throbber
Journal of Neuroimmunology 166 (2005) 132 – 143
`
`www.elsevier.com/locate/jneuroim
`
`Detoxication enzyme inducers modify cytokine production in rat
`mixed glial cells
`
`Anne Wierinckx a,1, John Breve´ a, Dominique Mercier a,2, Marianne Schultzberg b,
`Benjamin Drukarch a, Anne-Marie Van Dam a,*
`
`aInstitute for Clinical and Experimental Neurosciences, Department of Medical Pharmacology,
`VUmc, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
`bNeurotec Department, Division of Experimental Geriatrics, Karolinska Institute, Novum, SE-14186 Stockholm, Sweden
`
`Received 9 March 2005; accepted 27 May 2005
`
`Abstract
`
`interleukin-1h (IL-1h),
`factor-a (TNFa) as well as
`tumor necrosis
`interleukin-6 (IL-6),
`Pro-inflammatory cytokines, e.g.
`neurotoxic molecules such as nitric oxide (NO), that are produced and released by activated glial cells, play an important role in
`inflammation and oxidative stress occurring during Multiple Sclerosis (MS). Reduction of these processes could therefore be of
`therapeutic interest. Dimethylfumarate (DMF) and sulforaphane (SP) are well known for their detoxicating properties. Furthermore,
`they have anti-inflammatory effects as shown clinically by the treatment of inflammatory skin diseases. However, their detoxication
`and anti-inflammatory action on brain-derived cells
`is unknown.
`In the present
`study we have studied, within the same
`concentration range, the anti-inflammatory and detoxicating effects of DMF and SP on the production and release of mediators of
`inflammation and detoxication from lipopolysaccharide (LPS) activated primary co-cultures of rat microglial and astroglial cells.
`DMF and SP attenuated the LPS-induced production and release of TNFa, IL-1h, IL-6 and NO. In addition, DMF and SP increase
`both mRNA level and activity of NAD(P)H:quinone reductase (NQO-1), a detoxication enzyme, as well as the cellular glutathione
`content. We conclude that DMF or SP simultaneously can (1) reduce mediators of inflammation and (2) enhance detoxication
`enzymes in LPS stimulated co-cultures of astroglial and microglial cells. This double-sided effect could potentially be of therapeutic
`interest.
`D 2005 Elsevier B.V. All rights reserved.
`
`Keywords: Interleukin-1h; Interleukin-6; Tumor necrosis factor-a; Nitric oxide; Dimethylfumarate; Sulforaphane
`
`* Corresponding author. Tel.: +31 20 444 8095; fax: +31 20 444 8100.
`E-mail addresses: a.wierinckx@laposte.net (A. Wierinckx),
`jjp.breve@vumc.nl (J. Breve´), dominique.mercier@igmm.cnrs.fr
`(D. Mercier), schultzberg@nuerotec.ki.se (M. Schultzberg),
`b.drukarch@vumc.nl (B. Drukarch), amw.vandam@vumc.nl
`(A.-M. Van Dam).
`1 Present address: Unite´ Inserm U433-Neurobiologie Expe´rimentale et
`Physiopathologie Faculte´ Laennec, 8, rue G. Paradin 69372 Lyon cedex 08,
`France. Tel.: +33 4 72 91 34 92; fax: +33 4 72 68 49 55.
`2 Present address: CNRS-UMR 5535-IGMM, 1919 route de Mende, 34293
`Montpellier cedex 5, France. Tel.: +33 4 67 36 05; fax: +33 67 04 02 31.
`
`0165-5728/$ - see front matter D 2005 Elsevier B.V. All rights reserved.
`doi:10.1016/j.jneuroim.2005.05.013
`
`1. Introduction
`
`The etiology of chronic inflammatory, neurodegener-
`ative diseases such as Multiple Sclerosis (MS) is not well
`understood. Neuropathologically,
`the presence of
`infil-
`trated immune cells and activated glial cells is evident
`(Benveniste, 1997; Heneka and Feinstein, 2001). During
`inflammation these cells produce a variety of pro-
`inflammatory mediators including interleukin-1h (IL-1h),
`interleukin-6 (IL-6) and tumor necrosis factors-a (TNFa)
`as well as neurotoxic molecules such as nitric oxide (NO).
`All of these are known to contribute directly or indirectly
`to cellular dysfunction and/or cell death associated with
`neurodegenerative processes (Hantraye et al., 1996;
`
`Sawai (IPR2019-00789), Ex. 1035, p. 001
`
`

`

`A. Wierinckx et al. / Journal of Neuroimmunology 166 (2005) 132 – 143
`
`133
`
`Hooper et al., 1997; Oshima et al., 1996; Simonian and
`Coyle, 1996; Takahashi et al., 1997). In MS for example,
`cytokines and reactive oxygen species (ROS,
`including
`NO) are present in perivascular T-cells, macrophages and
`in surrounding activated glial cells. It is shown that their
`presence leads to demyelination,
`functional motor dis-
`turbances and finally axonal loss (Penkowa and Hildago,
`2003; Bar-Or et al., 1999; Brosnan et al., 1995; Cannella
`and Raine, 1995).
`Neuro-inflammation and oxidative stress, as detected in
`MS,
`leads to alterations in the activity/functionality of
`resident brain cells.
`In a chronic situation,
`it has a
`progressive effect on the course of the disease, emphasizing
`the importance of reducing the level of inflammation and
`reactive oxygen species within the brain. Several naturally
`occurring and synthetic compounds, including dimethylfu-
`marate (DMF) and sulforaphane (SP), are known to present
`detoxicating properties in mammalian cells. These com-
`pounds do not scavenge free radicals, but elevate the
`production of
`relevant detoxication enzymes including
`NAD(P)H:quinone reductase (NQO-1) and/or stimulate
`glutathione synthesis (Basten et al., 2002; Dringen et al.,
`1998; Gao et al., 2001; Khanna et al., 1998; McMahon et
`al., 2001; Nelson et al., 1999; Prochaska and Fernandes,
`1993; Spencer et al., 1990).
`Interestingly, DMF and SP have also been shown to
`affect inflammatory processes. DMF has been used for the
`systemic treatment of psoriasis and reduces itching and
`scaling and prevents extension of the disease (Nieboer et al.,
`1989). Mechanistically, DMF reduces cytokine expression
`in lymphocytes (de Jong et al., 1996; Ockenfels et al.,
`1998), has an anti-proliferative effect on keratinocytes (Thio
`et al., 1994) and,
`inhibits monocyte differentiation into
`dendritic cells (Zhu and Mrowietz, 2001). SP down-
`regulates bacterial lipopolysaccharide (LPS)-mediated pro-
`duction of several inflammatory proteins (e.g. iNOS, COX-
`2 and TNFa) in cultured macrophages (Heiss et al., 2001).
`Recently, Wu and collaborators have shown decreases in
`NFnB nuclear translocation and infiltration of activated
`macrophages in kidneys of spontaneous hypertensive rats
`fed with dried broccoli sprouts containing glucoraphanin,
`metabolized into SP (Wu et al., 2004).
`The compounds DMF and SP may represent promising
`therapeutic targets to enhance detoxication and reduce
`inflammation,
`if both actions occur within the same
`concentration range of the compound used. In the present
`study, we question whether DMF and SP show detoxication
`and anti-inflammatory activities in LPS-activated brain-
`derived glial cells. Until now, little is known of effects of
`DMF and SP on brain-derived cells. To address this question,
`rat mixed microglia and astrocytes were treated with LPS to
`stimulate production of pro-inflammatory mediators (e.g. IL-
`1h, TNFa, IL-6) and reactive oxygen species (e.g. NO).
`Subsequently, the anti-inflammatory effects of DMF and SP
`on IL-1h, TNFa, IL-6 and NO production by the glial cells
`were studied. In addition, the detoxicating effects of DMF
`
`and SP were studied by measuring glutathione (GSx) levels
`and NQO-1 activity in the cells.
`
`2. Materials and methods
`
`2.1. Mixed glial cell culture
`
`Primary co-cultures of astroglial and microglial cells were
`prepared from newborn (2 day-old) Wistar rats (Harlan CPB,
`Zeist, The Netherlands) (Vincent et al., 1996). Cerebral
`cortices were cleared from adhering meninges and blood
`vessels, and dissociated using 0.25% trypsin (Sigma
`Chemical Co., St Louis, MO) in phosphate-buffered saline
`(PBS). Cells (0.5 106/well) were plated in poly-l-lysine
`(15 Ag/ml; Sigma)-coated 12 wells plates (Nunclon, Ham-
`-
`strop, Denmark) and incubated at 37
`C in humidified air
`containing 5% CO2. The culture medium consisted of
`Dulbecco’s modified Eagles medium (DMEM)-F10 (Gibco,
`Life Technologies, Breda, The Netherlands), supplemented
`with 10% v/v heat-inactivated fetal calf serum (FCS)
`(Gibco), 2 mM l-glutamine (Sigma), 37.5 U/ml streptomy-
`cin (Sigma) and 100 U/ml penicillin (Sigma). The medium
`was changed 1 day after seeding. Cultures consisted mainly
`of microglial and astroglial cells with less than 0.01%
`oligodendrocytes. Experiments were carried out after 7 days
`of culture, when microglia showed short ramifications and
`cultures were confluent.
`
`2.2. Treatment of glial cells
`
`In a first series of experiments, co-cultures were incubated
`for 1, 2, 4, 6, 8, 16, 24, 48 or 72 h with LPS (100 ng/ml;
`Escherichia coli serotype 055-B5, Westphal, Difco, Detroit,
`MI) or media alone to determine the time-dependent
`cytokine production by activated glial cells. Culture super-
`natants were collected, divided into aliquots and stored at
` 20
`-
`C until assayed. The cells were lysed with a RNA
`extraction buffer (Promega Corporation, USA) and stored at
` 80
`-
`C. In subsequent series of experiments, astroglial –
`microglial cell co-cultures were incubated for 4 and 24 h
`with LPS (100 ng/ml). Incubation with LPS was carried out
`in the absence (vehicle: 0.0006% DMSO or 50 AM HCL) or
`presence of DMF (5, 15 or 30 AM in 0.0006% DMSO) or SP
`(1, 5 or 15 AM in 50 AM HCL). All conditions were tested in
`triplo and the experiments were repeated at least twice.
`Treatment of the cells did neither result in reduced cell
`viability nor in altered cellular protein levels within the
`timeframe of the experiments (data not shown). After
`incubation, culture supernatants were collected, divided into
`aliquots and stored at 20
`-
`C. Cells were lysed with a RNA
`extraction buffer (Promega Corporation) and stored at 80
`-
`C or lysed with Tris/EDTA and sonificated for NQO-1
`activity and stored at 20
`-
`C. For GSx measurements,
`cells were lysed with 2.5% salicilyc acid (SSA) and stored
`at 20
`-
`C.
`
`Sawai (IPR2019-00789), Ex. 1035, p. 002
`
`

`

`134
`
`A. Wierinckx et al. / Journal of Neuroimmunology 166 (2005) 132 – 143
`
`Table 1
`Rat primers and accession numbers of the rat DNA sequences used to design the primers
`
`Name
`
`GAPDH
`IL-1h
`IL-6
`iNOS
`NQO-1
`TNFa
`
`Primer FW
`5V-GAACATCATCCCTGCATCCA-3V
`5V-AAAGAAGAAGATGGAAAAGCGGTT-3V
`5V-CCCCAACTTCCAATGCTCTC-3V
`5V-AACTTGAGTGAGGAGCAGGTTGA-3V
`5V-AACGTCATTCTCTGGCCAATTC-3V
`5V-CCACACCGTCAGCCGATT-3V
`
`Primer Rev
`5V-CCAGTGAGCTTCCCGTTCA-3V
`5V-GGGAACTGTGCAGACTCAAACTC-3V
`5V-AGATGAGTTGGATGGTCTTGGTC-3V
`5V-CGCACCGAAGATATCCTCATGA-3V
`5V-GCCAATGCTGTACACCAGTTGA-3V
`5V-TCCTTAGGGCAAGGGCTCTT-3V
`
`Accession number
`
`NM_017008
`NM_031512
`NM_012589
`NM_012611
`NM_017000
`AJ002278
`
`2.3. Nitrite assay
`
`2.5. Rat IL-1b ELISA
`
`The nitrite concentration in the culture supernatant is
`used as a measure of NO production. Nitrite was measured
`by a colorimetric assay based on the Griess reaction, as
`described by Ding et al. (Ding et al., 1988). Briefly, 100 Al
`supernatant was mixed with 100 Al Griess reagent, consist-
`ing of 1% sulfanilamide (Jansen chimica, Geel, Belgium),
`0.1% naphtylethylene diamine dihydrochloride (Merck,
`Darmstadt, Germany), and 2.5% H3PO4 in water, and
`incubated at room temperature (RT)
`for 10 min. The
`absorbance was measured in a microtiter plate reader at
`550 nm (ICN Flow, MS2 Reader,
`ICN Biomedicals,
`Zoetermeer, The Netherlands). Sodium nitrite (Merck)
`dissolved in culture medium was used as the standard.
`Detection limit of the assay was 1 AM nitrite.
`
`2.4. Rat IL-6 ELISA
`
`IL-6 concentrations were measured in culture super-
`natant using an enzyme-linked immunosorbent assay
`(ELISA) specific for
`rat
`IL-6 (Rees et al., 1999a).
`Immunoaffinity-purified sheep anti-rat
`IL-6 antibodies
`were used as coating and detecting antibodies (S206/
`B1 : 2 mg/ml and 1 : 1000 dilution, respectively; NIBSC
`Potter Bar, UK). Recombinant rat IL-6 (NIBSC) diluted in
`culture medium/HPE (CLB, Amsterdam, The Netherlands)
`(50% v/v) was used as the standard. Detection limit of the
`assay was 8 pg IL-6/ml.
`
`IL-1h was measured using a
`The concentration of
`sandwich ELISA specific for rat IL-1h (Safieh-Garabedian
`et al., 1995). Purified polyclonal sheep anti-rat IL-1h
`antibodies were used as coating and detecting antibodies
`(S1002(110700): 1 Ag/ml and 1 : 1000 dilution, respectively;
`NIBSC). Standard curves, included in duplicate on each
`plate, were made with rat recombinant IL-1h (NIBSC)
`diluted in culture medium/HPE (CLB) (50% v/v) to measure
`IL-1h in the supernatant. Detection limit of the assay was 10
`pg IL-1h/ml.
`
`2.6. Rat TNFa ELISA
`
`The concentration of TNFa was measured using a
`rat TNFa (Rees et al.,
`sandwich ELISA specific for
`1999b). Purified polyclonal sheep anti-rat TNFa antibodies
`were used as coating and detecting antibodies
`(S54(050900): 1 Ag/ml and 1 : 500 dilution, respectively;
`NIBSC). Standard curves, included in duplicate on each
`plate, were made with rat recombinant TNFa (NIBSC)
`diluted in culture medium/HPE (CLB) (50% v/v). Detection
`limit of the assay was 10 pg TNFa/ml.
`
`2.7. NQO-1 enzyme activity measurement
`
`Cells were washed with PBS and lysed in ice cold 25
`mM Tris – HCL and 1 mM EDTA (pH = 7.4). After 5 s of
`
`Table 2
`IL-1h, IL-6, TNFa and nitrite levels produced by mixed astroglial – microglial cells
`
`Protein
`
`1 h
`
`IL-1b (pg/ml)
`Control
`< dl
`LPS
`< dl
`
`2 h
`
`4 h
`
`6 h
`
`8 h
`
`24 h
`
`48 h
`
`72 h
`
`< dl
`3.53
`
`T
`
`0.55
`
`< dl
`12.77
`
`T
`
`1.44
`
`< dl
`30.80
`
`T
`
`3.97
`
`< dl
`36.57
`
`T
`
`9.13
`
`< dl
`75.83
`
`T
`
`5.91
`
`< dl
`83.33
`
`T
`
`3.79
`
`< dl
`123.13
`
`T
`
`10.61
`
`T
`
`T
`
`T
`
`T
`
`IL-6 (pg/ml)
`Control
`< dl
`LPS
`< dl
`
`TNFa (pg/ml)
`Control
`< dl
`LPS
`139
`
`T
`
`Nitrite (lM)
`Control
`< dl
`LPS
`< dl
`
`< dl
`< dl
`
`< dl
`392.70
`
`T
`
`18.23
`
`< dl
`870.93
`
`T
`
`92.55
`
`< dl
`1019.83
`
`88.78
`
`< dl
`3711.67
`
`88.50
`
`< dl
`5164.60
`
`245.22
`
`< dl
`5775.50
`
`451.42
`
`62.13
`
`< dl
`1167
`
`T
`
`96.42
`
`< dl
`3110
`
`T
`
`97.54
`
`< dl
`3612
`
`T
`
`152.26
`
`< dl
`3716.1
`
`T
`
`292.10
`
`< dl
`4274.267
`
`T
`
`165.39
`
`< dl
`1918.6
`
`T
`
`97.92
`
`< dl
`1275.6
`
`T
`
`101.33
`
`< dl
`< dl
`
`< dl
`< dl
`
`< dl
`< dl
`
`< dl
`< dl
`
`< dl
`6.97
`
`T
`
`0.51
`
`< dl
`24.10
`
`T
`
`1.23
`
`< dl
`35.17
`
`T
`
`1.00
`
`< dl = below detection limit.
`
`Sawai (IPR2019-00789), Ex. 1035, p. 003
`
`

`

`A. Wierinckx et al. / Journal of Neuroimmunology 166 (2005) 132 – 143
`
`135
`
`sonification, cytosolic supernatants were collected, ali-
`quoted and stored at 20
`-
`C prior to use.
`The enzymatic activity of NQO-1 was determined
`spectrophotometrically by measuring the NADPH-depend-
`ent, menadiol-mediated reduction of 3-(4,5-dimethylthiazol-
`2yl)-2,5-diohenyltetrazolium bromide (MTT) as described
`before (Prochaska and Santamaria, 1988) with minor
`modifications (van Muiswinkel et al., 2000).
`
`rabbit IgGs (1 : 100; Jackson Immunoresearch Laboratories)
`and Alexa fluor 488 labelled streptavidin (1 : 100; Molecular
`Probes, or biotinylated donkey anti-rabbit IgGs (1 : 100;
`Jackson Immunoresearch Laboratories) and FITC labelled
`donkey anti-goat IgGs (1 : 25; Nordic, Denmark). Finally,
`the cells double-labelled for NQO-1 and GFAP were
`incubated with Texas Red labelled avidin (1 : 100; Vector
`Laboratories).
`
`2.8. Cellular localization of NQO-1
`
`2.9. Glutathione level measurement
`
`Mixed glial cells were cultured in a 12 wells plate and
`treated for 24 h with LPS (100 ng/ml), LPS and DMF (30
`AM) or LPS and sulforaphane (15 AM). After treatment, the
`cells were washed with PBS and fixed for 20 min with 4%
`paraformaldehyde. The cells were rinsed with TBS and
`subsequently incubated for 1 h at room temperature with a
`rabbit anti-rat NQO-1 antibody (MS-14; Dr. M. Schultzberg,
`Karolinska Institute, Stockholm, Sweden), diluted 1 : 10,000
`in TBS containing 0.1% Tween-20,
`followed by an
`incubation with biotinylated goat anti-rabbit IgGs (1 : 100;
`Jackson Immunoresearch Laboratories, USA) and finally an
`incubation with ABC (1 : 400; Vector Laboratories, USA).
`After a final wash in Tris/HCl, a nickel-enhanced DAB
`reaction was performed using H2O2 as a substrate to identify
`NQO-1 positive cells.
`A double-labeling immunocytochemical procedure was
`performed to determine which glial cell
`type expresses
`NQO-1. Therefore, the cells were incubated with a mixture
`of either rabbit anti-rat NQO-1 (1 : 10,000) and biotinylated
`mouse anti-rat ED-1 (1 : 250; gift from Prof. Dr. C.D.
`Dijkstra, Department of Molecular Cell Biology, VUmc,
`Amsterdam) or rabbit anti-rat NQO-1 (1 : 10,000) and goat
`anti-rat GFAP (1 : 100; Santa Cruz). After washing, the cells
`were incubated with either Texas Red labelled goat anti-
`
`To measure the total intracellular glutathione (GSx = 1
`GSH + 2 GSSG) content
`in cell
`lysates, we used a
`spectrophotometrical assay as described previously (Dru-
`karch et al., 1996). The amount of GSx in a cell lysate was
`determined according to a standard curve of 0, 1, 2, 3, 4 and
`5 AM GSx in 2.5% SSA, and calculated as Amol/mg protein.
`
`2.10. RNA extraction
`
`Cells were homogenized in 175 Al of SV RNA Lysis
`Buffer (SV Total RNA Isolation System, Promega Corpo-
`ration, USA). Total RNA was extracted according to the
`manufacturer’s protocol. The RNA samples were eluted in
`sterile water and concentrations were measured at A260 with
`a spectrophotometer (SPECTRAmax 250, Molecular Devi-
`ces, USA). The purity of RNA was valued on by a ratio
`A260 / A280 of 1.9 – 2.1.
`
`2.11. Reverse transcription
`
`One microgram of total RNA was reverse transcribed
`using the Avian Myeloblastosis Virus (AMV) Reverse
`Transcriptase (Reverse Transcription System, Promega
`Corporation). The absence of contaminating genomic
`
`Table 3
`Relative mRNA expression of IL-1h, IL-6, TNFa, iNOS and NQO-1 measured in mixed astroglial – microglial cells
`
`mRNA
`
`1 h
`
`2 h
`
`4 h
`
`6 h
`
`8 h
`
`24 h
`
`48 h
`
`72 h
`
`IL-1b (Relative expression to GAPDH, in percent)
`T
`T
`Control
`0.46
`0.03
`0.64
`0.002
`0.67
`T
`T
`LPS
`77.23
`1.56
`130.15
`6.32
`233.55
`
`T
`T
`
`0.03
`3.76
`
`0.26
`163.62
`
`T
`T
`
`0.003
`3.43
`
`0.11
`21.21
`
`T
`T
`
`T
`T
`
`0.07
`2.62
`
`0.01
`0.93
`
`T
`T
`
`T
`T
`
`0.30
`151.4
`
`0.09
`19.85
`
`0.13
`5.26
`
`0.07
`151.07
`
`0.01
`1.4
`
`0.04
`19.19
`
`T
`T
`
`T
`T
`
`0.004
`5.39
`
`0.08
`118.33
`
`0.01
`0.67
`
`0.01
`13.72
`
`T
`T
`
`T
`T
`
`0.01
`5.22
`
`0.16
`119.12
`
`0.001
`1.00
`
`0.03
`11.14
`
`T
`T
`
`T
`T
`
`0.02
`4.88
`
`0.001
`0.6
`
`IL-6 (Relative expression to GAPDH, in percent)
`T
`T
`Control
`0.21
`0.0001
`0.22
`0.02
`T
`T
`LPS
`1.35
`0.002
`16.07
`1.03
`
`0.11
`67.7
`
`TNFa (Relative expression to GAPDH, in percent)
`T
`T
`Control
`0.62
`0.05
`0.65
`0.03
`0.43
`T
`T
`LPS
`51.29
`2.49
`64.96
`3.50
`39.89
`
`T
`T
`
`0.03
`2.14
`
`0.30
`9.09
`
`iNOS (Relative expression to GAPDH, in percent)
`T
`T
`Control
`0.0075
`0.0008
`0.0123
`0.004
`0.0166
`T
`T
`LPS
`0.04
`0.01
`0.76
`0.07
`10.95
`
`T
`T
`
`0.0009
`0.85
`
`0.0093
`8.41
`
`NQO-1 (Relative expression to GAPDH, in percent)
`T
`T
`Control
`1.55
`0.05
`1.69
`0.04
`1.61
`T
`T
`LPS
`1.61
`0.01
`1.70
`0.01
`1.45
`
`T
`T
`
`0.07
`0.06
`
`1.81
`1.29
`
`T
`T
`
`T
`T
`
`T
`T
`
`0.03
`0.46
`
`0.33
`9.64
`
`0.0006
`0.32
`
`0.0040
`3.77
`
`0.04
`0.04
`
`1.68
`1.43
`
`T
`T
`
`T
`T
`
`T
`T
`
`0.04
`0.29
`
`0.14
`3.05
`
`0.001
`0.04
`
`0.0002
`12.05
`
`0.04
`0.06
`
`1.80
`3.66
`
`T
`T
`
`T
`T
`
`T
`T
`
`0.01
`0.14
`
`0.14
`1.49
`
`0.0001
`0.12
`
`0.0002
`4.75
`
`0.16
`0.09
`
`1.10
`4.57
`
`T
`T
`
`T
`T
`
`T
`T
`
`0.01
`0.05
`
`0.12
`0.92
`
`0.0002
`0.10
`
`0.0005
`3.89
`
`0.09
`0.15
`
`1.95
`4.56
`
`T
`T
`
`T
`T
`
`T
`T
`
`0.01
`0.07
`
`0.0001
`0.51
`
`0.27
`0.44
`
`Sawai (IPR2019-00789), Ex. 1035, p. 004
`
`

`

`136
`
`A. Wierinckx et al. / Journal of Neuroimmunology 166 (2005) 132 – 143
`
`DNA and the efficiency of the RT reactions were controlled
`by PCR on RNA and cDNA using GAPDH Forward and
`Reverse primers (Table 1).
`
`2.12. Quantitative PCR
`
`The cDNA synthesized was assayed using quantitative
`real time PCR (SYBR Green PCR, ABI 7700, PE Applied
`Biosystem, California, USA). With this method, direct
`detection of PCR products is monitored by measuring the
`increase in fluorescence caused by the binding of SYBR
`Green to double stranded DNA. The resulting relative
`increase in reporter fluorescent dye emission was monitored
`in real time during PCR amplification using the Sequence
`Detection System (ABI PRISM 7700 Sequence Detection
`system and software, PE Applied Biosystems).
`Primers (Table 1) were designed from rat corresponding
`sequences using Primer Express Software (PE Applied
`Biosystems).
`Quantitative PCR was carried out in a final volume of 20
`Al with 6.5 ng of cDNA, 3 mM MgCl2, 0.2 mM dATP,
`dCTP and dGTP, 0.4 mM dUTP, 0.75 mM of each primer,
`
`0.3 U of AmpliTaq Gold DNA and 0.12 U of Amperase
`uracil-N-glycosylase (all reagents from PE Applied Bio-
`-
`-
`system). PCR conditions were: 50
`C for 2 min, 95
`C for
`-
`-
`10 min followed by 45 cycles of 95
`C for 15 s and 60
`C
`for 1 min.
`Threshold cycle (Ct) values provide an index of the
`mRNA level. The level of GAPDH mRNA was used as an
`internal standard to control the amplification variations due
`to differences in the starting mRNA concentrations. The
`relative expression level of cytokine mRNA for each tissue
`was computed from the Ct values obtained for the gene of
`interest and GAPDH using the following formula:
`Relative mRNA expression of gene of interest (in percent
`compared to the GAPDH)
`¼ 2 Ct

`
`interest geneCt GAPDH
`

`
`2.13. Statistical analysis
`
`Using the NCSS 2000 statistical program (NCSS,
`Kaysville, Utah, US),
`two-way ANOVA was performed
`
`Fig. 1. Effects of DMF and SP on lipopolysaccharide (LPS)-induced production of IL-1h in mixed glial cells. Cells were treated with LPS (100 ng/ml) for 4 and
`24 h. IL-1h protein was measured in the media after 4 and 24 h and IL-1h mRNA was measured after 4 h. a, effect of DMF on IL-1h protein level, b, effect of
`SP on IL-1h protein level, c, effect of DMF on IL-1h mRNA level, d, effect of SP on IL-1h mRNA level. Data are expressed as percentage of levels measured
`T
`in cultures co-incubated with LPS and solvent (control), and represent mean
`S.E.M. of 2 – 3 independent experiments (n = 6 – 12). *P < 0.01 vs. control.
`
`MYLAN PHARMS. INC. EXHIBIT 1035 PAGE 5
`
`Sawai (IPR2019-00789), Ex. 1035, p. 005
`
`

`

`A. Wierinckx et al. / Journal of Neuroimmunology 166 (2005) 132 – 143
`
`137
`
`followed by a t-test for independent measurements (Fish-
`er’s LSD test). When the data did not follow a normal
`distribution,
`the analysis was performed on log-trans-
`formed data. P values < 0.01 were considered to indicate
`a significant difference.
`
`3. Results
`
`3.1. Time-dependent induction of IL-1b, IL-6, TNFa, iNOS,
`nitrite and NQO-1
`
`to measure
`time point(s)
`To establish the optimal
`inflammatory mediators produced by glial cells after LPS
`stimulation, the astroglial – microglial cell co-cultures were
`treated for 1 up to 72 h with LPS.
`At the protein level, TNFa was the first cytokine to be
`detected in the media after 1 h of LPS stimulation, IL-1h
`was detectable after 2 h, IL-6 was elevated after 4 h and
`nitrite after 24 h (Table 2). The concentration of TNFa
`reached its maximum at 24 h after LPS treatment. However,
`
`IL-1h, IL-6 and NO increased progressively until the end
`(72 h) of the experiment.
`The maximal mRNA levels for IL-1h, IL-6 and iNOS
`were observed after 4 h of LPS stimulation after which they
`attenuated, except
`for
`iNOS mRNA which remained
`elevated (Table 3). The mRNA level of TNFa reached its
`maximum after 2 h and decreased after 6 h (Table 3). NQO-
`1 mRNA was induced by LPS after 24 h to reach a plateau at
`48 h (Table 3).
`Based on these data, protein levels were measured at 4
`and 24 h after LPS incubation and mRNA levels at 4 h after
`LPS stimulation in all further experiments. Nitrite was
`measured at 24 h only.
`
`3.2. Effect of the compounds on LPS-induced IL-1b, IL-6
`and TNFa and nitrite levels
`
`The LPS-induced IL-1h protein levels measured in the
`culture supernatant were 15 pg/ml at 4 h after LPS
`treatment and 80 pg/ml at 24 h after LPS treatment. At 4 h
`after co-incubation of the cells, DMF suppressed slightly,
`
`Fig. 2. Effects of DMF and SP on lipopolysaccharide (LPS)-induced production of IL-6 by mixed glial cells. Cells were treated with LPS (100 ng/ml) for 4 and
`24 h. IL-6 protein was measured in the media after 4 and 24 h, and IL-6 mRNA was measured after 4 h. a, effect of DMF on IL-6 protein level, b, effect of SP
`on IL-6 protein level, c, effect of DMF on IL-6 mRNA level, d, effect of SP on IL-6 mRNA level. Data are expressed as percentage of levels measured in
`T
`cultures co-incubated with LPS and solvent (control), and represent mean
`S.E.M. of 2 – 3 independent experiments (n = 6 – 12). *P < 0.01 vs. control.
`
`Sawai (IPR2019-00789), Ex. 1035, p. 006
`
`

`

`138
`
`A. Wierinckx et al. / Journal of Neuroimmunology 166 (2005) 132 – 143
`
`but significantly LPS-induced IL-1h levels (Fig. 1a),
`whereas SP did not have any effect (Fig. 1b). At 24 h,
`IL-1h levels in the culture media were reduced by both
`DMF and SP (Fig. 1a, b).
`The LPS-induced IL-6 protein levels measured in the
`culture supernatant were 450 pg/ml at 4 h and 4200 pg/ml
`at 24 h after LPS treatment. Both DMF and SP reduce IL-
`6 levels in the media of LPS stimulated glia mix at 4 and
`24 h (Fig. 2a, b).
`LPS-induced TNFa protein levels measured in the
`culture supernatant were 2900 pg/ml at 4 h and 4100 pg/
`ml at 24 h after LPS treatment. DMF and SP affect TNFa
`production of LPS stimulated glia co-cultures at both time
`points measured (Fig. 3a, b).
`The LPS-induced nitrite levels measured in culture
`supernatant was 10 AM at 24 h after LPS treatment. Only
`the two highest doses of DMF and SP decreased nitrite
`LPS-induced nitrite levels measured at 24 h after incuba-
`tion (Fig. 4).
`
`3.3. Effect of the compounds on LPS-induced IL-1b, IL-6,
`TNFa and iNOS mRNA levels
`
`The mRNA levels of the cytokines and iNOS measured
`were detectable in non-LPS treated cells (Table 2) and were
`not modified by DMF or SP treatment (data not shown).
`LPS treatment dramatically enhanced cytokine and iNOS
`mRNA levels as shown in Table 2.
`Co-incubation of LPS with DMF inhibited the mRNA
`levels of all cytokines and of
`iNOS (Figs. 1c-4c).
`However, only the highest dose (15 AM) of SP reduced
`the transcription of LPS-induced inflammatory mediators
`(Figs. 1d-4d).
`
`3.4. Effect of the compounds on NQO-1 mRNA level and
`NQO-1 activity
`
`The average LPS-induced cellular NQO-1 activity was
`48 and 65 dE/min/mg of total protein at 4 and 24 h after LPS
`
`Fig. 3. Effects of DMF and SP on lipopolysaccharide (LPS)-induced production of TNFa by mixed glial cells. Cells were treated with LPS (100 ng/ml) for 4
`and 24 h. TNFa protein was measured in the media after 4 and 24 h, and TNFa mRNA was measured after 4 h. a, effect of DMF on TNFa protein level, b,
`effect of SP on TNFa protein level, c, effect of DMF on TNFa mRNA level, d, effect of SP on TNFa mRNA level. Data are expressed as percentage of levels
`T
`measured in cultures co-incubated with LPS and solvent (control), and represent mean
`S.E.M. of 2 – 3 independent experiments (n = 6 – 12). *P < 0.01 vs.
`control.
`
`Sawai (IPR2019-00789), Ex. 1035, p. 007
`
`

`

`A. Wierinckx et al. / Journal of Neuroimmunology 166 (2005) 132 – 143
`
`139
`
`Fig. 4. Effects of DMF and SP on lipopolysaccharide (LPS)-induced production of nitric oxide in mixed glial cells. Cells were treated with LPS (100 ng/ml)
`for 4 and 24 h. Nitrite was measured in the media after 24 h, and iNOS mRNA was measured after 4 h. a, effect of DMF on the nitrite level, b, effect of SP
`on the nitrite level, c, effect of DMF on the iNOS mRNA level, d, effect of SP on the iNOS mRNA level. Data are expressed as percentage of levels
`T
`measured in cultures co-incubated with LPS and solvent (control), and represent mean
`S.E.M. of 2 – 3 independent experiments (n = 6 – 12). *P < 0.01 vs.
`control.
`
`treatment, respectively. DMF and SP induced similar effects
`on NQO-1 mRNA level and activity (Fig. 5). At 4 h, the
`activity of NQO-1 was not modified by the presence of
`DMF or SP in cultures treated with LPS (Fig. 5a, b). After
`24 h of co-incubation a significant
`increase in NQO-1
`activity was observed with all doses of DMF and SP used
`(Fig. 5a,). In contrast to NQO-1 activity, NQO-1 mRNA was
`already increased at 4 h after co-incubation of the cells with
`LPS and either of the two compounds (Fig. 5c, d).
`
`activity of the detoxication enzyme NQO-1 was enhanced
`200% – 300% by DMF and SP, whereas the glutathione
`synthesis was maximally 60% enhanced, we studied the
`cellular localization of NQO-1 in the mixed glial cell
`culture. NQO-1 positive cells were clearly detected in cells
`with various morphological shapes (Fig. 6a). The subse-
`quent double labeling procedure showed that NQO-1 can be
`produced by both microglial and astroglial cell types (Fig.
`6b, c, d, e).
`
`3.5. Cellular localization of NQO-1
`
`3.6. Effects of the compounds on glutathione levels
`
`We and others observed in LPS-activated mixed glial
`cells, that pro-inflammatory mediators are mainly produced
`by microglial cells (Vincent et al., 1996, 1997; Nakamura et
`al., 1999; Ledeboer et al., 2000). As the production and
`
`In LPS activated glial cells, the GSx levels were 12.1
`Amoles/mg at 4 h and 16.1 Amoles/mg at 24 h. Co-
`incubation with DMF or SP for 4 h dramatically depleted
`GSx from the cells whereas after 24 h of co-incubation, both
`
`Sawai (IPR2019-00789), Ex. 1035, p. 008
`
`

`

`140
`
`A. Wierinckx et al. / Journal of Neuroimmunology 166 (2005) 132 – 143
`
`Fig. 5. Effects of DMF and SP on NQO-1 activity and mRNA level in lipopolysaccharide (LPS)-treated mixed glial cells. Cells were treated with LPS (100 ng/
`ml) for 4 and 24 h. Intracellular NQO-1 activity was measured after 4 and 24 h, NQO-1 mRNA was measured after 4 h. a, effect of DMF on NQO-1 activity, b,
`effect of SP on NQO-1 activity, c, effect of DMF on NQO-1 mRNA level, d, effect of SP on NQO-1 mRNA level. Data are expressed as percentage of levels
`T
`measured in cultures co-incubated with LPS and solvent (control), and represent mean
`S.E.M. of 2 – 3 independent experiments (n = 6 – 12). *P < 0.01 vs.
`control.
`
`compounds significantly increased the level of GSx in the
`cells (Fig. 7).
`
`4. Discussion
`
`Microglia and astrocytes are important sources of
`inflammatory mediators under neuropathological conditions
`(Hirsch et al., 2003, 1998; Liu and Hong, 2003; McGeer and
`McGeer, 1998) as well as under activated conditions in vitro
`(Gadient and Otten, 1997; Lee et al., 1993; Vincent et al.,
`1996). We have previously shown that interactions between
`activated glial cells determine the level of inflammatory
`mediators produced (Vincent et al., 1997, 1996). Therefore,
`a co-culture of astrocytes and microglia represents a relevant
`model to study neuro-inflammatory responses in vitro.
`In the present study, we observed for the first time that
`DMF and SP exert a dual action on brain-derived cells
`within the concentration range and timeframe measured, i.e.
`they reduce cytokine levels and, enhance NQO-1 mRNA
`
`and activity levels, as well as increase the cellular
`glutathione content of activated mixed glial cells.
`At first it was shown that LPS induced time-dependent
`mRNA production and subsequent release of IL-1h, IL-6,
`TNFa and NO. TNFa was produced and released within 1 h
`after LPS treatment, followed by IL-1h, IL-6 and finally
`NO. This sequence of cytokine and NO production fits
`nicely with previous in vitro observations (Nakamura et al.,
`1999), and allowed for the establishment of the optimal
`time-points to measure the above mentioned mediators after
`LPS treatment. Subsequently,
`the effects of
`the anti-
`inflammatory compounds DMF and SP were studied on
`mRNA production (at 4 h) and release of cytokines (4 and
`24 h) and NO (24 h). Co-incubation of mixed glial cells with
`LPS and either DMF or SP reduced cytokine and iNOS
`mRNA production as well as release of their concomitant
`proteins. DMF exerted the strongest effect on IL-6 followed
`by NO, TNFa and finally IL-1h production and release. SP
`reduced production and release of mentioned mediators in
`the same order, but mRNA levels were slightly less affected
`
`Sawai (IPR2019-00789), Ex. 1035, p. 009
`
`

`

`A. Wierinckx et al. / Journal of Neuroimmunology 166 (2005) 132 – 143
`
`141
`
`produced by both astrocytes and microglial cells in vitro is
`supported by the observation that glial cells in the brain are
`the main source of NQO-1 (van Muiswinkel et al., 2004). In
`addition, NQO-1 has been shown to be induced by DMF
`and SP demonstrating the potential value of these com-
`pounds as protective agents against chemical carcinogenesis
`and other forms of electrophile toxicity (Begleiter et al.,
`2001; Brooks et al., 2001; Cao et al., 2003; Kim et al., 2003;
`Kwak et al., 2001, 2003; Nelson et al., 1999; Otieno et al.,
`2000; Prochaska and Fernandes, 1993; Spencer et al., 1990).
`Our study now shows that the cellular glutathione content,
`depleted by DMF and SP at 4 h (Kim et al., 2003; Nelson et
`al., 1999), is significantly increased by both compounds
`after 24 h of treatment. Glutathione, produced by astrocytes
`(Dringen and Hirrlinger, 2003; Cooper and Kristal, 1997;
`Langeveld et al., 1996), plays a prominent role in protecting
`neurons from free radical-induced damage (Pompella et al.,
`2003). Indeed, because NQO-1 and glutathione are well
`known to protect numerous cells against redox cycling,
`oxidative stress and neoplasia (Jaiswal, 2000; Joseph et al.,
`1994), the strong induction of NQO-1 mRNA and activity,
`as well as the increase in glutathione content,
`in LPS-
`stimulated co-cultures of astrocytes and microglia by DMF
`and SP suggest they could constitute powerful anti-oxidant
`effe

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket