Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
2.
PLoS One ; 18(10): e0293218, 2023.
Article in English | MEDLINE | ID: mdl-37883475

ABSTRACT

The Lamc2jeb junctional epidermolysis bullosa (EB) mouse model has been used to demonstrate that significant genetic modification of EB symptoms is possible, identifying as modifiers Col17a1 and six other quantitative trait loci, several with strong candidate genes including dystonin (Dst/Bpag1). Here, CRISPR/Cas9 was used to alter exon 23 in mouse skin specific isoform Dst-e (Ensembl GRCm38 transcript name Dst-213, transcript ID ENSMUST00000183302.5, protein size 2639AA) and validate a proposed arginine/glutamine difference at amino acid p1226 in B6 versus 129 mice as a modifier of EB. Frame shift deletions (FSD) in mouse Dst-e exon 23 (Dst-eFSD/FSD) were also identified that cause mice carrying wild-type Lamc2 to develop a phenotype similar to human EB simplex without dystonia musculorum. When combined, Dst-eFSD/FSD modifies Lamc2jeb/jeb (FSD+jeb) induced disease in unexpected ways implicating an altered balance between DST-e (BPAG1e) and a rarely reported rodless DST-eS (BPAG1eS) in epithelium as a possible mechanism. Further, FSD+jeb mice with pinnae removed are found to provide a test bed for studying internal epithelium EB disease and treatment without severe skin disease as a limiting factor while also revealing and accelerating significant nasopharynx symptoms present but not previously noted in Lamc2jeb/jeb mice.


Subject(s)
Dystonia , Dystonic Disorders , Epidermolysis Bullosa Simplex , Epidermolysis Bullosa, Junctional , Epidermolysis Bullosa , Animals , Mice , Dystonia/genetics , Dystonia/metabolism , Dystonic Disorders/metabolism , Dystonin/metabolism , Epidermolysis Bullosa/genetics , Epidermolysis Bullosa Simplex/diagnosis , Epidermolysis Bullosa Simplex/genetics , Epidermolysis Bullosa Simplex/metabolism , Epidermolysis Bullosa, Junctional/genetics , Epidermolysis Bullosa, Junctional/diagnosis , Epidermolysis Bullosa, Junctional/metabolism , Skin/metabolism
3.
PLoS One ; 18(10): e0292456, 2023.
Article in English | MEDLINE | ID: mdl-37796769

ABSTRACT

Previous work strongly implicated Collagen 17a1 (Col17a1) as a potent genetic modifier of junctional epidermolysis bullosa (JEB) caused by a hypomorphic mutation (Lamc2jeb) in mice. The importance of the noncollagenous domain (NC4) of COLXVII was suggested by use of a congenic reduction approach that restricted the modifier effect to 2-3 neighboring amino acid changes in that domain. The current study utilizes TALEN and CRISPR/Cas9 induced amino acid replacements and in-frame indels nested to NC4 to further investigate the role of this and adjoining COLXVII domains both as modifiers and primary risk effectors. We confirm the importance of COLXVI AA 1275 S/G and 1277 N/S substitutions and utilize small nested indels to show that subtle changes in this microdomain attenuate JEB. We further show that large in-frame indels removing up to 1482 bp and 169 AA of NC6 through NC1 domains are surprisingly disease free on their own but can be very potent modifiers of Lamc2jeb/jeb JEB. Together these studies exploiting gene editing to functionally dissect the Col17a1 modifier demonstrate the importance of epistatic interactions between a primary disease-causing mutation in one gene and innocuous 'healthy' alleles in other genes.


Subject(s)
Epidermolysis Bullosa, Junctional , Animals , Mice , Epidermolysis Bullosa, Junctional/genetics , Non-Fibrillar Collagens/genetics , Non-Fibrillar Collagens/metabolism , Collagen/genetics , Mutation , Amino Acids/genetics
4.
PLoS One ; 18(7): e0288263, 2023.
Article in English | MEDLINE | ID: mdl-37437067

ABSTRACT

Epidermolysis Bullosa (EB) is a group of rare genetic disorders that compromise the structural integrity of the skin such that blisters and subsequent erosions occur after minor trauma. While primary genetic risk of all subforms of EB adhere to Mendelian patterns of inheritance, their clinical presentations and severities can vary greatly, implying genetic modifiers. The Lamc2jeb mouse model of non-Herlitz junctional EB (JEB-nH) demonstrated that genetic modifiers can contribute substantially to the phenotypic variability of JEB and likely other forms of EB. The innocuous changes in an 'EB related gene', Col17a1, have shown it to be a dominant modifier of Lamc2jeb. This work identifies six additional Quantitative Trait Loci (QTL) that modify disease in Lamc2jeb/jeb mice. Three QTL include other known 'EB related genes', with the strongest modifier effect mapping to a region including the epidermal hemi-desmosomal structural gene dystonin (Dst-e/Bpag1-e). Three other QTL map to intervals devoid of known EB-associated genes. Of these, one contains the nuclear receptor coactivator Ppargc1a as its primary candidate and the others contain related genes Pparg and Igf1, suggesting modifier pathways. These results, demonstrating the potent disease modifying effects of normally innocuous genetic variants, greatly expand the landscape of genetic modifiers of EB and therapeutic approaches that may be applied.


Subject(s)
Epidermolysis Bullosa, Junctional , Animals , Mice , Skin , Blister , Epidermis , Quantitative Trait Loci/genetics
5.
Dis Model Mech ; 13(5)2020 05 21.
Article in English | MEDLINE | ID: mdl-32482619

ABSTRACT

Loss-of-function mutations in dystonin (DST) can cause hereditary sensory and autonomic neuropathy type 6 (HSAN-VI) or epidermolysis bullosa simplex (EBS). Recently, DST-related diseases were recognized to be more complex than previously thought because a patient exhibited both neurological and skin manifestations, whereas others display only one or the other. A single DST locus produces at least three major DST isoforms: DST-a (neuronal isoform), DST-b (muscular isoform) and DST-e (epithelial isoform). Dystonia musculorum (dt) mice, which have mutations in Dst, were originally identified as spontaneous mutants displaying neurological phenotypes. To reveal the mechanisms underlying the phenotypic heterogeneity of DST-related diseases, we investigated two mutant strains with different mutations: a spontaneous Dst mutant (Dstdt-23Rbrc mice) and a gene-trap mutant (DstGt mice). The Dstdt-23Rbrc allele possesses a nonsense mutation in an exon shared by all Dst isoforms. The DstGt allele is predicted to inactivate Dst-a and Dst-b isoforms but not Dst-e There was a decrease in the levels of Dst-a mRNA in the neural tissue of both Dstdt-23Rbrc and DstGt homozygotes. Loss of sensory and autonomic nerve ends in the skin was observed in both Dstdt-23Rbrc and DstGt mice at postnatal stages. In contrast, Dst-e mRNA expression was reduced in the skin of Dstdt-23Rbrc mice but not in DstGt mice. Expression levels of Dst proteins in neural and cutaneous tissues correlated with Dst mRNAs. Because Dst-e encodes a structural protein in hemidesmosomes (HDs), we performed transmission electron microscopy. Lack of inner plaques and loss of keratin filament invasions underneath the HDs were observed in the basal keratinocytes of Dstdt-23Rbrc mice but not in those of DstGt mice; thus, the distinct phenotype of the skin of Dstdt-23Rbrc mice could be because of failure of Dst-e expression. These results indicate that distinct mutations within the Dst locus can cause different loss-of-function patterns among Dst isoforms, which accounts for the heterogeneous neural and skin phenotypes in dt mice and DST-related diseases.


Subject(s)
Dystonic Disorders/genetics , Dystonin/genetics , Mutation/genetics , Protein Isoforms/genetics , Animals , Desmosomes/metabolism , Desmosomes/ultrastructure , Disease Models, Animal , Dystonin/metabolism , Gene Expression Regulation , Homozygote , Mice , Neurons/pathology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Skin/pathology
6.
J Neurosci ; 40(23): 4576-4585, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32341096

ABSTRACT

An impediment to the development of effective therapies for neurodegenerative disease is that available animal models do not reproduce important clinical features such as adult-onset and stereotypical patterns of progression. Using in vivo magnetic resonance imaging and behavioral testing to study male and female decrepit mice, we found a stereotypical neuroanatomical pattern of progression of the lesion along the limbic system network and an associated memory impairment. Using structural variant analysis, we identified an intronic mutation in a mitochondrial-associated gene (Mrpl3) that is responsible for the decrepit phenotype. While the function of this gene is unknown, embryonic lethality in Mrpl3 knock-out mice suggests it is critical for early development. The observation that a mutation linked to energy metabolism precipitates a pattern of neurodegeneration via cell death across disparate but linked brain regions may explain how stereotyped patterns of neurodegeneration arise in humans or define a not yet identified human disease.SIGNIFICANCE STATEMENT The development of novel therapies for adult-onset neurodegenerative disease has been impeded by the limitations of available animal models in reproducing many of the clinical features. Here, we present a novel spontaneous mutation in a mitochondrial-associated gene in a mouse (termed decrepit) that results in adult-onset neurodegeneration with a stereotypical neuroanatomical pattern of progression and an associated memory impairment. The decrepit mouse model may represent a heretofore undiagnosed human disease and could serve as a new animal model to study neurodegenerative disease.


Subject(s)
Genetic Variation/genetics , Memory Disorders/diagnostic imaging , Memory Disorders/genetics , Mitochondrial Proteins/genetics , Neurodegenerative Diseases/diagnostic imaging , Neurodegenerative Diseases/genetics , Ribosomal Proteins/genetics , Age Factors , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
7.
J Immunol ; 204(5): 1091-1100, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31988182

ABSTRACT

Almost a decade has passed since the approval of belimumab, an mAb directed against B lymphocyte stimulation and the first targeted therapy approved for systemic lupus erythematous (SLE) in over 50 y. Although well tolerated, the efficacy of belimumab remains limited and is not labeled for patients suffering from nephritis, the leading cause of patient mortality. We sought to explore alternative targets of autoreactive B lymphocytes through manipulation of affinity maturation. The BXSB/MpJ mouse, a well-established model of human SLE, develops elevated antinuclear Abs and immune complex-mediated nephritis along with other manifestations of SLE-like disease. To limit interfering with critical background genetics, we used CRISPR-Cas9 to disrupt activation-induced cytidine deaminase (AID; Aicda) directly in BXSB zygotes. Homozygous null mice demonstrated significantly prolonged survival compared with wild-type. Although mice continued to develop plasma cells, splenic follicular structure was restored, and renal pathology was reduced. Mice developed expanded germinal center B lymphocyte populations as in other models of AID deficiency as well as increased populations of CD73+ B lymphocytes. Treatment with the small molecule inhibitor of RAD51, 4,4'-diisothiocyano-2,2'-stilbenedisulfonic acid, resulted in minimal changes in disease markers in BXSB mice. The prolonged survival in AID-deficient BXSB mice appears attributed primarily to the reduced renal pathology, warranting further exploration, as current therapeutics targeting lupus nephritis are limited and, thus, in great demand.


Subject(s)
B-Lymphocyte Subsets/immunology , Cytidine Deaminase/immunology , Lupus Erythematosus, Systemic/immunology , Animals , B-Lymphocyte Subsets/pathology , CRISPR-Cas Systems , Cytidine Deaminase/genetics , Disease Models, Animal , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Knockout
8.
Cancer Biol Ther ; 20(2): 169-182, 2019.
Article in English | MEDLINE | ID: mdl-30183475

ABSTRACT

Targeting the early steps of the glycolysis pathway in cancers is a well-established therapeutic strategy; however, the doses required to elicit a therapeutic effect on the cancer can be toxic to the patient. Consequently, numerous preclinical and clinical studies have combined glycolytic blockade with other therapies. However, most of these other therapies do not specifically target cancer cells, and thus adversely affect normal tissue. Here we first show that a diverse number of cancer models - spontaneous, patient-derived xenografted tumor samples, and xenografted human cancer cells - can be efficiently targeted by 2-deoxy-D-Glucose (2DG), a well-known glycolytic inhibitor. Next, we tested the cancer-cell specificity of a therapeutic compound using the MEC1 cell line, a chronic lymphocytic leukemia (CLL) cell line that expresses activation induced cytidine deaminase (AID). We show that MEC1 cells, are susceptible to 4,4'-Diisothiocyano-2,2'-stilbenedisulfonic acid (DIDS), a specific RAD51 inhibitor. We then combine 2DG and DIDS, each at a lower dose and demonstrate that this combination is more efficacious than fludarabine, the current standard- of- care treatment for CLL. This suggests that the therapeutic blockade of glycolysis together with the therapeutic inhibition of RAD51-dependent homologous recombination can be a potentially beneficial combination for targeting AID positive cancer cells with minimal adverse effects on normal tissue. Implications: Combination therapy targeting glycolysis and specific RAD51 function shows increased efficacy as compared to standard of care treatments in leukemias.


Subject(s)
4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxyglucose/pharmacology , Neoplasms/drug therapy , Rad51 Recombinase/antagonists & inhibitors , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/administration & dosage , Animals , Cell Line, Tumor , Deoxyglucose/administration & dosage , Drug Synergism , Female , Glycolysis/drug effects , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Neoplasms/metabolism , Rad51 Recombinase/metabolism , Xenograft Model Antitumor Assays
9.
Cell Rep ; 21(1): 208-221, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28978474

ABSTRACT

Interleukin 21 (IL-21) plays key roles in humoral immunity and autoimmune diseases. It is known to function in mature CD4+ T follicular B cell helper (TFH) cells, but its potential involvement in early T cell ontogeny is unclear. Here, we find that a significant population of newly activated thymic and peripheral CD4+ T cells functionally expresses IL-21 soon after birth. This naturally occurring population, termed natural (n)TH21 cells, exhibits considerable similarity to mature TFH cells. nTH21 cells originating and activated in the thymus are strictly dependent on autoimmune regulator (AIRE) and express high levels of NUR77, consistent with a bias toward self-reactivity. Their activation/expansion in the periphery requires gut microbiota and is held in check by FoxP3+ TREG cells. nTH21 cells are the major thymic and peripheral populations of IL-21+ cells to expand in an IL-21-dependent humoral autoimmune disease. These studies link IL-21 to T cell ontogeny, self-reactivity, and humoral autoimmunity.


Subject(s)
Arthritis/genetics , Autoimmunity/genetics , Interleukins/genetics , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Arthritis/immunology , Arthritis/pathology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gastrointestinal Microbiome/immunology , Gene Expression Regulation , Immunity, Humoral , Interleukins/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/immunology , Signal Transduction , T-Lymphocytes, Helper-Inducer/pathology , T-Lymphocytes, Regulatory/pathology , Transcription Factors/genetics , Transcription Factors/immunology , AIRE Protein
10.
Methods Mol Biol ; 1438: 103-14, 2016.
Article in English | MEDLINE | ID: mdl-27150086

ABSTRACT

Therapeutic monoclonal antibodies are widely recognized to be a most promising means to treat an increasing number of human diseases, including cancers and autoimmunity. To a large extent, the efficacy of monoclonal antibody treatment is because IgG antibodies have greatly extended persistence in vivo. However, conventional rodent models do not mirror human antibody pharmacokinetics. The key molecule responsible for the extended persistence antibodies is the major histocompatibility complex class I family Fc receptor, FcRn. We describe human FcRn transgenic mouse models and how they can be exploited productively for the preclinical pharmacokinetic evaluation of therapeutic antibodies.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Receptors, Fc/genetics , Receptors, Fc/metabolism , Animals , Drug Evaluation, Preclinical , Humans , Immunoglobulins, Intravenous/pharmacokinetics , Mice , Mice, Transgenic
11.
PLoS One ; 11(4): e0153059, 2016.
Article in English | MEDLINE | ID: mdl-27050763

ABSTRACT

IL6 is a multifunctional cytokine that drives terminal B cell differentiation and secretion of immunoglobulins. IL6 also cooperates with IL21 to promote differentiation of CD4+ T follicular helper cells (TFH). Elevated serum levels of IL6 correlate with disease flares in patients with systemic lupus erythematosus (SLE). We previously reported that IL21 produced by TFH plays a critical role in the development of the SLE-like disease of BXSB.Yaa mice. To examine the possible contributions of IL6 to disease, we compared disease parameters in IL6-deficient and IL6-competent BXSB.Yaa mice. We report that survival of IL6-deficient BXSB.Yaa mice was significantly prolonged in association with significant reductions in a variety of autoimmune manifestations. Moreover, B cells stimulated by co-engagement of TLR7 and B cell receptor (BCR) produced high levels of IL6 that was further augmented by stimulation with Type I interferon (IFN1). Importantly, the frequencies of TFH and serum levels of IL21 were significantly reduced in IL6-deficient mice. These findings suggest that high-level production of IL6 by B cells induced by integrated signaling from the IFN1 receptor, TLR7 and BCR promotes the differentiation of IL21-secreting TFH in a signaling sequence that drives the lethal autoimmune disease of BXSB.Yaa mice.


Subject(s)
Interleukin-6/physiology , Lupus Erythematosus, Systemic/mortality , Animals , B-Lymphocytes/metabolism , Female , Interleukin-6/blood , Interleukin-6/genetics , Lupus Erythematosus, Systemic/physiopathology , Male , Mice , Signal Transduction
12.
Am J Pathol ; 185(11): 3102-14, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26363366

ABSTRACT

SJL/J mice exhibit a high incidence of mature B-cell lymphomas that require CD4(+) T cells for their development. We found that their spleens and lymph nodes contained increased numbers of germinal centers and T follicular helper (TFH) cells. Microarray analyses revealed high levels of transcripts encoding IL-21 associated with high levels of serum IL-21. We developed IL-21 receptor (IL21R)-deficient Swiss Jim Lambart (SJL) mice to determine the role of IL-21 in disease. These mice had reduced numbers of TFH cells, lower serum levels of IL-21, and few germinal center B cells, and they did not develop B-cell tumors, suggesting IL-21-dependent B-cell lymphomagenesis. We also noted a series of features common to SJL disease and human angioimmunoblastic T-cell lymphoma (AITL), a malignancy of TFH cells. Gene expression analyses of AITL showed that essentially all cases expressed elevated levels of transcripts for IL21, IL21R, and a series of genes associated with TFH cell development and function. These results identify a mouse model with features of AITL and suggest that patients with the disease might benefit from therapeutic interventions that interrupt IL-21 signaling.


Subject(s)
Immunoblastic Lymphadenopathy/pathology , Interleukin-21 Receptor alpha Subunit/metabolism , Interleukins/metabolism , Lymphoma, B-Cell/pathology , Lymphoma, T-Cell/pathology , Signal Transduction , Animals , B-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/pathology , Cytokines/blood , Disease Models, Animal , Female , Gene Expression Profiling , Germinal Center/pathology , Humans , Immunoblastic Lymphadenopathy/prevention & control , Immunoglobulin G/blood , Interleukin-21 Receptor alpha Subunit/genetics , Interleukins/genetics , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Sequence Analysis, DNA , Spleen/pathology
13.
Nat Commun ; 6: 7994, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26272364

ABSTRACT

CD8(+) T cells activated without CD4(+) T-cell help are impaired in memory expansion. To understand the underlying cellular mechanism, here we track the dynamics of helper-deficient CD8(+) T-cell response to a minor histocompatibility antigen by phenotypic and in vivo imaging analyses. Helper-deficient CD8(+) T cells show reduced burst expansion, rapid peripheral egress, delayed antigen clearance and continuous activation, and are eventually exhausted. Contrary to the general consensus that CD4 help encodes memory programmes in CD8(+) T cells and helper-deficient CD8(+) T cells are abortive, these cells can differentiate into effectors and memory precursors. Importantly, accelerating antigen clearance or simply increasing the burst effector size enables generation of memory cells by CD8(+) T cells, regardless of CD4 help. These results suggest that the memory programme is CD8(+) T-cell-intrinsic, and provide insight into the role of CD4 help in CD8(+) T-cell responses.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Immunologic Memory/physiology , Adoptive Transfer , Animals , Antibodies , Cell Differentiation , Flow Cytometry , Gene Expression Regulation/physiology , Interferon-gamma/genetics , Interferon-gamma/metabolism , Mice , Mice, Inbred Strains , Microspheres , Minor Histocompatibility Antigens/immunology , RNA/genetics , RNA/metabolism
14.
Eur J Immunol ; 45(8): 2312-23, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25959828

ABSTRACT

Infection with the helminth Schistosoma mansoni results in hepatointestinal granulomatous inflammation mediated by CD4 T cells directed against parasite eggs. The severity of disease varies greatly in humans and mice; however, the genetic basis of such a heterogenous immune response remains poorly understood. Here we show that, despite their close genetic relationship, C57BL/10SnJ (B10) mice developed significantly more pronounced immunopathology and higher T helper 17 cell responses than C57BL/6J (B6) mice. Similarly, live egg-stimulated B10-derived dendritic cells (DCs) produced significantly more IL-1ß and IL-23, resulting in higher IL-17 production by CD4 T cells. Gene expression analysis disclosed a heightened proinflammatory cytokine profile together with a strikingly lower expression of Ym1 in B10 versus B6 mice, consistent with failure of B10 DCs to attain alternative activation. To genetically dissect the differential response, we developed and analyzed congenic mouse strains that capture major regions of allelic variation, and found that the level of inflammation was controlled by a relatively small number of genes in a locus mapping to chromosome 4 117-143 MB. Our study has thus identified novel genomic regions that regulate the severity of the schistosome infection by way of controlling the mode of DC activation and consequent CD4 T-cell subset development.


Subject(s)
Chromosomes, Mammalian , Dendritic Cells/immunology , Genetic Loci/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni , Th17 Cells/immunology , Animals , Chromosomes, Mammalian/genetics , Chromosomes, Mammalian/immunology , Cytokines/genetics , Cytokines/immunology , Female , Gene Expression Regulation/immunology , Lectins/genetics , Lectins/immunology , Mice , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/immunology , Species Specificity , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/immunology
15.
MAbs ; 7(2): 344-51, 2015.
Article in English | MEDLINE | ID: mdl-25654695

ABSTRACT

Serum albumin is the major determinant of blood colloidal osmotic pressure acting as a depot and distributor of compounds including drugs. In humans, serum albumin exhibits an unusually long half-life mainly due to protection from catabolism by neonatal Fc receptor (FcRn)-mediated recycling. These properties make albumin an attractive courier of therapeutically-active compounds. However, pharmaceutical research and development of albumin-based therapeutics has been hampered by the lack of appropriate preclinical animal models. To overcome this, we developed and describe the first mouse with a genetic deficiency in albumin and its incorporation into an existing humanized FcRn mouse model, B6.Cg-Fcgrt(tm1Dcr) Tg(FCGRT)32Dcr/DcrJ (Tg32). Albumin-deficient strains (Alb(-/-)) were created by TALEN-mediated disruption of the albumin (Alb) gene directly in fertilized oocytes derived from Tg32 mice and its non-transgenic background control, C57BL/6J (B6). The resulting Alb(-/-) strains are analbuminemic but healthy. Intravenous administration of human albumin to Tg32-Alb(-/-) mFcRn(-/-) hFcRn(Tg/Tg)) mice results in a remarkably extended human albumin serum half-life of ∼24 days, comparable to that found in humans, and in contrast to half-lives of 2.6-5.8 d observed in B6, B6-Alb(-/-) and Tg32 strains. This striking increase can be explained by the absence of competing endogenous mouse albumin and the presence of an active human FcRn. These novel albumin-deficient models provide unique tools for investigating the biology and pathobiology of serum albumin and are a more appropriate rodent surrogates for evaluating human serum albumin pharmacokinetics and albumin-based compounds.


Subject(s)
Models, Biological , Serum Albumin/pharmacology , Serum Albumin/pharmacokinetics , Animals , Half-Life , Humans , Mice , Mice, Knockout , Serum Albumin/genetics
16.
PLoS Genet ; 10(2): e1004068, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24550734

ABSTRACT

Epidermolysis Bullosa (EB) encompasses a spectrum of mechanobullous disorders caused by rare mutations that result in structural weakening of the skin and mucous membranes. While gene mutated and types of mutations present are broadly predictive of the range of disease to be expected, a remarkable amount of phenotypic variability remains unaccounted for in all but the most deleterious cases. This unexplained variance raises the possibility of genetic modifier effects. We tested this hypothesis using a mouse model that recapitulates a non-Herlitz form of junctional EB (JEB) owing to the hypomorphic jeb allele of laminin gamma 2 (Lamc2). By varying normally asymptomatic background genetics, we document the potent impact of genetic modifiers on the strength of dermal-epidermal adhesion and on the clinical severity of JEB in the context of the Lamc2(jeb) mutation. Through an unbiased genetic approach involving a combination of QTL mapping and positional cloning, we demonstrate that Col17a1 is a strong genetic modifier of the non-Herlitz JEB that develops in Lamc2(jeb) mice. This modifier is defined by variations in 1-3 neighboring amino acids in the non-collagenous 4 domain of the collagen XVII protein. These allelic variants alter the strength of dermal-epidermal adhesion in the context of the Lamc2(jeb) mutation and, consequentially, broadly impact the clinical severity of JEB. Overall the results provide an explanation for how normally innocuous allelic variants can act epistatically with a disease causing mutation to impact the severity of a rare, heritable mechanobullous disorder.


Subject(s)
Autoantigens/genetics , Epidermolysis Bullosa, Junctional/genetics , Epistasis, Genetic , Laminin/genetics , Non-Fibrillar Collagens/genetics , Animals , Disease Models, Animal , Epidermolysis Bullosa, Junctional/etiology , Epidermolysis Bullosa, Junctional/pathology , Genetic Variation , Mice , Mutation , Collagen Type XVII
17.
J Immunol ; 191(9): 4581-8, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24078696

ABSTRACT

The pleiotropic cytokine IL-21 is implicated in the pathogenesis of human systemic lupus erythematosus by polymorphisms in the molecule and its receptor (IL-21R). The systemic lupus erythematosus-like autoimmune disease of BXSB.Yaa mice is critically dependent on IL-21 signaling, providing a model for understanding IL-21/IL-21R signaling in lupus pathogenesis. In this study, we generated BXSB.Yaa mice selectively deficient in IL-21R on B cells, on all T cells, or on CD8(+) T cells alone and examined the effects on disease. We found that IL-21 signaling to B cells is essential for the development of all classical disease manifestations, but that IL-21 signaling also supports the expansion of central memory, CD8(+) suppressor cells and broadly represses the cytokine activity of CD4(+) T cells. These results indicate that IL-21 has both disease-promoting and disease-suppressive effects in the autoimmune disease of BXSB.Yaa mice.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Interleukins/metabolism , Lupus Erythematosus, Systemic/immunology , Receptors, Interleukin-21/metabolism , Animals , B-Lymphocytes , CD4-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Interleukins/genetics , Male , Mice , Mice, Transgenic , Receptors, Interleukin-21/genetics , Signal Transduction/immunology
18.
Exp Dermatol ; 21(6): 453-5, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22507638

ABSTRACT

Epidermolysis bullosa (EB) describes a spectrum of rare, incurable, inherited mechanobullous disorders unified by the fact that they are caused by structural defects in the basement membrane zone which disrupt adhesion between the epidermis and dermis. Mouse models provide valuable tools to define the molecular basis of these diseases and to test novel therapeutic approaches. There is need for rapid, quantitative tests that measure the integrity of dermal-epidermal adhesions in such models. To address this need, we describe a novel quantitative method to determine the mechanical strength of the adhesion between tail skin epidermis and dermis. We show that this test reliably measures the force required to cause dermal-epidermal separation in tails of mice that are genetically predisposed to a form of non-Herlitz Junctional EB which develops as the result of a hypomorphic mutation in the laminin gamma 2 gene (Lamc2(jeb) ). This simple, quantitative method of directly measuring the tensile strength of dermal-epidermal adhesion provides a novel dimension to the pathophysiological screening, evaluation, and therapeutic treatment of mice that may develop progressive forms of EB and potentially other disorders that compromise cutaneous integrity.


Subject(s)
Epidermolysis Bullosa/physiopathology , Skin/physiopathology , Animals , Dermatology/methods , Disease Models, Animal , Mice , Mice, Inbred C57BL , Tail , Tensile Strength
19.
Mol Cells ; 33(4): 393-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22441676

ABSTRACT

TCR of CD8 T cells recognizes peptides of 8-9 amino acids in length (epitope) complexed with MHC class I. Peptide ligands differing from an epitope by one or two amino acids are thought to modulate the immune response specific to that epitope. H60 is a minor histocompatibility antigen for which the specific CD8 T-cell response dominates during alloresponse after MHC-matched allogeneic transplantation. In the present study, we developed a transgenic mouse (designated H60H Tg) expressing a variant of H60, designated H60H, in which the arginine residue at position 4 of the H60 epitope sequence (LTFNYRNL) is replaced by a histidine residue (LTFHYRNL). Immunization of female C57BL/6 mice with splenocytes from male H60H Tg induced a CD8 T cell primary response and memory response after re-challenge. The response was CD4 help-dependent, demonstrating the potency of H60H as a cellular antigen. The response induced by the H60H cellular antigen was comparable to that induced by H60 in its peak magnitude and overall immune kinetics. H60H challenge recruited broadly diverse TCRs to the specific response, shaping a TCR repertoire different from that of the natural H60 epitope. However, some of the TCRs did overlap between the H60H- and H60-specific CD8 T cells, suggesting that H60H might modulate the H60-specific response. These results may provide a basis for the modulation of the H60-specific CD8 T-cell response.


Subject(s)
CD8 Antigens/immunology , Epitopes , Histocompatibility Antigens Class II/immunology , Minor Histocompatibility Antigens/immunology , Receptors, Antigen, T-Cell/immunology , Amino Acid Substitution/genetics , Amino Acid Substitution/immunology , Animals , CD8 Antigens/genetics , Epitopes/genetics , Epitopes/immunology , Female , Histocompatibility Antigens Class II/genetics , Immunization , Mice , Mice, Inbred C57BL , Mice, Transgenic , Minor Histocompatibility Antigens/genetics , Peptides/immunology , Transplantation, Homologous/immunology
20.
J Immunol ; 187(9): 4695-704, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21964024

ABSTRACT

Dysregulation of the T cell-dependent Ab response can lead to numerous immunological disorders, ranging from systemic lupus erythematosus to B cell lymphomas. Cellular processes governed by MHC class II proteins play a major role in this response and its dysregulation. The extent to which processes controlled by the diverse family of MHC class I proteins impact such autoimmune and neoplastic disorders, however, is less clear. In this study, we genetically dissect the contributions of individual MHC class I family members and the pathological processes under their control in the systemic lupus erythematosus-like disease of BXSB.Yaa mice and B cell lymphomagenesis of SJL mice. This study reveals a powerful repressive regulatory axis comprised of MHC class I-dependent CD8(+) T cells and NK cells. These results indicate that the predominant role of the MHC class I protein family in such immunological disorders is to protect from more aggressive diseases.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Histocompatibility Antigens Class I/physiology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/prevention & control , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/prevention & control , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , H-2 Antigens/genetics , Histocompatibility Antigen H-2D , Histocompatibility Antigens Class I/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Lupus Erythematosus, Systemic/mortality , Lymphoma, B-Cell/mortality , Mice , Mice, Inbred Strains , Moloney murine leukemia virus/immunology , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...