CA2000695A1 - Cytokine - Google Patents

Cytokine

Info

Publication number
CA2000695A1
CA2000695A1 CA 2000695 CA2000695A CA2000695A1 CA 2000695 A1 CA2000695 A1 CA 2000695A1 CA 2000695 CA2000695 CA 2000695 CA 2000695 A CA2000695 A CA 2000695A CA 2000695 A1 CA2000695 A1 CA 2000695A1
Authority
CA
Canada
Prior art keywords
cells
monokine
hours
factor
macrophage
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA 2000695
Other languages
French (fr)
Inventor
Nancy M. Hedberg
Neil Hunter
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Sydney
Original Assignee
University of Sydney
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Sydney filed Critical University of Sydney
Publication of CA2000695A1 publication Critical patent/CA2000695A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2833Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against MHC-molecules, e.g. HLA-molecules
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Biotechnology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

CYTOKINE
ABSTRACT

A monokine which is released by macrophages in response to stimulation with lipopolysaccharide, a method of producing the monokine, a method of bioassay of the monokine, and a factor which is produced by thymic epithelial cells in response to stimulation with the monokine. The monokine is distinct from macrophage - derived factors such as IL-1 and TNF.

Description

CYTORIWE
This invention relates to cytokines, and in particular to a polypeptide factor derived from macrophages.

Background and Prior Art Publications referred to herein are identified 1~ in full at the end of this specification.
It is well recognized that macrophages play an essential role in immune and inflammatory processes, both at a cellular level and via the release of mediators such ,;, ; ~, - ;, , . - , ~

~,, ~ . , , as interleukin-l (IL-l), colony stimulating factor ~CSF) and tumour necrosis factor (TNF) (Dinarello, 1984;
Burgess and Metcalf, 1980; Le and Vilcek, 1987).
The thymus is a key organ in the generation and 5 maturation of lymphocytes in mammals, particularly during foetal and neonatal life. Processing by the thymus is required for the production of T lymphocytes, of which various subsets are required for specific types of immune response. These subsets, such as helper T cells, 10 suppressor T cells, cytotoxic T cells etc. may be identified by the specific antigens present on their cell surfaces.
Thymus cell suspensions may be divided into light and dense cell populations by fractionation in 15 density gradients of Percoll (Salisbury et. al., 1979;
Percoll is a trade mark of Pharmacia AB).
In rats, the dense cell fraction comprises immature thymocytes which are unable to respond to Concanavalin A, and thymic epithelial cells, which 20 produce keratin.
The viability of dense, immature rat thymocytes was found by the present inventors to decrease markedly when a suspension of the cells in complete nutrient medium was incubated in vitro over a four hour period.
This was attributed to the phenomenon of programmed cell death, or apoptosis.
It has been estimated that approximately 90% of the cells which are generated within the thymus die in situ (Kinnon et. al, 1986). This population of 30 thymocytes is derived from the thymic cortex, and the cells are characterized by their high density and susceptibility to cortisone-mediated apoptotic death (Weissman, 1986). The essential role of thymic epithelial cells in thymocyte maturation is well 35 documented by studies in nude mice and rats in which the ectodermally derived epithelial cells have failed to , :: ~ ' ' ~.'' ' ~ .' .,. ~
. ~ .

:

develop (Douglas-Jones et. al., 1981). In the cortex of the normai thymus, the majority of the stromal cellq are epithelial in nature, and have long processes which reticulate among the cortical thymoctyes. In addition, 5 the cortex contains a small number of macrophages which are primarily located in the region adjacent to the medulla (Adkins et. al., 1981).
The epithelial cells in both the cortex and the medulla express both class I and class II
10 histocompatibility antigens (Van Ejwik et. al., 1980) and are thought to induce or select for self class II antigen recognition by thymo~ytes (Berrih et. al., 1985). Thymic epithelial cells have also been shown to be the source of thymic hormones which induce the appearance of maturation 15 markers on the thymocytes (Berrih et. al., 1985). More recently, the epithelial cells have also been shown to be a source of IL-l (Le et. al., 1987).
We have now surprisingly found that a factor released by macrophages in response to stimulation with 20 lipopolysaccharide is able to protect thymocytes from apoptotic death.
The factor appears to stimulate immature, non-proliferating thymocytes to differentiate to a stage at which they are able to survive.
Without wishing to be bound by any proposed mechanism for the observed beneficial effect, it is thought that the macrophage-derived factor binds to a common determinant of the Ia antigen complex on thymic epithelial cells.
This in turn stimulates the thymic epithelial cells to release a factor which promotes survival of thymocytes.
~ The macrophage-derived factor is clearly different in its biochemical and functional properties 35 from previously known factors such as tumour necrosis factor and interleukin-l.

.~

. . . ~., : , ~'s :. i - i :
> , ~ .- ~ ., :
, ....~ .
:,~;:: . : - :

~`

Summary of the Invention According to one aspect of the present invention there is provided a monokine released by macrophages in response to stimulation with 5 lipopolysaccharide, said monokine having the following properties:
Relative molecular weight 36000 kD;
Isoelectric point 6.3 - 6.4;
Stable at temperatures up to 70C;
Stable at pH 2 to 10;
Activity destroyed by: reduction with 2-mercaptoethanol treatment with proteinases, or treatment with urea;
Does not stimulate proliferation of fibroblasts; and Binds to common determinant of Ia antigen on thymic epithelial cells.
According to a second aspect of the invention, there is provided a method of producing said monokine, 20 comprising the steps of incubating macrophages in nutrient medium in the presence of bacterial lipopolysaccharide or-muramyl depeptide for 5 minutes to 2 hours, and recovering the monokine.
Preferably lipopolysaccharide is used to 25 stimulate monokine production.
Preferably incubation is for 2 hours.
Preferably recovering the monokine comprises the steps of:
recovering medium from macrophage cultures, removing lipopolysaccharide or muramyl dipeptide, removing material of Mr less than about 20,000, subjecting the remainder to sequential steps of gel filtration, ion exchange chromatography, and hydrophobic 35 interaction chromatography, and recovering fractions having monokine activity.

'`F~

,;, . . .
..: -. ~: ' ,.
~, .
., ~ .

Further purification may be effected by methods such as preparative gel electrophoresis and/or high performance liquid chromatography, which are well known to persons skilled in the art.
According to a third aspect of the invention there is provided a factor produced by thymic epithelial cells in response to stimulation with said monokine, said factor having the following properties:
Relative molecular weight 320,000;
Activity destroyed by trypsin treatment or by boiling;
Does not require active protein synthesis for production;
Binds to immature thymocytes;
Protects immature thymocytes from apoptotic death; and Binding to thymocytes inhibited by preincubation with antibody to CD4 antigen.
According to a fourth aspect of the invention, 20 there is provided a method of bioassay of the monokine comprising the steps of:
(a) adding a sample of fluid containing or suspec~ed of containing monokine to either dense thymic cells, or whole thymus suspension depleted of adherent 25 cells, in nutrient medium in the absence of serum and indicator dye for a period of 3 to 16 hours, preferably 3 to 7 hours, ;
(b) adding MTT (3-(4,5-dimethylthiazol-2-yl)- -~
2,5-diphenyl tetrazolium bromide) (c) incubating for 1 hour at 37C, (d) removing untransformed MTT, (e) adding isopropanol to the samples, (f) estimating extinction at 540 or 560 nm, and (g) calculating the amount of monokine in the sample.

~, ....... . ..

.: ~ -: .. , :.
:.: .~, ' ~ - ' , ~ -... ~ .... : . . . .

Preferably the assay is carried out in 96 well microtitre piates, and each well contain~ 0.5 to 5 x 106, more preferably 2 x 106, dense thymic cells in 100 ~1 medium, suitably RPMl 1640 medium containing no phenol 5 red or serum.
Preferably 100 ,ul of sample to be assayed is used per 2 x 107 cells.
The preferred incubation time before adding MTT
is 3 hours if adherent cells are not first depleted, and 10 7 hours if adherent cells have been depleted.

Detailed Description of the Invention The invention will now be described in detail by way of reference only to the following non-limiting examples, and to the accompanying drawings, in which:
Fig. 1 shows viability of the dense thymic cells, alone or in the presence of stimulated macrophage supernatant. Percentage cell viability is shown as a function of time. Results are the mean + s.e.m. of 25 experiments. By Student's t-test, *P c0.05 and 20 **P~ 0.01;
Fig. 2 shows results of molecular weight and isoelectric point measurement of the monokine (a) Molecular weight determination. Sephacryl S200 column run. A column (100 cm x 1~6 cm) calibrated with 25 Combithek II proteins in DPBS with 0.1 mol/l NaCl was run with a 5 ml sample under identical total conditions.
Flow rate 10.8 ml/h, sensitivity x 0.1, chart speed 2 cm/h., run time 17 hours. All of the fractions were tested and MF activity was detected in only one (O.8 ml) 30 fraction. Hatched area indicates activity. (b) Iso-electric point. LKB broad range ampholine gel (pH
3-9). Initial run, activity localized between 6 and 7 with reference to a standard curve. Second run, samples applied as a 10 cm band with standard proteins at either 35 end. Standard curves were identical and 2 mm strips of t .~. ~ ," ~ . , .

':

, ~ ~ ' .'., ` :
: ', ` .

gel were taken between pH 6 and 7 and the activity was localized to one strip: pH 6.3 - 6.4. Hatched area indicates activity;
Fig. 3 shows the production of the macrophage 5 factor in response to varying levels of two different stimuli. Production was determined by the titre resulting in 100% protection in the thymocyte viability assay. No protection refers to anything less than 100%
protection. The supernatant was collected at three 10 different times as shown. Results are the mean of four experiments;
Fig. 4 shows the time course for the production of the macrophage factor. Supernatants from macrophages stimulated with 20 ,ug/ml LPS were collected at various 15 time intervals after stimulation as shown. Production was expressed as the titre of macrophage factor resulting in 100% protection in the thymocyte viability assay. Any protection less than total was referred to as no protection. Representative experiment of two 20 experiments;
Fig. 5 shows the effect of protein synthesis inhibitors on the production of the macrophage factor.
Three protein synthesis inhibitors, actinomycin D (1 ~g/ml), cycloheximide (10 ,ug/ml) or puromycin (100 ~ug/ml) 25 were added to macrophage cultures simultaneously with LPS
(20 ~g/ml) and supernatants were collected at six intervals between five minutes and two hours for testing in the thymocyte viability assay. Protective activity of the macrophage factor at the various times is indicated 30 by the percentage viability of the thymyocytes at three hours. Results are the mean of three experiments;
Fig. 6 shows the effect of control supernatants on the production of the macrophage factor. Three control supernatants, lysed macrophage supernatant, 35 trypsin treated lysed macrophages and unstimulated macrophage supernatants were compared to macrophage - - ~., . .~ . .
, . . ~ ., . :~ .
- ~ . , . . - : . -... , . . ~. :: . :: :

supernatant from macrophages stimulated with 20 ~g/ml LPS. The percèntage viability of the thymocytes over four hours is shown. Results are the mean of five experiments;
Fig. 7 shows the effect of restimulation of macrophage cultures with LPS on the production of the macrophage factor. Supernatants were collected at two hourly intervals and cultures were restimulated with either LPS only (20 ~g/ml) or LPS (20 pg/ml) and 10 indomethacin (10 5M) in new medium. Production of the factor was expressed as the titre resulting in 100 protection in the thymocyte viability assay for supernatants collected at two, four, six and eight hours after initial stimulation;
Fig. 8 shows the role of the Ia molecule as a receptor for the monokine (representative experiments are shown). In every experiment, the unprotected populations were less than 70% of the control population by 4 hours incubation.
(a) Stimulated macrophage supernatant can be mimicked by a monoclonal antibody to Ia (MRC OX6). This effect is also mediated via an epithelial cell. Control DCF; DCF + 2.5 ~ug/ml MRC OX6 added (reproduced in four experiments): Ia-negative DCF with 2.5 ~g/ml MRC OX6 25 added (reproduced in three experiments); Ia-negative DCF
with supernatant from thymic epithelial cells stimulated with 2.5 ,ug/ml MRC OX6; and DCF + 2.5,ug/ml MRC OX3, the monoclonal antibody to rat Ia which is strain-specific in its recognition (reproduced in three experiments), are 30 compared.
(b) Removal of MF activity by its absorption on to Ia-positive cells. MF absorbed on to thymic epithelial cells prior to addition to DCF (reproduced in four experiments); MF absorbed on to rat spleen cells 35 (reproduced in three experiments); MF absorbed on to rat spleen cells depleted of Ia-positive cells (using MRC

. : - . .
:: . ~, . ..
~: ,~, - . .. ::
,~"~-"."" ~ ", , - , ~ ~ ,' ~ ' ,~ ,,, ,' ' . ~

.: . ., , - , - ~ . :

OX6/complement lysis) (reproduced in three e~periments);
and MF absorbed on to rat RBC (reproduced in two experiments) were compared.
(c) The binding site of the MF on spleen 5 cells is blocked by an antibody to Ia (MRC OX6). MF were absorbed on to spleen cells (as in Fig. 3b) but spleen cells were previously incubated with 2.5 ~g/ml of MRC
OX6. The possibility of carry over of MRC OX6 into the assay system was eliminated by labelling the antibody 10 with 125I (reproduced in three experiments). MF absorbed on to spleen cells previously incubated with rat-specific antibody to Ia (MRC OX3) (reproduced in three experiments);
Fig. 9 shows the role of the Fab portion of MRC
15 OX6 in stimulating protective activity and in blocking the binding of MF. Populations compared were DCF + MRC OX6; DCF + Fab MRC OX6;
DCF previously incubated with Fab MRC OX 6 +
MF;
Fig. 10 shows the role of thymic epithelial cells in thymocyte viability. A representative ~-experiment of percentage cell viability as a function-of - -time is shown. Note that in every experiment the unprotected populations were less than 70% of the control 25 population by 4 hours incubation. Populations compared were Control, DCF depleted of Ia-positive cells (using MRC OX6 and complement in a lysis technique);
Ia-negative DCF (i.e. DCF after the depletion 30 of Ia-positive cells) with the addition of MF (reproduced over four experiments);
Ia-negative DCF with l hour supernatant from unstimulated thymic epithelial cells (reproduced in two experiments);

Ia-negative DCF with supernatant from thymic epithelial cèlls incubated for 1 hour at 37C with MF
(reproduced in two experiments);
Fig. 11 shows results of molecular weight 5 determination of the thymic epithelial cell factor using Sepharose Cl-6B chromatography;
Fig. 12 shows the time course for the production of the thymic epithelial cell factor;
Fig. 13 shows the enhanced proliferation of 10 dense immature thymocytes in response to either whole - macrophage supernatant or an antibody to rat Ia (2.5 ~g/ml of clone MRC OX6 or clone MRC OX3). Representative experiment of eight experiments showing the mean +
standard deviation for quadruplicate cultures;
Fig. 14 shows the enhanced proliferation of immature thymocytes in response to partially purified macrophage supernatant. The addition of partially purified macrophage supernatant to dense immature thymocytes results in an increase in proliferation over 20 that of a control population. Pooled data from three preparations of purified Mr 36 000 monokine showing means values + standard error of the mean;
Fig. 15 shows the effect of purified macrophage factor on thymocyte proliferation. Addition of macrophage 25 supernatants which have been treated in one of three different ways restores the proliferative response of the adherent cell depleted population to that of a control population. Representative of two experiments showing mean ~ standard deviations; and -Fig. 16 shows the characteristics of the thymocyte response. The response of divided, recombined and whole thymus to varying levels of Con-A in the presence of 0.5 ~g of LPS is shown. Representative experiment of five experiments showing the mean +
35 standard deviations of quadruplicate cultures.

.

, ;,.. ~ ~ . .. :
. .... . ... .
~, . ., . . . ., . , -J . . ', ` , . ' ~
~, ~ ' : ' ;!~:, `, ,,. ~ .
:~` ~ . .' . ' "' ~.~'1,," " . "

Abbreviations used hereina~ter are deflned as follows:
Con-A Concanavalin-A
DCF Dense cell fraction of thymus DPBS Dulbecco's phosphate-buffered saline DTEC Dense thymic epithelial cells FCS Foetal calf serum IL-I Interleukin-l LPS Lipolysaccharide MDP Muramyl dipeptide MF Macrophage factor (monokine) Mr Relative molecular weight RBC Red blood cells TECF Macrophage-induced thymic - epithelial cell factor TNF TumouF necrosis factor Example 1 Preparation of macrophage supernatant Macrophage cultures were prepared from the 20 peritoneal washings of 6-8 week old Wistar-Furth rats.
The cells in RPMI-1640 medium containing 10% FCS were seeded into tissue culture flasks at a density of 1 x 106 cells/ml (5 x 106 per flask), and allowed to adhere for 1 hour at 37C. The monolayers were then washed five times 25 with DPBS, 5 ml serum-free medium was added per flask, and the cells were stimulated with 20 ~ug/ml lipopolysaccharide from Salmonella enteritidis (Sigma).
The supernatant was harvested at 2 hours, filtered through an XM300 filter (relative molecular weight (Mr) 30 300,000 cut off) to remove high Mr components, dialysed against DPBS and concentrated over a PM10 membrane filter (Mr 10,000 cut off). Both XM300 and PM10 ultra-filtration membranes were from Amicon Ltd. By titration it was shown that the described MF activity was 35 increased at least 500 times over that in control, ~ .

- ~
~, " ,~ ' ' - ' ' ' ' !;. - -unstimulated macrophage cultures. The culturedperitoneal cèlls were identi~ied as macrophages hy the criteria of being adherent cells, with 99.6% showing a positive reaction for lysozyme.

5 Example 2 Effect of Macrophage Supernatant on the Viability of Thymus Cells Thymus glands were removed from 4-8 week old male and female rats, taking care to dissect the thymus free from surrounding tissues including lymph nodes.
10 Thymuses were washed in RPMI-1640 containing lO~ FCS, sliced and pressed through a 0.5 mm pore size stainless steel wire mesh. The resulting thymic suspension was fractionated on a gradient of Percoll according to a described method (Salisbury et. al., 1979) The dense 15 fraction was then washed three times in RPMI-1640 with 10% FCS.
Purified dense thymic epithelial cells and dense thymocytes were prepared by treating whole thymus suspensions with antibody and complement prior to 20 fractionation on Percoll. Thymus cell suspensions containing 1 x 108 cells in 5 ml RPMI-1640 were treated ; with monoclonal antibody (MRC OX52, a rat pan T cell and thymocyte marker; Robinson et. al., 1986 for thymocytes, or MRC OX6, directed against a common determinant on rat 25 Ia, obtained from Sera Labs, for Ia-positive cells) at 2.5 pg/ml final concentration, together with complement at 2% final concentration. The preparations were incubated for 1 hour at 37C and then layered on to Percoll.
The isolated populations were washed and characterized by indirect immunofluorescence and by en~yme histochemistry. Results for 10 thymus preparations are summarized in Table 1.

~' .
j, , ,. "", ,, : . -o ~ + c E ~ E 3 E " ~ ~`~ o ~:: ~ ~ v~ 8 + + + ~ ~
c ,~e~ O O O I i ~ a~ > ~ O ~ o _. ~ C o. V C~ ~ ~ C ~
+~ +I X _ . a ~ X +l +l +I J o ~ o o ~aP
_~ ~~ ~~

~V7 ~, C
o ~.~ _ ~ u ~ a ~ ~ , c aJ ~, C

n i _ ~ oO ~ o s o _ - CJ _ X ~ X ~ o 1~
0 _ o.~ ~.~ ~ ~ V

a LL ^ ca ~_ o c ~1 0 3 ~ -~ 'J_ -O E~Xnl ~ j o--C~ ~ . ~ V V
~ ~ O
C C ~',> C~ .
_ CJ ~O'_ ~ (J .. -.
:~ C~ ~CJ .~

~",~ ' .

Aliquots of DCF were counted, and the number of viable cells (excluding trypan ~lue) was expressed as a percentage o~ the starting population. The results shown in Fig. 1 indicate a rapid loss of viability. This was 5 typically in the range of 40-70% of the starting population after 4 hours incubation.
The addition of 50 ~l/ml of supernatant from macrophage cultures which had been stimulated with 20 ~g lipopolysaccharide/ml 2 hours previously protected the 10 dense cell fraction from loss of viability. Neither lipopolysaccharide (from 1-20 ~g/ml) alone, nor supernatant from unstimulated macrophages protected the DCF. The supernatant from stimulated macrophages was titrated, and was found to be protective at an end-point 15 dilution of 1/500. Approximately half of the peritoneal cells which were seeded into culture flasks became adherent macrophages. This gave 2.5 x 106 cells per flask in 5 ml medium. Therefore 50 ~1 of supernatant represents the release from 2.5 x 104 macrophages. This 20 equates to this number of macrophages releasing enough activity to protect 500 x 107 dense thymic cells, or one macrophage protecting 2 x 105 dense thymic cells.
The loss of viability in vitro was confined to the dense, immature population and did not affect the 25 buoyant fraction or the thymic epithelial cells alone.
The protective effect was found in whole supernatant, and in the fraction retained above the PM10 membrane. Although these membranes have a nominal cut off of Mr 10,000, it was found that under the conditions 30 used, soybean trypsin inhibitor (Mr 23,000) was filtered out while bovine serum albumin (Mr 67,000) was retained.
Repeated assessment of macrophage cultures by examining at least 200 cells per culture showed that 99.6 + 0.6% of the adherent cells were positive for lysozyme 35 by indirect immunofluorescence staining. The specificity of the anti-rat lysozyme serum was demonstrated by a .:

... ~ . . . .

.. . . - ~. - .

-: . .. . . . .
.. ,.;... . . . . .

single arc in immunoelectrophoresis against a rat neutrophil lysate and again~t the purified protein used for immunization. A control or second stage antibody alone was negative. The cells in the cultures were 100%
5 positive for acid phosphatase, the reaction being characterized by diffuse cytoplasmic staining of variable intensity. For further studies a batch of fractionated macrophage supernatant was prepared from the peritoneal washings of 30 Wistar-Furth rats. This preparation (MF) 10 was used at a concentration equivalent to a 1/250 dilution of whole supernatant in subsequent protection studies.

Example 3 Characterization of Monokine (MF) (a) Physical properties.
The Mr of the monokine was determined by gel filtration fractionation on Sephacryl S200. The results are shown in Fig. 2a. MF eluted at Mr 36,000.
The isoelectric point of the MF was determined by isoelectric focusing. Standard proteins and 100 ~ul of 20 MF were loaded on to a broad range (pH 3-9) LKB ampholine gel and run to equilibrium. A standard curve was prepared from the reference proteins as shown in Fig. 2b and strips of the gel were cut at known intervals in relation to this. The strips were homogenized, dialysed 25 against DPBS and tested. A second run was used to narrow the range further. The activity was localized between pH
6.3 and 6.4.
The effect of heating was assessed by treating aliquots of MF for 30 minutes at 40, 56, 60, 70 and 80C.
30 Activity was destroyed at 80C, but was stable at lower temperatures. The effect of pH on the stability was tested by adding MF to appropriate 0.1 mol/l buffers at pH 2, 4, 6, 8 and 10 for 60 min at 37C. Samples were ~,-, :. ` '' then dialysed against DPE3S and tested for activity. MF
was stable over this pH range. MF activity was lost following reduction with 5% 2-mercaptoethanol.
The e~fect of proteinases on MF activity was 5 assessed by adding MF to 10 }lg/ml solutions of enzyme for 60 minutes at 37C. The proteinases were papain, pronase, thermolysin and trypsin, which were used in RPMI-1640, pH 7.4, and pepsin in RPMI-1640 adjusted to pH
4.0 with 1 mol/l HCl. The enzyme-MF mixtures or enzyme 10 controls were then added to DCF in RPMI-1640 containing 10% FCS as a source of proteinase inhibitors.
Proteinases alone had no effect on thymocyte survival, whereas all five proteinases destroyed MF activity. Thus the monokine is at least partly protein in nature. The 15 results of these studies are summarized in Table 2.

Physical Characteristics of Monokine.

Parameter Monokine Molecular weight Mr 36,000 Isoelectric point - 6.3 - 6.4 Heat stability Stable at 70C, activity lost - --at 80C ~.
25 pH stability Stable between pH 2 and 10 - -Proteinases Activity destroyed by papain, pepsin, pronase, thermolysin ;~ -and trypsin 5% 2-Mercaptoethanol Destroys activity `~
30 8 mol/l urea Destroys activity - -(b) Functional Properties.
MF was te~ted for its ability to stimulate 3T3 fibroblasts to proliferate both in the presence and the absence of FCS. Proliferation of fibroblasts was 5 expressed as the mean of quadruplicates of counts per minute of [3H]-thymidine uptake by the cells. As controls, fibroblasts alone and fibroblasts with whole macrophage supernatant were tested. The results given in Table 3 indicate that while the whole supernatant has 10 stimulating activity both in serum-containing and in serum-free conditions, MF has none under either condition.

r Effect of Macrophage Culture Supernatants 15 on Fibroblast Proliferation Sample Serum-free 10% FCS
(cpm) (cpm) 20 Control (fibroblasts alone) 1010 + 991880 + 315 Whole macrophage supernatant 5075 + 4344305 + 1300 Partially purified macrophage 903 + 125 1586 + 588 supernatant (MF) .

25 Example 4 Thymocyte Viability Assay Dense thymus cell fractions prepared as described in Example 2 were divided into one ml aliquots containing 1 x 107 cells. Test supernatants were added as 50 pl samples to these cells and viability was 30 assessed at hourly intervals by the exclusion of trypan blue (0.4% solution). Viability was expressed as a percentage of the original cell number.

'~ :
.

The titre of the activity was defined as the reciprocal of the maximum dilution of unfractionated macrophage supernatant which gave complete protection at four hours in the thymocyte viability assay. This 5 applied to 100 ,ul of supernatant added to 1 x 107 cells in a total volume of 1 ml.
Unfractionated thymus cell suspensions show no decrease in cell viability with time. However, depletion of macrophages, e.g. by adherence to plastic, results in 10 apoptotic death of immature thymocytes in the suspension.
This was prevented by addition of the Mr 36,000 monokine.
Surprisingly, if the thymocytes were first treated with corticosterone, cell death was increased.
The monokine evidently induces differentiation in a 15 population of Ia+ prothymocytes, which thereby become corticosterone-sensitive. This is supported by results described below with Con A.

Example 5 Further Characterisation of Synthesis and -Release of the Monokine.
The time course of production for the factor, --together with the effect of different stimuli at various -- -~
doses and at various times was examined. In addition, ~ -the nature of the production of the macrophage factor was studied using three different protein synthesis 25 inhibitors and trypsin treated and untreated lysed macrophage preparations.
(a) Macrophages were prepared as in Example 1, and were stimulated with either LPS (1-20 ug/ml) or MDP
(0.5-2.5 ~ug/ml). Supernatants were collected at various ---~
30 time intervals between five minutes and forty-eight hours -according to the experiment. In the case of the unstimulated macrophage supernatant, an identical procedure was followed without the addition of the stimulant, and the supernatant was collected two hours 35 after the replacement of serum-free medium.
~ ' - :

~.;.: ,.. - , ., .. ,, .- - :, ., - . : - - - -Varying concentrations of LPS and MDP were added to freshly established peritoneal macrophage cultures, and the supernatant was collected after two, eight or twenty-four hours incubation and tested for 5 activity in the thymocyte viability assay. The results are shown in Figure 3. For LPS, there was a proportionate increase in titre from 1 to 10 ~g LPS and then an abrupt increase from ten units at 10 ,ug LPS to 500 units at 20 ,ug LPS. There was no demonstrable 10 activity at eight or at twenty-four hours after the addition of LPS. The response to MDP was quite different in both the titre achieved and in the persistence of the response, so that for 2.5 yg MDP, although the titre was only one unit, it persisted for at 15 least twenty-four hours. Controls were set up to test the effect of the stimulants alone or in combination with the partially purified macrophage factor. Neither stimulant protected the thymocytes or affected the activity of the macrophage factor.
20 (b) The time course for the release of the macrophage factor was examined using 20 ,ug/ml LPS as the stimulant. As shown in Figure 4, low level activity was --~
released from the macrophages within five minutes after the addition of the stimulant. After one hour the 25 activity had increased ten-fold, with the peak response --of 500 units being reached at two hours. The activity decreased to 100 units at three hours, while from four hours to forty-eight hours there was no demonstrable activity. The yield as assessed by the number of units 30 of protective activity in the thymocyte viability assay is greatest in response to 20 ~g/ml LPS when the supernatant is collected two hours after the cells are challenged. Increasing the culture time beyond two hours led to a decrease in the number of units of activity at 35 three hours and a loss of protective activity beyond that time and up to forty-eight hours.

,.~

(c) Effect of Inhibitors of Protein Synthesis.
Macrophage supernatants produced in the presence of inhibitors of protei~ synthesis were prepared as follows: the macrophages were allowed to adhere and S were then washed to remove any traces of serum. The protein synthesis inhibitors were used in the following concentrations: actinomycin D 1 ~g/ml, puromycin 100 ~ug/ml, cycloheximide 10 ~g/ml.
Inhibitors were added to macrophage cultures at 10 the same time as 20 ~ug LPS and the supernatants were collected at time intervals up to two hours. They were dialysed to remove low molecular weight inhibitors and then tested for protective activity in the thymocyte viability assay. The results shown in Figure 5 show that 15 in the presence of each of the three protein synthesis inhibitors, protective aetivity was demonstrable in the culture supernatants at five and ten minutes after LPS
challenge. By fifteen minutes, however, activity had been lost in the puromycin and cycloheximide treated 20 cultures, whereas it was still present in the cultures which had been incubated with actinomycin D. From thirty -minutes to two hours, activity was lost in all of the treated cultures. -~
The early release of low level activity from -~- -25 five minutes onwards suggested that the factor was already present within the macrophages and was released.
In addition it was shown that lysates of unstimulated cells also contained protective activity, although this activity was not titratable. Addition of the protein - -~
30 synthesis inhibitors, actinomycin, puromycin and cycloheximide to cultures at the same time as the LPS
demonstrated that de novo protein synthesis was necessary for production of the factor. In contrast to cultures -with LPS alone, protective activity could not be detected 35 beyond ten minutes with puromycin and cycloheximide or after fifteen minutes with actinomycin. Hydrolysis of IP

~-"v .. ~ " . ,, . : ,, surface proteins by trypsinization prior to washing or by lysis of thè cells failed to remove the protective activity, indicating that the factor was not a surface protein on the macrophage, but wa9 probably cytosolic.
5 These results are shown in Figure 6.

Example 6 Effect of Restimulation of Macrophage Cultures Restimulation of cultures with LPS was examined to see whether yield of the monokine could be increased.
In addition, the abrupt decrease in titre from a peak at two hours after stimulation to no detectable protective effect at four hours was studied by examining the effects of restimulation with LPS at two, four and six hours of culture.
The abrupt decrease in titre of protective activity after 2 hours could have been due to prostaglandin production by the macrophages. The spontaneous production of prostaglandin E2 by thymic phagocytes in culture is greatly enhanced by adding LPS
20 to the cultures; the prostaglandin response to LPS is reduced 10-fold by indomethacin (Papiernik and Homo-Delarche, 1983).
We found a further peak of protective activity when 10 5M indomethacin was added to cultures at 4 hours 25 together with LPS and new medium, but not when cultures ; were restimulated in the absence of indomethacin.
-~; Supernatants were taken after two hours from cultures stimulated with 20,ug/ml LPS. At this time, the cultures were restimulated with 20 ~g/ml LPS alone or in 30 combination with 10 5M indomethacin. The supernatants were collected at four hours and the cultures were restimulated. This procedure was repeated at six hours and the final supernatants were collected at eight hours.

~' , .,", . ~
~'s.:; . .. :

... ~ .

The results are shown in Figure 7, and indicate that macrophages were anergic to further stimulation. In this context, it was noted that 10 nM of prostaglandin E2, added together with LPS at 20 ~g/ml to fresh 5 macrophage cultures, totally removed the activity at two hours. Indomethacin (10 5M), added together with LPS to fresh cultures, did not alter the peak titre at two hours; however, in the presence of this inhibitor of prostaglandin synthesis macrophages which were 10 restimulated at four hours did release low titre activity at six hours of incubation. There was no effect of indomethacin, however, on restimulation at two hours or at six hours.

Example 7 Partial Purification of the Monokine Macrophage supernatant was prepared as in Example 1, and filtered through an XM 300 membrane, to -~
remove the bulk of the lipopolysaccharide, and then through a PM 10 membrane. The fraction above the ~ ~-membrane was retained and sterile filtered as described. -~
20 The molecular weight was determined by gel filtration fractionation on Sephacryl S200, and the activity was -localized to an Mr 36,000 fraction (Yield = 80% of loaded ~
sample). The macrophage factor was further purified by - -ion exchange chromatography on DEAE Sephacel (Pharmacia). -~
25 The active fraction (Sml) from Sephacryl S200 was loaded ~
onto a DEAE Sephacel column (1.6 x 60 cm), and the ~ -activity eluted from this column in 2 x 2 ml fractions at - -120-124 ml after a gradient of 200 mls of 0.05 M Tris (pH
8.0) + 200 mls of the same buffer with 0.5M NaCl was 30 applied (Yield = 50% of loaded sample). The active --fractions were pooled and sterile filtered, and half of the active fraction was dialysed against lM (NH4)2S04 at pH 7.0 in preparation for chromatography on Phenyl-Sepharose CL-4B (Pharmacia). A 7 ml sample was 35 applied to 10 mls of Phenyl-Sepharose in a 0.9 cm diameter column with adaptor, the matrix having been equilibrated with lM (NH4)2S04, tpH 7.0). The flow rate was 20 ml/hour and a 100 ml gradient of lM ~NH4)2S04, (pH
7.0) + 100 mls H20 was applied immediately after the 5 addition of the sample and 2 ml fractions were collected.
The activity was recovered in 2 fractions, which represented 2~ of the total gradient (Yield = 27% of loaded sample).

Example 8 Receptor Site for the Monokine.
During the course of optimizing procedures for the depletion of Ia-positive cells it was observed that MRC OX6, in the absence of complement, promoted thymocyte survival (Figure 8a), thus mimicking the effect of MF.
As expected, this antibody had no effect on the survival 15 of the Ia-negative DCF. When incubated with isolated thymic epithelial cells at 1.2 x 106 per ml, it promoted the release of thymocyte survival activity. In contrast, MRC OX3, which recognizes a rat strain-specific epitope on Ia, failed to protect.
The possibility that MF also bound Ia was probed initially by attempting to deplete MF activity by absorption with different types of cells, as outlined earlier. MF was absorbed at 0C, using 3 cycles of --incubation for 1 hour with 1 x 107 cells. As shown in 25 Figure 8b, the DTEC effectively removed the MF activity.
Spleen cells also removed the activity, whereas spleen cells depleted of Ia-positive cells did not. RBC were also ineffective. Similarly, MF was absorbed on to spleen cells which had previously been incubated at 0C
30 with MRC OX6 or MRC OX3, and the supernatant was tested for protective activity. In order to negate the possibility of carry over of MRC OX6 into the viability assay, the antibody was labelled with 125I. The specific activity of the dialysed product was 2.09 x 105 35 disintegrations/min per ~g protein. No carry-over of ~5"` ". ". ,' ., ' ~ " , , ,'', . ' " ' ~ ' ' ' ' , " ' . ' ':' . " . . '', ' ' :
''.',"'.' :" ',', ' , radioactivity was detected. The results shown in Figure 8c indicate that prior binding of MRC OX3 did not prevent absorption of MF, whereas prior binding of MRC OX6 did so. This provided evidence that MF bound to surface Ia 5 at a site which was the same as or nearby the site recognized by MRC OX6. The finding that MRC OX3 did not block absorption of MF mitigated against an intermolecular steric blocking of MF by MRC OX6. The binding site of the MF was further examined by studying 10 the competitive effect of univalent Fab MRC OX6 antibodies prepared as described. Fab OX6 at 2.5 ~g/ml failed to protect the DCF in the thymocyte viability assay. Pre-incubation of the DCF with this Fab antibody ~
preparation for 15 min. at 37C did, however, effectively -;
15 block the protective action of MF, indicating a close -~
relationship between the binding site of the antibody and the macrophage product (Figure 9).
':,: " ,~
Example 9 Mediation of the Protective Effect Via Thymic Epithelial Cells The DCF was depleted of Ia-positive cells by treatment with MRC OX6 and complement as described -~
earlier. Similarly, DTEC were prepared by lysis of -~
thymocytes using MRC OX52 and complement. Cell ~-populations were characterized as described in Table 1. ~-~
There was good agreement between the number of cells positive for keratin and those remaining after -lysis of thymocytes in the DCF. The presence of macrophages and non-epithelial dendritic cells in this fraction was excluded on the basis of the buoyant nature ~ -~30 of these cells and that all of the dense cells were accounted for by either anti-keratin staining of epithelial cells (12.8%) or MRC OX52 staining of -thymocytes (87.2%). In addition, none of the cells in this fraction gave a positive reaction for lysozyme or 35 for acid phosphatase. Furthermore, the purified :- '. -~i~ ' ' :

epithelial cells appeared to be a uniform population by electron microscopy. The DTEC maintained viability over 24 hours in culture while the dense Ia-negative thymocytes showed 100% mortality over this time period.
5 Aliquots of MF (50 ~1) failed to protect 1 x 107 thymocytes depleted of Ia-positive cells (Figure 2)., The incubation of 50,ul MF with 1.2 x 106 DTEC (in proportion to the percentage in the DCF) did, however, result in the release of protective activity by these 10 cells (Figure 10).
Thus removal of the Ia-positive cells in the DCF left a population which displayed the described loss of viability over 4 hours. The addition of macrophage supernatant failed to prevent cell death, as did the 15 addition of supernatant from unstimulated thymic epithelial cells. The addition of supernatant from thymic epithelial cells which had previously been incubated with MF was protective, suggesting that a cascade was occurring in which macrophage products were 20 acting on thymic epithelial cells, resulting in the release of a factor responsible for the survival of thymocytes.

Example 10 Production of the Thymic Epithelial Cell - Factor The thymic epithelial cell factor was induced with either the macrophage factor or an antibody to a common determinant on Ia, as follows: Whole thymus cell suspensions were treated with MRC OX52 and complement for thirty minutes at 37C. The remaining cells were then 30 fractionated on Percoll and the dense cell fraction which contained keratin-positive thymic epithelial cells was collected. These cells were divided into three aliquots and were cultured in tissue culture flasks at a density of 1 x 108 cells/ml in RPMI 1640 medium. The first 35 aliquot served as a control, the second aliquot was ~
.

~,, . ~ .
.- .. ~
"

; - 26 -stimulated with 2.5 pg/ml of MRC OX6 and the remaining suspension was stimulated with the macrophage factor ~at dilution equivalent to 1/250 of the original macrophage supernatant). The cultures were incubated for three 5 hours, after which the supernatants were collected. The resultant supernatants were tested in the thymocyte viability assay using a dense cell fraction depleted of Ia positive cells. The macrophage-induced thymic epithelial cell factor (MF-TECF) was active at a 10 concentration of 1/5000 and the antibody-induced -epithelial cell factor ~MRC OX6-TECF) protected the thymocytes at a dilution of 1/750 in the thymocyte viability assay. The control supernatant from unstimulated thymic epithelial cells was inactive in the 15 thymocyte viability assay.
.,. ~,. . .
Example 11 Properties of the Thymic Epithelial Cell - ;
Factor The protective activity of the macrophage factor-thymic epithelial cell factor was destroyed by 20 incubating the factor (at a ljlO00 dilution) with 1 mg/ml -of trypsin. Activity was also destroyed by boiling the factor prior to testing it in the thymocyte viability -~
assay, suggesting that it was probably protein. ~ -The molecular weights of both MF-TECF and MRC
25 OX6-TECF were determined by gel filtration fractionation on Sepharose CL-6B (Pharmacia). The results are shown in Figure 11. A column (100 cm x 2.6 cm) was calibrated -with ferritin (Mr 440,000), aldolase (Mr 158,000), albumin (Mr 68,000~ and ovalbumin (MR 43,000) in PBS with 30 0.1 M NaCl and was run with a 1 ml sample under total identical conditions. The MF-TECF eluted at a molecular weight of 320,000 and the MRC OX6-TECF eluted at 760,000.
A possible binding site for the MF-TECF was suggested by experiments using a monoclonal antibody to 35 rat T helper cells which bound at CD4 (Sera Lab, Clone W3/25). Whole thymus suspensions were incubated with this antibody, Ia positive cells were removed and the remaining cells were fractionated on Percoll for use in the thymocyte viability assay. The addition of MF-TECF
5 to this dense cell fraction failed to protect the thymocytes, suggesting that the availability of CD4 molecules was important in this process.

Example 12 Characteristics of the Release of the Thymic Epithelial Cell Factor.
The MF-TECF was prepared as previously described but supernatants were collected from cultures at various intervals between five minutes and three hours and tested in the thymocyte viability assay. As shown in Figure 12, low titres of activity are produced in the 15 first fifteen minutes following which the titre increases steadily up to three hours. The addition of one of three protein synthesis inhibitors, actinomycin D, -cycloheximide or puromycin had no effect on the production of MF-TECF, indicating that protein synthesis 20 was not required.
These experiments suggest that the thymic epithelial cell factor, when induced by the macrophage factor, has a molecular weight of around 320,000.
Induction of the thymic epithelial cell factor with MRC
25 OX6 (the antibody to a common determinant on Ia), results in the release of a protein of molecular weight 760,000.
The large molecular weight of the protein together with the fact that the molecular weight of the product varied according to the method by which it was induced, 30 suggested that the thymic epithelial cell factor might in fact be an Ia-ligand complex that is shed from the thymic epithelial cell. If this does occur, then the selection of thymocytes for which apoptosis is arrested could be based on their ability to bind the Ia-ligand complqx that 35 is shed. This appears to be related to the expression of ,. . . .
:G' ., ~ ~

,,. , : .: ': !

CD4 by the thymocytes since pre-incubation of immature thymocytes with anti-CD4 antibodies inhibits the ob~erved protective efect of the monokine-induced thymic epithelial cell factor.
It was not possible to absorb the monokine with the monoclonal antibody W3/25 to CD4, indicating that it is unlikely that this macrophage product is soluble CD4.

Example 13 Stimulation of Thymocyte Maturation by the Monokine.
The immature thymocytes in the dense cell fraction do not proliferate in response to Con A. The effect of the monokine on their ability to respond to Con A was therefore tested.
The dense thymic fraction was incubated at 1 x 15 106 cells per well with 2.5 ,ug of MRC OX6 or MRC OX3.
The proliferative response to Con A was measured. While MRC OX3 had only a slight effect, there was a substantial increase in proliferation with both MRC OX6 and 2 hours whole macrophage supernatant, and an additive 20 effect when the two treatments were combined, as shown in Figure 13. This trend was reproduced over eight experiments.
Incubation of the dense thymic cell fraction with partially purified macrophage supernatant (Mr 36,000 25 fraction from chromatography on Sephacryl S200) also resulted in an increase in the proliferative response to Con A. This fraction was used at a concentration which demonstrated protective activity in the thymocyte viability assay, and three different preparations were 30 tested. All preparations showed a similar increase in proliferation, as shown in Figure 14, and the effect was reproduced over five experiments.
The depletion of adherent cells from whole thymus suspensions resulted in a decrease in thymocyte 35 proliferation in response to varying levels o~ Con A and ' 0.5 ~g o~ LPS compared with a control population. The mean o~ the ratios of the control population to the depleted population was 2.27 + 0.6. The addition to each well of 25 ~1 of whole macrophage supernatant (prepared 5 from 1 x 106 cells stimulated with 20 ,ug/ml LPS and harvested at 2 hours) restored the response to that of the control. The purity of the macrophage cultures was confirmed by staining for lysozyme and acid phosphatase.
The means of the ratio of the control response to the 10 restored response was 1.0 + 0.07.
The restoration of the response with macrophage supernatant did not appear to be due to IL-l.
Supernatant treated with 10 mmol/l phenylglyoxal to remove IL-l activity was still capable of restoring the 15 response to control levels. Similarly, the response was restored by the addition of Mr 36,000 factor from chromatography on Sephacryl S200, at a concentration which was active in the thymocyte viability assay. These results are shown in Figure 15.
In other experiments it was found that 0.5 ~g LPS increased thymidine uptake in response to a range of concentrations of Con A, but that the shape of the dose-response profile was unchanged. Similar results were obtained with MDP (1 ,ug/106 cells) or streptococcal 25 cell wall fragments (0.5 ,ug/106 celIs). Conversely, in the presence of a constant amount of Con A (0.25 ~ug/well), 1 pg of LPS sharply stimulated thymidine uptake, with gradual further increase up to 1 mg LPS.
Again, similar results were obtained using MDP or 30 streptococcal cell wall fragments.
Fractionation of the thymus cell suspension on a density gradient of Percoll confirmed that the buoyant fraction which contained mature thymocytes, macrophages, epithelial cells and non-epithelial dendritic cells 35 proliferated in response to Con A and 0.5 ~g of LPS. The dense fraction containing immature thymocytes and dense ~i~

r",',' ..
~ .

~~. ' ' " , .

' ., ' '~ .

epithelial cells responded only weakly to Con A and LPS.
Recombination of the two fractions restored the response to a level equivalent to that of whole thymus, showing that cells had not been lost in the separation procedure.
5 Figure 16 shows the response of the buoyant, dense, recombined and whole fractions to Con A and LPS.
These results indicate that macrophage products are responsible for the increased proliferative responses of thymocytes to co-stimulation with Con A and microbial -~
10 products. The data support the efficient uptake of picogram quantities of a variety of bacterial components by macrophages, which comprised less than 0.1~ of the thymic cell suspension. The evidence for the r~le for macrophages in the augmentation of the proliferative 15 response was provided by a diminished response following depletion of adherent cells, and by the restoration of the activity by products from highly purified peritoneal macrophages. The active macrophage component was shown to be the Mr 36,000 protein, which did not have IL-l 20 activity as determined by a fibroblast proliferation assay.
. .
Example 14 Effects of the Monokine in vivo.
It is known that the cytokines IL-l, TNF, and -~-interleukin-6 can induce liver epithelial cells to 25 secrete acute-phase plasma proteins. Macrophages are a ~; major source of these cytokines. ~ -A single intravenous injection of the -~ -partially-purified Mr 36,000 protein into rats induced a significant elevation of plàsma fibrinogen and ; 30 ~2-macroglobulin levels.An injection of heated protein -`
caused no response.
The amount of protein injected was equivalent ;; to that released by 1 x 105 macrophages. This small amount of monokine produces a substantial effect in the - -.:: ' `i animal by a cascade phenomenon whereby responding macrophages secrete one or more cytokines, which in turn activate the acute phase response.

Example 15 An Automated Bioassay for the Monokine.
In order to monitor large numbers of samples conveniently, an assay which can be carried out in microtitre plates and in which many samples can read simultaneously is desirable.
We have developed an assay based on the 10 conversion of a soluble, yellow compound ~MTT) to an insoluble blue formazan by enzymes in living cells. The insoluble product is dissolved in isopropanol, and estimated spectrophotometrically. The assay can be carried out in 96 well microtitre plates of the type used 15 for ELISA assays.
Two complementary variations in technique were used. The assay is based on the cleavage of the tetrazolium salt MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) into a blue 20 coloured product (formazan) by the mitochondrial enzyme succinate dehydrogenase (Denizot and Lang, 1986). The conversion takes place only in living cells, and the amount of formazan produced is proportional to the number of cells present.
Thymic suspensions were either fractionated on Percoll as described earlier, or depleted of adherent cells by incubating 1 x 108 thymic cells in Sml RPMI 1640 medium in a 75 cm2 tissue culture flask for one hour at 37C. This depletes the suspension of thymic macrophages 30 and destabilizes the population so that the immature, dense thymocytes undergo apoptotic death.
Using the dense ce-ll fraction from Percoll gradient separation, cells were washed into RPMI 1640 medium, without serum or phenol red, at a density of 2 x 35 107 per 0.9 ml. Dilutions of macrophage supernatant or ..,~

.,, ~ , :

of the fractions containing monokine activity were added at 100 ~ul/2 x 107 cells. The cells were then aliquoted in replicates of eight into flat bottom 96 well culture plates at 2 x 106 cells/100 ,ul. The effective range is 5 0.5 x 106 to 5 x 106 cells/well but 2 x 106 cells are preferred. The final row of cells received cycloheximide at 10 ,ug/ml instead of macrophage products. This arrests the protein synthesis necessary for apoptotic death, and provides a positive control at 100% viability. The 10 plates were incubated at 37 for three hours and then MTT
was added as 50 ,ul of a 1 mg/ml solution in RPMI 1640 medium. After one hour of incubation at 37C, the plates were centrifuged at 800 x g for 10 minutes and the untransformed MTT was removed by inverting, flicking and 15 blotting the tray. Two hundred microlitres of isopropanol was then added to each well, the plates briefly shaken and the formazan read at 540 or 560 nm (preferred) test wavelength against a 690 nm reference wavelength in an ELISA reader.
Formazan generation in test samples was read with reference to both cycloheximide-treated positive controls and unprotected controls. The conditions for using adherent cell-depleted whole thymic populations were identical, except that the incubation step was for 25 seven hours or sixteen hours (preferably seven) before adding the MTT.
The assay using fractionated dense thymic cells involves rapid apoptotic death and therefore a short incubation period while the assay using depleted adherent 30 cells involves a slower rate of death and longer incubation time. It does, however, have the advantage that it is easy to establish, and is applicable to -~;-screening large number of samples. ^`

Example 16 Distinction Between the Monokine and Previously known Macrophage-derived Factors.
The characteristlcs of IL-l and TNF in the rat 5 have been described ("Lovett et al, 1983; Schmitt et al, 1986; Vilcek et al. 1986; Rupp et al. 1986).
The failure of the monokine of this invention to stimulate fibroblast proliferation in either the presence or absence of serum (Example 3(b)) suggests that 10 it is neither IL-l nor TNF. This is supported by the homogeneity of the factor with respect to both Mr and isoelectric point; both IL-l and TNF have a number of molecular forms (Smith et al., 1986; Wood et al. 1985).
Phenylglyoxal is an inhibitor of IL-l activity 15 (Klamfeldt, 1985; Krakauer, 1985). Whole macrophage supernatant from cultures incubated for 2 hours with 20 ,ug LPS/ml were treated with 10 mmol phenylglyoxal/ml.
This treated supernatant was able to restore the response of adherent cell-depleted thymus cell cultures to Con A, 20 as shown in Figure 15.
The time course for the production of this factor differs from that of IL-l or lymphocyte activating factor in mice, humans and rats. Mizel (1981) showed that mouse peritoneal macrophages stimulated with phorbol 25 myristate acetate reach a peak level of production of IL-l around twenty-four hours, and that this then plateaus. Similarly, Wood e~ al, (1983) collected supernatants from a Balb/c macrophage line, 2-3 days after the cells were stimulated with LPS from Escherichia 30 coli in order to harvest lymphocyte activating factor for testing in the thymocyte proliferation assay. Human monocytes and macrophages stimulated with LPS show a maximum yield of lymphocyte activating factor at twenty-four hours with very little production prior to 35 two hours as assessed by the ability of the supernatants to augment proliferation of mouse thymocytes in response .,, ~, . -~" .

to phytohaemagglutinin (Treves et al. 1983). Bird et al.
(198S) prepared IL-1 from Sprague-Dawley rat peritoneal macrophages by culturing the cells for forty-eight hours in the presence of 10 Jug/ml LPS. Examination of the time 5 requirement for TNF production by LPS induction of human monocytes showed that cytotoxicity reached plateau values within two hours (Kornbluth and Edgington, 1986). The monocytes, however, need to be primed two to three days prior to challenging with LPS, with an agent such as 10 tubercle bacilli (Carswell, 1975; Mestan, 1986). This was not necessary for the early production of the factor of the present invention.
While it is recognized that the macrophages will produce a large number of soluble factors in 15 response to challenge with LPS, the early release together with the abrupt decrease in production at two hours of this factor would appear to be different from other well recognised monokines.

::
In summary, the monokine of this invention acts 20 on thymic epithelial cells to cause the release of a second factor, thought to be a complex between the monokine and Ia antigen, which in turn promotes the survival and differentiation of immature thymocytes.
The monokine would be useful in:
~ -,- ,. . .
(a) Stimulating the maturation of the immune -~
system e.g. in children with congenital or acquired ---immune deficiency; AIDS patients; patients being treated -~
with radiotherapy or cytotoxic drugs;
(b) Selective stimulation of immunity;
(c) Selective suppression of inflammation or ;
autoimmunity.
It will be clearly understood that the invention in its general aspects is not limited to the ~-specific details referred to hereinabove.

~ .

REFERENCES

1. Adkins, B., Mueller, C., Okada, C.Y. and Reichert, R.A. (1987). Early events in T-cell maturation.
Ann. Rev. Immunol. 5: 325-365.

2. Berrih, S., Savino, W. and Cohen S. (1985).
Extracellular matrix of human thymus; Immunofluorescence studies on frozen sections and cultured epithelial cells.
J. Histochem. Cytochem. 33: 655-664.

3. Bird, J., Sheng, Y.J., Florentin, I., and Giroud, J.P. (1985). Release of interleukin and low-molecular-weight lymphocyte-activating factors by rat peritoneal macrophages and its enhancement by acute non-specific inflammatory processes. Br. J. Exper. Path., 66: 271-277.

4. Burgess, A.W. and Metcalf, D. (1980). The nature and action of granulocyte-macrophage colony stimulating factors. Blood 56: 947-958.

5. Carswell, E.A., Old, L.J., Kassell, R.L., Green, S., Fiore, N., and Williamson, B. (1975). An endotoxin-induced serum factor that causes necrosis of tumours. Proc. Natl. Acad. Sci. USA, 72: 3666-3670.
6. Denizot, F. and Lang, R. (1986). Rapid colorimetric assay for cell growth and survival.
Modifications to the tetrazolium dye procedure giving improved sensitivity and reliability. J. Immunol.
Methods. 89:271-277.
7. Dinarello, C.A. (1984). Interleukin-l. Rev.
Infect. Dis. 6: 51-95.

~.' : . ~ ,, :
,,~ - .
, . ~ . .
8. Douglas-Jones, A., Nelson, J., Jansen, V. and Miller, T. (i981). Characterization o~ the ~rnunZ) nude rat. Morphological characteristics of the lymphoid system. Aust. J. Exp. Biol. Med. Sci. 53: 277-286.
9. Kinnon, C., Diamond, R.A. and Rothenberg, E.V.
(1986). Activation of T-cell antigen receptor ~- and ~ -chain genes in the thymus; implications for the lineages of developing cortical thymocytes. J. Immunol.
137: 4010-4015.
10. Klamfeldt, A. (1985). Enhanced breakdown in vitro of bovine articular cartilage proteoglycans by conditioned synovial medium. The effect of phenylglyoxal. Scand. J. Rheumatol. 14: 217-223.
11. Kornbluth, R.S. and Edgington, T.S. (1986).
Tumour necrosis factor production by human monocytes is a regulated event: induction of TNF-a-mediated cellular cytotoxicity by endotoxin. J. Immunol., 137(8):
2585-2591. - --~-12. Krakauer, T. (1985). Biochemical characterization of interleukin 1 from a human monocytic -cell line. J. Leuk. Biol. 37: 511-518.
13. Le, J. and Vilcek, J. (1987). Biology of disease. Tumour necrosis factor and interleukin 1:
cytokines with multiple overlapping biological activities. Lab. Invest. 56: 234-248.
i 14. Le. P.T., Tuck, D.T., Dinarello, C.A., Haynes, B.F., and Singer, X.H. (1987). Human thymic epithelial cells produce interleukin 1. J. Immunol. 138: 2520-2526.

`: - - ~ . : ' ' :
15. Lo, D. and Sprent, J. (1986). Exogenous control of I-a expression in foetal thymus explants. J. Immunol.
137: 1772-1775.
16. Lovett, D.H., Ryan, J.L. and Sterzel, R.B.
(1983) Stimulation of rat mesangial cell proliferation by macrophage interleukin 1. ~. Immunol. 131: 2830-2836.
17. Mestan, J., Digel, W., Mittnacht, S., Hillen, H., Blohm, D., Moller, A., Jacobsen, and Kirchner, H.
(1986). Antiviral effects of recombinant tumour necrosis factor in vitro. Nature, 323: 816-819.
18. Mizel, S.B. (1981). Production and quantitation of lymphocyte-activating factor (interleukin 1), in Manual of Macrophage Methodology (H.B. Herscowitz, H.T.
Holden, J.A. Bellanti and A. Ghaffor, eds.) Marcel Dekker Inc., New York.
19. Papiernik, M. and Homo-Delarche, F. (1983).
Thymic reticulum in mice III. Phagocytic cells of the thymic reticulum in culture secrete both prostaglandin E2 and interleukin 1 which regulate thymocyte proliferation.
Eur. J. Immmunol., 13: 689-692.
20. Robinson, A.P., Puklavec, M. and Mason, D.W.
(1986) MRC OX-52; a rat T-cell antigen. Immunology 57:
527-531.
21. Rupp, E.A., Cameron, P.M., Ranawat, C.S., Schmidt, J.A., and Bayne, E.K. (1986). Speific bioactivites of monocyte derived interleukin 1 alpha and interleukin 1 beta are similar to each other on cultured human connective tissue cells. J. Clin. Invest. 78:
836-839.
22. Salisbury, J.G., Graham, J.M. and Pasternak, C.A., (1979). A rapid method for the separation of large and small thymocytes from rats and mice. J. Biochem.
~iophys. Meth. 1:341-347.
23. Schmitt, A., Hauser, C., Jaunin, F., Dayer, J.M., and Saurat, J.H. (1986). Normal epidermis containq high amounts of natural tissues II-l, biochemical analysis by HPLC identifies a Mw approximately 17Kd with -a Pi 5.7 and an Mw 30 Kd form. Lymph. Res. 5: 105-118.
24. Smith, R.A., Xirstein, M., Fiers, W. and Boglioni, C. (1986). Species specificity of human and murine tumour necrosis factor. A comparative study of tumour necrosis factor receptors. J. Biol. Chem. 261: 14 871-14 876. ~;
25. Treves, A.J., Barak, V., Tal, T., and Fuks, Z. -(1983). Constitutive secretion of interleukin 1 by human monocytes. Europ. J. Immmunol., 13: 647-651.
~ , 26. Van Ewijk, N., Rouse, R.V. and Weissman, I.L. -~
(1980). Distribution of the H-2 microenvironments in the mouse thymus. Immunoelectron microscopic identification of the I-A and H-2K bearing cells. J. Histochem.
Cytochem. 28: 1098-1099.
27. Vilcek, J., Palombella, V.J., Henriksen-Di Stefano, D., Swenson, C., Feinman, R., Hirai, M. and Tsujimoto, M. (1986). Fibroblast growth enhancing activity of tumour necrosis factor and its relationship to other polypeptide growth factors. J. Exp. Med.
163:632-643.
::
28. Weissman, I.L. (1986). Nursing the thymus (editorial) Lab. Invest. 55: 1-4.

:
~ ~ .

- . ~
29. Wood, P.R. Andrus, L. and Clark I.A. (1983).
Production o~ lymphocyte activating factor in vivo.
Immunol 50: 637-644.

~ ,'''' :-' , ' ` ' . ' ~. .: `

Claims (20)

1. A monokine which is released by macrophages in response to stimulation with lipopolysaccharide, said monokine having the following properties:
Relative molecular weight 36000 kD;
Isoelectric point 6.3 - 6.4;
Stable at temperatures up to 70°C;
Stable at pH 2 to 10;
Activity destroyed by: reduction with
2-mercaptoethanol treatment with proteinases, or treatment with urea;
Does not stimulate proliferation of fibroblasts; and Binds to common determinant of Ia antigen on thymic epithelial cells.
2. A method of producing the monokine defined in Claim 1, comprising the steps of incubating macrophages in nutrient medium in the presence of bacterial lipopolysaccharide or muramyl depeptide for 5 minutes to 2 hours, and recovering the monokine.
3. A method according to Claim 2, in which the macrophages are incubated with bacterial lipopolysaccharide.
4. A method according to Claim 2 or Claim 3, in which incubation is carried out for 2 hours.
5. A method according to any one of Claims 2 to 4, in which monokine is recovered by the steps of:
(a) recovering medium from macrophage cultures, (b) removing lipopolysaccharide or muramyl dipeptide, (c) removing material of Mr less than about 20,000, (d) subjecting the remainder to sequential steps of gel filtration, ion exchange chromatography, and hydrophobic interaction chromatography, and (e) recovering fractions having monokine activity.
6. A method according to Claim 5, in which steps (b) and (c) are performed by ultrafiltration.
7. A method according to Claim 5 or Claim 6 in which gel filtration is performed using Sephacryl S200.
8. A method according to any one of Claims 5 to 7 in which ion exchange chromatography is performed using DEAE Sephacel.
9. A method according to any one of Claims 5 to 8 in which hydrophobic interaction chromatography is performed using Phenyl-Sepharose CL-4B.
10. A method according to any one of Claims 5 to 9 in which further purification is effected using preparative gel electrophoresis or high performance liquid chromatography.
11. A monokine as defined in Claim 1, produced by the method of any one of Claims 2 to 10.
12. A method of bioassay of the monokine defined in Claim 1, comprising the steps of:
(a) adding a sample of fluid containing or suspected of containing monokine to either dense thymic cells, or whole thymus suspension depleted of adherent cells, in nutrient medium in the absence of serum and indicator dye and incubating for a period of 3 to 16 hours, (b) adding MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide);
(c) incubating for 1 hour at 37°C;
(d) removing untransformed MTT;
(e) adding isopropanol to the samples;
(f) estimating extinction at 540 or 560 nm;
and (g) calculating the amount of monokine in the sample.
13. A method of bioassay according to Claim 12 in which incubation in step (a) is for 3 to 7 hours.
14. A method of bioassay according to Claim 12 in which dense thymic cells are used, and incubation in step (a) is for 3 hours.
15. A method of bioassay according to Claim 12 in which a thymus suspension depleted of adherent cells is used, and incubation is for 7 hours.
16. A method of bioassay according to any one of Claims 12 to 15 in which MTT is incubated with samples each containing 0.5 to 5 x 106 cells.
17. A method of bioassay according to Claim 16 in which each sample contains 2 x 106 cells.
18. A factor which is produced by thymic epithelial cells in response to stimulation with the monokine defined in Claim 1, said factor having the following properties:
Relative molecular weight 320,000;
Activity destroyed by trypsin treatment or by boiling;
Does not require active protein synthesis for production;
Binds to immature thymocytes;
Protects immature thymocytes from apoptotic death; and Binding to thymocytes inhibited by preincubation with antibody to CD4 antigen.
19. A pharmaceutical composition comprising the monokine defined in Claim 1, together with a pharmaceutically acceptable carrier, diluent or excipient.
20. A pharmaceutical composition according to Claim 19, which also comprises a thymic epithelial cell factor as defined in Claim 18.
CA 2000695 1989-03-16 1989-10-13 Cytokine Abandoned CA2000695A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
AUPJ3248 1989-03-16
AUPJ324889 1989-03-16

Publications (1)

Publication Number Publication Date
CA2000695A1 true CA2000695A1 (en) 1990-09-16

Family

ID=3773786

Family Applications (1)

Application Number Title Priority Date Filing Date
CA 2000695 Abandoned CA2000695A1 (en) 1989-03-16 1989-10-13 Cytokine

Country Status (2)

Country Link
CA (1) CA2000695A1 (en)
WO (1) WO1990010650A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5198428A (en) * 1991-04-05 1993-03-30 Board Of Regents, The Univ. Of Texas System Macrophage cytotoxin compositions and methods of preparation

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS60105621A (en) * 1983-11-14 1985-06-11 Agency Of Ind Science & Technol Physiologically active substance having differentiation inducing activity to cell and its preparation
US4923807A (en) * 1984-05-18 1990-05-08 New England Medical Center Hospitals Inc. Arg-Serpin human plasminogen activator inhibitor designated PAI-2
FR2596651B1 (en) * 1986-04-08 1989-11-10 Centre Nat Rech Scient NOVEL LIPOSOMES BASED ON PHOSPHATIDYLINOSITOLMANNOSIDES, AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
FR2615622B1 (en) * 1987-05-19 1994-05-06 Ire Medgenix Sa PLASMATIC ASSAY OF MONOKINES
JPS6490199A (en) * 1987-09-30 1989-04-06 Hiromi Fujiwara T cell growth factor derived from thymic interstitial cell and production thereof
JPH01132381A (en) * 1987-11-19 1989-05-24 Kyodo Nyugyo Kk Heparitinase and production thereof

Also Published As

Publication number Publication date
WO1990010650A1 (en) 1990-09-20

Similar Documents

Publication Publication Date Title
Blanton et al. Regulation of interleukin 1 and its receptor in human keratinocytes.
Togawa et al. Characterization of lymphocyte-activating factor (LAF) produced by human mononuclear cells: biochemical relationship of high and low molecular weight forms of LAF
CA2086264C (en) Surface complexed lymphotoxin
Gately et al. Synergy between recombinant interleukin 2 (rIL 2) and IL 2-depleted lymphokine-containing supernatants in facilitating allogeneic human cytolytic T lymphocyte responses in vitro.
Lefrancois et al. Antigen-independent activation of memory cytotoxic T cells by interleukin 2.
JP2006312640A (en) Activation of t cell
JPH04502769A (en) Method for controlling human endothelial cell proliferation and effector function using oncostatin M
MXPA01010332A (en) Methods and compositions for modulating an immune response.
CA2125181A1 (en) Trophoblast interferons and their use
Grufman et al. Innate and adaptive immunity to tumors: IL‐12 is required for optimal responses
CA2000695A1 (en) Cytokine
Borges et al. Peptide‐based analysis of the amino acid sequence important to the immunoregulatory function of Trypanosoma cruzi Tc52 virulence factor
Nissen-Meyer et al. Comparison of recombinant tumor necrosis factor and the monocyte-derived cytotoxic factor involved in monocyte-mediated cytotoxicity
Peleg et al. 1, 25-Dihydroxyvitamin D3 and its analogues inhibit acute myelogenous leukemia progenitor proliferation by suppressing interleukin-1beta production.
Sorg Modulation of macrophage functions by lymphokines
Heemskerk et al. Differential activation of mouse hepatitis virus-specific CD4+ cytotoxic T cells is defined by peptide length.
Lin et al. Increased cAMP and cAMP-Dependent Protein Kinase Activity Mediate Anti-CD2 Induced Suppression of Anti-CD3-Driven lnterleukin-2 Production and CD25 Expression
Hirayama et al. Accessory cell functions of dendritic cells and macrophages in the thymic T-cell response to Con A.
Gallicchio et al. Synergistic action of recombinant-derived murine interleukin-1 on the augmentation of colony stimulating activity on murine granulocyte-macrophage hematopoietic stem cells in vitro
Krane Collagen degradation
Cohen et al. Adherent Ia+ murine cell lines with characteristics of dendritic cells: II. Characteristics of I region-restricted antigen presentation
Parra et al. Inhibition of mitogenesis induced by phytohemagglutinin and Lens culinaris lectin in adherent-cell supernatants treated with protein extract of Mycobacterium tuberculosis
Snyderman et al. Biologic aspects of leukocyte chemotaxis
Papageorgiou Human Lymphoid Cell Lines: in Vitro Models for Study of the Immune Response
Nocera et al. In vitro production of different interferon types by cloned human NK cells.

Legal Events

Date Code Title Description
FZDE Dead