Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(7)2023 Mar 30.
Article in English | MEDLINE | ID: mdl-37047449

ABSTRACT

Our laboratory has identified and developed a unique human-engineered domain (HED) structure that was obtained from the human Alpha-2-macroglobulin receptor-associated protein based on the three-dimensional structure of the Z-domain derived from Staphylococcal protein A. This HED retains µM binding activity to the human IgG1CH2-CH3 elbow region. We determined the crystal structure of HED in association with IgG1's Fc. This demonstrated that HED preserves the same three-bundle helix structure and Fc-interacting residues as the Z domain. HED was fused to the single chain variable fragment (scFv) of mAb 4D5 to produce an antibody-like protein capable of interacting with the p185Her2/neu ectodomain and the Fc of IgG. When further fused with murine IFN-γ (mIFN-γ) at the carboxy terminus, the novel species exhibited antitumor efficacy in vivo in a mouse model of human breast cancer. The HED is a novel platform for the therapeutic utilization of engineered proteins to alleviate human disease.


Subject(s)
Breast Neoplasms , Single-Chain Antibodies , Humans , Animals , Mice , Female , Single-Chain Antibodies/genetics , Staphylococcal Protein A/chemistry
2.
Am J Respir Crit Care Med ; 204(9): 1060-1074, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34346860

ABSTRACT

Rationale: Primary graft dysfunction (PGD) is a severe form of acute lung injury, leading to increased early morbidity and mortality after lung transplant. Obesity is a major health problem, and recipient obesity is one of the most significant risk factors for developing PGD. Objectives: We hypothesized that T-regulatory cells (Tregs) are able to dampen early ischemia-reperfusion events and thereby decrease the risk of PGD, whereas that action is impaired in obese recipients. Methods: We evaluated Tregs, T cells, and inflammatory markers, plus clinical data, in 79 lung transplant recipients and 41 liver or kidney transplant recipients and studied two groups of mice on a high-fat diet (HFD), which did ("inflammatory" HFD) or did not ("healthy" HFD) develop low-grade inflammation with decreased Treg function. Measurements and Main Results: We identified increased levels of IL-18 as a previously unrecognized mechanism that impairs Tregs' suppressive function in obese individuals. IL-18 decreases levels of FOXP3, the key Treg transcription factor, decreases FOXP3 di- and oligomerization, and increases the ubiquitination and proteasomal degradation of FOXP3. IL-18-treated Tregs or Tregs from obese mice fail to control PGD, whereas IL-18 inhibition ameliorates lung inflammation. The IL-18-driven impairment in Tregs' suppressive function before transplant was associated with an increased risk and severity of PGD in clinical lung transplant recipients. Conclusions: Obesity-related IL-18 induces Treg dysfunction that may contribute to the pathogenesis of PGD. Evaluation of Tregs' suppressive function together with evaluation of IL-18 levels may serve as a screening tool to identify obese individuals with an increased risk of PGD before transplant.


Subject(s)
Acute Lung Injury/etiology , Interleukin-18/metabolism , Lung Transplantation/adverse effects , Obesity/complications , Primary Graft Dysfunction/etiology , Reperfusion Injury/etiology , T-Lymphocytes, Regulatory/metabolism , Acute Lung Injury/physiopathology , Adult , Aged , Aged, 80 and over , Animals , Female , Humans , Male , Mice , Mice, Obese , Middle Aged , Primary Graft Dysfunction/physiopathology , Reperfusion Injury/physiopathology
3.
Front Immunol ; 12: 703632, 2021.
Article in English | MEDLINE | ID: mdl-34290714

ABSTRACT

The Mads/Mef2 (Mef2a/b/c/d) family of transcription factors (TFs) regulates differentiation of muscle cells, neurons and hematopoietic cells. By functioning in physiological feedback loops, Mef2 TFs promote the transcription of their repressor, Hdac9, thereby providing temporal control of Mef2-driven differentiation. Disruption of this feedback is associated with the development of various pathologic states, including cancer. Beside their direct involvement in oncogenesis, Mef2 TFs indirectly control tumor progression by regulating antitumor immunity. We recently reported that in CD4+CD25+Foxp3+ T-regulatory (Treg) cells, Mef2d is required for the acquisition of an effector Treg (eTreg) phenotype and for the activation of an epigenetic program that suppresses the anti-tumor immune responses of conventional T and B cells. We now report that as with Mef2d, the deletion of Mef2c in Tregs switches off the expression of Il10 and Icos and leads to enhanced antitumor immunity in syngeneic models of lung cancer. Mechanistically, Mef2c does not directly bind the regulatory elements of Icos and Il10, but its loss-of-function in Tregs induces the expression of the transcriptional repressor, Hdac9. As a consequence, Mef2d, the more abundant member of the Mef2 family, is converted by Hdac9 into a transcriptional repressor on these loci. This leads to the impairment of Treg suppressive properties in vivo and to enhanced anti-cancer immunity. These data further highlight the central role played by the Mef2/Hdac9 axis in the regulation of CD4+Foxp3+ Treg function and adds a new level of complexity to the analysis and study of Treg biology.


Subject(s)
Histone Deacetylases/immunology , Immune Tolerance , Lung Neoplasms/immunology , Neoplasms, Experimental/immunology , Repressor Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Histone Deacetylases/genetics , Immunity, Cellular , Lung Neoplasms/genetics , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasms, Experimental/genetics , Repressor Proteins/genetics
4.
Sci Rep ; 11(1): 9018, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33907245

ABSTRACT

Histone/protein deacetylases (HDAC) 1 and 2 are typically viewed as structurally and functionally similar enzymes present within various co-regulatory complexes. We tested differential effects of these isoforms in renal ischemia reperfusion injury (IRI) using inducible knockout mice and found no significant change in ischemic tolerance with HDAC1 deletion, but mitigation of ischemic injury with HDAC2 deletion. Restriction of HDAC2 deletion to the kidney via transplantation or PAX8-controlled proximal renal tubule-specific Cre resulted in renal IRI protection. Pharmacologic inhibition of HDAC2 increased histone acetylation in the kidney but did not extend renal protection. Protein analysis demonstrated increased HDAC1-associated CoREST protein in HDAC2-/- versus WT cells, suggesting that in the absence of HDAC2, increased CoREST complex occupancy of HDAC1 can stabilize this complex. In vivo administration of a CoREST inhibitor exacerbated renal injury in WT mice and eliminated the benefit of HDAC2 deletion. Gene expression analysis of endothelin showed decreased endothelin levels in HDAC2 deletion. These data demonstrate that contrasting effects of HDAC1 and 2 on CoREST complex stability within renal tubules can affect outcomes of renal IRI and implicate endothelin as a potential downstream mediator.


Subject(s)
Co-Repressor Proteins/metabolism , Histone Deacetylase 2/metabolism , Kidney Tubules, Proximal/metabolism , Reperfusion Injury/prevention & control , Animals , Co-Repressor Proteins/antagonists & inhibitors , Endothelins/metabolism , Enzyme Inhibitors/pharmacology , Female , Gene Deletion , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 2/genetics , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Kidney Tubules, Proximal/drug effects , Male , Mice , Mice, Knockout
5.
J Clin Invest ; 130(12): 6242-6260, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32790649

ABSTRACT

The transcription factor MEF2D is important in the regulation of differentiation and adaptive responses in many cell types. We found that among T cells, MEF2D gained new functions in Foxp3+ T regulatory (Treg) cells due to its interactions with the transcription factor Foxp3 and its release from canonical partners, like histone/protein deacetylases. Though not necessary for the generation and maintenance of Tregs, MEF2D was required for the expression of IL-10, CTLA4, and Icos, and for the acquisition of an effector Treg phenotype. At these loci, MEF2D acted both synergistically and additively to Foxp3, and downstream of Blimp1. Mice with the conditional deletion in Tregs of the gene encoding MEF2D were unable to maintain long-term allograft survival despite costimulation blockade, had enhanced antitumor immunity in syngeneic models, but displayed only minor evidence of autoimmunity when maintained under normal conditions. The role played by MEF2D in sustaining effector Foxp3+ Treg functions without abrogating their basal actions suggests its suitability for drug discovery efforts in cancer therapy.


Subject(s)
Graft Survival/immunology , Heart Transplantation , Lymphocyte Activation , Neoplasms, Experimental/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Graft Survival/genetics , HEK293 Cells , Humans , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasms, Experimental/genetics , T-Lymphocytes, Regulatory/pathology , Transplantation, Isogeneic
6.
Sci Rep ; 10(1): 9292, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32518311

ABSTRACT

Vascularized composite allotransplantation (VCA) allows tissue replacement after devastating loss but is currently limited in application and may be more widely performed if maintenance immunosuppression was not essential for graft acceptance. We tested whether peri-transplant costimulation blockade could prolong VCA survival and required donor bone-marrow cells, given that bone-marrow might promote graft immunogenicity or graft-versus-host disease. Peritransplant CD154 mAb/rapamycin (RPM) induced long-term orthotopic hindlimb VCA survival (BALB/c->C57BL/6), as did CTLA4Ig/RPM. Surprisingly, success of either protocol required a bone-marrow-associated, radiation-sensitive cell population, since long-bone removal or pre-transplant donor irradiation prevented long-term engraftment. Rejection also occurred if Rag1-/- donors were used, or if donors were treated with a CXCR4 inhibitor to mobilize donor BM cells pre-transplant. Donor bone-marrow contained a large population of Foxp3+ T-regulatory (Treg) cells, and donor Foxp3+ Treg depletion, by diphtheria toxin administration to DEREG donor mice whose Foxp3+ Treg cells expressed diphtheria toxin receptor, restored rejection with either protocol. Rejection also occurred if CXCR4 was deleted from donor Tregs pre-transplant. Hence, long-term VCA survival is possible across a full MHC disparity using peritransplant costimulation blockade-based approaches, but unexpectedly, the efficacy of costimulation blockade requires the presence of a radiation-sensitive, CXCR4+ Foxp3+ Treg population resident within donor BM.


Subject(s)
Bone Marrow Transplantation , Extremities/transplantation , Graft Survival/physiology , T-Lymphocytes, Regulatory/immunology , Vascularized Composite Allotransplantation/methods , Abatacept/pharmacology , Animals , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , CD40 Ligand/immunology , Diphtheria Toxin/pharmacology , Forkhead Transcription Factors/metabolism , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft vs Host Disease/pathology , Graft vs Host Disease/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Sirolimus/pharmacology
7.
Autophagy ; 13(12): 2056-2071, 2017.
Article in English | MEDLINE | ID: mdl-28981387

ABSTRACT

Lysosomal autophagy inhibitors (LAI) such as hydroxychloroquine (HCQ) have significant activity in a subset of cancer cell lines. LAIs are being evaluated in cancer clinical trials, but genetic determinants of sensitivity to LAIs are unknown, making it difficult to predict which tumors would be most susceptible. Here we characterize differentially expressed genes in HCQ-sensitive (-S) and -resistant (-R) cancer cells. Notably, expression of canonical macroautophagy/autophagy genes was not associated with sensitivity to HCQ. Expression patterns of ALDH1A1 (aldehyde dehydrogenase 1 family member A1) and HLTF (helicase like transcription factor) identified HCQ-S (ALDH1A1high HLTFlow; ALDH1A1low HLTFlow) and HCQ-R (ALDH1A1low HLTFhigh) cells. ALDH1A1 overexpression was found to enhance LAI cell entry and cytotoxicity without directly affecting lysosome function or autophagic flux. Expression of HLTF allows repair of DNA damage caused by LAI-induced reactive oxygen species, leading to HCQ resistance. Sensitivity to HCQ is increased in cells where HLTF is silenced by promoter methylation. HLTF overexpression blunted the antitumor efficacy of chloroquine derivatives in vitro and in vivo. Analysis of tumor RNA sequencing data from >700 patients in the Cancer Genome Atlas identified cancers including colon cancer, renal cell carcinoma, and gastric cancers, that were enriched for the HCQ-S or HCQ-R signature. These results provide mechanistic insights into LAI efficacy, and guidance for LAI clinical development.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Autophagy , DNA-Binding Proteins/metabolism , Lysosomes/metabolism , Transcription Factors/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Autophagy/drug effects , Cell Line, Tumor , Chloroquine/pharmacology , DNA Damage , Drug Resistance, Neoplasm/drug effects , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxychloroquine/pharmacology , Lysosomes/drug effects , Mice, Nude , Reactive Oxygen Species/metabolism , Reproducibility of Results , Retinal Dehydrogenase
8.
Sci Rep ; 7(1): 8626, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819166

ABSTRACT

Current interest in Foxp3+ T-regulatory (Treg) cells as therapeutic targets in transplantation is largely focused on their harvesting pre-transplant, expansion and infusion post-transplantation. An alternate strategy of pharmacologic modulation of Treg function using histone/protein deacetylase inhibitors (HDACi) may allow more titratable and longer-term dosing. However, the effects of broadly acting HDACi vary, such that HDAC isoform-selective targeting is likely required. We report data from mice with constitutive or conditional deletion of HDAC11 within Foxp3+ Treg cells, and their use, along with small molecule HDAC11 inhibitors, in allograft models. Global HDAC11 deletion had no effect on health or development, and compared to WT controls, Foxp3+ Tregs lacking HDAC11 showed increased suppressive function, and increased expression of Foxp3 and TGF-ß. Likewise, compared to WT recipients, conditional deletion of HDAC11 within Tregs led to long-term survival of fully MHC-mismatched cardiac allografts, and prevented development of transplant arteriosclerosis in an MHC class II-mismatched allograft model. The translational significance of HDAC11 targeting was shown by the ability of an HDAC11i to promote long-term allograft allografts in fully MHC-disparate strains. These data are powerful stimuli for the further development and testing of HDAC11-selective pharmacologic inhibitors, and may ultimately provide new therapies for transplantation and autoimmune diseases.


Subject(s)
Forkhead Transcription Factors/immunology , Histone Deacetylases/immunology , Histones/immunology , T-Lymphocytes, Regulatory/immunology , Allografts , Animals , Forkhead Transcription Factors/metabolism , Gene Expression Profiling/methods , Graft Survival/drug effects , Graft Survival/genetics , Graft Survival/immunology , HEK293 Cells , Heart Transplantation/methods , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocytes, Regulatory/metabolism
9.
EBioMedicine ; 13: 99-112, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27769803

ABSTRACT

Foxp3+ T-regulatory (Treg) cells are known to suppress protective host immune responses to a wide variety of solid tumors, but their therapeutic targeting is largely restricted to their transient depletion or "secondary" modulation, e.g. using anti-CTLA-4 monoclonal antibody. Our ongoing studies of the post-translational modifications that regulate Foxp3 demonstrated that the histone/protein acetyltransferase, Tip60, plays a dominant role in promoting acetylation, dimerization and function in Treg cells. We now show that the ubiquitin-specific protease, Usp7, controls Treg function largely by stabilizing the expression and promoting the multimerization of Tip60 and Foxp3. Genetic or pharmacologic targeting of Usp7 impairs Foxp3+ Treg suppressive functions, while conventional T cell responses remain intact. As a result, pharmacologic inhibitors of Usp7 can limit tumor growth in immunocompetent mice, and promote the efficacy of antitumor vaccines and immune checkpoint therapy with anti-PD1 monoclonal antibody in murine models. Hence, pharmacologic therapy with Usp7 inhibitors may have an important role in future cancer immunotherapy.


Subject(s)
Forkhead Transcription Factors/metabolism , Histone Acetyltransferases/metabolism , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Trans-Activators/metabolism , Ubiquitin-Specific Proteases/antagonists & inhibitors , Animals , Autoimmunity/genetics , Autoimmunity/immunology , Cell Line, Tumor , Disease Models, Animal , Gene Expression , Immunity , Lymphocyte Activation/immunology , Lysine Acetyltransferase 5 , Mice , Mice, Knockout , Mice, Transgenic , Neoplasms/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Burden , Ubiquitin-Specific Peptidase 7 , Ubiquitin-Specific Proteases/genetics
10.
J Biol Chem ; 290(33): 20211-20, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-25987564

ABSTRACT

Regulation of the extent of immune responses is a requirement to maintain self-tolerance and limit inflammatory processes. CD4(+)CD25(+)Foxp3(+) regulatory T (Treg) cells play a role in regulation. The Foxp3 transcription factor is considered a dominant regulator for Treg cell development and function. Foxp3 function itself is directly regulated by multiple posttranslational modifications that occur in response to various external stimuli. The Foxp3 protein is a component of several dynamic macromolecular regulatory complexes. The complexes change constituents over time and through different signals to regulate the development and function of regulatory T cells. Here we identified a mechanism regulating Foxp3 level and activity that operates through discrete phosphorylation. The Pim-2 kinase can phosphorylate Foxp3, leading to decreased suppressive functions of Treg cells. The amino-terminal domain of Foxp3 is modified at several sites by Pim-2 kinase. This modification leads to altered expression of proteins related to Treg cell functions and increased Treg cell lineage stability. Treg cell suppressive function can be up-regulated by either pharmacologically inhibiting Pim-2 kinase activity or by genetically knocking out Pim-2 in rodent Treg cells. Deficiency of Pim-2 activity increases murine host resistance to dextran sodium sulfate-induced colitis in vivo, and a Pim-2 small molecule kinase inhibitor also modified Treg cell functions. Our studies define a pathway for limiting the regulation of Foxp3 function because the Pim-2 kinase represents a potential therapeutic target for modulating the Treg cell suppressive activities in controlling immune responses.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , Amino Acid Sequence , Animals , Mice , Mice, Knockout , Molecular Sequence Data , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics
11.
Cell Rep ; 7(5): 1471-1480, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24835996

ABSTRACT

The human FOXP3 molecule is an oligomeric transcriptional factor able to mediate activities that characterize T regulatory cells, a class of lymphocytes central to the regulation of immune responses. The activity of FOXP3 is regulated at the posttranslational level, in part by two histone acetyltransferases (HATs): TIP60 and p300. TIP60 and p300 work cooperatively to regulate FOXP3 activity. Initially, p300 and TIP60 interactions lead to the activation of TIP60 and facilitate acetylation of K327 of TIP60, which functions as a molecular switch to allow TIP60 to change binding partners. Subsequently, p300 is released from this complex, and TIP60 interacts with and acetylates FOXP3. Maximal induction of FOXP3 activities is observed when both p300 and TIP60 are able to undergo cooperative interactions. Conditional knockout of TIP60 in Treg cells significantly decreases the Treg population in the peripheral immune organs, leading to a scurfy-like fatal autoimmune disease.


Subject(s)
E1A-Associated p300 Protein/metabolism , Forkhead Transcription Factors/metabolism , Histone Acetyltransferases/metabolism , Trans-Activators/metabolism , Amino Acid Sequence , Animals , Autoimmune Diseases/metabolism , E1A-Associated p300 Protein/genetics , Forkhead Transcription Factors/genetics , HEK293 Cells , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/genetics , Humans , Lysine/metabolism , Lysine Acetyltransferase 5 , Mice , Molecular Sequence Data , Mutation , Protein Binding , T-Lymphocytes, Regulatory/metabolism , Trans-Activators/chemistry , Trans-Activators/genetics
12.
Proc Natl Acad Sci U S A ; 109(21): 8253-8, 2012 May 22.
Article in English | MEDLINE | ID: mdl-22566612

ABSTRACT

Autophagy is a lysosome-dependent degradative process that protects cancer cells from multiple stresses. In preclinical models, autophagy inhibition with chloroquine (CQ) derivatives augments the efficacy of many anticancer therapies, but CQ has limited activity as a single agent. Clinical trials are underway combining anticancer agents with hydroxychloroquine (HCQ), but concentrations of HCQ required to inhibit autophagy are not consistently achievable in the clinic. We report the synthesis and characterization of bisaminoquinoline autophagy inhibitors that potently inhibit autophagy and impair tumor growth in vivo. The structural motifs that are necessary for improved autophagy inhibition compared with CQ include the presence of two aminoquinoline rings and a triamine linker and C-7 chlorine. The lead compound, Lys01, is a 10-fold more potent autophagy inhibitor than HCQ. Compared with HCQ, Lys05, a water-soluble salt of Lys01, more potently accumulates within and deacidifies the lysosome, resulting in impaired autophagy and tumor growth. At the highest dose administered, some mice develop Paneth cell dysfunction that resembles the intestinal phenotype of mice and humans with genetic defects in the autophagy gene ATG16L1, providing in vivo evidence that Lys05 targets autophagy. Unlike HCQ, significant single-agent antitumor activity is observed without toxicity in mice treated with lower doses of Lys05, establishing the therapeutic potential of this compound in cancer.


Subject(s)
Aminoquinolines/pharmacology , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Lysosomes/drug effects , Polyamines/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Aminoquinolines/chemical synthesis , Aminoquinolines/toxicity , Animals , Antimalarials/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Autophagy/genetics , Autophagy-Related Proteins , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Carrier Proteins/genetics , Cell Death/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm , Glioblastoma/genetics , Glioblastoma/pathology , HT29 Cells , Humans , Hydroxychloroquine/pharmacology , Intestinal Obstruction/chemically induced , Intestinal Obstruction/genetics , Mice , Mice, Nude , Polyamines/chemical synthesis , Polyamines/toxicity , Xenograft Model Antitumor Assays
13.
Hum Gene Ther ; 21(10): 1259-71, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20497038

ABSTRACT

Adeno-associated viral (AAV) manufacturing at scale continues to hinder the application of AAV technology to gene therapy studies. Although scalable systems based on AAV-adenovirus, AAV-herpesvirus, and AAV-baculovirus hybrids hold promise for clinical applications, they require time-consuming generation of reagents and are not highly suited to intermediate-scale preclinical studies in large animals, in which several combinations of serotype and genome may need to be tested. We observed that during production of many AAV serotypes, large amounts of vector are found in the culture supernatant, a relatively pure source of vector in comparison with cell-derived material. Here we describe a high-yielding, recombinant AAV production process based on polyethylenimine (PEI)-mediated transfection of HEK293 cells and iodixanol gradient centrifugation of concentrated culture supernatant. The entire process can be completed in 1 week and the steps involved are universal for a number of different AAV serotypes. Process conditions have been optimized such that final purified yields are routinely greater than 1 x 10(14) genome copies per run, with capsid protein purity exceeding 90%. Initial experiments with vectors produced by the new process demonstrate equivalent or better transduction both in vitro and in vivo when compared with small-scale, CsCl gradient-purified vectors. In addition, the iodixanol gradient purification process described effectively separates infectious particles from empty capsids, a desirable property for reducing toxicity and unwanted immune responses during preclinical studies.


Subject(s)
Dependovirus , Genetic Vectors , Polyethyleneimine , Adenoviridae/genetics , Baculoviridae/genetics , Capsid Proteins/genetics , Centrifugation, Density Gradient , Dependovirus/genetics , Dependovirus/growth & development , Dependovirus/isolation & purification , Genetic Therapy , HEK293 Cells , Helper Viruses/genetics , Herpesviridae/genetics , Humans , Transfection , Transgenes , Triiodobenzoic Acids , Virus Cultivation
14.
Proc Natl Acad Sci U S A ; 105(37): 14023-7, 2008 Sep 16.
Article in English | MEDLINE | ID: mdl-18779564

ABSTRACT

Expression of FOXP3, a potent gene-specific transcriptional repressor, in regulatory T cells is required to suppress autoreactive and alloreactive effector T cell function. Recent studies have shown that FOXP3 is an acetylated protein in a large nuclear complex and FOXP3 actively represses transcription by recruiting enzymatic corepressors, including histone modification enzymes. The mechanism by which extracellular stimuli regulate the FOXP3 complex ensemble is currently unknown. Although TGF-beta is known to induce murine FOXP3(+) Treg cells, TGF-beta in combination with IL-6 attenuates the induction of FOXP3 functional activities. Here we show that TCR stimuli and TGF-beta signals modulate the disposition of FOXP3 into different subnuclear compartments, leading to enhanced chromatin binding in human CD4(+)CD25(+) regulatory T cells. TGF-beta treatment increases the level of acetylated FOXP3 on chromatin and site-specific recruitment of FOXP3 on the human IL-2 promoter. However, the proinflammatory cytokine IL-6 down-regulates FOXP3 binding to chromatin in the presence of TGF-beta. Moreover, histone deacetylation inhibitor (HDACi) treatment abrogates the down-regulating effects of IL-6 and TGF-beta. These studies indicate that HDACi can enhance regulatory T cell function via promoting FOXP3 binding to chromatin even in a proinflammatory cellular microenvironment. Collectively, our data provide a framework of how different signals affect intranuclear redistribution, posttranslational modifications, and chromatin binding patterns of FOXP3.


Subject(s)
Chromatin/genetics , Forkhead Transcription Factors/metabolism , Interleukin-6/pharmacology , Promoter Regions, Genetic/genetics , Signal Transduction/drug effects , Signal Transduction/immunology , Transforming Growth Factor beta/pharmacology , Acetylation , Cells, Cultured , Down-Regulation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Humans , Protein Binding
15.
Curr Opin Immunol ; 19(5): 583-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17703930

ABSTRACT

While mutations in human FOXP3 predispose individuals to autoimmune conditions, it is unclear how the mutant protein fails to function as a transcriptional regulator. There is also limited detail of how FOXP3 itself interacts with the transcriptional machinery and which components of the FOXP3 ensembles exert phenotypic changes to render cells able to mediate suppression. Increasing evidence indicates that the level and duration of FOXP3 expression plays a crucial role in the development and function of natural regulatory T cells (Tregs). Our studies focus on the post-translational modification of the FOXP3 protein, and how the FOXP3 complex ensemble, containing histone modification and chromatin-remodeling enzymes, defines its functional role in regulatory T cells. Understanding the molecular mechanisms underlying FOXP3 activity will provide therapeutic implications for transplantation, allergy, autoimmune disease and cancer.


Subject(s)
Cytokines/metabolism , Forkhead Transcription Factors/metabolism , Immune Tolerance , T-Lymphocytes, Regulatory/immunology , Animals , Chromatin/metabolism , Cytokines/immunology , Gene Expression Regulation , Humans , T-Lymphocytes, Regulatory/metabolism , Transcription, Genetic
16.
Int Immunol ; 19(7): 825-35, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17586580

ABSTRACT

We have found that FOXP3 is an oligomeric component of a large supramolecular complex. Certain FOXP3 mutants with single amino acid deletions in the leucine zipper domain of FOXP3 are associated with the X-linked autoimmunity-allergic dysregulation (XLAAD) and immunodysregulation, polyendocrinopathy and enteropathy, X-linked (IPEX) syndrome in humans. We report that the single amino acid deletion found in human XLAAD/IPEX patients within the leucine zipper domain of FOXP3 does not disrupt its ability to join the larger protein complex, but eliminates FOXP3 homo-oligomerization as well as heteromerization with FOXP1. We found that the zinc finger-leucine zipper domain region of FOXP3 is sufficient to mediate both homodimerization and homotetramerization. However, the same domain region from XLAAD/IPEX FOXP3 containing an E251 deletion prevents oligomerizaton and the protein remains monomeric. We also found that wild-type FOXP3 directly binds to the human IL-2 promoter, but the E251 deletion in FOXP3 in XLAAD/IPEX patient's T cells disrupts its association with the IL-2 promoter in vivo and in vitro, and limits repression of IL-2 transcription after T-cell activation. Our results suggest that compromising FOXP3 homo-oligomerization and hetero-oligomerization with the FOXP1 protein impairs DNA-binding properties leading to distinct biochemical phenotypes in humans with the XLAAD/IPEX autoimmune syndrome. This study explains some features of the pathogenesis of a disease syndrome that arises as a consequence of specific assembly failure of a transcriptional repressor due to certain mutations within the FOXP3 leucine zipper.


Subject(s)
Forkhead Transcription Factors/genetics , Gene Deletion , Genetic Diseases, X-Linked/genetics , Immunologic Deficiency Syndromes/genetics , Cell Line , Forkhead Transcription Factors/metabolism , Humans , Interleukin-2/genetics , Interleukin-2/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Syndrome , T-Lymphocytes/immunology , Transcription, Genetic
17.
Proc Natl Acad Sci U S A ; 104(11): 4571-6, 2007 Mar 13.
Article in English | MEDLINE | ID: mdl-17360565

ABSTRACT

The forkhead family protein FOXP3 acts as a repressor of transcription and is both an essential and sufficient regulator of the development and function of regulatory T cells. The molecular mechanism by which FOXP3-mediated transcriptional repression occurs remains unclear. Here, we report that transcriptional repression by FOXP3 involves a histone acetyltransferase-deacetylase complex that includes histone acetyltransferase TIP60 (Tat-interactive protein, 60 kDa) and class II histone deacetylases HDAC7 and HDAC9. The N-terminal 106-190 aa of FOXP3 are required for TIP60-FOXP3, HDAC7-FOXP3 association, as well as for the transcriptional repression of FOXP3 via its forkhead domain. FOXP3 can be acetylated in primary human regulatory T cells, and TIP60 promotes FOXP3 acetylation in vivo. Overexpression of TIP60 but not its histone acetyltransferase-deficient mutant promotes, whereas knockdown of endogenous TIP60 relieved, FOXP3-mediated transcriptional repression. A minimum FOXP3 ensemble containing native TIP60 and HDAC7 is necessary for IL-2 production regulation in T cells. Moreover, FOXP3 association with HDAC9 is antagonized by T cell stimulation and can be restored by the protein deacetylation inhibitor trichostatin A, indicating a complex dynamic aspect of T suppressor cell regulation. These findings identify a previously uncharacterized complex-based mechanism by which FOXP3 actively mediates transcriptional repression.


Subject(s)
Forkhead Transcription Factors/metabolism , Histone Acetyltransferases/metabolism , CD4 Antigens/biosynthesis , Cell Nucleus/metabolism , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/biosynthesis , Lysine Acetyltransferase 5 , Models, Biological , Protein Binding , Protein Structure, Tertiary , Repressor Proteins/metabolism , Transcription, Genetic
18.
Immunol Rev ; 212: 99-113, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16903909

ABSTRACT

Our recent studies have identified dynamic protein ensembles containing forkhead box protein 3 (FOXP3) that provide insight into the molecular complexity of suppressor T-cell activities, and it is our goal to determine how these ensembles regulate FOXP3's transcriptional activity in vivo. In this review, we summarize our current understanding of how FOXP3 expression is induced and how FOXP3 functions in vivo as a transcriptional regulator by assembling a multisubunit complex involved in histone modification as well as chromatin remodeling.


Subject(s)
Chromatin Assembly and Disassembly , Forkhead Transcription Factors/metabolism , Histones/metabolism , T-Lymphocytes, Regulatory/immunology , Transcription, Genetic , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/therapy , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/genetics , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Humans , Immunotherapy , Mice , T-Lymphocytes, Regulatory/chemistry , T-Lymphocytes, Regulatory/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...