Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Nucleic Acids Res ; 52(10): 5732-5755, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38597682

ABSTRACT

Expansion of a G4C2 repeat in the C9orf72 gene is associated with familial Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia (FTD). To investigate the underlying mechanisms of repeat instability, which occurs both somatically and intergenerationally, we created a novel mouse model of familial ALS/FTD that harbors 96 copies of G4C2 repeats at a humanized C9orf72 locus. In mouse embryonic stem cells, we observed two modes of repeat expansion. First, we noted minor increases in repeat length per expansion event, which was dependent on a mismatch repair pathway protein Msh2. Second, we found major increases in repeat length per event when a DNA double- or single-strand break (DSB/SSB) was artificially introduced proximal to the repeats, and which was dependent on the homology-directed repair (HDR) pathway. In mice, the first mode primarily drove somatic repeat expansion. Major changes in repeat length, including expansion, were observed when SSB was introduced in one-cell embryos, or intergenerationally without DSB/SSB introduction if G4C2 repeats exceeded 400 copies, although spontaneous HDR-mediated expansion has yet to be identified. These findings provide a novel strategy to model repeat expansion in a non-human genome and offer insights into the mechanism behind C9orf72 G4C2 repeat instability.


Subject(s)
C9orf72 Protein , DNA Repeat Expansion , Genomic Instability , Animals , Humans , Mice , Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein/genetics , Disease Models, Animal , DNA Breaks, Double-Stranded , DNA Repeat Expansion/genetics , Frontotemporal Dementia/genetics , Gene Knock-In Techniques , Genomic Instability/genetics , MutS Homolog 2 Protein/genetics
2.
Neurobiol Pain ; 14: 100136, 2023.
Article in English | MEDLINE | ID: mdl-38099276

ABSTRACT

The artemin-GFRα3 signaling pathway has been implicated in various painful conditions including migraine, cold allodynia, hyperalgesia, inflammatory bone pain, and mouse knees contain GFRα3-immunoreactive nerve endings. We developed high affinity mouse (REGN1967) and human (REGN5069) GFRα3-blocking monoclonal antibodies and, following in vivo evaluations in mouse models of chronic joint pain (osteoarthritic-like and inflammatory), conducted a first-in-human phase 1 pharmacokinetics (PK) and safety trial of REGN5069 (NCT03645746) in healthy volunteers, and a phase 2 randomized placebo-controlled efficacy and safety trial of REGN5069 (NCT03956550) in patients with knee osteoarthritis (OA) pain. In three commonly used mouse models of chronic joint pain (destabilization of the medial meniscus, intra-articular monoiodoacetate, or Complete Freund's Adjuvant), REGN1967 and REGN5069 attenuated evoked behaviors including tactile allodynia and thermal hyperalgesia without discernably impacting joint pathology or inflammation, prompting us to further evaluate REGN5069 in humans. In the phase 1 study in healthy subjects, the safety profiles of single doses of REGN5069 up to 3000 mg (intravenous) or 600 mg (subcutaneous) were comparable to placebo; PK were consistent with a monoclonal antibody exhibiting target-mediated disposition. In the phase 2 study in patients with OA knee pain, two doses of REGN5069 (100 mg or 1000 mg intravenous every 4 weeks) for 8 weeks failed to achieve the 12-week primary and secondary efficacy endpoints relative to placebo. In addition to possible differences in GFRα3 biology between mice and humans, we highlight here differences in experimental parameters that could have contributed to a different profile of efficacy in mouse models versus human OA pain. Additional research is required to more fully evaluate any potential role of GFRα3 in human pain.

3.
Sci Adv ; 9(15): eadf4490, 2023 04 14.
Article in English | MEDLINE | ID: mdl-37058568

ABSTRACT

Liver steatosis is an increasing health issue with few therapeutic options, partly because of a paucity of experimental models. In humanized liver rodent models, abnormal lipid accumulation in transplanted human hepatocytes occurs spontaneously. Here, we demonstrate that this abnormality is associated with compromised interleukin-6 (IL-6)-glycoprotein 130 (GP130) signaling in human hepatocytes because of incompatibility between host rodent IL-6 and human IL-6 receptor (IL-6R) on donor hepatocytes. Restoration of hepatic IL-6-GP130 signaling, through ectopic expression of rodent IL-6R, constitutive activation of GP130 in human hepatocytes, or humanization of an Il6 allele in recipient mice, substantially reduced hepatosteatosis. Notably, providing human Kupffer cells via hematopoietic stem cell engraftment in humanized liver mice also corrected the abnormality. Our observations suggest an important role of IL-6-GP130 pathway in regulating lipid accumulation in hepatocytes and not only provide a method to improve humanized liver models but also suggest therapeutic potential for manipulating GP130 signaling in human liver steatosis.


Subject(s)
Fatty Liver , Interleukin-6 , Humans , Mice , Animals , Interleukin-6/metabolism , Cytokine Receptor gp130/metabolism , Lipid Droplets/metabolism , Hepatocytes/metabolism , Glycoproteins , Lipids
4.
Cell Rep ; 41(10): 111769, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36476866

ABSTRACT

Monocytes are highly plastic immune cells that modulate antitumor immunity. Therefore, identifying factors that regulate tumor monocyte functions is critical for developing effective immunotherapies. Here, we determine that endogenous cancer cell-derived type I interferons (IFNs) control monocyte functional polarization. Guided by single-cell transcriptomic profiling of human and mouse tumors, we devise a strategy to distinguish and separate immunostimulatory from immunosuppressive tumor monocytes by surface CD88 and Sca-1 expression. Leveraging this approach, we show that cGAS-STING-regulated cancer cell-derived IFNs polarize immunostimulatory monocytes associated with anti-PD-1 immunotherapy response in mice. We also demonstrate that immunosuppressive monocytes convert into immunostimulatory monocytes upon cancer cell-intrinsic cGAS-STING activation. Consistently, we find that human cancer cells can produce type I IFNs that polarize monocytes, and our immunostimulatory monocyte gene signature is enriched in patient tumors that respond to anti-PD-1 immunotherapy. Our work exposes a role for cancer cell-derived IFNs in licensing monocyte functions that influence immunotherapy outcomes.


Subject(s)
Interferon Type I , Neoplasms , Humans , Mice , Animals , Monocytes
5.
J Am Assoc Lab Anim Sci ; 61(3): 256-269, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35277210

ABSTRACT

The exclusion of opportunistic pathogens is important for protecting animal health and ensuring desired research outcomes in highly immunodeficient mice. Proteus mirabilis has been associated with gastrointestinal tract lesions, septicemia, pyelonephritis, splenomegaly, and hepatitis and can influence select mouse models. To inform health-surveillance practices after we experienced difficulty in excluding P. mirabilis from our mouse colony, we aimed to determine the likelihood of detecting P. mirabilis-positive immunocompromised (SRG), immunovague (Fbn1+/-), and immunocompetent (CD1) colony mice through culture and PCR testing; to evaluate transmission via 2 sentinel-based approaches (direct contact and indirect dirty-bedding transfer); and to further characterize associated pathology. We hypothesized that immunocompromised mice would be better detectors and transmitters of P. mirabilis. Multiple logistic regression models were used for analysis and included PCR copy number, repeated testing, age, sex, and antibiotic-treated (trimethoprim-sulfamethoxazole) diet as covariates. Repeated testing over 10 wk showed that P. mirabilis -colonized immunocompromised colony mice were 95 times more likely than immunocompetent mice to test positive by culture and 30 times more likely by PCR assay. Sentinel mice were 15 times more likely to test positive by PCR assay for P. mirabilis when exposed by direct contact compared with dirty bedding and 18 times more likely to test positive when exposed to positive immunocompromised as compared with immunocompetent colony mice. After 10 wk of exposure, 3.8% of dirty-bedding sentinel PCR tests were positive, as compared with 30.7% of contact sentinels. Only immunocompromised mice on antibiotic diet (37.5%) developed lesions of the urogenital tract and abdominal cavity consistent with known pathology of P. mirabilis. Our findings suggest that PCR testing of dirty-bedding sentinels alone is not sufficient for the detection of P. mirabilis in mouse colonies. Direct-contact sentinels and testing of colony mice-especially if immunocompromised-with adjunct culture may facilitate successful bioexclusion.


Subject(s)
Rodent Diseases , Animals , Anti-Bacterial Agents , Bedding and Linens , Housing, Animal , Mice , Proteus mirabilis , Rodent Diseases/diagnosis
6.
Sci Immunol ; 6(66): eabj4026, 2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34919442

ABSTRACT

Despite the enormous promise of T cell therapies, the isolation and study of human T cell receptors (TCRs) of dedicated specificity remains a major challenge. To overcome this limitation, we generated mice with a genetically humanized system of T cell immunity. We used VelociGene technology to replace the murine TCRαß variable regions, along with regions encoding the extracellular domains of co-receptors CD4 and CD8, and major histocompatibility complex (MHC) class I and II, with corresponding human sequences. The resulting "VelociT" mice have normal myeloid and lymphoid immune cell populations, including thymic and peripheral αß T cell subsets comparable with wild-type mice. VelociT mice expressed a diverse TCR repertoire, mounted functional T cell responses to lymphocytic choriomeningitis virus infection, and could develop experimental autoimmune encephalomyelitis. Immunization of VelociT mice with human tumor-associated peptide antigens generated robust, antigen-specific responses and led to identification of a TCR against tumor antigen New York esophageal squamous cell carcinoma-1 with potent antitumor activity. These studies demonstrate that VelociT mice mount clinically relevant T cell responses to both MHC-I­ and MHC-II­restricted antigens, providing a powerful new model for analyzing T cell function in human disease. Moreover, VelociT mice are a new platform for de novo discovery of therapeutic human TCRs.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Animals , Humans , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/genetics
7.
Comp Med ; 71(6): 485-491, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34782034

ABSTRACT

Gastrointestinal microbiota are affected by a wide variety of extrinsic and intrinsic factors. In the husbandry of laboratory mice and design of experiments, controlling these factors where possible provides more reproducible results. However, the microbiome is dynamic, particularly in the weeks immediately after weaning. In this study, we characterized the baseline gastrointestinal microbiota of immunocompromised mice housed under standard conditions for our facility for 6 weeks after weaning, with housing either in an isolator or in individually ventilated cages and a common antibiotic diet (trimethoprim sulfamethoxazole). We compared these conditions to a group fed a standard diet and a group that was weaned to a standard diet then switched to antibiotic diet after 2 weeks. We found no clear effect of diet on richness and α diversity of the gastrointestinal microbiota. However, diet did affect which taxa were enriched at the end of the experiment. The change to antibiotic diet during the experiment did not convert the gastrointestinal microbiome to a state similar to mice consistently fed antibiotic diet, which may highlight the importance of the initial post-weaning period in the establishment of the gastrointestinal microbiome. We also observed a strong effect of housing type (isolator compared with individually ventilated cage) on the richness, α diversity, ß diversity, and taxa enriched over the course of the experiment. Investigating whether the diet or microbiome affects a certain strain's phenotype is warranted in some cases. However, our findings do not suggest that maintaining immunocompromised mice on antibiotic feed has a clinical benefit when potential pathogens are operationally excluded, nor does it result in a more consistent or controlled microbiome in the post-weaning period.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Animals , Anti-Bacterial Agents/pharmacology , Diet , Housing Quality , Mice
8.
Commun Biol ; 4(1): 913, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34312491

ABSTRACT

Tissue-resident γδ intraepithelial lymphocytes (IELs) orchestrate innate and adaptive immune responses to maintain intestinal epithelial barrier integrity. Epithelia-specific butyrophilin-like (Btnl) molecules induce perinatal development of distinct Vγ TCR+ IELs, however, the mechanisms that control γδ IEL maintenance within discrete intestinal segments are unclear. Here, we show that Btnl2 suppressed homeostatic proliferation of γδ IELs preferentially in the ileum. High throughput transcriptomic characterization of site-specific Btnl2-KO γδ IELs reveals that Btnl2 regulated the antimicrobial response module of ileal γδ IELs. Btnl2 deficiency shapes the TCR specificities and TCRγ/δ repertoire diversity of ileal γδ IELs. During DSS-induced colitis, Btnl2-KO mice exhibit increased inflammation and delayed mucosal repair in the colon. Collectively, these data suggest that Btnl2 fine-tunes γδ IEL frequencies and TCR specificities in response to site-specific homeostatic and inflammatory cues. Hence, Btnl-mediated targeting of γδ IEL development and maintenance may help dissect their immunological functions in intestinal diseases with segment-specific manifestations.


Subject(s)
Butyrophilins/genetics , Ileum/immunology , Immunity, Innate/genetics , Immunity, Mucosal/genetics , Intraepithelial Lymphocytes/metabolism , Animals , Butyrophilins/metabolism , Female , Mice , Mice, Inbred C57BL
9.
Sci Transl Med ; 12(549)2020 06 24.
Article in English | MEDLINE | ID: mdl-32581132

ABSTRACT

Monoclonal antibodies that block the programmed cell death 1 (PD-1) checkpoint have revolutionized cancer immunotherapy. However, many major tumor types remain unresponsive to anti-PD-1 therapy, and even among responsive tumor types, most of the patients do not develop durable antitumor immunity. It has been shown that bispecific antibodies activate T cells by cross-linking the TCR/CD3 complex with a tumor-specific antigen (TSA). The class of TSAxCD3 bispecific antibodies have generated exciting results in early clinical trials. We have recently described another class of "costimulatory bispecifics" that cross-link a TSA to CD28 (TSAxCD28) and cooperate with TSAxCD3 bispecifics. Here, we demonstrate that these TSAxCD28 bispecifics (one specific for prostate cancer and the other for epithelial tumors) can also synergize with the broader anti-PD-1 approach and endow responsiveness-as well as long-term immune memory-against tumors that otherwise do not respond to anti-PD-1 alone. Unlike CD28 superagonists, which broadly activate T cells and induce cytokine storm, TSAxCD28 bispecifics display little or no toxicity when used alone or in combination with a PD-1 blocker in genetically humanized immunocompetent mouse models or in primates and thus may provide a well-tolerated and "off the shelf" combination approach with PD-1 immunotherapy that can markedly enhance antitumor efficacy.


Subject(s)
Antibodies, Bispecific , Neoplasms , Animals , Antibodies, Bispecific/therapeutic use , CD28 Antigens , Humans , Immunotherapy , Mice , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor
10.
Eur J Hum Genet ; 28(9): 1243-1264, 2020 09.
Article in English | MEDLINE | ID: mdl-32376988

ABSTRACT

Previously we reported the identification of a homozygous COL27A1 (c.2089G>C; p.Gly697Arg) missense variant and proposed it as a founder allele in Puerto Rico segregating with Steel syndrome (STLS, MIM #615155); a rare osteochondrodysplasia characterized by short stature, congenital bilateral hip dysplasia, carpal coalitions, and scoliosis. We now report segregation of this variant in five probands from the initial clinical report defining the syndrome and an additional family of Puerto Rican descent with multiple affected adult individuals. We modeled the orthologous variant in murine Col27a1 and found it recapitulates some of the major Steel syndrome associated skeletal features including reduced body length, scoliosis, and a more rounded skull shape. Characterization of the in vivo murine model shows abnormal collagen deposition in the extracellular matrix and disorganization of the proliferative zone of the growth plate. We report additional COL27A1 pathogenic variant alleles identified in unrelated consanguineous Turkish kindreds suggesting Clan Genomics and identity-by-descent homozygosity contributing to disease in this population. The hypothesis that carrier states for this autosomal recessive osteochondrodysplasia may contribute to common complex traits is further explored in a large clinical population cohort. Our findings augment our understanding of COL27A1 biology and its role in skeletal development; and expand the functional allelic architecture in this gene underlying both rare and common disease phenotypes.


Subject(s)
Abnormalities, Multiple/genetics , Fibrillar Collagens/genetics , Founder Effect , Hip Dislocation/genetics , Scoliosis/genetics , Abnormalities, Multiple/pathology , Adolescent , Animals , Bone Development , Child , Child, Preschool , Consanguinity , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Fibrillar Collagens/metabolism , Gene Frequency , Hip Dislocation/pathology , Homozygote , Humans , Male , Mice , Mice, Inbred C57BL , Mutation , Pedigree , Scoliosis/pathology , Syndrome
11.
PLoS One ; 15(5): e0231892, 2020.
Article in English | MEDLINE | ID: mdl-32384086

ABSTRACT

Complement is a key component of the innate immune system. Inappropriate complement activation underlies the pathophysiology of a variety of diseases. Complement component 5 (C5) is a validated therapeutic target for complement-mediated diseases, but the development of new therapeutics has been limited by a paucity of preclinical models to evaluate the pharmacokinetic (PK) and pharmacodynamic (PD) properties of candidate therapies. The present report describes a novel humanized C5 mouse and its utility in evaluating a panel of fully human anti-C5 antibodies. Surprisingly, humanized C5 mice revealed marked differences in clearance rates amongst a panel of anti-C5 antibodies. One antibody, pozelimab (REGN3918), bound C5 and C5 variants with high affinity and potently blocked complement-mediated hemolysis in vitro. In studies conducted in both humanized C5 mice and cynomolgus monkeys, pozelimab demonstrated prolonged PK and durable suppression of hemolytic activity ex vivo. In humanized C5 mice, a switch in dosing from in-house eculizumab to pozelimab was associated with normalization of serum C5 concentrations, sustained suppression of hemolytic activity ex vivo, and no overt toxicity. Our findings demonstrate the value of humanized C5 mice in identifying new therapeutic candidates and treatment options for complement-mediated diseases.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Complement C5/immunology , Animals , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacology , Antigen-Antibody Reactions , Binding Sites , Complement Activation/drug effects , Complement C5/chemistry , Complement C5/genetics , Genetic Variation , Half-Life , Hemolysis/drug effects , Humans , Inhibitory Concentration 50 , Macaca fascicularis , Mice , Protein Structure, Quaternary
12.
Sci Transl Med ; 12(525)2020 01 08.
Article in English | MEDLINE | ID: mdl-31915305

ABSTRACT

T cell activation is initiated upon binding of the T cell receptor (TCR)/CD3 complex to peptide-major histocompatibility complexes ("signal 1"); activation is enhanced by engagement of a second "costimulatory" receptor, such as the CD28 receptor on T cells binding to its cognate ligand(s) on the target cell ("signal 2"). CD3-based bispecific antibodies act by replacing conventional signal 1, linking T cells to tumor cells by binding a tumor-specific antigen (TSA) with one arm of the bispecific and bridging to TCR/CD3 with the other. Although some of these so-called TSAxCD3 bispecifics have demonstrated promising antitumor efficacy in patients with cancer, their activity remains to be optimized. Here, we introduce a class of bispecific antibodies that mimic signal 2 by bridging TSA to the costimulatory CD28 receptor on T cells. We term these TSAxCD28 bispecifics and describe two such bispecific antibodies: one specific for ovarian and the other for prostate cancer antigens. Unlike CD28 superagonists, which broadly activate T cells and resulted in profound toxicity in early clinical trials, these TSAxCD28 bispecifics show limited activity and no toxicity when used alone in genetically humanized immunocompetent mouse models or in primates. However, when combined with TSAxCD3 bispecifics, they enhance the artificial synapse between a T cell and its target cell, potentiate T cell activation, and markedly improve antitumor activity of CD3 bispecifics in a variety of xenogeneic and syngeneic tumor models. Combining this class of CD28-costimulatory bispecific antibodies with the emerging class of TSAxCD3 bispecifics may provide well-tolerated, off-the-shelf antibody therapies with robust antitumor efficacy.


Subject(s)
Antibodies, Bispecific/immunology , CD28 Antigens/immunology , CD3 Complex/immunology , Neoplasms/immunology , Animals , Antigens, Neoplasm/immunology , Cell Line, Tumor , Cell Proliferation , Cytokines/metabolism , Cytotoxicity, Immunologic , Female , HEK293 Cells , Humans , Immunological Synapses/metabolism , Lymphocyte Activation/immunology , Macaca fascicularis , Mice , Neoplasms/pathology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
13.
Sci Immunol ; 5(54)2020 12 18.
Article in English | MEDLINE | ID: mdl-33443029

ABSTRACT

Deficiency in interleukin-36R (IL-36R) antagonist caused by loss-of-function mutations in IL-36RN leads to DITRA (deficiency of IL-36 receptor antagonist), a rare inflammatory human disease that belongs to a subgroup of generalized pustular psoriasis (GPP). We report a functional genetic mouse model of DITRA with enhanced IL-36R signaling analogous to that observed in patients with DITRA, which provides new insight into our understanding of the IL-36 family of molecules in regulating barrier integrity across multiple tissues. Humanized DITRA-like mice displayed increased skin inflammation in a preclinical model of psoriasis, and in vivo blockade of IL-36R pathway using anti-human IL-36R antibody ameliorated imiquimod-induced skin pathology as both prophylactic and therapeutic treatments. Deeper characterization of the humanized DITRA-like mice revealed that deregulated IL-36R signaling promoted tissue pathology during intestinal injury and led to impairment in mucosal restoration in the repair phase of chronic dextran sulfate sodium (DSS)-induced colitis. Blockade of IL-36R pathway significantly ameliorated DSS-induced intestinal inflammation and rescued the inability of DITRA-like mice to recover from mucosal damage in vivo. Our results indicate a central role for IL-36 in regulating proinflammatory responses in the skin and epithelial barrier function in the intestine, suggesting a new therapeutic potential for targeting the IL-36R axis in psoriasis and at the later stages of intestinal pathology in inflammatory bowel disease.


Subject(s)
Dermatitis/etiology , Dermatitis/metabolism , Gastroenteritis/etiology , Gastroenteritis/metabolism , Receptors, Interleukin-1/metabolism , Signal Transduction , Animals , Biomarkers , Dermatitis/pathology , Disease Models, Animal , Disease Susceptibility , Gastroenteritis/pathology , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Skin/metabolism , Skin/pathology
14.
Proc Natl Acad Sci U S A ; 117(1): 292-299, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31879340

ABSTRACT

We describe a Kappa-on-Heavy (KoH) mouse that produces a class of highly diverse, fully human, antibody-like agents. This mouse was made by replacing the germline variable sequences of both the Ig heavy-chain (IgH) and Ig kappa (IgK) loci with the human IgK germline variable sequences, producing antibody-like molecules with an antigen binding site made up of 2 kappa variable domains. These molecules, named KoH bodies, structurally mimic naturally existing Bence-Jones light-chain dimers in their variable domains and remain wild-type in their antibody constant domains. Unlike artificially diversified, nonimmunoglobulin alternative scaffolds (e.g., DARPins), KoH bodies consist of a configuration of normal Ig scaffolds that undergo natural diversification in B cells. Monoclonal KoH bodies have properties similar to those of conventional antibodies but exhibit an enhanced ability to bind small molecules such as the endogenous cardiotonic steroid marinobufagenin (MBG) and nicotine. A comparison of crystal structures of MBG bound to a KoH Fab versus a conventional Fab showed that the KoH body has a much deeper binding pocket, allowing MBG to be held 4 Å further down into the combining site between the 2 variable domains.


Subject(s)
Antibodies/chemistry , Antibodies/immunology , Antigens/immunology , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/immunology , Immunoglobulin kappa-Chains/chemistry , Animals , Antibodies/genetics , Antibodies/therapeutic use , Base Sequence , Binding Sites, Antibody/genetics , Bufanolides , Genetic Engineering , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Immunoglobulin kappa-Chains/genetics , Mice , Models, Molecular , Nicotine , Protein Conformation
15.
Mol Cancer Ther ; 18(11): 2051-2062, 2019 11.
Article in English | MEDLINE | ID: mdl-31395688

ABSTRACT

In the tumor microenvironment, multiple inhibitory checkpoint receptors can suppress T-cell function, thereby enabling tumor immune evasion. Blockade of one of these checkpoint receptors, PD-1, with therapeutic antibodies has produced positive clinical responses in various cancers; however, the efficacy of this approach can be further improved. Simultaneously targeting multiple inhibitory checkpoint receptors has emerged as a promising therapeutic strategy. Here, we report the development and characterization of REGN3767, a fully human IgG4 antibody targeting LAG-3, another inhibitory receptor on T cells. REGN3767 binds human and monkey LAG-3 with high affinity and specificity and blocks the interaction of LAG-3 with its ligand, MHC class II. In an engineered T-cell/antigen-presenting cell bioassay, REGN3767 alone, or in combination with cemiplimab (REGN2810, human anti-PD-1 antibody), blocked inhibitory signaling to T cells mediated by hLAG-3/MHCII in the presence of PD-1/PD-L1. To test the in vivo activity of REGN3767 alone or in combination with cemiplimab, we generated human PD-1xLAG-3 knockin mice, in which the extracellular domains of mouse Pdcd1 and Lag3 were replaced with their human counterparts. In these humanized mice, treatment with cemiplimab and REGN3767 showed increased efficacy in a mouse tumor model and enhanced the secretion of proinflammatory cytokines by tumor-specific T cells. The favorable pharmacokinetics and toxicology of REGN3767 in nonhuman primates, together with enhancement of antitumor efficacy of anti-PD-1 antibody in preclinical tumor models, support its clinical development.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antigens, CD/chemistry , Antigens, CD/genetics , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/genetics , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, CD/metabolism , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Gene Knock-In Techniques , Haplorhini , Histocompatibility Antigens Class II/metabolism , Humans , Mice , Neoplasms/genetics , Protein Binding/drug effects , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays , Lymphocyte Activation Gene 3 Protein
16.
Biol Reprod ; 100(3): 686-696, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30289441

ABSTRACT

The Adisintegrin and metalloprotease domain-containing (ADAM) family of proteins is involved in cell adhesion, migration, proteolysis, and signaling. Many ADAMs are required for reproduction; however, the role of Adam6 has remained largely unknown. In the course of humanizing the mouse immunoglobulin heavy chain (IgH) locus, we generated Adam6-deficient mice that demonstrate severe subfertility. We decided to elucidate the role of ADAM6 in fertility and explore the underlying mechanisms. Despite normal sperm development and motility, Adam6-deficient mice display diminished male fertility, have abnormal sperm adhesion, and most importantly cannot transition from uterus to oviduct. To test whether ADAM6 is required for sperm's binding to extracellular matrix (ECM) components, we used a panel of ECM components and showed that unlike normal sperm, Adam6-deficient sperm cannot bind fibronectin, laminin, and tenascin. Reintroduction of Adam6 into these deficient mice repaired sperm interaction with ECM, restored male fertility, and corrected the sperm transport deficit. Together, our data suggest that ADAM6, either alone or in complex with other proteins, aids sperm transport through the female reproductive tract by providing a temporary site of attachment of sperm to ECM components prior to ascent into the oviduct.


Subject(s)
ADAM Proteins/metabolism , Infertility, Male/genetics , Sperm Motility/physiology , Spermatozoa/physiology , ADAM Proteins/genetics , Animals , Female , Male , Mice , Mice, Knockout , Oviducts , Sperm Motility/genetics
17.
Sci Immunol ; 3(29)2018 11 02.
Article in English | MEDLINE | ID: mdl-30389797

ABSTRACT

Most patients with cancer do not develop durable antitumor responses after programmed cell death protein 1 (PD-1) or programmed cell death ligand 1(PD-L1) checkpoint inhibition monotherapy because of an ephemeral reversal of T cell dysfunction and failure to promote long-lasting immunological T cell memory. Activating costimulatory pathways to induce stronger T cell activation may improve the efficacy of checkpoint inhibition and lead to durable antitumor responses. We performed single-cell RNA sequencing of more than 2000 tumor-infiltrating CD8+ T cells in mice receiving both PD-1 and GITR (glucocorticoid-induced tumor necrosis factor receptor-related protein) antibodies and found that this combination synergistically enhanced the effector function of expanded CD8+ T cells by restoring the balance of key homeostatic regulators CD226 and T cell immunoreceptor with Ig and ITIM domains (TIGIT), leading to a robust survival benefit. Combination therapy decreased CD8+ T cell dysfunction and induced a highly proliferative precursor effector memory T cell phenotype in a CD226-dependent manner. PD-1 inhibition rescued CD226 activity by preventing PD-1-Src homology region 2 (SHP2) dephosphophorylation of the CD226 intracellular domain, whereas GITR agonism decreased TIGIT expression. Unmasking the molecular pathways driving durable antitumor responses will be essential to the development of rational approaches to optimizing cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Glucocorticoid-Induced TNFR-Related Protein/immunology , Immunologic Memory/immunology , Immunotherapy , Neoplasms/therapy , Programmed Cell Death 1 Receptor/immunology , Animals , Antigens, Differentiation, T-Lymphocyte/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/pathology , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Phenotype
18.
Mol Cancer Ther ; 16(5): 861-870, 2017 05.
Article in English | MEDLINE | ID: mdl-28265006

ABSTRACT

The Programmed Death-1 (PD-1) receptor delivers inhibitory checkpoint signals to activated T cells upon binding to its ligands PD-L1 and PD-L2 expressed on antigen-presenting cells and cancer cells, resulting in suppression of T-cell effector function and tumor immune evasion. Clinical antibodies blocking the interaction between PD-1 and PD-L1 restore the cytotoxic function of tumor antigen-specific T cells, yielding durable objective responses in multiple cancers. This report describes the preclinical characterization of REGN2810, a fully human hinge-stabilized IgG4(S228P) high-affinity anti-PD-1 antibody that potently blocks PD-1 interactions with PD-L1 and PD-L2. REGN2810 was characterized in a series of binding, blocking, and functional cell-based assays, and preclinical in vivo studies in mice and monkeys. In cell-based assays, REGN2810 reverses PD-1-dependent attenuation of T-cell receptor signaling in engineered T cells and enhances responses of human primary T cells. To test the in vivo activity of REGN2810, which does not cross-react with murine PD-1, knock-in mice were generated to express a hybrid protein containing the extracellular domain of human PD-1, and transmembrane and intracellular domains of mouse PD-1. In these mice, REGN2810 binds the humanized PD-1 receptor and inhibits growth of MC38 murine tumors. As REGN2810 binds to cynomolgus monkey PD-1 with high affinity, pharmacokinetic and toxicologic assessment of REGN2810 was performed in cynomolgus monkeys. High doses of REGN2810 were well tolerated, without adverse immune-related effects. These preclinical studies validate REGN2810 as a potent and promising candidate for cancer immunotherapy. Mol Cancer Ther; 16(5); 861-70. ©2017 AACR.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Cell Proliferation/drug effects , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/genetics , Animals , Antibodies, Monoclonal, Humanized/immunology , Cell Line, Tumor , Gene Knock-In Techniques , Humans , Immunotherapy , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
19.
Sci Rep ; 6: 23204, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26979938

ABSTRACT

The expansion of a hexanucleotide (GGGGCC) repeat in C9ORF72 is the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Both the function of C9ORF72 and the mechanism by which the repeat expansion drives neuropathology are unknown. To examine whether C9ORF72 haploinsufficiency induces neurological disease, we created a C9orf72-deficient mouse line. Null mice developed a robust immune phenotype characterized by myeloid expansion, T cell activation, and increased plasma cells. Mice also presented with elevated autoantibodies and evidence of immune-mediated glomerulonephropathy. Collectively, our data suggest that C9orf72 regulates immune homeostasis and an autoimmune response reminiscent of systemic lupus erythematosus (SLE) occurs in its absence. We further imply that haploinsufficiency is unlikely to be the causative factor in C9ALS/FTD pathology.


Subject(s)
Autoantibodies/biosynthesis , Autoimmunity , Glomerulonephritis, Membranoproliferative/genetics , Guanine Nucleotide Exchange Factors/genetics , Animals , Autoantibodies/blood , C9orf72 Protein , Cytokines/blood , Female , Glomerulonephritis, Membranoproliferative/blood , Glomerulonephritis, Membranoproliferative/immunology , Guanine Nucleotide Exchange Factors/metabolism , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation , Lymphoid Tissue/pathology , Macrophages/immunology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/immunology , Sequence Analysis, RNA , Transcriptome
20.
PLoS One ; 10(4): e0125522, 2015.
Article in English | MEDLINE | ID: mdl-25909911

ABSTRACT

In a survey of 20 knockout mouse lines designed to examine the biological functions of large intergenic non-coding RNAs (lincRNAs), we have found a variety of phenotypes, ranging from perinatal lethality to defects associated with premature aging and morphological and functional abnormalities in the lungs, skeleton, and muscle. Each mutant allele carried a lacZ reporter whose expression profile highlighted a wide spectrum of spatiotemporal and tissue-specific transcription patterns in embryos and adults that informed our phenotypic analyses and will serve as a guide for future investigations of these genes. Our study shows that lincRNAs are a new class of encoded molecules that, like proteins, serve essential and important functional roles in embryonic development, physiology, and homeostasis of a broad array of tissues and organs in mammals.


Subject(s)
RNA, Long Noncoding/genetics , Transcription, Genetic/genetics , Transcriptome/genetics , Alleles , Animals , Embryonic Development/genetics , Female , Genes, Reporter/genetics , Male , Mammals/genetics , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...