Methods for regulating T cell subsets by modulating transcription factor activity

a transcription factor and activity-modulating technology, applied in the direction of peptides, drug compositions, immunological disorders, etc., can solve the problems of unwanted side effects of cytokines or antibodies, and achieve the effects of stimulating expression or activity, modulating the activity of transcription factors, and enhancing the expression or activity of transcription facto

Inactive Publication Date: 2008-02-14
PRESIDENT & FELLOWS OF HARVARD COLLEGE
View PDF0 Cites 0 Cited by
  • Summary
  • Abstract
  • Description
  • Claims
  • Application Information

AI Technical Summary

Benefits of technology

[0010] The modulatory methods of the invention generally involve contacting a cell with an agent that modulates the expression or activity of a transcription factor(s) such that production of the Th2-associated cytokine by a cell is modulated. In particular, preferred agents of the invention act intracellularly to modulate the activity of the transcription factor. In one embodiment, the modulatory method of the invention stimulates production of a Th2-associated cytokine. For example, Th2-associated cytokine production can be stimulated in Th1 cells, B cells or non-lymphoid cells. In another embodiment, the modulatory method of the invention inhibits production of a Th2-associated cytokine. A Th2-associated cytokine modulated in the method preferably is interleukin-4.
[0011] A variety of agents can be used to stimulate the expression or activity of a transcription factor that regulates expression of a Th2-associated cytokine gene. For example, a stimulatory agent of the invention can be a nucleic acid molecule encoding the transcription factor that is introduced into and expressed in the cell. Alternatively, chemical agents that enhance the expression or activity of the transcription facto can be used as stimulatory agents.
[0012] A variety of agents can be used to inhibit the expression or activity of a transcription factor that regulates expression of a Th2-associated cytokine gene. Examples of suitable inhibitory agents include antisense nucleic acid molecules that are complementary to a gene encoding the transcription factor, intracellular antibodies that bind the transcription factor (e.g., in the cell nucleus), inhibitory forms of the transcription factor (e.g., dominant-negative forms) and chemical agents that inhibit the expression or activity of the transcription factor.
[0013] Combination methods, involving modulation of the expression or activity of two, three or more transcription factors that regulate Th2-associated cytokine gene expression, are also encompassed by the invention. Accordingly, in other embodiments of the invention, a cell is contacted with at least one additional agent that modulates the activity of at least one additional transcription factor that contributes to the regulation of the Th2-associated cytokine gene. Preferably, the at least one additional transcription factor whose expression or activity is modulated is selected from the group consisting of NF-AT family proteins, NF-AT-interacting proteins, maf family proteins and AP-1 family proteins.
[0014] Cytokine production by a cell can be modulated in vitro or in vivo in accordance with the methods of the invention. In one embodiment, a cell is contacted with a modulating agent(s) in vitro and then the cell is administered to a subject to thereby regulate the development of Th1 and / or Th2 subsets in the subject. Accordingly, in another aspect, the invention provides methods for regulating the development of Th1 or Th2 subsets in a subject. In addition to the embodiment wherein ex vivo modified cells are administered to the subject, in another embodiment, these methods involve direct administration to the subject of an agent that modulates the activity of one or more transcription factors that regulate expression of a Th2-associated cytokine gene such that development of Th1 or Th2 cells in the subject is modulated.
[0015] The modulatory methods of the invention can be used to manipulate Th1:Th2 ratios in a variety of clinical situations. For example, inhibition of Th2 formation may be useful in allergic diseases, malignancies and infectious diseases whereas enhancement of Th2 formation may be useful in autoimmune diseases and organ transplantation.

Problems solved by technology

However, as therapeutic options, systemic administration of cytokines or antibodies may have unwanted side effects and, accordingly, alternative approaches to manipulating Th1 / Th2 subsets are still needed.
However, none of these AP-1 proteins is expressed in a Th1- or Th2-specific manner and there is no evidence for the differential recruitment of AP-1 family members to the IL-2 or IL-4 composite sites (Rooney, J. et al.

Method used

the structure of the environmentally friendly knitted fabric provided by the present invention; figure 2 Flow chart of the yarn wrapping machine for environmentally friendly knitted fabrics and storage devices; image 3 Is the parameter map of the yarn covering machine
View more

Image

Smart Image Click on the blue labels to locate them in the text.
Viewing Examples
Smart Image
  • Methods for regulating T cell subsets by modulating transcription factor activity
  • Methods for regulating T cell subsets by modulating transcription factor activity
  • Methods for regulating T cell subsets by modulating transcription factor activity

Examples

Experimental program
Comparison scheme
Effect test

example 1

Cytokine Specificity is Due to a Positive Transacting Factor and not to a Repressor

[0205] Tissue specificity can be achieved through the action of repressor or silencer proteins. Thus it was possible that the IL-2 and IL-4 genes were actively repressed in Th2 and Th1 cells respectively. To test for the existence of repressor proteins, somatic cell fusions were performed between a Th1(D1.1) and a Th2 (D 10) clone of differing MHC Class I haplotypes. The Th1 clone D1.1 (Kd) and the Th2 clone D10 (Kk) were fused according to the “suspension cell fusion” procedure (Lane, R. D. et al. (1986) Methods Enzymol. 121:183-192). After fusion, the cells were allowed to recover for 8 hours and then double-stained using PE-conjugated anti-Kk and FITC-conjugated anti-Kd antibodies (Pharmingen, La Jolla, Calif.). Cells were then sorted on the basis of size to distinguish unfused cells from hetero and homokaryons and by fluorescence to identify single-positive and double-positive cells. As indicated...

example 2

Isolation of a Th2-Specific c-maf Gene from a cDNA Library Prepared from an Anti-CD3 Activated Th2 Clone

[0208] In the course of screening a cDNA library prepared from an anti-CD3 activated Th2 clone, D10, for NF-AT-interacting proteins by the yeast two-hybrid system (for descriptions of this system, see e.g., Field U.S. Pat. No. 5,283,173; Zervos et al. (1993) Cell 72:223-232; Madura et al. (1993) J. Biol. Chem. 268:12046-12054; Bartel et al. (1993) Biotechniques 14:920-924; and Iwabuchi et al. (1993) Oncogene 8:1693-1696), multiple cDNAs were isolated, all of which were extremely weak interactors. All cDNAs obtained in this screen were next evaluated for Th-specific expression by Northern blot analysis using a panel of Th1 and Th2 clones. One such cDNA, which was repeatedly isolated (60 of 140) detected transcripts only in RNA prepared from Th2 clones (D10, CDC35) and not from either Th1 clones (AR5, OS6, D1) or from a B cell lymphoma, M12, as illustrated in the Northern blot anal...

example 3

Ectopic Expression of c-Maf in Th1 and B Cells Results in Activation of the IL-4 Promoter

[0213] The identification of the isolated cDNA described in Example 2 as a member of the AP-1 / CREB / ATF gene family, together with its selective expression in Th2 cells raised the possibility that c-Maf controlled the tissue-specific transcription of the IL-4 gene. Additionally, the presence of transcripts encoding c-maf correlated well with IL-4 expression in Th2 cells and in three of four transformed mast cell lines examined. To test whether c-Maf could transactivate the IL-4 promoter, cotransfection experiments were performed.

[0214] Th1 clones and the B lymphoma M12.4.C3 (M12) neither express c-maf nor transcribe the IL-4 gene. If c-Maf is the transcription factor critical for controlling IL-4 gene expression, then forced expression in these cells should permit IL-4 gene expression. To test this, the full-length (4.3 kb) c-maf cDNA clone was inserted into the SalI site of the pMex-NeoI mamma...

the structure of the environmentally friendly knitted fabric provided by the present invention; figure 2 Flow chart of the yarn wrapping machine for environmentally friendly knitted fabrics and storage devices; image 3 Is the parameter map of the yarn covering machine
Login to view more

PUM

PropertyMeasurementUnit
densityaaaaaaaaaa
timeaaaaaaaaaa
pHaaaaaaaaaa
Login to view more

Abstract

Methods for modulating production of a T helper type 2 (Th2)-associated cytokine, in particular interleukin-4, by modulating the activity of one or more transcription factors that cooperate with NF-AT family proteins to regulate expression of a Th2-associated cytokine gene are disclosed. In one embodiment, the activity of a maf family protein (e.g., c-Maf or a small maf protein, such as p18) is modulated. In another embodiment, the activity of a protein that interacts with an NF-AT family protein (e.g., NIP45) is modulated. Combination methods, for example wherein the activities of a maf family protein and an NF-AT protein are modulated or the activities of a maf protein and NF-AT-interacting protein are modulated, are also encompassed by the invention. Methods for modulating development of T helper type 1 (Th1) or T helper type 2 (Th2) subsets in a subject using agents that modulate transcription factor activity are also disclosed.

Description

RELATED APPLICATIONS [0001] This application is a continuation application of U.S. Ser. No. 10 / 236,629, entitled “Methods and Compositions for Regulating T cell Subsets by Modulating Transcription Factor Activity”, filed Sep. 5, 2002, which is a continuation application of U.S. Ser. No. 08 / 755,592, entitled “Methods and Compositions for Regulating T cell Subsets by Modulating Transcription Factor Activity”, filed Nov. 25, 1996, which is a continuation-in-part of U.S. Ser. No. 08 / 636,602, entitled “Methods and Compositions for Regulating T cell Subsets by Modulating Transcription Factor Activity”, filed Apr. 23, 1996, the entire contents of which are expressly incorporated herein by reference. This application is also related to U.S. Ser. No. 08 / 755,584, entitled “NF-AT Interacting Protein NIP45 and Methods of Use Therefor”, filed Nov. 25, 1996 (Attorney Docket No. HUI-026), the entire contents of which are expressly incorporated herein by reference.GOVERNMENT FUNDING [0002] Work des...

Claims

the structure of the environmentally friendly knitted fabric provided by the present invention; figure 2 Flow chart of the yarn wrapping machine for environmentally friendly knitted fabrics and storage devices; image 3 Is the parameter map of the yarn covering machine
Login to view more

Application Information

Patent Timeline
no application Login to view more
Patent Type & Authority Applications(United States)
IPC IPC(8): A61K35/12A61P37/02C12N15/85C12N5/02A61K38/00C07K14/47C07K14/52C07K14/54C07K14/82C12N15/12
CPCA01K2217/05A61K38/00C07K14/4702G01N33/505C07K14/5406C07K14/82C07K2319/00C07K14/52A61P37/02
Inventor GLIMCHER, LAURIE H.HO, I-CHENG
Owner PRESIDENT & FELLOWS OF HARVARD COLLEGE
Who we serve
  • R&D Engineer
  • R&D Manager
  • IP Professional
Why Eureka
  • Industry Leading Data Capabilities
  • Powerful AI technology
  • Patent DNA Extraction
Social media
Try Eureka
PatSnap group products