacerations may affect the rank order of susceptibility to HIV but cannot be assessed in humans. We measured relative virus transmissibility across intact mucosae in macaques using a single stock of SHIV-1157ipd3N4, a simian-human immunodeficiency virus encoding a primary R5 HIV clade C env (SHIV-C). RESULTS: The penetrability of rhesus macaque mucosae differed significantly, with rectal challenge requiring the least virus, followed by vaginal and then oral routes (P = .031, oral vs vaginal; P < .001 rectal vs vaginal). These findings imply that intrinsic mucosal properties are responsible for the differential mucosal permeability. The latter paralleled the rank order reported for humans, with relative risk estimates within the range of epidemiological human studies. To test whether inflammation facilitates virus transmission--as predicted from human studies--we established a macaque model of localized buccal inflammation. Systemic infection occurred across inflamed but not normal buccal mucosa. CONCLUSION: Our primate data recapitulate virus transmission risks observed in humans, thus establishing R5 SHIV-1157ipd3N4 in macaques as a robust model system to study cofactors involved in human mucosal HIV transmission and its prevention.


Subject(s)
HIV-1/pathogenicity , Mucous Membrane/virology , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/pathogenicity , Animals , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes , Disease Progression , Disease Susceptibility/virology , Female , HIV Infections/transmission , Humans , Inflammation/virology , Intestinal Mucosa/virology , Macaca mulatta/virology , Mouth Mucosa/virology , Rectum/virology , Vagina/virology , Viral Load
12.
PLoS Pathog ; 4(2): e1000003, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18389077

ABSTRACT

The TRIM family proteins share a conserved arrangement of three adjacent domains, an N-terminal RING domain, followed by one or two B-boxes and a coiled-coil, which constitutes the tripartite-motif for which the family is named. However, the C-termini of TRIM proteins vary, and include at least nine evolutionarily distinct, unrelated protein domains. Antiviral restriction factor TRIM5alpha has a C-terminal B30.2/SPRY domain, which is the major determinant of viral target specificity. Here, we describe the evolution of a cyclophilin-A encoding exon downstream of the TRIM5 locus of Asian macaques. Alternative splicing gives rise to chimeric transcripts encoding the TRIM motif fused to a C-terminal CypA domain (TRIM5-CypA). We detected TRIM5-CypA chimeric transcripts in primary lymphocytes from two macaque species. These were derived in part from a CypA pseudogene in the TRIM5 locus, which is distinct from the previously described CypA insertion in TRIM5 of owl monkeys. The CypA insertion is linked to a mutation in the 3' splice site upstream of exon 7, which may prevent or reduce expression of the alpha-isoform. All pig-tailed macaques (M. nemestrina) screened were homozygous for the CypA insertion. In contrast, the CypA-containing allele was present in 17% (17/101) of rhesus macaques (M. mulatta). The block to HIV-1 infection in lymphocytes from animals bearing the TRIM5-CypA allele was weaker than that in cells from wild type animals. HIV-1 infectivity remained significantly lower than SIV infectivity, but was not rescued by treatment with cyclosporine A. Thus, unlike owl monkey TRIMCyp, expression of the macaque TRIM5-CypA isoform does not result in increased restriction of HIV-1. Despite its distinct evolutionary origin, Macaca TRIM5-CypA has a similar domain arrangement and shares approximately 80% amino-acid identity with the TRIMCyp protein of owl monkeys. The independent appearance of TRIM5-CypA chimeras in two primate lineages constitutes a remarkable example of convergent evolution. Based on the presence of the CypA insertion in separate macaque lineages, and its absence from sooty mangabeys, we estimate that the Macaca TRIM5-CypA variant appeared 5-10 million years ago in a common ancestor of the Asian macaques. Whether the formation of novel genes through alternative splicing has played a wider role in the evolution of the TRIM family remains to be investigated.


Subject(s)
Cyclophilin A/genetics , Evolution, Molecular , HIV Infections/genetics , Monkey Diseases/genetics , Proteins/genetics , Amino Acid Sequence , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cercopithecidae , Cyclophilin A/metabolism , Genotype , HIV Infections/immunology , HIV Infections/metabolism , HIV-1 , Homozygote , Immunity, Innate , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Molecular Sequence Data , Monkey Diseases/immunology , Monkey Diseases/metabolism , Mutant Chimeric Proteins/genetics , Phytohemagglutinins/pharmacology , Polymorphism, Single Nucleotide , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Proteins/metabolism , Ubiquitin-Protein Ligases
13.
Cytokine ; 51(3): 232-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20663688

ABSTRACT

IL-18 is a proinflammatory cytokine that is important for host defense, but is also involved in the pathogenesis of a number of disease processes, ranging from autoimmune disorders to atherosclerosis. IL-18 binding protein (IL-18BP) is a constitutively expressed glycoprotein that specifically neutralizes the effects of IL-18, resulting in decreased production of IFN-gamma and reduction in Th1 immune responses. In this study we cloned and sequenced a full-length cDNA of the rhesus IL-18BP (RhIL-18BP) from the spleen of rhesus macaques (Macaca mulatta) and compared its nucleotide and amino acid sequences to the functional murine and human IL-18BP orthologues. In addition, we fused RhIL-18BP to the Fc portion of human IgG1 to make recombinant RhIL-18BP x Fcgamma1 in order to facilitate its detection by Western blot analysis and determined the approximate molecular weight of RhIL-18BP x Fcgamma1 to be 66 kD. With this fusion protein, we showed that RhIL-18BP was functional and could significantly reduce murine IL-18 and LPS-induced IFN-gamma production by murine splenocytes. Furthermore, we demonstrated the expression of IL-18BP in atherosclerotic lesions in a rhesus model of atherosclerosis, underscoring the need to fully understand the role of this protein as a primary negative regulator of IL-18 in multiple disease processes.


Subject(s)
Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-18/antagonists & inhibitors , Macaca mulatta/genetics , Amino Acid Sequence , Animals , Atherosclerosis/metabolism , Base Sequence , Cloning, Molecular , Humans , Immunoglobulin G/metabolism , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/chemistry , Interferon-gamma/biosynthesis , Interleukin-18/pharmacology , Lipopolysaccharides/pharmacology , Mice , Molecular Sequence Data , Molecular Weight , Protein Transport/drug effects , Recombinant Fusion Proteins/metabolism , Sequence Analysis, Protein , Spleen/cytology , Spleen/drug effects , Spleen/metabolism
14.
Toxicol Pathol ; 38(4): 606-18, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20448081

ABSTRACT

Exposure to moderately selective p38alpha mitogen-activated protein kinase (MAPK) inhibitors in the Beagle dog results in an acute toxicity consisting of mild clinical signs (decreased activity, diarrhea, and fever), lymphoid necrosis and depletion in the gut-associated lymphoid tissue (GALT), mesenteric lymph nodes and spleen, and linear colonic and cecal mucosal hemorrhages. Lymphocyte apoptosis and necrosis in the GALT is the earliest and most prominent histopathologic change observed, followed temporally by neutrophilic infiltration and acute inflammation of the lymph nodes and spleen and multifocal mucosal epithelial necrosis and linear hemorrhages in the colon and cecum. These effects are not observed in the mouse, rat, or cynomolgus monkey. To further characterize the acute toxicity in the dog, a series of in vivo, in vitro, and immunohistochemical studies were conducted to determine the relationship between the lymphoid and gastrointestinal (GI) toxicity and p38 MAPK inhibition. Results of these studies demonstrate a direct correlation between p38alpha MAPK inhibition and the acute lymphoid and gastrointestinal toxicity in the dog. Similar effects were observed following exposure to inhibitors of MAPK-activated protein kinase-2 (MK2), further implicating the role of p38alpha MAPK signaling pathway inhibition in these effects. Based on these findings, the authors conclude that p38alpha MAPK inhibition results in acute lymphoid and GI toxicity in the dog and is unique among the species evaluated in these studies.


Subject(s)
Gastrointestinal Diseases/chemically induced , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Lymphatic Diseases/chemically induced , Protein Kinase Inhibitors/toxicity , Protein Serine-Threonine Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , B-Lymphocytes/metabolism , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Colon/drug effects , Colon/pathology , Dogs , Female , Gastrointestinal Diseases/pathology , Gastrointestinal Hemorrhage/chemically induced , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/metabolism , Linear Models , Lymph Nodes/drug effects , Lymph Nodes/pathology , Lymphatic Diseases/pathology , Macaca fascicularis , Male , Mice , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Spleen/cytology , Spleen/metabolism , T-Lymphocytes/metabolism , Toxicity Tests, Acute , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Retrovirology ; 5: 94, 2008 Oct 17.
Article in English | MEDLINE | ID: mdl-18928523

ABSTRACT

BACKGROUND: Infection of nonhuman primates with simian immunodeficiency virus (SIV) or chimeric simian-human immunodeficiency virus (SHIV) strains is widely used to study lentiviral pathogenesis, antiviral immunity and the efficacy of AIDS vaccine candidates. SHIV challenges allow assessment of anti-HIV-1 envelope responses in primates. As such, SHIVs should mimic natural HIV-1 infection in humans and, to address the pandemic, encode HIV-1 Env components representing major viral subtypes worldwide. RESULTS: We have developed a panel of clade C R5-tropic SHIVs based upon env of a Zambian pediatric isolate of HIV-1 clade C, the world's most prevalent HIV-1 subtype. The parental infectious proviral clone, SHIV-1157i, was rapidly passaged through five rhesus monkeys. After AIDS developed in the first animal at week 123 post-inoculation, infected blood was infused into a sixth monkey. Virus reisolated at this late stage was still exclusively R5 tropic and mucosally transmissible. Here we describe the long-term follow-up of this initial cohort of six monkeys. Two have remained non-progressors, whereas the other four gradually progressed to AIDS within 123-270 weeks post-exposure. Two progressors succumbed to opportunistic infections, including a case of SV40 encephalitis. CONCLUSION: These data document the disease progression induced by the first mucosally transmissible, pathogenic R5 non-clade B SHIV and suggest that SHIV-1157i-derived viruses, including the late-stage, highly replication-competent SHIV-1157ipd3N4 previously described (Song et al., 2006), display biological characteristics that mirror those of HIV-1 clade C and support their expanded use for AIDS vaccine studies in nonhuman primates.


Subject(s)
Acquired Immunodeficiency Syndrome/transmission , HIV-1/pathogenicity , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/pathogenicity , Viral Envelope Proteins/genetics , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/pathology , Acquired Immunodeficiency Syndrome/virology , Amino Acid Sequence , Animals , Cell Line , HIV-1/genetics , HIV-1/physiology , Humans , Macaca mulatta , Molecular Sequence Data , Sequence Alignment , Serial Passage , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/physiology , Viral Envelope Proteins/metabolism , Viral Load , Virus Replication
16.
Dev Comp Immunol ; 32(12): 1510-21, 2008.
Article in English | MEDLINE | ID: mdl-18606180

ABSTRACT

Studies were undertaken to determine whether previously described reductions in splenic DC-SIGN expression in simian acquired immune deficiency syndrome (AIDS) are limited to pathogenic simian immunodeficiency virus (SIV) infection. DC-SIGN expression was evaluated by immunohistochemistry in lymphoid tissues from AIDS-susceptible Asian macaque monkeys as compared with AIDS-resistant sooty mangabey monkeys in the presence and absence of SIV infection. The phenotype of DC-SIGN+ cells in susceptible and resistant species was identical and most consistent with macrophage identity. Significantly lower levels of DC-SIGN expression were identified in spleen, mesenteric lymph node, and bone marrow of macaques with AIDS (P<0.05). Reduced levels of splenic DC-SIGN correlated significantly with CD4T cell depletion in long-term pathogenic infection of macaques (P<0.01), whereas SIV-infected mangabeys retained high levels of DC-SIGN expression in spleen despite persistent infection. Reduced expression of DC-SIGN in spleen specifically characterizes pathogenic forms of SIV infection, correlates with disease progression, and may contribute to SIV pathogenesis.


Subject(s)
Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/biosynthesis , Down-Regulation/immunology , Lectins, C-Type/antagonists & inhibitors , Lectins, C-Type/biosynthesis , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/biosynthesis , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/immunology , Animals , Cell Adhesion Molecules/genetics , Cercocebus atys , Disease Progression , Lectins, C-Type/genetics , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Lymphoid Tissue/virology , Macaca mulatta , Macaca nemestrina , Mesentery , Organ Specificity/immunology , Receptors, Cell Surface/genetics , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Immunodeficiency Virus/pathogenicity
17.
Curr Protoc Toxicol ; 76(1): e51, 2018 05.
Article in English | MEDLINE | ID: mdl-30040239

ABSTRACT

Cellular development and homeostasis are regulated via programmed cell death (PCD; apoptosis), which is a genetically regulated cellular process. Accidental cell death (ACD; necrosis) can be triggered by chemical, physical, or mechanical stress. Necrosis is the presence of dead tissues or cells in a living organism regardless of the initiating process and can be observed in infectious and non-infectious diseases and toxicities. This article describes tissue-based immunohistotechnical protocols used for assessing PCD and necrosis in formalin-fixed tissues obtained from preclinical species used in investigative and toxicologic pathology. Two commonly employed protocols for the identification of PCD and necrosis are described in this article: immunohistochemistry (IHC) for cleaved caspase 3, and terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling (TUNEL). TUNEL has been used to detect DNA fragmentation by labeling the terminal ends of nucleic acids in necrotic and apoptotic cells. © 2018 by John Wiley & Sons, Inc.


Subject(s)
Apoptosis , Cell Death , Necrosis/pathology , Pathology/methods , Toxicology/methods , Humans , Immunohistochemistry/methods , Necrosis/chemically induced
18.
Bioanalysis ; 10(18): 1487-1500, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30198746

ABSTRACT

AIM: Tools for mapping and quantifying monoclonal antibody (mAb) and peptide biotherapeutics distribumtion were evaluated by comparing data from three independent methods conducted at the whole body, organ or tissue, and cellular levels. MATERIALS & METHODS: [3H]-mAb1 and [3H]-peptide A were administered intravenously to rats followed by quantitative whole-body autoradiography, kidney macro-autoradiography and micro-autoradiography. RESULTS: [3H]-mAb1 and [3H]-peptide A concentrations were measured in anatomical regions ranging from whole body to whole organ to sub-organ level, such as the kidney glomerulus, with increasing resolution. The tissue/blood [3H]-mAb1 concentrations in selected kidney microenvironments were comparable among the three quantitative methods. CONCLUSION: Quantitative whole-body autoradiography, tissue macro-autoradiography and micro-autoradiography all provide useful tools for quantifying the concentrations of biotherapeutics at different anatomical levels in tissues, facilitating better predictions of efficacy and toxicity.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Autoradiography , Kidney/metabolism , Oncogene Protein pp60(v-src)/pharmacokinetics , Peptide Fragments/pharmacokinetics , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Male , Oncogene Protein pp60(v-src)/metabolism , Oncogene Protein pp60(v-src)/therapeutic use , Peptide Fragments/metabolism , Peptide Fragments/therapeutic use , Rats , Rats, Sprague-Dawley , Tissue Distribution
19.
J Clin Invest ; 111(10): 1505-18, 2003 May.
Article in English | MEDLINE | ID: mdl-12750400

ABSTRACT

Systemic lupus erythematosus (SLE) is a CD4(+) T cell-dependent, immune complex-mediated, autoimmune disease that primarily affects women of childbearing age. Generation of high-titer affinity-matured IgG autoantibodies, specific for double-stranded DNA and other nuclear antigens, coincides with disease progression. Current forms of treatment of SLE including glucocorticosteroids are often inadequate and induce severe side effects. Immunological approaches for treating SLE in mice using anti-CD4 mAb's or CTLA4-Ig and anti-CD154 mAb's have proven to be effective. However, like steroid treatment, these regimens induce global immunosuppression, and their withdrawal allows for disease progression. In this report we show that lupus-prone NZB x NZW F(1) mice given three injections of anti-CD137 (4-1BB) mAb's between 26 and 35 weeks of age reversed acute disease, blocked chronic disease, and extended the mice's lifespan from 10 months to more than 2 years. Autoantibody production in recipients was rapidly suppressed without inducing immunosuppression. Successful treatment could be traced to the fact that NZB x NZW F(1) mice, regardless of their age or disease status, could not maintain pathogenic IgG autoantibody production in the absence of continuous CD4(+) T cell help. Our data support the hypothesis that CD137-mediated signaling anergized CD4(+) T cells during priming at the DC interface.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Receptors, Nerve Growth Factor/antagonists & inhibitors , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Acute Disease , Adoptive Transfer , Animals , Antibodies, Antinuclear/blood , Antibody Formation/drug effects , Antigens, CD , Autoantibodies/blood , Autoantibodies/drug effects , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Chronic Disease , Crosses, Genetic , Dendritic Cells/immunology , Dendritic Cells/transplantation , Disease Models, Animal , Immunoglobulin G/blood , Immunoglobulin M/blood , Interleukin-2/biosynthesis , Interleukin-4/biosynthesis , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred NZB , Mice, Inbred Strains , Proteinuria/prevention & control , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/immunology , Treatment Outcome , Tumor Necrosis Factor Receptor Superfamily, Member 9
20.
Int J Mol Imaging ; 2016: 5768312, 2016.
Article in English | MEDLINE | ID: mdl-28050284

ABSTRACT

Human plasma-derived α1-antitrypsin (AAT) delivered by intravenous infusion is used as augmentation therapy in patients with emphysema who have a genetic mutation resulting in deficiency of AAT. Inhalation is an alternative route of administration that can potentially increase the efficacy and convenience of treatment. This study was conducted to determine whether delivery to the lungs, initially via the intratracheal (IT) route of administration, would deliver efficacious levels of a recombinant AAT (rAAT) to the site of action in the lungs in mice. 125I-radiolabeled rAAT, fluorophore-conjugated rAAT (rAAT-Alexa488), and NE680 (neutrophil elastase 680, a silent fluorescent substrate of neutrophil elastase which fluoresces in the near-infrared range upon activation by neutrophil elastase) were used to characterize the pharmacokinetics and tissue distribution profile, distribution of rAAT within the lung, and efficacy of rAAT to inhibit neutrophil elastase at the site of action, respectively. The study has demonstrated that rAAT was able to gain access to locations where neutrophil elastase was localized. The histochemical quantification of rAAT activity relative to dose at the site of action provided here will improve confidence in predicting the human dose via the inhalation route.

SELECTION OF CITATIONS
SEARCH DETAIL