Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Biol Chem ; 294(37): 13580-13592, 2019 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-31285261

RESUMO

Antigen receptor assembly in lymphocytes involves stringently-regulated coordination of specific DNA rearrangement events across several large chromosomal domains. Previous studies indicate that transcription factors such as paired box 5 (PAX5), Yin Yang 1 (YY1), and CCCTC-binding factor (CTCF) play a role in regulating the accessibility of the antigen receptor loci to the V(D)J recombinase, which is required for these rearrangements. To gain clues about the role of CTCF binding at the murine immunoglobulin heavy chain (IgH) locus, we utilized a computational approach that identified 144 putative CTCF-binding sites within this locus. We found that these CTCF sites share a consensus motif distinct from other CTCF sites in the mouse genome. Additionally, we could divide these CTCF sites into three categories: intergenic sites remote from any coding element, upstream sites present within 8 kb of the VH-leader exon, and recombination signal sequence (RSS)-associated sites characteristically located at a fixed distance (∼18 bp) downstream of the RSS. We noted that the intergenic and upstream sites are located in the distal portion of the VH locus, whereas the RSS-associated sites are located in the DH-proximal region. Computational analysis indicated that the prevalence of CTCF-binding sites at the IgH locus is evolutionarily conserved. In all species analyzed, these sites exhibit a striking strand-orientation bias, with >98% of the murine sites being present in one orientation with respect to VH gene transcription. Electrophoretic mobility shift and enhancer-blocking assays and ChIP-chip analysis confirmed CTCF binding to these sites both in vitro and in vivo.


Assuntos
Fator de Ligação a CCCTC/metabolismo , Cadeias Pesadas de Imunoglobulinas/genética , Imunidade Adaptativa/genética , Animais , Sítios de Ligação , Fator de Ligação a CCCTC/genética , Cromatina/genética , Cromatina/metabolismo , Proteínas de Ligação a DNA/genética , Rearranjo Gênico , Humanos , Cadeias Pesadas de Imunoglobulinas/metabolismo , Região Variável de Imunoglobulina , Células K562 , Camundongos , Camundongos Knockout , Células NIH 3T3 , Motivos de Nucleotídeos , Sequências Reguladoras de Ácido Nucleico , Proteínas Repressoras/metabolismo
2.
AAPS J ; 26(4): 68, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38862748

RESUMO

Bispecific and multispecific agents have become increasingly utilized in cancer treatment and immunotherapy, yet their complex design parameters present a challenge in developing successful therapeutics. Bispecifics that crosslink receptors on two opposing cells can provide specific activation of a receptor only when these cells are in close spatial proximity, such as an immune cell and cancer cell in a tumor. These agents, including T cell activating bispecifics, can avoid off-tumor toxicity through activation only in the tumor microenvironment by utilizing a tumor target to cluster T-cell receptors for a selective costimulatory signal. Here, we investigate a panel of PD-1/CD137 targeted Humabody VH domains to determine the key factors for T cell activation, such as affinity, valency, expression level, domain orientation, and epitope location. Target expression is a dominant factor determining both specificity and potency of T cell activation. Given an intrinsic expression level, the affinity can be tuned to modulate the level of activation and IC50 and achieve specificity between low and high expression levels. Changing the epitope location and linker length showed minor improvements to activation at low expression levels, but increasing the valency for the target decreased activation at all expression levels. By combining non-overlapping epitopes for the target, we achieved higher receptor activation at low expression levels. A kinetic model was able to capture these trends, offering support for the mechanistic interpretation. This work provides a framework to quantify factors for T cell activation by cell-crosslinking bispecific agents and guiding principles for the design of new agents.


Assuntos
Anticorpos Biespecíficos , Ativação Linfocitária , Receptor de Morte Celular Programada 1 , Linfócitos T , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral , Anticorpos Biespecíficos/farmacologia , Anticorpos Biespecíficos/imunologia , Humanos , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Linfócitos T/imunologia , Linfócitos T/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Receptor de Morte Celular Programada 1/imunologia , Reagentes de Ligações Cruzadas/química , Desenho de Fármacos
3.
Neoplasia ; 48: 100962, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38183712

RESUMO

Bispecific agents are a rapidly growing class of cancer therapeutics, and immune targeted bispecific agents have the potential to expand functionality well beyond monoclonal antibody agents. Humabodies⁎ are fully human single domain antibodies that can be linked in a modular fashion to form multispecific therapeutics. However, the effect of heterogeneous delivery on the efficacy of crosslinking bispecific agents is currently unclear. In this work, we utilize a PSMA-CD137 Humabody with an albumin binding half-life extension (HLE) domain to determine the impact of tissue penetration on T cell activating bispecific agents. Using heterotypic spheroids, we demonstrate that increased tissue penetration results in higher T cell activation at sub-saturating concentrations. Next, we tested the effect of two different albumin binding moieties on tissue distribution using albumin-specific HLE domains with varying affinities for albumin and a non-specific lipophilic dye. The results show that a specific binding mechanism to albumin does not influence tissue penetration, but a non-specific mechanism reduced both spheroid uptake and distribution in the presence of albumin. These results highlight the potential importance of tissue penetration on bispecific agent efficacy and describe how the design parameters including albumin-binding domains can be selected to maximize the efficacy of bispecific agents.


Assuntos
Anticorpos Biespecíficos , Antineoplásicos , Neoplasias , Humanos , Linfócitos T , Anticorpos Biespecíficos/farmacologia , Anticorpos Biespecíficos/química , Albuminas/uso terapêutico , Neoplasias/tratamento farmacológico , Antineoplásicos/uso terapêutico
4.
Clin Cancer Res ; 30(8): 1595-1606, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38593226

RESUMO

PURPOSE: CD137 is a T- and NK-cell costimulatory receptor involved in consolidating immunologic responses. The potent CD137 agonist urelumab has shown clinical promise as a cancer immunotherapeutic but development has been hampered by on-target off-tumor toxicities. A CD137 agonist targeted to the prostate-specific membrane antigen (PSMA), frequently and highly expressed on castration-resistant metastatic prostate cancer (mCRPC) tumor cells, could bring effective immunotherapy to this immunologically challenging to address disease. EXPERIMENTAL DESIGN: We designed and manufactured CB307, a novel half-life extended bispecific costimulatory Humabody VH therapeutic to elicit CD137 agonism exclusively in a PSMA-high tumor microenvironment (TME). The functional activity of CB307 was assessed in cell-based assays and in syngeneic mouse antitumor pharmacology studies. Nonclinical toxicology and toxicokinetic properties of CB307 were assessed in a good laboratory practice (GLP) compliant study in cynomolgus macaques. RESULTS: CB307 provides effective CD137 agonism in a PSMA-dependent manner, with antitumor activity both in vitro and in vivo, and additional activity when combined with checkpoint inhibitors. A validated novel PSMA/CD137 IHC assay demonstrated a higher prevalence of CD137-positive cells in the PSMA-expressing human mCRPC TME with respect to primary lesions. CB307 did not show substantial toxicity in nonhuman primates and exhibited a plasma half-life supporting weekly clinical administration. CONCLUSIONS: CB307 is a first-in-class immunotherapeutic that triggers potent PSMA-dependent T-cell activation, thereby alleviating toxicologic concerns against unrestricted CD137 agonism.


Assuntos
Neoplasias de Próstata Resistentes à Castração , Masculino , Humanos , Camundongos , Animais , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/patologia , Imunoterapia/métodos , Microambiente Tumoral
5.
Wound Repair Regen ; 20(3): 414-24, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22564233

RESUMO

The innate immune system differentially regulates the expression of host defense peptides to combat infection during wound healing. We enhanced the expression of a host defense peptide, human beta defensin-3 (hBD-3), in keratinocytes to generate a three-dimensional biologic dressing to improve healing of infected wounds. The NIKS human keratinocyte cell line was stably transfected ex vivo with a construct containing an epidermis-specific promoter driving hBD-3 (NIKS(hBD) (-3) ) using nonviral methods. Levels of hBD-3 mRNA and protein in three-dimensional skin tissue produced from NIKS(hBD) (-3) were determined using quantitative polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. Tissue architecture was characterized by hematoxylin and eosin staining and by indirect immunofluorescence using proliferation and keratinocyte differentiation markers. Antimicrobial activity was assessed using an in vitro bacterial growth assay and in vivo using a murine burn infection model. Three-dimensional full thickness skin tissues containing epidermal NIKS(hBD) (-3) or control NIKS possessed histologic features of interfollicular epidermis and exhibited normal tissue growth and differentiation. NIKS(hBD) (-3) tissue contained approximately fivefold more hBD-3 protein than tissue containing unmodified control NIKS. In vitro studies showed that NIKS(hBD) (-3) tissue produced a significant reduction in the growth of Staphylococcus aureus multiple peptide resistance factor (mprF) compared with control tissue. In an in vivo infected murine burn model, NIKS(hBD) (-3) tissue resulted in a 90% reduction in bacterial growth. These results demonstrate that sustained delivery of hBD-3 by a bioengineered skin tissue results in a therapeutically relevant reduction in growth of a S. aureus strain in an animal model of infected third-degree burn wounds.


Assuntos
Queimaduras/metabolismo , Infecções Cutâneas Estafilocócicas/metabolismo , Staphylococcus aureus/patogenicidade , Infecção dos Ferimentos/metabolismo , beta-Defensinas/metabolismo , Animais , Western Blotting , Queimaduras/microbiologia , Células Cultivadas , Modelos Animais de Doenças , Expressão Gênica , Humanos , Camundongos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Infecções Cutâneas Estafilocócicas/microbiologia , Cicatrização/genética , Infecção dos Ferimentos/microbiologia
6.
PLoS Genet ; 4(1): e9, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18208332

RESUMO

X chromosome inactivation in female mammals results in dosage compensation of X-linked gene products between the sexes. In humans there is evidence that a substantial proportion of genes escape from silencing. We have carried out a large-scale analysis of gene expression in lymphoblastoid cell lines from four human populations to determine the extent to which escape from X chromosome inactivation disrupts dosage compensation. We conclude that dosage compensation is virtually complete. Overall expression from the X chromosome is only slightly higher in females and can largely be accounted for by elevated female expression of approximately 5% of X-linked genes. We suggest that the potential contribution of escape from X chromosome inactivation to phenotypic differences between the sexes is more limited than previously believed.


Assuntos
Mecanismo Genético de Compensação de Dose , Genética Populacional , Linfócitos/citologia , Linhagem Celular , Cromossomos Humanos X , Cromossomos Humanos Y , Feminino , Expressão Gênica , Genes Ligados ao Cromossomo X , Haplótipos , Heterozigoto , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Polimorfismo de Nucleotídeo Único , Inativação do Cromossomo X
7.
J Immunother Cancer ; 9(4)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33795386

RESUMO

BACKGROUND: Chimeric antigen receptor (CAR) T cells are effective in B-cell malignancies. However, heterogeneous antigen expression and antigen loss remain important limitations of targeted immunotherapy in solid tumors. Therefore, targeting multiple tumor-associated antigens simultaneously is expected to improve the outcome of CAR-T cell therapies. Due to the instability of single-chain variable fragments, it remains challenging to develop the simultaneous targeting of multiple antigens using traditional single-chain fragment variable (scFv)-based CARs. METHODS: We used Humabody VH domains derived from a transgenic mouse to obtain fully human prostate-specific membrane antigen (PSMA) VH and mesothelin (MSLN) VH sequences and redirect T cell with VH based-CAR. The antitumor activity and mode of action of PSMA VH and MSLN VH were evaluated in vitro and in vivo compared with the traditional scFv-based CARs. RESULTS: Human VH domain-based CAR targeting PSMA and MSLN are stable and functional both in vitro and in vivo. VH modules in the bispecific format are capable of binding their specific target with similar affinity as their monovalent counterparts. Bispecific CARs generated by joining two human antibody VH domains can prevent tumor escape in tumor with heterogeneous antigen expression. CONCLUSIONS: Fully human antibody VH domains can be used to generate functional CAR molecules, and redirected T cells elicit antitumoral responses in solid tumors at least as well as conventional scFv-based CARs. In addition, VH domains can be used to generate bispecific CAR-T cells to simultaneously target two different antigens expressed by tumor cells, and therefore, achieve better tumor control in solid tumors.


Assuntos
Antígenos de Superfície/imunologia , Glutamato Carboxipeptidase II/imunologia , Região Variável de Imunoglobulina/imunologia , Imunoterapia Adotiva , Mesotelina/imunologia , Neoplasias da Próstata/terapia , Receptores de Antígenos Quiméricos/imunologia , Anticorpos de Cadeia Única/imunologia , Linfócitos T/transplante , Animais , Linhagem Celular Tumoral , Técnicas de Cocultura , Citocinas/metabolismo , Citotoxicidade Imunológica , Humanos , Região Variável de Imunoglobulina/genética , Ativação Linfocitária , Masculino , Camundongos Endogâmicos NOD , Fenótipo , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/metabolismo , Receptores de Antígenos Quiméricos/genética , Anticorpos de Cadeia Única/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Mol Ther ; 17(3): 562-9, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19190595

RESUMO

When skin is compromised, a cascade of signals initiates the rapid repair of the epidermis to prevent fluid loss and provide defense against invading microbes. During this response, keratinocytes produce host defense peptides (HDPs) that have antimicrobial activity against a diverse set of pathogens. Using nonviral vectors we have genetically modified the novel, nontumorigenic, pathogen-free human keratinocyte progenitor cell line (NIKS) to express the human cathelicidin HDP in a tissue-specific manner. NIKS skin tissue that expresses elevated levels of cathelicidin possesses key histological features of normal epidermis and displays enhanced antimicrobial activity against bacteria in vitro. Moreover, in an in vivo infected burn wound model, this tissue results in a two log reduction in a clinical isolate of multidrug-resistant Acinetobacter baumannii. Taken together, these results suggest that this genetically engineered human tissue could be applied to burns and ulcers to counteract bacterial contamination and prevent infection.


Assuntos
Acinetobacter baumannii/fisiologia , Peptídeos Catiônicos Antimicrobianos/metabolismo , Farmacorresistência Bacteriana Múltipla , Expressão Gênica , Engenharia de Proteínas/métodos , Pele/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Queimaduras/genética , Queimaduras/microbiologia , Queimaduras/terapia , Células Cultivadas , Modelos Animais de Doenças , Terapia Genética , Vetores Genéticos/genética , Humanos , Queratinócitos/metabolismo , Camundongos , Camundongos Nus , Catelicidinas
9.
N Biotechnol ; 55: 65-76, 2020 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-31600579

RESUMO

We describe the 'Crescendo Mouse', a human VH transgenic platform combining an engineered heavy chain locus with diverse human heavy chain V, D and J genes, a modified mouse Cγ1 gene and complete 3' regulatory region, in a triple knock-out (TKO) mouse background devoid of endogenous immunoglobulin expression. The addition of the engineered heavy chain locus to the TKO mouse restored B cell development, giving rise to functional B cells that responded to immunization with a diverse response that comprised entirely 'heavy chain only' antibodies. Heavy chain variable (VH) domain libraries were rapidly mined using phage display technology, yielding diverse high-affinity human VH that had undergone somatic hypermutation, lacked aggregation and showed enhanced expression in E. coli. The Crescendo Mouse produces human VH fragments, or Humabody® VH, with excellent bio-therapeutic potential, as exemplified here by the generation of antagonistic Humabody® VH specific for human IL17A and IL17RA.


Assuntos
Anticorpos/imunologia , Cadeias Pesadas de Imunoglobulinas/imunologia , Região Variável de Imunoglobulina/imunologia , Animais , Formação de Anticorpos/imunologia , Fenômenos Biofísicos , Humanos , Camundongos Knockout
10.
J Immunol ; 176(7): 4221-34, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16547259

RESUMO

The mechanisms that regulate variable (V) gene selection during the development of the mouse IgH repertoire are not fully understood, due in part to the absence of the complete locus sequence. To better understand these processes, we have assembled the entire 2.5-Mb mouse IgH (Igh) V region sequence of the C57BL/6 strain from public sequences and present the first complete annotated map of the region, including V genes, pseudogenes, repeats, and nonrepetitive intergenic sequences. In so doing, we have discovered a new V gene family, VH16. We have identified clusters of conserved region-specific intergenic sequences and have verified our assembly by genic and intergenic Southern blotting. We have observed that V pseudogenes are not evenly spread throughout the V region, but rather cluster together. The largest J558 family, which spans more than half of the locus, has two strikingly different domains, which suggest points of evolutionary divergence or duplication. The 5' end contains widely spaced J558 genes interspersed with 3609 genes and is pseudogene poor. The 3' end contains closely spaced J558 genes, no 3609 genes, and is pseudogene rich. Each occupies a different branch of the phylogenetic tree. Detailed analysis of 500-bp upstream of all functional genes has revealed several conserved binding sites, general and B cell-specific, as well as key differences between families. This complete and definitive assembly of the mouse Igh V region will facilitate detailed study of promoter function and large-scale mechanisms associated with V(D)J recombination including locus contraction and antisense intergenic transcription.


Assuntos
Cadeias Pesadas de Imunoglobulinas/genética , Região Variável de Imunoglobulina/genética , Animais , Sequência de Bases , Sítios de Ligação , Southern Blotting , Camundongos , Camundongos Endogâmicos C57BL , Família Multigênica/genética , Filogenia , Pseudogenes/genética , Análise de Sequência de DNA
11.
Genomics ; 80(2): 236-44, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12160738

RESUMO

Dosage compensation in mammals is accomplished by the transcriptional silencing of a single X chromosome in female cells, a process termed X inactivation. A cytogenetically defined region of the X chromosome, the X-inactivation center (Xic), is necessary in cis for this process. Although the precise nature of the Xic remains unknown, a key component, the Xist gene, has been shown to be essential for X inactivation. In XX somatic cells, Xist RNA is specifically transcribed from the inactive X chromosome, which is otherwise essentially heterochromatic. Previous studies aimed at defining the proximal limit of the Xic have indicated that it lies within 30 kb upstream of the Xist promoter. Here we describe a novel gene, Enox (expressed neighbor of Xist), that maps to an unmethylated CpG island 10 kb upstream of Xist. Enox transcripts are antisense relative to Xist, highly heterogeneous, and apparently noncoding. In female somatic tissue Enox partially escapes from X inactivation. We discuss the implications of these findings in relation to our understanding of the Xic.


Assuntos
Mecanismo Genético de Compensação de Dose , RNA não Traduzido/genética , Fatores de Transcrição/genética , Cromossomo X , Animais , Antígenos de Neoplasias/genética , Ilhas de CpG , DNA Complementar , Feminino , Camundongos , Reação em Cadeia da Polimerase , RNA/metabolismo , RNA Longo não Codificante
12.
Genes Dev ; 17(17): 2177-90, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12952890

RESUMO

The X-inactive-specific transcript (Xist) locus is a cis-acting switch that regulates X chromosome inactivation in mammals. Over recent years an important goal has been to understand how Xist is regulated at the initiation of X inactivation. Here we report the analysis of a series of targeted mutations at the 5' end of the Xist locus. A number of these mutations were found to cause preferential inactivation, to varying degrees, of the X chromosome bearing the targeted allele in XX heterozygotes. This phenotype is similar to that seen with mutations that ablate Tsix, an antisense RNA initiated 3' of Xist. Interestingly, each of the 5' mutations causing nonrandom X inactivation was found to exhibit ectopic sense transcription in embryonic stem (ES) cells. The level of ectopic transcription was seen to correlate with the degree of X inactivation skewing. Conversely, targeted mutations which did not affect randomness of X inactivation also did not exhibit ectopic sense transcription. These results indicate that X chromosome choice is determined by the balance of Xist sense and antisense transcription prior to the onset of random X inactivation.


Assuntos
Mecanismo Genético de Compensação de Dose , Regulação da Expressão Gênica/fisiologia , RNA não Traduzido/genética , RNA não Traduzido/fisiologia , Transcrição Gênica/fisiologia , Animais , Hibridização In Situ , Camundongos , Mutagênese , Regiões Promotoras Genéticas , RNA Longo não Codificante , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
13.
Nat Immunol ; 5(6): 630-7, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15107847

RESUMO

Antigen receptor genes undergo variable, diversity and joining (V(D)J) recombination, which requires ordered large-scale chromatin remodeling. Here we show that antisense transcription, both genic and intergenic, occurs extensively in the V region of the immunoglobulin heavy chain locus. RNA fluorescence in situ hybridization demonstrates antisense transcription is strictly developmentally regulated and is initiated during the transition from DJ(H) to VDJ(H) recombination and terminates concomitantly with VDJ(H) recombination. Our data show antisense transcription is specific to the V region and suggest transcripts extend across several genes. We propose that antisense transcription remodels the V region to facilitate V(H)-to-DJ(H) recombination. These findings have wider implications for V(D)J recombination of other antigen receptor loci and developmental regulation of multigene loci.


Assuntos
DNA Antissenso/metabolismo , DNA Intergênico/metabolismo , Rearranjo Gênico/fisiologia , Genes de Imunoglobulinas/fisiologia , Transcrição Gênica/fisiologia , Animais , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Hibridização in Situ Fluorescente , Camundongos , Camundongos Endogâmicos C57BL , RNA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA